Next Article in Journal
A New Approach in Lipase-Octyl-Agarose Biocatalysis of 2-Arylpropionic Acid Derivatives
Previous Article in Journal
Temperature-Wise Calibration Increases the Accuracy of DNA Methylation Levels Determined by High-Resolution Melting (HRM)
Previous Article in Special Issue
How Can Imbalance in Oral Microbiota and Immune Response Lead to Dental Implant Problems?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

About a Possible Impact of Endodontic Infections by Fusobacterium nucleatum or Porphyromonas gingivalis on Oral Carcinogenesis: A Literature Overview

by
Luca Ciani
1,†,
Antonio Libonati
2,†,
Maria Dri
3,
Silvia Pomella
1,
Vincenzo Campanella
1 and
Giovanni Barillari
1,*
1
Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
2
Department of Surgical Sciences, Catholic University of Our Lady of Good Counsel of Tirane, 1001 Tirana, Albania
3
Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(10), 5083; https://doi.org/10.3390/ijms25105083
Submission received: 21 March 2024 / Revised: 2 May 2024 / Accepted: 4 May 2024 / Published: 7 May 2024
(This article belongs to the Special Issue Oral Microbiome and Oral Diseases 2.0)

Abstract

:
Periodontitis is linked to the onset and progression of oral squamous cell carcinoma (OSCC), an epidemiologically frequent and clinically aggressive malignancy. In this context, Fusobacterium (F.) nucleatum and Porphyromonas (P.) gingivalis, two bacteria that cause periodontitis, are found in OSCC tissues as well as in oral premalignant lesions, where they exert pro-tumorigenic activities. Since the two bacteria are present also in endodontic diseases, playing a role in their pathogenesis, here we analyze the literature searching for information on the impact that endodontic infection by P. gingivalis or F. nucleatum could have on cellular and molecular events involved in oral carcinogenesis. Results from the reviewed papers indicate that infection by P. gingivalis and/or F. nucleatum triggers the production of inflammatory cytokines and growth factors in dental pulp cells or periodontal cells, affecting the survival, proliferation, invasion, and differentiation of OSCC cells. In addition, the two bacteria and the cytokines they induce halt the differentiation and stimulate the proliferation and invasion of stem cells populating the dental pulp or the periodontium. Although most of the literature confutes the possibility that bacteria-induced endodontic inflammatory diseases could impact on oral carcinogenesis, the papers we have analyzed and discussed herein recommend further investigations on this topic.

1. Introduction

The proper functioning of the human body is aided by its colonization by saprophytic bacteria synthesizing homeostatic factors (e.g., vitamins) or antagonizing pathogenic microorganisms [1].
Mainly because of its direct connection with the external environment, the oral cavity is one of the areas of the human body richest in bacteria: the latter vary in type and number depending on factors specific to the host individual, such as age, sex, eating habits, and geographical area of origin and/or residence [2].
In healthy individuals that keep a good oral hygiene, saprophytic bacteria predominantly populate the oral cavity [3]. Poor oral hygiene, especially when prolonged over time, favors the prevalence of pathogenic bacteria over saprophytes, causing caries, abscesses, gingivitis, and/or periodontitis [3].
In particular, periodontitis develops following the formation of a microfilm on the teeth (the so termed dental plaque) in which obligate anaerobic Gram-negative bacteria, such as Fusobacterium nucleatum (F. nucleatum) and Porphyromonas gingivalis (P. gingivalis), are frequently found [4]. Using their membrane receptors, both F. nucleatum and P. gingivalis adhere to the cells of the oral cavity, penetrate them, and, at the same time, interact with each other or with other bacterial species that are concomitantly present therein [4]. Regarding such interactions, it is well established that a pathogenic bacterial strain increases the virulence of another pathogenic strain, and that saprophytic bacteria counter the pathogenic ones [3].
Of importance, periodontitis has been found to be linked to the development and clinical progression of squamous cell carcinoma (SCC), an aggressive malignancy which constitutes over 90% of oral cavity neoplasms (oral SCC, OSCC) [5,6]. This pathogenic association is not surprising, given the well-known pro-tumor effects of chronic inflammation: the latter, in fact, implies the long-lasting production of molecules altering cell survival, growth, differentiation or motility, and concomitantly promoting tissue matrix remodeling [7]. However, while the carcinogenic effects of some families of viruses are widely documented, those of bacteria are poorly defined.
Regarding the OSCC, however, we have some more information. In fact, evidence indicates that both P. gingivalis and F. nucleatum are endowed with tumorigenic activities which are likely to favor OSCC onset and/or progression [5]. Confirming this, the dysplastic and/or hyperplastic lesions that often precede OSCC development (oral pre-malignant diseases, OPMDs) [8] are densely populated by pathogenic bacteria, including F. nucleatum or P. gingivalis, and display a reduced number of saprophytic bacteria as compared with healthy oral mucosa [9,10]. In OSCCs, the presence of F. nucleatum and P. gingivalis becomes even more evident, and its intensity parallels the clinical progression of the disease, positively correlating with the tumor size and lymph node metastases [9,10].
It is noteworthy that F. nucleatum and P. gingivalis are also present in endodontic diseases such as pulpitis, apical granuloma, and radicular cyst [11,12]. That being so, herein we evaluated whether there is any link between endodontic infections by these two bacteria and oral carcinogenesis.
Specifically, in the present review, we first summarize published data concerning F. nucleatum or P. gingivalis impact on the onset and on the clinical evolution of OPMDs and OSCC. Then, the literature is examined regarding the effects that infection of dental pulp and/or periodontium by F. nucleatum or P. gingivalis could have on molecular or cellular events leading to the development or the progression of OSCC, endodontic tumors, and/or odontogenic tumors.
Data were searched for in the PubMed Central electronic database of the National Library of Medicine (National Institutes of Health, Bethesda, Maryland, United States of America). The search and the analysis of the retrieved articles were carried out from November 2022 to January 2024. In total, 90 articles were selected for full text screening, and 64 of them were included in the final study. The other articles discussed and cited in this review served to complete the description of the topics herein considered.

2. Activities of P. gingivalis and F. nucleatum Leading to the Development of OPMDs

When they colonize the oral cavity, P. gingivalis and F. nucleatum release enzymes that digest both the cells and the extracellular matrix, thus dismantling the tissue [5,6]. This is followed by an intense inflammatory response during which leukocytes are recruited to smash the bacteria and the tissue debris that bacteria have generated [5,6,13].
Amidst the recruited leukocytes are monocytes that, once they reach the inflamed site, differentiate into macrophages [5,6,13]. Of note, F. nucleatum binds the toll-like receptors (TLR)-2 and -4 that are expressed on the membrane of macrophages: this binding is followed by the activation of the nuclear factor kappa-B (NF-kB) transcription factor that, in turn, promotes the expression of inflammatory cytokines including interleukin (IL)-6 and tumor necrosis factor (TNF) α by macrophages [14] (Table 1).
Additional pro-inflammatory mediators produced by the leukocytes upon their arrival in the injured oral tissue include IL-1 and IL-8 [5,6,65].
Apart from the leukocytes, the inflamed oral tissue is infiltrated by fibroblasts directed at repairing the damaged tissue [65]. Those fibroblasts synthesize the transforming growth factor (TGF)-β1, while nearby macrophages release the epidermal growth factor (EGF) [66]: both types of cytokines deeply influence the proliferation and differentiation of epithelial cells [66].
It is noteworthy that the pro-inflammatory IL-1, IL-6, IL-8, and TNFα, as well as TGF-β1 and EGF, are almost absent from the healthy oral cavity, are found at low levels in periodontitis, are upregulated in OPMDs, and are expressed at even higher levels in OSCCs [65,66], especially when F. nucleatum and/or P. gingivalis are present [5,6].
In fact, in the inflamed oral mucosa, not only leukocytes, macrophages, and fibroblasts, but also epithelial cells infected by F. nucleatum or P. gingivalis produce inflammatory cytokines and growth factors [14,15,16,17,18,19,20,21,22,23,24] (Table 1). Consequently, epithelial cells lining the inflamed, bacteria-infected oral cavity are exposed for prolonged times to cytokines which, particularly when combined with each other, promote epithelial-to-mesenchymal transition (EMT) [65,67].
The latter is a multistep process that occurs during the inflammation that precedes and accompanies the repair of a wounded epithelium [65,67]. EMT implies that epithelial cells lose their peculiar static and polarized phenotype and acquire a motile one which resembles that of mesenchymal cells [65,67]. Such a change allows epithelial cells migration that is required for wound healing [65,67].
Briefly, IL-1, IL-6, IL-8, TNF, EGF, and TGF-β trigger intracellular signaling pathways such as the Wingless-related integration site (Wnt)/β catenin, the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT), and the mitogen-activated protein kinases (MAPK)/extracellular regulated kinases (ERK) [65,67]. This leads to the activation of transcription factors such as NF-kB, zinc finger snail homolog (SNAI), basic helix–loop–helix twist homolog (TWIST), and zinc finger E-box binding homeobox (ZEB) [65,67,68,69]. These transcription factors, in turn, promote the expression of mesenchymal proteins (e.g., vimentin or neuronal cadherin) and pro-invasive proteolytic enzymes, while repressing the expression of epithelial markers such as epithelial (E)-cadherin [65,67,68,69].
Of note, P. gingivalis and F. nucleatum can activate SNAI and/or ZEB either directly or by inducing the synthesis of IL-1β, TNF-α, EGF, and/or TGF-β [19,25,26,27] (Table 1).
Specifically, when P. gingivalis and F. nucleatum make contact with epithelial cells, they trigger the EMT-associated transcriptional activity of SNAI, ZEB, TWIST, and β-catenin, thereby upregulating vimentin and pro-invasive enzymes while concomitantly downregulating E-cadherin [26,70,71,72]. This phenomenon, which is particularly evident when oral epithelial cells are infected with P. gingivalis and/or F. nucleatum [26], explains why the physiological, reversible EMT arising during the transient inflammation that goes along with the repair of the damaged oral mucosa is exacerbated and becomes stable in chronic bacterial periodontitis [9,72].
In this context, it must be highlighted that the EMT arising in the oral cavity upon the infection by pathogenic bacteria is among the main inducers of OPMDs development [9,72].
Indeed, in the inflamed oral cavity, the trans-differentiation of epithelial cells can be accompanied by their proliferation. Specifically, IL-1 directly triggers the growth of dysplastic oral keratinocytes [73]. At the same time, IL-1 stimulates keratinocytes to synthesize IL-6 and IL-8 [73] that, in turn, intensify the EMT process [74]. Moreover, F. nucleatum and P. gingivalis induce the production of EGF [19] (Table 1), which sparks both the EMT and the proliferation of oral keratinocytes [75]. In addition, F. nucleatum and P. gingivalis promote, in oral keratinocytes, the synthesis of factors stimulating cell cycle progression (e.g., the cyclin-dependent kinases), and repress the expression or the activity of growth inhibitors such as p53 [32,33,34,35,36] (Table 1). In doing so, F. nucleatum and P. gingivalis amplify the proliferative stimulus that EGF exerts on keratinocytes.
Altogether, these molecular and cellular events explain why the presence of F. nucleatum and P. gingivalis in the oral cavity associates with the development of dysplastic/hyperplastic lesions such as OPMDs [9,10].

3. Effects of P. gingivalis and F. nucleatum Leading to the Onset of OSCC

OSCC results from the malignant transformation of oral keratinocytes: this event is facilitated when the carcinogen is acting on trans-differentiated and/or proliferating keratinocytes, i.e., on an OPMD [76,77].
OSCC is induced mainly by chemical pathogens (e.g., polycyclic hydrocarbons or alcohol), although microbial agents such as the human papilloma viruses certainly contribute to oral carcinogenesis [78]. Also, P. gingivalis and F. nucleatum are likely to play a direct role in OPMD evolution to OSCC, as they release mutagenic substances (e.g., hydrogen sulfide) into the oral tissue they have colonized [28,29,30,31] (Table 1).
Like all carcinomas, OSCC consists of transformed epithelial cells that display a strong resistance to programmed cell death (apoptosis) [79]. In this regard, it must be highlighted that the intracellular signaling pathways stimulated by F. nucleatum and P. gingivalis hamper apoptosis promoters such as p53 or Bad [40,44] in oral keratinocytes (Table 1). At the same time, signaling by F. nucleatum and P. gingivalis upregulates the expression of cell survival factors such as AKT, Bcl-2, heat-shock protein 27, superoxide dismutase 2, baculoviral IAP repeat-containing protein 3, or the signal transducers and activators of transcription (STATs) transcription factors [37,38,39,41,42,43,45,46,80] (Table 1).
STATs are ignited by the Janus-associated kinases (JAKs) [6]. In bacterial-infected and inflamed oral cavity, JAKs can be triggered by F. nucleatum and P. gingivalis [43,46] or upon the binding of IL-1β, IL-6, IL-8, TNF-α, EGF, or TGF-β to their membrane receptors [81,82,83,84,85,86,87]. In this context, it must be underlined that, while they directly activate the JAKs, F. nucleatum and P. gingivalis stimulate OSCC cells to produce the abovementioned JAK-activating cytokines [14,15,16,17,18,19,20,21,22,23,24].
STATs actuation results in the induction of the expression of genes that impact not only on cell survival, but also on cell proliferation, motility, and differentiation [88]. All these activities well explain the fact that STATs dysregulation accompanies the onset or progression of a variety of human malignancies [88,89].
Among STAT family members, STAT3 is over-activated in OSCCs, where it strengthens the viability and promotes the proliferation of OSCC cells [82,83,86,90].
Despite this, the establishment and growth of the OSCC can still be hampered by host immune reactions directed against the developing tumor [13]. In fact, the growing OSCC undergoes infiltration by immune cells such as B, T, and NK lymphocytes, dendritic cells, as well as two types of tumor-associated macrophages (TAMs): M1 and M2 [13]. The former has an anti-tumor action as it engulfs tumor cells and processes their antigens by presenting them to the lymphocytes in association with class II major histocompatibility complex (MHC-II) molecules which they display at high levels [13]. In contrast, the M2 macrophages bear low levels of MHC-II, are poorly capable of engulfing the OSCC cells, and produce high amounts of pro-tumor molecules [13]. This explains why a high number of infiltrating M2 macrophages correlates with the poor prognosis of OSCC patients [91].
Of relevance, STAT3, which is triggered by F. nucleatum or P. gingivalis [37,43,46,80], promotes the synthesis of cytokines in OSCCs that depress the response of cytotoxic T lymphocytes against the carcinoma cells [90].
Of utmost interest for the present review, P. gingivalis induces the polarization of TAMS toward the M2 phenotype [13], stimulates the generation of myeloid-derived dendritic suppressor cells from monocytes [56,58], and causes T cells anergy and apoptosis [57] (Table 1). For its part, F. nucleatum degrades immunoglobulins [55] and protects tumors from immune cells attack by activating the immune inhibitory receptors TIGIT (T cell immunoreceptor with Ig and ITIM domains) and CEACAM1 (CEA Cell Adhesion Molecule 1) [59,60] (Table 1).

4. Effects of P. gingivalis and F. nucleatum Leading to OSCC Progression

When they escape anti-tumor immune responses, OSCC cells proliferate uncontrollably, thereby infiltrating the tissue in which they have developed and replacing pre-existing normal cells [92,93]. This is by reason that OSCC cells are invasive, that is, they produce proteolytic enzymes capable of degrading the intercellular junctions and both the interstitial and the peritumoral extracellular matrix [94].
Cellular invasiveness is a basic feature of the EMT phenotype [65,67]. In this regard, it must be underscored that, when F. nucleatum and P. gingivalis penetrate the OSCC cells, they activate ZEB and SNAI, thus directly promoting the pro-invasive EMT phenotype of those cancerous cells [19] (Table 1).
Among the proteolytic enzymes produced by OSCC cells, the matrix metalloproteases (MMPs) play a preponderant role in OSCC invasiveness [94]. In this context, both F. nucleatum and P. gingivalis stimulate MMP activity and OSCC cell invasion (Table 1). Specifically, these bacteria trigger intracellular signaling pathways including Wnt, integrin/FAK, or p38 MAPK that, in turn, lead to MMPs expression and actuation [44,48,49,52,53,54,63]. Additional ways through which P. gingivalis and F. nucleatum provoke OSCC invasion include by producing sodium butyrate [95], a metabolite that promotes MMPs synthesis [96]. Moreover, F. nucleatum can also degrade the extracellular matrix directly, that is, via its bacterial proteases [55], while P. gingivalis stimulates the locomotion of the carcinoma cells [51].
In summary, F. nucleatum and P. gingivalis exasperate EMT in OSCC cells, further reducing the levels of E-cadherin and dramatically increasing those of MMPs [69,72].
The combination of E-cadherin downregulation and MMP overexpression loosens intercellular adhesions, causing the detachment of OSCC cells from the tumor and their centrifugal migration, which will eventually expand the OSCC mass [97].
The enlarging OSCC requires greater amounts of oxygen and nutrients that local vessels are unable to provide: the formation of new blood vessels from pre-existing ones (the so-termed angiogenesis) is, therefore, triggered [76,98,99]. In this context, the GroEL protein of P. gingivalis has been reported to promote new vessel formation in vivo [61], while infection by F. nucleatum impacts angiogenesis-related genes [62].
In OSCC, STAT3 induces the expression of cytokines that promote angiogenesis, hence supporting the growth of the tumor [90]. Among STAT3-induced angiogenic cytokines are the vascular endothelial growth factor (VEGF) and the fibroblast growth factor (FGF)-2 [100,101]. Definitely, P. gingivalis capability of turning on STAT could explain the angiogenic effect of the bacterium. Still in this regard, F. nucleatum transiently increases the expression of VEGF and its type 1 receptor by endothelial cells [102].
Moreover, F. nucleatum stimulates endothelial cells to produce and release IL-1 and TNFα, hence further increasing the concentration of these inflammatory mediators in the microenvironment [102]. Therefore, as the tumor progresses, EMT-promoting transcription factors are more and more activated, until the carcinoma cells acquire a phenotype that is very similar to that of stem cells (cancer stem cells, CSCs) [103,104,105].
Specifically, CSCs appearance in an OSCC is often preceded by the EMT of the carcinoma cells and/or resident stem cells [103,105]. In agreement with P. gingivalis capability of sparking pro-EMT transcription factors, the P. gingivalis-infected oral epithelial cells express stem cell markers such as CD44 and CD133 [50] (Table 1). Similar effects have been described for F. nucleatum in other types of carcinomas [106]. F. nucleatum further inhibits the differentiation of gingival stem cells and triggers events linked to their neoplastic transformation [22,64] (Table 1).
Since they are very invasive and plastic, that is, adaptable to the characteristics of tissues other than that in which they have originated, CSCs are very metastatic [105]. Specifically, CSCs rapidly migrate through the peritumoral matrix, degrade the basement membrane, reach the lymphatic or blood capillaries, and penetrate them [65].
However, when they circulate in the blood or lymph, OSCC cells lack the anchorage to a solid substrate which, analogously to any other adherent cell type, they need to survive [107]. Consequently, circulating OSCC cells may undergo a peculiar, fast-occurring type of apoptosis that is termed “anoikis” [107]. In this regard, the activation of the PI3K/AKT signaling pathway promoted by P. gingivalis [39] or F. nucleatum [47] makes OSCC cells resistant to anoikis [107], thereby effectively favoring OSCC metastasization.
It is noteworthy that, by virtue of their high resistance to apoptosis, CSCs can survive even ionizing radiation or cytotoxic drugs [108,109]: this renders the OSCCs that are rich in CSCs poorly sensitive to anti-tumor therapies [110].

5. Possible Impact of Endodontic Infections by P. gingivalis or F. nucleatum on OSCCs

While the oral cavity hosts hundreds of different microbial species, the dental pulp is, under physiological conditions, sterile [111]. This is because the pulp is separated and protected from the external environment by the dentin and enamel [111].
Damage to these coatings, resulting from traumatic events or tooth decay, allows oral bacteria to penetrate the dental pulp [112]. There, bacteria find nutrients and oxygen that favor the replication of saprophytic species [111,113,114]. The latter then consume the oxygen and the nutrients and produce catabolites: all this changes the characteristics of the microenvironment in such a way as to favor the prevalence of pathogenic anaerobic bacteria, P. gingivalis and F. nucleatum included [111,113,114].
However, the pulp reacts against the invading microorganisms [11]. Specifically, when bacteria arrive in the pulp, cells that are present therein such as the odontoblasts lining the pulp chamber toward the dentine, the dendritic cells, and the fibroblasts produce chemokines that recruit, to the site of the lesion, the leukocytes that will fight the bacteria [11]. As an example of this, when P. gingivalis or F. nucleatum binds the microbial recognition receptors TLR2 and TLR4 on the surface of odontoblasts, NF-kB is actuated, resulting in odontoblast production of the leukocyte-activating TNFα and IL-8 [115] (Figure 1). Other cytokines whose synthesis by dental pulp cells is triggered by pathogenic bacteria include IL-1β, IL-6, and TGF-β1 [116,117].
In addition to the odontoblasts, the dendritic cells, and the fibroblasts, the dental pulp is populated by stem cells (dental pulp stem cells, DPSCs): these are characterized by a high replicative index and by their capability of differentiating into a wide variety of cell types, which include odontoblasts, osteoblasts, chondrocytes, adipocytes, neural cells, and endothelial cells [118,119,120,121,122]. DPSCs carry out reactive, defensive, and regenerative actions that are mediated by the molecules these stem cells produce [123]. Specifically, DPSCs have been shown to synthesize and release inflammatory mediators such as TNFα and growth factors including VEGF, FGF-2, and TGF-β1 [124].
It is noteworthy that both P. gingivalis and F. nucleatum are capable of infecting DPSCs where they trigger NF-κB, which, in turn, activates the expression of inflammatory mediators, increasing their local concentrations [117,125,126] (Figure 1). The same effect can result from the bare exposure of the oral stem cells to P. gingivalis, without the need for them to be infected by the bacterium [127]. This is because either P. gingivalis or its lipopolysaccharides (LPSs) trigger the phosphorylation of ERK and p38 MAPK in DPSCs, this phenomenon being followed by the production of pro-inflammatory cytokines [127]. In this regard, MAPK/ERK activation is also known to lead to VEGF and TGF-1β expression [128,129]. In addition to VEGF and TGF-1β, FGF-2 as well could likely be upregulated in the infected pulp, as found for other inflamed tissues [130,131]. Therefore, the levels of inflammatory cytokines and/or growth factors could get high in a bacteria-infected and inflamed pulp, where they would be simultaneously released by DPSCs, odontoblasts, dendritic cells, fibroblasts, and by the pulp-infiltrating leukocytes. Given the mitogenic, pro-EMT, and pro-invasive effect of the abovementioned cytokines, these findings suggest that an infection of the dental pulp may impact OSCC (Figure 1).
This hypothesis is corroborated by the finding that, upon their release by DPSCs, the VEGF, FGF-2, TNF-α, and TGF-β1 promote the proliferation of OSCC cells [124] (Figure 1). Consistently, other studies have described the positive impact of each of the aforecited molecules on OSCC cell growth [56,132,133]. In addition, the same cytokines could favor OSCC progression also because of their capability of stimulating the invasiveness of carcinoma cells and of maintaining or even exacerbating their dedifferentiation status [132,133,134,135,136] (Figure 1). Moreover, due to their immunosuppressive properties, VEGF, FGF-2, and TGF-β1 could support OSCC growth also in an indirect fashion, that is, by inhibiting the immune response that OSCC-infiltrating leukocytes exert against the cancer cells [137,138,139] (Figure 1). Last but not least, the VEGF and FGF-2 released by DPSCs could enlarge the OSCC mass by promoting angiogenesis [76,98,99] (Figure 1).
Taken together, all these data and considerations strengthen the hypothesis of a pathogenetic link between pulp bacterial infections and OSCC.
In the absence of the proper endodontic therapy, a deep dental caries causes pulp necrosis that, in turn, permits bacteria to replicate in the root canals and reach the alveolar bone through the apical foramen [11,112,140].
Upon bacterial infection of the root canals, inflammation develops at the interface between the infected radicular pulp and the periodontal ligament, eventually resulting in the destruction of periodontal tissues and the resorption of the alveolar bone [112,140]. In this context, it is noteworthy that the inflammatory reaction promoted by P. gingivalis inhibits the mineralization capability of cementoblasts [141]. In addition, P. gingivalis can directly promote the apoptosis of cementoblasts [142], while its LPSs inhibit cementoblast growth [143].
Such a disruption of the periodontium defines the “apical periodontitis”, which can lead to the formation of apical granulomas and, subsequently, radicular cysts [11] (Figure 2).
Radicular cysts are lined by a stratified epithelium that derives from the proliferation of the so-called “epithelial cell rests of Malassez” (ERM) residing in the periodontal ligament [144,145]. ERMs are embryonic epithelial remnants that maintain the characteristics of stem cells, being able to differentiate into various cytotypes during periodontal repair [146].
In radicular cysts, ERMs proliferate because they are stimulated by inflammatory cytokines secreted by the various cytotypes that populate the cyst [144,147,148,149,150,151] (Figure 2). Specifically, in addition to epithelial cells, the cyst contains fibroblasts, lymphocytes, monocytes/macrophages, and Langerhans cells [149,150,152,153].
Due to the biosynthetic activity of all those cell types, IL-1β, IL-6, IL-8, and TNFα are expressed in radicular cysts together with growth factors (e.g., TGF-β1, FGF-2, VEGF) or growth factor receptors (e.g., EGF receptor) [151,154,155,156]. In the cysts, MMPs are also detected and are known to be induced by IL-1β, IL-6, IL-8, TNFα, TGF-β1, FGF-2, VEGF, or EGF [147]: these results indicate that inflammatory cytokines and growth factors effectively act on the cells of the cysts, in both an autocrine and paracrine fashion, eventually promoting pro-tumor events such as cell proliferation and invasion (Figure 2).
In most cases, apical lesions heal upon the disinfection and filling of the dental pulp and root canals [112]. In such a contingency, inflammatory cytokines and growth factors are no longer produced or their levels strongly diminish, thus causing the apoptosis of the ERMs that line the radicular cyst [157].
However, sometimes root canals cannot be cleaned and filled in full and this causes infection to abide [112]. In that case, inflammation of the pulp lasts, keeping on IL-1, IL-6, IL-8, and/or TNF production and release [11]. In the eventuality that also the cyst will persist, its cells would prolongedly release these same inflammatory cytokines together with TGF-β1, FGF-2, and VEGF (Figure 2).
In addition to promoting cell growth and invasiveness, these cytokines inhibit cell differentiation or, at the very least, promote EMT, especially when they are all present in the microenvironment at the same time (Figure 2).
Previous work has shown that ERMs can acquire the EMT phenotype [158]. Others have found that DPSCs express genes involved in EMT [159], raising the possibility that DPSCs may also undergo this process.
Given that, the infection of such poorly differentiated cells by pathogenic bacteria could very likely favor the onset of an OSCC (Figure 2). In accordance with this hypothesis, the case of a patient with an infected radicular cyst of the mandible in which OSCC developed has been recently described [160].

6. Endodontic Infections by P. gingivalis or F. nucleatum and Endodontic or Odontogenic Tumors

Despite the simultaneous presence of mitogenic, EMT-inducing, and/or pro-invasive cytokines and that of pro-carcinogenic bacteria, to date no malignant evolution of pulpitis or apical granuloma has been reported in the literature [11,12].
This could depend on several reasons. To begin with, the mesenchymal stromal cells of the dental pulp secrete antibacterial substances [161]. Moreover, cariogenic bacteria produce lactic acid [162], a compound known to exert anti-inflammatory and antitumor actions [163]: this could explain the inverse correlation existing between the presence of tooth decay and the development of head and neck SCC [162]. Furthermore, one should also consider that the onset of a neoplasm in the dental pulp would lead to a reactive pulpitis which sooner or later would be treated as such [12]. In this context, it is likely that the expansion of the tumor mass in the narrow space of the dental chamber would irritate the odontoblasts, inducing them to produce and release new dentin: this would lead to a further shrinking of the dental chamber and, finally, to pulp necrosis [12]. Nonetheless, if one may believe that the small size of the pulp chamber prevents the development of a tumor inside it, this is denied by the finding that cancer metastases can colonize dental pulp [164,165].
Certainly, however, important differences exist between the pathogenetic mechanisms of oral cavity tumors and those of endodontic inflammatory diseases. One of these differences concerns epigenetic alterations such as, for example, those regarding the DNA methylation status [166,167]. Specifically, altered genes in OSCC are those regulating cell survival and proliferation [167], that is, the ones generally most involved in neoplastic induction and progression [166]. In contrast, periodontitis- or pulpitis-associated epigenetic alterations have turned out to mainly affect the genes coding for inflammatory mediators [167]. As such, these results confute, at least in part, the possibility of a neoplastic evolution of an endodontic pathology.
Anyway, regardless of all these considerations, it should be remembered that chronic inflammation can predispose to the development of a neoplasm [7]. It must also be highlighted that the dental pulp is populated by cells that are susceptible to neoplastic transformation such as, for example, the odontoblasts, the fibroblasts, and, above all, the DPSCs [11,168,169]. In this regard, the neoplastic transformation of DPSCs is believed to give origin to the odontogenic myxoma, an invasive and highly recurrent dental tumor [170,171]. Yet, for DPSCs transformation to occur, the intervention of a carcinogen is required [168,169], since these stem cells do not behave like neoplastic cells either in vitro or in vivo [172].
Nevertheless, considering the importance of inflammation and cellular stemness in cancerogenesis [7,76,77,173], it is noteworthy that, upon their release by P. gingivalis-infected or F. nucleatum-infected DPSCs, the inflammatory cytokines IL-1β, IL-6, and TNFα maintain DPSCs stemness by activating the Wnt/β-catenin signaling [119,120,121] (Table 2). The same effect is also detectable in DPSCs not infected by P. gingivalis, but only exposed to the LPSs of the bacterium [118] (Table 2).
P. gingivalis, its LPSs, or the inflammatory cytokines induced by this bacterium affect the differentiative potential, but not the survival or growth, of DPSCs [118,119,127]. In contrast, the LPSs of Escherichia coli, another bacterium involved in periodontitis [181], increase DPSCs viability [182], possibly favoring endodontic carcinogenesis.
In addition to DPSCs, F. nucleatum can also infect the stem cells of the periodontal ligament and apical papilla, thereby halting their differentiation [174] (Table 2). This is due to F. nucleatum ability to downregulate the expression of WDR5, a promoter of histone methylation [174]. Still in the stem cells of the periodontal ligament and apical papilla, F. nucleatum upregulates the expression of TBX3 and NFIL3, two transcription factors that are involved in embryonic development and are overexpressed in a wide variety of cancers where they functionally hamper cell growth inhibitors, repress cell differentiation, and stimulate cell invasion [174].
As observed in DPSCs, also in the stem cells of the periodontal ligament and of the apical papilla, the LPSs of P. gingivalis spark Wnt/β-catenin or p38 MAPK signaling and activate NF-kB, thus triggering the expression of inflammatory cytokines [175,176] (Table 2). Similarly, F. nucleatum promotes the synthesis of IL-8 and IL-10 by apical papilla stem cells [177] (Table 2).
Altogether, because of their impact on cellular stemness, these activities of F. nucleatum and P. gingivalis may have relevance for the onset of dental tumors.
Concerning the effects that P. gingivalis exerts on other types of dental cells, results from a previous study indicate that the bacterium triggers both the PI3K/AKT and the MAPK signaling pathways in cementoblasts, thus inhibiting the differentiation of these cells [142] (Table 2).
Others have shown that the LPSs of P. gingivalis upregulate IL-1 and MMP-2 expression in cementoblasts [143] (Table 2). This could be of importance for dental tumorigenesis, as IL-1β inhibits DPSCs differentiation [120] and MMP-2 mediates SCs invasion [178] and it is overexpressed in odontogenic neoplasms, contributing to their invasive behavior [171].
Once again regarding a possible role for pathogenic bacteria in odontogenic tumors, evidence suggests a link between endodontic infections and ameloblastoma, the most common odontogenic tumor [183]. In this regard, it is useful to remind that, although it is benign, ameloblastoma can rapidly grow and be locally invasive, thereby causing significant morbidity [183].
Of note, the odontogenic ameloblast-associated protein (ODAM) is diffusely and highly expressed in the P. gingivalis-infected sub-gingival regions, as compared to the uninfected ones [180] (Table 2). In physiologic conditions, ODAM is expressed at low levels, mediating the adhesion of the junctional epithelium to the tooth surface [184]. As for pathologic settings, ODAM is overexpressed in ameloblastoma as well as in a variety of carcinomas such as that of the stomach, lung, or mammary gland [179].
Furthermore, although it is not known whether F. nucleatum or P. gingivalis can infect the ameloblasts, the same molecules that are upregulated or activated in OSCC cells infected with F. nucleatum or P. gingivalis (e.g., ZEB, SNAI, TWIST, β-catenin, the MMPs, IL-8, and vimentin) are also overexpressed in ameloblastoma [185,186,187,188].

7. Conclusions and Future Directions

Results from clinical–epidemiological studies indicate that OSCC development and progression are favored by bacterial periodontitis [5,6]. In this context, P. gingivalis and F. nucleatum, two causative agents of periodontitis, colonize OPMDs and OSCCs [9,10] where they exert tumorigenic activities. In particular, P. gingivalis and F. nucleatum support the onset of OPMDs and their progression to OSCCs by promoting the transdifferentiation, survival, proliferation, and invasiveness of oral epithelial cells [19,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,63,64]. In addition, P. gingivalis and F. nucleatum actions can sustain the growth and metastatization of an established OSCC [13,55,56,57,58,59,60,61,62]. All these effects of P. gingivalis and F. nucleatum are directly promoted by the two bacteria and/or are mediated by the cytokines they induce [14,15,16,17,18,19,20,21,22,23,24].
Given that P. gingivalis and F. nucleatum are also present in pulpitis, apical granulomas, or radicular cysts [11,12], here we discuss the literature concerning any eventual pathogenetic link between endodontic infections by the abovecited bacteria and oral carcinogenesis.
Specifically, here we report that P. gingivalis and/or F. nucleatum spark, in dental pulp cells or periodontal cells, the synthesis of cytokines that, in turn, trigger the survival, growth, and invasion of OSCC cells [115,116,117,124,125,126,127,132,133,134,135,136]. Furthermore, the two bacteria and the cytokines induced by them stop the differentiation of DPSCs and periodontal stem cells while concomitantly stimulating their growth and invasiveness [118,119,120,121,174,175,176,177] and releasing mutagenic substances [28,29,30,31].
In conclusion, the set of data discussed here, recovered from the fragmented literature produced on this specific topic, lead to further investigation into the effects that a bacterial endodontic infection could have on oral carcinogenesis.
Noteworthy is the fact that many of the pro-tumor activities of P. gingivalis and F. nucleatum depend on the capability that the two bacteria have to activate the PI3K/AKT intracellular signaling pathway that is key to OSCC development and progression [39,47,189].
Since several human malignancies display PI3K/AKT dysregulation, antagonists of this pathway have been developed that exert anticancer activity [190]. As for OSCC, PI3K/AKT inhibitors hamper the proliferation and invasion of the carcinoma cells and increase their sensitivity to conventional antitumor therapies [191,192,193,194].
However, regarding a clinical use of PI3K/AKT inhibitors against OSCC, one should consider that, in this tumor, AKT is continuously reactivated due to the peculiar cellular and molecular characteristics of the tumor microenvironment [189]. This has suggested the usage of combinatorial therapeutic regimens directed against OSCC. Consistently, treatment of OSCC patients with PI3K/AKT inhibitors combined with cytostatic/cytotoxic chemotherapeutics has provided promising results [189,190].
Based on the data herein described, OSCC patients could take conventional chemotherapeutics and PI3K/AKT inhibitors together with antibiotics and anti-inflammatory drugs. Because the oral cavity is accessed effortlessly, anti-OSCC drugs would be administered also topically, thus minimizing their collateral effects.

Author Contributions

All the authors have searched for and analyzed the articles that are discussed herein. L.C., A.L., V.C. and G.B. wrote the manuscript; M.D. and S.P. completed some paragraphs. Authors have read and agreed to the submitted version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

Supported by the Italian Ministry of University and Research (MUR), University Scientific Research Projects (RSA) 2021, grant. no. E83C22002040005. S.P. is funded by a grant from MUR, Research and Innovation Projects (PON).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Aggarwal, N.; Kitano, S.; Puah, G.R.Y.; Kittelmann, S.; Hwang, I.Y.; Chang, M.W. Microbiome and human health: Current understanding, engineering, and enabling technologies. Chem. Rev. 2023, 123, 31–72. [Google Scholar] [CrossRef] [PubMed]
  2. Sedghi, L.; Di Massa, V.; Harrington, A.; Lynch, S.V.; Kapila, Y.L. The oral microbiome: Role of key organisms and complex networks in oral health and disease. Periodontol. 2000 2021, 87, 107–131. [Google Scholar] [CrossRef] [PubMed]
  3. Radaic, A.; Kapila, Y.L. The oralome and its dysbiosis: New insights into oral microbiome-host interactions. Comput. Struct. Biotechnol. J. 2021, 19, 1335–1360. [Google Scholar] [CrossRef] [PubMed]
  4. Gasmi Benahmed, A.; Kumar Mujawdiya, P.; Noor, S.; Gasmi, A. Porphyromonas gingivalis in the Development of Periodontitis: Impact on Dysbiosis and Inflammation. Arch. Razi Inst. 2022, 77, 1539–1551. [Google Scholar] [CrossRef]
  5. Irfan, M.; Delgado, R.Z.R.; Frias-Lopez, J. The oral microbiome and cancer. Front. Immunol. 2020, 11, 591088. [Google Scholar] [CrossRef] [PubMed]
  6. Hoare, A.; Soto, C.; Rojas-Celis, V.; Bravo, D. Chronic inflammation as a link between periodontitis and carcinogenesis. Mediators Inflamm. 2019, 2019, 1029857. [Google Scholar] [CrossRef]
  7. Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef] [PubMed]
  8. Khan, M.M.; Frustino, J.; Villa, A.; Nguyen, B.C.; Woo, S.B.; Johnson, W.E.; Varelas, X.; Kukuruzinska, M.; Monti, S. Total RNA sequencing reveals gene expression and microbial alterations shared by oral pre-malignant lesions and cancer. Hum. Genomics 2023, 17, 72. [Google Scholar] [CrossRef] [PubMed]
  9. La Rosa, G.R.M.; Gattuso, G.; Pedullà, E.; Rapisarda, E.; Nicolosi, D.; Salmeri, M. Association of oral dysbiosis with oral cancer development. Oncol. Lett. 2020, 19, 3045–3058. [Google Scholar] [CrossRef] [PubMed]
  10. Pignatelli, P.; Curia, M.C.; Tenore, G.; Bondi, D.; Piattelli, A.; Romeo, U. Oral bacteriome and oral potentially malignant disorders: A systematic review of the associations. Arch. Oral Biol. 2024, 160, 105891. [Google Scholar] [CrossRef] [PubMed]
  11. Galler, K.M.; Weber, M.; Korkmaz, Y.; Widbiller, M.; Feuerer, M. Inflammatory response mechanisms of the dentine-pulp complex and the periapical tissues. Int. J. Mol. Sci. 2021, 22, 1480. [Google Scholar] [CrossRef] [PubMed]
  12. Neuhaus, K.W. Teeth: Malignant neoplasms in the dental pulp? Lancet Oncol. 2007, 8, 75–78. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, S.; Zhou, X.; Peng, X.; Li, M.; Ren, B.; Cheng, G.; Cheng, L. Porphyromonas gingivalis promotes immunoevasion of oral cancer by protecting cancer from macrophage attack. J. Immunol. 2020, 205, 282–289. [Google Scholar] [CrossRef] [PubMed]
  14. Park, S.R.; Kim, D.J.; Han, S.H.; Kang, M.J.; Lee, J.Y.; Jeong, Y.J.; Lee, S.J.; Kim, T.H.; Ahn, S.G.; Yoon, J.H.; et al. Diverse Toll-like receptors mediate cytokine production by Fusobacterium nucleatum and Aggregatibacter actinomycetemcomitans in macrophages. Infect. Immun. 2014, 82, 1914–1920. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, Q.; Sztukowska, M.; Ojo, A.; Scott, D.A.; Wang, H.; Lamont, R.J. FOXO responses to Porphyromonas gingivalis in epithelial cells. Cell Microbiol. 2015, 17, 1605–1617. [Google Scholar] [CrossRef] [PubMed]
  16. Ramage, G.; Lappin, D.F.; Millhouse, E.; Malcolm, J.; Jose, A.; Yang, J.; Bradshaw, D.J.; Pratten, J.R.; Culshaw, S. The epithelial cell response to health and disease associated oral biofilm models. J. Periodontal Res. 2017, 52, 325–333. [Google Scholar] [CrossRef] [PubMed]
  17. Milward, M.R.; Chapple, I.L.; Wright, H.J.; Millard, J.L.; Matthews, J.B.; Cooper, P.R. Differential activation of NF-kappaB and gene expression in oral epithelial cells by periodontal pathogens. Clin. Exp. Immunol. 2007, 148, 307–324. [Google Scholar] [CrossRef] [PubMed]
  18. Yee, M.; Kim, S.; Sethi, P.; Düzgüneş, N.; Konopka, K. Porphyromonas gingivalis stimulates IL-6 and IL-8 secretion in GMSM-K, HSC-3 and H413 oral epithelial cells. Anaerobe 2014, 28, 62–67. [Google Scholar] [CrossRef]
  19. Abdulkareem, A.A.; Shelton, R.M.; Landini, G.; Cooper, P.R.; Milward, M.R. Periodontal pathogens promote epithelial-mesenchymal transition in oral squamous carcinoma cells in vitro. Cell Adhes. Migr. 2018, 12, 127–137. [Google Scholar] [CrossRef] [PubMed]
  20. Aral, K.; Milward, M.R.; Gupta, D.; Cooper, P.R. Effects of Porphyromonas gingivalis and Fusobacterium nucleatum on inflammasomes and their regulators in H400 cells. Mol. Oral Microbiol. 2020, 35, 158–167. [Google Scholar] [CrossRef] [PubMed]
  21. Hung, S.C.; Huang, P.R.; Almeida-da-Silva, C.L.C.; Atanasova, K.R.; Yilmaz, O.; Ojcius, D.M. NLRX1 modulates differentially NLRP3 inflammasome activation and NF-κB signaling during Fusobacterium nucleatum infection. Microbes Infect. 2018, 20, 615–625. [Google Scholar] [CrossRef] [PubMed]
  22. Kang, W.; Ji, X.; Zhang, X.; Tang, D.; Feng, Q. Persistent exposure to Fusobacterium nucleatum triggers chemokine/cytokine release and inhibits the proliferation and osteogenic differentiation capabilities of human gingiva-derived mesenchymal stem cells. Front. Cell Infect. Microbiol. 2019, 9, 429. [Google Scholar] [CrossRef] [PubMed]
  23. Kang, W.; Jia, Z.; Tang, D.; Zhang, Z.; Gao, H.; He, K.; Feng, Q. Fusobacterium nucleatum Facilitates apoptosis, ROS generation, and inflammatory cytokine production by activating AKT/MAPK and NF-κB signaling pathways in human gingival fibroblasts. Oxid. Med. Cell Longev. 2019, 2019, 1681972. [Google Scholar] [CrossRef] [PubMed]
  24. Kurgan, Ş.; Kansal, S.; Nguyen, D.; Stephens, D.; Koroneos, Y.; Hasturk, H.; Van Dyke, T.E.; Kantarci, A. Strain-specific impact of Fusobacterium nucleatum on neutrophil Function. J. Periodontol. 2017, 88, 380–389. [Google Scholar] [CrossRef] [PubMed]
  25. Sztukowska, M.N.; Ojo, A.; Ahmed, S.; Carenbauer, A.L.; Wang, Q.; Shumway, B.; Jenkinson, H.F.; Wang, H.; Darling, D.S.; Lamont, R.J. Porphyromonas gingivalis initiates a mesenchymal-like transition through ZEB1 in gingival epithelial cells. Cell Microbiol. 2016, 180, 844–858. [Google Scholar] [CrossRef] [PubMed]
  26. Lee, J.; Roberts, J.S.; Atanasova, K.R.; Chowdhury, N.; Han, K.; Yilmaz, Ö. Human primary epithelial cells acquire an epithelial-mesenchymal-transition phenotype during long-term infection by the oral opportunistic pathogen, Porphyromonas gingivalis. Front. Cell Infect. Microbiol. 2017, 7, 493. [Google Scholar] [CrossRef] [PubMed]
  27. Abdulkareem, A.A.; Shelton, R.M.; Landini, G.; Cooper, P.R.; Milward, M.R. Potential role of periodontal pathogens in compromising epithelial barrier function by inducing epithelial-mesenchymal transition. J. Periodontal Res. 2018, 53, 565–574. [Google Scholar] [CrossRef] [PubMed]
  28. Nguyen, L.H.; Ma, W.; Wang, D.D.; Cao, Y.; Mallick, H.; Gerbaba, T.K.; Lloyd-Price, J.; Abu-Ali, G.; Hall, A.B.; Sikavi, D.; et al. Association between sulfur-metabolizing bacterial communities in stool and risk of distal colorectal cancer in men. Gastroenterology 2020, 158, 1313–1325. [Google Scholar] [CrossRef] [PubMed]
  29. Ma, Z.; Bi, Q.; Wang, Y. Hydrogen sulfide accelerates cell cycle progression in oral squamous cell carcinoma cell lines. Oral Dis. 2015, 21, 156–162. [Google Scholar] [CrossRef] [PubMed]
  30. Vital, M.; Howe, A.C.; Tiedje, J.M. Revealing the bacterial butyrate synthesis pathways by analyzing (meta)genomic data. mBio 2014, 5, e00889. [Google Scholar] [CrossRef] [PubMed]
  31. Zhang, S.; Bian, H.; Li, X.; Wu, H.; Bi, Q.; Yan, Y.; Wang, Y. Hydrogen sulfide promotes cell proliferation of oral cancer through activation of the COX2/AKT/ERK1/2 axis. Oncol. Rep. 2016, 35, 2825–2832. [Google Scholar] [CrossRef] [PubMed]
  32. Kuboniwa, M.; Hasegawa, Y.; Mao, S.; Shizukuishi, S.; Amano, A.; Lamont, R.J.; Yilmaz, O. P. gingivalis accelerates gingival epithelial cell progression through the cell cycle. Microbes Infect. 2008, 10, 122–128. [Google Scholar] [CrossRef] [PubMed]
  33. Chang, C.; Wang, H.; Liu, J.; Pan, C.; Zhang, D.; Li, X.; Pan, Y. Porphyromonas gingivalis infection promoted the proliferation of oral squamous cell carcinoma cells through the miR-21/PDCD4/AP-1 negative signaling pathway. ACS Infect. Dis. 2019, 5, 1336–1347. [Google Scholar] [CrossRef] [PubMed]
  34. Pan, C.; Xu, X.; Tan, L.; Lin, L.; Pan, Y. The effects of Porphyromonas gingivalis on the cell cycle progression of human gingival epithelial cells. Oral Dis. 2014, 20, 100–108. [Google Scholar] [CrossRef] [PubMed]
  35. Geng, F.; Zhang, Y.; Lu, Z.; Zhang, S.; Pan, Y. Fusobacterium nucleatum Caused DNA Damage and Promoted Cell Proliferation by the Ku70/p53 Pathway in Oral Cancer Cells. DNA Cell Biol. 2020, 39, 144–151. [Google Scholar] [CrossRef]
  36. Binder Gallimidi, A.; Fischman, S.; Revach, B.; Bulvik, R.; Maliutina, A.; Rubinstein, A.M.; Nussbaum, G.; Elkin, M. Periodontal pathogens Porphyromonas gingivalis and Fusobacterium nucleatum promote tumor progression in an oral-specific chemical carcinogenesis model. Oncotarget 2015, 6, 22613–22623. [Google Scholar] [CrossRef] [PubMed]
  37. Gao, S.; Liu, Y.; Duan, X.; Liu, K.; Mohammed, M.; Gu, Z.; Ren, J.; Yakoumatos, L.; Yuan, X.; Lu, L.; et al. Porphyromonas gingivalis infection exacerbates oesophageal cancer and promotes resistance to neoadjuvant chemotherapy. Br. J. Cancer 2021, 125, 433–444. [Google Scholar] [CrossRef] [PubMed]
  38. Lee, J.; Roberts, J.S.; Atanasova, K.R.; Chowdhury, N.; Yilmaz, Ö. A novel kinase function of a nucleoside-diphosphate-kinase homologue in Porphyromonas gingivalis is critical in subversion of host cell apoptosis by targeting heat-shock protein 27. Cell Microbiol. 2018, 20, e12825. [Google Scholar] [CrossRef] [PubMed]
  39. Yilmaz, O.; Jungas, T.; Verbeke, P.; Ojcius, D.M. Activation of the phosphatidylinositol 3-kinase/Akt pathway contributes to survival of primary epithelial cells infected with the periodontal pathogen Porphyromonas gingivalis. Infect. Immun. 2004, 72, 3743–3751. [Google Scholar] [CrossRef] [PubMed]
  40. Yao, L.; Jermanus, C.; Barbetta, B.; Choi, C.; Verbeke, P.; Ojcius, D.M.; Yilmaz, O. Porphyromonas gingivalis infection sequesters pro-apoptotic Bad through Akt in primary gingival epithelial cells. Mol. Oral Microbiol. 2010, 25, 89–101. [Google Scholar] [CrossRef]
  41. Nakhjiri, S.F.; Park, Y.; Yilmaz, O.; Chung, W.O.; Watanabe, K.; El-Sabaeny, A.; Park, K.; Lamont, R.J. Inhibition of epithelial cell apoptosis by Porphyromonas gingivalis. FEMS Microbiol. Lett. 2001, 200, 145–149. [Google Scholar] [CrossRef] [PubMed]
  42. Hoppe, T.; Kraus, D.; Probstmeier, R.; Jepsen, S.; Winter, J. Stimulation with Porphyromonas gingivalis enhances malignancy and initiates anoikis resistance in immortalized oral keratinocytes. J. Cell Physiol. 2019, 234, 21903–21914. [Google Scholar] [CrossRef] [PubMed]
  43. Li, J.; Li, L.; Wang, X.; Xiao, L. Porphyromonas gingivalis inhibition of microRNA-205-5p expression modulates proinflammatory cytokines in gingival epithelial cells. Biochem. Genet. 2020, 58, 566–579. [Google Scholar] [CrossRef] [PubMed]
  44. Da, J.; Wang, X.; Li, L.; Xu, Y. Fusobacterium nucleatum Promotes Cisplatin-Resistance and Migration of Oral Squamous Carcinoma Cells by Up-Regulating Wnt5a-Mediated NFATc3 Expression. Tohoku J. Exp. Med. 2021, 253, 249–259. [Google Scholar] [CrossRef] [PubMed]
  45. Rath-Deschner, B.; Nogueira, A.V.B.; Memmert, S.; Nokhbehsaim, M.; Augusto Cirelli, J.; Eick, S.; Miosge, N.; Kirschneck, C.; Kesting, M.; Deschner, J.; et al. Regulation of anti-apoptotic SOD2 and BIRC3 in periodontal cells and tissues. Int. J. Mol. Sci. 2021, 22, 591. [Google Scholar] [CrossRef] [PubMed]
  46. Duan, C.; Tang, X.; Wang, W.; Qian, W.; Fu, X.; Deng, X.; Han, C.; Hou, X. L-fucose ameliorates the carcinogenic properties of Fusobacterium nucleatum in colorectal cancer. Oncol. Lett. 2021, 21, 143. [Google Scholar] [CrossRef] [PubMed]
  47. Shen, S.; Sun, T.; Ding, X.; Gu, X.; Wang, Y.; Ma, X.; Li, Z.; Gao, H.; Ge, S.; Feng, Q. The exoprotein Gbp of Fusobacterium nucleatum promotes THP-1 cell lipid deposition by binding to CypA and activating PI3K-AKT/MAPK/NF-κB pathways. J. Adv. Res. 2024, 57, 93–105. [Google Scholar] [CrossRef] [PubMed]
  48. Inaba, H.; Sugita, H.; Kuboniwa, M.; Iwai, S.; Hamada, M.; Noda, T.; Morisaki, I.; Lamont, R.J.; Amano, A. Porphyromonas gingivalis promotes invasion of oral squamous cell carcinoma through induction of proMMP9 and its activation. Cell Microbiol. 2014, 16, 131–145. [Google Scholar] [CrossRef]
  49. Inaba, H.; Amano, A.; Lamont, R.J.; Murakami, Y. Involvement of protease-activated receptor 4 in over-expression of matrix metalloproteinase 9 induced by Porphyromonas gingivalis. Med. Microbiol. Immunol. 2015, 204, 605–612. [Google Scholar] [CrossRef]
  50. Ha, N.H.; Park, D.G.; Woo, B.H.; Kim, D.J.; Choi, J.I.; Park, B.S.; Kim, Y.D.; Lee, J.H.; Park, H.R. Porphyromonas gingivalis increases the invasiveness of oral cancer cells by upregulating IL-8 and MMPs. Cytokine 2016, 86, 64–72. [Google Scholar] [CrossRef] [PubMed]
  51. Meng, F.; Li, R.; Ma, L.; Liu, L.; Lai, X.; Yang, D.; Wei, J.; Ma, D.; Li, Z. Porphyromonas gingivalis promotes the motility of esophageal squamous cell carcinoma by activating NF-κB signaling pathway. Microbes Infect. 2019, 21, 296–304. [Google Scholar] [CrossRef] [PubMed]
  52. Harrandah, A.M.; Chukkapalli, S.S.; Bhattacharyya, I.; Progulske-Fox, A.; Chan, E.K.L. Fusobacteria modulate oral carcinogenesis and promote cancer progression. J. Oral Microbiol. 2020, 13, 1849493. [Google Scholar] [CrossRef] [PubMed]
  53. Kamarajan, P.; Ateia, I.; Shin, J.M.; Fenno, J.C.; Le, C.; Zhan, L.; Chang, A.; Darveau, R.; Kapila, Y.L. Periodontal pathogens promote cancer aggressivity via TLR/MyD88 triggered activation of Integrin/FAK signaling that is therapeutically reversible by a probiotic bacteriocin. PLoS Pathog. 2020, 16, e1008881. [Google Scholar] [CrossRef]
  54. Uitto, V.J.; Baillie, D.; Wu, Q.; Gendron, R.; Grenier, D.; Putnins, E.E.; Kanervo, A.; Firth, J.D. Fusobacterium nucleatum increases collagenase 3 production and migration of epithelial cells. Infect. Immun. 2005, 73, 1171–1179. [Google Scholar] [CrossRef] [PubMed]
  55. Bachrach, G.; Rosen, G.; Bellalou, M.; Naor, R.; Sela, M.N. Identification of a Fusobacterium nucleatum 65 kDa serine protease. Oral Microbiol. Immunol. 2004, 19, 155–159. [Google Scholar] [CrossRef] [PubMed]
  56. Arjunan, P.; Meghil, M.M.; Pi, W.; Xu, J.; Lang, L.; El-Awady, A.; Sullivan, W.; Rajendran, M.; Rabelo, M.S.; Wang, T.; et al. Oral pathobiont activates anti-apoptotic pathway, promoting both immune suppression and oncogenic cell proliferation. Sci. Rep. 2018, 8, 16607. [Google Scholar] [CrossRef] [PubMed]
  57. Groeger, S.; Domann, E.; Gonzales, J.R.; Chakraborty, T.; Meyle, J. B7-H1 and B7-DC receptors of oral squamous carcinoma cells are upregulated by Porphyromonas gingivalis. Immunobiology 2011, 216, 1302–1310. [Google Scholar] [CrossRef] [PubMed]
  58. 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] [PubMed]
  59. Gur, C.; Ibrahim, Y.; Isaacson, B.; Yamin, R.; Abed, J.; Gamliel, M.; Enk, J.; Bar-On, Y.; Stanietsky-Kaynan, N.; Coppenhagen-Glazer, S.; 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. [Google Scholar] [CrossRef] [PubMed]
  60. Gur, C.; Maalouf, N.; Shhadeh, A.; Berhani, O.; Singer, B.B.; Bachrach, G.; Mandelboim, O. Fusobacterium nucleatum supresses anti-tumor immunity by activating CEACAM1. OncoImmunology 2019, 8, e1581531. [Google Scholar] [CrossRef] [PubMed]
  61. Lin, F.Y.; Huang, C.Y.; Lu, H.Y.; Shih, C.M.; Tsao, N.W.; Shyue, S.K.; Lin, C.Y.; Chang, Y.J.; Tsai, C.S.; Lin, Y.W.; et al. The GroEL protein of Porphyromonas gingivalis accelerates tumor growth by enhancing endothelial progenitor cell function and neovascularization. Mol. Oral Microbiol. 2015, 30, 198–216. [Google Scholar] [CrossRef] [PubMed]
  62. Li, Z.; Liu, Y.; Guo, P.; Wei, Y. Construction and validation of a novel angiogenesis pattern to predict prognosis and immunotherapy efficacy in colorectal cancer. Aging 2023, 15, 12413–12450. [Google Scholar] [CrossRef] [PubMed]
  63. Ha, N.H.; Woo, B.H.; Kim, D.J.; Ha, E.S.; Choi, J.I.; Kim, S.J.; Park, B.S.; Lee, J.H.; Park, H.R. Prolonged and repetitive exposure to Porphyromonas gingivalis increases aggressiveness of oral cancer cells by promoting acquisition of cancer stem cell properties. Tumour Biol. 2015, 36, 9947–9960. [Google Scholar] [CrossRef] [PubMed]
  64. Kang, W.; Sun, T.; Tang, D.; Zhou, J.; Feng, Q. Time-Course Transcriptome Analysis of Gingiva-Derived Mesenchymal Stem Cells Reveals That Fusobacterium nucleatum Triggers Oncogene Expression in the Process of Cell Differentiation. Front. Cell Dev. Biol. 2020, 7, 359. [Google Scholar] [CrossRef] [PubMed]
  65. Barillari, G.; Melaiu, O.; Gargari, M.; Pomella, S.; Bei, R.; Campanella, V. The multiple roles of CD147 in the development and progression of oral squamous cell carcinoma: An overview. Int. J. Mol. Sci. 2022, 23, 8336. [Google Scholar] [CrossRef] [PubMed]
  66. Wenghoefer, M.; Pantelis, A.; Najafi, T.; Deschner, J.; Allam, J.P.; Novak, N.; Reich, R.; Martini, M.; Bergé, S.; Fischer, H.P.; et al. Gene expression of oncogenes, antimicrobial peptides, and cytokines in the development of oral leukoplakia. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2010, 110, 351–356. [Google Scholar] [CrossRef] [PubMed]
  67. Ling, Z.; Cheng, B.; Tao, X. Epithelial-to-mesenchymal transition in oral squamous cell carcinoma: Challenges and opportunities. Int. J. Cancer 2021, 148, 1548–1561. [Google Scholar] [CrossRef] [PubMed]
  68. Kim, S.R.; Lee, E.Y.; Kim, D.J.; Kim, H.J.; Park, H.R. Quercetin inhibits cell survival and metastatic ability via the EMT-mediated pathway in oral squamous cell carcinoma. Molecules 2020, 25, 757. [Google Scholar] [CrossRef] [PubMed]
  69. Fang, Y.; Yang, Y.; Liu, C. Evolutionary relationships between dysregulated genes in oral squamous cell carcinoma and oral microbiota. Front. Cell Infect. Microbiol. 2022, 12, 931011. [Google Scholar] [CrossRef]
  70. Liu, M.; Liu, Q.; Fan, S.; Su, F.; Jiang, C.; Cai, G.; Wang, Y.; Liao, G.; Lei, X.; Chen, W.; et al. LncRNA LTSCCAT promotes tongue squamous cell carcinoma metastasis via targeting the miR-103a-2-5p/SMYD3/TWIST1 axis. Cell Death Dis. 2021, 12, 144. [Google Scholar] [CrossRef]
  71. Lu, X.; Xu, Q.; Tong, Y.; Zhang, Z.; Dun, G.; Feng, Y.; Tang, J.; Han, D.; Mao, Y.; Deng, L.; et al. Long non-coding RNA EVADR induced by Fusobacterium nucleatum infection promotes colorectal cancer metastasis. Cell Rep. 2022, 40, 111127. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, S.; Li, C.; Liu, J.; Geng, F.; Shi, X.; Li, Q.; Lu, Z.; Pan, Y. Fusobacterium nucleatum promotes epithelial-mesenchymal transiton through regulation of the lncRNA MIR4435-2HG/miR-296-5p/Akt2/SNAI1 signaling pathway. FEBS J. 2020, 287, 4032–4047. [Google Scholar] [CrossRef] [PubMed]
  73. Lee, C.H.; Chang, J.S.; Syu, S.H.; Wong, T.S.; Chan, J.Y.; Tang, Y.C.; Yang, Z.P.; Yang, W.C.; Chen, C.T.; Lu, S.C.; et al. IL-1β promotes malignant transformation and tumor aggressiveness in oral cancer. J. Cell Physiol. 2015, 230, 875–884. [Google Scholar] [CrossRef] [PubMed]
  74. Kim, D.; Illeperuma, R.P.; Kim, J. The Protective Effect of Antioxidants in Areca Nut Extract-Induced Oral Carcinogenesis. Asian Pac. J. Cancer Prev. 2020, 21, 2447–2452. [Google Scholar] [CrossRef] [PubMed]
  75. Kobayashi, R.; Hoshikawa, E.; Saito, T.; Suebsamarn, O.; Naito, E.; Suzuki, A.; Ishihara, S.; Haga, H.; Tomihara, K.; Izumi, K. The EGF/EGFR axis and its downstream signaling pathways regulate the motility and proliferation of cultured oral keratinocytes. FEBS Open Bio 2023, 13, 1469–1484. [Google Scholar] [CrossRef] [PubMed]
  76. Mohtasham, N.; Babakoohi, S.; Salehinejad, J.; Montaser-Kouhsari, L.; Shakeri, M.T.; Shojaee, S.; Sistani, N.S.; Firooz, A. Mast cell density and angiogenesis in oral dysplastic epithelium and low- and high-grade oral squamous cell carcinoma. Acta Odontol. Scand. 2010, 68, 300–304. [Google Scholar] [CrossRef]
  77. Bai, Y.; Sha, J.; Kanno, T. The role of carcinogenesis-related biomarkers in the Wnt pathway and their effects on epithelial-mesenchymal transition (EMT) in oral squamous cell carcinoma. Cancers 2020, 12, 555. [Google Scholar] [CrossRef]
  78. Shigeishi, H. Association between human papillomavirus and oral cancer: A literature review. Int. J. Clin. Oncol. 2023, 28, 982–989. [Google Scholar] [CrossRef] [PubMed]
  79. Scheurer, M.J.J.; Seher, A.; Steinacker, V.; Linz, C.; Hartmann, S.; Kübler, A.C.; Müller-Richter, U.D.A.; Brands, R.C. Targeting inhibitors of apoptosis in oral squamous cell carcinoma in vitro. J. Craniomaxillofac Surg. 2019, 47, 1589–1599. [Google Scholar] [CrossRef]
  80. Mao, S.; Park, Y.; Hasegawa, Y.; Tribble, G.D.; James, C.E.; Handfield, M.; Stavropoulos, M.F.; Yilmaz, O.; Lamont, R.J. Intrinsic apoptotic pathways of gingival epithelial cells modulated by Porphyromonas gingivalis. Cell Microbiol. 2007, 9, 1997–2007. [Google Scholar] [CrossRef]
  81. Ravichandran, K.; Tyagi, A.; Deep, G.; Agarwal, C.; Agarwal, R. Interleukin-1beta-induced iNOS expression in human lung carcinoma A549 cells: Involvement of STAT and MAPK pathways. Indian J. Exp. Biol. 2011, 49, 840–847. [Google Scholar] [PubMed]
  82. Liu, C.; Zhou, J.; Zhang, S.; Fu, J.; Li, Y.; Hao, Y.; Yuan, J.; Tang, F.; Ge, W.; He, H.; et al. Mesenchymal stem cells-derived IL-6 promotes invasion and metastasis of oral squamous cell carcinoma via JAK-STAT3 signalling. Oral Dis. 2023, 1–13. [Google Scholar] [CrossRef] [PubMed]
  83. Wei, L.Y.; Lin, H.C.; Tsai, F.C.; Ko, J.Y.; Kok, S.H.; Cheng, S.J.; Lee, J.J.; Chia, J.S. Effects of Interleukin-6 on STAT3-regulated signaling in oral cancer and as a prognosticator of patient survival. Oral Oncol. 2022, 124, 105665. [Google Scholar] [CrossRef] [PubMed]
  84. Jin, G.; Yang, Y.; Liu, K.; Zhao, J.; Chen, X.; Liu, H.; Bai, R.; Li, X.; Jiang, Y.; Zhang, X.; et al. Combination curcumin and (-)-epigallocatechin-3-gallate inhibits colorectal carcinoma microenvironment-induced angiogenesis by JAK/STAT3/IL-8 pathway. Oncogenesis 2017, 6, e384. [Google Scholar] [CrossRef]
  85. Mi, C.; Ma, J.; Wang, K.S.; Wang, Z.; Li, M.Y.; Li, J.B.; Li, X.; Piao, L.X.; Xu, G.H.; Jin, X. Amorfrutin A inhibits TNF-α induced JAK/STAT signaling, cell survival and proliferation of human cancer cells. Immunopharmacol. Immunotoxicol. 2017, 39, 338–347. [Google Scholar] [CrossRef] [PubMed]
  86. Duan, M.; Du, X.; Ren, G.; Zhang, Y.; Zheng, Y.; Sun, S.; Zhang, J. Obovatol inhibits the growth and aggressiveness of tongue squamous cell carcinoma through regulation of the EGF-mediated JAK-STAT signaling pathway. Mol. Med. Rep. 2018, 18, 1651–1659. [Google Scholar] [CrossRef]
  87. Liu, R.Y.; Zeng, Y.; Lei, Z.; Wang, L.; Yang, H.; Liu, Z.; Zhao, J.; Zhang, H.T. JAK/STAT3 signaling is required for TGF-β-induced epithelial-mesenchymal transition in lung cancer cells. Int. J. Oncol. 2014, 44, 1643–1651. [Google Scholar] [CrossRef] [PubMed]
  88. Hu, Y.; Dong, Z.; Liu, K. Unraveling the complexity of STAT3 in cancer: Molecular understanding and drug discovery. J. Exp. Clin. Cancer Res. 2024, 43, 23. [Google Scholar] [CrossRef]
  89. Jill, N.; Bhootra, S.; Kannanthodi, S.; Shanmugam, G.; Rakshit, S.; Rajak, R.; Thakkar, V.; Sarkar, K. Interplay between signal transducers and activators of transcription (STAT) proteins and cancer: Involvement, therapeutic and prognostic perspective. Clin. Exp. Med. 2023, 23, 4323–4339. [Google Scholar] [CrossRef]
  90. Jiang, M.; Li, B. STAT3 and Its Targeting Inhibitors in Oral Squamous Cell Carcinoma. Cells 2022, 11, 3131. [Google Scholar] [CrossRef] [PubMed]
  91. Weber, M.; Iliopoulos, C.; Moebius, P.; Büttner-Herold, M.; Amann, K.; Ries, J.; Preidl, R.; Neukam, F.W.; Wehrhan, F. Prognostic significance of macrophage polarization in early stage oral squamous cell carcinomas. Oral Oncol. 2016, 52, 75–84. [Google Scholar] [CrossRef] [PubMed]
  92. Ding, L.; Fu, Y.; Zhu, N.; Zhao, M.; Ding, Z.; Zhang, X.; Song, Y.; Jing, Y.; Zhang, Q.; Chen, S.; et al. OXTRHigh stroma fibroblasts control the invasion pattern of oral squamous cell carcinoma via ERK5 signaling. Nat. Commun. 2022, 13, 5124. [Google Scholar] [CrossRef] [PubMed]
  93. Yang, J.; Wei, D.; Wang, W.; Shen, B.; Xu, S.; Cao, Y. TRAF4 enhances oral squamous cell carcinoma cell growth, invasion and migration by Wnt-β-catenin signaling pathway. Int. J. Clin. Exp. Pathol. 2015, 8, 11837–11846. [Google Scholar]
  94. Mishev, G.; Deliverska, E.; Hlushchuk, R.; Velinov, N.; Aebersold, D.; Weinstein, F.; Djonov, V. Prognostic value of matrix metalloproteinases in oral squamous cell carcinoma. Biotechnol. Biotechnol. Equip. 2014, 28, 1138–1149. [Google Scholar] [CrossRef] [PubMed]
  95. Abe, K. Butyric acid induces apoptosis in both human monocytes and lymphocytes equivalently. J. Oral Sci. 2012, 54, 7–14. [Google Scholar] [CrossRef] [PubMed]
  96. Zang, W.; Liu, J.; Geng, F.; Liu, D.; Zhang, S.; Li, Y.; Pan, Y. Butyrate promotes oral squamous cell carcinoma cells migration, invasion and epithelial-mesenchymal transition. Peer J. 2022, 10, e12991. [Google Scholar] [CrossRef]
  97. Goswami, P.R.; Singh, G. Perineural Invasion (PNI) Definition, histopathological parameters of PNI in oral squamous cell carcinoma with molecular insight and prognostic significance. Cureus 2023, 15, e40165. [Google Scholar] [CrossRef] [PubMed]
  98. Joseph, J.P.; Harishankar, M.K.; Pillai, A.A.; Devi, A. Hypoxia induced EMT: A review on the mechanism of tumor progression and metastasis in OSCC. Oral Oncol. 2018, 80, 23–32. [Google Scholar] [CrossRef] [PubMed]
  99. Mărgăritescu, C.; Pirici, D.; Stîngă, A.; Simionescu, C.; Raica, M.; Mogoantă, L.; Stepan, A.; Ribatti, D. VEGF expression and angiogenesis in oral squamous cell carcinoma: An immunohistochemical and morphometric study. Clin. Exp. Med. 2010, 10, 209–214. [Google Scholar] [CrossRef]
  100. Xu, C.; Li, X.; Guo, P.; Wang, J. Hypoxia-induced activation of JAK/STAT3 signaling pathway promotes trophoblast cell viability and angiogenesis in preeclampsia. Med. Sci. Monit. 2017, 23, 4909–4917. [Google Scholar] [CrossRef] [PubMed]
  101. Yang, M.; Wang, L.; Wang, X.; Wang, X.; Yang, Z.; Li, J. IL-6 promotes FSH-induced VEGF expression through JAK/STAT3 signaling pathway in bovine granulosa cells. Cell Physiol. Biochem. 2017, 44, 293–302. [Google Scholar] [CrossRef] [PubMed]
  102. Mendes, R.T.; Nguyen, D.; Stephens, D.; Pamuk, F.; Fernandes, D.; Van Dyke, T.E.; Kantarci, A. Endothelial Cell Response to Fusobacterium nucleatum. Infect. Immun. 2016, 84, 2141–2148. [Google Scholar] [CrossRef] [PubMed]
  103. Lu, M.Y.; Liao, Y.W.; Chen, P.Y.; Hsieh, P.L.; Fang, C.Y.; Wu, C.Y.; Yen, M.L.; Peng, B.Y.; Wang, D.P.; Cheng, H.C.; et al. Targeting LncRNA HOTAIR suppresses cancer stemness and metastasis in oral carcinomas stem cells through modulation of EMT. Oncotarget 2017, 8, 98542–98552. [Google Scholar] [CrossRef] [PubMed]
  104. Mărgăritescu, C.; Pirici, D.; Simionescu, C.; Stepan, A. The utility of CD44, CD117 and CD133 in identification of cancer stem cells (CSC) in oral squamous cell carcinomas (OSCC). Rom. J. Morphol. Embryol. 2011, 52, 985–993. [Google Scholar] [PubMed]
  105. Patel, S.; Shah, K.; Mirza, S.; Daga, A.; Rawal, R. Epigenetic regulators governing cancer stem cells and epithelial-mesenchymal transition in oral squamous cell carcinoma. Curr. Stem Cell Res. Ther. 2015, 10, 140–152. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, H.; Du, J.; Chao, S.; Li, S.; Cai, H.; Zhang, H.; Chen, G.; Liu, P.; Bu, P. Fusobacterium nucleatum promotes colorectal cancer cell to acquire stem cell-like features by manipulating lipid droplet-mediated numb degradation. Adv. Sci. 2022, 9, e2105222. [Google Scholar] [CrossRef]
  107. Kupferman, M.E.; Patel, V.; Sriuranpong, V.; Amornphimoltham, P.; Jasser, S.A.; Mandal, M.; Zhou, G.; Wang, J.; Coombes, K.; Multani, A.; et al. Molecular analysis of anoikis resistance in oral cavity squamous cell carcinoma. Oral Oncol. 2007, 43, 440–454. [Google Scholar] [CrossRef] [PubMed]
  108. Kaseb, H.O.; Lewis, D.W.; Saunders, W.S.; Gollin, S.M. Cell division patterns and chromosomal segregation defects in oral cancer stem cells. Genes Chromosomes Cancer 2016, 55, 694–709. [Google Scholar] [CrossRef] [PubMed]
  109. Notarstefano, V.; Belloni, A.; Sabbatini, S.; Pro, C.; Orilisi, G.; Monterubbianesi, R.; Tosco, V.; Byrne, H.J.; Vaccari, L.; Giorgini, E. Cytotoxic effects of 5-azacytidine on primary tumour cells and cancer stem cells from oral squamous cell carcinoma: An in vitro FTIRM analysis. Cells 2021, 10, 2127. [Google Scholar] [CrossRef]
  110. Biddle, A.; Gammon, L.; Liang, X.; Costea, D.E.; Mackenzie, I.C. Phenotypic plasticity determines cancer stem cell therapeutic resistance in oral squamous cell carcinoma. EBioMedicine 2016, 4, 138–145. [Google Scholar] [CrossRef] [PubMed]
  111. Wong, J.; Manoil, D.; Näsman, P.; Belibasakis, G.N.; Neelakantan, P. Microbiological aspects of root canal infections and disinfection strategies: An update review on the current knowledge and challenges. Front. Oral Health 2021, 2, 672887. [Google Scholar] [CrossRef]
  112. Nair, P.N. Pathogenesis of apical periodontitis and the causes of endodontic failures. Crit. Rev. Oral Biol. Med. 2004, 15, 348–381. [Google Scholar] [CrossRef] [PubMed]
  113. Lee, L.W.; Lee, Y.L.; Hsiao, S.H.; Lin, H.P. Bacteria in the apical root canals of teeth with apical periodontitis. J. Formos. Med. Assoc. 2017, 116, 448–456. [Google Scholar] [CrossRef] [PubMed]
  114. Shetty, A.; Anand, K.V.; Rai, N.; Shetty, A.; Pradeep, K.; Kumar, A.A. In vitro comparative study of antimicrobial efficacy of endodontic sealers against common pathogens in the dental pulp. J. Conserv. Dent. 2023, 26, 216–220. [Google Scholar] [PubMed]
  115. Horst, O.V.; Tompkins, K.A.; Coats, S.R.; Braham, P.H.; Darveau, R.P.; Dale, B.A. TGF-beta1 Inhibits TLR-mediated odontoblast responses to oral bacteria. J. Dent. Res. 2009, 88, 333–338. [Google Scholar] [CrossRef] [PubMed]
  116. Lan, C.; Chen, S.; Jiang, S.; Lei, H.; Cai, Z.; Huang, X. Different expression patterns of inflammatory cytokines induced by lipopolysaccharides from Escherichia coli or Porphyromonas gingivalis in human dental pulp stem cells. BMC Oral Health 2022, 22, 121. [Google Scholar] [CrossRef] [PubMed]
  117. Zhang, W.; Xu, T.; Li, X.; Zhang, Y.; Zou, X.; Chen, F.; Yue, L. Single-cell atlas of dental pulp stem cells exposed to the oral bacteria Porphyromonas gingivalis and Enterococcus faecalis. Front. Cell Dev. Biol. 2023, 11, 1166934. [Google Scholar] [CrossRef]
  118. Rothermund, K.; Calabrese, T.C.; Syed-Picard, F.N. Differential effects of Escherichia coli- versus Porphyromonas gingivalis-derived lipopolysaccharides on dental pulp stem cell differentiation in scaffold-free engineered tissues. J. Endod. 2022, 48, 1378–1386.e2. [Google Scholar] [CrossRef] [PubMed]
  119. Qin, Z.; Fang, Z.; Zhao, L.; Chen, J.; Li, Y.; Liu, G. High dose of TNF-α suppressed osteogenic differentiation of human dental pulp stem cells by activating the Wnt/β-catenin signaling. J. Mol. Histol. 2015, 46, 409–420. [Google Scholar] [CrossRef] [PubMed]
  120. Sonmez Kaplan, S.; Sazak Ovecoglu, H.; Genc, D.; Akkoc, T. TNF-α, IL-1B and IL-6 affect the differentiation ability of dental pulp stem cells. BMC Oral Health 2023, 23, 555. [Google Scholar] [CrossRef] [PubMed]
  121. Li, M.; Wang, Y.; Xue, J.; Xu, Q.; Zhang, Y.; Liu, J.; Xu, H.; Guan, Z.; Bian, C.; Zhang, G.; et al. Baicalin can enhance odonto/osteogenic differentiation of inflammatory dental pulp stem cells by inhibiting the NF-κB and β-catenin/Wnt signaling pathways. Mol. Biol. Rep. 2023, 50, 4435–4446. [Google Scholar] [CrossRef] [PubMed]
  122. Luzuriaga, J.; Pastor-Alonso, O.; Encinas, J.M.; Unda, F.; Ibarretxe, G.; Pineda, J.R. Human dental pulp stem cells grown in neurogenic media differentiate into endothelial cells and promote neovasculogenesis in the mouse brain. Front. Physiol. 2019, 10, 347. [Google Scholar] [CrossRef]
  123. Xie, Z.; Shen, Z.; Zhan, P.; Yang, J.; Huang, Q.; Huang, S.; Chen, L.; Lin, Z. Functional dental pulp regeneration: Basic research and clinical translation. Int. J. Mol. Sci. 2021, 22, 8991. [Google Scholar] [CrossRef] [PubMed]
  124. Raj, A.T.; Kheur, S.; Khurshid, Z.; Sayed, M.E.; Mugri, M.H.; Almasri, M.A.; Al-Ahmari, M.M.; Patil, V.R.; Bhandi, S.; Testarelli, L.; et al. The growth factors and cytokines of dental pulp mesenchymal stem cell secretome may potentially aid in oral cancer proliferation. Molecules 2021, 26, 5683. [Google Scholar] [CrossRef]
  125. Mojtahedi, H.; Hossein-Khannazer, N.; Mahmoud Hashemi, S.; Masoudnia, M.; Askarzadeh, M.; Khojasteh, A.; Sattari, M. Effects of lipopolysaccharide from Porphyromonas gingivalis and Escherichia coli on gene expression levels of Toll-like receptors and inflammatory cytokines in human dental pulp stem cells. Iran. J. Immunol. 2022, 19, 299–310. [Google Scholar] [CrossRef] [PubMed]
  126. Wang, Y.; Wang, L.; Sun, T.; Shen, S.; Li, Z.; Ma, X.; Gu, X.; Zhang, X.; Peng, A.; Xu, X.; et al. Study of the inflammatory activating process in the early stage of Fusobacterium nucleatum infected PDLSCs. Int. J. Oral Sci. 2023, 15, 8. [Google Scholar] [CrossRef] [PubMed]
  127. Ko, Y.J.; Kwon, K.Y.; Kum, K.Y.; Lee, W.C.; Baek, S.H.; Kang, M.K.; Shon, W.J. The anti-inflammatory effect of human telomerase-derived peptide on P. gingivalis lipopolysaccharide-induced inflammatory cytokine production and its mechanism in human dental pulp cells. Mediators Inflamm. 2015, 2015, 385127. [Google Scholar] [CrossRef] [PubMed]
  128. Nicolas, S.; Abdellatef, S.; Haddad, M.A.; Fakhoury, I.; El-Sibai, M. Hypoxia and EGF stimulation regulate vegf expression in human glioblastoma multiforme (GBM) cells by differential regulation of the PI3K/Rho-GTPase and MAPK pathways. Cells 2019, 8, 1397. [Google Scholar] [CrossRef] [PubMed]
  129. Breitkopf, K.; Sawitza, I.; Gressner, A.M. Characterization of intracellular pathways leading to coinduction of thrombospondin-1 and TGF-beta1 expression in rat hepatic stellate cells. Growth Factors 2005, 23, 77–85. [Google Scholar] [CrossRef]
  130. Catar, R.; Witowski, J.; Wagner, P.; Annett Schramm, I.; Kawka, E.; Philippe, A.; Dragun, D.; Jörres, A. The proto-oncogene c-Fos transcriptionally regulates VEGF production during peritoneal inflammation. Kidney Int. 2013, 84, 1119–1128. [Google Scholar] [CrossRef]
  131. Takizawa, H.; Araki, Y.; Fujishiro, M.; Tomita, S.; Kishikawa, S.; Hashizume, A.; Mitsumori, T.; Nitta, H.; Iizuka-Honma, H.; Sawada, T.; et al. Role of TGF-beta1 and TNF-alpha1 produced by neoplastic cells in the pathogenesis of fibrosis in patients with hematologic neoplasms. J. Clin. Exp. Hematop. 2023, 63, 83–89. [Google Scholar] [CrossRef] [PubMed]
  132. Lai, K.C.; Liu, C.J.; Lin, T.J.; Mar, A.C.; Wang, H.H.; Chen, C.W.; Hong, Z.X.; Lee, T.C. Blocking TNF-α inhibits angiogenesis and growth of IFIT2-depleted metastatic oral squamous cell carcinoma cells. Cancer Lett. 2016, 370, 207–215. [Google Scholar] [CrossRef] [PubMed]
  133. Ma, P.; He, M.; Lian, H.; Li, J.; Gao, Y.; Wu, J.; Men, K.; Men, Y.; Li, C. Systemic and local administration of a dual-siRNA complex efficiently inhibits tumor growth and bone invasion in oral squamous cell carcinoma. Mol. Pharm. 2024, 21, 661–676. [Google Scholar] [CrossRef] [PubMed]
  134. Martin, C.E.; Nguyen, A.; Kang, M.K.; Kim, R.H.; Park, N.H.; Shin, K.H. DYRK1A is required for maintenance of cancer stemness, contributing to tumorigenic potential in oral/oropharyngeal squamous cell carcinoma. Exp. Cell Res. 2021, 405, 112656. [Google Scholar] [CrossRef] [PubMed]
  135. Tang, D.; Tao, D.; Fang, Y.; Deng, C.; Xu, Q.; Zhou, J. TNF-Alpha Promotes Invasion and Metastasis via NF-Kappa B Pathway in Oral Squamous Cell Carcinoma. Med. Sci. Monit. Basic. Res. 2017, 23, 141–149. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, D.; Peng, Y.; Lin, G.; Xiao, Q.; Liu, H.; Nan, X.; Han, J.; Zhao, R.; Wang, Y.; Liu, J.; et al. TGF-βRII/EP300/SMAD4 cascade signaling pathway promotes invasion and glycolysis in oral squamous cell carcinoma. J. Oral Pathol. Med. 2023, 52, 483–492. [Google Scholar] [CrossRef] [PubMed]
  137. Geindreau, M.; Ghiringhelli, F.; Bruchard, M. Vascular Endothelial Growth Factor, a Key Modulator of the Anti-Tumor Immune Response. Int. J. Mol. Sci. 2021, 22, 4871. [Google Scholar] [CrossRef] [PubMed]
  138. Im, J.H.; Buzzelli, J.N.; Jones, K.; Franchini, F.; Gordon-Weeks, A.; Markelc, B.; Chen, J.; Kim, J.; Cao, Y.; Muschel, R.J. FGF2 alters macrophage polarization, tumour immunity and growth and can be targeted during radiotherapy. Nat. Commun. 2020, 11, 4064. [Google Scholar] [CrossRef] [PubMed]
  139. Kondoh, N.; Mizuno-Kamiya, M.; Umemura, N.; Takayama, E.; Kawaki, H.; Mitsudo, K.; Muramatsu, Y.; Sumitomo, S. Immunomodulatory aspects in the progression and treatment of oral malignancy. Jpn. Dent. Sci. Rev. 2019, 55, 113–120. [Google Scholar] [CrossRef]
  140. Jansson, L.; Ehnevid, H.; Blomlöf, L.; Weintraub, A.; Lindskog, S. Endodontic pathogens in periodontal disease augmentation. J. Clin. Periodontol. 1995, 22, 598–602. [Google Scholar] [CrossRef]
  141. Ma, L.; Wang, X.; Liu, H.; Jiang, C.; Liao, H.; Xu, S.; Guo, Y.; Cao, Z. CXXC5 mediates p. gingivalis-suppressed cementoblast functions partially via MAPK Signaling Network. Int. J. Biol. Sci. 2019, 15, 1685–1695. [Google Scholar] [CrossRef] [PubMed]
  142. Ma, L.; Liu, H.; Wang, X.; Jiang, C.; Yao, S.; Guo, Y.; Wang, H.; Cao, Z. CXXC5 orchestrates Stat3/Erk/Akt signaling networks to modulate P. gingivalis-elicited autophagy in cementoblasts. Biochim. Biophys. Acta Mol. Cell Res. 2021, 1868, 118923. [Google Scholar] [CrossRef]
  143. Bozkurt, S.B.; Tuncer Gokdag, I.; Hakki, S.S. Porphyromonas gingivalis-Lipopolysaccharide induces cytokines and enzymes of the mouse cementoblasts. Cytokine 2021, 138, 155380. [Google Scholar] [CrossRef] [PubMed]
  144. Nagano, R.; Nakako, Y.; Fujii, S.; Kawano, S.; Maeda, H.; Kiyoshima, T. The IL-1β-p65 axis stimulates quiescent odontogenic epithelial cell rests via TGF-β signalling to promote cell proliferation of the lining epithelia in radicular cysts: A laboratory investigation. Int. Endod. J. 2024, 57, 344–354. [Google Scholar] [CrossRef]
  145. Nair, P.N.; Pajarola, G.; Luder, H.U. Ciliated epithelium-lined radicular cysts. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2002, 94, 485–493. [Google Scholar] [CrossRef] [PubMed]
  146. Keinan, D.; Cohen, R.E. The significance of epithelial rests of Malassez in the periodontal ligament. J. Endod. 2013, 39, 582–587. [Google Scholar] [CrossRef] [PubMed]
  147. França, G.M.; Medeiros, C.K.S.; Almeida, D.R.M.F.; Pinheiro, J.C.; de Andrade, A.L.D.L.; Santos, P.P.A.; Galvão, H.C. Release of matrix metalloproteinases by macrophages in radicular cysts and residual radicular cysts. Appl. Immunohistochem. Mol. Morphol. 2022, 30, 291–297. [Google Scholar] [CrossRef] [PubMed]
  148. Rios Osorio, N.; Caviedes-Bucheli, J.; Mosquera-Guevara, L.; Adames-Martinez, J.S.; Gomez-Pinto, D.; Jimenez-Jimenez, K.; Avendano Maz, H.; Bornacelly-Mendoza, S. The paradigm of the inflammatory radicular cyst: Biological aspects to be considered. Eur. Endod. J. 2023, 8, 20–36. [Google Scholar] [CrossRef]
  149. Wu, Q.; Asif, M.; Qari, I.H.; Qazi, J.A. Role of interleukin-1 in pathogenesis of radicular cyst. J. Ayub Med. Coll. Abbottabad 2010, 22, 86–87. [Google Scholar]
  150. Qureshi, W.U.; Idris, M.; Khan, S.A. Role of tumour necrosis factor in pathogenesis of radicular cyst. J. Ayub Med. Coll. Abbottabad 2011, 23, 87–89. [Google Scholar]
  151. Ohshima, M.; Nishiyama, T.; Tokunaga, K.; Sato, S.; Maeno, M.; Otsuka, K. Profiles of cytokine expression in radicular cyst-lining epithelium examined by RT-PCR. J. Oral Sci. 2000, 42, 239–246. [Google Scholar] [CrossRef] [PubMed]
  152. Suzuki, T.; Kumamoto, H.; Ooya, K.; Motegi, K. Immunohistochemical analysis of CD1a-labeled Langerhans cells in human dental periapical inflammatory lesions—Correlation with inflammatory cells and epithelial cells. Oral Dis. 2001, 7, 336–343. [Google Scholar] [CrossRef] [PubMed]
  153. Gao, Z.; Mackenzie, I.C.; Rittman, B.R.; Korszun, A.K.; Williams, D.M.; Cruchley, A.T. Immunocytochemical examination of immune cells in periapical granulomata and odontogenic cysts. J. Oral Pathol. 1988, 17, 84–90. [Google Scholar] [CrossRef] [PubMed]
  154. Li, T.J.; Browne, R.M.; Matthews, J.B. Expression of epidermal growth factor receptors by odontogenic jaw cysts. Virchows Arch. A Pathol. Anat. Histopathol. 1993, 423, 137–144. [Google Scholar] [CrossRef] [PubMed]
  155. Lin, L.M.; Wang, S.L.; Wu-Wang, C.; Chang, K.M.; Leung, C. Detection of epidermal growth factor receptor in inflammatory periapical lesions. Int. Endod. J. 1996, 29, 179–184. [Google Scholar] [CrossRef] [PubMed]
  156. Leonardi, R.; Caltabiano, M.; Pagano, M.; Pezzuto, V.; Loreto, C.; Palestro, G. Detection of vascular endothelial growth factor/vascular permeability factor in periapical lesions. J. Endod. 2003, 29, 180–183. [Google Scholar] [CrossRef] [PubMed]
  157. Lin, L.M.; Huang, G.T.; Rosenberg, P.A. Proliferation of epithelial cell rests, formation of apical cysts, and regression of apical cysts after periapical wound healing. J. Endod. 2007, 33, 908–916. [Google Scholar] [CrossRef]
  158. Lee, J.H.; Nam, H.; Um, S.; Lee, J.; Lee, G.; Seo, B.M. Upregulation of GM-CSF by TGF-β1 in epithelial mesenchymal transition of human HERS/ERM cells. In Vitro Cell Dev. Biol. Anim. 2014, 50, 399–405. [Google Scholar] [CrossRef] [PubMed]
  159. Bueno, D.F.; Sunaga, D.Y.; Kobayashi, G.S.; Aguena, M.; Raposo-Amaral, C.E.; Masotti, C.; Cruz, L.A.; Pearson, P.L.; Passos-Bueno, M.R. Human stem cell cultures from cleft lip/palate patients show enrichment of transcripts involved in extracellular matrix modeling by comparison to controls. Stem Cell Rev. Rep. 2011, 7, 446–457. [Google Scholar] [CrossRef] [PubMed]
  160. Han, X.; Shao, X.; Lin, X.; Du, Z.; Wang, Y. Squamous cell carcinoma arising from odontogenic cyst epithelium or oral mucosa epithelium after marsupialization of odontogenic radicular cyst in posterior mandible? J. Craniofac Surg. 2023, 34, e423–e425. [Google Scholar] [CrossRef] [PubMed]
  161. Ravenscroft, H.; El Karim, I.; Krasnodembskaya, A.D.; Gilmore, B.; About, I.; Lundy, F.T. Novel antibacterial properties of the human dental pulp multipotent mesenchymal stromal cell secretome. Am. J. Pathol. 2022, 192, 956–969. [Google Scholar] [CrossRef]
  162. Tezal, M.; Scannapieco, F.A.; Wactawski-Wende, J.; Meurman, J.H.; Marshall, J.R.; Rojas, I.G.; Stoler, D.L.; Genco, R.J. Dental caries and head and neck cancers. JAMA Otolaryngol. Head. Neck Surg. 2013, 139, 1054–1060. [Google Scholar] [CrossRef] [PubMed]
  163. Zhang, Q.; Zhao, Q.; Li, T.; Lu, L.; Wang, F.; Zhang, H.; Liu, Z.; Ma, H.; Zhu, Q.; Wang, J.; et al. Lactobacillus plantarum-derived indole-3-lactic acid ameliorates colorectal tumorigenesis via epigenetic regulation of CD8+ T cell immunity. Cell Metab. 2023, 35, 943–960.e9. [Google Scholar] [CrossRef] [PubMed]
  164. Hunter, H.A.; Poyton, H.G. Pulpal metastases from Ewing’s sarcoma. Report of a case. Oral Surg. Oral Med. Oral Pathol. 1963, 16, 136–139. [Google Scholar] [CrossRef]
  165. Shankar, S. Dental pulp metastases and pan-osseous mandibular involvement with mammary adenocarcinoma. Br. J. Oral Maxillofac. Surg. 1984, 22, 455–461. [Google Scholar] [CrossRef]
  166. Feinberg, A.P.; Levchenko, A. Epigenetics as a mediator of plasticity in cancer. Science 2023, 379, eaaw3835. [Google Scholar] [CrossRef]
  167. Emfietzoglou, R.; Pachymanolis, E.; Piperi, C. Impact of epigenetic alterations in the development of oral diseases. Curr. Med. Chem. 2021, 28, 1091–1103. [Google Scholar] [CrossRef] [PubMed]
  168. Panagakos, F.S. Transformation and preliminary characterization of primary human pulp cells. J. Endod. 1998, 24, 171–175. [Google Scholar] [CrossRef]
  169. Lee, M.S.; Yuan, H.; Jeon, H.; Zhu, Y.; Yoo, S.; Shi, S.; Krueger, B.; Renne, R.; Lu, C.; Jung, J.U.; et al. Human mesenchymal stem cells of diverse origins support persistent infection with Kaposi’s sarcoma-associated herpesvirus and manifest distinct angiogenic, invasive, and transforming phenotypes. mBio 2016, 7, e02109-15. [Google Scholar] [CrossRef] [PubMed]
  170. Barker, B.F. Odontogenic myxoma. Semin. Diagn. Pathol. 1999, 16, 297–301. [Google Scholar] [PubMed]
  171. Miyagi, S.P.; Maranduba, C.M.; Silva Fde, S.; Marques, M.M. Dental pulp stem cells express proteins involved in the local invasiveness of odontogenic myxoma. Braz. Oral Res. 2012, 26, 139–144. [Google Scholar] [CrossRef]
  172. Wilson, R.; Urraca, N.; Skobowiat, C.; Hope, K.A.; Miravalle, L.; Chamberlin, R.; Donaldson, M.; Seagroves, T.N.; Reiter, L.T. Assessment of the tumorigenic potential of spontaneously immortalized and hTERT-immortalized cultured dental pulp stem cells. Stem Cells Transl. Med. 2015, 4, 905–912. [Google Scholar] [CrossRef]
  173. Barillari, G.; Bei, R.; Manzari, V.; Modesti, A. Infection by High-Risk Human Papillomaviruses, epithelial-to-mesenchymal transition and squamous pre-malignant or malignant lesions of the uterine cervix: A series of chained events? Int. J. Mol. Sci. 2021, 22, 13543. [Google Scholar] [CrossRef] [PubMed]
  174. Razghonova, Y.; Zymovets, V.; Wadelius, P.; Rakhimova, O.; Manoharan, L.; Brundin, M.; Kelk, P.; Romani Vestman, N. Transcriptome analysis reveals modulation of human stem cells from the apical papilla by species associated with dental root canal infection. Int. J. Mol. Sci. 2022, 23, 14420. [Google Scholar] [CrossRef] [PubMed]
  175. Diomede, F.; Zingariello, M.; Cavalcanti, M.F.X.B.; Merciaro, I.; Pizzicannella, J.; De Isla, N.; Caputi, S.; Ballerini, P.; Trubiani, O. MyD88/ERK/NFkB pathways and pro-inflammatory cytokines release in periodontal ligament stem cells stimulated by Porphyromonas gingivalis. Eur. J. Histochem. 2017, 61, 2791. [Google Scholar] [CrossRef]
  176. Wang, J.; Dai, J.; Liu, B.; Gu, S.; Cheng, L.; Liang, J. Porphyromonas gingivalis lipopolysaccharide activates canonical Wnt/β-catenin and p38 MAPK signalling in stem cells from the apical papilla. Inflammation 2013, 36, 1393–2402. [Google Scholar] [CrossRef] [PubMed]
  177. Rakhimova, O.; Schmidt, A.; Landström, M.; Johansson, A.; Kelk, P.; Romani Vestman, N. Cytokine secretion, viability, and real-time proliferation of apical-papilla stem cells upon exposure to oral bacteria. Front. Cell Infect. Microbiol. 2021, 10, 620801. [Google Scholar] [CrossRef] [PubMed]
  178. Neth, P.; Ries, C.; Karow, M.; Egea, V.; Ilmer, M.; Jochum, M. The Wnt signal transduction pathway in stem cells and cancer cells: Influence on cellular invasion. Stem Cell Rev. 2007, 3, 18–29. [Google Scholar] [CrossRef] [PubMed]
  179. Kestler, D.P.; Foster, J.S.; Macy, S.D.; Murphy, C.L.; Weiss, D.T.; Solomon, A. Expression of odontogenic ameloblast-associated protein (ODAM) in dental and other epithelial neoplasms. Mol. Med. 2008, 14, 318–326. [Google Scholar] [CrossRef] [PubMed]
  180. Nakayama, Y.; Kobayashi, R.; Matsui, S.; Matsumura, H.; Iwai, Y.; Noda, K.; Yamazaki, M.; Kurita-Ochiai, T.; Yoshimura, A.; Shinomura, T.; et al. Localization and expression pattern of amelotin, odontogenic ameloblast-associated protein and follicular dendritic cell-secreted protein in the junctional epithelium of inflamed gingiva. Odontology 2017, 105, 329–337. [Google Scholar] [CrossRef] [PubMed]
  181. Gupta, I.; Das, N.; Ranjan, P.; Sujatha, R.; Gupta, R.; Gupta, N. A preliminary study on the evaluation of in-vitro inhibition potential of antimicrobial efficacy of raw and commercial honey on escherichia coli: An emerging periodontal pathogen. Mymensingh Med. J. 2021, 30, 547–554. [Google Scholar] [PubMed]
  182. Pedrosa, M.D.S.; Vilela, H.D.S.; Rahhal, J.G.; Bueno, N.P.; Lima, F.S.; Nogueira, F.N.; Sipert, C.R. Exposure to lipopolysaccharide and calcium silicate-based materials affects the behavior of dental pulp cells. Braz. Dent. J. 2022, 33, 9–17. [Google Scholar] [CrossRef] [PubMed]
  183. Bredenkamp, J.K.; Zimmerman, M.C.; Mickel, R.A. Maxillary ameloblastoma. A potentially lethal neoplasm. Arch. Otolaryngol. Head. Neck Surg. 1989, 115, 99–104. [Google Scholar] [CrossRef]
  184. Lee, H.K.; Ji, S.; Park, S.J.; Choung, H.W.; Choi, Y.; Lee, H.J.; Park, S.Y.; Park, J.C. Odontogenic ameloblast-associated protein (ODAM) mediates junctional epithelium attachment to teeth via integrin-ODAM-Rho guanine nucleotide exchange factor 5 (ARHGEF5)-RhoA signaling. J. Biol. Chem. 2015, 290, 14740–14753. [Google Scholar] [CrossRef] [PubMed]
  185. Zhang, J.; Wang, Y.; Fan, C.; Xiao, X.; Zhang, Q.; Xu, T.; Jiang, C. Interleukin-8/β-catenin mediates epithelial-mesenchymal transition in ameloblastoma. Oral Dis. 2019, 25, 1964–1971. [Google Scholar] [CrossRef] [PubMed]
  186. Hao, F.; Liu, J.; Zhong, M.; Wang, J.; Liu, J. Expression of E-cadherin, vimentin and β-catenin in ameloblastoma and association with clinicopathological characteristics of ameloblastoma. Int. J. Clin. Exp. Pathol. 2018, 11, 199–207. [Google Scholar] [PubMed]
  187. Ribeiro, B.F.; Iglesias, D.P.; Nascimento, G.J.; Galvão, H.C.; Medeiros, A.M.; Freitas, R.A. Immunoexpression of MMPs-1, -2, and -9 in ameloblastoma and odontogenic adenomatoid tumor. Oral Dis. 2009, 15, 472–477. [Google Scholar] [CrossRef] [PubMed]
  188. Siar, C.H.; Ng, K.H. Differential expression of transcription factors Snail, Slug, SIP1, and Twist in ameloblastoma. J. Oral Pathol. Med. 2014, 43, 45–52. [Google Scholar] [CrossRef] [PubMed]
  189. Marquard, F.E.; Jücker, M. PI3K/AKT/mTOR signaling as a molecular target in head and neck cancer. Biochem. Pharmacol. 2020, 172, 113729. [Google Scholar] [CrossRef] [PubMed]
  190. Ghareghomi, S.; Atabaki, V.; Abdollahzadeh, N.; Ahmadian, S.; Hafez Ghoran, S. Bioactive PI3-kinase/Akt/mTOR inhibitors in targeted lung cancer therapy. Adv. Pharm. Bull. 2023, 13, 24–35. [Google Scholar] [CrossRef] [PubMed]
  191. Kashyap, T.; Pramanik, K.K.; Nath, N.; Mishra, P.; Singh, A.K.; Nagini, S.; Rana, A.; Mishra, R. Crosstalk between Raf-MEK-ERK and PI3K-Akt-GSK3β signaling networks promotes chemoresistance, invasion/migration and stemness via expression of CD44 variants (v4 and v6) in oral cancer. Oral Oncol. 2018, 86, 234–243. [Google Scholar] [CrossRef] [PubMed]
  192. Ito, K.; Ota, A.; Ono, T.; Nakaoka, T.; Wahiduzzaman, M.; Karnan, S.; Konishi, H.; Furuhashi, A.; Hayashi, T.; Yamada, Y.; et al. Inhibition of Nox1 induces apoptosis by attenuating the AKT signaling pathway in oral squamous cell carcinoma cell lines. Oncol. Rep. 2016, 36, 2991–2998. [Google Scholar] [CrossRef] [PubMed]
  193. Pai, S.; Yadav, V.K.; Kuo, K.T.; Pikatan, N.W.; Lin, C.S.; Chien, M.H.; Lee, W.H.; Hsiao, M.; Chiu, S.C.; Yeh, C.T.; et al. PDK1 Inhibitor BX795 Improves Cisplatin and Radio-Efficacy in Oral Squamous Cell Carcinoma by Downregulating the PDK1/CD47/Akt-Mediated Glycolysis Signaling Pathway. Int. J. Mol. Sci. 2021, 22, 11492. [Google Scholar] [CrossRef] [PubMed]
  194. Smolensky, D.; Rathore, K.; Bourn, J.; Cekanova, M. Inhibition of the PI3K/AKT Pathway Sensitizes Oral Squamous Cell Carcinoma Cells to Anthracycline-Based Chemotherapy In Vitro. J. Cell Biochem. 2017, 118, 2615–2624. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Theoretical effects that dental pulp infection by P. gingivalis and/or F. nucleatum could have on OSCCs. Arrows symbolize the direction of connections. Abbreviations: CSC, cancer stem cell; EMT, epithelial-to-mesenchymal transition; FGF, fibroblast growth factor; I.C., inflammatory cytokines; TGF, transforming growth factor; VEGF, vascular endothelial growth factor. Created with BioRender.com.
Figure 1. Theoretical effects that dental pulp infection by P. gingivalis and/or F. nucleatum could have on OSCCs. Arrows symbolize the direction of connections. Abbreviations: CSC, cancer stem cell; EMT, epithelial-to-mesenchymal transition; FGF, fibroblast growth factor; I.C., inflammatory cytokines; TGF, transforming growth factor; VEGF, vascular endothelial growth factor. Created with BioRender.com.
Ijms 25 05083 g001
Figure 2. Theoretical effects that an apical periodontitis by P. gingivalis and/or F. nucleatum could have on OSCCs. Arrows symbolize the directions of connections. Abbreviations: ERMs, epithelial cell rests of Malassez; G.F., growth factors; I.C., inflammatory cytokines. Created with BioRender.com.
Figure 2. Theoretical effects that an apical periodontitis by P. gingivalis and/or F. nucleatum could have on OSCCs. Arrows symbolize the directions of connections. Abbreviations: ERMs, epithelial cell rests of Malassez; G.F., growth factors; I.C., inflammatory cytokines. Created with BioRender.com.
Ijms 25 05083 g002
Table 1. Direct pro-tumor effects of P. gingivalis and F. nucleatum.
Table 1. Direct pro-tumor effects of P. gingivalis and F. nucleatum.
Pro-Tumor EffectP. gingivalisF. nucleatum
Triggering of the synthesis of inflammatory cytokines or growth factors by infected cellsWang Q et al. [15]; Ramage G et al. [16]; Milward MR et al. [17]; Yee M et al. [18]; Abdulkareem AA et al. [19]Park SR et al. [14]; Abdulkareem AA et al. [19]; Aral K et al. [20]; Hung SC et al. [21]; Kang W et al. [22]; Kang W et al. [23]; Kurgan S et al. [24]
EMT inductionAbdulkareem AA et al. [19]; Sztukowska MN et al. [25]; Lee J et al. [26]; Abdulkareem AA et al. [27]Abdulkareem AA et al. [19]; Abdulkareem AA et al. [27]
Release of mutagenic substancesNguyen LH et al. [28]Ma Z et al. [29]; Vital M et al. [30]; Zhang S et al. [31]
Stimulation of cell proliferationKuboniwa M et al. [32]; Chang C et al. [33]; Pan C et al. [34]Geng F et al. [35]; Binder Gallimidi A et al. [36]
Upregulation of cell survival factorsGao S et al. [37]; Lee J et al. [38]; Yilmaz O et al. [39]; Yao L et al. [40]; Nakhjiri SF et al. [41]; Hoppe T et al. [42]; Li J et al. [43]Da J et al. [44]; Rath-Deschner B et al. [45]; Duan C et al. [46]
Induction/enhancement of cell invasionShen S et al. [47]; Inaba H et al. [48]; Inaba H et al. [49]; Ha NH et al. 2016 [50]; Meng F et al. [51]Da J et al. [44]; Harrandah AM et al. [52]; Kamarajan P et al. [53]; Uitto VJ et [54]; Bachrach G et al. [55]
Impairment of anti-tumor immunityLiu S et al. [13]; Arjunan P et al. [56]; Groeger S et al. [57]; Wen L et al. [58]Bachrach G et al. [55]; Gur C et al. [59]; Gur C et al. [60]
Triggering of tumor angiogenesisLin FY et al. [61]Li Z et al. [62]
Promotion of cellular stemnessHa NH et al. [63]Kang W et al. [23]; Kang W et al. [64]
Table 2. P. gingivalis or F. nucleatum effects possibly affecting dental tumorigenesis.
Table 2. P. gingivalis or F. nucleatum effects possibly affecting dental tumorigenesis.
EventEffectReferences
Upon their infection by (or exposure to) Fn or Pg, the dental pulp SCs release ICIC inhibit the differentiation of dental pulp SCsRothermund K et al. [118]; Qin Z et al. [119]; Sonmez Kaplan S et al. [120]; Li M et al. [121];
Upon their infection by (or exposure to) Fn or Pg, the SCs of the periodontal ligament or apical papilla release ICIC inhibit the differentiation of periodontal ligament or apical papilla SCsRazghonova Y et al. [174]; Diomede F et al. [175]; Wang J et al. [176]; Rakhimonova O et al. [177]
Fn and Pg upregulate both IL-1 and MMP-2 expression by cementoblastsIL-1 inhibits the differentiation of SCs and cementoblasts. MMP-2 mediates cementoblasts invasionSonmez Kaplan S et al. [120]; Ma L et al. [142]; Bozkurt SB et al. [143]; Miyagi 2012 [171]; Neth P et al. [178]
Pg upregulates the expression of the ODAM-associated proteinPossible impact on the onset and/or progression of ameloblastomasKestler DP et al. [179]; Nakayama Y et al. [180]
Abbreviations: IC, inflammatory cytokines; IL, interleukin; Fn, Fusobacterium nucleatum; MMP, matrix metalloproteinase; ODAM, odontogenic ameloblast; Pg, Porphyromonas gingivalis; SCs, stem cells.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ciani, L.; Libonati, A.; Dri, M.; Pomella, S.; Campanella, V.; Barillari, G. About a Possible Impact of Endodontic Infections by Fusobacterium nucleatum or Porphyromonas gingivalis on Oral Carcinogenesis: A Literature Overview. Int. J. Mol. Sci. 2024, 25, 5083. https://doi.org/10.3390/ijms25105083

AMA Style

Ciani L, Libonati A, Dri M, Pomella S, Campanella V, Barillari G. About a Possible Impact of Endodontic Infections by Fusobacterium nucleatum or Porphyromonas gingivalis on Oral Carcinogenesis: A Literature Overview. International Journal of Molecular Sciences. 2024; 25(10):5083. https://doi.org/10.3390/ijms25105083

Chicago/Turabian Style

Ciani, Luca, Antonio Libonati, Maria Dri, Silvia Pomella, Vincenzo Campanella, and Giovanni Barillari. 2024. "About a Possible Impact of Endodontic Infections by Fusobacterium nucleatum or Porphyromonas gingivalis on Oral Carcinogenesis: A Literature Overview" International Journal of Molecular Sciences 25, no. 10: 5083. https://doi.org/10.3390/ijms25105083

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