Next Article in Journal
Application of Inflatable Video-Assisted Mediastinoscopic Transhiatal Esophagectomy in Individualized Treatment of Esophageal Cancer
Previous Article in Journal
Editorial of Special Issue “Oral Cancer: From Pathophysiology to Novel Therapeutic Approaches”
Previous Article in Special Issue
Oxidative Stress Biomarkers in Cystic Fibrosis and Cystic Fibrosis-Related Diabetes in Children: A Literature Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Complicated Relationship of Short-Chain Fatty Acids and Oral Microbiome: A Narrative Review

by
Georgy E. Leonov
1,*,
Yurgita R. Varaeva
1,
Elena N. Livantsova
1 and
Antonina V. Starodubova
1,2
1
Federal Research Center of Nutrition, Biotechnology and Food Safety, 109240 Moscow, Russia
2
Therapy Faculty, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(10), 2749; https://doi.org/10.3390/biomedicines11102749
Submission received: 13 September 2023 / Revised: 30 September 2023 / Accepted: 3 October 2023 / Published: 11 October 2023

Abstract

:
The human oral microbiome has emerged as a focal point of research due to its profound implications for human health. The involvement of short-chain fatty acids in oral microbiome composition, oral health, and chronic inflammation is gaining increasing attention. In this narrative review, the results of early in vitro, in vivo, and pilot clinical studies and research projects are presented in order to define the boundaries of this new complicated issue. According to the results, the current research data are disputable and ambiguous. When investigating the role of SCFAs in human health and disease, it is crucial to distinguish between their local GI effects and the systemic influences. Locally, SCFAs are a part of normal oral microbiota metabolism, but the increased formation of SCFAs usually attribute to dysbiosis; excess SCFAs participate in the development of local oral diseases and in oral biota gut colonization and dysbiosis. On the other hand, a number of studies have established the positive impact of SCFAs on human health as a whole, including the reduction of chronic systemic inflammation, improvement of metabolic processes, and decrease of some types of cancer incidence. Thus, a complex and sophisticated approach with consideration of origin and localization for SCFA function assessment is demanded. Therefore, more research, especially clinical research, is needed to investigate the complicated relationship of SCFAs with health and disease and their potential role in prevention and treatment.

1. Introduction

Nowadays, the human body microbiome is causing an increasing research interest [1]. The number of original papers devoted to the investigation of biota-associated axes has rocketed in the past decades due to the development of new ‘omics’ technologies. At the same time, the high prevalence of oral cavity pathology, including dental caries, periodontitis, and oral cancer, is also causing great concern worldwide [2]. Thus, the new focus for research is the oral flora, which has physiological, metabolic, immunological, mucosal protective, nutritional, and detoxification functions [3].
The human oral microbiome is composed of bacteria, archaea, viruses, fungi, and protozoa [4]. The oral cavity harbors 774 bacterial species, according to the expanded Human Oral Microbiome Database (HOMD), with 58% having formal names, 16% being unnamed but cultivated, and 26% known solely as uncultivated phylotypes [5]. Predominantly, Actinobacteria (genera Corynebacterium, Rothia, and Actinomyces), Bacteroidetes (genera Prevotella, Capnocytophaga, and Porphyromonas), Firmicutes (genera Streptococcus and Granulicatella), Fusobacteria (genus Fusobacterium), and Proteobacteria (genera Neisseria and Haemophilus) are found in the oral microbiomes of healthy individuals [6]. Furthermore, there is a reliable relationship between oral microbiota and systemic diseases, including inflammatory bowel disease (IBD), cancers, cardiovascular diseases, Alzheimer’s disease, diabetes, rheumatoid arthritis, and preterm birth [7,8,9,10,11,12,13].
The species diversity of oral microorganisms is influenced by various factors such as host genetics, gender, age, oral health, smoking, diet, socioeconomic factors, and medications [14,15,16,17,18,19,20].
Chronic inflammation and oxidative stress play a critical role in the pathogenesis of damage in both systemic and localized oral diseases [21]. Short-chain fatty acids (SCFAs) have an important anti-inflammatory function in the regulation of immune activity, contributing to the prevention of several chronic inflammatory conditions [22]. They can directly influence neutrophils by decreasing their production of reactive oxygen species (ROS) and myeloperoxidase (MPO) while also enhancing their apoptosis [23,24]. SCFAs, also known as volatile fatty acids, consist of organic linear carboxylic acids containing fewer than six carbons, which include formic acid (C1), acetic acid (C2), propionic acid (C3), isobutyric and butyric acids (C4), and isovaleric and valeric acids (C5) [25]. These SCFAs are generated by the bacterial fermentation of dietary fiber and resistant starch. Butyrate, propionate, and acetate make up approximately 90% to 95% of the SCFAs found in the colon [26].
The composition of a healthy gut microbiota is primarily composed of two phyla, Firmicutes and Bacteroidetes, accounting for nearly 90%, alongside less-represented phyla like Proteobacteria, Verrucomicrobia, or Actinobacteria. Acetate and propionate are the primary products of the Bacteroidetes phylum, while Firmicutes produces butyrate as its primary metabolic end product. SCFAs contribute to the integrity and permeability of the gut barrier through various mechanisms [27]. Primarily, butyrate increases the concentration of tight junction proteins, such as claudin-1 (CLDN1), zonula occludens-1 (ZO-1), and occludin, by activating the gene expression for encoding these proteins [28]. Additionally, butyrate reinforces the mucosal layer of the intestinal epithelium by upregulating the expression of mucin 2 [29].
SCFAs play a crucial role in regulating various physiological pathways within the nervous system. They modulate systemic and neuroinflammation by influencing the functions and structures of microglial cells, which subsequently impact emotions, cognitive functions, and mental disorders. Furthermore, it appears that high concentrations of SCFAs are associated with the core expression of neurotrophic factors. [30,31]. SCFAs, especially acetate, also play a critical role in appetite and human metabolism regulation [32]. Animal models have shown that diets high in fermentable carbohydrates, which are broken down in the colon to produce SCFAs, lead to appetite reduction [33]. SCFAs are the host’s energy metabolites that provide nearly 10% of the daily energy requirements for humans [34].
The mentioned data are not only theoretically valuable but also applicable to resolving various human health issues. The involvement of short-chain fatty acids in oral microbiome composition, oral health, and chronic inflammation, as well as the potential development of SCFA-based biomarkers and therapies aimed at improving oral microbiome health, oral disease, and chronic inflammation, are gaining increasing attention. To analyze the problem, we conducted a search on PubMed/MEDLINE, SCOPUS, and Google Scholar with the following MESH terms/keywords: “Oral microbiota AND short-chain fatty acids”, “Oral microbiota AND (oral diseases OR periodontitis OR oral cancer OR systemic diseases)”. In this narrative review, identified results of early in vitro, in vivo, and pilot clinical studies and research projects are presented in order to define the boundaries of this new complicated issue.

2. SCFAs and Oral Health

The oral bacteria are not pure planktonic in saliva but consist primarily of a structured community of aggregated bacterial cells (either from the same species or multi-species) embedded and enclosed in a self-produced extracellular polymeric matrix and adherent to an inert or living surface (the biofilms) [35]. Biofilms occur in the oral cavity as a normal physiological process, as well as in the colon or female reproductive tract, notwithstanding that biofilms are commonly associated with human pathology [36]. Inside biofilms, there are gradients of nutrients, signaling compounds, and bacterial waste products that result from environmental conditions and the physiological responses of bacteria to the local environment. Thus, within a consortium, bacteria can achieve multicellular metabolic coordination and expansion by utilizing additional metabolic pathways or engaging in cross-feeding of metabolic intermediates [37,38]. These adaptive responses heavily rely on the perception and processing of chemical information from the environment, which plays a central role in their regulatory control. Microorganisms that establish colonies in the oral cavity possess a diverse range of adaptive mechanisms encoded in their genes, enabling the oral microbiota to seamlessly adapt and integrate into an oral biofilm lifestyle. This adaptation involves optimizing their phenotypic characteristics to suit the specific environment [39]. As a consequence, microorganisms within biofilms exhibit distinct phenotypic traits compared to their planktonic counterparts.
The segregation of the oral microbiome into different niches can be attributed to several factors, including pH, salinity, redox potential, oxygen levels, and nutritional availability [40]. The microbial composition varies across various regions of the oral cavity, including the gingival sulcus, tongue, cheek, hard and soft palate, floor of the mouth, throat, saliva, and teeth, with each area presenting unique characteristics [41]. These interconnected intra-oral habitats can harbor different microbial profiles, collectively forming the oral microbiome as a meta-microbial community. Moreover, based on cluster analysis of microbial genera distribution in different oral niches, three distinct ‘metaniches’ with similar compositions have been identified. These metaniches are categorized based on the composition of bacteria and include the following: supragingival plaque and gingival crevicular fluid (P-GCF); saliva-tongue-hard palate (S-T-HP); and cheek and sublingual area (C-U). In saliva, the predominant species include Streptococcus, Prevotella, Veillonella, Neisseria, and Haemophilus. At the genus level, Streptococcus, Gemella, Veillonella, Haemophilus, Neisseria, Porphyromonas, Fusobacterium, Actinomyces, and Prevotella are consistently found across all healthy human oral sites, regardless of their niche location. Notably, Streptococcus is the dominant genus, followed by Haemophilus in the buccal mucosa, Actinomyces in the supragingival plaque, and Prevotella in the subgingival plaque. In the “P-GCF” metaniche, strong positive correlations were observed between cytokine levels and the relative abundance of genera such as Aggregatibacter, Fusobacterium, Gemella, and Streptococcus, while negative correlations were evident with Tannerella, Leptotrichia, Corynebacterium, Capnocytophaga, Saccharimononadae, and Neisseriacae. In the “S-T-HP” metaniche, Gemella spp. exhibited the highest positive correlations with the cytokine levels, and variable positive connections were identified for the genera Capnocytophaga, Streptococcus, Porphyromonas, and Campylobacter [42].
Within the oral microbiota, there exists a substantial number, approximately 1839, of biosynthetic gene clusters responsible for generating a diverse array of metabolites through various mechanisms [43]. SCFAs can be synthesized in the oral cavity using two primary approaches: carbohydrate hydrolysis or amino acid metabolism. Carbohydrates undergo fermentation, leading to the formation of monosaccharides, followed by the conversion to pyruvate through either the pentose phosphate pathway (C5) or glycolysis (C6) [44]. Bacterial species with the capability to convert sugars into SCFAs include Streptococcus, Actinomyces, Lactobacillus, Propionibacterium, and Prevotella [45]. In addition to food sources, oral bacteria can use salivary mucins, which cover the surface of the oral mucosa and contain glycans, as a nutrient source after they are fermented by sialidases [46]. Amino acids also offer a substrate for SCFA production through the process of transamination. The microbiota employs proteases and peptidases to break down proteins, and subsequent deamination of the resulting peptides and amino acids yields SCFAs in the oral cavity. Actinomyces, Veillonella, and Fusobacterium predominantly serve as the primary proteolytic bacteria [47]. Typical concentrations of SCFAs in the human oral cavity fall within the ranges of 6.3–16.2 mM for acetate, 1.2–3.1 mM for propionate, and 0.0–0.4 mM for butyrate [48]. The nature of the relationship between oral commensals, whether mutualistic or competitive, can be determined by the antimicrobial potential of these SCFAs. The antimicrobial potential of SCFAs can alter the structure of the oral microbiome to both support and inhibit bacterial growth [49].
Thus, high dietary carbohydrate intake leads to the proliferation of saccharolytic and acidogenic bacteria in supragingival plaques due to different strains. Eventually, this may cause the formation of ecosystems with increased cariogenic potential, which contributes to the demineralization of tooth surfaces and the development of oral diseases [50]. A recent study showed that the concentrations of SCFAs, as well as acetate alone, correlate with the abundance of the following bacterial species: T. denticola, Treponema socranskii, Filifactor alocis, T. forsythia, P. gingivalis, Porphyromonas endodontalis, Prevotella dentalis, and F. nucleatum. Butyrate has been linked to T. denticola, F. alocis, T. socraskii, F. nucleatum, T. forsythia, and P. gingivalis, while propionate has only been associated with T. denticola and F. alocis [51]. The data on certain oral bacteria production of SCFAs is given in Table 1.
Elevated concentrations of SCFAs have been associated with soft tissue damage and an increased inflammatory response within the oral cavity [55]. Substantial and reliable evidence suggests that SCFAs can exert adverse effects on periodontal cells [56]. Notably, exposure to SCFAs has been observed to induce oral epithelial cell death, primarily through pyroptosis and apoptosis mechanisms [57]. In an in vitro model, it was observed that acetate, propionate, and butyrate, each at a concentration of 12.5 mM, induced cell death. Propionate exhibited the highest level of toxicity, while acetate displayed the lowest toxicity levels [58,59]. Moreover, millimolar concentrations of butyrate were found to impair the integrity of the gingival epithelial barrier, resulting in the disruption of cell–cell junctions, an upregulation of caspase-3 and gasedermin-E expression, and the induction of cell pyroptosis [60]. Furthermore, the study revealed that treatment with butyrate and propionate led to an increased relative abundance of keratin proteins in gingival epithelial cells, particularly keratin K17 [61]. Additionally, butyrate demonstrated the capacity to reduce the cytokine-induced expression of adhesion molecule 1 (ICAM-1) in epithelial cells [62]. In addition, SCFAs have been found to suppress the expression of various components, including connexin 26 and 43, cadherin-1, adhesion molecule-1, claudin-1 and 4, as well as desmoglein-1 and desmocollin-2. Such suppression can result in increased epithelial permeability [63].
Oxidative stress has also been implicated in epithelial cell damage, particularly in gingival fibroblasts. Prolonged butyrate exposure to gingival fibroblasts may lead to cell apoptosis, an increase in BAX expression, and the activation of caspase 8 and caspase 9 [64,65]. Moreover, an in vivo study demonstrated that prolonged treatment with butyrate led to iron non-accumulation, ROS formation, glutathione depletion, lipid peroxidation, and eventual ferroptosis in periodontal pocket tissues. It should be highlighted that butyrate also plays a role in stimulating T-cell apoptosis, promoting osteoblast differentiation, and releasing pro-inflammatory cytokines from neutrophils [66].
Nevertheless, several studies have indicated the situational benefit of SCFAs. For instance, the reduction in local immune response caused by long-term antibiotic treatment of Candida albicans infections was partially compensated by the application of SCFAs. These fatty acids have been suggested to inhibit C. albicans growth and stimulate the expression of Foxp3 and IL-17A in CD4+ T cells [67]. Therefore, SCFAs play a complicated role in oral cavity health. SCFAs are synthesized by the oral microbiota as a part of normal local metabolic processes, whereas the increased formation of SCFAs may be considered a marker of oral dysbiosis. Furthermore, excess SCFAs participate in local oral cavity pathology development as well as in pro-inflammatory signaling.

3. SCFAs and Oral Diseases

Increased levels of SCFAs have been linked to the development of numerous oral diseases, including dental caries, periodontitis, oral cancer, and various viral infections [68]. SCFAs take part in the regulation of bacterial growth by reshaping the microbiome structure, thereby promoting the proliferation of predominantly periodontal pathogens. Furthermore, specific SCFAs contribute to the formation of biofilms on the oral cavity’s surface [69]. The mentioned harm inflicted on oral tissues, primarily by butyrate and propionate, also contributes to the progression of oral diseases. A common underlying mechanism associated with oral lesions involves an intensified inflammatory response and heightened oxidative stress [70]. Acetate, propionate, and butyrate have been identified as ligands for transmembrane G-protein receptors known as FFAR2 (GPR43), FFAR3 (GPR41), and GPR109a [71]. These receptor types are expressed in cells throughout the body and serve various functions. Among the diverse SCFAs, FFAR2 shows a preference for acetate and propionate as ligands, while FFAR3 primarily binds to butyrate. For instance, one of the functions of FFAR2 is to signal about excess energy intake, inhibit insulin signaling in adipocytes, and thus reduce fat accumulation in adipose tissue [72]. Also, activation of FFAR3 induces changes in the hematopoietic activity of the bone marrow, characterized by an increased production of macrophages and dendritic cell precursors. As a result, highly phagocytic dendritic cells can infiltrate the lung tissue during allergic inflammation, for instance, where they establish the immune microenvironment and influence the severity of the reaction [73]. Notably, within the context of the oral cavity, the FFAR2 receptor is expressed in various immune cell types, including monocytes, neutrophils, eosinophils, and regulatory T cells (Treg). The finding that FFAR2 activation, particularly through SCFA administration such as acetate, confers resistance against several bacterial and viral infections is of particular significance [74]. A common feature observed in these SCFA-mediated processes is the direct or indirect stimulation of leukocytes via FFAR2 activation [75]. The detailed data of some recent studies investigating the association of SCFAs with oral health and disease is presented in Table 2.
Describing the contribution of SCFAs to the pathogenesis of oral cavity diseases, it should be considered that they are generally multifactorial pathologies, and SCFAs participate in the development of the diseases as an element of the sophisticated pathological chain consistent with the list of numerous factors working in joint action.

3.1. SCFAs and Dental Caries

Dental caries, a chronic infectious disease, results in the demineralization of dental hard tissues. This demineralization arises from a complex interplay between the resident flora and fermentable carbohydrates found in plaque [76]. The pathological processes associated with caries involve numerous microorganisms, including Actinomyces gerencseriae, Bifidobacterium, S. mutans, Veillonella, S. salivarius, S. constellatus, S. parasanguinis, and Lactobacillus fermentum [77].
Table 2. Recent studies investigating the association of SCFAs with oral health and disease.
Table 2. Recent studies investigating the association of SCFAs with oral health and disease.
DiseaseAuthorsYearSCFAsPopulationBrief Results
PeriodontitisK Hatanaka et al. [78]2022C2 and
C3–C6
10 healthy participants and 10 participants with mild and severe periodontal diseaseNo significant difference in saliva was observed between healthy participants and patients. However, C3–C6 significantly differed between mild and severe periodontal disease.
JC Provenzano et al. [79]2014All18 adult patients (ages ranging from 20 to 39 years) with asymptomatic apical periodontitisBoth propionate and butyrate were found in most of the root canal tissue samples examined. Before treatment, the predominant bacteria were F. nucleatum and members of the Actinobacteria phylum, and after treatment, Streptococcus.
OJ Park et al. [80]2023ButyrateTHP-1 cell line and six-week-old female Sprague Dawley ratsButyrate combined with lipoteichoic acid significantly increased caspase-1 activation and IL-1β secretion both in vitro and in vivo in a rat model of apical periodontitis.
AD Rudin et al. [81]2021AllPrimary human neutrophilsP. gingivalis from both lab strains and clinical specimens has been shown to produce significant amounts of SCFAs. A similar mixture of SCFAs induces Ca2+ signaling and chemotaxis in human neutrophils through activation of FFAR2.
L. Qiqiang et al. [82]2012All37 individuals (21 patients with chronic periodontitis and 16 periodontally healthy controls)Periodontal treatment decreased the concentration of lactic, propionic, butyric, and isovaleric acids in the gingival crevicular fluid to the level of a healthy control group. The formic acid concentration increased. A rebound effect was observed for all SCFAs within 2–6 months.
R Lu et al.
[48]
2014All40 individuals (20 with generalized aggressive periodontitis, 20 healthy controls)Formic acid concentration increased significantly, and acetic, propionic, and butyric acid concentrations decreased after conservative treatment of periodontitis. Sites containing P gingivalis, T denticola, P intermedia, or F. nucleatum showed a similar correlation.
N Murakami et al. [83]2022ButyrateMale C57BL/6 miceButyrate modulates periodontal mechanical nociception via FFAR3 signaling in P. gingivalis-induced periodontitis.
ME Cueno et al. [84]2018Butyrate10-week-old male Wistar ratsAdministration of butyrate (5 mM) resulted in increased NADPH-related oxidative stress and inflammation, presumably mediated by MMP-9, in a rat model of periodontitis.
S Ji et al.
[85]
2023All16 individuals (8 with periodontitis, 8 healthy controls)A total of 570 human proteins associated with inflammation, cell death, and metabolism were found to be differentially expressed in periodontitis. Microbial proteins associated with butyrate metabolism were upregulated in the periodontitis group.
R Lu et al.
[86]
2013All34 individuals (20 with generalized aggressive periodontitis, 14 healthy controls)Patients with periodontitis had significantly higher concentrations of succinic, acetic, propionic, butyric, and isovaleric acids and a higher -abundance of both P. gingivalis and T. denticola. The level of SCFAs correlated positively with the number of these bacteria.
HS Na et al.
[51]
2021All112 individuals (79 with periodontitis, 33 healthy controls)SCFAs correlated with the abundance of the following bacterial species in the periodontitis cohort: T. denticola, Treponema socranskii, Filifactor alocis, T. forsythia, P. gingivalis, Porphyromonas endodontalis, Prevotella dentalis, and F. nucleatum.
AD Rudin et al. [87]2021Acetate and ButyratePrimary human neutrophilsPredominantly acetate and butyrate, which are released in large amounts as end products of F. nucleatum metabolism, induce human neutrophil chemotaxis and cytosolic Ca 2+ signals via the FFAR2 receptor.
Oral cancerZ Nouri et al. [88]2023All309 adult cancer patients and 745 healthy controlsLeuconostoc, Streptococcus, Abiotrophia, and Prevotella were decreased in the cancer group, while Haemophilus and Neisseria were increased. Total SCFA and FFAR2 expression levels were higher in the control group, while TNFAIP8, IL6, and STAT3 levels were higher in the cancer group.
Y Miyazaki et al. [89]2010ButyrateCa9-22, HSC-2, -3, and -4 cellsButyrate induced the expression of podoplanin in HSC-2 and -3 cells and vimentin in Ca9-22 cells. Cell migration was stimulated at low concentrations of butyrate, especially in HSC-3 cells, while it was inhibited in HSC-2 and -4 cells.
HIVK Imai et al. [90]2009ButyrateACH-2 and U1 cellsP. gingivalis produces high levels of butyric acid, which acts as an HDAC inhibitor and induces histone acetylation, leading to the induction of RNA polymerase II and the reactivation of HIV-1.
Epstein–Barr virusK Imai et al.
[91]
2012ButyrateDaudi cell lineThe butyric acid of P. gingivalis stimulates the expression of ZEBRA and inhibits HDAC, which results in increased histone acetylation, increased activity of the BZLF1 gene transcription, and the induction of EBV reactivation.
KSHVX Yu et al.
[92]
2014AllBCBL1 cellsSCFAs dose-dependently and synergistically stimulate lytic expression of KSHV genes. By transactivating viral chromatin, SCFAs inhibit HDAC, suppress the expression of SIRT1, EZH2, and SUV39H1, increase acetylation, and decrease histone trimethylation.
Dental cariesJ Wu et al.
[93]
2022AcetateBiofilm in vitroLactobacillus casei in a multi-species biofilm has a competitive advantage over S. mutans due to acetate production.
T Park et al. [94]2021AllBiofilm in vitroA mix of SCFAs inhibits S. gordonii biofilm formation by downregulating comD and comE mRNAs, which are regulators of the CSP pathway.
The extent of contribution by oral bacteria to dental caries hinges on metabolic pathways that encompass the absorption and metabolism of carbohydrates, including the production of SCFAs. Among these microorganisms, S. mutans plays a pivotal role as a major initiator of dental caries. This is due to its capacity to synthesize exopolysaccharides, metabolize carbohydrates into organic acids, particularly lactic acid, and maintain a low pH environment within the oral cavity [93]. SCFAs, as organic acids, also contribute to the environment conducive to the proliferation of acidogenic bacteria. On the other hand, the most relevant producers of SCFAs in the oral cavity are generally periodontal pathogens such as P. gingivalis and F. nucleatum. These bacteria compete with the main saccharolytic bacteria and actually increase the pH of the environment through amino acid metabolism, whereas they also directly inhibit the growth of cariogenic bacteria caused by SCFAs [95].
Studies centered on oral biofilms have revealed that acetate, propionate, and butyrate exhibit effective inhibitory activity against the formation of S. gordonii biofilms [94]. Furthermore, research has demonstrated that Lactobacillus casei possesses the capability to suppress the growth of S. mutans, including the conversion of lactate into acetate [95]. While it has been hypothesized that Veillonella’s conversion of lactic acid, produced by streptococci, into less potent acids like acetic acid may reduce the host’s susceptibility to caries, experimental evidence supporting this notion is lacking. Conversely, a molecular study suggests that Veillonella coexists with Streptococcus in carious lesions [96]. Understanding the complex relationships between oral bacteria and their metabolic pathways is essential for developing effective strategies for dental caries prevention and management.

3.2. SCFAs and Periodontitis

Periodontitis is a chronic inflammatory disease characterized by the destruction of the alveolar bone and periodontal tissues, leading to clinical manifestations such as deepening gum pockets and tooth loss. Early signs of periodontitis may involve bleeding during tooth-brushing, tooth mobility during eating, and conditions like halitosis (bad mouth odor), with additional signs emerging at later stages [97]. The dysbiosis of the oral microbiome, particularly within the periodontal pocket, represents a primary factor in the pathogenesis of periodontitis [98]. The most important periodontal pathogens are Gram-negative rod-shaped bacteria such as P. gingivalis, P. intermedia, T. forsithia, T. denticola, and F. nucleatum [99]. The development of periodontal diseases is significantly influenced by the formation of biofilms. Butyrate has been shown to stimulate fimbrilin-dependent colonization of Actinomyces oris and promote biofilm growth in the early stages of biofilm formation [100]. Higher expression of enzymes involved in F. nucleatum butyrate production is observed during biofilm formation compared to planktonic growth, suggesting the importance of butyrate as a virulence factor for certain bacteria [101]. SCFA-producing bacteria such as Porphyromonas gingivalis, Tannerella forsythia, Prevotella intermedia, and Treponema denticola also contribute to the multi-species expansion of biofilms [102]. Research has demonstrated significant alterations in local concentrations of SCFAs among subjects with periodontitis. Specifically, in the gingival crevicular fluid of individuals with chronic periodontitis, notably higher levels of propionate (11.68 ± 8.84 vs. 5.87 ± 3.35 mM) and butyrate (3.11 ± 1.86 vs. 1.10 ± 0.87 mM) have been observed when compared to their healthy counterparts. Furthermore, individuals with aggressive periodontitis exhibited elevated concentrations of acetic, propionic, and butyric acids (26.0 versus 11.3, 8.8 versus 2.1, and 2.5 versus 0.0 mM, respectively) [48]. The evidence suggests that periodontal treatment leads to a reduction in the concentrations of lactic, propionic, butyric, and isovaleric acids in the sulcular fluid, effectively returning them to levels comparable to those of a healthy control group. Although the post-treatment concentrations of formic acid tend to increase, these effects persist for several months [82]. Patients with periodontitis have significantly higher concentrations of succinic, acetic, propionic, butyric, and isovaleric acids, as well as a predominance of both P. gingivalis and T. denticola, compared to healthy individuals. The level of SCFAs positively correlates with the number of these bacteria [86]. In addition to the periodontal pocket, SCFAs, including propionate and butyrate, were detected in tissue samples from the root canals during the treatment of apical periodontitis. It should be pointed out that prior to treatment, the predominant bacterial species included F. nucleatum and members of the Actinobacteria phylum, but after treatment, representatives of the Streptococcus genus were prevalent [79]. In a study on a murine periodontitis model, butyrate was shown to modulate periodontal mechanical nociception through FFAR3 signaling, which may explain the often observed absence of pain in periodontitis [83]. Studies in vitro have shown that mainly acetate and butyrate, which are released in large amounts as metabolic end products of F. nucleatum and P. gingivalis, induce human neutrophil chemotaxis and cytosolic Ca2+ signals via the FFAR2 receptor [81,87]. However, SCFAs in the oral cavity may not only act locally; a recent study in rats found that butyrate injected directly into the gum was able to enter the systemic circulation and adversely affect distant organs [84]. To summarize, SCFAs play a critical role in periodontitis pathogenesis.

3.3. SCFAs and Oral Cancer

Oral cancer comprises a substantial portion of head and neck cancers, accounting for 48%, with approximately 90% of oral cancer cases being histologically classified as oral squamous cell carcinoma [103]. Acidogenic and aciduric species play a significant role in facilitating the invasion and metastasis of malignant cells. This is achieved through the promotion of an acidic tumor microenvironment via acid production [104]. Numerous in vitro studies have underscored the significance of acidic conditions in amplifying the metastatic potential of tumor cells [105]. Furthermore, SCFAs have been implicated in inhibiting the development of an effective immune response against tumor cells. They accomplish this process by attracting myeloid suppressive cells, thus expanding the population of immunosuppressive cells within the tumor microenvironment [106]. Butyrate has been observed to suppress the cytokine-induced expression of intercellular ICAM-1 on oral squamous carcinoma cells [62], which results in the promotion of leukocyte transmigration to pathologically affected areas and the activation of macrophage polarization toward the M2 tumor phenotype [107].
Human studies indicate a potential connection between the oral microbiota and carcinogenesis in several organs, including the gastrointestinal tract, head and neck, oral cavity, and pancreas. Moreover, recent research on the genera Lactobacillus and Streptococcus has identified the production of volatile sulfur compounds, SCFAs, reactive oxygen species, reactive nitrogen species, hydrogen peroxide, and lactate, which are associated with carcinogenesis, chronic inflammation, genomic instability, and tumor angiogenesis [88]. Also, investigations have shown that P. gingivalis promotes the development and progression of oral cancer by promoting oral cell proliferation and inducing the expression of key molecules such as nuclear factor kappa B (NF-κB), IL-6, signal transducer and activator of transcription 3 (STAT3), cyclin D1, and matrix metallopeptidase 9 (MMP-9) [108]. The relationship between F. nucleatum and the development of oral cancer has been extensively researched. Epidemiological studies indicate that F. nucleatum is the most prevalent species colonizing oral mucosal sites affected by tumors. Although F. nucleatum is positively correlated with oral cancer, the bacterium is specifically less common in advanced cancers and is also associated with better survival [109]. F. nucleatum is a significant producer of SCFAs, especially propionate and butyrate, and while there has been previous evidence of F. nucleatum’s ability to activate neutrophils via the FFAR2 pathway, there is no direct evidence of F. nucleatum’s effect on cancer development via SCFAs [110]. In a recent study investigating the correlation between oral microbiota, their metabolites, and the development of different cancer types, it was discovered that cancer patients had lower levels of Leuconostoc, Streptococcus, Abiotropia, and Prevotella. Conversely, the concentration of Haemophilus and Neisseria increased in this group. The control group presented higher expression levels of SCFA and FFAR2, while the cancer group had higher levels of TNFAIP8, IL6, and STAT3 [88]. Therefore, early-stage studies suggest that some SCFA-producing bacteria may promote cancer development in the oral cavity. However, SCFAs’ effects are associated with possible activation of immune suppression of carcinogenesis.

3.4. SCFAs and Viral Disease

The oral cavity is a site of active viral replication and shedding, including human immunodeficiency viruses (HIV) and herpesviruses, Kaposi’s sarcoma-associated herpesvirus (KSHV), and Epstein–Barr virus (EBV) [111]. Although these viruses are the causative agents of their respective infections, the oral microbiome can have a significant impact on them. Disruption of host-microbial homeostasis in the epithelial tissues of the oral cavity contributes to viral replication [112], which is characterized by a change in the composition of the polymicrobial community of the oral cavity towards a dysbiotic and often pathogenic one. Consequently, the described shifting contributes to overactivation of the immune system and inflammatory conditions. S. mutans, Lactobacillus, Candida, Haemophilus parahaemolyticus, Actinomyces, Neisseria subflava, and Corynebacterium diphtheriae species are more prevalent in the saliva of HIV patients, while Streptococcus mitis is less frequently found [113]. Furthermore, the potential increase in opportunistic bacterial load, which can facilitate HIV reactivation and expedite the progression of AIDS, exists in the context of AIDS-associated immunosuppression. Notably, periodontitis-associated bacteria-produced SCFAs have demonstrated the capability to reactivate KSHV. This reactivation process is mediated through the inhibition of histone deacetylase (HDAC) and the downregulation of various epigenetic modulators (EZH2, SUV39H1) responsible for histone trimethylation repression. As a result, exposition of SCFAs to KSHV-infected cells exhibits histone hyperacetylation, thereby facilitating the transactivation of viral chromatin [92]. Consequently, individuals afflicted with periodontal diseases may exhibit increased lytic reactivation of KSHV and a higher risk of the development of oral Kaposi’s sarcoma. Comparable effects of butyrate were described for Epstein–Barr virus activation [91].
Overall, in some medical conditions, viruses can replicate in the oral cavity, inducing complicated relationships with the oral microbiome. SCFAs are one of the factors that determine the interaction between the virus and the epithelial cells of the oral cavity due to immune mechanisms and epigenetic modifications.

4. SCFAs and the Oral–Gut Axis

The gut’s production of SCFAs is contingent on the composition of the gut microbiota, available nutrients, and overall gut health [114]. SCFAs are absorbed within the cecum and colon through mechanisms involving proton-ion-dependent nonionic diffusion and active transport mediated by Na+- and H+-conjugated monocarboxylate transporters [115]. Within the intestine, SCFA concentrations range from 20 to 140 mM and exhibit segmental variation, with higher levels typically detected in the proximal colon [116]. The colon’s SCFA rates are higher than in the oral cavity, even in the presence of periodontitis. Nevertheless, this disparity does not result in damage to the cells of the intestinal epithelium. This apparent paradox can be elucidated by examining the structural differences between the oral and intestinal mucosa [117] (Figure 1). In contrast, the intestine is composed of a simple columnar monolayer epithelium characterized by high permeability [118]. SCFAs are actively absorbed by intestinal cells, where they are partially metabolized as an energy source and partially transported and disseminated through the bloodstream. Conversely, the oral mucosa lacks the capacity to modulate SCFA levels, thereby maintaining high local concentrations. Moreover, both the intestinal and oral mucosa are coated with a layer of mucus, a viscous secretion primarily composed of mucins. Mucus serves a protective role for epithelial tissue and can also retain specific substances. The thickness of the oral mucosa is estimated at 70–100 µm, whereas the intestinal mucosal layer comprises a loosely attached outer layer approximately 150–400 µm thick and an inner mucosal layer estimated at 800–900 µm thick [119,120]. These structural differences between the oral and intestinal mucosa indicate that the intestinal epithelium boasts a diverse array of adaptive mechanisms aimed at minimizing the adverse effects of SCFAs [121].
Thus, an appreciation of these varieties highlights the importance of context-specific considerations when studying the effects of SCFAs on different mucosal tissues.

4.1. Oral Bacteria and SCFAs Production in the Gut

The acidic environment of the stomach, the alkaline environment of the gut, and the abundance of digestive enzymes provide a natural barrier for oral microbes to penetrate the lower gastrointestinal tract. Another important factor is the resistance to colonization mediated by the commensal gut microbiota, which eliminates oral bacteria [122]. There are conflicting data about the possibility of colonization of the gut microbiome by oral microorganisms in healthy individuals [123]. It has been noted that pathological conditions of the stomach, such as achlorhydria, promote colonization [124]. Moreover, barrier function was found to decrease with age and the presence of diseases such as alcoholism, cirrhosis, HIV infection, inflammatory bowel disease, colorectal cancer, and rheumatoid arthritis [125,126,127,128,129,130,131]. In a murine model transplant of the human oral microbiome, it has been demonstrated that Actinomyces, Streptococcus, Haemophilus, Veillonella, Fusobacterium, and Trichococcus are capable of colonizing the distal colon [132].
Moreover, in a rodent model of periodontitis induced by F. nucleatum, researchers detected the migration of oral bacteria to the intestinal tract, concomitant with shifts in the gut microbiota. Specifically, the gut ecosystem exhibited modifications, with a notable reduction in Verrucomicrobia and an elevation in Proteobacteria evident within a two-week timeframe, followed by subsequent increases in Bacteroidetes and Firmicutes observed to the four-week time point [133]. For in vivo study on the C57BL/6 mouse strain, the amount of Klebsiella and Enterobacter species in the oral cavity and intestine increased in periodontitis; pathobionts promoted Th17 cell activation and colitis development [134]. In another in vivo study, a model of P. gingivalis periodontitis found an increase in Bacteroidetes and a decrease in Firmicutes, but subsequent analysis did not detect specific P. gingivalis DNA in the gut. In contrast, in another murine study, a single injection of P. gingivalis similarly altered the gut microbiota after 48 h in comparison to a control group [135,136].
Pilot clinical studies demonstrate comparable data. A clinical study of 44 participants showed that Firmicutes, Proteobacteria, Verrucomicrobia, and Euryarchaeota were increased, while Bacteroidetes were decreased in patients with chronic periodontitis. Representatives of Mogibacteriaceae, Ruminococcaceae, and Prevotella significantly differentiated individuals with oral disease from healthy controls [137]. Another study found a correlation between Veillonella levels in saliva and feces, indicating intestinal colonization by this microorganism; hypertension was also detected as a factor contributing to the spread of Veillonella in the gut [138]. A. actinomycetemcomitans has been shown to colonize the intestine and negatively affect lipid metabolism and insulin resistance [139]. Patients with type 1 diabetes have been shown to have decreased S. salivarius and increased S. mutans, reflected by an increased abundance of facultative anaerobes, including Enterobacteria [140].
Alterations in the composition of the microbiota may lead to changes in the production of SCFAs. Although there is evidence that the Bacteroides/Firmicutes ratio does not affect the total concentration of SCFAs, an increase in this ratio appears to lead to a relative elevation in the concentration of acetate and propionate and a decrease in butyrate [141]. Two-week oral administration of S. mitis, S. salivarius, P. gingivalis, or P. nigrescens to mice led to an increase in Staphylococcus and Bacteroides and a decrease in Lactobacillus spp in the gut, as well as a pronounced decrease in the level of butyric acid in the feces [142]. In human studies, Lachnospiraceae, a family of butyrate-producing clostridia, were associated with fewer oral bacteria in the gut, apparently improving resistance to colonization [143] in patients with colorectal cancer. Peptostreptococcus, Parvimonas, and Fusobacterium were associated with tumor growth in this group but were also found in healthy individuals.
From the above, it can be concluded that oral bacteria, especially opportunistic species, can colonize the intestine and affect the gut microbiome, leading to changes in the production of various metabolites, including SCFAs. This may contribute to the development of diseases of the digestive tract.

4.2. Oral and Systemic Effects of Gut Bacteria

Although the spread of oral microorganisms to the gut is fairly obvious due to the anatomical features of the digestive tract, the ability of gut bacteria to influence the oral microbiome is not so straightforward. Emerging evidence suggests that SCFAs produced in the gut support overall bone health. An increase in serum SCFA levels correlates directly with enhanced production of insulin-like growth factor 1 (IGF-1) and the proliferation and differentiation of regulatory T cells, which, in turn, diminish osteoclast activity and foster the production of WNT10b [144,145]. Consequently, research has shown that administration of SCFAs or implementation of a high-fiber diet can mitigate postmenopausal and inflammation-induced bone loss in murine models. Remarkably, propionate and butyrate exhibit the ability to inhibit osteoclast activity via the suppression of NFATc1 and TRAF6 gene expression, both in vitro and in vivo [146]. In an osteoporosis model on rats, the treatment of the gut with periodontitis patients’ saliva resulted in increased bone loss, possibly attributable to alterations in the lipolysis processes of tryptophan metabolism [147]. Considering a high-fat diet, its administration led to a reduction in Bacteroidetes and an increase in Firmicutes populations within the gut microbiota of C57BL/6J mice. This dietary shift subsequently resulted in decreased gingival blood flow and diminished blood SCFA levels, potentially culminating in long-term alveolar bone deterioration and the onset of periodontal disease [148]. Exploring the connection between intestinal microbiome composition and periodontitis risk, nine taxa were identified as associated with an increased risk of periodontitis: Prevotella, Lachnospiraceae, Enterobacteriales, Pasteurellales, Enterobacteriaceae, Pasteurellaceae, Bacteroidales, Alistipes, and Eisenbergiella groups. Conversely, Butyricicoccus and Ruminiclostridium were linked to a reduced risk of periodontitis [149].
Investigating the links between gut bacteria and the development of periodontitis, a clinical study demonstrated significant differences in the oral microbiome between individuals with IBD and healthy controls. The IBD group was characterized by higher levels of the Veillonella and Prevotella genera, while Neisseria, Streptococcus, Haemophilus, and Fusobacterium were associated with a healthy gut microbiome. It should be highlighted that Streptococcus was the sole microorganism simultaneously found in the saliva and feces of IBD patients [150].
The research data point out the complex interplay between oral and gut microbiota and their potential impact on systemic health, particularly in relation to bone health and periodontal disease. Further research projects are demanded to clarify the precise mechanisms underlying these interactions and their clinical implications.

5. Conclusions

The complicated relationship between the human oral microbiome, chronic inflammation, and SCFAs, as well as the health implications, is causing a growing research interest nowadays. The current research data assessing the effects of SCFAs on human health are disputable and ambiguous. When investigating the role of SCFAs in human health and disease, it is crucial to distinguish between their local GI effects and the systemic influences. Locally, SCFAs are a part of normal oral microbiota metabolism, but the increased formation of SCFAs usually attributes to dysbiosis. Moreover, excess SCFAs participate in the development of local diseases, including microbiome structure alterations, periodontal pathogen growth, a direct cytotoxic effect on the oral mucosa, consecutive damage to oral tissues, etc. On the other hand, a number of studies have established the positive impact of SCFAs on human health as a whole, including the reduction of chronic systemic inflammation and the improvement of carbohydrates and lipid metabolism. Furthermore, according to current data, serum SCFA levels are epidemiologically associated with a lower incidence of some types of cancer.
Thus, a complex and sophisticated approach with consideration of origin and localization for SCFA function assessment is demanded. Therefore, more research, especially clinical research, is needed to investigate the complicated relationship of SCFAs with health and disease and their potential role in prevention and treatment.

Author Contributions

Design and the main concept of the paper, G.E.L., Y.R.V. and A.V.S.; writing—original draft, G.E.L.; visual material preparation, G.E.L.; funding acquisition, A.V.S.; writing—review and editing, G.E.L., Y.R.V., E.N.L. and A.V.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Russian Science Foundation, grant number 22-15-00252, https://www.rscf.ru/en/project/22-15-00252/ (accessed on 31 August 2023).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors acknowledge the BioRender team for providing the artwork creation online service.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the writing of the manuscript.

References

  1. Tidjani Alou, M.; Naud, S.; Khelaifia, S.; Bonnet, M.; Lagier, J.-C.; Raoult, D. State of the Art in the Culture of the Human Microbiota: New Interests and Strategies. Clin. Microbiol. Rev. 2020, 34, e00129-19. [Google Scholar] [CrossRef] [PubMed]
  2. Peres, M.A.; Macpherson, L.M.D.; Weyant, R.J.; Daly, B.; Venturelli, R.; Mathur, M.R.; Listl, S.; Celeste, R.K.; Guarnizo-Herreño, C.C.; Kearns, C.; et al. Oral Diseases: A Global Public Health Challenge. Lancet 2019, 394, 249–260. [Google Scholar] [CrossRef] [PubMed]
  3. Bacali, C.; Vulturar, R.; Buduru, S.; Cozma, A.; Fodor, A.; Chiș, A.; Lucaciu, O.; Damian, L.; Moldovan, M.L. Oral Microbiome: Getting to Know and Befriend Neighbors, a Biological Approach. Biomedicines 2022, 10, 671. [Google Scholar] [CrossRef]
  4. Morrison, A.G.; Sarkar, S.; Umar, S.; Lee, S.T.M.; Thomas, S.M. The Contribution of the Human Oral Microbiome to Oral Disease: A Review. Microorganisms 2023, 11, 318. [Google Scholar] [CrossRef] [PubMed]
  5. HOMD. Human Oral Microbiome Database. Available online: https://www.homd.org/ (accessed on 31 August 2023).
  6. Li, X.; Liu, Y.; Yang, X.; Li, C.; Song, Z. The Oral Microbiota: Community Composition, Influencing Factors, Pathogenesis, and Interventions. Front. Microbiol. 2022, 13, 895537. [Google Scholar] [CrossRef] [PubMed]
  7. Read, E.; Curtis, M.A.; Neves, J.F. The Role of Oral Bacteria in Inflammatory Bowel Disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 731–742. [Google Scholar] [CrossRef]
  8. Irfan, M.; Delgado, R.Z.R.; Frias-Lopez, J. The Oral Microbiome and Cancer. Front. Immunol. 2020, 11, 591088. [Google Scholar] [CrossRef]
  9. Tonelli, A.; Lumngwena, E.N.; Ntusi, N.A.B. The Oral Microbiome in the Pathophysiology of Cardiovascular Disease. Nat. Rev. Cardiol. 2023, 20, 386–403. [Google Scholar] [CrossRef]
  10. Maitre, Y.; Mahalli, R.; Micheneau, P.; Delpierre, A.; Amador, G.; Denis, F. Evidence and Therapeutic Perspectives in the Relationship between the Oral Microbiome and Alzheimer’s Disease: A Systematic Review. Int. J. Environ. Res. Public. Health 2021, 18, 11157. [Google Scholar] [CrossRef]
  11. Qin, H.; Li, G.; Xu, X.; Zhang, C.; Zhong, W.; Xu, S.; Yin, Y.; Song, J. The Role of Oral Microbiome in Periodontitis under Diabetes Mellitus. J. Oral Microbiol. 2022, 14, 2078031. [Google Scholar] [CrossRef]
  12. Chu, X.J.; Cao, N.W.; Zhou, H.Y.; Meng, X.; Guo, B.; Zhang, H.Y.; Li, B.Z. Oral and Gut Microbiome in Rheumatoid Arthritis Patients: A Systematic Review. Rheumatology 2021, 60, 1054–1066. Available online: https://academic.oup.com/rheumatology/article/60/3/1054/6101642 (accessed on 31 August 2023). [CrossRef] [PubMed]
  13. Jang, H.; Patoine, A.; Wu, T.T.; Castillo, D.A.; Xiao, J. Oral Microflora and Pregnancy: A Systematic Review and Meta-Analysis. Sci. Rep. 2021, 11, 16870. [Google Scholar] [CrossRef]
  14. Mukherjee, C.; Moyer, C.O.; Steinkamp, H.M.; Hashmi, S.B.; Beall, C.J.; Guo, X.; Ni, A.; Leys, E.J.; Griffen, A.L. Acquisition of Oral Microbiota Is Driven by Environment, Not Host Genetics. Microbiome 2021, 9, 54. [Google Scholar] [CrossRef] [PubMed]
  15. Zhao, J.; Zhou, Y.-H.; Zhao, Y.-Q.; Feng, Y.; Yan, F.; Gao, Z.-R.; Ye, Q.; Chen, Y.; Liu, Q.; Tan, L.; et al. Gender Variations in the Oral Microbiomes of Elderly Patients with Initial Periodontitis. J. Immunol. Res. 2021, 2021, 7403042. [Google Scholar] [CrossRef] [PubMed]
  16. Schwartz, J.L.; Peña, N.; Kawar, N.; Zhang, A.; Callahan, N.; Robles, S.J.; Griebel, A.; Adami, G.R. Old Age and Other Factors Associated with Salivary Microbiome Variation. BMC Oral Health 2021, 21, 490. [Google Scholar] [CrossRef]
  17. Wu, J.; Peters, B.A.; Dominianni, C.; Zhang, Y.; Pei, Z.; Yang, L.; Ma, Y.; Purdue, M.P.; Jacobs, E.J.; Gapstur, S.M.; et al. Cigarette Smoking and the Oral Microbiome in a Large Study of American Adults. ISME J. 2016, 10, 2435–2446. [Google Scholar] [CrossRef]
  18. Santonocito, S.; Giudice, A.; Polizzi, A.; Troiano, G.; Merlo, E.M.; Sclafani, R.; Grosso, G.; Isola, G. A Cross-Talk between Diet and the Oral Microbiome: Balance of Nutrition on Inflammation and Immune System’s Response during Periodontitis. Nutrients 2022, 14, 2426. [Google Scholar] [CrossRef]
  19. Renson, A.; Jones, H.E.; Beghini, F.; Segata, N.; Zolnik, C.P.; Usyk, M.; Moody, T.U.; Thorpe, L.; Burk, R.; Waldron, L.; et al. Sociodemographic Variation in the Oral Microbiome. Ann. Epidemiol. 2019, 35, 73–80.e2. [Google Scholar] [CrossRef]
  20. Cheng, X.; He, F.; Si, M.; Sun, P.; Chen, Q. Effects of Antibiotic Use on Saliva Antibody Content and Oral Microbiota in Sprague Dawley Rats. Front. Cell. Infect. Microbiol. 2022, 12, 721691. [Google Scholar] [CrossRef]
  21. Kumar, J.; Teoh, S.L.; Das, S.; Mahakknaukrauh, P. Oxidative Stress in Oral Diseases: Understanding Its Relation with Other Systemic Diseases. Front. Physiol. 2017, 8, 693. [Google Scholar] [CrossRef]
  22. Zhang, Z.; Zhang, H.; Chen, T.; Shi, L.; Wang, D.; Tang, D. Regulatory Role of Short-Chain Fatty Acids in Inflammatory Bowel Disease. Cell Commun. Signal. CCS 2022, 20, 64. [Google Scholar] [CrossRef]
  23. Sun, Y.; Zhou, C.; Chen, Y.; He, X.; Gao, F.; Xue, D. Quantitative Increase in Short-Chain Fatty Acids, Especially Butyrate Protects Kidney from Ischemia/Reperfusion Injury. J. Investig. Med. 2022, 70, 29–35. [Google Scholar] [CrossRef]
  24. Andrade-Oliveira, V.; Amano, M.T.; Correa-Costa, M.; Castoldi, A.; Felizardo, R.J.F.; de Almeida, D.C.; Bassi, E.J.; Moraes-Vieira, P.M.; Hiyane, M.I.; Rodas, A.C.D.; et al. Gut Bacteria Products Prevent AKI Induced by Ischemia-Reperfusion. J. Am. Soc. Nephrol. JASN 2015, 26, 1877–1888. [Google Scholar] [CrossRef] [PubMed]
  25. Huang, C.B.; Altimova, Y.; Myers, T.M.; Ebersole, J.L. Short- and Medium-Chain Fatty Acids Exhibit Antimicrobial Activity for Oral Microorganisms. Arch. Oral Biol. 2011, 56, 650–654. [Google Scholar] [CrossRef]
  26. Kim, K.N.; Yao, Y.; Ju, S.Y. Short Chain Fatty Acids and Fecal Microbiota Abundance in Humans with Obesity: A Systematic Review and Meta-Analysis. Nutrients 2019, 11, 2512. [Google Scholar] [CrossRef]
  27. Hou, K.; Wu, Z.-X.; Chen, X.-Y.; Wang, J.-Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in Health and Diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef] [PubMed]
  28. Lyu, W.; Yang, H.; Li, N.; Lu, L.; Yang, C.; Jin, P.; Xiao, Y. Molecular Characterization, Developmental Expression, and Modulation of Occludin by Early Intervention with Clostridium butyricum in Muscovy Ducks. Poult. Sci. 2021, 100, 101271. [Google Scholar] [CrossRef] [PubMed]
  29. Hodgkinson, K.; Abbar, F.E.; Dobranowski, P.; Manoogian, J.; Butcher, J.; Figeys, D.; Mack, D.; Stintzi, A. Butyrate’s Role in Human Health and the Current Progress towards Its Clinical Application to Treat Gastrointestinal Disease. Clin. Nutr. 2023, 42, 61–75. [Google Scholar] [CrossRef]
  30. Caetano-Silva, M.E.; Rund, L.; Hutchinson, N.T.; Woods, J.A.; Steelman, A.J.; Johnson, R.W. Inhibition of Inflammatory Microglia by Dietary Fiber and Short-Chain Fatty Acids. Sci. Rep. 2023, 13, 2819. [Google Scholar] [CrossRef]
  31. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef]
  32. Frost, G.; Sleeth, M.L.; Sahuri-Arisoylu, M.; Lizarbe, B.; Cerdan, S.; Brody, L.; Anastasovska, J.; Ghourab, S.; Hankir, M.; Zhang, S.; et al. The Short-Chain Fatty Acid Acetate Reduces Appetite via a Central Homeostatic Mechanism. Nat. Commun. 2014, 5, 3611. [Google Scholar] [CrossRef] [PubMed]
  33. Lange, O.; Proczko-Stepaniak, M.; Mika, A. Short-Chain Fatty Acids—A Product of the Microbiome and Its Participation in Two-Way Communication on the Microbiome-Host Mammal Line. Curr. Obes. Rep. 2023, 12, 108–126. [Google Scholar] [CrossRef] [PubMed]
  34. Hurst, N.R.; Kendig, D.M.; Murthy, K.S.; Grider, J.R. The Short Chain Fatty Acids, Butyrate and Propionate, Have Differential Effects on the Motility of the Guinea Pig Colon. Neurogastroenterol. Motil. 2014, 26, 1586–1596. [Google Scholar] [CrossRef] [PubMed]
  35. Radaic, A.; Kapila, Y.L. The Oralome and Its Dysbiosis: New Insights into Oral Microbiome-Host Interactions. Comput. Struct. Biotechnol. J. 2021, 19, 1335–1360. [Google Scholar] [CrossRef]
  36. Perry, E.K.; Tan, M.-W. Bacterial biofilms in the human body: Prevalence and impacts on health and disease. Front. Cell. Infect. Microbiol. 2023, 13, 1237164. [Google Scholar] [CrossRef]
  37. Ng, H.M.; Kin, L.X.; Dashper, S.G.; Slakeski, N.; Butler, C.A.; Reynolds, E.C. Bacterial Interactions in Pathogenic Subgingival Plaque. Microb. Pathog. 2016, 94, 60–69. [Google Scholar] [CrossRef]
  38. Uruén, C.; Chopo-Escuin, G.; Tommassen, J.; Mainar-Jaime, R.C.; Arenas, J. Biofilms as Promoters of Bacterial Antibiotic Resistance and Tolerance. Antibiotics 2020, 10, 3. [Google Scholar] [CrossRef]
  39. Rossi, E.; Paroni, M.; Landini, P. Biofilm and Motility in Response to Environmental and Host-related Signals in Gram Negative Opportunistic Pathogens. J. Appl. Microbiol. 2018, 125, 1587–1602. [Google Scholar] [CrossRef]
  40. Kilian, M.; Chapple, I.L.C.; Hannig, M.; Marsh, P.D.; Meuric, V.; Pedersen, A.M.L.; Tonetti, M.S.; Wade, W.G.; Zaura, E. The Oral Microbiome—An Update for Oral Healthcare Professionals. Br. Dent. J. 2016, 221, 657–666. [Google Scholar] [CrossRef]
  41. Giordano-Kelhoffer, B.; Lorca, C.; March Llanes, J.; Rábano, A.; del Ser, T.; Serra, A.; Gallart-Palau, X. Oral Microbiota, Its Equilibrium and Implications in the Pathophysiology of Human Diseases: A Systematic Review. Biomedicines 2022, 10, 1803. [Google Scholar] [CrossRef]
  42. Seidel, C.L.; Gerlach, R.G.; Wiedemann, P.; Weider, M.; Rodrian, G.; Hader, M.; Frey, B.; Gaipl, U.S.; Bozec, A.; Cieplik, F.; et al. Defining Metaniches in the Oral Cavity According to Their Microbial Composition and Cytokine Profile. Int. J. Mol. Sci. 2020, 21, 8218. [Google Scholar] [CrossRef] [PubMed]
  43. Donia, M.S.; Cimermancic, P.; Schulze, C.J.; Wieland Brown, L.C.; Martin, J.; Mitreva, M.; Clardy, J.; Linington, R.G.; Fischbach, M.A. A Systematic Analysis of Biosynthetic Gene Clusters in the Human Microbiome Reveals a Common Family of Antibiotics. Cell 2014, 158, 1402–1414. [Google Scholar] [CrossRef] [PubMed]
  44. Takahashi, N. Oral Microbiome Metabolism: From “Who Are They?” to “What Are They Doing?”. J. Dent. Res. 2015, 94, 1628–1637. [Google Scholar] [CrossRef] [PubMed]
  45. Li, Y.; Zhu, M.; Liu, Y.; Luo, B.; Cui, J.; Huang, L.; Chen, K.; Liu, Y. The Oral Microbiota and Cardiometabolic Health: A Comprehensive Review and Emerging Insights. Front. Immunol. 2022, 13, 1010368. [Google Scholar] [CrossRef] [PubMed]
  46. Ndeh, D.; Gilbert, H.J. Biochemistry of Complex Glycan Depolymerisation by the Human Gut Microbiota. FEMS Microbiol. Rev. 2018, 42, 146–164. [Google Scholar] [CrossRef]
  47. Norimatsu, Y.; Kawashima, J.; Takano-Yamamoto, T.; Takahashi, N. Nitrogenous Compounds Stimulate Glucose-Derived Acid Production by Oral Streptococcus and Actinomyces. Microbiol. Immunol. 2015, 59, 501–506. [Google Scholar] [CrossRef]
  48. Lu, R.; Meng, H.; Gao, X.; Xu, L.; Feng, X. Effect of Non-Surgical Periodontal Treatment on Short Chain Fatty Acid Levels in Gingival Crevicular Fluid of Patients with Generalized Aggressive Periodontitis. J. Periodontal Res. 2014, 49, 574–583. [Google Scholar] [CrossRef]
  49. Xu, Y.; Zhu, Y.; Li, X.; Sun, B. Dynamic Balancing of Intestinal Short-Chain Fatty Acids: The Crucial Role of Bacterial Metabolism. Trends Food Sci. Technol. 2020, 100, 118–130. [Google Scholar] [CrossRef]
  50. Tennert, C.; Reinmuth, A.; Bremer, K.; Al-Ahmad, A.; Karygianni, L.; Hellwig, E.; Vach, K.; Ratka-Krüger, P.; Wittmer, A.; Woelber, J.P. An Oral Health Optimized Diet Reduces the Load of Potential Cariogenic and Periodontal Bacterial Species in the Supragingival Oral Plaque: A Randomized Controlled Pilot Study. MicrobiologyOpen 2020, 9, e1056. [Google Scholar] [CrossRef]
  51. Na, H.S.; Kim, S.; Kim, S.; Yu, Y.; Kim, S.Y.; Kim, H.-J.; Lee, J.Y.; Lee, J.-H.; Chung, J. Molecular Subgroup of Periodontitis Revealed by Integrated Analysis of the Microbiome and Metabolome in a Cross-Sectional Observational Study. J. Oral Microbiol. 2021, 13, 1902707. [Google Scholar] [CrossRef]
  52. Ochiai, K.; Kurita-Ochiai, T. Effects of Butyric Acid on the Periodontal Tissue. Jpn. Dent. Sci. Rev. 2009, 45, 75–82. [Google Scholar] [CrossRef]
  53. Okumura, S.; Konishi, Y.; Narukawa, M.; Sugiura, Y.; Yoshimoto, S.; Arai, Y.; Sato, S.; Yoshida, Y.; Tsuji, S.; Uemura, K.; et al. Gut Bacteria Identified in Colorectal Cancer Patients Promote Tumourigenesis via Butyrate Secretion. Nat. Commun. 2021, 12, 5674. [Google Scholar] [CrossRef] [PubMed]
  54. Mirković, B.; Murray, M.A.; Lavelle, G.M.; Molloy, K.; Azim, A.A.; Gunaratnam, C.; Healy, F.; Slattery, D.; McNally, P.; Hatch, J.; et al. The Role of Short-Chain Fatty Acids, Produced by Anaerobic Bacteria, in the Cystic Fibrosis Airway. Am. J. Respir. Crit. Care Med. 2015, 192, 1314–1324. [Google Scholar] [CrossRef] [PubMed]
  55. Tsuda, H.; Ochiai, K.; Suzuki, N.; Otsuka, K. Butyrate, a Bacterial Metabolite, Induces Apoptosis and Autophagic Cell Death in Gingival Epithelial Cells. J. Periodontal Res. 2010, 45, 626–634. [Google Scholar] [CrossRef] [PubMed]
  56. Magrin, G.L.; Strauss, F.J.; Benfatti, C.A.M.; Maia, L.C.; Gruber, R. Effects of Short-Chain Fatty Acids on Human Oral Epithelial Cells and the Potential Impact on Periodontal Disease: A Systematic Review of In Vitro Studies. Int. J. Mol. Sci. 2020, 21, 4895. [Google Scholar] [CrossRef]
  57. Guan, X.; Li, W.; Meng, H. A Double-Edged Sword: Role of Butyrate in the Oral Cavity and the Gut. Mol. Oral Microbiol. 2021, 36, 121–131. [Google Scholar] [CrossRef]
  58. Zhang, J.; Kashket, S. Cytotoxic Effects of Short-Chain Carboxylic Acids on Human Gingival Epithelial Cells. Oral Microbiol. Immunol. 1997, 12, 345–349. [Google Scholar] [CrossRef]
  59. Shirasugi, M.; Nishioka, K.; Yamamoto, T.; Nakaya, T.; Kanamura, N. Normal Human Gingival Fibroblasts Undergo Cytostasis and Apoptosis after Long-Term Exposure to Butyric Acid. Biochem. Biophys. Res. Commun. 2017, 482, 1122–1128. [Google Scholar] [CrossRef]
  60. Liu, J.; Wang, Y.; Meng, H.; Yu, J.; Lu, H.; Li, W.; Lu, R.; Zhao, Y.; Li, Q.; Su, L. Butyrate Rather than LPS Subverts Gingival Epithelial Homeostasis by Downregulation of Intercellular Junctions and Triggering Pyroptosis. J. Clin. Periodontol. 2019, 46, 894–907. [Google Scholar] [CrossRef]
  61. Pöllänen, M.T.; Salonen, J.I. Effect of Short Chain Fatty Acids on Human Gingival Epithelial Cell Keratins in Vitro. Eur. J. Oral Sci. 2000, 108, 523–529. [Google Scholar] [CrossRef]
  62. Magrin, G.L.; Di Summa, F.; Strauss, F.-J.; Panahipour, L.; Mildner, M.; Magalhães Benfatti, C.A.; Gruber, R. Butyrate Decreases ICAM-1 Expression in Human Oral Squamous Cell Carcinoma Cells. Int. J. Mol. Sci. 2020, 21, 1679. [Google Scholar] [CrossRef] [PubMed]
  63. Alamri, A.; Semlali, A.; Jacques, É.; Alanazi, M.; Zakrzewski, A.; Chmielewski, W.; Rouabhia, M. Long-Term Exposure of Human Gingival Fibroblasts to Cigarette Smoke Condensate Reduces Cell Growth by Modulating Bax, Caspase-3 and P53 Expression. J. Periodontal Res. 2015, 50, 423–433. [Google Scholar] [CrossRef] [PubMed]
  64. Santos, A.F.P.; Cervantes, L.C.C.; Panahipour, L.; Souza, F.Á.; Gruber, R. Proof-of-Principle Study Suggesting Potential Anti-Inflammatory Activity of Butyrate and Propionate in Periodontal Cells. Int. J. Mol. Sci. 2022, 23, 11006. [Google Scholar] [CrossRef] [PubMed]
  65. Zhao, Y.; Li, J.; Guo, W.; Li, H.; Lei, L. Periodontitis-Level Butyrate-Induced Ferroptosis in Periodontal Ligament Fibroblasts by Activation of Ferritinophagy. Cell Death Discov. 2020, 6, 119. [Google Scholar] [CrossRef] [PubMed]
  66. Whibley, N.; MacCallum, D.M.; Vickers, M.A.; Zafreen, S.; Waldmann, H.; Hori, S.; Gaffen, S.L.; Gow, N.A.R.; Barker, R.N.; Hall, A.M. Expansion of Foxp3+ T-Cell Populations by Candida albicans Enhances Both Th17-Cell Responses and Fungal Dissemination after Intravenous Challenge. Eur. J. Immunol. 2014, 44, 1069–1083. [Google Scholar] [CrossRef] [PubMed]
  67. Dekaboruah, E.; Suryavanshi, M.V.; Chettri, D.; Verma, A.K. Human Microbiome: An Academic Update on Human Body Site Specific Surveillance and Its Possible Role. Arch. Microbiol. 2020, 202, 2147–2167. [Google Scholar] [CrossRef]
  68. Hojo, K.; Nagaoka, S.; Ohshima, T.; Maeda, N. Bacterial Interactions in Dental Biofilm Development. J. Dent. Res. 2009, 88, 982–990. [Google Scholar] [CrossRef]
  69. Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 227. [Google Scholar]
  70. Martin-Gallausiaux, C.; Marinelli, L.; Blottière, H.M.; Larraufie, P.; Lapaque, N. SCFA: Mechanisms and Functional Importance in the Gut. Proc. Nutr. Soc. 2021, 80, 37–49. [Google Scholar] [CrossRef]
  71. May, K.S.; den Hartigh, L.J. Gut Microbial-Derived Short Chain Fatty Acids: Impact on Adipose Tissue Physiology. Nutrients 2023, 15, 272. [Google Scholar] [CrossRef]
  72. Corrêa-Oliveira, R.; Fachi, J.L.; Vieira, A.; Sato, F.T.; Vinolo, M.A.R. Regulation of Immune Cell Function by Short-Chain Fatty Acids. Clin. Transl. Immunol. 2016, 5, e73. [Google Scholar] [CrossRef]
  73. Ney, L.-M.; Wipplinger, M.; Grossmann, M.; Engert, N.; Wegner, V.D.; Mosig, A.S. Short Chain Fatty Acids: Key Regulators of the Local and Systemic Immune Response in Inflammatory Diseases and Infections. Open Biol. 2023, 13, 230014. [Google Scholar] [CrossRef] [PubMed]
  74. Sencio, V.; Barthelemy, A.; Tavares, L.P.; Machado, M.G.; Soulard, D.; Cuinat, C.; Queiroz-Junior, C.M.; Noordine, M.-L.; Salomé-Desnoulez, S.; Deryuter, L.; et al. Gut Dysbiosis during Influenza Contributes to Pulmonary Pneumococcal Superinfection through Altered Short-Chain Fatty Acid Production. Cell Rep. 2020, 30, 2934–2947.e6. [Google Scholar] [CrossRef] [PubMed]
  75. Schlatterer, K.; Peschel, A.; Kretschmer, D. Short-Chain Fatty Acid and FFAR2 Activation—A New Option for Treating Infections? Front. Cell. Infect. Microbiol. 2021, 11, 785833. [Google Scholar] [CrossRef]
  76. Valm, A.M. The Structure of Dental Plaque Microbial Communities in the Transition from Health to Dental Caries and Periodontal Disease. J. Mol. Biol. 2019, 431, 2957–2969. [Google Scholar] [CrossRef]
  77. Peters, B.M.; Jabra-Rizk, M.A.; O’May, G.A.; Costerton, J.W.; Shirtliff, M.E. Polymicrobial Interactions: Impact on Pathogenesis and Human Disease. Clin. Microbiol. Rev. 2012, 25, 193–213. [Google Scholar] [CrossRef] [PubMed]
  78. Hatanaka, K.; Shirahase, Y.; Yoshida, T.; Kono, M.; Toya, N.; Sakasegawa, S.; Konishi, K.; Yamamoto, T.; Ochiai, K.; Takashiba, S. Enzymatic Measurement of Short-Chain Fatty Acids and Application in Periodontal Disease Diagnosis. PLoS ONE 2022, 17, e0268671. [Google Scholar] [CrossRef] [PubMed]
  79. Provenzano, J.C.; Rôças, I.N.; Tavares, L.F.D.; Neves, B.C.; Siqueira, J.F. Short-Chain Fatty Acids in Infected Root Canals of Teeth with Apical Periodontitis before and after Treatment. J. Endod. 2015, 41, 831–835. [Google Scholar] [CrossRef]
  80. Park, O.-J.; Ha, Y.-E.; Sim, J.-R.; Lee, D.; Lee, E.-H.; Kim, S.-Y.; Yun, C.-H.; Han, S.H. Butyrate Potentiates Enterococcus faecalis Lipoteichoic Acid-Induced Inflammasome Activation via Histone Deacetylase Inhibition. Cell Death Discov. 2023, 9, 107. [Google Scholar] [CrossRef]
  81. Dahlstrand Rudin, A.; Khamzeh, A.; Venkatakrishnan, V.; Persson, T.; Gabl, M.; Savolainen, O.; Forsman, H.; Dahlgren, C.; Christenson, K.; Bylund, J. Porphyromonas gingivalis Produce Neutrophil Specific Chemoattractants Including Short Chain Fatty Acids. Front. Cell. Infect. Microbiol. 2021, 10, 620681. [Google Scholar] [CrossRef]
  82. Qiqiang, L.; Huanxin, M.; Xuejun, G. Longitudinal Study of Volatile Fatty Acids in the Gingival Crevicular Fluid of Patients with Periodontitis before and after Nonsurgical Therapy. J. Periodontal Res. 2012, 47, 740–749. [Google Scholar] [CrossRef]
  83. Murakami, N.; Yoshikawa, K.; Tsukada, K.; Kamio, N.; Hayashi, Y.; Hitomi, S.; Kimura, Y.; Shibuta, I.; Osada, A.; Sato, S. Butyric Acid Modulates Periodontal Nociception in Porphyromonas Gingivalis-Induced Periodontitis. J. Oral Sci. 2022, 64, 91–94. Available online: https://www.jstage.jst.go.jp/article/josnusd/64/1/64_21-0483/_article/-char/ja/ (accessed on 31 August 2023). [CrossRef] [PubMed]
  84. Cueno, M.E.; Ochiai, K. Gingival Periodontal Disease (PD) Level-Butyric Acid Affects the Systemic Blood and Brain Organ: Insights Into the Systemic Inflammation of Periodontal Disease. Front. Immunol. 2018, 9, 1158. [Google Scholar] [CrossRef]
  85. Ji, S.; Kook, J.-K.; Park, S.-N.; Lim, Y.K.; Choi, G.H.; Jung, J.-S. Characteristics of the Salivary Microbiota in Periodontal Diseases and Potential Roles of Individual Bacterial Species To Predict the Severity of Periodontal Disease. Microbiol. Spectr. 2023, 11, e0432722. Available online: https://journals.asm.org/doi/full/10.1128/spectrum.04327-22 (accessed on 31 August 2023). [CrossRef]
  86. Lu, R.; Feng, L.; Gao, X.; Meng, H.; Feng, X. Relationship between volatile fatty acids and Porphyromonas gingivalis and Treponema denticola in gingival crevicular fluids of patients with aggressive periodontitis. Beijing Da Xue Xue Bao 2013, 45, 12–16. [Google Scholar] [PubMed]
  87. Dahlstrand Rudin, A.; Khamzeh, A.; Venkatakrishnan, V.; Basic, A.; Christenson, K.; Bylund, J. Short Chain Fatty Acids Released by Fusobacterium nucleatum Are Neutrophil Chemoattractants Acting via Free Fatty Acid Receptor 2 (FFAR2). Cell. Microbiol. 2021, 23, e13348. [Google Scholar] [CrossRef] [PubMed]
  88. Nouri, Z.; Choi, S.W.; Choi, I.J.; Ryu, K.W.; Woo, S.M.; Park, S.-J.; Lee, W.J.; Choi, W.; Jung, Y.-S.; Myung, S.-K.; et al. Exploring Connections between Oral Microbiota, Short-Chain Fatty Acids, and Specific Cancer Types: A Study of Oral Cancer, Head and Neck Cancer, Pancreatic Cancer, and Gastric Cancer. Cancers 2023, 15, 2898. [Google Scholar] [CrossRef] [PubMed]
  89. Miyazaki, Y.; Kikuchi, K.; González-Alva, P.; Inoue, H.; Noguchi, Y.; Tsuchiya, H.; Hayashi, J.; Shin, K.; Ochiai, K.; Kusama, K. Association of Butyric Acid Produced by Periodontopathic Bacteria with Progression of Oral Cancer. J. Cancer Sci. Ther. 2010, 2, 26–32. [Google Scholar] [CrossRef]
  90. Imai, K.; Ochiai, K.; Okamoto, T. Reactivation of Latent HIV-1 Infection by the Periodontopathic Bacterium Porphyromonas gingivalis Involves Histone Modification1. J. Immunol. 2009, 182, 3688–3695. [Google Scholar] [CrossRef]
  91. Imai, K.; Inoue, H.; Tamura, M.; Cueno, M.E.; Inoue, H.; Takeichi, O.; Kusama, K.; Saito, I.; Ochiai, K. The Periodontal Pathogen Porphyromonas gingivalis Induces the Epstein–Barr Virus Lytic Switch Transactivator ZEBRA by Histone Modification. Biochimie 2012, 94, 839–846. [Google Scholar] [CrossRef]
  92. Yu, X.; Shahir, A.-M.; Sha, J.; Feng, Z.; Eapen, B.; Nithianantham, S.; Das, B.; Karn, J.; Weinberg, A.; Bissada, N.F.; et al. Short-Chain Fatty Acids from Periodontal Pathogens Suppress Histone Deacetylases, EZH2, and SUV39H1 To Promote Kaposi’s Sarcoma-Associated Herpesvirus Replication. J. Virol. 2014, 88, 4466–4479. [Google Scholar] [CrossRef] [PubMed]
  93. Wu, J.; Jiang, X.; Yang, Q.; Zhang, Y.; Wang, C.; Huang, R. Inhibition of Streptococcus mutans Biofilm Formation by the Joint Action of Oxyresveratrol and Lactobacillus casei. Appl. Environ. Microbiol. 2022, 88, e02436-21. [Google Scholar] [CrossRef] [PubMed]
  94. Park, T.; Im, J.; Kim, A.R.; Lee, D.; Jeong, S.; Yun, C.-H.; Han, S.H. Short-Chain Fatty Acids Inhibit the Biofilm Formation of Streptococcus Gordonii through Negative Regulation of Competence-Stimulating Peptide Signaling Pathway. J. Microbiol. 2021, 59, 1142–1149. [Google Scholar] [CrossRef]
  95. Nyvad, B.; Takahashi, N. Integrated Hypothesis of Dental Caries and Periodontal Diseases. J. Oral Microbiol. 2020, 12, 1710953. [Google Scholar] [CrossRef] [PubMed]
  96. Mashima, I.; Nakazawa, F. Interaction between Streptococcus Spp. and Veillonella tobetsuensis in the Early Stages of Oral Biofilm Formation. J. Bacteriol. 2015, 197, 2104–2111. [Google Scholar] [CrossRef] [PubMed]
  97. Mann, J.; Bernstein, Y.; Findler, M. Periodontal Disease and Its Prevention, by Traditional and New Avenues. Exp. Ther. Med. 2020, 19, 1504. [Google Scholar] [CrossRef]
  98. Siddiqui, R.; Badran, Z.; Boghossian, A.; Alharbi, A.M.; Alfahemi, H.; Khan, N.A. The Increasing Importance of the Oral Microbiome in Periodontal Health and Disease. Future Sci. OA 2023, 9, FSO856. [Google Scholar] [CrossRef]
  99. Mohanty, R.; Asopa, S.J.; Joseph, M.D.; Singh, B.; Rajguru, J.P.; Saidath, K.; Sharma, U. Red Complex: Polymicrobial Conglomerate in Oral Flora: A Review. J. Fam. Med. Prim. Care 2019, 8, 3480–3486. [Google Scholar] [CrossRef]
  100. Suzuki, I.; Shimizu, T.; Senpuku, H. Short Chain Fatty Acids Induced the Type 1 and Type 2 Fimbrillin-Dependent and Fimbrillin-Independent Initial Attachment and Colonization of Actinomyces oris Monoculture but Not Coculture with Streptococci. BMC Microbiol. 2020, 20, 329. [Google Scholar] [CrossRef]
  101. Llama-Palacios, A.; Potupa, O.; Sánchez, M.C.; Figuero, E.; Herrera, D.; Sanz, M. Proteomic Analysis of Fusobacterium nucleatum Growth in Biofilm versus Planktonic State. Mol. Oral Microbiol. 2020, 35, 168–180. [Google Scholar] [CrossRef]
  102. Zhu, Y.; Dashper, S.G.; Chen, Y.-Y.; Crawford, S.; Slakeski, N.; Reynolds, E.C. Porphyromonas gingivalis and Treponema denticola Synergistic Polymicrobial Biofilm Development. PLoS ONE 2013, 8, e71727. [Google Scholar] [CrossRef]
  103. Irani, S. New Insights into Oral Cancer—Risk Factors and Prevention: A Review of Literature. Int. J. Prev. Med. 2020, 11, 202. [Google Scholar] [CrossRef] [PubMed]
  104. Karpiński, T.M. Role of Oral Microbiota in Cancer Development. Microorganisms 2019, 7, 20. [Google Scholar] [CrossRef] [PubMed]
  105. Liu, C.; Wang, M.; Zhang, H.; Li, C.; Zhang, T.; Liu, H.; Zhu, S.; Chen, J. Tumor Microenvironment and Immunotherapy of Oral Cancer. Eur. J. Med. Res. 2022, 27, 198. [Google Scholar] [CrossRef]
  106. Kostic, A.D.; Chun, E.; Robertson, L.; Glickman, J.N.; Gallini, C.A.; Michaud, M.; Clancy, T.E.; Chung, D.C.; Lochhead, P.; Hold, G.L.; et al. Fusobacterium nucleatum Potentiates Intestinal Tumorigenesis and Modulates the Tumor-Immune Microenvironment. Cell Host Microbe 2013, 14, 207–215. [Google Scholar] [CrossRef] [PubMed]
  107. Ji, J.; Shu, D.; Zheng, M.; Wang, J.; Luo, C.; Wang, Y.; Guo, F.; Zou, X.; Lv, X.; Li, Y.; et al. Microbial Metabolite Butyrate Facilitates M2 Macrophage Polarization and Function. Sci. Rep. 2016, 6, 24838. [Google Scholar] [CrossRef]
  108. Wang, S.; Zhang, X.; Wang, G.; Cao, B.; Yang, H.; Jin, L.; Cui, M.; Mao, Y. Syndecan-1 Suppresses Cell Growth and Migration via Blocking JAK1/STAT3 and Ras/Raf/MEK/ERK Pathways in Human Colorectal Carcinoma Cells. BMC Cancer 2019, 19, 1160. [Google Scholar] [CrossRef]
  109. Fan, Z.; Tang, P.; Li, C.; Yang, Q.; Xu, Y.; Su, C.; Li, L. Fusobacterium nucleatum and Its Associated Systemic Diseases: Epidemiologic Studies and Possible Mechanisms. J. Oral Microbiol. 2022, 15, 2145729. [Google Scholar] [CrossRef]
  110. Brennan, C.A.; Clay, S.L.; Lavoie, S.L.; Bae, S.; Lang, J.K.; Fonseca-Pereira, D.; Rosinski, K.G.; Ou, N.; Glickman, J.N.; Garrett, W.S. Fusobacterium nucleatum Drives a Pro-Inflammatory Intestinal Microenvironment through Metabolite Receptor-Dependent Modulation of IL-17 Expression. Gut Microbes 2021, 13, 1987780. [Google Scholar] [CrossRef]
  111. Johnson, N.W.; Anaya-Saavedra, G.; Webster-Cyriaque, J. Viruses and Oral Diseases in HIV-Infected Individuals on Long-Term Antiretroviral Therapy: What Are the Risks and What Are the Mechanisms? Oral Dis. 2020, 26, 80–90. [Google Scholar] [CrossRef]
  112. Coker, M.O.; Cairo, C.; Garzino-Demo, A. HIV-Associated Interactions Between Oral Microbiota and Mucosal Immune Cells: Knowledge Gaps and Future Directions. Front. Immunol. 2021, 12, 676669. [Google Scholar] [CrossRef] [PubMed]
  113. Coker, M.O.; Mongodin, E.F.; El-Kamary, S.S.; Akhigbe, P.; Obuekwe, O.; Omoigberale, A.; Langenberg, P.; Enwonwu, C.; Hittle, L.; Blattner, W.A.; et al. Immune Status, and Not HIV Infection or Exposure, Drives the Development of the Oral Microbiota. Sci. Rep. 2020, 10, 10830. [Google Scholar] [CrossRef] [PubMed]
  114. Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023, 15, 2211. [Google Scholar] [CrossRef]
  115. Deleu, S.; Machiels, K.; Raes, J.; Verbeke, K.; Vermeire, S. Short Chain Fatty Acids and Its Producing Organisms: An Overlooked Therapy for IBD? eBioMedicine 2021, 66, 103293. [Google Scholar] [CrossRef] [PubMed]
  116. Cong, J.; Zhou, P.; Zhang, R. Intestinal Microbiota-Derived Short Chain Fatty Acids in Host Health and Disease. Nutrients 2022, 14, 1977. [Google Scholar] [CrossRef]
  117. Gomez-Casado, C.; Sanchez-Solares, J.; Izquierdo, E.; Díaz-Perales, A.; Barber, D.; Escribese, M.M. Oral Mucosa as a Potential Site for Diagnosis and Treatment of Allergic and Autoimmune Diseases. Foods 2021, 10, 970. [Google Scholar] [CrossRef]
  118. Kulkarni, D.H.; Newberry, R.D. Intestinal Macromolecular Transport Supporting Adaptive Immunity. Cell. Mol. Gastroenterol. Hepatol. 2019, 7, 729–737. [Google Scholar] [CrossRef]
  119. Derrien, M.; van Passel, M.W.J.; van de Bovenkamp, J.H.B.; Schipper, R.; de Vos, W.; Dekker, J. Mucin-Bacterial Interactions in the Human Oral Cavity and Digestive Tract. Gut Microbes 2010, 1, 254–268. [Google Scholar] [CrossRef]
  120. Atuma, C.; Strugala, V.; Allen, A.; Holm, L. The Adherent Gastrointestinal Mucus Gel Layer: Thickness and Physical State in Vivo. Am. J. Physiol.-Gastrointest. Liver Physiol. 2001, 280, G922–G929. [Google Scholar] [CrossRef]
  121. Cueno, M.E.; Ochiai, K. Re-Discovering Periodontal Butyric Acid: New Insights on an Old Metabolite. Microb. Pathog. 2016, 94, 48–53. [Google Scholar] [CrossRef]
  122. Kampmann, C.; Dicksved, J.; Engstrand, L.; Rautelin, H. Composition of Human Faecal Microbiota in Resistance to Campylobacter Infection. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2016, 22, 61.e1–61.e8. [Google Scholar] [CrossRef] [PubMed]
  123. Schmidt, T.S.; Hayward, M.R.; Coelho, L.P.; Li, S.S.; Costea, P.I.; Voigt, A.Y.; Wirbel, J.; Maistrenko, O.M.; Alves, R.J.; Bergsten, E.; et al. Extensive Transmission of Microbes along the Gastrointestinal Tract. eLife 2019, 8, e42693. [Google Scholar] [CrossRef] [PubMed]
  124. Sarker, S.A.; Ahmed, T.; Brüssow, H. Hunger and Microbiology: Is a Low Gastric Acid-induced Bacterial Overgrowth in the Small Intestine a Contributor to Malnutrition in Developing Countries? Microb. Biotechnol. 2017, 10, 1025–1030. [Google Scholar] [CrossRef] [PubMed]
  125. Singh, H.; Torralba, M.G.; Moncera, K.J.; DiLello, L.; Petrini, J.; Nelson, K.E.; Pieper, R. Gastro-Intestinal and Oral Microbiome Signatures Associated with Healthy Aging. GeroScience 2019, 41, 907–921. [Google Scholar] [CrossRef] [PubMed]
  126. Hu, L.; Ni, Z.; Zhao, K.; Li, X.; Gao, X.; Kang, Y.; Yu, Z.; Qin, Y.; Zhao, J.; Peng, W.; et al. The Association between Oral and Gut Microbiota in Male Patients with Alcohol Dependence. Front. Microbiol. 2023, 14, 1203678. [Google Scholar] [CrossRef]
  127. Qin, N.; Yang, F.; Li, A.; Prifti, E.; Chen, Y.; Shao, L.; Guo, J.; Le Chatelier, E.; Yao, J.; Wu, L.; et al. Alterations of the Human Gut Microbiome in Liver Cirrhosis. Nature 2014, 513, 59–64. [Google Scholar] [CrossRef]
  128. Lozupone, C.A.; Li, M.; Campbell, T.B.; Flores, S.C.; Linderman, D.; Gebert, M.J.; Knight, R.; Fontenot, A.P.; Palmer, B.E. Alterations in the Gut Microbiota Associated with HIV-1 Infection. Cell Host Microbe 2013, 14, 329–339. [Google Scholar] [CrossRef]
  129. Rojas-Tapias, D.F.; Brown, E.M.; Temple, E.R.; Onyekaba, M.A.; Mohamed, A.M.T.; Duncan, K.; Schirmer, M.; Walker, R.L.; Mayassi, T.; Pierce, K.A.; et al. Inflammation-Associated Nitrate Facilitates Ectopic Colonization of Oral Bacterium Veillonella parvula in the Intestine. Nat. Microbiol. 2022, 7, 1673–1685. [Google Scholar] [CrossRef]
  130. Zheng, X.; Liu, R.; Zhou, C.; Yu, H.; Luo, W.; Zhu, J.; Liu, J.; Zhang, Z.; Xie, N.; Peng, X.; et al. ANGPTL4-Mediated Promotion of Glycolysis Facilitates the Colonization of Fusobacterium nucleatum in Colorectal Cancer. Cancer Res. 2021, 81, 6157–6170. [Google Scholar] [CrossRef]
  131. Kitamura, K.; Shionoya, H.; Suzuki, S.; Fukai, R.; Uda, S.; Abe, C.; Takemori, H.; Nishimura, K.; Baba, H.; Katayama, K.; et al. Oral and Intestinal Bacterial Substances Associated with Disease Activities in Patients with Rheumatoid Arthritis: A Cross-Sectional Clinical Study. J. Immunol. Res. 2022, 2022, e6839356. [Google Scholar] [CrossRef]
  132. Li, B.; Ge, Y.; Cheng, L.; Zeng, B.; Yu, J.; Peng, X.; Zhao, J.; Li, W.; Ren, B.; Li, M.; et al. Oral Bacteria Colonize and Compete with Gut Microbiota in Gnotobiotic Mice. Int. J. Oral Sci. 2019, 11, 1–9. [Google Scholar] [CrossRef] [PubMed]
  133. Haraga, H.; Sato, T.; Watanabe, K.; Hamada, N.; Tani-Ishii, N. Effect of the Progression of Fusobacterium nucleatum–Induced Apical Periodontitis on the Gut Microbiota. J. Endod. 2022, 48, 1038–1045. [Google Scholar] [CrossRef] [PubMed]
  134. Kitamoto, S.; Nagao-Kitamoto, H.; Jiao, Y.; Gillilland, M.G.; Hayashi, A.; Imai, J.; Sugihara, K.; Miyoshi, M.; Brazil, J.C.; Kuffa, P.; et al. The Intermucosal Connection between the Mouth and Gut in Commensal Pathobiont-Driven Colitis. Cell 2020, 182, 447–462.e14. [Google Scholar] [CrossRef] [PubMed]
  135. 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]
  136. 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. Available online: https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0134234 (accessed on 6 September 2023). [CrossRef]
  137. Lourenςo, 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]
  138. Chen, B.-Y.; Lin, W.-Z.; Li, Y.-L.; Bi, C.; Du, L.-J.; Liu, Y.; Zhou, L.-J.; Liu, T.; Xu, S.; Shi, C.-J.; et al. Roles of Oral Microbiota and Oral-Gut Microbial Transmission in Hypertension. J. Adv. Res. 2023, 43, 147–161. [Google Scholar] [CrossRef]
  139. 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]
  140. Kunath, B.J.; Hickl, O.; Queirós, P.; Martin-Gallausiaux, C.; Lebrun, L.A.; Halder, R.; Laczny, C.C.; Schmidt, T.S.B.; Hayward, M.R.; Becher, D.; et al. Alterations of Oral Microbiota and Impact on the Gut Microbiome in Type 1 Diabetes Mellitus Revealed by Integrated Multi-Omic Analyses. Microbiome 2022, 10, 243. [Google Scholar] [CrossRef]
  141. Fei, N.; Zhao, L. An Opportunistic Pathogen Isolated from the Gut of an Obese Human Causes Obesity in Germfree Mice. ISME J. 2013, 7, 880–884. [Google Scholar] [CrossRef]
  142. 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]
  143. Flemer, B.; Warren, R.D.; Barrett, M.P.; Cisek, K.; Das, A.; Jeffery, I.B.; Hurley, E.; O’Riordain, M.; Shanahan, F.; O’Toole, P.W. The Oral Microbiota in Colorectal Cancer Is Distinctive and Predictive. Gut 2018, 67, 1454–1463. [Google Scholar] [CrossRef]
  144. Tyagi, A.M.; Yu, M.; Darby, T.M.; Vaccaro, C.; Li, J.-Y.; Owens, J.A.; Hsu, E.; Adams, J.; Weitzmann, M.N.; Jones, R.M.; et al. The Microbial Metabolite Butyrate Stimulates Bone Formation via T Regulatory Cell-Mediated Regulation of WNT10B Expression. Immunity 2018, 49, 1116–1131.e7. [Google Scholar] [CrossRef]
  145. Yan, J.; Herzog, J.W.; Tsang, K.; Brennan, C.A.; Bower, M.A.; Garrett, W.S.; Sartor, B.R.; Aliprantis, A.O.; Charles, J.F. Gut Microbiota Induce IGF-1 and Promote Bone Formation and Growth. Proc. Natl. Acad. Sci. USA 2016, 113, E7554–E7563. [Google Scholar] [CrossRef]
  146. Lucas, S.; Omata, Y.; Hofmann, J.; Böttcher, M.; Iljazovic, A.; Sarter, K.; Albrecht, O.; Schulz, O.; Krishnacoumar, B.; Krönke, G.; et al. Short-Chain Fatty Acids Regulate Systemic Bone Mass and Protect from Pathological Bone Loss. Nat. Commun. 2018, 9, 55. [Google Scholar] [CrossRef]
  147. Wang, N.; Zheng, L.; Qian, J.; Wang, M.; Li, L.; Huang, Y.; Zhang, Q.; Li, Y.; Yan, F. Salivary Microbiota of Periodontitis Aggravates Bone Loss in Ovariectomized Rats. Front. Cell. Infect. Microbiol. 2022, 12, 983608. [Google Scholar] [CrossRef]
  148. Stanisic, D.; Jeremic, N.; Majumder, S.; Pushpakumar, S.; George, A.; Singh, M.; Tyagi, S.C. High Fat Diet Dysbiotic Mechanism of Decreased Gingival Blood Flow. Front. Physiol. 2021, 12, 625780. [Google Scholar] [CrossRef]
  149. Luo, S.; Li, W.; Li, Q.; Zhang, M.; Wang, X.; Wu, S.; Li, Y. Causal Effects of Gut Microbiota on the Risk of Periodontitis: A Two-Sample Mendelian Randomization Study. Front. Cell. Infect. Microbiol. 2023, 13, 1160993. [Google Scholar] [CrossRef]
  150. Abdelbary, M.M.H.; Hatting, M.; Bott, A.; Dahlhausen, A.; Keller, D.; Trautwein, C.; Conrads, G. The Oral-Gut Axis: Salivary and Fecal Microbiome Dysbiosis in Patients with Inflammatory Bowel Disease. Front. Cell. Infect. Microbiol. 2022, 12, 1010853. [Google Scholar] [CrossRef]
Figure 1. SCFAs and the oral–gut axis. SCFA-producing oral bacteria can also colonize the gut and influence the metabolism of SCFAs by the gut microbiota. The paradox of the negative effect of SCFAs in the oral cavity is demonstrated. The major contribution is made by a significantly higher thickness of the mucus layer and the type of intestinal epithelium, which provide a decrease in the local concentration of SCFAs. Increased growth of SCFA-producing bacteria and the active formation of biofilms on the surface of the oral mucosa lead to cell damage and death. SCFAs are absorbed in the intestine, enter the systemic circulation, affect glucose and lipid metabolism, reduce appetite, and have a predominantly anti-inflammatory effect. Moreover, SCFAs support osteogenesis and reduce osteoclast activity, including in the alveolar bone. * Notes. Specific conditions: digestive tract pathology (achlorhydria, cirrhosis, colorectal cancer, etc.), rheumatoid arthritis, etc. In normal physiological conditions, oral–gut microbiota colonization is still disputable.
Figure 1. SCFAs and the oral–gut axis. SCFA-producing oral bacteria can also colonize the gut and influence the metabolism of SCFAs by the gut microbiota. The paradox of the negative effect of SCFAs in the oral cavity is demonstrated. The major contribution is made by a significantly higher thickness of the mucus layer and the type of intestinal epithelium, which provide a decrease in the local concentration of SCFAs. Increased growth of SCFA-producing bacteria and the active formation of biofilms on the surface of the oral mucosa lead to cell damage and death. SCFAs are absorbed in the intestine, enter the systemic circulation, affect glucose and lipid metabolism, reduce appetite, and have a predominantly anti-inflammatory effect. Moreover, SCFAs support osteogenesis and reduce osteoclast activity, including in the alveolar bone. * Notes. Specific conditions: digestive tract pathology (achlorhydria, cirrhosis, colorectal cancer, etc.), rheumatoid arthritis, etc. In normal physiological conditions, oral–gut microbiota colonization is still disputable.
Biomedicines 11 02749 g001
Table 1. Ratio of SCFAs produced by certain oral bacteria [52,53,54].
Table 1. Ratio of SCFAs produced by certain oral bacteria [52,53,54].
SpeciesSCFAs
AceticPropanoicButyricIsobutyricIsovaleric
P. gingivalis+++++++++
P. asaccharolytica+++++++++
P. intermedia++++++++
F. nucleatum++++++++
A. actinomycetemcomitans+++----
V. parvula+++++---
S. sanguinis++----
“+++”—relatively high concentration of SCFA in the bacterial isolate, “++”—relatively medium concentration of SCFA in the bacterial isolate, “+”—relatively low concentration of SCFA in the bacterial isolate, “-”—SCFA were not detected in the bacterial isolate.
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

Leonov, G.E.; Varaeva, Y.R.; Livantsova, E.N.; Starodubova, A.V. The Complicated Relationship of Short-Chain Fatty Acids and Oral Microbiome: A Narrative Review. Biomedicines 2023, 11, 2749. https://doi.org/10.3390/biomedicines11102749

AMA Style

Leonov GE, Varaeva YR, Livantsova EN, Starodubova AV. The Complicated Relationship of Short-Chain Fatty Acids and Oral Microbiome: A Narrative Review. Biomedicines. 2023; 11(10):2749. https://doi.org/10.3390/biomedicines11102749

Chicago/Turabian Style

Leonov, Georgy E., Yurgita R. Varaeva, Elena N. Livantsova, and Antonina V. Starodubova. 2023. "The Complicated Relationship of Short-Chain Fatty Acids and Oral Microbiome: A Narrative Review" Biomedicines 11, no. 10: 2749. https://doi.org/10.3390/biomedicines11102749

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

Article Metrics

Back to TopTop