Skip to main content

Understanding the characteristics of the host genome and microbiome interaction in oral squamous cell carcinoma: a narrative review

Abstract

Background

Oral health status is directly associated with microbes present within it. The abundance of microbes at the OSCC site is more than at its control site, representing its possible role in the progression of OSCC development. Dysbiosis of oral microbiota could be a crucial etiological risk factor in the elevation of OSCC. This study aimed to analyze and assess: a) positive regulator microbes of oral cancer and their abundance at the cancer site, b) pathways involved in positive regulator microbes, and c) identification of the most virulent oral oncogenic microbe.

Main body

It is obtained from several studies that microbes belonging to Prevotella, Fusobacterium, Alloprevotella, Capnocytophaga, Porphyromonas, Campylobacter, and Aggregatibacter are detected to be more in number contrast to healthy sites. Fusobacterium nucleatum, Porphyromonas gingivalis, and Candida albicans show molecular pathways linked with OSCC development. Genes encoding for virulent factors like FimA, Gingipains, lipopolysaccharide (P. gingivalis), FadA, Fap2 (F. nucleatum), and zymosan (C. Albicans) are directly involved in elevating oral cancer.

Conclusion

Mostly, the genes that are involved in promoting oral cancer are the genes that generally encode cell wall proteins. The cell wall proteins that is FadA, Fap, and FimA interact with the host's cell and hamper the normal regulation pathway, which leads to activation of cell proliferating pathways, down-regulates apoptotic pathways, cytoskeleton rearrangement, and upregulates the cell cycle checkpoint regulators; as a result, progression of oral cancer occurs.

1 Background

The buccal cavity is the most diverse reservoir of microbes comprehending symbiotic, commensal, and pathogenic organisms. The microbes in the buccal cavity/oral cavity determine the fitness of oral health. Over time with the progress of techniques, it has been evaluated that about 600 species encompass the oral ecological community. Gerald et al. [1].

The oral cavity is predominantly enriched by Proteobacteria and Firmicutes, which is about 19.26% and 53.83%, respectively. It is espied that there is a shift of oral microbiome in preoral cancer and oral cancer patients compared with normal individuals, and microflora changes metabolic pathway to influence oral health. Zixuan et al. [2].

OSCC—Oral squamous cell carcinoma, is one of the most common types of oral cancer, approximately ninety percent of total oral cancer Tendon et al. [3]. Wang et al. [4] The reoccurrence rate in OSCC ranges from 32.7 to 44.9% [4]. Mathur et al. [5] consumption of alcohol, poor oral hygiene, tobacco smoking, unhealthy diet, and microbial infection are the etiological factor that boosts the risk of OSCC [5]. Some unknown etiological factors are responsible for OSCC development as it has been seen that persons who have never drank or smoked also face OSCC Kruse et al. [6]. It is obtained from the data that genes of OSCC-associated bacterium have a potential role in bacterial chemotaxis, flagellar assembly, and lipopolysaccharide (LPS) synthesis are concentrated in the tumor site [7].

Streptococcus spp. is found to be significantly less in number on the surface of oral squamous cell carcinoma-OSCC lesions. The tumor site of the oral cavity is hiked up with Campylobacter, Fusobacterium, Capnocytophaga, Prevotella, and Peptostreptococcus. Su et al. [8]. Carnobacteriaceae, Actinomycetaceae, Micrococcaceae, and Streptococcaceae are low in number in OSCC patients. Zhang et al. [9].

Periodontitis patients are more prone to develop oral squamous cell carcinoma. Yufei et al. [10]. Considering the carcinogenic property, a study was conducted to determine the relationship between OSCC microflora and periodontitis. The positive and negative regulator microbes of periodontitis were tested against the HSC-3 cell line. Positive regulator microbes of periodontitis express a high amount of IL-6, cyclin-D1, IL-8, and MMP-9 and show more proliferative activity than control and negative regulator microbes of periodontitis. Xiaoyu Hu et al. [11]. This review analyzes the oral microbiota of OSCC patients and the pathway associated with the OSCC advancement to establish the link between the particular positive regulator of OSCC bacteria and molecular mechanisms. These positive regulator microorganisms might be employed as biomarkers to identify and prevent OSCC in its early stages.

2 Pathway used by the positive regulator of oral cancer

2.1 Porphyromonas gingivalis

Porphyromonas gingivalis resides in the subgingival site and is a Gram-negative anaerobic bacteria involved in promoting anti-apoptotic pathway by increasing the expression of anti-apoptotic genes and by blocking the pro-apoptotic genes, as represented in Fig. 1. Porphyromonas gingivalis activates PI3/Akt pathway, which enhances cancer. Mao et al. [12].

Fig. 1
figure 1

Porphyromonas gingivalis carcinogenic signaling pathway

Sandros et al. [13] Some essential virulent factors like endotoxin, gingipains, and fimbriae of Porphyromonas gingivalis provoke secretion of various chemokines and tumor necrosis factor-alpha (TNF-α). Yilmaz et al. [14] FimA fimbriae found on the surface of Porphyromonas gingivalis, which have a role in rearranging cytoskeleton by interacting with beta 1 integrin on the host cell membrane and cause invasion of the cell. Amano et al. [15] Fimbriae also cause inflammation by inducing the host cell secretion of IL-1, IL-6, IL-8, and TNF-α. Inaba et al. [16] Gingipains convert ProMMP9 to activated MMP9, and MMP9 makes carcinoma cells migrate and show invasiveness property by degrading the basement membrane.

Tang et al. [17] Porphyromonas gingivalis alters the pathway, which is linked with the regulation of cell cycles like cyclins, PI3K, and p53. LPS (lipopolysaccharide) of P. gingivalis is involved in the downregulation of p53. Groeger et al. and Kuboniwa et al. [18, 19] Porphyromonas gingivalis activate NF-κB and MAPK pathways which are associated with cell proliferation.

Metabolites like “Butyric acid” (Chang et al. [20]) and “Acetaldehyde” (Olsen et al. [21]) are synthesized by PG, whose effect can promote cancer by causing mutation in the nucleotides. Kurita-Ochiai et al. [22] Butyric acid also provokes apoptosis of T- cell and B-cells in the absence of P53.

Gabrilovich et al. [23] Myeloid-derived suppressor cells are linked with metastasis and found abundantly at the OSCC site, inhibiting the activation of T cells. PG induces the growth of myeloid-derived suppressor cells at the OSCC site through cytokine and interleukins-mediated pathway.

Nucleoside diphosphate kinase is secreted by P. gingivalis, which blocks the ATP-dependent pathway via purinergic receptor P2X7 [23].

Porphyromonas gingivalis adhere to the host cell membrane through integrin-mediated receptor binding. It activates Akt/ PI3K pathway and promotes the cell proliferation process. It upregulates the anti-apoptosis gene expression and down-regulates pro-apoptotic gene expression.

2.2 Fusobacterium nucleatum

Peyret-Lacombe et al. [24] F. nucleatum is a Gram-negative anaerobic bacteria involved in producing various chemokines like IL-6, IL8, IL1β, IL6, and IL1α interact with oral epithelial cell and promotes carcinogenesis by the 'START' mediated pathway as depleted in Fig. 2. START, in turn, inhibits the expression of apoptotic genes and upregulates the cell survival genes. F. nucleatum also interacts with toll-like receptors present on the oral epithelial cell, and its entry is straightforward for the invasion process. Like PG, F. nucleatum is found copiously at the OSCC site than at the control site [25].

Fig. 2
figure 2

Carcinogenic pathway associated with Fusobacterium nucleatum

Fardini et al. [26] With the help of FadA, the adhesion molecule, F. nucleatum, binds with the host cell, disrupts the host's membrane integrity by disrupting its tight junction proteins and is also able to invade cells like colonic and placental epithelial cells, keratinocytes, T-cells and macrophages by expressing Fap2 and FadA Han et al. [27]. Abed et al. [28] F. nucleatum attaches to the host cell through an adhesion molecule that is Fap2, which binds with the oligonucleotide that is Gal-GalNAc of the host cell. Coppenhagen-Glazer et al. [29] Fap2 is also associated with oral biofilm formation.

F. nucleatum activates P38, which activates a series of proteins like matrix metalloproteinase-9 and MMP-13 and heat shock protein-27 (HSP-27). MMP-9 and MMP-13 are observed to cause metastasis [30]. F. nucleatum also promotes the JNK and TLR-2 signaling pathways [31].

LPS (lipopolysaccharide) at the cell wall of F. nucleatum upregulates the expression of genes encoding IL-8, which in turn causes inflammation and lesion made in the gingiva cell through cytokine-mediated damaging. [32, 33].

Yost et al. [34] Fusobacterium shows high RNA expression at tumor sites than at healthy sites. Mostly virulent expressing genes transcript more at OSCC site.

BD-1-beta defensin-1and BD-2-beta defensin-2 are antimicrobial peptides generally produced by epithelial cells of salivary glands whose expression is elevated when it detects the presence of LPS and TNF-alpha. [33,34,35].

F. nucleatum and Porphyromonas gingivalis act cooperatively to skip from the host's immune system and found that F. Nucleatum can increase the invasive potential of Porphyromonas gingivalis [36, 37]. Huang et al. [38] F. nucleatum produces ammonia from aspartate and glutamate and makes the environment neutral, which is suitable for P.gingivalis colonization as P. gingivalis is an acid intolerant microbe.

2.3 Candida albicans

Candida albicans is a saprophytic fungus generally found in the oral cavity, vagina, and GI tract (gastrointestinal) [39]. The presence of Candida in the deeper site of OSCC, which is about 74%, clarifies its role in OSCC occurrence [40]. Under certain specific conditions, Candida albicans become pathogenic and can cause acute and chronic infection (candidiasis), especially in immunosuppressive patients [41]. According to Chen et al. [42] , Candida albicans contains zymosan in the cell wall, is glycan in nature and can be easily recognized by pattern recognition receptors. OSCC expresses TLR (mostly TLR3 and TLR4). Activation of TLR leads to activation of adaptor molecules like MyD88, which ultimately triggers the NF-κB pathway that promotes tumor progression. Abdullah et al. [43] Leukoplakic lesions, a pre-cancerous state, are caused by Candida albicans. The ECE1 gene of Candida albicans encodes candidalysin toxin and triggers NF-κB and MAPK pathways. ECE1gene expression was found to be more during macrophage attack by the host cell and escapes from macrophage attack by switching its morphological form in the phagosome [44, 45] (Fig. 3).

Fig. 3
figure 3

Candida albicans carcinogenic signaling pathway

The ECE1 gene of Candida albicans encodes candidalysin protein, which is very toxic. It damages the host's cell membrane. As a result, cytokine secretion triggers APCs to differentiate and form a lesion, which ultimately turns into cancer. Candida albicans develops hyphae and secretes candidalysin when it is swallowed by macrophages, causing additional harm to the host immune cell. Candida albicans activate the NF-kB and MAPK pathways via TLR and adaptor protein activation.CA: Candida albicans (Table 1).

Table 1 Microbial profile at OSCC site against control site

3 Positive regulators and their interaction with the host at the OSCC site

It is observed that the deeper tissue is generally sterile, and microbes colonize on the surface layer of mucosal epithelium in healthy individuals. However, microbes can evade the mucosal barrier and seem to colonize themselves on the deeper tissue under certain circumstance. [46, 47]. Aerobic bacteria, obligate anaerobic bacteria, and facultative anaerobic species are abundantly present at the OSCC site, but facultative anaerobic species are more frequently found [48].

The expression of genes regulating bacterial mobility, LPS synthesis, bacterial chemotaxis, and flagella assembly was high and played an essential role at the OSCC site (7). Moghimi et al. [49] found an up-regulation of microbial genes like bspA, fadA, and interpain A, which correlates with CXCL10, MMP9, NCLN, and DIAPH1 genes.

4 Interaction of virulent microbes at the OSCC site

P. gingivalis and F. nucleatum employ different invasion methods. According to studies, P. gingivalis enters host cells using the endocytic route and lipid rafts; likewise, F. nucleatum enters host cells through numerous adhesins and a "zipper" mechanism [50].

FomA of F. nucleatum assists in bacterial co-aggregation and biofilm formation and is advantageous for bacterial invasion into host cells. It binds to the Fc portion of human IgG [51]. F. nucleatum assists P. gingivalis in invading host cells [52].

However, on the other hand, it has been found that P. gingivalis prevented F. nucleatum from invading oral epithelial cells by decreasing the production of the adhesion-related proteins FadA and FomA by secreting proteases [50].

Although P. gingivalis inhibits the invasion process of F. nucleatum, those bacterial mixtures abundantly found at the OSCC site are now a question of research. It may be due to the involvement of a particular strain of P. gingivalis that inhibit the F. nucleatum invasion process. More research is needed focusing on those areas.

A study report observed that C. albicans SN152's hyphal development is inhibited by F. nucleatum ATCC 23,726 in a contact-dependent manner [53]. Whether all strains of C. albicans inhibit F. nucleatum or not, which particular strain of Candida albicans is involved in promoting oral cancer needs to be studied. These are the research gap that needs to be focused on in future research.

5 Conclusion

Although many microbes are found to be more at the OSCC site than the control site, few positive regulator microbes like Porphyromonas gingivalis, Candida albicans, and Fusobacterium nucleatum are associated with host cell membrane damage and tissue invasion, which activates signaling pathways like MAPK and PI3K. Among these microbes, Porphyromonas gingivalis and Fusobacterium nucleatum are found to be more virulent oncogenic bacteria. Moreover, the interrelation between these microbes of different strains needs to be screened. The virulent strains of these microbes may lead to dysplasia of the oral cavity, which might further lead to various inflammatory pathways and later develops into cancer. Positive regulator microbes of OSCC activate pathways associated with cell growth, proliferation, survival, differentiation, and motility, increasing the risk of OSCC. These microbes suppress the pro-apoptotic gene expression and upregulate the anti-apoptotic gene expression; as a result, the survival chance of cells increases. The drug can be developed as a future therapeutic intervention strategy by targeting virulent expressing genes responsible for promoting cancer. The validation of the OSCC-associated microbiota as a biomarker might have significant implications for future oral cancer screening, early detection and treatment.

Availability of Data and Material

All the data were collected from Scopus, google scholar and PubMed for this study.

Abbreviations

OSCC:

Oral squamous cell carcinoma

LPS:

Lipopolysaccharide

IL:

Interleukin

MMP 9:

Matrix metallopeptidase 9

CXCL10:

C-X-C Motif chemokine ligand 10

TNF:

Tumor necrosis factor

References

  1. Gerald PC (2013) Oral microbiome homeostasis: the new frontier in oral care therapies. J Dent Oral Disord Ther 1(1):3. https://doi.org/10.15226/jdodt.2013.00105

    Article  Google Scholar 

  2. Li Z, Chen G, Wang P, Sun M, Zhao J, Li A, Sun Q (2021) Alterations of the oral microbiota profiles in chinese patient with oral cancer. Front Cell Infect Microbiol. https://doi.org/10.3389/fcimb.2021.780067

    Article  PubMed  PubMed Central  Google Scholar 

  3. Tandon P, Dadhich A, Saluja H, Bawane S, Sachdeva S (2017) The prevalence of squamous cell carcinoma in different sites of oral cavity at our Rural Health Care Centre in Loni, Maharashtra–a retrospective 10-year study. Contemp Oncol 21(2):178. https://doi.org/10.5114/wo.2017.68628

    Article  Google Scholar 

  4. Wang B, Zhang S, Yue K, Wang XD (2013) The recurrence and survival of oral squamous cell carcinoma: a report of 275 cases. Chin J Cancer 32(11):614. https://doi.org/10.5732/cjc.012.10219

    Article  PubMed  PubMed Central  Google Scholar 

  5. Mathur R, Singhavi HR, Malik A, Nair S, Chaturvedi P (2019) Role of poor oral hygiene in causation of oral cancer—a review of literature. Indian J Surg Oncol 10(1):184–195. https://doi.org/10.1007/s13193-018-0836-5

    Article  PubMed  Google Scholar 

  6. Kruse AL, Bredell M, Grätz KW (2010) Oral squamous cell carcinoma in non-smoking and non-drinking patients. Head Neck Oncol 2(1):1–3. https://doi.org/10.1186/1758-3284-2-24

    Article  Google Scholar 

  7. Al-Hebshi NN, Nasher AT, Maryoud MY, Homeida HE, Chen T, Idris AM, Johnson NW (2017) Inflammatory bacteriome featuring Fusobacterium nucleatum and Pseudomonas aeruginosa identified in association with oral squamous cell carcinoma. Sci Rep 7(1):1–10

    Article  CAS  Google Scholar 

  8. Su SC, Chang LC, Huang HD, Peng CY, Chuang CY, Chen YT, Lu MY, Chiu YW, Chen PY, Yang SF (2021) Oral microbial dysbiosis and its performance in predicting oral cancer. Carcinogenesis 42(1):127–135. https://doi.org/10.1093/carcin/bgaa062

    Article  CAS  PubMed  Google Scholar 

  9. Zhang L, Liu Y, Zheng HJ, Zhang CP (2020) The oral microbiota may have influence on oral cancer. Front Cell Infect Microbiol. https://doi.org/10.3389/fcimb.2019.00476

    Article  PubMed  PubMed Central  Google Scholar 

  10. Yao Y, Liu W, Li J, Zhou M, Qu C, Wang K (2021) MPI-based bioinformatic analysis and co-inhibitory therapy with mannose for oral squamous cell carcinoma. Med Oncol 38(9):1–11. https://doi.org/10.1007/s12032-021-01552-4

    Article  CAS  Google Scholar 

  11. Hu X, Shen X, Tian J (2021) The effects of periodontitis associated microbiota on the development of oral squamous cell carcinoma. Biochem Biophys Res Commun 576:80–85. https://doi.org/10.1016/j.bbrc.2021.07.092

    Article  CAS  PubMed  Google Scholar 

  12. Mao S, Park Y, Hasegawa Y, Tribble GD, James CE, Handfield M, Stavropoulos MF, Yilmaz Ö, Lamont RJ (2007) Intrinsic apoptotic pathways of gingival epithelial cells modulated by Porphyromonas gingivalis. Cell Microbiol 9(8):1997–2007. https://doi.org/10.1111/j.1462-5822.2007.00931.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sandros J, Karlsson C, Lappin DF, Madianos PN, Kinane DF, Papapanou PN (2000) Cytokine responses of oral epithelial cells to Porphyromonas gingivalis infection. J Dent Res 79(10):1808–1814. https://doi.org/10.1177/00220345000790101301

    Article  CAS  PubMed  Google Scholar 

  14. Yilmaz Ö, Watanabe K, Lamont RJ (2002) Involvement of integrins in fimbriae-mediated binding and invasion by Porphyromonas gingivalis. Cell Microbiol 4(5):305–314. https://doi.org/10.1046/j.1462-5822.2002.00192.x

    Article  CAS  PubMed  Google Scholar 

  15. Amano A, Nakagawa I, Okahashi N, Hamada N (2004) Variations of Porphyromonas gingivalis fimbriae in relation to microbial pathogenesis. J Periodontal Res 39(2):136–142. https://doi.org/10.1111/j.1600-0765.2004.00719.x

    Article  PubMed  Google Scholar 

  16. Inaba H, Sugita H, Kuboniwa M, Iwai S, Hamada M, Noda T, Morisaki I, Lamont RJ, Amano A (2014) Porphyromonas gingivalis promotes invasion of oral squamous cell carcinoma through induction of pro MMP 9 and its activation. Cell Microbiol 16(1):131–145. https://doi.org/10.1111/cmi.12211

    Article  CAS  PubMed  Google Scholar 

  17. Tang X, Asano M, O’Reilly A, Farquhar A, Yang Y, Amar S (2012) p53 is an important regulator of CCL2 gene expression. Curr Mol Med 12(8):929–943. https://doi.org/10.2174/156652412802480844

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Groeger S, Jarzina F, Domann E, Meyle J (2017) Porphyromonas gingivalis activates NFκB and MAPK pathways in human oral epithelial cells. BMC Immunol 18(1):1–11. https://doi.org/10.1186/s12865-016-0185-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kuboniwa M, Hasegawa Y, Mao S, Shizukuishi S, Amano A, Lamont RJ, Yilmaz Ö (2008) P. gingivalis accelerates gingival epithelial cell progression through the cell cycle. Microbes Infect 10(2):122–128. https://doi.org/10.1016/j.micinf.2007.10.011

    Article  CAS  PubMed  Google Scholar 

  20. Chang MC, Tsai YL, Chen YW, Chan CP, Huang CF, Lan WC, Lin CC, Lan WH, Jeng JH (2013) Butyrate induces reactive oxygen species production and affects cell cycle progression in human gingival fibroblasts. J Periodontal Res 48(1):66–73. https://doi.org/10.1111/j.1600-0765.2012.01504.x

    Article  CAS  PubMed  Google Scholar 

  21. Olsen I, Yilmaz Ö (2019) Possible role of Porphyromonas gingivalis in orodigestive cancers. J Oral Microbiol 11(1):1563410. https://doi.org/10.1080/20002297.2018.1563410

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kurita-Ochiai T, Ochiai K, Fukushima K (2000) Butyric-acid-induced apoptosis in murine thymocytes and splenic T-and B-cells occurs in the absence of p53. J Dent Res 79(12):1948–1954. https://doi.org/10.1177/00220345000790120501

    Article  CAS  PubMed  Google Scholar 

  23. Gabrilovich DI, Nagaraj S (2009) Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 9(3):162–174

    Article  CAS  Google Scholar 

  24. Peyret-Lacombe A, Brunel G, Watts M, Charveron M, Duplan H (2009) TLR2 sensing of F. nucleatum and S. sanguinis distinctly triggered gingival innate response. Cytokine 46(2):201–210. https://doi.org/10.1016/j.cyto.2009.01.006

    Article  CAS  PubMed  Google Scholar 

  25. McIlvanna E, Linden GJ, Craig SG, Lundy FT, James JA (2021) Fusobacterium nucleatum and oral cancer: a critical review. BMC Cancer 21(1):1–11. https://doi.org/10.1186/s12885-021-08903-4

    Article  CAS  Google Scholar 

  26. Fardini Y, Wang X, Témoin S, Nithianantham S, Lee D, Shoham M, Han YW (2011) Fusobacterium nucleatum adhesin FadA binds vascular endothelial cadherin and alters endothelial integrity. Mol Microbiol 82(6):1468–1480. https://doi.org/10.1111/j.1365-2958.2011.07905.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Han YW, Ikegami A, Rajanna C, Kawsar HI, Zhou Y, Li M, Sojar HT, Genco RJ, Kuramitsu HK, Deng CX (2005) Identification and characterization of a novel adhesin unique to oral fusobacteria. J Bacteriol 187(15):5330–5340. https://doi.org/10.1128/JB.187.15.5330-5340.2005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Abed J, Emgård JE, Zamir G, Faroja M, Almogy G, Grenov A, Sol A, Naor R, Pikarsky E, Atlan KA, Mellul A, Chaushu S, Manson AL, Earl AM, Ou N, Brennan CA, Garrett WS, Bachrach G (2016) Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed Gal-GalNAc. Cell Host Microbe 20(2):215–225. https://doi.org/10.1016/j.chom.2016.07.006

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Coppenhagen-Glazer S, Sol A, Abed J, Naor R, Zhang X, Han YW, Bachrach G (2015) Fap2 of Fusobacterium nucleatum is a galactose-inhibitable adhesin involved in co-aggregation, cell adhesion, and preterm birth. Infect Immun 83(3):1104–1113

    Article  CAS  Google Scholar 

  30. Kao SY, Chen YW, Chang KW, Liu TY (2009) Detection and screening of oral cancer and pre-cancerous lesions. J Chin Med Assoc 72(5):227–233. https://doi.org/10.1016/S1726-4901(09)70062-0

    Article  PubMed  Google Scholar 

  31. Gholizadeh P, Eslami H, Kafil HS (2017) Carcinogenesis mechanisms of Fusobacterium nucleatum. Biomed Pharmacother 89:918–925. https://doi.org/10.1016/j.biopha.2017.02.102

    Article  CAS  PubMed  Google Scholar 

  32. Hamada S, Koga T, Nishihara T, Fujiwara T, Okahashi N (1988) Characterization and immunobiologic activities of lipopolysaccharides from periodontal bacteria. Adv Dent Res 2(2):284–291. https://doi.org/10.1177/08959374880020021301

    Article  CAS  PubMed  Google Scholar 

  33. Krisanaprakornkit S, Kimball JR, Weinberg A, Darveau RP, Bainbridge BW, Dale BA (2000) Inducible expression of human β-defensin 2 by Fusobacterium nucleatum in oral epithelial cells: multiple signaling pathways and role of commensal bacteria in innate immunity and the epithelial barrier. Infect Immun 68(5):2907–2915. https://doi.org/10.1128/IAI.68.5.2907-2915.2000

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yost S, Stashenko P, Choi Y, Kukuruzinska M, Genco CA, Salama A, Frias-Lopez J (2018) Increased virulence of the oral microbiome in oral squamous cell carcinoma revealed by metatranscriptome analyses. Int J Oral Sci 10(4):1–10

    Article  CAS  Google Scholar 

  35. Abiko Y, Sato T, Mayanagi G, Takahashi N (2010) Profiling of subgingival plaque biofilm microflora from periodontally healthy subjects and from subjects with periodontitis using quantitative real-time PCR. J Periodontal Res 45(3):389–395. https://doi.org/10.1111/j.1600-0765.2009.01250.x

    Article  CAS  PubMed  Google Scholar 

  36. Metzger Z, Lin YY, Dimeo F, Ambrose WW, Trope M, Arnold RR (2009) Synergistic pathogenicity of Porphyromonas gingivalis and Fusobacterium nucleatum in the mouse subcutaneous chamber model. J Endod 35(1):86–94. https://doi.org/10.1016/j.joen.2008.10.015

    Article  PubMed  Google Scholar 

  37. Saito A, Kokubu E, Inagaki S, Imamura K, Kita D, Lamont RJ, Ishihara K (2012) Porphyromonas gingivalis entry into gingival epithelial cells modulated by Fusobacterium nucleatum is dependent on lipid rafts. Microb Pathog 53(5–6):234–242. https://doi.org/10.1016/j.micpath.2012.08.005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Huang R, Li M, Gregory RL (2011) Bacterial interactions in dental biofilm. Virulence 2(5):435–444. https://doi.org/10.4161/viru.2.5.16140

    Article  PubMed  PubMed Central  Google Scholar 

  39. Ruiz-Herrera J, Victoria Elorza M, Valentín E, Sentandreu R (2006) Molecular organization of the cell wall of Candida albicans and its relation to pathogenicity. FEMS Yeast Res 6(1):14–29. https://doi.org/10.1111/j.1567-1364.2005.00017.x

    Article  CAS  PubMed  Google Scholar 

  40. Jahanshahi G, Shirani S (2015) Detection of Candida albicans in oral squamous cell carcinoma by fluorescence staining technique. Dent Res J 12(2):115

    Google Scholar 

  41. Nelson RD, Shibata N, Podzorski RP, Herron MJ (1991) Candida mannan: chemistry, suppression of cell-mediated immunity, and possible mechanisms of action. Clin Microbiol Rev 4(1):1–19

    Article  CAS  Google Scholar 

  42. Chen X, Luo Q, Ding J, Yang M, Zhang R, Chen F (2020) Zymosan promotes proliferation, Candida albicans adhesion and IL-1β production of oral squamous cell carcinoma in vitro. Infect Agents Cancer 15(1):1–8. https://doi.org/10.1186/s13027-020-00315-6

    Article  CAS  Google Scholar 

  43. Abidullah M, Kiran G, Gaddikeri K, Raghoji S (2014) Leuloplakia-Review of a potentially malignant disorder. J Clin Diagn Research JCDR 8(8):ZE01. https://doi.org/10.7860/JCDR/2014/10214.4677

    Article  Google Scholar 

  44. Wilson D, Naglik JR, Hube B (2016) The missing link between Candida albicans hyphal morphogenesis and host cell damage. PLoS Pathog 12(10):1005867. https://doi.org/10.1371/journal.ppat.1005867

    Article  CAS  Google Scholar 

  45. Kasper L, König A, Koenig PA, Gresnigt MS, Westman J, Drummond RA, Lionakis MS, Groß O, Ruland J, Naglik JR, Hube B (2018) The fungal peptide toxin Candidalysin activates the NLRP3 inflammasome and causes cytolysis in mononuclear phagocytes. Nat Commun 9(1):1–20

    Article  CAS  Google Scholar 

  46. Choi YS, Kim Y, Yoon HJ, Baek KJ, Alam J, Park HK, Choi Y (2016) The presence of bacteria within tissue provides insights into the pathogenesis of oral lichen planus. Sci Rep 6(1):1–13

    CAS  Google Scholar 

  47. Ribet D, Cossart P (2015) How bacterial pathogens colonize their hosts and invade deeper tissues. Microbes Infect 17(3):173–183. https://doi.org/10.1016/j.micinf.2015.01.004

    Article  CAS  PubMed  Google Scholar 

  48. Al-Hebshi NN, Nasher AT, Idris AM, Chen T (2015) Robust species taxonomy assignment algorithm for 16S rRNA NGS reads: application to oral carcinoma samples. J Oral Microbiol 7(1):28934. https://doi.org/10.3402/jom.v7.28934

    Article  CAS  PubMed  Google Scholar 

  49. Moghimi M, Bakhtiari R, Mehrabadi JF, Jamshidi N, Jamshidi N, Siyadatpanah A, Mitsuwan W, Nissapatorn V (2020) Interaction of human oral cancer and the expression of virulence genes of dental pathogenic bacteria. Microb Pathog 149:104464. https://doi.org/10.1016/j.micpath.2020.104464

    Article  CAS  PubMed  Google Scholar 

  50. Zhang Z, Liu S, Zhang S, Li Y, Shi X, Liu D, Pan Y (2022) Porphyromonas gingivalis outer membrane vesicles inhibit the invasion of Fusobacterium nucleatum into oral epithelial cells by downregulating FadA and FomA. J Periodontol 93(4):515–525. https://doi.org/10.1002/JPER.21-0144

    Article  CAS  PubMed  Google Scholar 

  51. Gu X, Song LJ, Li LX, Liu T, Zhang MM, Li Z, Zuo XL (2020) Fusobacterium nucleatum causes microbial dysbiosis and exacerbates visceral hypersensitivity in a colonization-independent manner. Front Microbiol 11:1281. https://doi.org/10.3389/fmicb.2020.01281

    Article  PubMed  PubMed Central  Google Scholar 

  52. Li Y, Guo H, Wang X, Lu Y, Yang C, Yang P (2015) Coinfection with Fusobacterium nucleatum can enhance the attachment and invasion of Porphyromonas gingivalis or Aggregatibacter actinomycetemcomitans to human gingival epithelial cells. Arch Oral Biol 60(9):1387–1393. https://doi.org/10.1016/j.archoralbio.2015.06.017

    Article  PubMed  Google Scholar 

  53. Bor B, Cen L, Agnello M, Shi W, He X (2016) Morphological and physiological changes induced by contact-dependent interaction between Candida albicans and Fusobacterium nucleatum. Sci Rep 6(1):1–11

    Article  Google Scholar 

  54. Zhang Z, Yang J, Feng Q, Chen B, Li M, Liang C, Chen W (2019) Compositional and functional analysis of the microbiome in tissue and saliva of oral squamous cell carcinoma. Front Microbiol. https://doi.org/10.3389/fmicb.2019.01439

    Article  PubMed  PubMed Central  Google Scholar 

  55. Pushalkar S, Ji X, Li Y, Estilo C, Yegnanarayana R, Singh B, Saxena D (2012) Comparison of oral microbiota in tumor and non-tumor tissues of patients with oral squamous cell carcinoma. BMC Microbiol 12(1):1–15. https://doi.org/10.1186/1471-2180-12-144

    Article  Google Scholar 

  56. Torralba MG, Aleti G, Li W, Moncera KJ, Lin YH, Yu Y, Masternak MM, Golusinski W, Golusinski P, Lamperska K, Nelson KE (2021) Oral microbial species and virulence factors associated with oral squamous cell carcinoma. Microb Ecol 82(4):1030–1046. https://doi.org/10.1007/s00248-020-01596-5

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

The authors would like to express their gratitude to the president of Siksha 'O' Anusandhan deemed to be university, India, for giving the required facilities to carry out the aforementioned study

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

The concept was given by RB. She also examined and revised the paper. SL conducted the literature review and wrote the manuscript. SB guided in making diagrams. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Ruchi Bhuyan.

Ethics declarations

Ethics Approval and Consent to Participate

Not applicable.

Consent for Publication

Not applicable.

Competing interests

The authors declare that they have no Competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lenka, S., Bhuyan, S.K. & Bhuyan, R. Understanding the characteristics of the host genome and microbiome interaction in oral squamous cell carcinoma: a narrative review. Beni-Suef Univ J Basic Appl Sci 11, 126 (2022). https://doi.org/10.1186/s43088-022-00306-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43088-022-00306-z

Keywords