The Possible Role of Chronic Periodontal Disease in Neurodegenerative Diseases and Glioblastoma Pathogenesis- An Essential Review Download PDF

Journal Name : SunText Review of Dental Sciences

DOI : doi.org/10.51737/2766-4996.2024.183

Article Type : Research Article

Authors : Chrysanthakopoulos NA and Vryzaki E

Keywords : Periodontal disease; Tumorigenesis; Cancer; Fusobacterium nucleatum; Porphyromonas gingivalis; Neurodegenerative Diseases; Immune Response

Abstract

Chronic periodontal disease (PD), and especially periodontitis is a multifactorial disease caused by dental plaque accumulation, which formed by pathogenic bacteria that are able to trigger an immune response in susceptible hosts. Pathogenic bacteria in oral cavity are a source of chronic immunological reaction which can result in local and peripheral production of pro-inflammatory cytokines such as IL-1ß, IL-6 and TNF-?, inflammatory mediators, and bacterial products such as lipopolysaccaride endotoxin. Similarly, viruses, such as herpes and Epstein-Barr, have also been detected in periodontal pockets. In a similar way, a chronic systemic inflammation has been linked with an increased risk of brain pathological conditions such as cerebrovascular diseases, abscesses, etc. Therefore, it is reasonable, that a chronic infection and inflammation disease, such as periodontitis, may affect the?a central nervous system (CNS). It is well evidenced that oral pathogenic bacteria may cause systemic infection by transient or persistent bacteraemia, and in-filtrate distal locations and organs. It has also been suggested that in susceptible populations and under certain circumstances, bacterial and viral infections may enter the brain from the blood circulation. After entering the brain periodontal bacteria and their products may affect brain’s vascular integrity. Therefore, a potential role of chronic periodontitis in the development and progression of cerebral infection and inflammation could be suggested. No direct evidence has been revealed regarding a possible causal association between periodontal pathogens and brain diseases, however several researches have detected periodontal bacteria survived in the brain. It is possible that both inflammatory conditions may share a causal association with common risk factors and complex multifactorial aetiologies. Nevertheless, the possibility that pathogenic oral bacteria spread to the blood circulation and reach the brain, initiating or exacerbating existing cerebral diseases, could not be ignored. Moreover, the pro-inflammatory factors, and biomarkers induced systemically by a chronic periodontal inflammation, may play a role in CNS pathological conditions, such as Alzheimer’s Disease, Parkinson’s Disease, Multiple Sclerosis, etc. Systemic infection and/or inflammation, and especially chronic inflammation has also been associated with an increased risk of diverse types of cancer in organs such as liver, colorectal, pancreatic, lung, etc. Gliomas, and especially Glioblastoma Multiforme (GBM) is the most common type of primary malignant brain tumor, which currently has no effective treatments. The etiology of GBM has not been fully elucidated and it still remains unclear. Genetic influences in combination with environmental risk factors have been suggested as pathogenic factors of GBM. Viruses, such as Human Cytomegalovirus (HCMV), is also considered to be among the etiologic factors for gliomas development as exhibits tropism for glial cells. Human studies have recorded that oral pathogens are closely associated with cancers, however, whether those pathogens play a role in gliomas development remains unclear. The current review highlights the possible role of chronic periodontitis in brain, and especially in GBM development, focused on the role of chronic inflammation in carcinogenesis, in the light of recent literature.


Introduction

Glioma is the most conventional form of brain malignant tumors, accounting for 25.1% of the primary tumors in the central nervous system (CNS) [1], and are categorized into 4 histological grades according to the malignancy level. WHO grades I-II gliomas (low-grade gliomas, LGG) exhibit low aggressive tendencies and have a better prognosis, whereas WHO grades III-IV gliomas (high-grade gliomas, HGG) show a high rate of deterioration and a poor prognosis [2]. Glioma patients with the isocitrate dehydrogenase 1(IDH1) mutation have a more favourable prognosis, and the mutation is frequently expressed in LGG patients but rarely observed in WHO grade IV glioma patients [3]. The LGG, WHO grades II and III, could progress in Glioblastoma Multiforme (GBM), WHO grade IV, along with the progression of the tumor [4]. GBM is the most frequent and malignant glioma type, with an extremely poor prognosis because of its histopathological characteristics [5]. GBM, also known as diffuse astrocytoma, shows a great morphological and genetical heterogeneity. GBM’s incidence is about 5-6 cases/100,000 population and its frequency varies between 12.0%-15.0% of all intracranial tumors [6,7]. The mean survival is under 15 months and the five-year rate is under 10%. The poor prognosis of GBM could be attributed to a highly abnormal vascularization, resistant to the common chemotherapy and radiotherapy and to the fact that general is difficult to be completely removed surgi-cally [8]. GBM is mainly diagnosed at advanced ages, with a mean age on diagnosis of 64 years [9]. Its etiology still remains unclear, however, genetic influences in combination with environmental risk factors have been suggested as GBM pathogenic factors [10]. It has also been suggested that is associated with constant exposure to ionizing radiation or chemical agents such as polycyclic aromatic hydrocarbons (PAH), electromagnetic fields and certain metals [11-13]. Viruses infe-ctions with human cytomegalovirus (CMV), genetic diseases such as tuberous sclerosis, multiple endocrine neoplasia (MEN) type IIA, Turcot syndrome, and neurofibromatos is type I, NF1 [14-16]. Moreover, acquired head traumas, which occurred as a result of a brain contusion, may predispose to the GBM development [17]. GBM is classified as a primary or as a secondary tumor as a result of a malignant transformation from a lower grade brain tumor and/or with mutation in the IDH gene and is classified in diverse histopathological types such as Classical, Proneural, Neural and Mesenchymal according to the gene expression profile [18]. Periodontitis, is a chronic inflammatory condition characterized by the disruption of tissues surrounding and supporting the teeth [19]. Periodontal Disease (PD) in Europe affects 5-20% of adults aged 35-44 years old and 40% of the elderly aged 65-74 [20,21] whereas chronic periodontitis affects approximately 50% of the adult individuals and its incidence and severity increase with age, showing a prevalence of 70% of over-65 years-old in the USA [22]. PD or periodontitis is caused by the host’s immunological response to periodontal pathogens [23], and leads to local inflammation, ultimately contributing to chronic systemic inflammation. Periodontal infection individuals exhibit increased circulating inflammatory biomarkers levels indicating the systemic implications of periodontal infection [23,24]. PD is significantly associated with other pathological conditions such as cardiovascular disease, rheumatoid arthritis, pneumonia, chronic obstructive pulmonary disease, metabolic syndrome, obesity, chronic kidney disease, and cancer [25]. Individuals with chronic inflammatory diseases such as the mentioned may be at greater risk of cancer [26].The controversial differences between the association of PD and systemic diseases in different studies could be attributed to the heterogeneity in the definitions. The susceptibility to PD is individual, as it depends on possible dysbiosis and immune response to the bacteria accumulation, genetics, oral hygiene and the chronic disease [27]. Gram-negative bacteria are responsible for the dysbiosis in PD [28]. In PD the most conventional species are Porphyromonas gingivalis, Treponema dent cola, and Tanneralla forsythia, also known as the red-complex [28]. Moreover, P. gingivalis, Aggregatibacter actionmycetemcomitans, Fusobacterium spp., and Prevotella intermedia/nigrescens, have been described as the most common subgingival pathogens detected in chronic PD patients [29, 30]. PD also results in the development of more diverse microbiota population, potentially caused by an increase of different nutrients available to microorganisms due to ongoing inflammation and weakened immune response, not sufficient to control bacterial proliferation [28]. Chronic inflammation has been suggested to be implicated in tumor initiation and progression [13]. For more than five decades the relationship between PD and increased cancer risk has been investigated, however, findings to date have limited practical significance as cancer prevention indices, despite the fact that useful knowledge have been acquired regarding the role of PD treatment in reducing the risk of different types of cancer [31]. Recently, an increasing interest exists in examining the possible association between PD variables, and cancer risk, in several organs and systems. Epidemiological studies have suggested significant associations between periodontitis indices, tooth loss and cancer risk of diverse organs and locations such as head and neck region, lungsupper gastro-intestinal system, pancreas, etc. [25,32-35]. During the PD active phase P. gingivalis partially disrupts the periodontal tissue, enters the blood circulation, and causes bacteremia, which leads to a great number of inflammatory mediators release, eventually inducing a prolonged low-grade inflammatory response in distant organs [36]. P.gingivalis is the most conventional periopathogenic bacteria associated with periodontitis [24,37], and it plays a crucial role in tumor initiation and progression. Significantly increased numbers of P.gingivalis have been detected in oral squamous cell carcinoma [38], orodigestive [34], and pancreatic cancer [39]. The substances produced by oral microbiota may be carcinogenic [40,41]. Chronic periodontitis exposes organs to bacterial endotoxins, enzymes, metabolic by-products and continuously stimulates the immune response and production of cytokines, chemokines, prostaglandins, and other inflammatory biomarkers [42]. Chronic inflammation lengthens the cell cycle, stimulates proliferation, angiogenesis and migration, and inhibits apoptosis. Oxidative stress destroys the mucosa making it more susceptible to other carcinogens such as tobacco, alcohol, HPV and EBV [43]. All of the mentioned factors may predispose individuals to the development of diverse types of cancer [44]. Recent reports have recorded that PD may affect the development and progression of some brain disorders. Examining the association between periodontitis and cancer, it is interesting to identify whether periodontal infections are potentially associated with glioma. Evidence from human studies has indicated that oral microbiota is closely related to cancers [45,46], however, whether oral microbiota is involved in glioma malignancy remains unclear

P. gingival is has been detected in the brains of patients with Alzheimer’s disease and intracranial aneurysm [47-49]. P. gingival is lipopolysaccharide (LPS) has been identified in brain tissue using immunofluorescence labeling [50], whereas is frequently used to examine how PD affects cancers [47-50] and CNS diseases [51,52]. Experimental studies have shown that P. gingival is or its LPS is able to cross the blood-brain barrier, enter brain tissue, and stimulate the proliferation and migration of glioma cells at diverse concentrations [36]. P. gingival is associated with glioma grading and also shows a significant association with IDH1 mutations in gliomas [53]. Those observations suggest that periodontal pathogens may have a significant role in glioma development. However, the precise mechanisms through which PD contributes to the initiation and progression of GBM remain incompletely understood. No previous studies have specifically investigated the possible association between PD and GBM. However, recently PD and GBM have been associated with an increased activity of CMV [54,55], leading to a possible relationship between both. The mentioned increased activity of CMV has been also detected in others inflammatory diseases such as cardiovascular disease, rheumatoid arthritis, and diabetes mellitus [55,56].

The aim of the current review was to explore the common pathogenesis of Neurodegenerative Diseases and GBM, in an effort to detect the possible role of PD as an etiologic or risk factor for their development.

P. gingivalis andF. nucleatum Molecular Mechanisms in Cancer Pathogenesis

Recent epidemiological researches have suggested an increase in the risk of cancer incidence and /or mortality in PD individuals [42]. Dysbiosis in chronic periodontitis is attributed to oral pathogens [57], and Porphyromonas gingivalis and Fusobacterium nucleatum are the main microbial pathogens in its pathogenesis [58]. Those bacteria also play an essential role in initiation and promotion of carcinogenesis [59]. Resent interest has focused on the role of P. gingivalis in cancer due to its ability to evade the immune system whereas maintains a persisting chronic inflammation condition in the surrounding environment [60]. In a similar way, but to a lesser extent, the role of F. nucleatum in carcinogenesis has been a central point due to its ability to coaggregate with oral biofilm colonizers and to regulate other bacteria’s crossing of the host’s epithelial and endothelial cells barrier [61-63].

Role of P. gingivalis in Mediating Cellular Transformation

Long-term infections of P. gingivalis in human immortalized oral epithelial cells [64] showed that the infected cells ultrastructure was indicated by aberrant nucleoli and heterochromatin and weakened cellular junctions highlighted by desmosomes scarcity, known morphological characteristics of cancer cells. In P. gingivalis infected cells the plakophilin 1 (PKP1), which stabilizes desmosomes, was decreased [65]. The following biomarkers, Colony-Stimulating Factor 1 (CSF1), Friend Leukemia Virus Integration 1 (FLI1), Growth Arrest Specific 6 (GAS6), Programmed Cell Death 1 Ligand 2 (PDCD1LG2), CD274, Colon-Cancer-Associated Transcript 1 (CCAT1) and Nicotinamide N-Methyltransferase (NNMT), which are tumorigenesis markers, were up-regulated in P. gingivalis infected cells. In addition, proMMP9 and activated MMP9, which are involved in cellular invasion, were increased in P. gingivalis infected cells [64]. GroEL, a Heat Shock Protein (HSP) 60 family member, is considered one of the virulent factors released by P. gingivalis [66]. That member is responsible for induction neo-angiogenesis in epithelial progenitor cells and promotes their migration and progression by up-regulating E-selectin via activation of the SAPK/JNK, PI3K, and p38MAPK signaling pathways and also to a lesser extent through the NOS-related pathways [67]. P. gingivalis activates the PI3K/Akt and JAK/STAT signaling pathways and inhibits the apoptotic intrinsic pathway by preventing mitochondrial membrane depolarization and blocking cytochrome C release followed by pro-apoptotic down-regulation (caspase 3, caspase 9, Bad and Bax) and anti-apoptotic genes up-regulation (survivin, Bcl-2, bcl-XL and Bfl-1) in gingival epithelial cells [68-71]. P. gingivalis also up-regulates Cyclin A, CDK4 and CDK6 expression and activates CDK2, down-regulates the Cyclin D and INK4 expression, decreases p53’s concentrations and activation, and also decreases the levels of the following kinases Chk2, CK1delta,CK1 epsilon and Aurora A. Moreover, it increases the levels of PI3K, PDK1, p70S6K and p90RSK whereas inactivates PTEN by phosphorylation at s370 [72]. P. gingivalis induces the inflammatory cytokines IL-6, IL-8, sICAM-1 and MCP-1 production and their increase may be in part dependent on RgpA-Kgp activity, whereas the MIP-1? and IL1? post-infection secretion were found to be independent of RgpA-Kgp proteinase-adhesin complex [73,74]. Those events are responsible for an inflammatory environment which promotes tumor development. P. gingivalis also increases Toll-Like Receptor 2 (TLR2) signaling in gingival epithelial cells through the miR-105 down-regulation. TLR2 increased levels lead to IL-6 and TNF-? production and the NF-kB activation, which promotes pro-inflammation, contributing to an adequate tumor microenvironment [75]. Infected gingival epithelial cells by P. gingivalis upregulate the mi RNA-203 expression which applies its silencing effect on the cytokine signaling 3 (SOCS3) and SOCS6 suppressor, which leads to an increase in STAT3 and results in increased inflammation, a perfect tumorigenic microenvironment [76]. A P. gingivalis post-infection increase in Cyclin D1 and Cyclin E, which are implicated in promoting the transition from the G1 to S phase, simultaneously with a decrease in p21has been detected [73,77].

Role of P. gingivalis and F. nucleatum in Exacerbating Malignancy

P. gingivalis and F. nucleatum co-infection leads to an inflammatory response reflected by an increase in TNF-? and IL-1? [78]. The same co-infection led to tumor growth, invasion and proliferation in oral carcinoma in mouse model. TLR2 and TLR4, induce the IL-6 increase which is possible to activate STAT3 and NF-kB. STAT3 leads to Cyclin D1transcription, which promotes cellular proliferation [59]. F. nucleatum promotes tumor development and proliferation in vivo and in vitro in colorectal cancer cases, via FadA-binding to E-cadherin and the ?-catenin pathway signaling activation [79]. FadA binds to region 3 of the E-cadherin extracellular domain 5 (EC5), which is activated and internalized by clathrin and activates the ?-catenin which is translocated to the nucleus, and activates inflammatory genes NF-kB1 and NF-kB2, IL-6, IL-8 and IL18, Cyclin D1 and Myc oncogenes, transcription factors LEF/TCF and Wnt genes WNT7a, WNT7b and WNT9a [79]. 


Central Nervous System Cells Functions

 Astrocytes, the glia dominant neural type in the CNS, and microglial cells, originating from the hematopoietic lineage with monocyte/macrophage precursors are distributed uniformly throughout the CNS parenchyma [74]. Those CNS cells cooperate with one another in normal and pathological conditions. Astrocytes are not immune cells, however their morphology allow them to perform diverse important functions, such as providing metabolic support to neurons, regulazing neuronal activity, maintaining the extracellular fluids balance, and isolating excitable cells electrically [76]. Astrocytes also undergo proliferation and secrete matrix ingredients, resulting in a “glial scar” formation which surrounds the affected location. Moreover, they secrete proinflammatory and chemotactic mediators [75]. The neurons contribute to the processes implicated in neuro-inflammation, through constant communication with microglia, endothelial cells and the CNS blood vessels pericytes [80]. Microglia are the permanent mononuclear phagocyte population in the CNS, which shares phenotypical and functional features with macrophages, and are essential to the brain’s immune and inflammatory response [81,82]. Upon exogenous stimulation or micro-environment alterations, microglia are activated, release pro-inflammatory cytokines, such as IL-1?, IL-6, TNF-? and cytotoxic agents such as Reactive Oxidative Species (ROS) [74]. The increased level of ROS can cause neurotoxicity that affects diverse cellular proteins and homeostasis in the cells [83]. Microglia are also critical for phagocytosis of pathogens and debris [84-86], functions which are regulated by CD68, CD14, CX3CR1, and toll-like receptors (TLRs) [82,87,88]. TLRs are associated with microglial recognition of patterns on bacterial pathogens, where CD14 acts as a co-receptor for transmembrane TLR2 and TLR4, presenting antigens to them [82,89]. In microglial cells downstream signaling is mediated by NF-?B activation and the pro-inflammatory genes transcription such as, TNF-?,IL-1?, IL-6 [88]. Thus, microglia are the multi-tasking first line of defense in the brain, contributing to the inflammatory response upon detecting any danger signals presented by infectious stimuli or debris. Microglial cells in an activated state undergo morphological alterations and exhibit increased proliferation, acquire migration capacity, and display increased phagocytic activity [82].


Chronic Neuro-inflammation and Neurodegenerative Disorders Pathogenesis

Until now, the exact cause of the most prevalent neurodegenerative disorders, such as Alzheimer’s Disease (AD) and Parkinson’s Disease (PSD), is still unknown. However, chronic neuro inflammation seems to be a potentially significant risk factor for those diseases. Moreover, evidence indicates that systemic inflammation may potentially trigger the neuro-inflammation appearance [81]. Microglial cells are crucial for the neuro-inflammatory process within the brain [81,90,91]. In neurodegenerative diseases, such as AD, where neuro-inflammation is a part of the pathogenesis, microglial cells become chronically activated, release pro-inflammatory cytokines and display abnormal phagocytic ability of proteins, such as A? peptides, leading to further microglial activation [81,90,92]. Microbial ingredients are also able to trigger microglial activation and initiate the neuro-inflammatory response. Recent reports have suggested that oral microbes and/ or their virulence factors may contribute to this process [93-95], thus linking oral health status with neuro-inflammation risk.

Periodontitis is linked with neurodegenerative diseases and neuro-inflammatory processes through circulating mediators or the oral microbes direct access to the CNS via systemic circulation [96-98]. Treponema different species associated with periodontitis were revealed in AD cases brains [99]. Animal studies have indicated its migration from the oral cavity to the CNS, and post-mortem human studies in human AD have confirmed its presence intra-cerebrally [48, 100]. P. gingivalis, LPS and gingipains were also detected in AD patients brains [48,50,81,101,102]. P. gingivalis has unique abilities as is able to escape from the immune system, displays invasive properties, proteolytic nature and aggressive virulence factors, such as LPS and gingipains. The bacteria and its gingipains can directly invade astrocytes, microglia and neurons and mediate their neuro-inflammatory activity [103]. Moreover, gingipains is able to initiate physical injury to the cerebral microvasculature with degeneration of endothelial tight junctions and blood-brain barrier (BBB) increased permeability [103-105]. Neuro-inflammation and amyloid plaque formation developed after repeated oral infection of P. gingivalis in mice [93], indicating that infectious and inflammatory mechanisms are reasonable in the association between PD and neurodegenerative processes. However, it remains unclear whether PD-associated pathogens can directly activate the microglial function. In addition to P. gingivalis, other periodontal pathogens, such as T. denticola, A. actinomycetem- comitans), and F. nucleatum, might have a potential role in neuro-inflammation, especially in AD [106]. Animals studies have revealed that Treponema species have the ability to invade CNS and produce amyloid, A. actinomycetemcomitans serotype b triggers pro-inflammatory cytokines secretion by microglia and F. nucleatum-induced periodontitis can result in the worsening of AD symptoms in mice [107]. Neuro-inflammation is considered to be essential in PSD pathogenesis. It is suggested that an inflammatory response in the intestine may be responsible for initiating the disease. It has been supposed that bacteria form the intestine could access the brain via the vagus nerve, subsequently resulting in brain inflammation, especially in the substantia nigra region which disrupts the dopamine production, which is a hallmark element of PSD [101,102]. Neuro-inflammation is also thought to play a critical role in the appearance and progression of Multiple Sclerosis (MS). It has been found that inflammatory cytokines disrupt the blood-brain barrier, allowing B and plasma cells to enter the CNS, which then damage the myelin sheath, resulting in demyelination, a primary symptom of the disease [101,102].


Pathogenesis of Periodontitis and Its Role in Neuro-inflammation

In susceptible individuals with periodontitis, oral microbial dysbiosis triggers exaggerated chro-nic inflammation [108]. Possible direct and indirect mechanisms through which periodontitis may contribute to the appearance and progression of neuro-degeneration diseases have been recorded in the literature. It has been suggested that humoral, neuronal and cellular pathway are e low-grade systemic inflammation or locally released in periodontitis can enter the brain via the blood circularesponsible for the possible role of periodontitis in neuro-inflammation. Pro-inflammatory cytokines induced by thtion. Periodontal pathogens can reach the intestine by swallowing or via the blood circulation, are able to disrupt the intestine microbiota. The infected intestine epithelial cells release pro-inflammatory mediators that enter the brain, i.e., “oral-gut-brain axis” through the blood circulation. Another way to enter the brain is via the vagus nerve, whereas periodontal pathogens can reach the brain through the trigeminal nerve. Those pathogens might trigger trained myelopoiesis (trained immunity) which might induce hyper-inflammatory response that could affect the brain [80].


Molecular Mechanisms between Periodontitis and Glioblastoma

A previous survey has suggested a strong association between GBM and PD [36]. However, the specific pathogenic mechanism and crosstalk genes remain unclear. Another recent study [109] revealed that Chemokine (C-X-Cmotif) Receptor 4(CXCR4), Lymphocyte antigen 96 (LY96), and C3 genes play a crucial role in the co-pathogenesis of both diseases. CXCR4, a G protein-coupled receptor, binds its typical ligand Stromal cell-Derived Factor 1 (SDF-1). Although CXCR4 signaling is crucial for individual development and organ repair [110], high CXCR4 expression has been associated with an increased risk of cancer [111]. It was also found that CXCR4 was overexpressed in GBM and was associated with a poorer prognosis [112], consistent with previous findings [109]. In GBM, ligand binding to CXCR4 leads to conformational alterations which activate PI3K-AKT, JAK/STAT, and MEK1/2-Erk1/2 signaling pathways, resulting in the STAT3 activation, an oncogenic transcription factor implicated in GBM development [113]. MEK-ERK1/2 signaling inhibition amplifies the glioma cells adhesion to the extracellular matrix and reduces cell proliferation and migration [114]. Moreover, CXCR4 was found to be overexpressed in periodontitis gingival tissue [115]. CXCR4 activation by P. gingivalis results in crosstalk with TLR2, which disrupts the monocytes or macrophages killing function by inhibiting NO production and increasing cAMP-dependent protein kinase A (PKA) signaling [116]. In addition, PI3K-dependent adhesion pathway activation via CXCR4 in macrophages and monocytes results in CR3 activation, which is used by P. gingivalis and other pathogens as a safe entry portal to increase their intracellular survival [117]. Lymphocyte antigen 96 (LY96, MD2) is a critical component required for the TLR4 activation by LPS in the outer wall of P. gingivalis. It acts as the first defense line against bacterial infection [118]. LY96 expression is significantly increased in gingival tissues in periodontitis patients [119], leading to the TLR4-LY96-CD14 complexes formation which trigger the MyD88 signaling pathway, resulting in the production of TNF-?, IL-6, IL-8, and IL-2 [120]. Recent reports have detected that LY96 is closely associated with tumorigenesis and progression in diverse types of cancer, such as colon cancer [121], and GBM [122], with the highest expression being observed in GBM [123,124]. In GBM, TLR4 is commonly expressed on glioma tissues and microglia/macrophages [125]. The TLR4/MD2 complex signalling stimulation by LPS may involve tumour suppressor PTEN [126] loss or mutation, which can have a significant impact on cancer susceptibility and tumorigenesis. Complement C3 is an essential ingredient where classical, lectin, and alternative pathways connect, producing effector molecules such as C3a and C5a that activate C3aR and C5aR, respectively, resulting in leukocyte mobilization and activation [127]. Histological findings indicated that C3-activated complement fragments are in abundance in the periodontitis gingival crevices and significantly associated with inflammatory indices. After periodontitis treatment, levels of complement C3 significantly decrease (Top 5% genes), whereas they are present at lower levels or absent in healthy individuals [127]. Mechanistically, C3 activation may promote periodontal inflammation commonly by increasing vascular permeability and inflammatory cells chemotactic recruitment through C5aR activation, increasing vascular permeability and inflammatory exudates flow and inflammatory cells [128] chemotactic recruitment, however it is not able to control the infection [129]. Consequently, C3 is currently identified as one of the 21 most promising candidate genes for treatment of periodontitis [130]. Complement-activating proteins high levels may be beneficial for tumors [131]. It has been observed that C3 deposition was revealed in GBM tissues, suggesting local activation of complement in GBM, and confirmed the complement C3 protective effect on GBM development and progression [132]. It is important to highlight that glioma stem cells (GCS) may activate C3 with the alternative pathways assist and activate STAT-3, ERK2/1, and PI3K/Akt/mTOR signaling pathways to retain their pluripotent status [132]. Moreover, hypoxic conditions contribute to C3 activation and amplify C3a-C3aR effects [133], generating an additional effector mechanism for GSC survival, self-renewal, and tumour growth. A recent report analysed the immune characteristics of GBM and PD using immune infiltration, and showed decreased macrophage M2 polarization in PD, which negatively associated with key crosstalk gene expression and was consistent with a PD pro-inflammatory demonstration [134]. Increased macrophage M2 polarization in GBM positively associated with key cross-talk gene expression and was consistent with an immunosuppressive tumour microenvironment in GBM [135].The discrepancies in gene effects may primarily be attributed to the separate cells in which interacting genes function and the diverse inflammatory factors secretion. For instance, in GBM cells, the CXCR4 up-regulation recruit’s glioma-associated microglia/macrophages (GAMs) and leads to macrophages M2 polarization [136]. On the contrary, in periodontal inflammation, the CXCR4 up-regulation inhibits TLR4-induced NF-?B activation through the LPS-CXCR4 axis, thereby suppressing macrophages M2 polarization [137]. Moreover, it is also associated with the lack of other immune cell types, such as insufficiently activated CD8+ T cells and functionally impaired microglia in GBM [138,139]. Those observations indicate that these shared differentially expressed genes may bridge the common pathogenesis of GBM and PD by affecting immune cells. The exploration of the mechanism of crosstalk between PD and GBM [109] showed that P. gin-givalis, the primary causative factor of PD, activates complement C3 or CXCR4, deteriorate the killing capacity of macrophages and neutrophils and fail to control the infection [127,129,140]. Therefore P. gingivalis proliferates excessively in an inflammatory environment. Moreover, P. gingivalis spreads distantly with the blood circulation. Thereafter, LPS binding to LY96 leads to the blood-brain barrier disruption, triggering chronic and insidious inflammation in the brain, promoting glial cell carcinogenesis and exacerbating tumor development [141]. Although glioma-associated microglia and macrophages (GAMs) and MDSC are recruited into the glioma microenvironment and release diverse growth factors and cytokines, CXCR4 and C3 abnormal expression results in immunosuppression [142], leading to cancer cells limited clearance.


Conclusions

PD may are responsible for production of pro-inflammatory cytokines and bacterial products in the brain. Those products affect the brain as are able to increase the BBB permeability directly or indirectly by inducing the recruitment of other inflammatory cells. Chronic neuro-inflammation is triggered by systemic inflammation as a consequence of PD. AD is the most common reported neurodegenerative disease which has been associated with periodontitis, and that association is overall significant. For PSD and MS, those association appear to be weak. The possible role of PD in GBM pathogenesis remains unclear, whereas in one study only was recorded that three genes may play a role in the crosstalk between those diseases through immune pathways and provided new perspective data regarding the co-pathogenesis of PD and GBM.


References

  1. Ostrom QT, Patil N, Cioffi G, Waite K, Kruchko C, Barnholtz-Sloan JS. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013-2017. Neuro Oncol. 2020; 22: iv1-iv96.
  2. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016; 131: 803-820.
  3. Ceccarelli M, Barthel FP, Malta TM, Sabedot TS, Salama SR, Murray B. A, et al. Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse Glioma. Cell. 2016; 164: 550-563.
  4. Brat DJ, Verhaak RG, Aldape KD, Yung WK, Salama, SR, Cooper LA, et al. Cancer Genome Atlas Research Network. Comprehensive, Integrative Genomic Analysis of Diffuse Lower Grade Gliomas. N Engl J Med. 2015; 372: 2481-2498.
  5. Tan AC, Ashley DM, Lopez GY, Malinzak M, Friedman HS, Khasraw M. Management of glioblastoma: State of the art and future directions. CA Cancer J Clin. 2020; 70: 299-312.
  6. Alifieris C, Trafalis DT. Glioblastoma multiforme: Pathogenesis and treatment. Pharmacol Thera. 2016; 152: 63-82.
  7. Ohba S, Hirose Y. Current and future drug treatments for glioblastomas. Curr Med Chem. 2016; 23: 4309-4316.
  8. Weller M, Bent M, Hopkins K, Tonn JC, Stupp R, Falini A, et al. EANO guideline for the diagnosis and treatment of anaplastic gliomas and glioblastoma. Lancet Oncol. 2014; 15: 395-403.
  9. Gil-Gil MJ, Mesia C, Rey M, Bruna J. Bevacizumab for the treatment of glioblastoma. Clin Med Insights Oncol. 2013; 7:123-135.
  10. Daniels LB, Shaya M, Nordberg ML, Shorter CD, Fowler M, Nanda A. Glioblastoma multi forme in two non-nuclear family members. J La State Med Soc. 2007; 159: 215-322.
  11. Cobbs CS. Evolving evidence implicates cytomegalovirus as a promoter of malignant glioma Pathogenesis. Herpesviridae. 2011; 2:10.
  12. Spinelli V, Chinot O, Cabaniols C, Giorgi R, Alla P, Michel LMP. Occupational and Environmental risk factors for brain cancer: a pilot case-control study in France. La Press Med. 2010; 39: 35-44.
  13. Rakoff-Nahoum S. Why cancer and inflammation? Yale J Biol Med. 2006; 79: 123-130.
  14. Padmalath C, Harruff RC, Ganick D, Hafez GB. Glioblastoma multiforme with tuberous sclerosis. Report of a case. Arch Pathol Lab Med. 1980; 104: 649-650.
  15. Grips E, Wentzensen N, Sutter C, Sedlaczek O, Gebert J, Weigel R, et al. Glioblastoma multiforme as a manifestation of Turcot syndrome. Nervenarzt. 2002; 73: 177-182.
  16. Ortiga SR, Carreno BE, Perez MO, Alfonso PA. Glioblastoma multiforme and multiple endocrine neoplasic type 2 A. Med Clin Barc. 2009; 133: 196-197.
  17. Moorthy RK, Rajshekhar V. Development of glioblastoma multiforme following traumatic cerebral contusion: case report and review of literature. Surg Neurol. 2004; 61: 180-184.
  18. Kleihues P, Burger PC, Aldape KD. Glioblastoma, in: WHO classification of tumors of the central nervous system 4th, International Agency for Research on Cancer IACC, 2007.
  19. Peres MA, Macpherson LMD, Weyant RJ, Daly B, Venturelli R, Mathur MR, et al. Oral diseases: A global public health challenge. Lancet. 2019; 394: 249-260.
  20. WHO Regional Office for Europe. 2021.
  21. Gopinath D, Menona RK, Veettil SK, Botelho MG, Johnson NW. Periodontal diseases as putative risk factors for head and neck cancer: Systematic review and meta-analysis. Cancers. 2020: 12, 1893.
  22. Eke PI, Dye BA, Wei L, Slade GD, Thornton-Evans GO, Borgnakke WS, et al. Update on Prevalence of Periodontitis in Adults in the United States: NHANES 2009 to 2012. J Periodontol. 2015; 86: 611-622.
  23. Bartold PM, Van Dyke TE. Periodontitis: a host-mediated disruption of microbial homeostasis. Unlearning learned concepts. Periodontol 2000. 2013; 62: 203-217.
  24. Slots J. Periodontitis: facts, fallacies and the future. Periodontol 2000. 2017; 75: 7-23.
  25. Linden GJ, Lyons A, Scannapieco FA. Periodontal systemic associations: review of the evidence. J Periodontol. 2013; 84: S8-S19.
  26. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002; 420 (6917): 860-867.
  27. Cardoso EM, Reis C, Manzanares-Cespedes MC. Chronic periodontitis, inflammatory cytokines, and interrelationship with other chronic diseases. Postgrad Med. 2018; 130: 98-104.
  28. Curtis MA, Diaz PI, van Dyke TE. The role of the microbiota in periodontal disease. Periodontology 2000. 2020; 83: 14-25.
  29. Jaramillo A, Arce RM, Herrera D, Betancourth M, Botero JE, Contreras A. Clinical and and microbiological characterization of periodontal abscesses. J Clin Periodontol. 2005; 32, 1213-1218.
  30. Highfield J. Diagnosis and Classification of Periodontal Disease. Aust Dent J. 2009; 54: S11-S26.
  31. Hwang IM, Sun LM, Lin CL, Lee C-F, Kao C-H. Periodontal disease with treatment reduces subsequent cancer risks. QJM. 2014; 107:805-812.
  32. Zeng XT, Xia LY, Zhang YG, Li S, Leng WD, Kwong JS. Periodontal disease and incident lung cancer risk: a meta-analysis of cohort studies. J Periodontol. 2016; 87: 1158-64.
  33. Fitzpatrick SG, Katz J. The association between periodontal disease and cancer: a review of the literature. J Dent. 2010; 38: 83-95.
  34. Ahn J, Segers S, Hayes RB. Periodontal disease, Porphyromonas gingivalis serum antibody levels and orodigestive cancer mortality. Carcinogenesis. 2012; 33:1055-1058.
  35. Corbella S, Veronesi P, Galimberti V, Weinstein R, Del Fabbro M, Francetti L. Is periodontitis a risk indicator for cancer? A meta-analysis. PLoS One. 2018; 13: e0195683.
  36. Gao Z, Weng X, Yu D, Pan Z, Zhao M, Cheng B, et al. Porphyromonas gingivalis-derived lipopolysaccharide promotes glioma cell proliferation and migration via activating Akt signaling pathways. Cells. 2022; 11: 4088.
  37. Yao G, Zhang W, Yang M, Yang H, Wang J, Zhang H, et al. Micro Pheno DB Associates Meta-genomic Data with Pathogenic Microbes, Microbial Core Genes, and Human Disease Phenotypes. Genom Proteom Bioinform. 2020; 18: 760-772.
  38. Nagy KN, Sonkodi I, Szoke I, Nagy E, Newman HN. The microflora associated with human oral carcinomas. Oral Oncol. 1998; 34: 304-308.
  39. Michaud DS. Role of bacterial infections in pancreatic cancer. Carcinogenesis. 2013; 34: 2193-2197.
  40. Kurkivuori J, Salaspuro V, Kaihovaara P, Kari K, Rautemaa R, Gronroos L, et al. Aceraldehyde production from ethanol by oral streptococci. Oral Oncol. 2007; 43: 181-186.
  41. Meurman JH, Uittamo J. Oral micro-organisms in the etiology of cancer. Acta Odontol Scand. 2008; 66: 321-326.
  42. Nwizu N, Wactawski-Wende J, Genco RJ. Periodontal disease and cancer: Epidemiologic studies and possible mechanisms. Periodontology 2000. 2020; 83: 213-233.
  43. Gholizadeh P, Eslami H, Yousefi M, Asgharzadeh M, Aghazadeh M, Kafil HS. Role of oral microbiome on oral cancers, a review. Biomed Pharmacother. 2016; 84: 552-558.
  44. Bansal M, Rastogi S, Vineeth NS. Influence of periodontal disease on systemic disease: Inversion of a paradigm: A review. J Med Life 2013; 6: 126-130.
  45. Michaud DS, Izard J, Wilhelm-Benartzi CS, You DH, Grote VA, Tjonneland A, et al. Plasma antibodies to oral bacteria and risk of pancreatic cancer in a large European prospective co-hort study. Gut. 2013; 62: 1764-1770.
  46. Fan X, Alekseyenko AV, Wu J, Peters BA, Jacobs EJ., Gapstur SM, et al. Human oral microbiome and prospective risk for pancreatic cancer: a population-based nested case-control study. Gut. 2018; 67: 120-127.
  47. Kamer AR, Pirraglia E, Tsui W, Rusinek H, Vallabhajosula S, Mosconi L, et al. Periodontal disease associates with higher brain amyloid load in normal elderly. Neurobiol Aging. 2015; 36: 627-633.
  48. Dominy SS, Lynch C, Ermini F, Benedyk M, Marczyk A, Konradi A, et al. Porphyromonas gingivalis in Alzheimer’s disease brains: Evidence for disease causation and treatment with small-molecule inhibitors. Sci Adv. 2019; 5: eaau3333.
  49. Hallikainen J, Lindgren A, Savolainen J, Selander T, Jula A, Narhi M, et al. Periodontitis and gingival bleeding associate with intracranial aneurysms and risk of aneurysmal subarachnoid hemorrhage. Neurosurg Rev. 2020; 43: 669-679.
  50. Poole S, Singhrao SK, Kesavalu L, Curtis MA, Crean S. Determining the Presence of Periodontopathic Virulence Factors in Short-Term Postmortem Alzheimer’s Disease Brain Tissue. J Alzheimers Dis. 2013; 36: 665-677.
  51. Costa MJF, de Araujo IDT, da Rocha Alves L, da Silva RL, Dos Santos Calderon P, Borges BCD, et al. Relationship of Porphyromonas gingivalis and Alzheimer’s disease: A systematic review of pre-clinical studies. Clin Oral Investig. 2021; 25: 797-806.
  52. Sato N, Matsumoto T, Kawaguchi S, Seya K, Matsumiya T, Ding J, et al. Porphyromonas gingivalis lipopolysaccharide induces interleukin-6 and c-c motif chemokine ligand 2 expression in cultured hCMEC/D3 human brain microvascular endothelial cells. Gerodontology. 2022; 39: 139-147.
  53. Wen Y, Feng L, Wang H, Zhou H, Li Q, Zhang W, et al. Association between oral microbiota and human brain glioma grade: A case-control study. Front Microbiol. 2021; 12:746568.
  54. Zhu C, Li F, Wong MC, Feng XP, Lu HX, Xu W. Association between Herpesviruses and Chronic Periodontitis: A Meta-Analysis Based on Case-Control Studies. PloS one. 2015; 10(12):e0144319.
  55. Mohammad AA, Rahbar A, Lui WO, Davoudi B, Catrina A, Stragliotto G, et al. Detection of circulating hcmv-miR-UL112-3p in patients with glioblastoma, rheumatoid arthritis, diabetes mellitus and healthy controls. PloS one. 2014; 9(12): e113740.
  56. Yaiw KC, Ovchinnikova O, Taher C, Mohammad AA, Davoudi B, Shlyakhto E, et al. High prevalence of human cytomegalovirus in carotid atherosclerotic plaques obtained from Russian patients undergoing carotid endarterectomy. Herpesviridae. 2013; 4: 3.
  57. Nath SG, Raveendran R. Microbial dysbiosis in periodontitis. J Indian Soc Periodontol. 2013; 17: 543-545.
  58. Kamarajan P, Ateia I, Shin JM, Fenno JC, Le C, Zhan L, et al. 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.
  59. Gallimidi AB, Fischman S, Revach B, Bulvik R, Rubinstein AM, Nussbaum G, et al. Periodontal pathogens Porphyromonas gingivalis and Fusobacterium nucleatum promote tumor progression in an oral-specific chemical carcinogenesis model. Oncotarget. 2015; 6: 22613.
  60. Makkawi H, Hoch S, Burns E, Hosur K, Hajishengallis G, Kirschning CJ, et al. Porphyromonas gingivalis Stimulates TLR2-PI3K Signaling to escape immune clearance and induce bone resorption independently of MyD88. Front Cell Infect Microbiol. 2017; 7: 359.
  61. Han YW, Shi W, Huang GTJ, Kinder Haake S, Park NH, Kuramitsu H, et al. Interactions between periodontal bacteria and human oral epithelial cells: Fusobacterium nucleatum adheres to and invades epithelial cells. Infect Immun. 2000; 68: 3140-3146.
  62. Fardini Y, Chung P, Dumm R, Joshi N, Han YW. Transmission of diverse oral bacteria to murine placenta: Evidence for the oral microbiome as a potential source of intrauterine infection. Infect Immun. 2010; 78: 1789-1796.
  63. Kolenbrander PE, Palmer RJ, Periasamy S, Jakubovics NS. Oral multispecies biofilm development and the key role of cell-cell distance. Nat Rev Microbiol. 2010; 8: 471-480.
  64. Geng F, Liu J, Guo Y, Li C, Wang H, Wang, H, et al. Persistent exposure to Porphyromonas gingivalis promotes proliferative and invasion capabilities, and tumorigenic properties of human immortalized oral epithelial cells. Front Cell Infect Microbiol. 2017; 7: 57
  65. Harris TM, Du P, Kawachi N, Belbin TJ, Wang Y, Schlecht NF, et al. Proteomic analysis of oral cavity squamous cell carcinoma specimens identifies patient outcome-associated proteins. Arch Pathol Lab Med. 2015; 139: 494-507.
  66. Jeong E, Kim K, Kim JH, Cha GS, Kim SJ, Kang HS, et al. Porphyromonas gingivalis HSP 60 peptides have distinct roles in the development of atherosclerosis. Mol Immunol.2015; 63: 489-496.
  67. Lin FY, Huang CY, Lu HY, Shih CM, Tsao NW, Shyue SK, 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.
  68. Mao S, Park Y, Hasegawa Y, Tribble GD, James CE, Handfield M, et al. Intrinsic apoptotic pathways of gingival epithelial cells modulated by Porphyromonas gingivalis. Cell Microbiol. 2007; 9:1997-2007.
  69. Yilmaz O, Jungas T, Verbeke P, Ojcius DM. 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.
  70. Nakhjiri SF, Park Y, Yilmaz O, Chung WO, Watanabe K, El-Sabaeny A, et al. Inhibition of epithelial cell apoptosis by Porphyromonas gingivalis. FEMS Microbiol Lett. 2001; 200: 145 -149.
  71. Yao L, Jermanus C, Barbetta B, Choi C, Verbeke P, Ojcius DM, et al. Porphyromonas gingivalis infection sequesters pro-apoptotic bad through akt in primary gingival epithelial cells. Mol Oral Microbiol. 2010; 25: 89-101.
  72. Kuboniwa M, Hasegawa Y, Mao S, Shizukuishi S, Amano A, Lamont RJ, et al. P. gingivalis accelerates gingival epithelial cell progression through the cell cycle. Microbes Infect. 2008;10: 122-128.
  73. Liu J, Tang X, Li C, Pan C, Li Q, Geng F, et al. Porphyromonas gingivalis promotes the cell cycle and inflammatory cytokine production in periodontal ligament fibroblasts. Arch Oral Biol. 2015; 60: 1153-1161.
  74. O Brien-Simpson NM, Pathirana RD, Walker GD, Reynolds EC. Porphyromonas gingivalis RgpA-Kgp proteinase-adhesin complexes penetrate gingival tissue and induce proinflammatory cytokines or apoptosis in a concentration-dependent manner. Infect Immun. 2009; 77: 1246-1261.
  75. Benakanakere MR, Li Q, Eskan MA, Singh AV, Zhao J, Galicia JC, et al. Modulation of TLR 2 protein expression by MiR-105 in human oral keratinocytes. J Biol Chem. 2009; 284: 23107-23115.
  76. Moffatt CE, Lamont RJ. Porphyromonas gingivalis induction of MicroRNA-203 expression controls suppressor of cytokine signaling 3 in gingival epithelial cells. Infect Immun. 2011; 79: 2632-2637.
  77. 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.
  78. Polak D, Wilensky A, Shapira L, Halabi A, Goldstein D, Weiss EI, et al. Mouse model of experimental periodontitis induced by Porphyromonas gingivalis/Fusobacterium nucleatum infection: Bone loss and host response. J Clin Periodontol. 2009; 36: 406-410.
  79. Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-Cadherin/?-Catenin Signaling via its FadA adhesin. Cell Host Microbe. 2013; 14; 195-206.
  80. Mu W, Jia Y, Chen X, Li H, Wang Z, Cheng B. Intracellular Porphyromonas gingivalis promotes the proliferation of colorectal cancer cells via the MAPK/ERK signaling pathway. Front Cell Infect Microbiol. 2020; 10: 812.
  81. Nizami S, Hall-Roberts H, Warrier S, Cowley SA, Di Daniel E. Microglial inflammation and phagocytosis in Alzheimer’s disease: potential therapeutic targets. Br J Pharmacol. 2019; 176(18):3515-3532.
  82. Jurga AM, Paleczna M, Kuter KZ. Overview of general and discriminating markers of differential microglia phenotypes. Front Cell Neurosci. 2020.
  83. Li X, Kiprowska M, Kansara T, Kansara P, Li P. Neuroinflammation: A Distal Consequence of Periodontitis. J Dent Res. 2022; 101: 1441-1449.
  84. Wilkinson K, El Khoury J. Microglial scavenger receptors and their roles in the pathogenesis of Alzheimer’s disease. Int J Alzheimers Dis. 2012; 489456.
  85. Doens D, Fernández PL. Microglia receptors and their implications in the response to amyloid ? for Alzheimer’s disease pathogenesis. J Neuroinflamm. 2014; 11: 48.
  86. Schetters STT, Gomez-Nicola D, Garcia-Vallejo JJ, Van Kooyk Y. Neuroinflammation: microglia and T cells get ready to Tango. Front Immunol. 2018.
  87. Luo P, Chu SF, Zhang Z, Xia CY, Chen NH. Fractalkine/CX3CR1 is involved in the crosstalk between neuron and glia in neurological diseases. Brain Res Bull. 2019; 146: 12-21.
  88. Kumar V. Toll-like receptors in the pathogenesis of neuroinflammation. J Neuroimmunol. 2019; 332:16-30.
  89. Reed-Geaghan EG, Savage JC, Hise AG, Landreth GE. CD14 and toll-like receptors 2 and 4 are required for fibrillar A?-stimulated microglial activation. J Neurosci. 2009; 29:11982-11992.
  90. Hemonnot AL, Hua J, Ulmann L, Hirbec H. Microglia in Alzheimer disease: well-known targets and new opportunities. Front Aging Neurosci. 2019; 11: 233.
  91. Ghosh P, Singh R, Ganeshpurkar A, Pokle AV, Singh RB, Singh SK, et al. Cellular and molecular influencers of neuro-inflammation in Alzheimer’s disease: recent concepts and roles. Neurochem Int. 2021; 151: 105212.
  92. Leng F, Edison P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat Rev Neurol. 2021; 17: 157-172.
  93. Ilievski V, Zuchowska PK, Green SJ, Toth PT, Ragozzino ME, Le K, et al. Chronic oral application of a periodontal pathogen results in brain inflammation, neuro-degeneration and amyloid beta production in wild type mice. PLoS ONE 2018, 13, e0204941.
  94. Kantarci A, Tognoni CM, Yaghmoor W, Marghalani A, Stephens D, Ahn JY, et al. Microglial response to experimental periodontitis in a murine model of Alzheimer’s disease. Sci Rep. 2020; 10: 18561.
  95. Hu Y, Li H, Zhang J, Zhang X, Xia X, Qiu C, et al. Periodontitis induced by P. gingivalis- LPS is associated with neuro-inflammation and learning and memory impairment in Sprague Dawley rats. Front Neurosci. 2020; 14: 658.
  96. Bell JS, Spencer JI, Yates RL, Yee SA, Jacobs BM, DeLuca GC. Invited review: from nose to gut-the role of the microbiome in neurological disease. Neuropathol Appl Neurobiol. 2029; 45: 195-215.
  97. Sureda A, Daglia M, Arguelles Castilla S, Sanadgol N, Fazel Nabavi S, Khan H, et al. Oral microbiota and Alzheimer’s disease: do all roads lead to Rome? Pharmacol Res. 2020; 151: 104582.
  98. Teixeira FB, Saito MT, Matheus FC, Prediger RD, Yamada ES, Maia CSF, et al. Periodontitis and Alzheimer’s disease: a possible comorbidity between oral chronic inflammatory condition and neuroinflammation. Front Aging Neurosci. 2017; 9: 327.
  99. Riviere GR, Riviere KH, Smith KS. Molecular and immunological evidence of oral Treponema in the human brain and their association with Alzheimer’s disease. Oral Microbiol Immunol. 2002; 17: 113-118.
  100. Mao S, Huang C-P, Lan H, Lau H-G, Chiang C-P, Chen Y-W. Association of periodontitis and oral microbiomes with Alzheimer’s disease: A narrative systematic review. J Dent Sci. 2022; 17: 1762-1779.
  101. Bazan N, Halabi A, Ertel M, Petasis N. Neuroinflammation. In Basic Neurochemistry:Molecular, Cellular,and Medical Aspects,8th ed.; McGraw Hill: New York, NY, USA, 2012; pp. 610-620.
  102. Pizza V, Agresta A, D’Acunto CW, Festa M, Capasso A. Neuro-inflammaging and neurodegenerative diseases: An overview. CNS Neurol Disord Drug Targets. 2011; 10: 621-634.
  103. Dorn BR, Dunn WA, Progulske-Fox A. Porphyromonas gingivalis Traffics to Autophagosomes in Human Coronary Artery Endothelial Cells. Infect Immun. 2001; 69: 5698-5708.
  104. Singhrao SK, Chukkapalli S, Poole S, Velsko I, Crean SJ, Kesavalu L. Chronic Porphyromonas gingivalis infection accelerates the occurrence of age-related granules in ApoE-/- mice brains. J Oral Microbiol. 2017; 9: 1270602.
  105. Singhrao SK, Olsen I. Assessing the role of Porphyromonas gingivalis in periodontitis to determine a causative relationship with Alzheimer’s disease. J Oral Microbiol. 2019; 11:1563405.
  106. Wu DT, Cho YW, Spalti MD, Bishara M, Nguyen TT. The link between periodontitis and Alzheimer’s disease-Emerging clinical evidence. Dent Rev. 2023; 3: 100062.
  107. Wu H, Qiu W, Zhu X, Li X, Xie Z, Carreras I, et al. The Periodontal Pathogen Fusobacterium nucleatum Exacerbates Alzheimer’s Pathogenesis via Specific Pathways. Front Aging Neurosci. 2022; 14: 912709.
  108. Hajishengallis, G. Immunomicrobial pathogenesis of periodontitis: Keystones, pathobionts, and host response. Trends Immunol. 2013, 35, 3-11.
  109. Huang J-H, Chen Y, Kang Y-B, Yao Z-J, Song J-Y. The potential crosstalk genes and molecular mechanisms between glioblastoma and periodontitis. Sci Rep. 2024; 14:5970
  110. Das S, Goldstone AB, Wang H, Farry J, D'Amato G, Paulsen MJ, et al. A unique collateral artery development program promotes neonatal heart regeneration. Cell. 2019; 176: 1128-1142.e1118.
  111. Wu Y, Zhang C, Xu W, Zhang J, Zheng Y, Lu Z, et al. CXC motif chemokine receptor 4 gene polymorphism and cancer risk. Medicine (Baltimore). 2016; 95: e5317.
  112. Khan AB, Lee S, Harmanci AS, Patel R, Latha K, Yang Y, et al. CXCR4 expression is associated with proneural-to-mesenchymal transition in glioblastoma. Int J Cancer. 2023; 152: 713-724.
  113. Le Rhun E, Preusser M, Roth P, Reardon DA, van den Bent M, Wen P, et al. Molecular targeted therapy of glioblastoma. Cancer Treat Rev. 2019; 80: 101896.
  114. Ramaswamy P, Nanjaiah ND, Borkotokey M. Role of MEK-ERK signaling mediated adhesion of glioma cells to extra-cellular matrix: Possible implication on migration and proliferation. Ann Neurosci. 2019; 26: 52-56.
  115. Nagashima H, Shinoda M, Honda K, Kamio N, Watanabe M, Suzuki T, et al. CXCR4 signaling in macrophages contributes to periodontal mechanical hypersensitivity in Porphyromonas gingivalis-induced periodontitis in mice. Mol Pain 2017; 13: 1744806916689269.
  116. Pierce DL, Nishiyama S-I, Liang S, Wang M, Triantafilou M, Triantafilou K, et al. Host adhesive activities and virulence of novel fimbrial proteins of Porphyromonas gingivalis. Infect Immun. 2009; 77: 3294-3301.
  117. Hajishengallis G, McIntosh ML. Nishiyama SI, Yoshimura F. Mechanism and implications of CXCR4-mediated integrin activation by Porphyromonas gingivalis. Mol Oral Microbiol. 2013; 28: 239-249.
  118. Dou T, Fu M, Wang Y, Wang Z, Wang Z, Bian Z, et al. Signatures of positive selection in LY96 gene in vertebrates. J Biosci. 2013; 38: 899-904.
  119. Li J, Chen Y, Ng CHC, Fung M-L, Xu A, Cheng B, et al. Differential expression of Toll-like receptor 4 in healthy and diseased human gingiva. J Periodontal Res.2014; 49: 845-854.
  120. Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity. 2011; 34: 637-650.
  121. Rajamanickam V, Yan T, Xu S, Hui J, Xu X, Ren L, et al. Selective targeting of the TLR4 co-receptor, MD2, prevents colon cancer growth and lung metastasis. Int J Biol Sci. 2020; 16:1288-1302.
  122. Zhao M, Li X, Chen Y, Wang S. MD2 is a potential biomarker associated with immune cell infiltration in gliomas. Front Oncol. 2022; 12: 854598.
  123. Nie K, Li J, Peng L, Zhang M, Huang W. Pan-cancer analysis of the characteristics of LY96 in prognosis and immunotherapy across human cancer. Front Mol Biosci. 2022; 9: 837393.
  124. Moretti IF, Lerario AM, Trombetta-Lima M, Sola PR, da Silva Soares R, Oba-Shinjo SM, et al. Late p65 nuclear translocation in glioblastoma cells indicates non-canonical TLR4 signaling and activation of DNA repair genes. Sci Rep. 2021:11: 1333.
  125. Xun Y, Yang H, Kaminska B, You H. Toll-like receptors and toll-like receptor targeted immunotherapy against glioma. J Hematol Oncol. 2021; 14: 176.
  126. Moretti IF, Franco DG, de Almeida Galatro TF, Oba-Shinjo SM, Marie SKN. Plasmatic membrane toll-like receptor expressions in human astrocytomas. PLoS One. 2018; e0199211.
  127. Hajishengallis G. Complement and periodontitis. Biochem Pharmacol 2010; 80:1992-2001.
  128. Krauss JL, Potempa J, Lambris JD, Hajishengallis G. Complementary Tolls in the periodontium: How periodontal bacteria modify complement and Toll-like receptor responses to prevail in the host. Periodontol 2000. 2010; 52: 141-162.
  129. Delima AJ, Van Dyke TE. Origin and function of the cellular components in gingival crevice fluid. Periodontology 2000. 2003; 31: 55-76.
  130. Zhan Y, Zhang R, Lv H, Song X, Xu X, Chai L, et al. Prioritization of candidate genes for periodontitis using multiple computational tools. J Periodontol. 2014; 85:1059-1069.
  131. Markiewski MM, Lambris JD. Is complement good or bad for cancer patients? A new perspective on an old dilemma. Trends Immunol. 2009; 30: 286-292.
  132. Bouwens TA, Trouw LA, Veerhuis R, Dirven CMF, Lamfers MLM, Al-Khawaja H. Complement activation in glioblastoma multiforme pathophysiology: Evidence from serum levels and presence of complement activation products in tumor tissue. J Neuroimmunol. 2015; 271-276.
  133. Pedersen ED, Froyland E, Kvissel A-K, Pharo AM, Skalhegg BS, Rootwelt T, et al. Expression of complement regulators and receptors on human NT2-N neurons-Effect of hypoxia and reoxygenation. Mol Immunol. 2007; 44: 2459-2468.
  134. Almubarak A, Tanagala KKK, Papapanou PN, Lalla E, Momen-Heravi F. Disruption of monocyte and macrophage homeostasis in periodontitis. Front Immunol. 2020; 330.
  135. Walters MJ, Ebsworth K, Berahovich RD, Penfold MET, Liu S-C, Omran RAI, et al. Inhibition of CXCR7 extends survival following irradiation of brain tumours in mice and rats. Br J Cancer. 2014; 110: 1179-1188.
  136. Mercurio L, Ajmone-Cat MA, Cecchetti S, Ricci A, Bozzuto G, Molinari A, et al. Targeting CXCR4 by a selective peptide antagonist modulates tumor microenvironment and microglia reactivity in a human glioblastoma model. J Exp Clin Cancer Res.2016; 55.
  137. Lin YH, Wang Y-H, Peng Y-J, Liu F-C, Lin G-J, Huang S-H, et al. Interleukin 26 skews macrophage polarization towards M1 phenotype by activating cJUN and the NF-?B pathway. Cells. 2020; 9: 938.
  138. Qian, J. Feifei Luo, Jiao Yang, Jun Liu, Ronghua Liu , Luman Wang, et al. TLR2 promotes glioma immune evasion by downregulating MHC class II molecules in microglia. Cancer Immunol Res. 2018; 6: 1220-1233.
  139. Locarno CV, Simonelli M, Carenza C, Capucetti A, Stanzani E, Lorenzi E, et al. Role of myeloid cells in the immunosuppressive microenvironment in gliomas. Immunobiology. 2020; 225: 151853.
  140. Hajishengallis G, Lambris JD. Microbial manipulation of receptor crosstalk in innate immunity. Nat Rev Immunol. 2011; 11:187-200.
  141. Balkwill F, Mantovani A. Inflammation and cancer: Back to Virchow? Lancet. 2001; 539-545.
  142. Hambardzumyan D, Gutmann DH, Kettenmann H. The role of microglia and macrophages in glioma maintenance and progression. Nat Neurosci. 2016; 19: 20-27.