Korean J Helicobacter  Up Gastrointest Res Search

CLOSE


Korean J Helicobacter  Up Gastrointest Res > Volume 24(4); 2024 > Article
Park, Kim, Kim, Hwang, Lee, Hwang, and Hwang: Association Between Oral Microbiota and Gastrointestinal/Extra-Gastrointestinal Diseases

Abstract

Dental clinicians and researchers have recently recommended oral microbial examinations to more accurately diagnose and treat oral diseases, including periodontitis and dental caries. Theoretical and experimental evidence suggests that oral microbiota may also be associated with non-oral diseases, such as gastrointestinal and extra-gastrointestinal diseases. This review highlights studies demonstrating microbial alterations in the oral cavity associated with malignant tumors including gastric, colorectal, esophageal, and lung cancers, implying that these alterations may serve as early indicators for non-invasive diagnosis and risk assessment of cancer development. Furthermore, we addressed the implications of oral microbial co-occurrence with malignant tumors, such as Streptococcus anginosus, Fusobacterium nucleatum, and Veillonella parvula, which are recognized as tumor-enriched oral pathogens involved in the development and progression of cancers in the stomach, colon, and lungs, respectively. Notably, we explored the immune and inflammatory mechanisms underlying reciprocal interactions between oral microbiota and tumors, underscoring that targeting these mechanistic pathways can contribute to preventing cancer development.

INTRODUCTION

The oral cavity, the second-largest microbial habitat in the human body, is colonized by diverse microorganisms that inhabit the buccal mucosa, dental pellicle, tongue, gingiva, and saliv [1]. The microbiota in the oral cavity of newborns is predominantly composed of maternal vaginal or skin microbes, which are immediately replaced by oral microbiota that are common in all infants and adults [2]. The microbial distribution in the oral cavity is easily influenced by various factors, such as food consumption [3], smoking [4], nationality [5], and underlying diseases [6]. Importantly, the chronic dominance of pathogenic bacteria in the oral cavity, referred to as oral dysbiosis, has been widely recognized to contribute to the progression of various oral diseases, including periodontitis [7-16], dental caries [17], and oral cancer [18]. Furthermore, recent studies highlighted the association between oral microbiota and systemic, non-oral diseases mainly due to the microbial translocation to other organs, either by passing through the esophagus via swallowing [19] or entering the bloodstream via leaky gums [6]. This review aimed to explore recent research investigating the potential roles of the oral microbiota in both gastrointestinal and extra-gastrointestinal diseases.

ORAL MICROBIOTA AND GASTROINTESTINAL DISEASES

Oral microbial alterations associated with gastrointestinal diseases

Advances in metagenome sequencing technologies have enhanced our understanding of the strong association between oral microbial alterations and gastrointestinal diseases, including gastric and colorectal cancers (Table 1). Sun et al. [20] analyzed saliva or plaque samples from 37 gastric cancer patients and 13 healthy controls and reported considerable differential abundances in oral microbial compositions between the two groups. Specifically, compared with healthy controls, gastric cancer patients exhibited that relative abundance of Veillonella, Prevotella, Aggregatibacter, and Megasphaera increased, whereas Leptotrichia, Rothia, Capnocytophaga, Campylobacter, Tannerella, and Granulicatella decreased. Russo et al. [21] analyzed the saliva samples from 10 patients with colorectal cancer and 10 healthy controls using similar sequencing techniques and revealed an upregulation of Actinobacteria, Saccharibacteria, Proteobacteria, Fusobacteria, Firmicutes, and Bacteroidetes. These findings imply that oral microbial alterations may serve as non-invasive biomarkers for the early detection and prevention of gastrointestinal diseases. However, the precise mechanisms linking these oral microbial alterations and gastrointestinal diseases remains unclear.

Enrichment of oral microbiota in gastrointestinal cancer tissues

Gastrointestinal cancer tissues are highly colonized by oral microbiota [22-24], suggesting a reciprocal influence between these microbes and gastrointestinal cancer development via direct interaction (Table 2). To identify specific oral microbiota enriched in gastric cancer sites, gastric mucosal samples were collected from patients according to disease stages (20 gastric cancer, 17 intestinal metaplasia, 23 atrophic gastritis, and 21 superficial gastritis) to analyze the distribution of oral microbiota. The results revealed that the relative abundance of Streptococcus anginosus, Parvimonas micra, Dialister pneumosintes, Slackia exigua, Fusobacterium nucleatum, Prevotella intermedia, Catonella morbi, and Peptostreptococcus stomatis was significantly higher in samples from gastric cancer than in samples from all other stages [24], all of which belonged to the Human Oral Microbiome Database (HOMD, https://homd.org), developed through the Human Microbiome Project (Table 2). Similar findings have been observed in colorectal cancer tissues, which were also colonized by oral microbiota. Tran et al. [23] compared the microbial compositions of colorectal malignant tumors and non-malignant polyps and found that the compositional percentages of oral microbiota, including Gemella, Peptostreptococcus, F. nucleatum, Leptotrichia, Selenomonas sputigena, and Campylobacter rectus, were significantly higher in malignant tumors than in non-malignant polyps. Additionally, Loftus et al. [22] identified that oral microbiota, including F. nucleatum, P. stomatis, Gemella morbillorum, and P. micra, were differentially abundant in fecal samples from 74 colorectal cancer patients than in 178 healthy controls.

Interaction and relationship between oral microbiota and gastrointestinal microbiota

You et al. [25] elucidated the interaction between oral microbiota and gastrointestinal tumoral microbiota through simultaneous 16S rRNA metagenomic analysis of saliva and gastric juice samples from 141 patients with different gastric cancer stages (58 low-grade dysplasia, 33 high-grade dysplasia, and 41 gastric cancer) and 9 healthy controls. As shown in Table 3, several oral-origin bacteria were significantly enriched in the gastric juice samples of the disease groups than in the healthy controls. Notably, Gemella haemolysans was significantly enriched in both the oral cavity and stomach of the disease groups than in the control group, suggesting that the presence of G. haemolysans in the oral cavity could serve as a non-invasive marker for predicting gastric diseases. A key finding was the increased similarity in microbial compositions between the oral cavity and stomach with disease progression, indicating a potential role for oral-gastric bacterial interactions in gastric cancer development.
In addition to gastric cancer, the interactions between oral and stomach microbiota contribute to early-stage intramucosal esophageal squamous carcinoma (EIESC) [26]. Chen et al. [26] compared the 16S rRNA metagenome results of both saliva and gastric antrum tissues from 31 patients with EIESC with those obtained from 21 healthy controls and revealed a significant enrichment of Streptococcus in the gastric samples of the EIESC group than in the control group, whereas the salivary levels of Streptococcus did not differ between the two groups. Notably, Porphyromonas endodontalis was identified as a potential biomarker for differentiating EIESC patients from healthy controls. The higher abundance of P. endodontalis in the saliva samples of EIESC patients could aid in early diagnosis, although the gastric P. endodontalis did not differ between the two groups. These findings highlight the importance of the combined analysis of both oral and gastric microbiota for improving diagnosis and potential treatment strategies for EIESC.

Modulating effects of oral pathogens on gastrointestinal tumorigenesis

Role of S. anginosus in gastric cancer pathogenesis

Although Helicobacter pylori is a major risk factor for gastric adenocarcinoma [27], the involvement of non-H. pylori pathogens in gastric tumorigenesis has been consistently suggested because of a weak association between the gastric abundance of H. pylori and gastric cancer development [28]. Alternatively, stomach-colonizing oral pathogens have recently emerged as modulating factors of gastric tumorigenesis. Among these, five oral microbial genera, including Streptococcus, Prevotella, Haemophilus, Veillonella, and Neisseria, the abundances of which were ranked high in the HOMD, accounted for a considerable proportion of the gastric microbiota. Collectively, these genera represent 17.6% and 11.6% of the gastric microbiome in Mexicans and Chinese populations, respectively [29].
Recently, Lei et al. [30] highlighted the spatially distinct correlation of H. pylori and stomach-colonizing oral pathogens with gastric cancer development. A total of 223 gastric tumoral tissues and their matched non-tumoral tissue samples were collected from three different sites of gastric tumors, including 76, 33, and 114 samples from the upper, middle, and lower third, respectively. Subsequent 16S rRNA metagenome sequencing revealed that the relative abundance of H. pylori was significantly higher in the lower-third of tumor tissues, whereas oral-origin bacteria, including Veillonella parvula, Streptococcus oralis, S. anginosus, and P. intermedia, were significantly enriched in the upper-third sites of tumor tissues. Therefore, identifying the specific oral-origin pathogens contributing to the development of gastric tumorigenesis could lead to a breakthrough in the diagnosis and intervention of H. pylori-independent gastric cancer.
Notably, Fu et al. [31] investigated the role of S. anginosus in the onset and development of gastric tumorigenesis, using an N-methyl-N-nitrosourea-induced gastric cancer mouse model, divided into three experimental groups: S. anginosus-infected, H. pylori-infected, and non-infected groups. The results indicated a higher tumor incidence rate in the S. anginosus-infected group (92%) than in the other two groups (67% and 31% in H. pylori- and non-infected groups, respectively) [31], underscoring the causative role of S. anginosus in promoting gastric tumorigenesis. Further investigation revealed that the downstream pathway underlying S. anginosus-mediated gastric tumorigenesis involves activation of the mitogen-activated protein kinase signaling, followed by S. anginosus colonization of the gastric mucosa via the interaction between S. anginosus surface protein Treponema pallidum membrane protein C (TMPC) [32] and gastric epithelial receptor Annexin 2 [31]. These findings suggest that targeting TMPC or Annexin 2 could be a therapeutic strategy for mitigating gastric cancer development.

Role of F. nucleatum in colorectal cancer pathogenesis

F. nucleatum overabundance was observed in both gastric and colorectal cancer tissues (Table 2), indicating that gastrointestinal tract-colonizing F. nucleatum is closely associated with gastrointestinal tumorigenesis. Yamamura et al. [33] conducted an interesting study to determine whether F. nucleatum is also distributed in extra-gastrointestinal cancer tissues, using various cancer tissues collected from patients with esophageal, gastric, colorectal, pancreatic, and liver cancers. The results demonstrated that F. nucleatum was exclusively colonized in the gastrointestinal tumor tissues (esophageal, gastric, and colorectal cancer) and not extra-gastrointestinal tumor tissues (pancreatic and liver cancer), highlighting its specific association with gastrointestinal cancers.
Given the increased abundance of F. nucleatum in gastrointestinal cancer tumors, its causative role in the onset and progression of gastrointestinal cancer has been extensively investigated. For instance, experiments were conducted using ApcMin/+ mice, a specific animal model useful for colonic tumorigenesis [34], in which the mice were fed F. nucleatum or Streptococcus to analyze the number of colonic tumors and inflammatory responses using dissected colon tissues [35]. Compared with Streptococcus-fed ApcMin/+ mice, F. nucleatum-fed ApcMin/+ mice exhibited an increased number of colonic tumor tissues and upregulated expression of proinflammatory genes in colonic tumor tissues. Furthermore, F. nucleatum was enriched in colonic tumor tissues but not in the adjacent normal tissues of F. nucleatum-fed ApcMin/+ mice. These results suggest that therapeutic interventions targeting F. nucleatum can be used to prevent or treat colonic cancer.
To delineate the molecular mechanism underlying F. nucleatum-mediated colorectal tumorigenesis, Brennan et al. [36] focused on pro-inflammatory gene expressions in ApcMin/+ mice neonatally inoculated with F. nucleatum, which revealed significant upregulation of interleukin-17 (Il-17) expression in both the colonic epithelium and colonic lamina propria. The immune-modulating roles of F. nucleatum were further examined ex vivo by Kim et al. [37] using tumor-infiltrating lymphocytes to elucidate the role of F. nucleatum. Lymphocytes isolated from tumors were infected with F. nucleatum, resulting in higher FoxP3+ regulatory T cells and lower CD3+, CD8+, and CD45RO+ T lymphocytes, which are associated with poor prognosis [38] and low survival rates [39].
In particular, compared with healthy individuals, cancer patients are more susceptible to F. nucleatum infection due to the anti-tumor immune evasion, which is accompanied by the upregulation of immune checkpoint inhibitory receptors, including programmed cell death-1 (PD-1) and T cell immunoglobulin and ITIM domains (TIGIT). The enhanced interaction between the PD-1 receptor of T lymphocytes and programmed death-ligand 1 in colorectal tumors suppresses the efficacy of cancer immunotherapy [40]. Additionally, the binding of the TIGIT receptor on T cells to the Fap2 adhesin protein of F. nucleatum inactivates T cells, allowing cancer cells to evade the immune system [41,42].

Reassessment of association between colorectal cancer and F. nucleatum at a subspecies-level

Recent studies have highlighted that F. nucleatum subsp. animalis (Fna) is exclusively colonized in inflammatory environmental niches, such as odontogenic abscesses [43] and intra-colorectal tumor [44], whereas other F. nucleatum subspecies, including F. nucleatum subsp. polymorphum (Fnp), F. nucleatum subsp. nucleatum (Fnn), and F. nucleatum subsp. vincentii, did not. Zepeda-Rivera et al. [44] further classified Fna into two distinct clades, C1 and C2, and observed that Fna C2 was more prevalent in the colorectal cancer niche, which was validated using Fna C2-treated ApcMin/+ mice, which showed a higher number of colonic adenomas than those of Fna C1-treated mice. An in-depth understanding of taxonomic profiles and their functions in relation to F. nucleatum requires a reassessment of previous experimental findings. Russo et al. [21] performed quantitative polymerase chain reaction (qPCR) using a forward primer (5'-CTTAGGAATGAGACAGAGATG-3') to compare the amounts of the amplified 16S rRNA fragment of F. nucleatum in the oral cavity of colorectal cancer patients and healthy controls. Their results showed no significant difference in F. nucleatum abundance between the two groups. However, we aligned sequences between the primer and the 16S rRNA of the four F. nucleatum subspecies and observed that the primer amplified the 16S rRNA of Fna, Fnp, and Fnn, except for Fna, due to the presence of mismatched sequences between the primer and the 16S rRNA of Fna [21]. This suggests the need for more additional qPCR experiments targeting each F. nucleatum subspecies-specific primer to re-evaluate the differences in abundance of F. nucleatum at the subspecies level between the experimental groups.

ORAL MICROBIOTA AND EXTRA-GASTROINTESTINAL DISEASES

Oral microbial alterations associated with extra-gastrointestinal diseases

Park et al. [6] reported oral microbial composition is closely associated with extra-gastrointestinal diseases, including atherosclerotic cardiovascular disease, diabetes, and Alzheimer’s disease. In this review, we focused on lung cancer, a malignant disease with low survival rates. Most lung cancer patients are often diagnosed at an advanced stage because of the lack of early-stage symptoms [45]. A comparative study of salivary microbial taxa between 46 lung cancer patients and 45 healthy controls revealed an abundance of Rothia, Granulicatella, Parvimonas, Abiotrophia, and Eubacterium in lung cancer patients [46]. Furthermore, the relative abundance of Capnocytophaga, Selenomonas, and Veillonella in salivary samples was substantially higher in lung cancer patients (n=20) than in healthy controls (n=10), and all participants were heavy smokers for >10 years [47]. In particular, Veillonella dispar was identified as a potential biomarker for smoking-associated oral microbial disturbances contributing to lung cancer development, as demonstrated in two different studies: V. dispar was highly abundant in the buccal swab samples from 55 heavy smokers than in 50 non-smokers [48], and V. dispar was elevated in salivary samples obtained from three lung cancer patients (smokers) than in five healthy individuals (non-smokers) [49]. Moreover, oral microbial alterations were observed in non-smokers, with an increased abundance of salivary Sphingomonas and Blastomonas in 75 lung cancer patients than in 172 healthy controls [50]. Taken together, these findings suggest that lung cancer-associated oral microbial compositions can be used for the early and non-invasive detection of lung cancer, potentially lowering mortality rates (Table 4).

Enrichment of oral microbiota in lung cancer tissues

Lungs were previously believed to be sterile because of the inability to culture microbes from lower airway samples using traditional microbial culture systems [51]. However, advancements in metagenomic sequencing have contributed to the understanding that the lungs are commonly colonized by diverse microorganisms, including oral microbiota such as Prevotella, Streptococcus, Veillonella, and Neisseria [52]. Changes in the lung microbiota are closely associated with the deterioration of pulmonary disorders, such as lung cancer (Table 5). Lee et al. [53] showed that the abundance of oral microbiota, including Veillonella, Megasphaera, Atopobium, and Selenomonas, was significantly higher in the bronchoalveolar fluids of lung cancer patients (n=20) than in those with benign mass-like lesions (n=8). Further investigations based on the smoking history of 20 lung cancer patients showed that Streptococcus and Porphyromonas were significantly elevated in 12 ever-smokers than in 8 never-smokers [53]. Another study reported an overabundance of Streptococcus and Veillonella in the biopsy samples of malignant lung lesions than in benign pulmonary nodules [54]. Microbial elevation is mechanistically associated with the activation of phosphoinositide 3-kinase (PI3K) and extracellular signal–regulated kinase (ERK) signaling [54], an early pathogenetic event in lung cancer development that regulates cell proliferation, survival, and tissue invasion [55]. Taken together, these findings suggest that the ectopic colonization of oral microbiota in the lungs plays a key role in the onset and progression of lung cancer, which is associated with disturbances in host molecular signaling pathways related to cellular growth and proliferation.

Modulating effects of an oral pathogen on lung tumorigenesis

Role of Veillonella parvula in lung cancer pathogenesis

As shown in Table 5, two independent research groups have identified Veillonella as a malignant lung tumor-enriched taxon, suggesting its potential as a biomarker for diagnosing and treating lung cancer. To validate this, Tsay et al. [56] investigated whether V. parvula played a role in the development and progression of lung cancer using lung cancer mouse model (genotypically manipulated as KRASLSL-G12D;p53flox/flox, KP mice). The results demonstrated that KP mice inoculated with V. parvula exhibited increased tumor growth and upregulated PI3K/ERK pathways, whereas wild-type mice inoculated with V. parvula showed no such changes [56,57]. This could be attributed to the exaggerated inflammatory responses, marked by increased recruitment of T helper 17 cells, leading to elevated Il-17 production and PD-1 receptor expression [56]. Jin et al. [58] demonstrated that eliminating lung-resident bacteria from KP mice reduced tumor size and number by lowering Il-17-producing T cells, indicating that therapeutic interventions targeting both oral microbiota dysbiosis and Il-17 can ameliorate the risk of lung cancer development.
Zeng et al. [59] developed a different lung cancer mouse model by administrating Lewis lung carcinoma cells (LLC) into either the trachea or subcutaneous tissues of C57 BL/6J mice, followed by inoculation with V. parvula. The results showed that V. parvula promoted tumorigenesis induced by both LLC-intratracheal instillation and LLC-subcutaneous graft, as reflected by the increased number and size of tumors in LLC + V. parvula than in the LLC only model. Mechanistically, V. parvula alters the immune microenvironment by inhibiting the recruitment of tumor-infiltrating T lymphocytes and affecting the distribution of CD3+ and CD4+ T lymphocytes in the peripheral immune environment [59].

CONCLUDING REMARKS

This review highlights the association between oral microbiota and non-oral diseases, including gastric, colorectal, esophageal, and lung cancers. Cancer-associated microbial taxonomic alteration in the oral cavity can serve as early and non-invasive biomarkers for assessing disease. The enrichment of oral microbiota in malignant tissues emphasizes the role of oral microbiota in systemic disease spread via the esophagus, larynx, and bloodstream (Fig. 1) [60,61]. The specific oral microbiota can play a causative role in the development of diverse types of cancers by modulating host immune and inflammatory responses. In particular, regulation of IL-17 production is crucial for a balanced adjustment between its protective roles against extrinsic pathogens [62] and its detrimental roles in the development of various types of cancers [63,64].

Notes

Availability of Data and Material

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

Conflicts of Interest

The authors have no financial conflicts of interest.

Funding Statement

This work was supported in part by the Korea Biobank Network Program run by the Korea Disease Control and Prevention Agency (KBN4-A04-03).

Authors’ Contribution

Conceptualization: Do-Young Park. Data curation: Do-Young Park. Formal analysis: Do-Young Park. Funding acquisition: Inseong Hwang. Investigation: Young-Youn Kim. Methodology: Do-Young Park. Project administration: Do-Young Park. Resources: Chang Kee Kim. Software: Do-Young Park. Supervision: Do-Young Park. Validation: Do-Young Park. Visualization: Do-Young Park. Writing—original draft: Do-Young Park. Writing—review & editing: Do-Young Park, Jeong-Hoo Lee, Jiyoung Hwang, Ju-Yeong Hwang. Approval of final manuscript: all authors.

Acknowledgements

None

Fig. 1.
Malignant tumor-enriched oral microbiota according to different kinds of cancers.
kjhugr-2024-0051f1.jpg
Table 1.
Gastrointestinal disease-associated oral microbial alterations
Study Participants Samples Methods Oral microbes upregulated in the oral cavity of patients with gastrointestinal diseases Location
Sun et al. [20] 37 gastric cancer patients Saliva or plaque 16S rRNA metagenome Veillonella, Prevotella, Aggregatibacter, Megasphaera China
13 healthy controls
Russo et al. [21] 10 colorectal cancer patients Saliva 16S rRNA metagenome Actinobacteria, Saccharibacteria, Proteobacteria, Fusobacteria, Firmicutes, Bacteroidetes Italy
10 healthy controls
Table 2.
The enrichment of oral microbiota in gastrointestinal cancers
Study Participants Samples Methods Oral microbes enriched in gastrointestinal cancer tumors Location
Coker et al. [24] 20 gastric cancer patients Gastric mucosa 16S rRNA metagenome S. anginosus, P. micra, D. pneumosintes, S. exigua, F. nucleatum, P. intermedia, C. morbi, P. stomatis China
17 intestinal metaplasia patients
23 atrophic gastritis patients
21 superficial gastritis patients
Tran et al. [23] 43 colorectal cancer patients Colorectal tumor or polyp tissues 16S rRNA metagenome Gemella, Peptostreptococcus, F. nucleatum, Leptotrichia, S. sputigena, C. rectus Vietnam
25 colorectal non-cancerous polyp patients
Loftus et al. [22] 74 colorectal cancer patients Fecal samples Whole genome shotgun metagenome F. nucleatum, P. stomatis, G. morbillorum, P. micra Japan
178 healthy controls
Table 3.
Salivary and gastric microbiota in gastric disease patients and healthy controls
Study Participants Samples Microbiota significantly enriched in gastric disease groups Location
You et al. [25] 41 gastric cancer patients Saliva Prevotella melaninogenica, Gemella haemolysans Korea
33 high-grade dysplasia patients Gastric juice Schaalia odontolytica, Rothia mucilaginosa, P. stomatis, Streptococcus salivarius, Helicobacter pylori, Streptococcus sanguinis, G. haemolysans, Streptococcus australis, Streptococcus cristatus, Haemophilus parainfluenza, Prophyromonas pasteri
58 low-grade dysplasia patients
9 healthy controls
Table 4.
Lung cancer-associated oral microbial alterations
Study Participants (No. of smokers) Samples Methods Upregulated oral microbes of lung cancer patients Location
Jiang et al. [46] 46 lung cancer patients (24) Saliva 16S rRNA metagenome Rothia, Granulicatella, Parvimonas, Abiotrophia, Eubacterium China
45 healthy controls (13)
Yan et al. [47] 20 lung cancer patients (20) Saliva 16S rRNA metagenome Capnocytophaga, Selenomonas, Veillonella China
10 healthy controls (10)
Roy et al. [49] 5 lung cancer patients (3) Saliva 16S rRNA metagenome V. dispar, R. mucilaginosa, P. melaninogenica, Prevotella pallens, Prevotella copri, H. parainfluenzae, Neisseria bacilliformis, Aggregatibacter segnis India
5 healthy controls (0)
Yang et al. [50] 75 lung cancer patients (0) Saliva 16S rRNA metagenome Sphingomonas, Blastomonas China
172 healthy controls (0)
Table 5.
Enrichment of oral microbiota in lung cancers
Study Participants (No. of smokers) Samples Methods Oral microbes enriched in lung cancer patients Location
Lee et al. [53] 8 benign mass-like lesion patients (5) Bronchoalveolar fluid 16S rRNA metagenome Veillonella, Megasphaera, Atopobium, Selenomonas Korea
39 lung cancer patients (12)
Tsay et al. [54] 36 benign pulmonary nodule patients (11) Lower airway 16S rRNA metagenome Streptococcus, Veillonella United States
36 benign pulmonary nodule patients (11)

REFERENCES

1. Marsh PD, Do T, Beighton D, Devine DA. Influence of saliva on the oral microbiota. Periodontol 2000 2016;70:80–92.
crossref pmid
2. Sulyanto RM, Thompson ZA, Beall CJ, Leys EJ, Griffen AL. The predominant oral microbiota is acquired early in an organized pattern. Sci Rep 2019;9:10550.
crossref pmid pmc pdf
3. Michaud DS, Izard J, Rubin Z, et al. Lifestyle, dietary factors, and antibody levels to oral bacteria in cancer-free participants of a European cohort study. Cancer Causes Control 2013;24:1901–1909.
crossref pmid pmc pdf
4. Thomas AM, Gleber-Netto FO, Fernandes GR, et al. Alcohol and tobacco consumption affects bacterial richness in oral cavity mucosa biofilms. BMC Microbiol 2014;14:250.
crossref pmid pmc pdf
5. Mason MR, Nagaraja HN, Camerlengo T, Joshi V, Kumar PS. Deep sequencing identifies ethnicity-specific bacterial signatures in the oral microbiome. PLoS One 2013;8:e77287.
crossref pmid pmc
6. Park DY, Park JY, Lee D, Hwang I, Kim HS. Leaky gum: the revisited origin of systemic diseases. Cells 2022;11:1079.
crossref pmid pmc
7. Griffen AL, Beall CJ, Campbell JH, et al. Distinct and complex bacterial profiles in human periodontitis and health revealed by 16S pyrosequencing. ISME J 2012;6:1176–1185.
crossref pmid pmc pdf
8. Abusleme L, Dupuy AK, Dutzan N, et al. The subgingival microbiome in health and periodontitis and its relationship with community biomass and inflammation. ISME J 2013;7:1016–1025.
crossref pmid pmc pdf
9. Kirst ME, Li EC, Alfant B, et al. Dysbiosis and alterations in predicted functions of the subgingival microbiome in chronic periodontitis. Appl Environ Microbiol 2015;81:783–793.
crossref pmid pmc pdf
10. Hong BY, Furtado Araujo MV, Strausbaugh LD, Terzi E, Ioannidou E, Diaz PI. Microbiome profiles in periodontitis in relation to host and disease characteristics. PLoS One 2015;10:e0127077.
crossref pmid pmc
11. Nibali L, Sousa V, Davrandi M, et al. Differences in the periodontal microbiome of successfully treated and persistent aggressive periodontitis. J Clin Periodontol 2020;47:980–990.
crossref pmid pdf
12. Genco RJ, LaMonte MJ, McSkimming DI, et al. The subgingival microbiome relationship to periodontal disease in older women. J Dent Res 2019;98:975–984.
crossref pmid pmc pdf
13. Papapanou PN, Park H, Cheng B, et al. Subgingival microbiome and clinical periodontal status in an elderly cohort: the WHICAP ancillary study of oral health. J Periodontol 2020;91(Suppl 1): S56–S67.
crossref pmid pmc pdf
14. Ghensi P, Manghi P, Zolfo M, et al. Strong oral plaque microbiome signatures for dental implant diseases identified by strain-resolution metagenomics. NPJ Biofilms Microbiomes 2020;6:47.
crossref pmid pmc pdf
15. Ikeda E, Shiba T, Ikeda Y, et al. Japanese subgingival microbiota in health vs disease and their roles in predicted functions associated with periodontitis. Odontology 2020;108:280–291.
crossref pmid pdf
16. Diao J, Yuan C, Tong P, Ma Z, Sun X, Zheng S. Potential roles of the free salivary microbiome dysbiosis in periodontal diseases. Front Cell Infect Microbiol 2021;11:711282.
crossref pmid pmc
17. Struzycka I. The oral microbiome in dental caries. Pol J Microbiol 2014;63:127–135.
crossref pmid
18. Yang CY, Yeh YM, Yu HY, et al. Oral microbiota community dynamics associated with oral squamous cell carcinoma staging. Front Microbiol 2018;9:862.
crossref pmid pmc
19. Olsen I, Yamazaki K. Can oral bacteria affect the microbiome of the gut? J Oral Microbiol 2019;11:1586422.
crossref pmid pmc pdf
20. Sun JH, Li XL, Yin J, Li YH, Hou BX, Zhang Z. A screening method for gastric cancer by oral microbiome detection. Oncol Rep 2018;39:2217–2224.
crossref pmid
21. Russo E, Bacci G, Chiellini C, et al. Preliminary comparison of oral and intestinal human microbiota in patients with colorectal cancer: a pilot study. Front Microbiol 2018;8:2699.
crossref pmid pmc
22. Loftus M, Hassouneh SA, Yooseph S. Bacterial community structure alterations within the colorectal cancer gut microbiome. BMC Microbiol 2021;21:98.
crossref pmid pmc pdf
23. Tran HNH, Thu TNH, Nguyen PH, et al. Tumour microbiomes and Fusobacterium genomics in Vietnamese colorectal cancer patients. NPJ Biofilms Microbiomes 2022;8:87.
crossref pmid pmc pdf
24. Coker OO, Dai Z, Nie Y, et al. Mucosal microbiome dysbiosis in gastric carcinogenesis. Gut 2018;67:1024–1032.
crossref pmid pmc
25. You HS, Park JY, Seo H, Kim BJ, Kim JG. Increasing correlation between oral and gastric microbiota during gastric carcinogenesis. Korean J Intern Med 2024;39:590–602.
crossref pmid pmc pdf
26. Chen H, Jiang X, Zhu F, et al. Characteristics of the oral and gastric microbiome in patients with early-stage intramucosal esophageal squamous cell carcinoma. BMC Microbiol 2024;24:88.
crossref pmid pmc pdf
27. Uemura N, Okamoto S, Yamamoto S, et al. Helicobacter pylori infection and the development of gastric cancer. N Engl J Med 2001;345:784–789.
crossref pmid
28. Ferreira RM, Pereira-Marques J, Pinto-Ribeiro I, et al. Gastric microbial community profiling reveals a dysbiotic cancer-associated microbiota. Gut 2018;67:226–236.
crossref pmid pmc
29. Yu G, Torres J, Hu N, et al. Molecular characterization of the human stomach microbiota in gastric cancer patients. Front Cell Infect Microbiol 2017;7:302.
crossref pmid pmc
30. Lei L, Zhao LY, Cheng R, et al. Distinct oral-associated gastric microbiota and Helicobacter pylori communities for spatial microbial heterogeneity in gastric cancer. mSystems 2024;9:e0008924.
crossref pmid pmc pdf
31. Fu K, Cheung AHK, Wong CC, et al. Streptococcus anginosus promotes gastric inflammation, atrophy, and tumorigenesis in mice. Cell 2024;187:882–896; e17.
crossref pmid
32. Deka RK, Brautigam CA, Yang XF, et al. The PnrA (Tp0319; TmpC) lipoprotein represents a new family of bacterial purine nucleoside receptor encoded within an ATP-binding cassette (ABC)-like operon in Treponema pallidum. J Biol Chem 2006;281:8072–8081.
crossref pmid
33. Yamamura K, Baba Y, Miyake K, et al. Fusobacterium nucleatum in gastroenterological cancer: evaluation of measurement methods using quantitative polymerase chain reaction and a literature review. Oncol Lett 2017;14:6373–6378.
crossref pmid pmc
34. Ren J, Sui H, Fang F, Li Q, Li B. The application of ApcMin/+ mouse model in colorectal tumor researches. J Cancer Res Clin Oncol 2019;145:1111–1122.
crossref pmid pdf
35. Kostic AD, Chun E, Robertson L, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 2013;14:207–215.
crossref pmid pmc
36. Brennan CA, Clay SL, Lavoie SL, et al. Fusobacterium nucleatum drives a pro-inflammatory intestinal microenvironment through metabolite receptor-dependent modulation of IL-17 expression. Gut Microbes 2021;13:1987780.
crossref pmid pmc
37. Kim HS, Kim CG, Kim WK, et al. Fusobacterium nucleatum induces a tumor microenvironment with diminished adaptive immunity against colorectal cancers. Front Cell Infect Microbiol 2023;13:1101291.
crossref pmid pmc
38. Saito T, Nishikawa H, Wada H, et al. Two FOXP3+CD4+ T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med 2016;22:679–684.
crossref pmid pdf
39. Nosho K, Baba Y, Tanaka N, et al. Tumour-infiltrating T-cell subsets, molecular changes in colorectal cancer, and prognosis: cohort study and literature review. J Pathol 2010;222:350–366.
pmid pmc
40. Masugi Y, Nishihara R, Yang J, et al. Tumour CD274 (PD-L1) expression and T cells in colorectal cancer. Gut 2017;66:1463–1473.
crossref pmid pmc
41. Gur C, Ibrahim Y, Isaacson B, et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity 2015;42:344–355.
crossref pmid pmc
42. Coppenhagen-Glazer S, Sol A, Abed J, et al. Fap2 of Fusobacterium nucleatum is a galactose-inhibitable adhesin involved in coaggregation, cell adhesion, and preterm birth. Infect Immun 2015;83:1104–1113.
crossref pmid pmc pdf
43. Krieger M, AbdelRahman YM, Choi D, et al. Stratification of Fusobacterium nucleatum by local health status in the oral cavity defines its subspecies disease association. Cell Host Microbe 2024;32:479–488; e4.
crossref pmid
44. Zepeda-Rivera M, Minot SS, Bouzek H, et al. A distinct Fusobacterium nucleatum clade dominates the colorectal cancer niche. Nature 2024;628:424–432.
crossref pmid pmc pdf
45. Liu M, Liu X, Suo P, et al. The contribution of hereditary cancer-related germline mutations to lung cancer susceptibility. Transl Lung Cancer Res 2020;9:646–658.
crossref pmid pmc
46. Jiang X, Liu S, Zhang Y, et al. Free-flow isoelectric focusing for comprehensive separation and analysis of human salivary microbiome for lung cancer. Anal Chem 2020;92:12017–12025.
crossref pmid
47. Yan X, Yang M, Liu J, et al. Discovery and validation of potential bacterial biomarkers for lung cancer. Am J Cancer Res 2015;5:3111–3122.
pmid pmc
48. Al Bataineh MT, Dash NR, Elkhazendar M, et al. Revealing oral microbiota composition and functionality associated with heavy cigarette smoking. J Transl Med 2020;18:421.
pmid pmc
49. Roy P, Sarma A, Kataki AC, Rai AK, Chattopadhyay I. Salivary microbial dysbiosis may predict lung adenocarcinoma: a pilot study. Indian J Pathol Microbiol 2022;65:123–128.
pmid
50. Yang J, Mu X, Wang Y, et al. Dysbiosis of the salivary microbiome is associated with non-smoking female lung cancer and correlated with immunocytochemistry markers. Front Oncol 2018;8:520.
crossref pmid pmc
51. Marsland BJ, Yadava K, Nicod LP. The airway microbiome and disease. Chest 2013;144:632–637.
crossref pmid
52. Dickson RP, Erb-Downward JR, Martinez FJ, Huffnagle GB. The microbiome and the respiratory tract. Annu Rev Physiol 2016;78:481–504.
crossref pmid pmc
53. Lee SH, Sung JY, Yong D, et al. Characterization of microbiome in bronchoalveolar lavage fluid of patients with lung cancer comparing with benign mass like lesions. Lung Cancer 2016;102:89–95.
crossref pmid
54. Tsay JJ, Wu BG, Badri MH, et al. Airway microbiota is associated with upregulation of the PI3K pathway in lung cancer. Am J Respir Crit Care Med 2018;198:1188–1198.
crossref pmid pmc
55. Gustafson AM, Soldi R, Anderlind C, et al. Airway PI3K pathway activation is an early and reversible event in lung cancer development. Sci Transl Med 2010;2:26.ra25.
crossref pmid pmc
56. Tsay JJ, Wu BG, Sulaiman I, et al. Lower airway dysbiosis affects lung cancer progression. Cancer Discov 2021;11:293–307.
pmid
57. Riccardo F, Arigoni M, Buson G, et al. Characterization of a genetic mouse model of lung cancer: a promise to identify non-small cell lung cancer therapeutic targets and biomarkers. BMC Genomics 2014;15(Suppl 3): S1.
crossref pmid pmc pdf
58. Jin C, Lagoudas GK, Zhao C, et al. Commensal microbiota promote lung cancer development via γδ T cells. Cell 2019;176:998–1013.e16.
crossref pmid pmc
59. Zeng W, Wang Y, Wang Z, et al. Veillonella parvula promotes the proliferation of lung adenocarcinoma through the nucleotide oligomerization domain 2/cellular communication network factor 4/nuclear factor kappa B pathway. Discov Oncol 2023;14:129.
crossref pmid pmc pdf
60. Hernández-Cabanyero C, Vonaesch P. Ectopic colonization by oral bacteria as an emerging theme in health and disease. FEMS Microbiol Rev 2024;48:fuae012.
pmid pmc
61. Elghannam MT, Hassanien MH, Ameen YA, et al. Oral microbiome dysbiosis and gastrointestinal diseases: a narrative review. Egypt Liver Journal 2024;14:32.
crossref pdf
62. Mills KHG. IL-17 and IL-17-producing cells in protection versus pathology. Nat Rev Immunol 2023;23:38–54.
crossref pmid pmc pdf
63. Papotto PH, Ribot JC, Silva-Santos B. IL-17+ γδ T cells as kick-starters of inflammation. Nat Immunol 2017;18:604–611.
crossref pmid pdf
64. Wu D, Wu P, Huang Q, Liu Y, Ye J, Huang J. Interleukin-17: a promoter in colorectal cancer progression. Clin Dev Immunol 2013;2013:436307.
crossref pmid pmc pdf


Editorial Office
Lotte Gold Rose II Room 917, 31 Seolleung-ro 86-gil, Gangnam-gu, Seoul 06193, Korea
Tel: +82-2-717-5543    Fax: +82-2-565-9947    E-mail: kjhugr@kams.or.kr                

Copyright © 2025 by Korean College of Helicobacter and Upper Gastrointestinal Research.

Developed in M2PI

Close layer
prev next