Next Article in Journal
Role of Platelets in Osteoarthritis—Updated Systematic Review and Meta-Analysis on the Role of Platelet-Rich Plasma in Osteoarthritis
Previous Article in Journal
Pyroptosis and Sarcopenia: Frontier Perspective of Disease Mechanism
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Leaky Gum: The Revisited Origin of Systemic Diseases

1
DOCSmedi Co., Ltd., 4F, 143, Gangseong-ro, Ilsanseo-gu, Goyang-si 10387, Korea
2
Department of Gastrointestinal Endoscopy, Apple Tree Healthcare Center, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea
3
Department of Orthodontics, Apple Tree Dental Hospital, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea
4
Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea
*
Authors to whom correspondence should be addressed.
Cells 2022, 11(7), 1079; https://doi.org/10.3390/cells11071079
Submission received: 17 February 2022 / Revised: 21 March 2022 / Accepted: 22 March 2022 / Published: 23 March 2022

Abstract

:
The oral cavity is the gateway for microorganisms into your body where they disseminate not only to the directly connected respiratory and digestive tracts but also to the many remote organs. Oral microbiota, travelling to the end of the intestine and circulating in our bodies through blood vessels, not only affect a gut microbiome profile but also lead to many systemic diseases. By gathering information accumulated from the era of focal infection theory to the age of revolution in microbiome research, we propose a pivotal role of “leaky gum”, as an analogy of “leaky gut”, to underscore the importance of the oral cavity in systemic health. The oral cavity has unique structures, the gingival sulcus (GS) and the junctional epithelium (JE) below the GS, which are rarely found anywhere else in our body. The JE is attached to the tooth enamel and cementum by hemidesmosome (HD), which is structurally weaker than desmosome and is, thus, vulnerable to microbial infiltration. In the GS, microbial biofilms can build up for life, unlike the biofilms on the skin and intestinal mucosa that fall off by the natural process. Thus, we emphasize that the GS and the JE are the weakest leaky point for microbes to invade the human body, making the leaky gum just as important as, or even more important than, the leaky gut.

1. Introduction

Humans internalize the microbiota of this planet through the oral cavity, either temporarily or permanently. The oral cavity harbours the second most abundant microorganisms after the gastrointestinal (GI) tract in a variety of distinct habitats, such as teeth, tongue, gingival sulcus (GS), palate, saliva, buccal mucosa, and throat. The expanded Human Oral Microbiome Database (eHOMD v3, http://homd.org, accessed on 16 February 2022) established during the Human Microbiome Project enlists at least 774 microbial species to date.
As the old dogma that the lungs and placenta are sterile becomes obsolete [1,2,3,4,5], the oral microbiota has proven to be the primary source of the bacterial microbiota in human organs [6]. For one, microaspiration during respiratory activity, such as oral breathing, affects the lung microbiota [7]. In addition, dietary patterns dynamically affect the microbiome profile of the GI tract either by microbial contamination or by supplying specific nutrients for microbial commensals, even manipulating the pathophysiology of cancerous diseases [8,9] as well as regulating immune responses across the gut–brain axis [10,11]. As such, along with the revolution of human microbiome research, much effort has been dedicated to figuring out the relationship between the oral and gut microbiota, which has been dubbed the “oral–gut–brain axis” [12,13,14,15,16,17].
The microbiota in the gut seek opportunities to breach the dysfunctional gut mucosal barrier to reach the underlying immune system, resulting in “leaky gut” syndrome. This type of leak occurs through the cellular junctions between the intestinal epithelia. In the oral cavity, the microbes can easily colonize on the surface of the hard tissue of the tooth enamel to form biofilm and remain sclerotized until proper interventions come in. The growing body of the biofilm not only acts as a wedge disjoining the tooth and the gingiva, enlarging the GS depth but also deploy an ample number of microbes near the sulcus epithelium, enhancing the opportunity for microbial infiltration into the oral mucosa [18]. Thus, microbial infection in the oral cavity is accelerated by both physical and biological processes.
In this review, we gather current knowledge of disease-related oral pathogens and contrast the anatomical structures of oral versus gut mucosal layers in the context of microbial leak into a human body, embossing the role of oral pathogens in the development of systemic diseases.

2. Hyperpermeable Intestine—Leaky Gut

The largest portion of research funds has been digested by gut bacteria because the majority of human microbiota resides in the colon [19]. The findings of their roles in human immune systems have been extensively illuminating. The human intestine is the widest and longest space in contact with microorganisms compared to the oral cavity or the skin. It boasts almost 10 metres of length and 400 m2 of luminal surface area. In addition, it allows the passage of about 60 tons of food during a lifetime while processing digestion and absorption, making the role of gut bacteria even more important [20]. Especially, gut bacteria, numbering almost equivalent to the human cells [19], metabolize dietary fibers to yield short-chain fatty acids, an essential task of which humans are not capable. They not only provide intestinal cells with immune substances and vitamins but also keep the intestinal homeostasis. The homeostasis of the intestinal microbiome itself and between the intestinal microbiome and the host has been conceptualized as a “symbiosis” of the intestinal ecosystem [21].
Perturbation factors, such as stress, smoking, alcohol consumption, eating processed foods, and overuse of antibiotics, have a certain effect on the ecosystem of the intestinal microbiome. The disturbance is usually absorbed by the resilience of the intestinal ecosystem, but repeated exposures to such risk factors would lead to “dysbiosis”, a continuous status of imbalance between gut microbiota and their host [22]. For example, when antibiotics deplete intestinal bacteria who are responsible for converting primary bile acids into secondary bile acids, such as deoxycholate, lithocholate, ursodeoxycholate, hyodeoxycholate, and ω-muricholate, the resistance to Clostridium difficile decreases [23], resulting in pseudomembranous colitis and persistent diarrhoea that claim lives of tens of thousands of people in the US [24,25]. Likewise, it is now well-accepted that a dysbiosis of gut microbiome can affect not only inflammatory bowel disease (IBD), constipation, indigestion, and obesity but also the occurrence and prognosis of hypertension, diabetes, cancer, and cardiovascular diseases (CVD) [26,27,28,29,30,31,32,33].
During the golden era of gut microbiome research, the term “leaky gut”, previously proposed and used in the field of alternative medicine and dietetics, was revisited as a compatible explanation of “increased intestinal permeability” (Figure 1a,b) [34,35,36,37]. The rationale behind the term lies in the concurrent pathogenesis of intestinal and systemic diseases caused by the infiltration of enteric bacteria and virulence factors into the intestinal mucosal membrane when the epithelial barrier function is disrupted [36,38,39]. The intestinal hyperpermeability has often been accompanied by the changes of tight junction proteins in the epithelium or the increased bacterial endotoxin in the bloodstream, endotoxemia [36]. For instance, patients with IBD, irritable bowel syndrome (IBS), liver diseases, pancreatitis, diabetes, chronic heart failure, depression, and other chronic diseases often exhibit increased permeability and epithelial barrier dysfunction [40,41,42]. As evidence builds up and interests from diverse research fields expand further, the methodologies for the measurement of intestinal barrier function have been extensively developed [42].
The association between intestinal and systemic diseases can be found in extra-intestinal manifestations (EIMs) of intestinal diseases. For example, the EIMs of IBD affect joints, eyes, liver, lung, and pancreas [43]. About 15% of people with ulcerative colitis (UC) [44] and up to 40% of patients with Crohn’s disease [45,46,47,48], both are types of IBD, have skin issues. In some EIMs cases, such as peripheral arthritis, oral aphthous ulcers, episcleritis, and erythema nodosum, symptoms can improve on standard treatment of the intestinal inflammation [43]. In addition, IBD and periodontitis have been reported to affect each other, and several nutritional deficiencies and systemic diseases are known to be manifested in the oral cavity [49,50], which is supporting bidirectional influence in the context of an oral–gut axis. In the same vein, bacteria can travel via the bloodstream to reach and colonize in the tumour microenvironment (TME) of melanoma, lung, ovarian, glioblastoma, pancreatic, bone, and breast cancer where ~106 intratumoural bacteria per palpable 1-cm3 tumour can be found [32,51].

3. Gum and Gut Mucosal Barriers

The lumen of the digestive tract, a twisted hollow tube from the oral cavity to the anus, is continuously overloaded with the external environment [52]. Thus, just as the human skin protects our body, the oral and intestinal mucosa, which cover the inner surface of our body, should exert barrier function physically and physiologically [38,39]. Yet unlike the skin whose major function is building a physical barrier, the major function of the mucosal membrane comprises several physiological barriers. For example, saliva and mucus on the epithelium contain antibacterial substances, such as lactoferrin, lysozyme, commensal flora, and antibacterial peptides, which inhibit pathogen colonization [53,54].
The surface layers of the skin (epidermis), the oral cavity, and the gastrointestinal tract share both similar and dissimilar structures and functions. The epidermis comprises several cellular layers strongly bound with intercellular junctions of which the surface is covered by the stratum corneum [55]. The intestinal epithelial layer is also filled with cells interconnected with strong intercellular junctions (Table 1). The luminal side of the intestinal epithelial layer, however, is made up of a thinner monolayer, is supported by the connective tissue underneath only, and has no stratum corneum covering the layer. Thus, from an anatomical point of view, the intestinal mucosa is less tolerable to an environmental shift such as dysbiosis of a microbial community, which leads to bacteremia or endotoxemia through the increased intestinal permeability [56]. To compensate for the weakness of the epithelial layer, the intestinal mucosa always covers the epithelium with mucus that not only acts as a lubricant between the mucosa and the luminal passengers but also contains a lot of antibacterial substances, indicating that the gut mucosa has both chemical and biological barrier functions [57]. Pathogenic intruders who survive in the mucosal layer and leak through the epithelial barriers face mucosal-associated lymphoid tissue (MALT) that takes up ~70% of the entire immune system of a human body (Figure 1b). In addition, ~80% of plasma cells residing in gut-associated lymphoid tissue (GALT), a major part of MALT, wage a deliberate war against antigens originating from ~4 × 1013 commensal microbes and more than 30 kg of food proteins yearly [19,52,58]. Consequently, the gastrointestinal system plays a pivotal role in immune surveillance.
Mucosal immunity in the oral cavity is also gaining traction with the advance of microbiome research [59,60]. The oral mucosa, like the skin, is composed of both keratinized and non-keratinized tissues (Figure 1c). The thickness of the keratinized layers of the oral mucosa, however, is thinner than that of the skin. Thus, the oral mucosa, similar to the intestinal mucosa, supplements chemical and biological defence functions using saliva. The oral mucosa also consists of 3–5 cellular layers that are thicker than the intestinal mucosa monolayer. Anatomically and histologically, the oral mucosa appears as a transitional layer between the intestinal mucosa and the skin [61].

4. Gingival Sulcus and Junctional Epithelium

For humans, the primary teeth erupt through the mucous membrane from the inner alveolar bone at about six months of age, forming unique structures originating from the interface between the exposed teeth and the surrounding tissues. Those interface structures, the GS and the JE below the GS, are essential for the survival of animals that need mastication of ingested food (Figure 1d). For adults with permanent teeth, the healthy GS depth can reach 3 mm. Thus, below the GS there should be a sealing layer that binds the soft tissue, especially the JE, with the surface layer of the hard tissue (enamel and cementum), protecting the tissues from external challenges. The JE has the highest turnover rate (4–6 days) of all oral epithelia and remains undifferentiated and non-keratinized [62]. In the JE, the binding proteins are generated by the basal layer of soft tissues only to form HD with the hard tissues of a tooth (Figure 1d, magnified box). The internal basal lamina (IBL), the intercellular space in the HD, are relatively wide, allowing water-soluble substances to pass through them with ease. These structural limitations of the HD between the JE and the tooth can provide pathogens with a good opportunity to invade the human body [63,64,65]. To compensate for this inherent structural weakness, immune cells such as polymorphic nuclear leukocytes (PMNs) transude into the GS together with gingival crevicular fluid (GCF), taking constant vigilance even without any signs of inflammation [66,67]. Ironically, to allow immune cells to pass through a JE layer, the JE cells have fewer desmosomes that bind the cells vertically than the other oral epithelia, adding another structural instability to JE [68].
The GS provides the perfect space for biofilm accumulation. Biofilms on the skin, oral mucosa, and gut mucosa are washed out along with hygiene activities, digestive processes, exfoliation, and defecation. The surface layer of the tooth enamel, however, does not fall off because it lacks cell division and maintains its structure unless external physical and biological intervention is applied. Thus, if the GS is not properly managed, biofilms will inevitably accumulate during lifetime [69] even to the level of thickness enough to ward off antibiotics [70] (Figure 2a,b). The biofilm accumulation induces inflammatory responses that erode alveolar bone and increases the GS depth, resulting in the formation of the periodontal pocket (PP). The deepened PP in turn makes it difficult to remove the biofilm in the PP. This vicious cycle results in increased inflammatory reactions, i.e., periodontal diseases [18,71].
The important roles of the GS and the JE were embossed in a seminal study conducted on 417 patients at 11 nursing homes in Japan [72]. In this study, older patients who received oral care exhibited lower cases of pneumonia, febrile days, and death from pneumonia, while showing improved metrics of activities of daily living (ADL) and cognitive functions evaluated with Mini-Mental State Examination (MMSE). By contrast, the total mortality rate was greater in the dentate group (13.5%, 28/208) than in the edentate group (10%, 16/158). The mortality rate of dentate and edentate groups with oral care was similar (6% and 7%, respectively), but without oral care, the mortality rate of the dentate group (20%) was higher than that of the edentate group (13%) even if ADL and MMSE scores were slightly worse in the edentate group at the time of final evaluation. The reduced mortality rate in the edentate group without oral care, although not fully discussed in this study, may indicate that the edentate state is somehow advantageous for longevity if proper oral care cannot be administered. Indeed, the spot where the tooth is removed becomes covered with mucosal membrane and transforms like the mucosa of the skin and the intestine (Figure 2c). In other words, the absence of teeth may render more effective protection from bacterial infections by removing the vulnerable structures originating from the GS and the JE [73].

5. Focal Infection Theory and Leaky Gum

Concerns have already existed since the end of the 19th century that the oral cavity could be a source of human pathogenic microbes. In the 1890s, Willoughby D. Miller, who studied at the Koch Institute, warned of the dangers of oral microbes [74,75]. Miller, riding on the bandwagon of the “germ theory” of disease established at the end of the 19th century, suggested that the oral cavity, a breeding ground for many pathogens, could be an origin of many diseases of unknown aetiology, such as CVDs, pneumonia, angina pectoris, and foot gangrene [76]. Miller’s study has established a modern daily routine for oral care, such as brushing teeth three times a day and flossing. His argument was later accepted as “oral sepsis” by British surgeon William Hunter in the early 1900s [77] and expanded as “focal infection theory” by American physician Frank Billings in the 1910s [78]. It was further amplified by Henry Cotton who claimed that mental illness could be improved by tooth extraction or tonsillectomy [79]. Even accepted by the physicians at Johns Hopkins University and Mayo Clinic, the theory was implemented into routine clinical practice. The theory was so widely expanded that Russell Cecil, an eminent author of Cecil Essentials of Medicine, also joined the club. In the 1940s, however, Hobart A. Reinmann and W. Paul Havens pointed out, in their critical appraisal of focal infection in relation to systemic disease, that the theory lacks clinical evidence and the causative relationship of infections of teeth and tonsils to systemic disease is unproven [80]. Consequently, in the late 1950s, the theory gradually vanished and was regarded as fringe medicine.
In the 21st century, the focal infection theory began to be revisited from a different perspective [81]. For example, bacteremia, a temporary infiltration of bacteria into blood vessels, has been regarded as an illness resolved by immune responses within an hour [65]. Recent culture-independent microbial research techniques, however, have shown that bacteria or bacterial DNA are always present in the blood vessels of healthy people [82,83,84]. These findings suggest that bacteremia may not be a temporary nor a localized problem. Furthermore, it has been revealed that microbes can be found in the lungs of healthy people [5,85,86] and cancer patients [87], the placenta of healthy pregnant women, albeit controversial [1,2,3,4], and the brains in Alzheimer’s disease (AD) [88,89,90,91,92], which had long been considered sterile.

6. Oral Pathogens and Systemic Diseases

The origin of bacteria found in remote organs converges to the oral cavity [93,94,95,96] (Figure 3). For example, the placental microbiome profiles were most comparable to those of the oral microbiome [1]. The overlap of the unique members of oral microbes with other remote organs is consistent with previous clinical studies in which Fusobacterium nucleatum, a Gram-negative oral anaerobe, were clinically suspected to be a major risk factor in colorectal cancer [97,98,99], oral squamous cell carcinoma (OSCC) [100], and in preterm and term stillbirth [101,102]. Likewise, an infamous oral pathogenic bacterium, Porphyromonas gingivalis, is related to pancreatic cancer [103], colorectal cancer [104,105,106], liver health [107], rheumatoid arthritis [108,109], diabetes [110,111,112], OSCC [113,114], and neurodegenerative diseases [88,89,90,91,92,115,116,117,118]. In the case of atherosclerotic CVD, when the vascular tissues of the coronary and femoral arteries of the patients with CVD were examined, P. gingivalis was found in 42 out of 42 patients [119]. Thus, previously explained by the passive accumulation of fat, the aetiology of CVD is now leaning toward inflammatory responses of the vascular endothelium [120,121]. Notably, live P. gingivalis is known to traffic into endoplasmic reticulum-rich autophagosomes [122] and target host ectonucleotidase-CD73 [123] for its chronic survival, replication, and persistence in the dysbiotic human gingival epithelia [124].
Recently, Kitamoto et al. demonstrated the mechanistic underpinnings by which periodontal inflammation due to oral infection contributes to the pathogenesis of extra-oral diseases [12]. In this elaborated study using mice, they showed that periodontitis aggravates gut inflammation by translocating oral Klebsiella/Enterobacter species to the lower digestive tract where it colonizes ectopically to elicit colitis through IL-1β. In parallel, oral Th17 cells induced by oral pathobiont expansion migrate to the gut and promote colitis, constituting both microbial and immunological pathways that link oral and gut health. Furthermore, Dong et al. showed that the alterations of the oral microbiota, especially F. nucleatum colonization in CRC locus, can change the gut bacterial composition within tumours and influence the therapeutic efficacy and prognosis of radiotherapy for primary rectal cancer and CRC liver metastases in mouse models [125]. Kartal et al. applied shotgun metagenomic and 16S rRNA sequencing to faecal and saliva samples from pancreatic ductal adenocarcinoma (PDAC) to identify diagnostic classifiers [96]. While proving that the faecal metagenomic classifiers outperformed the saliva-based classifiers, they also confirmed that the strains of faecal PDAC-associated microbes originate from the oral cavity. Thus, the growing body of examples that show the close relationship of oral pathogens with a variety of systemic diseases enabled the introduction of the term “periodontal medicine”, to describe how periodontal infection and inflammation affect extraoral illness [126,127]. As such, the oral cavity needs to be re-evaluated as a more pivotal organ with the revolution of microbiology in the 21st century [71].
There are many disease model systems proving that the oral bacteria induce systemic diseases, such as CVD, type II diabetes mellitus (T2DM), OSCC, and AD (Table 2). For one, the infection of aortic lesions with P. gingivalis activates adhesion molecules such as intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1), leading to chronic inflammation via migration of more immune cells to the lesion sites [128]. The microarray analysis demonstrated that P. gingivalis-treated human aortic endothelial cells (HAECs) upregulated expression levels of ICAM-1, VCAM-1, and interleukin-6 (IL-6). As well as ICAM-1 and VCAM-1 upregulation, pathological enlargement of atherosclerotic lesion area were well demonstrated in hyperlipidemic (Apoe−/−) mice orally challenged with P. gingivalis [128,129]. As an effective molecule, lipopolysaccharide from P. gingivalis (PgLPS) was established to promote inflammatory response as increasing mononuclear cell adhesion to human umbilical vein endothelial cells (HUVECs) via ICAM-1 and Toll-like receptor 2 (TLR-2)-dependent mechanism [130]. Subcutaneous infection of obese pigs with P. gingivalis also showed enhanced aortic and coronary arterial atherosclerosis [63]. In addition to P. gingivalis, intravenous infection of hyperlipidemic mice with Aggregatibacter actinomycetemcomitans can promote and accelerate atherosclerotic plaques [131] and time-dependently elevate matrix metalloproteinase-9 (MMP-9) expression [132]. The MMP-9, derived from macrophage, has been highlighted as a risk factor of acute atherosclerosis due to its proteolytic activity of advanced atherosclerotic plaque rupture [133].
T2DM is a highly prevalent metabolic disease characterized by prolonged high glucose levels in the blood. Insulin resistance on peripheral tissues has been focused on as the major causing factor of T2DM [144]. Recently, many microbiologists designated gut dysbiosis as a critical factor of insulin resistance development in T2DM accompanied by gut barrier dysfunction [145]. Interestingly, oral infection of mice with P. gingivalis can also induce gut dysbiosis, leading to insulin resistance via a pathway through endotoxin entrance and chronic inflammation [134]. Mice pre-treated with P. gingivalis, F. nucleatum, and Prevotella intermedia showed accelerated insulin resistance after three months of high-fat diet (HFD) feeding [135]. The branched-chain amino acid (BCAA) biosynthesis activity of P. gingivalis is a suggested mechanism of insulin resistance development, as evident that BCAA aminotransferase-deficient (∆bcat) P. gingivalis strain can neither induce insulin resistance nor upregulate serum BCAA in HFD mice model [136].
OSCC is the most malignant cancer of the oral cavity with an increasing rate of incidence, and the risk factors for OSCC include alcohol consumption, smoke, and human papillomavirus [146]. Interestingly, two independent groups suggested that P. gingivalis administration can significantly increase the number and diameter of the lesions in tongue tissues of mice pre-treated with carcinogen 4-nitroquinoline-1 oxide (4NQO). They provided two pathways, dysregulated lipid metabolism and CD11b+ myeloid-derived suppressor cells (MDSCs) infiltration, involved with OSCC deterioration by the pathogen [113,114]. Indeed, abnormal lipid regulation by increased expressions of fatty acid-binding protein 4 (FABP4) and FABP5 has been reported to have a crucial role in OSCC development via activation of mitogen-activated protein kinase (MAPK) pathway and MMP-9 [147,148]. By contrast, CD11b+ is responsible for MDSCs migration to tumour microenvironment where the cells have an immunosuppressive role that favours tumour progression [149].
AD is one of the representative neurodegenerative diseases diagnosed with senile plaques and abundant neurofibrillary tangles, which can be deteriorated by oral pathogenic infection. Gingival-infected P. gingivalis was reported to exacerbate the accumulation of Aβ plaques and inflammatory cytokines in brain specimens of amyloid precursor protein (APP) transgenic mice [140]. Interestingly, the anatomic analysis demonstrated that P. gingivalis genomic DNA was detected in brain specimens of 9 out of 12 Apoe−/− mice orally challenged with P. gingivalis for 24 weeks [139], implicating that P. gingivalis can penetrate the gum and enter the blood-brain barrier (BBB). The result that intravenous injection of P. gingivalis into rats enhanced tau hyperphosphorylation in the hippocampus can reinforce the theory of BBB penetration of P. gingivalis [143]. As similar atherosclerotic CVD, PgLPS has also been designated as P. gingivalis-driven virulence factor affecting AD. It was reported that PgLPS treatment to rat brain neonatal microglial cells promoted the release of inflammatory mediators such as TNF-α and IL-6 [138], and palatal gingival infection of PgLPS into rats induced alveolar bone loss and increased serum Aβ levels [142]. Middle-aged wild-type (WT) mice intraperitoneally challenged with PgLPS for 5 weeks represented learning and memory deficit and microglia-mediated neuroinflammation, although age-matched mice deficient for cathepsin B (Catb−/−) were insensitive to PgLPS [141]. In addition to PgLPS, gingipain is an AD virulence factor, a unique class of cysteine proteinase that comprises Lys-gingipain (Kgp) and Arg-gingipain (Rgp). The modulatory role of the gingipain on neuroinflammation was well-established using Kgp and Rgp inhibitors (KYT1 and KYT36, respectively) or P. gingivalis KDP129, a gingipain-deficient mutant strain [137]. In this study, KYT1 and KYT36 treatment effectively inhibited P. gingivalis-driven increased expression of IL-6 and TNF-α in immortalized mouse microglial cell line MG6. Injection of P. gingivalis, but not KDP129 strain, into the somatosensory cortex of mice can recruit microglia to the injection site, revealing that gingipain is the effective factor for microglial migration and accumulation around P. gingivalis in the brain [137].

7. Conclusions and Perspectives

Thanks to rapid advances in gene sequencing technology combined with nanotechnology and information technology, the human microbiome proves to be present even in bodily sites previously known to be sterile. Intriguingly, the microbiome inside the human body mostly originates from the oral cavity, reminiscing the focal infection theory backed by more recent scientific proofs. Indeed, the oral cavity has unique mucosal structures such as the PP and the JE with innate vulnerability where oral pathogens can colonize for life and leak into blood vessels to circulate throughout the body, resulting in many systemic diseases in remote sites. As of now, although just a few molecules (PgLPS or gingipain) have been unravelled as oral pathogen-induced toxic molecules, we need to figure out many other causative candidates derived from oral pathogens to reinforce the attribution of leaky gum to various types of systemic diseases. Filled with anticipation for more causative evidence from well-designed empirical studies, we also need to focus on how to provide a leaky gum with a protective shield made of biological, not physicochemical, knowledge. By doing so, we can look forward to the realization of more prominent personalized medicine for systemic health by striking a balance between oral microbiota and its host.

Author Contributions

Conceptualization, H.-S.K.; resources, J.Y.P. and D.L.; writing—original draft preparation, D.-Y.P., I.H. and H.-S.K.; writing—review and editing, I.H.; visualization, I.H.; supervision, I.H. and H.-S.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

This work was supported in part by DOCSmedi Co., Ltd. and the Apple Tree Medical Foundation. The authors thank Hyo Jung Kwon and Kye Kyoung Park for their help in manuscript preparation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Aagaard, K.; Ma, J.; Antony, K.M.; Ganu, R.; Petrosino, J.; Versalovic, J. The placenta harbors a unique microbiome. Sci. Transl. Med. 2014, 6, 237ra265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Gomez-Arango, L.F.; Barrett, H.L.; McIntyre, H.D.; Callaway, L.K.; Morrison, M.; Nitert, M.D. Contributions of the maternal oral and gut microbiome to placental microbial colonization in overweight and obese pregnant women. Sci. Rep. 2017, 7, 2860. [Google Scholar] [CrossRef] [PubMed]
  3. Goffau, M.C.d.; Lager, S.; Sovio, U.; Gaccioli, F.; Cook, E.; Peacock, S.J.; Parkhill, J.; Charnock-Jones, D.S.; Smith, G.C.S. Human placenta has no microbiome but can contain potential pathogens. Nature 2019, 572, 329–334. [Google Scholar] [CrossRef] [PubMed]
  4. Ye, C.; Kapila, Y. Oral microbiome shifts during pregnancy and adverse pregnancy outcomes: Hormonal and Immunologic changes at play. Periodontol. 2000 2021, 87, 276–281. [Google Scholar] [CrossRef]
  5. Whiteside, S.A.; McGinniss, J.E.; Collman, R.G. The lung microbiome: Progress and promise. J. Clin. Investig. 2021, 131, e150473. [Google Scholar] [CrossRef]
  6. Huffnagle, G.B.; Dickson, R.P.; Lukacs, N.W. The respiratory tract microbiome and lung inflammation: A two-way street. Mucosal Immunol. 2017, 10, 299–306. [Google Scholar] [CrossRef] [Green Version]
  7. Dickson, R.P.; Erb-Downward, J.R.; Freeman, C.M.; McCloskey, L.; Falkowski, N.R.; Huffnagle, G.B.; Curtis, J.L.; Clemente, J.C.; Molyneaux, P.; Bogaert, D. Bacterial Topography of the Healthy Human Lower Respiratory Tract. MBio 2017, 8, e02287-16. [Google Scholar] [CrossRef]
  8. Mehta, R.S.; Nishihara, R.; Cao, Y.; Song, M.; Mima, K.; Qian, Z.R.; Nowak, J.A.; Kosumi, K.; Hamada, T.; Masugi, Y.; et al. Association of Dietary Patterns With Risk of Colorectal Cancer Subtypes Classified by Fusobacterium nucleatum in Tumor Tissue. JAMA Oncol. 2017, 3, 921–927. [Google Scholar] [CrossRef] [Green Version]
  9. Tao, J.; Li, S.; Gan, R.-Y.; Zhao, C.-N.; Meng, X.; Li, H.-B. Targeting gut microbiota with dietary components on cancer: Effects and potential mechanisms of action. Crit. Rev. Food Sci. Nutr. 2020, 60, 1025–1037. [Google Scholar] [CrossRef]
  10. Agirman, G.; Yu, K.B.; Hsiao, E.Y. Signaling inflammation across the gut-brain axis. Science 2021, 374, 1087–1092. [Google Scholar] [CrossRef]
  11. Morais, L.H.; Schreiber, H.L.; Mazmanian, S.K. The gut microbiota–brain axis in behaviour and brain disorders. Nat. Rev. Microbiol. 2021, 19, 241–255. [Google Scholar] [CrossRef]
  12. 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]
  13. Kitamoto, S.; Nagao-Kitamoto, H.; Hein, R.; Schmidt, T.M.; Kamada, N. The Bacterial Connection between the Oral Cavity and the Gut Diseases. J. Dent. Res. 2020, 99, 1021–1029. [Google Scholar] [CrossRef] [PubMed]
  14. Ray, K. The oral–gut axis in IBD. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 532. [Google Scholar] [CrossRef] [PubMed]
  15. Narengaowa; Kong, W.; Lan, F.; Awan, U.F.; Qing, H.; Ni, J. The Oral–Gut–Brain AXIS: The Influence of Microbes in Alzheimer’s Disease. Front. Cell. Neurosci. 2021, 15, 113. [Google Scholar] [CrossRef]
  16. Byrd, K.M.; Gulati, A.S. The “Gum–Gut” Axis in Inflammatory Bowel Diseases: A Hypothesis-Driven Review of Associations and Advances. Front. Immunol. 2021, 12, 620124. [Google Scholar] [CrossRef]
  17. Hu, S.; Png, E.; Gowans, M.; Ong, D.E.H.; de Sessions, P.F.; Song, J.; Nagarajan, N. Ectopic gut colonization: A metagenomic study of the oral and gut microbiome in Crohn’s disease. Gut Pathog. 2021, 13, 13. [Google Scholar] [CrossRef]
  18. Hall-Stoodley, L.; Costerton, J.W.; Stoodley, P. Bacterial biofilms: From the natural environment to infectious diseases. Nat. Rev. Microbiol. 2004, 2, 95–108. [Google Scholar] [CrossRef]
  19. Sender, R.; Fuchs, S.; Milo, R. Are We Really Vastly Outnumbered? Revisiting the Ratio of Bacterial to Host Cells in Humans. Cell 2016, 164, 337–340. [Google Scholar] [CrossRef] [Green Version]
  20. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef]
  21. Malard, F.; Dore, J.; Gaugler, B.; Mohty, M. Introduction to host microbiome symbiosis in health and disease. Mucosal Immunol. 2021, 14, 547–554. [Google Scholar] [CrossRef] [PubMed]
  22. Relman, D.A. The human microbiome: Ecosystem resilience and health. Nutr. Rev. 2012, 70, S2–S9. [Google Scholar] [CrossRef] [PubMed]
  23. Theriot, C.M.; Bowman, A.A.; Young, V.B. Antibiotic-Induced Alterations of the Gut Microbiota Alter Secondary Bile Acid Production and Allow for Clostridium difficile Spore Germination and Outgrowth in the Large Intestine. mSphere 2016, 1, e00045-15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Lessa, F.C.; Mu, Y.; Bamberg, W.M.; Beldavs, Z.G.; Dumyati, G.K.; Dunn, J.R.; Farley, M.M.; Holzbauer, S.M.; Meek, J.I.; Phipps, E.C.; et al. Burden of Clostridium difficile infection in the United States. N. Engl. J. Med. 2015, 372, 825–834. [Google Scholar] [CrossRef] [Green Version]
  25. CDC. Nearly Half a Million Americans Suffer from C. difficile Infections in Single Year. Available online: https://www.cdc.gov/media/releases/2015/p0225-clostridium-difficile.html (accessed on 16 February 2022).
  26. Zitvogel, L.; Galluzzi, L.; Viaud, S.; Vétizou, M.; Daillère, R.; Merad, M.; Kroemer, G. Cancer and the gut microbiota: An unexpected link. Sci. Transl. Med. 2015, 7, 271ps1. [Google Scholar] [CrossRef] [Green Version]
  27. Zhao, Y.; Yu, Y.-B. Intestinal microbiota and chronic constipation. Springerplus 2016, 5, 1130. [Google Scholar] [CrossRef] [Green Version]
  28. Wong, S.H.; Zhao, L.; Zhang, X.; Nakatsu, G.; Han, J.; Xu, W.; Xiao, X.; Kwong, T.N.Y.; Tsoi, H.; Wu, W.K.K.; et al. Gavage of Fecal Samples from Patients with Colorectal Cancer Promotes Intestinal Carcinogenesis in Germ-Free and Conventional Mice. Gastroenterology 2017, 153, 1621–1633. [Google Scholar] [CrossRef] [Green Version]
  29. Cao, H.; Liu, X.; An, Y.; Zhou, G.; Liu, Y.; Xu, M.; Dong, W.; Wang, S.; Yan, F.; Jiang, K.; et al. Dysbiosis contributes to chronic constipation development via regulation of serotonin transporter in the intestine. Sci. Rep. 2017, 7, 10322. [Google Scholar] [CrossRef] [Green Version]
  30. Ohkusa, T.; Koido, S.; Nishikawa, Y.; Sato, N. Gut Microbiota and Chronic Constipation: A Review and Update. Front. Med. 2019, 6, 19. [Google Scholar] [CrossRef] [Green Version]
  31. Ding, R.X.; Goh, W.R.; Wu, R.N.; Yue, X.Q.; Luo, X.; Khine, W.W.T.; Wu, J.R.; Lee, Y.K. Revisit gut microbiota and its impact on human health and disease. J. Food Drug Anal. 2019, 27, 623–631. [Google Scholar] [CrossRef]
  32. Atreya, C.E.; Turnbaugh, P.J. Probing the tumor micro(b)environment. Science 2020, 368, 938–939. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, S.; Wang, R.; Li, D.; Zhao, L.; Zhu, L. Role of gut microbiota in functional constipation. Gastroenterol. Rep. 2021, 9, 392–401. [Google Scholar] [CrossRef] [PubMed]
  34. Hollander, D. Intestinal permeability, leaky gut, and intestinal disorders. Curr. Gastroenterol. Rep. 1999, 1, 410–416. [Google Scholar] [CrossRef] [PubMed]
  35. Bischoff, S.C.; Barbara, G.; Buurman, W.; Ockhuizen, T.; Schulzke, J.-D.; Serino, M.; Tilg, H.; Watson, A.; Wells, J.M. Intestinal permeability—A new target for disease prevention and therapy. BMC Gastroenterol. 2014, 14, 189. [Google Scholar] [CrossRef] [Green Version]
  36. Fukui, H. Increased Intestinal Permeability and Decreased Barrier Function: Does It Really Influence the Risk of Inflammation? Inflamm. Intest. Dis. 2016, 1, 135–145. [Google Scholar] [CrossRef]
  37. Mu, Q.; Kirby, J.; Reilly, C.M.; Luo, X.M. Leaky gut as a danger signal for autoimmune diseases. Front. Immunol. 2017, 8, 598. [Google Scholar] [CrossRef] [Green Version]
  38. Kelly, J.R.; Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G.; Hyland, N.P. Breaking down the barriers: The gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front. Cell. Neurosci. 2015, 9, 392. [Google Scholar] [CrossRef] [Green Version]
  39. Odenwald, M.A.; Turner, J.R. The intestinal epithelial barrier: A therapeutic target? Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 9–21. [Google Scholar] [CrossRef]
  40. Fukui, H. Role of Gut Dysbiosis in Liver Diseases: What Have We Learned So Far? Diseases 2019, 7, 58. [Google Scholar] [CrossRef] [Green Version]
  41. Harbison, J.E.; Roth-Schulze, A.J.; Giles, L.C.; Tran, C.D.; Ngui, K.M.; Penno, M.A.; Thomson, R.L.; Wentworth, J.M.; Colman, P.G.; Craig, M.E.; et al. Gut microbiome dysbiosis and increased intestinal permeability in children with islet autoimmunity and type 1 diabetes: A prospective cohort study. Pediatr. Diabetes 2019, 20, 574–583. [Google Scholar] [CrossRef]
  42. Schoultz, I.; Keita, Å.V. The Intestinal Barrier and Current Techniques for the Assessment of Gut Permeability. Cells 2020, 9, 1909. [Google Scholar] [CrossRef] [PubMed]
  43. Rogler, G.; Singh, A.; Kavanaugh, A.; Rubin, D.T. Extraintestinal Manifestations of Inflammatory Bowel Disease: Current Concepts, Treatment, and Implications for Disease Management. Gastroenterology 2021, 161, 1118–1132. [Google Scholar] [CrossRef] [PubMed]
  44. Vavricka, S.R.; Schoepfer, A.; Scharl, M.; Lakatos, P.L.; Navarini, A.; Rogler, G. Extraintestinal Manifestations of Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2015, 21, 1982–1992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Gravina, A.; Federico, A.; Ruocco, E.; Lo Schiavo, A.; Romano, F.; Miranda, A.; Sgambato, D.; Dallio, M.; Ruocco, V.; Loguercio, C.; et al. Crohn’s disease and skin. United Eur. Gastroenterol. J. 2016, 4, 165–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Argyriou, K.; Khan, M.; Samuel, S. Multiple Unusual Ulcerated Skin Lesions in a Crohn’s Disease Patient. Gastroenterology 2018, 155, e17–e18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Chebli, J.M.F.; de Oliveira Moreira, B.; da Rocha Ribeiro, T.C. An Unusual Cause of Skin Rash in Crohn’s Disease. Gastroenterology 2018, 155, 618–620. [Google Scholar] [CrossRef] [Green Version]
  48. Chung-Yee Hui, R.; Kuo, K.L.; Yang, T.W. A Rare Skin Complication Associated With Crohn’s Disease. Gastroenterology 2021, 160, 669–670. [Google Scholar] [CrossRef]
  49. Rabiei, S.; Mohebbi, S.Z.; Patja, K.; Virtanen, J.I. Physicians’ knowledge of and adherence to improving oral health. BMC Public Health 2012, 12, 855. [Google Scholar] [CrossRef] [Green Version]
  50. Elad, S.; Zadik, Y.; Caton, J.G.; Epstein, J.B. Oral mucosal changes associated with primary diseases in other body systems. Periodontol. 2000 2019, 80, 28–48. [Google Scholar] [CrossRef]
  51. Del Monte, U. Does the cell number 109 still really fit one gram of tumor tissue? Cell Cycle 2009, 8, 505–506. [Google Scholar] [CrossRef] [Green Version]
  52. Vighi, G.; Marcucci, F.; Sensi, L.; Di Cara, G.; Frati, F. Allergy and the gastrointestinal system. Clin. Exp. Immunol. 2008, 153, 3–6. [Google Scholar] [CrossRef] [PubMed]
  53. Antoni, L.; Nuding, S.; Weller, D.; Gersemann, M.; Ott, G.; Wehkamp, J.; Stange, E.F. Human colonic mucus is a reservoir for antimicrobial peptides. J. Crohns Colitis 2013, 7, e652–e664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Vila, T.; Rizk, A.M.; Sultan, A.S.; Jabra-Rizk, M.A. The power of saliva: Antimicrobial and beyond. PLoS Pathog. 2019, 15, e1008058. [Google Scholar] [CrossRef] [PubMed]
  55. Wickett, R.R.; Visscher, M.O. Structure and function of the epidermal barrier. Am. J. Infect. Control 2006, 34, S98–S110. [Google Scholar] [CrossRef]
  56. Wells, J.M.; Brummer, R.J.; Derrien, M.; MacDonald, T.T.; Troost, F.; Cani, P.D.; Theodorou, V.; Dekker, J.; Méheust, A.; De Vos, W.M. Homeostasis of the gut barrier and potential biomarkers. Am. J. Physiol. Gastrointest. Liver. Physiol. 2017, 312, G171–G193. [Google Scholar] [CrossRef]
  57. Cornick, S.; Tawiah, A.; Chadee, K. Roles and regulation of the mucus barrier in the gut. Tissue Barriers 2015, 3, e982426. [Google Scholar] [CrossRef] [Green Version]
  58. Iweala, O.I.; Nagler, C.R. The Microbiome and Food Allergy. Annu. Rev. Immunol. 2019, 37, 377–403. [Google Scholar] [CrossRef]
  59. Zenobia, C.; Herpoldt, K.-L.; Freire, M. Is the oral microbiome a source to enhance mucosal immunity against infectious diseases? NPJ Vaccines 2021, 6, 80. [Google Scholar] [CrossRef]
  60. Lin, D.; Yang, L.; Wen, L.; Lu, H.; Chen, Q.; Wang, Z. Crosstalk between the oral microbiota, mucosal immunity, and the epithelial barrier regulates oral mucosal disease pathogenesis. Mucosal Immunol. 2021, 14, 1247–1258. [Google Scholar] [CrossRef]
  61. Adams, D. Keratinization of the oral epithelium. Ann. R. Coll. Surg. Engl. 1976, 58, 351–358. [Google Scholar]
  62. Walmsley, A.D.; Walsh, T.F.; Lumley, P.J.; Burke, F.J.T.; Shortall, A.C.C.; Hayes-Hall, R.; Pretty, I.A. Chapter 2—The healthy mouth. In Restorative Dentistry, 2nd ed.; Walmsley, A.D., Walsh, T.F., Lumley, P.J., Burke, F.J.T., Shortall, A.C.C., Hayes-Hall, R., Pretty, I.A., Eds.; Churchill Livingstone: Edinburgh, UK, 2007; pp. 3–11. [Google Scholar]
  63. Brodala, N.; Merricks, E.P.; Bellinger, D.A.; Damrongsri, D.; Offenbacher, S.; Beck, J.; Madianos, P.; Sotres, D.; Chang, Y.-L.; Koch, G.; et al. Porphyromonas gingivalis Bacteremia Induces Coronary and Aortic Atherosclerosis in Normocholesterolemic and Hypercholesterolemic Pigs. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 1446–1451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Forner, L.; Larsen, T.; Kilian, M.; Holmstrup, P. Incidence of bacteremia after chewing, tooth brushing and scaling in individuals with periodontal inflammation. J. Clin. Periodontol. 2006, 33, 401–407. [Google Scholar] [CrossRef] [PubMed]
  65. Lockhart, P.B.; Brennan, M.T.; Sasser, H.C.; Fox, P.C.; Paster, B.J.; Bahrani-Mougeot, F.K. Bacteremia associated with toothbrushing and dental extraction. Circulation 2008, 117, 3118–3125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Rosales, C.; Uribe-Querol, E. Neutrophil Role in Periodontal Disease; Khajah, M.A., Ed.; IntechOpen: London, UK, 2017; p. 67. [Google Scholar]
  67. Groeger, S.; Meyle, J. Oral Mucosal Epithelial Cells. Front. Immunol. 2019, 10, 208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Bosshardt, D.D.; Lang, N.P. The junctional epithelium: From health to disease. J. Dent. Res. 2005, 84, 9–20. [Google Scholar] [CrossRef]
  69. Lasserre, J.F.; Brecx, M.C.; Toma, S. Oral Microbes, Biofilms and Their Role in Periodontal and Peri-Implant Diseases. Materials 2018, 11, 1802. [Google Scholar] [CrossRef] [Green Version]
  70. Roberts, A.P.; Mullany, P. Oral biofilms: A reservoir of transferable, bacterial, antimicrobial resistance. Expert Rev. Anti-Infect. Ther. 2010, 8, 1441–1450. [Google Scholar] [CrossRef]
  71. Sharma, N.; Bhatia, S.; Sodhi, A.S.; Batra, N. Oral microbiome and health. AIMS Microbiol. 2018, 4, 42–66. [Google Scholar] [CrossRef]
  72. Yoneyama, T.; Yoshida, M.; Ohrui, T.; Mukaiyama, H.; Okamoto, H.; Hoshiba, K.; Ihara, S.; Yanagisawa, S.; Ariumi, S.; Morita, T. Oral care reduces pneumonia in older patients in nursing homes. J. Am. Geriatr. Soc. 2002, 50, 430–433. [Google Scholar] [CrossRef]
  73. Paju, S.; Scannapieco, F.A. Oral biofilms, periodontitis, and pulmonary infections. Oral Dis. 2007, 13, 508–512. [Google Scholar] [CrossRef] [Green Version]
  74. Miller, W.D. The Micro-Organisms of the Human Mouth; S.S. White Dental Manufacturing Co.: Philadelphia, PA, USA, 1890. [Google Scholar]
  75. He, X.S.; Shi, W.Y. Oral Microbiology: Past, Present and Future. Int. J. Oral Sci. 2009, 1, 47–58. [Google Scholar] [CrossRef] [PubMed]
  76. Miller, W.D. The human mouth as a focus of infection. Lancet 1891, 138, 340–342. [Google Scholar] [CrossRef] [Green Version]
  77. Hunter, W. Oral Sepsis as a Cause of Disease. Br. Med. J. 1900, 2, 215–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Billings, F. Chronic focal infections and their etiologic relations to arthritis and nephritis. JAMA Intern. Med. 1912, 9, 484–498. [Google Scholar] [CrossRef] [Green Version]
  79. Wessely, S. Surgery for the treatment of psychiatric illness: The need to test untested theories. J. R. Soc. Med. 2009, 102, 445–451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Reimann, H.A.; Havens, W.P. Focal infection and systemic disease: A critical appraisal: The case against indiscriminate removal of teeth and tonsils clinical lecture at St. Louis session. J. Am. Med. Assoc. 1940, 114, 1–6. [Google Scholar] [CrossRef]
  81. Gutmann, J.L. Focal Infection Revisited—The Swinging of the Pendulum. Dent. Hist. 2017, 62, 81–92. [Google Scholar]
  82. Potgieter, M.; Bester, J.; Kell, D.B.; Pretorius, E. The dormant blood microbiome in chronic, inflammatory diseases. FEMS Microbiol. Rev. 2015, 39, 567–591. [Google Scholar] [CrossRef] [Green Version]
  83. Païssé, S.; Valle, C.; Servant, F.; Courtney, M.; Burcelin, R.; Amar, J.; Lelouvier, B.J.T. Comprehensive description of blood microbiome from healthy donors assessed by 16S targeted metagenomic sequencing. Transfusion 2016, 56, 1138–1147. [Google Scholar] [CrossRef]
  84. Whittle, E.; Leonard, M.O.; Harrison, R.; Gant, T.W.; Tonge, D.P. Multi-Method Characterization of the Human Circulating Microbiome. Front. Microbiol. 2019, 9, 3266. [Google Scholar] [CrossRef] [Green Version]
  85. Dickson, R.P.; Huffnagle, G.B. The lung microbiome: New principles for respiratory bacteriology in health and disease. PLoS Pathog. 2015, 11, e1004923. [Google Scholar] [CrossRef] [PubMed]
  86. Man, W.H.; de Steenhuijsen Piters, W.A.A.; Bogaert, D. The microbiota of the respiratory tract: Gatekeeper to respiratory health. Nat. Rev. Microbiol. 2017, 15, 259–270. [Google Scholar] [CrossRef] [PubMed]
  87. Jin, C.; Lagoudas, G.K.; Zhao, C.; Bullman, S.; Bhutkar, A.; Hu, B.; Ameh, S.; Sandel, D.; Liang, X.S.; Mazzilli, S.; et al. Commensal Microbiota Promote Lung Cancer Development via γδ T Cells. Cell 2019, 176, 998–1013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Dominy, S.S.; Lynch, C.; Ermini, F.; Benedyk, M.; Marczyk, A.; Konradi, A.; Nguyen, M.; Haditsch, U.; Raha, D.; Griffin, C.; et al. Porphyromonas gingivalis in Alzheimer’s disease brains: Evidence for disease causation and treatment with small-molecule inhibitors. Sci. Adv. 2019, 5, eaau3333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Elwishahy, A.; Antia, K.; Bhusari, S.; Ilechukwu, N.C.; Horstick, O.; Winkler, V. Porphyromonas Gingivalis as a Risk Factor to Alzheimer’s Disease: A Systematic Review. J. Alzheimers Dis. Rep. 2021, 5, 721–732. [Google Scholar] [CrossRef]
  90. Nara, P.L.; Sindelar, D.; Penn, M.S.; Potempa, J.; Griffin, W.S.T. Porphyromonas gingivalis Outer Membrane Vesicles as the Major Driver of and Explanation for Neuropathogenesis, the Cholinergic Hypothesis, Iron Dyshomeostasis, and Salivary Lactoferrin in Alzheimer’s Disease. J. Alzheimers Dis. 2021, 82, 1417–1450. [Google Scholar] [CrossRef]
  91. Zhang, Z.; Liu, D.; Liu, S.; Zhang, S.; Pan, Y. The Role of Porphyromonas gingivalis Outer Membrane Vesicles in Periodontal Disease and Related Systemic Diseases. Front. Cell. Infect. Microbiol. 2021, 10, 585917. [Google Scholar] [CrossRef]
  92. Patel, S.; Howard, D.; Chowdhury, N.; Derieux, C.; Wellslager, B.; Yilmaz, Ö.; French, L. Characterization of Human Genes Modulated by Porphyromonas gingivalis Highlights the Ribosome, Hypothalamus, and Cholinergic Neurons. Front. Immunol. 2021, 12, 2165. [Google Scholar] [CrossRef]
  93. Fardini, Y.; Chung, P.; Dumm, R.; Joshi, N.; Han, Y.W. Transmission of diverse oral bacteria to murine placenta: Evidence for the oral microbiome as a potential source of intrauterine infection. Infect. Immun. 2010, 78, 1789–1796. [Google Scholar] [CrossRef] [Green Version]
  94. Kumar, P.S. From focal sepsis to periodontal medicine: A century of exploring the role of the oral microbiome in systemic disease. J. Physiol. 2017, 595, 465–476. [Google Scholar] [CrossRef]
  95. Kleinstein, S.; Nelson, K.; Freire, M. Inflammatory networks linking oral microbiome with systemic health and disease. J. Dent. Res. 2020, 99, 1131–1139. [Google Scholar] [CrossRef]
  96. Kartal, E.; Schmidt, T.S.B.; Molina-Montes, E.; Rodríguez-Perales, S.; Wirbel, J.; Maistrenko, O.M.; Akanni, W.A.; Alashkar Alhamwe, B.; Alves, R.J.; Carrato, A.; et al. A faecal microbiota signature with high specificity for pancreatic cancer. Gut 2022. [Google Scholar] [CrossRef] [PubMed]
  97. Wu, J.; Li, Q.; Fu, X. Fusobacterium nucleatum Contributes to the Carcinogenesis of Colorectal Cancer by Inducing Inflammation and Suppressing Host Immunity. Transl. Oncol. 2019, 12, 846–851. [Google Scholar] [CrossRef] [PubMed]
  98. Casasanta, M.A.; Yoo, C.C.; Udayasuryan, B.; Sanders, B.E.; Umaña, A.; Zhang, Y.; Peng, H.; Duncan, A.J.; Wang, Y.; Li, L.; et al. Fusobacterium nucleatum host-cell binding and invasion induces IL-8 and CXCL1 secretion that drives colorectal cancer cell migration. Sci. Signal. 2020, 13, eaba9157. [Google Scholar] [CrossRef] [PubMed]
  99. Loftus, M.; Hassouneh, S.A.-D.; Yooseph, S. Bacterial community structure alterations within the colorectal cancer gut microbiome. BMC Microbiol. 2021, 21, 98. [Google Scholar] [CrossRef] [PubMed]
  100. Zhang, S.; Li, C.; Zhang, Z.; Li, Y.; Li, Q.; Geng, F.; Liu, J.; Pan, Y. Analysis of differentially expressed genes in oral epithelial cells infected with Fusobacterium nucleatum for revealing genes associated with oral cancer. J. Cell. Mol. Med. 2021, 25, 892–904. [Google Scholar] [CrossRef]
  101. Han, Y.W.; Redline, R.W.; Li, M.; Yin, L.; Hill, G.B.; McCormick, T.S. Fusobacterium nucleatum induces premature and term stillbirths in pregnant mice: Implication of oral bacteria in preterm birth. Infect. Immun. 2004, 72, 2272–2279. [Google Scholar] [CrossRef] [Green Version]
  102. Vander Haar, E.L.; So, J.; Gyamfi-Bannerman, C.; Han, Y.W. Fusobacterium nucleatum and adverse pregnancy outcomes: Epidemiological and mechanistic evidence. Anaerobe 2018, 50, 55–59. [Google Scholar] [CrossRef]
  103. Fan, X.; Alekseyenko, A.V.; Wu, J.; Peters, B.A.; Jacobs, E.J.; Gapstur, S.M.; Purdue, M.P.; Abnet, C.C.; Stolzenberg-Solomon, R.; Miller, G.; et al. Human oral microbiome and prospective risk for pancreatic cancer: A population-based nested case-control study. Gut 2018, 67, 120–127. [Google Scholar] [CrossRef] [Green Version]
  104. 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]
  105. Wang, X.; Jia, Y.; Wen, L.; Mu, W.; Wu, X.; Liu, T.; Liu, X.; Fang, J.; Luan, Y.; Chen, P.; et al. Porphyromonas gingivalis Promotes Colorectal Carcinoma by Activating the Hematopoietic NLRP3 Inflammasome. Cancer Res. 2021, 81, 2745–2759. [Google Scholar] [CrossRef] [PubMed]
  106. Chen, Y.; Huang, Z.; Tang, Z.; Huang, Y.; Huang, M.; Liu, H.; Ziebolz, D.; Schmalz, G.; Jia, B.; Zhao, J. More Than Just a Periodontal Pathogen–the Research Progress on Fusobacterium nucleatum. Front. Cell. Infect. Microbiol. 2022, 12, 64. [Google Scholar] [CrossRef]
  107. Mohammed, H.; Varoni, E.; Cochis, A.; Cordaro, M.; Gallenzi, P.; Patini, R.; Staderini, E.; Lajolo, C.; Rimondini, L.; Rocchetti, V. Oral Dysbiosis in Pancreatic Cancer and Liver Cirrhosis: A Review of the Literature. Biomedicines 2018, 6, 115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Flak, M.B.; Colas, R.A.; Muñoz-Atienza, E.; Curtis, M.A.; Dalli, J.; Pitzalis, C. Inflammatory arthritis disrupts gut resolution mechanisms, promoting barrier breakdown by Porphyromonas gingivalis. JCI Insight 2019, 4, e125191. [Google Scholar] [CrossRef]
  109. Reyes, L. Porphyromonas gingivalis. Trends Microbiol. 2021, 29, 376–377. [Google Scholar] [CrossRef] [PubMed]
  110. Wang, J.; Qi, J.; Zhao, H.; He, S.; Zhang, Y.; Wei, S.; Zhao, F. Metagenomic sequencing reveals microbiota and its functional potential associated with periodontal disease. Sci. Rep. 2013, 3, 1843. [Google Scholar] [CrossRef]
  111. Padmalatha, G.; Bavle, R.; Satyakiran, G.; Paremala, K.; Sudhakara, M.; Makarla, S. Quantification of Porphyromonas gingivalis in chronic periodontitis patients associated with diabetes mellitus using real-time polymerase chain reaction. J. Oral Maxillofac. Pathol. 2016, 20, 413–418. [Google Scholar] [CrossRef]
  112. Matsha, T.E.; Prince, Y.; Davids, S.; Chikte, U.; Erasmus, R.T.; Kengne, A.P.; Davison, G.M. Oral Microbiome Signatures in Diabetes Mellitus and Periodontal Disease. J. Dent. Res. 2020, 99, 658–665. [Google Scholar] [CrossRef]
  113. Wu, J.S.; Zheng, M.; Zhang, M.; Pang, X.; Li, L.; Wang, S.S.; Yang, X.; Wu, J.B.; Tang, Y.J.; Tang, Y.L.; et al. Porphyromonas gingivalis Promotes 4-Nitroquinoline-1-Oxide-Induced Oral Carcinogenesis With an Alteration of Fatty Acid Metabolism. Front. Microbiol. 2018, 9, 2081. [Google Scholar] [CrossRef]
  114. Wen, L.; Mu, W.; Lu, H.; Wang, X.; Fang, J.; Jia, Y.; Li, Q.; Wang, D.; Wen, S.; Guo, J.; et al. Porphyromonas gingivalis Promotes Oral Squamous Cell Carcinoma Progression in an Immune Microenvironment. J. Dent. Res. 2020, 99, 666–675. [Google Scholar] [CrossRef]
  115. Riviere, G.R.; Riviere, K.; Smith, K. Molecular and immunological evidence of oral Treponema in the human brain and their association with Alzheimer’s disease. Oral Microbiol. Immunol. 2002, 17, 113–118. [Google Scholar] [CrossRef] [PubMed]
  116. Hajishengallis, G. Immunomicrobial pathogenesis of periodontitis: Keystones, pathobionts, and host response. Trends Immunol. 2014, 35, 3–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Hajishengallis, G.; Lamont, R.J. Dancing with the stars: How choreographed bacterial interactions dictate nososymbiocity and give rise to keystone pathogens, accessory pathogens, and pathobionts. Trends Microbiol. 2016, 24, 477–489. [Google Scholar] [CrossRef] [Green Version]
  118. Costa, M.J.F.; Araújo, I.D.T.d.; Alves, L.d.R.; Silva, R.L.d.; Calderon, P.d.S.; Borges, B.C.D.; Martins, A.R.L.d.A.; Gurgel, B.C.d.V.; Lins, R.D.A.U. Relationship of Porphyromonas gingivalis and Alzheimer’s disease: A systematic review of pre-clinical studies. Clin. Oral Investig. 2021, 25, 797–806. [Google Scholar] [CrossRef] [PubMed]
  119. Mougeot, J.L.C.; Stevens, C.B.; Paster, B.J.; Brennan, M.T.; Lockhart, P.B.; Mougeot, F.K.B. Porphyromonas gingivalis is the most abundant species detected in coronary and femoral arteries. J. Oral Microbiol. 2017, 9, 1281562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Slocum, C.; Kramer, C.; Genco, C. Immune dysregulation mediated by the oral microbiome: Potential link to chronic inflammation and atherosclerosis. J. Intern. Med. 2016, 280, 114–128. [Google Scholar] [CrossRef]
  121. Farrugia, C.; Stafford, G.P.; Potempa, J.; Wilkinson, R.N.; Chen, Y.; Murdoch, C.; Widziolek, M. Mechanisms of vascular damage by systemic dissemination of the oral pathogen Porphyromonas gingivalis. FEBS J. 2021, 288, 1479–1495. [Google Scholar] [CrossRef]
  122. Lee, K.; Roberts, J.S.; Choi, C.H.; Atanasova, K.R.; Yilmaz, Ö. Porphyromonas gingivalis traffics into endoplasmic reticulum-rich-autophagosomes for successful survival in human gingival epithelial cells. Virulence 2018, 9, 845–859. [Google Scholar] [CrossRef] [Green Version]
  123. Lee, J.S.; Chowdhury, N.; Roberts, J.S.; Yilmaz, Ö. Host surface ectonucleotidase-CD73 and the opportunistic pathogen, Porphyromonas gingivalis, cross-modulation underlies a new homeostatic mechanism for chronic bacterial survival in human epithelial cells. Virulence 2020, 11, 414–429. [Google Scholar] [CrossRef]
  124. Lee, J.S.; Spooner, R.; Chowdhury, N.; Pandey, V.; Wellslager, B.; Atanasova, K.R.; Evans, Z.; Yilmaz, Ö. In Situ Intraepithelial Localizations of Opportunistic Pathogens, Porphyromonas gingivalis and Filifactor alocis, in Human Gingiva. Curr. Res. Microb. Sci. 2020, 1, 7–17. [Google Scholar] [CrossRef]
  125. Dong, J.; Li, Y.; Xiao, H.; Zhang, S.; Wang, B.; Wang, H.; Li, Y.; Fan, S.; Cui, M. Oral microbiota affects the efficacy and prognosis of radiotherapy for colorectal cancer in mouse models. Cell Rep. 2021, 37, 109866. [Google Scholar] [CrossRef] [PubMed]
  126. Pizzo, G.; Guiglia, R.; Russo, L.L.; Campisi, G. Dentistry and internal medicine: From the focal infection theory to the periodontal medicine concept. Eur. J. Intern. Med. 2010, 21, 496–502. [Google Scholar] [CrossRef] [PubMed]
  127. Beck, J.D.; Papapanou, P.N.; Philips, K.H.; Offenbacher, S. Periodontal Medicine: 100 Years of Progress. J. Dent. Res. 2019, 98, 1053–1062. [Google Scholar] [CrossRef]
  128. Chou, H.H.; Yumoto, H.; Davey, M.; Takahashi, Y.; Miyamoto, T.; Gibson, F.C., 3rd; Genco, C.A. Porphyromonas gingivalis fimbria-dependent activation of inflammatory genes in human aortic endothelial cells. Infect. Immun. 2005, 73, 5367–5378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Lalla, E.; Lamster, I.B.; Hofmann, M.A.; Bucciarelli, L.; Jerud, A.P.; Tucker, S.; Lu, Y.; Papapanou, P.N.; Schmidt, A.M. Oral infection with a periodontal pathogen accelerates early atherosclerosis in apolipoprotein E-null mice. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 1405–1411. [Google Scholar] [CrossRef] [PubMed]
  130. Nakamura, N.; Yoshida, M.; Umeda, M.; Huang, Y.; Kitajima, S.; Inoue, Y.; Ishikawa, I.; Iwai, T. Extended exposure of lipopolysaccharide fraction from Porphyromonas gingivalis facilitates mononuclear cell adhesion to vascular endothelium via Toll-like receptor-2 dependent mechanism. Atherosclerosis 2008, 196, 59–67. [Google Scholar] [CrossRef] [PubMed]
  131. Zhang, T.; Kurita-Ochiai, T.; Hashizume, T.; Du, Y.; Oguchi, S.; Yamamoto, M. Aggregatibacter actinomycetemcomitans accelerates atherosclerosis with an increase in atherogenic factors in spontaneously hyperlipidemic mice. FEMS Immunol. Med. Microbiol. 2010, 59, 143–151. [Google Scholar] [CrossRef] [Green Version]
  132. Tuomainen, A.M.; Jauhiainen, M.; Kovanen, P.T.; Metso, J.; Paju, S.; Pussinen, P.J. Aggregatibacter actinomycetemcomitans induces MMP-9 expression and proatherogenic lipoprotein profile in apoE-deficient mice. Microb. Pathog. 2008, 44, 111–117. [Google Scholar] [CrossRef]
  133. Gough, P.J.; Gomez, I.G.; Wille, P.T.; Raines, E.W. Macrophage expression of active MMP-9 induces acute plaque disruption in apoE-deficient mice. J. Clin. Investig. 2006, 116, 59–69. [Google Scholar] [CrossRef] [Green Version]
  134. 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] [Green Version]
  135. Blasco-Baque, V.; Garidou, L.; Pomié, C.; Escoula, Q.; Loubieres, P.; Le Gall-David, S.; Lemaitre, M.; Nicolas, S.; Klopp, P.; Waget, A.; et al. Periodontitis induced by Porphyromonas gingivalis drives periodontal microbiota dysbiosis and insulin resistance via an impaired adaptive immune response. Gut 2017, 66, 872–885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Tian, J.; Liu, C.; Zheng, X.; Jia, X.; Peng, X.; Yang, R.; Zhou, X.; Xu, X. Porphyromonas gingivalis Induces Insulin Resistance by Increasing BCAA Levels in Mice. J. Dent. Res. 2020, 99, 839–846. [Google Scholar] [CrossRef] [PubMed]
  137. Liu, Y.; Wu, Z.; Nakanishi, Y.; Ni, J.; Hayashi, Y.; Takayama, F.; Zhou, Y.; Kadowaki, T.; Nakanishi, H. Infection of microglia with Porphyromonas gingivalis promotes cell migration and an inflammatory response through the gingipain-mediated activation of protease-activated receptor-2 in mice. Sci. Rep. 2017, 7, 11759. [Google Scholar] [CrossRef] [PubMed]
  138. Memedovski, Z.; Czerwonka, E.; Han, J.; Mayer, J.; Luce, M.; Klemm, L.C.; Hall, M.L.; Mayer, A.M.S. Classical and Alternative Activation of Rat Microglia Treated with Ultrapure Porphyromonas gingivalis Lipopolysaccharide In Vitro. Toxins 2020, 12, 333. [Google Scholar] [CrossRef] [PubMed]
  139. Poole, S.; Singhrao, S.K.; Chukkapalli, S.; Rivera, M.; Velsko, I.; Kesavalu, L.; Crean, S. Active invasion of Porphyromonas gingivalis and infection-induced complement activation in ApoE-/- mice brains. J. Alzheimers Dis. 2015, 43, 67–80. [Google Scholar] [CrossRef] [Green Version]
  140. Ishida, N.; Ishihara, Y.; Ishida, K.; Tada, H.; Funaki-Kato, Y.; Hagiwara, M.; Ferdous, T.; Abdullah, M.; Mitani, A.; Michikawa, M.; et al. Periodontitis induced by bacterial infection exacerbates features of Alzheimer’s disease in transgenic mice. NPJ Aging Mech. Dis. 2017, 3, 15. [Google Scholar] [CrossRef] [Green Version]
  141. Wu, Z.; Ni, J.; Liu, Y.; Teeling, J.L.; Takayama, F.; Collcutt, A.; Ibbett, P.; Nakanishi, H. Cathepsin B plays a critical role in inducing Alzheimer’s disease-like phenotypes following chronic systemic exposure to lipopolysaccharide from Porphyromonas gingivalis in mice. Brain Behav. Immun. 2017, 65, 350–361. [Google Scholar] [CrossRef]
  142. Leira, Y.; Iglesias-Rey, R.; Gómez-Lado, N.; Aguiar, P.; Campos, F.; D’Aiuto, F.; Castillo, J.; Blanco, J.; Sobrino, T. Porphyromonas gingivalis lipopolysaccharide-induced periodontitis and serum amyloid-beta peptides. Arch. Oral Biol. 2019, 99, 120–125. [Google Scholar] [CrossRef]
  143. Tang, Z.; Liang, D.; Cheng, M.; Su, X.; Liu, R.; Zhang, Y.; Wu, H. Effects of Porphyromonas gingivalis and Its Underlying Mechanisms on Alzheimer-Like Tau Hyperphosphorylation in Sprague-Dawley Rats. J. Mol. Neurosci. 2021, 71, 89–100. [Google Scholar] [CrossRef]
  144. Wu, H.; Ballantyne, C.M. Metabolic Inflammation and Insulin Resistance in Obesity. Circ. Res. 2020, 126, 1549–1564. [Google Scholar] [CrossRef]
  145. Sharma, S.; Tripathi, P. Gut microbiome and type 2 diabetes: Where we are and where to go? J. Nutr. Biochem. 2019, 63, 101–108. [Google Scholar] [CrossRef] [PubMed]
  146. Javed, F.; Warnakulasuriya, S. Is there a relationship between periodontal disease and oral cancer? A systematic review of currently available evidence. Crit. Rev. Oncol. Hematol. 2016, 97, 197–205. [Google Scholar] [CrossRef] [PubMed]
  147. Fang, L.Y.; Wong, T.Y.; Chiang, W.F.; Chen, Y.L. Fatty-acid-binding protein 5 promotes cell proliferation and invasion in oral squamous cell carcinoma. J. Oral Pathol. Med. 2010, 39, 342–348. [Google Scholar] [CrossRef] [PubMed]
  148. Lee, D.; Wada, K.; Taniguchi, Y.; Al-Shareef, H.; Masuda, T.; Usami, Y.; Aikawa, T.; Okura, M.; Kamisaki, Y.; Kogo, M. Expression of fatty acid binding protein 4 is involved in the cell growth of oral squamous cell carcinoma. Oncol. Rep. 2014, 31, 1116–1120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Zhang, Q.Q.; Hu, X.W.; Liu, Y.L.; Ye, Z.J.; Gui, Y.H.; Zhou, D.L.; Qi, C.L.; He, X.D.; Wang, H.; Wang, L.J. CD11b deficiency suppresses intestinal tumor growth by reducing myeloid cell recruitment. Sci. Rep. 2015, 5, 15948. [Google Scholar] [CrossRef]
Figure 1. Schematics of differences in the cellular junctions between the intestinal and oral epithelia. (a) The intestinal epithelia are interconnected and communicate with each other through junctions, such as tight junction (TJ), adherence junction (AJ), desmosome (DM), and gap junction (GJ, not shown). (b) When these barriers are disrupted because of epithelial damages, pathogens, and chemicals in the luminal side can leak through the damaged cellular gaps into the lamina propria (blue arrows) whereby the MALT implements immune responses, resulting in a leaky gut syndrome. (c) Keratinized and non-keratinized oral mucosa. (d) Unlike the intestinal leakage through the cell-to-cell junctions, the leakage in the oral mucosa occurs through the hemidesmosome (HD) between the basal layer of the junctional epithelium (JE) and the hard surface layer of a tooth, which is inevitably and more frequently exposed to the physical and biological challenges. The internal basal lamina (IBL), an HD interface, is inhabited with collagens and binding proteins, such as laminin-332 and integrin. The periodontal pocket (PP), a pathologically deepened gingival sulcus (GS), occurs with the detachment of the connective tissues of the gingiva from the tooth surface. The JE below the GS is ~0.15 mm wide and 1–2 mm high, remains non-keratinized and undifferentiated, and has the highest turnover rate (4–6 days) of all oral epithelia. The polymorphonuclear leukocytes (PMNs) are also secreted with gingival crevicular fluid (GCF) from the basal layer to keep a lookout for any hostile intruders. ZO-1: zonula occludens-1, JAM: junctional adhesion molecule.
Figure 1. Schematics of differences in the cellular junctions between the intestinal and oral epithelia. (a) The intestinal epithelia are interconnected and communicate with each other through junctions, such as tight junction (TJ), adherence junction (AJ), desmosome (DM), and gap junction (GJ, not shown). (b) When these barriers are disrupted because of epithelial damages, pathogens, and chemicals in the luminal side can leak through the damaged cellular gaps into the lamina propria (blue arrows) whereby the MALT implements immune responses, resulting in a leaky gut syndrome. (c) Keratinized and non-keratinized oral mucosa. (d) Unlike the intestinal leakage through the cell-to-cell junctions, the leakage in the oral mucosa occurs through the hemidesmosome (HD) between the basal layer of the junctional epithelium (JE) and the hard surface layer of a tooth, which is inevitably and more frequently exposed to the physical and biological challenges. The internal basal lamina (IBL), an HD interface, is inhabited with collagens and binding proteins, such as laminin-332 and integrin. The periodontal pocket (PP), a pathologically deepened gingival sulcus (GS), occurs with the detachment of the connective tissues of the gingiva from the tooth surface. The JE below the GS is ~0.15 mm wide and 1–2 mm high, remains non-keratinized and undifferentiated, and has the highest turnover rate (4–6 days) of all oral epithelia. The polymorphonuclear leukocytes (PMNs) are also secreted with gingival crevicular fluid (GCF) from the basal layer to keep a lookout for any hostile intruders. ZO-1: zonula occludens-1, JAM: junctional adhesion molecule.
Cells 11 01079 g001
Figure 2. Biofilms built on (a) a tooth’s surface and (b) extracted implants. The hard surface of a tooth root, an implant, and a crown prosthesis abutting an implant shaft provide a solid ground on which biofilms can accumulate for a lifetime if not well cared for. (c) The edentate oral cavity. The toothless oral mucosa is free of the GS and the JE, making it less vulnerable to infection.
Figure 2. Biofilms built on (a) a tooth’s surface and (b) extracted implants. The hard surface of a tooth root, an implant, and a crown prosthesis abutting an implant shaft provide a solid ground on which biofilms can accumulate for a lifetime if not well cared for. (c) The edentate oral cavity. The toothless oral mucosa is free of the GS and the JE, making it less vulnerable to infection.
Cells 11 01079 g002
Figure 3. The oral cavity as the origin of the internal microbiome in humans. The microbiome in the oral cavity can disseminate to the remote sites of the body, such as the brain, stomach, intestines, and heart, via hematogenous and enteric pathways. The PP, a pathologically deepened GS due to microbial infection and colonization, gradually allows detachment of the connective tissues of the JE from the tooth surface. The epithelial layer of the apical JE is thin enough for bacterial virulence factors as well as pathogenic bacteria, such as P. gingivalis, to infiltrate into the bloodstream, resulting in a leaky gum syndrome. The microbiome of the oral cavity also affects gut microbiome profiles by moving through the gastrointestinal tract, causing a variety of gut-related diseases, such as IBD, IBS, and colon cancer.
Figure 3. The oral cavity as the origin of the internal microbiome in humans. The microbiome in the oral cavity can disseminate to the remote sites of the body, such as the brain, stomach, intestines, and heart, via hematogenous and enteric pathways. The PP, a pathologically deepened GS due to microbial infection and colonization, gradually allows detachment of the connective tissues of the JE from the tooth surface. The epithelial layer of the apical JE is thin enough for bacterial virulence factors as well as pathogenic bacteria, such as P. gingivalis, to infiltrate into the bloodstream, resulting in a leaky gum syndrome. The microbiome of the oral cavity also affects gut microbiome profiles by moving through the gastrointestinal tract, causing a variety of gut-related diseases, such as IBD, IBS, and colon cancer.
Cells 11 01079 g003
Table 1. Comparison of skin, oral mucosa, and intestinal mucosa.
Table 1. Comparison of skin, oral mucosa, and intestinal mucosa.
EpitheliumSkinOralIntestinal
KERATINIZED TISSUEExistPartially existNot exist
EPITHELIAL LAYERMultiple layersMultiple layersSingle layer
INTERCELLULAR JUNCTIONSTight junctionExistExistExist
Adherence junctionExistExistExist
DesmosomeExistExistExist
Gap junctionExistExistExist
HemidesomosomeNot existExistNot exist
Table 2. The systemic disease models induced by oral pathogens.
Table 2. The systemic disease models induced by oral pathogens.
Oral PathogensModelsInfection MethodsExperimental ResultsYearRef.
Atherosclerotic CVD In vitro P. gingivalis 381HAECs6 h infectionIncreased ICAM-1, VCAM-1, and IL-6 expression2005[128]
P. gingivalis ATCC33277-driven PgLPSHUVECs24 h infectionIncreased adhesion of mononuclear cells to HUVECs via ICAM-1 and TLR-2 dependent mechanism.2008[130]
In vivo P. gingivalis 381Apoe−/− mice
  • Infected (n = 25)
  • Non-infected (n = 25)
Oral infection 5 times per week over 3 weeksIncreased aortic atherosclerosis.2003[129]
P. gingivalis 381Apoe−/− mice
  • Infected (n = 6)
  • Non-infected (n = 6)
Oral infection 5 times per week over 3 weeksIncreased aortic ICAM-1, VCAM-1 immunostaining.2005[128]
P. gingivalis 381 or A7436Pigs
  • Infected (n = 23)
  • Non-infected (n = 13)
Subcutaneously infection 3 times per week for 5 monthsIncreased aortic and coronary arterial atherosclerosis.2005[63]
A. actinomycetemcomitans AT445bApoe−/− mice
  • Infected (n = 9)
  • Non-infected (n = 9)
Intravenous infection once a week for 4, 6, or 8 weeksIncreased aortic MMP-9 expression and serum CRP.2008[132]
A. actinomycetemcomitans HK1651Apoeshl mice
  • Infected (n = 6)
  • Non-infected (n = 6)
Intravenous infection 3 times per week over 3 weeksIncreased atherosclerotic plaque, serum C-reactive protein (CRP), IL-6, and aortic ICAM-1.2014[134]
T2DM In vivo P. gingivalis W83MiceOral infection twice per week for 5 weeksIncreased gut dysbiosis, gut barrier invasion, serum endotoxin, insulin resistance.2014[134]
P. gingivalis ATCC33277,
F. nucleatum,
P. intermedia
Mice
  • Infected (n = 16)
  • Non-infected (n = 13)
Oral infection 4 times a week for 4 weeks, thereafter normal diet or HFD-fed for additional 3 monthsIncreased periodontal dysbiosis, insulin resistance in HFD-fed mice.2017[135]
P. gingivalis ATCC33277 (WT) or ∆bcatMice
  • WT infected (n = 6)
  • bcat infected (n = 6)
  • Non-infected (n = 6)
Oral infection twice per week for 4 weeks concomitantly HFD-fedP. gingivalis (∆bcat) cannot induce insulin resistance in HFD-fed mice.2020[136]
OSCC In vivo P. gingivalis 381Mice
  • Infected (n = 15)
  • 4NQO-treated (n = 20)
  • 4NQO-treated + infected (n = 20)
  • Control (n = 10)
4NQO treatment for 8 weeks, thereafter oral infection with P. gingivalis for 8 weeksEnhanced OSCC induction and dysregulated lipid metabolism in 4NQO-treated mice.2018[113]
P. gingivalis ATCC33277Mice
  • 4NQO-treated + infected (n = 12)
  • Non-infected (n = 6)
4NQO treatment for 16 weeks, thereafter oral infection with P. gingivalis for 10 weeksEnhanced OSCC induction and increased infiltration of CD11b+ MDSCs in 4NQO-treated mice.2020[114]
AD In vitro P. gingivalis ATCC33277Immortalized mouse microglial cell line MG63, 6, or 12 h infection of P. gingivalis in the presence and absence of KYT1 (Rgp inhibitor) and KYT36 (Kgp inhibitor)Increased expression levels of IL-6 and TNF-α, which was inhibited by KYT1 and KYT36 treatment.2017[137]
PgLPSRat brain neonatal microglia18 h infectionActivated microglial release of cytokine TNF-α, IL-6, and MMP-9.2020[138]
In vivo P. gingivalis 381, Treponema denticola ATCC 35404, Tannerella forsythia ATCC 43037, and F. nucleatum ATCC 49256Apoe−/− mice
  • Mono-infected (n = 12)
  • Multi-infected (n = 12)
  • Non-infected (n = 12)
Oral infection for 24 weeksP. gingivalis genomic DNA was detected in mice brain (9 out of 12 at 24 weeks).2015[139]
P. gingivalis ATCC33277APP transgenic mice
  • Infected (n = 14)
  • Non-infected (n = 12)
Gingival infectionExacerbated Aβ plaques and inflammatory cytokines in the brain of AD mouse model.2017[140]
PgLPSMice
  • Young WT mice (2 months, n = 6)
  • Middle-aged WT mice (12 months, n = 6)
  • Young Catb−/− mice (n = 6)
  • Middle-aged Catb−/− mice (n = 6)
Intraperitoneal infection daily for 5 weeksPgLPS induced learning and memory deficit in middle-aged WT mice, but not in young WT, young Catb−/−, and middle-aged Catb−/− mice.2017[141]
P. gingivalis ATCC33277,
Kgp-deficient P. gingivalis KDP129
Cx3cr1+/GFP miceInjection of P. gingivalis into the somatosensory cortex of miceGFP+ microglia accumulated around the injection site of P. gingivalis, but not of KDP129.2017[137]
PgLPSRats (n = 6)Palatal gingival infection 3 times for 2 weeksInduced alveolar bone loss and increased serum Aβ levels.2019[137,142]
P. gingivalis ATCC33277Rats
  • Infected for 4 weeks (n = 10)
  • Non-infected for 4 weeks (n = 10)
  • Infected for 12 weeks (n = 10)
  • Non-infected for 12 weeks (n = 10)
Intravenous infection 3 times a week for 4 or 12 weeksInduced tau hyperphosphorylation (pTau181 and pTau231) in the rat hippocampus.2021[143]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Park, D.-Y.; Park, J.Y.; Lee, D.; Hwang, I.; Kim, H.-S. Leaky Gum: The Revisited Origin of Systemic Diseases. Cells 2022, 11, 1079. https://doi.org/10.3390/cells11071079

AMA Style

Park D-Y, Park JY, Lee D, Hwang I, Kim H-S. Leaky Gum: The Revisited Origin of Systemic Diseases. Cells. 2022; 11(7):1079. https://doi.org/10.3390/cells11071079

Chicago/Turabian Style

Park, Do-Young, Jin Young Park, Dahye Lee, Inseong Hwang, and Hye-Sung Kim. 2022. "Leaky Gum: The Revisited Origin of Systemic Diseases" Cells 11, no. 7: 1079. https://doi.org/10.3390/cells11071079

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