Skip to main content
Erschienen in: BMC Oral Health 1/2023

Open Access 01.12.2023 | Research

A systematic review of the impact of Porphyromonas gingivalis on foam cell formation: Implications for the role of periodontitis in atherosclerosis

verfasst von: Saeed Afzoon, Mohammad Amin Amiri, Mostafa Mohebbi, Shahram Hamedani, Nima Farshidfar

Erschienen in: BMC Oral Health | Ausgabe 1/2023

Abstract

Background

The current literature suggests the significant role of foam cells in the initiation of atherosclerosis through the formation of a necrotic core in atherosclerotic plaques. Moreover, an important periodontal pathogen called Porphyromonas gingivalis (P. gingivalis) is indicated to play a significant role in this regard. Thus, the aim of this systematic review was to comprehensively study the pathways by which P. gingivalis as a prominent bacterial species in periodontal disease, can induce foam cells that would initiate the process of atherosclerosis formation.

Methods

An electronic search was undertaken in three databases (Pubmed, Scopus, and Web of Science) to identify the studies published from January 2000 until March 2023. The risk of bias in each study was also assessed using the QUIN risk of bias assessment tool.

Results

After the completion of the screening process, 11 in-vitro studies met the inclusion criteria and were included for further assessments. Nine of these studies represented a medium risk of bias, while the other two had a high risk of bias. All of the studies have reported that P. gingivalis can significantly induce foam cell formation by infecting the macrophages and induction of oxidized low-density lipoprotein (oxLDL) uptake. This process is activated through various mediators and pathways. The most important factors in this regard are the lipopolysaccharide of P. gingivalis and its outer membrane vesicles, as well as the changes in the expression rate of transmembrane lipid transportation channels, including transient receptor potential channel of the vanilloid subfamily 4 (TRPV4), lysosomal integral protein 2 (LIMP2), CD36, etc. The identified molecular pathways involved in this process include but are not limited to NF-κB, ERK1/2, p65.

Conclusion

Based on the results of this study, it can be concluded that P. gingivalis can effectively promote foam cell formation through various pathogenic elements and this bacterial species can affect the expression rate of various genes and the function of specific receptors in the cellular and lysosomal membranes. However, due to the moderate to high level of risk of bias among the studies, further studies are required in this regard.
Hinweise
Saeed Afzoon and Mohammad Amin Amiri contributed equally to this work.

Publisher’s Note

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

Background

Periodontal diseases are inflammatory conditions which affect the periodontal tissue and consequently result in soft tissue recession, bone loss, tooth loss, and mild elevation of systemic inflammatory factors [15]. Based on the current evidence, 20–50% of the global health population are affected by periodontal diseases; therefore, its high prevalence makes it an important public health issue [6, 7]. Approximately 700 species of bacteria are identified in the oral cavity and it is proposed that the interaction of periodontal pathogens and host response can lead to periodontal diseases [8, 9].
Many pathogens are associated with the development of periodontitis among which Porphyromonas gingivalis (P. gingivalis) acts as a critical factor in the progression of periodontal pathologies [10, 11]. This process is mediated through the modified expression of multiple growth factors in periodontal tissues [1214]. This bacterial species produces different virulence factors which could induce and sustain systemic inflammation [15]. P. gingivalis is also able to degenerate the tissue and cause local and systemic pathologies [15]. Recent studies have indicated a possible association of P. gingivalis with different systemic diseases, such as cardiovascular, cerebral, pulmonary, digestive, bone, and perinatal disease [16]. Among all the mentioned systemic conditions, one of the most noticeable diseases with high cardiovascular complications is atherosclerosis [17].
Atherosclerosis is a lipid-driven inflammatory disease caused by dysregulation of lipid metabolism resulting in the accumulation of lipid droplets in the matrix beneath an endothelial layer of arteries [18, 19]. This vascular pathology is one the main causes of cardiovascular diseases, heart failure, stroke, and myocardial infarction [20, 21]. It can also lead to vascular complications, such as coronary artery disease, carotid artery disease, and peripheral arterial disease [22]. One of the major processes playing a crucial role in the occurrence of atherosclerosis is foam cell formation [23]. The increase in cholesterol level makes the arteries more permeable which results in monocytes infiltration to the sub-endothelial layer where they convert into macrophages [15, 19, 24, 25]. Excessive uptake of lipids and oxidized low-density lipoprotein (oxLDL) stored in macrophage cytoplasm eventually changes the macrophage metabolism [23]. Consequently, immoderate accumulation of oxLDL in macrophage cytoplasm exceeds the capacity of macrophage to continue normal lipid metabolism [2628]. This process eventually results in macrophage apoptosis and gradual formation of foamy cells [2628].
Studies have declared a marked correlation between atherosclerosis and periodontitis [2932]. Therefore, the process of foam cell formation, as one of the major mechanisms of atherosclerosis, could be affected by the presence of P. gingivalis in patients who have developed periodontitis [33]. It has been reported that P. gingivalis has been found in arterial plaque in humans and mice [30, 34]. The abilities of this bacteria to circumvent the immune system could contribute to the induction and progression of atherosclerosis [30, 31]. Besides, it was found that P. gingivalis is able to accelerate lipid peroxidation and the progression of atherosclerosis in the presence of oxLDL [32]. Several studies have indicated that this process is carried out through the infection of macrophages in the arterial intima layer with P. gingivalis [3537]. In order to obtain a comprehensive insight into this process, we have performed a systematic review of the different mechanisms by which P. gingivalis can induce foam cell formation.

Materials and methods

Protocol development

This systematic review follows the guidelines recommended by The Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) statement 2020 [38, 39].
The PubMed, Scopus, and Web of Science databases were searched to identify the articles reporting the association of P. gingivalis with foam cell formation from January 1, 2000, until March 12, 2023. In addition, independently, a manual search was conducted by two authors, in an act of perusing reference lists of included papers to find further studies associated with the topic.
We searched the mentioned databases using the following combination of free-text terms:
(Foam Cells OR Macrophages) AND (Porphyromonas gingivalis OR Porphyromonas OR Bacteroides gingivalis).

Eligibility criteria

Table 1 illustrates the eligibility criteria for the aspects of participants, intervention, comparison, outcomes, and study design (PICOS). All in vitro studies investigating the possible effect of P. gingivalis on foam cell formation were included in this review. The quantity of foam cell formation had to be explored in the presence of P. gingivalis in comparison to the control group in which no P. gingivalis was present. Studies evaluating the effect of P. gingivalis in the absence of the control group were excluded. Additionally, studies other than in-vitro studies such as ex-vivo studies, in-vivo studies, etc. were excluded. Studies in languages other than English or Persian were excluded from our review considering the linguistic competency of the research team.
Table 1
Representation of the PICOS of the systematic review
PICOS
Inclusion Criteria
Exclusion Criteria
Population
Studies assessing cultured macrophage cells
Studies assessing cells other than macrophages
Intervention
Studies evaluating the effect of P. gingivalis
Studies evaluating the effect of bacteria other than P. gingivalis
Comparison
Studies evaluating a group of macrophages without exposure to any bacterial species
-
Outcome
Studies assessing the rate of foam cell formation from macrophage cells
-
Study Design
In-vitro studies
Case reports, narrative reviews, systematic reviews with or without meta-analysis, letters to the editors, short communications, in-vivo studies, ex-vivo studies, animal studies, and non-comparative studies.

Study selection

Based on the eligibility criteria, the authors (MAA, SA, and MM) screened the title and abstract of the retrieved articles independently. Furthermore, the retrieved articles were scrutinized for any possible predatory publication. In the case of disagreement, all the aforementioned researchers discussed the matter with other authors (SH and NF) to reach an agreement. The full texts of the selected articles were obtained, and studies meeting the inclusion criteria were included in our systematic review.

Data collection and data items

In a customized data extraction manner, the name of the authors, the year of publications, the type of evaluations, the evaluation methods, the main outcomes, the key molecular elements, and the mechanism of action were extracted.

Risk of bias assessment

In this systematic review, we used a novel risk of bias assessment tool named QUIN tool which was recently introduced by Sheth et al. in 2022 [40]. QUIN tool was mainly introduced for the evaluation of the risk of biases within the in-vitro studies in the field of dentistry. This risk of bias tool contains 12 criteria, and each of them, as represented in Table 2, can be scored as either 2 (adequately specified), 1 (poorly specified), 0 (not specified), or NA (not applicable). The total score is also estimated by the following formula:
$$\text{Final score}=\frac{Total score\times 100}{Number of applicable criteria\times 2}$$
Table 2
Assessment of risk of bias in each study using the QUIN tool
Criteria number
Criteria
Qi et al.
(2003)
[35]
Kuramitsu et al.
(2003)
[45]
Miyakawa et al.
(2004)
[36]
Giacona et al.
(2004)
[37]
Shaik-Dasthagirisaheb et al.
(2013)
[42]
Li et al.
(2013)
[47]
Shaik-Dasthagirisheb et al.
(2016)
[43]
Liang et al.
(2016)
[46]
Kim et al.
(2018)
[48]
Gupta et al.
(2019) [44]
Yang et al.
(2020)
[41]
1
Clearly stated aims/objectives
2
1
2
2
2
2
1
2
2
2
2
2
Detailed explanation of sample size calculation
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
3
Detailed explanation of the sampling technique
NA
NA
NA
2
2
NA
2
2
2
2
2
4
Details of the comparison group
2
1
1
1
2
1
1
2
2
2
1
5
Detailed explanation of the methodology
2
2
2
2
2
2
2
2
2
2
2
6
Operator details
0
0
0
0
0
0
0
0
0
0
0
7
Randomization
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
8
Method of measurement of outcome
2
2
2
2
2
2
2
2
2
2
2
9
Outcome assessor details
0
0
0
0
0
0
0
0
0
0
0
10
Blinding
0
0
0
0
0
0
0
0
0
0
0
11
Statistical analysis
0
0
0
2
2
2
0
2
2
2
2
12
Presentation of results
2
2
2
2
2
2
0
2
2
2
2
Total score
55.55%
44.44%
50%
65%
70%
61.11%
40%
70%
70%
70%
65%
Risk of bias
Medium
High
Medium
Medium
Medium
Medium
High
Medium
Medium
Medium
Medium
A total score above 70% indicates a low risk of bias, a total score between 50 and 70% suggests a medium risk of bias, and a total score less than 50% represents a high risk of bias in the study.

Results

Study selection

The initial search of three databases identified 2654 studies in total. After duplicate removal, 1656 abstracts and titles remained and underwent screening. A total of 1479 papers were excluded due to a mismatch with our search criteria and 177 articles were retained for eligibility assessment and full-text review among which 166 were screened by title and abstract and didn’t meet the eligibility criteria mentioned in Table 1. Finally, 11 original articles were included in this systematic review. The PRISMA chart below briefly represents the aforementioned process (Fig. 1).

Study characteristics

There were 11 studies eligible for the systemic review. Table 3 presents detailed individual characteristics, including study groups, type, and method of evaluation, and the outcomes of each study.
Table 3
Summary of included studies
Author
(Year)
Type(s) of Evaluation
Method(s)
Main Outcome(s)
References
Yang et al.
(2020)
Foam cell formation
Oil Red O staining
The knockdown of limp2 reduces the rate of foam cell formation and enhances cholesterol export. The interaction of LIMP2 and caveolin-1 (CAV1) in the lysosome of macrophages may play a key role in this regard.
[41]
Cathepsin L activity
Magic red cathepsin L assay
RNA sequencing
RT-PCR
Protein detection
Western blot
Protein detection
Co-immunoprecipitation
Gupta et al.
(2019)
Binding and uptake of oxLDL
Fluorescence intensity microscopy
TRPV4 plays a key part in foam cell formation and inflammatory genes upregulation, which is subsequent to LDL oxidation. This process was also induced by P. gingivalis LPS.
[44]
Foam cell formation
Oil Red O Staining
Expression levels of TRPV4, actin, or CD36
Immunoblot and immunofluorescence assay
Kim et al.
(2018)
Oxidation extent of HDL or LDL
TBARS assay
HDL incubated with P. gingivalis showed significantly higher oxidation levels and TNF- α production.
P. gingivalis induces HDL oxidation, by proinflammatory response in interaction with macrophages.
[48]
TNF-α
ELISA
The activity of MMPs, and Gelatinase
Electrophoresis,
Gelatin zymography
Foam cells
Oil Red O staining
Liang et al.
(2016)
Foam cell formation
Oil Red O staining
P. gingivalis can induce foam cell formation through the upregulation of CD36 expression in macrophages. CD36 expression in the presence of P. gingivalis is mediated by NF-κB, ERK1/2, and p65.
[46]
NF-κB activity
RT-PCR
CD36 protein levels
Western blot
NF-κB activity
Luciferase reporter assay
The interaction of NF-κB and CD36 promoters
EMSA
The interaction of NF-κB and CD36 promoters
Chromatin immunoprecipitation assay
CD36 protein levels
Flow cytometry
Shaik-Dasthagirisheb et al.
(2016)
Foam cell formation
Oil Red O Staining
Both P. gingivalis and C. pneumonia can induce foam cell formation in macrophages.
[43]
Lipid peroxidation
TBARS assay for level of oxidized LDL
P. gingivalis enhances LDL oxidation while no statistical difference was reported between the species.
Inflammatory cytokines secretion
ELISA
Both P. gingivalis and C. pneumonia enhance TNF-α and IL-6 secretion from LDL-treated macrophages.
Gene expression
PCR
Despite the differences between P. gingivalis and C. pneumonia, they indicate a similar pattern in activation and down-regulation of genes in macrophages.
Li et al.
(2013)
Foam cell formation
Oil Red O staining
P. gingivalis LPS can promote foam cell formation in ox-LDL-treated macrophages. P. gingivalis LPS could enhance CD36 mRNA expression which acts as a mediator receptor for lipid uptake and decrease the cholesterol efflux by down-regulation of ABCA1.
[47]
Cholesterol efflux
Cholesterol efflux assay
Expression of ABCA1, CD36
RT-PCR
HO-shRNA level
Western blot
Shaik-Dasthagirisaheb et al.
(2013)
Foam cell formation
Oil Red O staining
The sole addition of P. gingivalis to macrophages could enhance foam cell formation; however, the sole addition of LDL did not demonstrate the same effect. Moreover, heat-killed P. gingivalis had a similar effect on foam cell formation compared to alive P. gingivalis, regardless of the presence or the absence of LDL.
[42]
MyD88 and lps2 gene’s role in foam cell formation
Oil Red O staining
In both concurrent and uncoupled methods, MyD88 gene knockout demonstrated substantial reductions in a number of foam cells compared to the naïve types. However, in the presence of LDL lps2-knockout mice formed foam cells similar to naïve types.
Effect of P. gingivalis dose on Foam cell formation
Oil Red O staining
Enhanced concentrations of P. gingivalis (MOI of 1, 10, and 100), regardless of the concurrent or uncoupled LDL treatment, elicited a greater percentage of foam cells
Effect of LDL on the production of inflammatory cytokines
ELISA
The elevated levels of LDL significantly decrease the pro-inflammatory cytokine production by macrophages cultured with P. gingivalis.
Giacona et al.
(2004)
Foam cell formation
Oil Red O staining
The results indicate the higher effect of naïve P.g compared to fimbria-deficient P. gingivalis to induce foam cell formation.
[37]
Recovery of viable P. gingivalis from antibiotic-treated macrophages
Antibiotic protection assay
Recovery of naïve P. gingivalis species was significantly higher than the fimbria-deficient ones.
Uptake of P. gingivalis by macrophages
Transmission electron microscopy
The naïve P. gingivalis types are more capable in adhering and entering the macrophage cells than the fimbria-deficient ones.
Miyakawa et al.
(2004)
Foam cell formation by aggregated LDL
Oil Red O staining
P. gingivalis and its OMVs induce dose-dependent LDL aggregation and eventually foam cell formation, which is in part performed by the proteolysis of apo B-100 protein that is involved in the transportation of LDL.
[36]
LDL aggregation
Transmission electron microscope
SDS–PAGE and western blotting
LDL modification
Relative electrophoresis mobility
(REM) shift assays
Kuramitso et al.
(2003)
Foam cell formation
Oil Red O staining
P. gingivalis promotes foam cell formation which the most important element in this regard seems to be the P. gingivalis LPS. Moreover, P. gingivalis can induce MCP-1 secretion in endothelial cells.
[45]
MCP-1
ELISA
Qi et al.
(2003)
Effect of P. g foam cell formation
Oil Red O staining
P. gingivalis LPS alone cannot induce foam cell formation by itself. The presence of P. gingivalis and its’ OMVs can modify LDL and induce foam cell formation.
[35]
Effect of OMV on foam cell formation
Effect of LPS on foam cell formation
Effect of LDL-uptake on foam cell formation
Fluorescence imaging of LDL binding to macrophages
LDL modification by P. gingivalis during foam cell formation
Agarose gel electrophoresis
LDL peroxidation induced by P. gingivalis
TBARS assay
Abbreviations: ABCA1: ATP-binding cassette transporter A1, ELISA: Enzyme-linked immunosorbent assay, LDL: Low-density lipoprotein, MCP-1: Monocyte chemoattractant protein-1, MMP: matrix metalloproteinase, MOI: Multiciplity of Infection, P. gingivalis: Porphyromonas gingivalis, OMV: Outer Membrane Vesicles, oxLDL: oxidized low-density lipoprotein, SDS-PAGE: Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis, TBARS: Thiobarbituric acid-reactive substances assay
Out of 11 studies, four studies used murine bone marrow-derived macrophages [4144], two studies used J-774 murine macrophage-like cells [35, 36, 45], one study used peritoneal macrophages [46] and three of them used THP-1-derived macrophages/monocytes [37, 47, 48]. Eight out of 11 studies used LDL [3537, 4245, 48] or oxLDL [41, 4648] and only one study used high-density lipoprotein (HDL) [48].
Furthermore, four studies used P. gingivalis lipopolysaccharide (LPS) [35, 44, 45, 47] and others used P. gingivalis [3537, 41, 42, 45, 46, 48]. All the studies evaluated foam cell formation by Oil Red O staining [2838]. Three studies assessed cholesterol accumulation due to P. gingivalis [35, 41, 47], and three studies evaluated inflammatory cytokines formation [42, 43, 48].

Results of individual studies

All studies showed foam cell formation as P. gingivalis or P. gingivalis LPS were used [2838]. Eight studies evaluated foam cell formation in the presence of LDL and showed its necessity for the formation of foam cells promoted by P. gingivalis [3537, 4245, 48]. Two studies showed increased LDL oxidization induced by P. gingivalis [35, 43] and one study reported the same results for HDL [48]. Three studies showed that P. gingivalis increases cholesterol accumulation [35, 41, 47]. On the other hand, three studies that evaluated inflammatory cytokines levels showed P. gingivalis-induced promotion of inflammatory cytokines [42, 43, 48]. Table 3 illustrates the variables of each study, as well as their methods of evaluation and main outcomes. Moreover, Table 4 was added to emphasize the type of pathogenic element on which each study has focused as well as their mechanism of action.
Table 4
Review of the key molecular elements and their mechanism of action used by P. gingivalis to induce foam cell formation
Authors
(Year)
Key molecular element
Mechanism of Action
References
Yang et al.
(2020)
LIMP2
P. gingivalis induces foam cell formation via NF-κB and JNK pathways, which enhance the expression of LIMP2, caveolin-1 (CAV-1), and their interactions.
[41]
Gupta et al.
(2019)
TRPV4
TRPV4 can regulate oxLDL uptake in macrophages and this mechanosensitive channel is sensitive to the extracellular matrix stiffness induced by P. gingivalis LPS.
[44]
Kim et al.
(2018)
HDL
P. gingivalis can induce HDL oxidation, which prevents its athero-protective effects and promotes athero-inductive effects by eliciting pro-inflammatory cytokines secretion.
[48]
Liang et al.
(2016)
CD36, NF-κB, ERK1/2, and p65
The P. gingivalis infection can cause CD36 upregulation through the pathways mediated by NF-κB, ERK1/2, and p65.
[46]
Shaik-Dasthagirisaheb et al.
(2016)
Modification of genes subsequent in macrophage-infected P. gingivalis
P. gingivalis can up-regulate and down-regulate the genes involved in lipid uptake and efflux, respectively. P. gingivalis can also enhance the expression of genes associated with inflammatory biomarkers, cell adhesion, and ECM modification.
[43]
Li et al.
(2013)
P. gingivalis LPS, CD36, ABCA-1, calpain, HO-1
P. gingivalis LPS induces foam cell formation through HO-1 expression, which results in the activation of the cJun/AP-1 pathway that can promote upregulation of CD36 and downregulation of ABCA-1via upregulation of calpain activity.
[47]
Shaik-Dasthagirisaheb et al.
(2013)
P. gingivalis LPS, Myeloid differentiation factor 88 (MyD88)
P. gingivalis LPS can induce foam cell formation, regardless of the presence or the absence of LDL. Moreover, the knockout of the MyD88 gene can markedly reduce foam cell formation.
[42]
Miyakawa et al.
(2004)
OMV
P. gingivalis and its OMVs could induce LDL aggregation in a dose-dependent manner by proteolysis of apo B-100 protein and modification of LDL to induce higher mobility of the final LDLs.
[36]
Giacona et al.
(2004)
P. gingivalis fimbria
The major fimbria of P. gingivalis plays a key role in inducing foam cell formation and P. gingivalis invasion into the macrophage cells. Moreover, the major fimbria enhances the recovery of P. gingivalis in the presence of antibiotics.
[37]
Kuramitso et al.
(2003)
P. gingivalis fimbria, P. gingivalis LPS, MCP-1
The induction of MCP-1 secretion from the endothelial cells, caused by P. gingivalis, can attract more monocytes to the site and accelerate the process of foam cell formation and eventually, atherosclerosis.
[45]
Qi et al.
(2003)
P. gingivalis LPS, OMV
Induction of cholesterol binding and intake by macrophages
[35]
Abbreviations: ECM: Extra-cellular matrix, HDL: High-density lipoprotein, HO-1: heme oxygenase-1, LIMP2: lysosomal integral protein 2, P. gingivalis: Porphyromonas gingivalis, OMV: Outer Membrane Vesicles, TRPV4: Transient receptor potential channel of the vanilloid subfamily 4

Risk of bias assessment

Out of 11 studies included in this study, nine studies represented a medium risk of bias [3537, 41, 42, 44, 4648] while two studies had a high risk of bias [43, 45] (Table 2).

Discussion

Based on the results of this study, P. gingivalis plays an imperative role in macrophage foam cell formation. This process is clearly described in Fig. 2. Although the exact mechanisms through which this process takes place are not thoroughly uncovered, several pathways and molecules are suggested to have a significant role in this process [43, 44, 46, 47, 4951].
The first and foremost pathway which was suggested by several studies is the scavenger receptors [41, 43, 47]. The macrophage scavenger receptors attach to the modified lipoproteins and enhance cellular cholesterol accumulation [52]. In this regard, CD36 is shown to increase in macrophage after exposure to P. gingivalis [52]. In a study by Li et al. [47], the P. gingivalis LPS could induce LDL accumulation and inhibit cholesterol efflux during the process of foam cell formation. In more detail, during the macrophage foam cell formation process, P. gingivalis LPS promotes CD36 mRNA and its protein expression, as well as inhibiting ATP–binding cassette transporter A1 (ABCA1) [47]. The P. gingivalis LPS-induced CD36 expression and ABCA1 inhibition are mediated through the activation of c–Jun-AP/1 and increased calpain activity [47]. Moreover, c–Jun-AP/1 is found to be the key transcriptional factor in P. gingivalis LPS–induced CD36 upregulation [47]. It is worth mentioning that LPS of P. gingivalis did not seem to have any effect on scavenger receptor A (SRA), scavenger receptor BI, and ATP-binding cassette transporter G1 (ABCG1) [47]. Furthermore, in a study by Liang et al. [46], it was indicated that P. gingivalis induced foam cell formation and CD36 upregulation through NF-κB, and ERK 1/2 pathways and nuclear translocation of p65 [46]. In contrast to the previous study [47], the upregulation of CD36 was merely reported by the exposure to P. gingivalis, whereas macrophage exposure to Escherichia coli (E. coli) and LPS did not exert any significant effect on CD36 [46]. The proposed mechanism by which the CD36 induces lipid accumulation is by the activation of the nuclear hormone receptor of peroxisome proliferator-activated receptor–gamma (PPAR-γ) by oxLDL [53, 54]. The activation of PPAR-γ positively affects the CD36 expression, which will accelerate the oxLDL internalization by foam cells. The role of PPAR–γ in foam cell formation is further confirmed by Luo et al. [55]. They [55] have indicated that the activation of the PPAR signaling pathway is an important factor in promoting adipogenic differentiation genes and the resultant intracellular lipid accumulation which makes it an important factor in foam cell formation.
As mentioned earlier, impaired lipid transportation is one of the main mechanisms by which P. gingivalis could induce foam cell formation [46, 47]. In this regard, Yang et al. [41], have reported that P. gingivalis can enhance lysosomal integral membrane protein 2 (LIMP2) expression levels in macrophages through NF–κB and JNK pathways. Moreover, it was shown that LIMP2 knockdown can contribute to enhanced cholesterol efflux and decreased foam cell formation [41]. It is postulated that the interaction of LIMP2 and caveolin–1 would explain part of the underlying mechanism of foam cell formation [41]. In addition, P. gingivalis inhibits the ABCA1 and ABCG1 cooperation which mediates the cholesterol efflux; therefore, cholesterol will not be removed from the lysosomes, and consequently results in aggravated intracellular cholesterol [41].
Moreover, in a study by Gupta et al. [44], it was shown that P. gingivalis and matrix stiffness, which is induced by P. gingivalis LPS, can enhance the expression of a Ca2+ influx channel called Transient receptor potential channel of the vanilloid subfamily 4 (TRPV4). The knockdown of this mechanosensitive receptor is shown to have inhibitory effects on P. gingivalis LPS–induced foam cell formation [44]. The authors [44] reported that TRPV4 mediates oxidized – LDL internalization but not its cell surface binding in macrophages. It would seem that P. gingivalis LPS is able to enhance Ca2+ influx in macrophages by upregulating the expression of TRPV4. On the other hand, TRPV4 in endothelial cells is shown to have athero-protective effects by inhibiting monocytes adhesion to endothelial cells and activation of endothelial NO synthase (eNOS) [49]. In contrast, TRPV4 channels insufficiency would lead to reduced foam cell formation, endothelial impairment, and vascular disease [51, 5658].
Another mechanism by which P. gingivalis prompts foam cell formations is through fimbria [37]. The fimbria–deficient P. gingivalis is shown to be unable to adhere to and invade cells [5961] and induce alveolar bone loss in the oral cavity [62]. Concerning their effects on macrophages, fimbria–deficient P. gingivalis are unable to promote foam cell formation and macrophage invasion [37]. The fimbria of P. gingivalis is proven to enhance the proinflammatory cytokines in macrophages [37]. In this regard, CD18 is shown to have an important role in signal transduction [63, 64]. The P. gingivalis minor fimbria are proven to enhance proinflammatory cytokines, including interleukin-6 (IL–6) through CD14 and toll–like receptor 2 (TLR–2) [65, 66]. The P. gingivalis major fimbria are also shown to have a significant role in foam cell formation [37]. The P. gingivalis fimbria’s interaction with β2–integrin of macrophage is reported to be important in the P. gingivalis internalization [64]. The fimbria–deficient P. gingivalis have exerted reduced catalytic activity compared to the wild–type P. gingivalis due to gingipain activity [67].
In addition, TLRs are suggested to have an imperative role in inflammatory response against P. gingivalis [50]. These molecules support innate immune recognition of pathogen–associated molecular patterns, such as lipopolysaccharides (TLR4), lipoproteins (TLR2), etc. [68]. In order to ignite the intracellular cascade by TLRs, myeloid differentiation factor 88 (MyD88), TRIF (TLR–domain–containing adaptor–inducing interferon–β), and so forth should be recruited [69]. Based on the results of Shaik–Dasthagirisaheb et al. [50], MyD88 and lps2 (the gene of TRIF) play a significant role in foam cell induction by P. gingivalis. Moreover, it was shown that heat–killed P. gingivalis and alive P. gingivalis exert the same ability in inducing foam cell formation [50]. This finding suggests the possible role of LPS of P. gingivalis in foam cell formation. Moreover, it was shown that the presence of P. gingivalis and P. gingivalis + LDL can significantly enhance the TNF–α and IL– 6 productions by macrophages [50]. However, the combination of LDL and P. gingivalis seemed to reduce the cytokine release compared to P. gingivalis alone [50].
Another important factor in inducing foam cell formation by P. gingivalis is the multiplicity of infection (MOI) of bacteria. Shaik–Dasthagirisaheb et al. [50], have shown that the higher the MOI of P. gingivalis, the higher rate of foam cell formation. In addition, in another study by Shaik–Dasthagirisaheb et al., it was reported that P. gingivalis and Chlamydia pneumoniae (C. pneumonia) can induce foam cell formation in bone marrow–derived macrophages (BMDMs) in MOI of 100 and 10, respectively [43]. The exposure of these pathogens to LDL–treated BMDM elevated the tumor necrosis factor-α (TNF–α), IL–6, and IL–1β (this factor was exclusively enhanced by C. pneumonia) [43]. Similarly, as mentioned in the previous study, LDL downregulates cytokines secretion [50]. The analysis of the cytokine release profile indicated that the cytokine response is not identical for all the pathogens that can cause foam cell formation [43].
Another mechanism concerning the possible effect of P. gingivalis on foam cell formation and atherosclerosis is through the suppression of heme oxygenase–1 (HO–1) [47]. This enzyme plays an imperative role in the prevention of vascular inflammation through anti–oxidant, anti–inflammatory, anti–proliferative, anti–apoptotic, and immunomodulatory effects which have shown athero–protective effects [70]. According to Li et al. [47], the HO–1 knockdown results in CD36 and ABCA1 downregulation, and activation of c-Jun-AP/1. In other words, inhibition of HO-1 exacerbates the effect of P. gingivalis LPS and aggravates the intracellular lipid content [47].
Aside from the mechanisms mentioned above, another pathway that the authors of this review believe to play a role in this regard is the metabolic changes in macrophages due to P. gingivalis infection [71, 72]. P. gingivalis can increase the activity of the lactic acid cycle while decreasing oxidative phosphorylation [71, 72]. This process can enhance cellular lactic acid storage, decline mitochondrial oxygen usage, and increase the load of ROS [71, 72]. The enhancement of cellular ROS in macrophages results in a higher rate of lipid oxidation and oxLDL production [71, 72].
This systematic review highlights the importance of periodontal pathogens, especially P. gingivalis, in the progression of atherosclerosis which can have significant clinical implications in the long term. Concerning the level of risk of bias, the studies have shown moderate to high levels. Therefore, for future studies in this field, we suggest further well-designed in-vitro studies with low risk of bias and equal in-vitro settings. This would aid future systematic reviews to be able to estimate a predictable correlation between the infection of P. gingivalis and the rate of foam cell formation. Moreover, grey literature was not assessed in the current systematic review. Thus, we recommend adding the grey literature in the search strategy of future systematic reviews to provide comprehensive data for screening.
Moreover, according to the compiled outcomes of all the included studies it might be possible to suggest that periodontal treatment procedures could avoid the process of foam cell formation in the arterial intima layer by minimizing the population of P. gingivalis in the subgingival plaque area [9, 73]. In this regard, the adjunct application of inflammation-modulatory agents, including nutraceutical agents could also be a treatment option to lessen the severity of the periodontal disease as well [7477]. In this regard, we strongly recommend further in-vitro, in-vivo, and clinical experiments to assess the reliability of our hypothesis. It is also important to note that the connection of periodontal diseases with atherosclerosis cannot be solely explained based on the effect of P. gingivalis on foam cell formation. When interpreting these results, one should be aware that the connection between periodontitis and atherosclerosis is reported to be mediated through various mechanisms, including bacterial species, miRNAs, and so on. Concerning the types of periodontal microbiota P. gingivalis is indicated to be the most significant one [73, 78]. On the other hand, the release of certain types of miRNAs into the gingival crevicular fluid can lead to higher susceptibility to cardiovascular diseases by altering gene expression in cardiovascular tissues [79]. Although foam cell formation is central to atherosclerosis, it doesn’t involve the whole mechanism of pathogenesis and development of atherosclerosis [80]. Therefore, we can conclude that P. gingivalis could contribute to the process of foam cell formation and this periodontal pathogen may enhance the likelihood of developing atherosclerosis.

Conclusion

Our study has shed light on the mechanisms by which P. gingivalis can promote the process of foam cell formation. Based on the gathered evidence, P. gingivalis affects the macrophages’ environment, their gene expression patterns, and cellular mechanisms through which macrophages enhance their lipid uptake and transform into foam cells. The changes in the environment include the effect of P. gingivalis on endothelial cells to gather more monocytes to the site and changes in the mechanical and biological properties of the ECM. Moreover, the changes in the gene expression patterns in macrophages can outweigh the equilibrium of lipid transportation into more lipid influx and less lipid efflux. Besides, P. gingivalis leads the process of foam cell formation through various cellular mechanisms, including pro-inflammatory cytokines secretion, modification of LDL and HDL, ignition of various cellular signaling pathways, and cell receptor activities. All these processes are ascribed to four marked characteristics in P. gingivalis, including MOI of P. gingivalis, P. gingivalis LPS, P. gingivalis major fimbria, and P. gingivalis OMV which have demonstrated significant impacts. Since the risk of bias of the included studies in this systematic review are moderate to high, future well-organized studies are required to further confirm the current results.

Acknowledgements

The authors wish to thank Dr. Delara Amiri (DDS) for her assistance and useful advice.

Declarations

Competing interests

NF is the Editorial Board Member of BMC Oral Health. All other authors declares that they have no competing interest.
Not applicable.
Not applicable.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Kinane DF, Stathopoulou PG, Papapanou PN. Periodontal diseases. Nat Rev Dis Prim. 2017;3:17038.PubMedCrossRef Kinane DF, Stathopoulou PG, Papapanou PN. Periodontal diseases. Nat Rev Dis Prim. 2017;3:17038.PubMedCrossRef
3.
Zurück zum Zitat Srimaneepong V, Heboyan A, Zafar MS, Khurshid Z, Marya A, Fernandes GVO, et al. Fixed prosthetic restorations and periodontal health: a narrative review. J Funct Biomater. 2022;13:15.PubMedPubMedCentralCrossRef Srimaneepong V, Heboyan A, Zafar MS, Khurshid Z, Marya A, Fernandes GVO, et al. Fixed prosthetic restorations and periodontal health: a narrative review. J Funct Biomater. 2022;13:15.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Yadalam PK, Sivasankari T, Rengaraj S, Mugri MH, Sayed M, Khan SS, et al. Gene interaction network analysis reveals IFI44L as a drug target in rheumatoid arthritis and periodontitis. Molecules. 2022;27:2749.PubMedPubMedCentralCrossRef Yadalam PK, Sivasankari T, Rengaraj S, Mugri MH, Sayed M, Khan SS, et al. Gene interaction network analysis reveals IFI44L as a drug target in rheumatoid arthritis and periodontitis. Molecules. 2022;27:2749.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Barzegar PEF, Ranjbar R, Yazdanian M, Tahmasebi E, Alam M, Abbasi K et al. The current natural/chemical materials and innovative technologies in periodontal diseases therapy and regeneration: a narrative review. Mater Today Commun. 2022;:104099. Barzegar PEF, Ranjbar R, Yazdanian M, Tahmasebi E, Alam M, Abbasi K et al. The current natural/chemical materials and innovative technologies in periodontal diseases therapy and regeneration: a narrative review. Mater Today Commun. 2022;:104099.
6.
Zurück zum Zitat Nazir MA. Prevalence of periodontal disease, its association with systemic diseases and prevention. Int J Health Sci (Qassim). 2017;11:72–80.PubMed Nazir MA. Prevalence of periodontal disease, its association with systemic diseases and prevention. Int J Health Sci (Qassim). 2017;11:72–80.PubMed
7.
8.
9.
Zurück zum Zitat Tahmasebi E, Keshvad A, Alam M, Abbasi K, Rahimi S, Nouri F, et al. Current infections of the Orofacial Region: treatment, diagnosis, and Epidemiology. Life. 2023;13:269.PubMedPubMedCentralCrossRef Tahmasebi E, Keshvad A, Alam M, Abbasi K, Rahimi S, Nouri F, et al. Current infections of the Orofacial Region: treatment, diagnosis, and Epidemiology. Life. 2023;13:269.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Mohanty R, Asopa SJ, Joseph MD, Singh B, Rajguru JP, Saidath K, et al. Red complex: polymicrobial conglomerate in oral flora: a review. J Fam Med Prim care. 2019;8:3480–6.CrossRef Mohanty R, Asopa SJ, Joseph MD, Singh B, Rajguru JP, Saidath K, et al. Red complex: polymicrobial conglomerate in oral flora: a review. J Fam Med Prim care. 2019;8:3480–6.CrossRef
11.
Zurück zum Zitat Mosaddad SA, Hussain A, Tebyaniyan H. Green Alternatives as Antimicrobial Agents in Mitigating Periodontal Diseases: a narrative review. Microorganisms. 2023;11:1269.PubMedPubMedCentralCrossRef Mosaddad SA, Hussain A, Tebyaniyan H. Green Alternatives as Antimicrobial Agents in Mitigating Periodontal Diseases: a narrative review. Microorganisms. 2023;11:1269.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Zhang B, Elmabsout AA, Khalaf H, Basic VT, Jayaprakash K, Kruse R, et al. The periodontal pathogen Porphyromonas gingivalis changes the gene expression in vascular smooth muscle cells involving the TGFbeta/Notch signalling pathway and increased cell proliferation. BMC Genomics. 2013;14:1–12.CrossRef Zhang B, Elmabsout AA, Khalaf H, Basic VT, Jayaprakash K, Kruse R, et al. The periodontal pathogen Porphyromonas gingivalis changes the gene expression in vascular smooth muscle cells involving the TGFbeta/Notch signalling pathway and increased cell proliferation. BMC Genomics. 2013;14:1–12.CrossRef
13.
Zurück zum Zitat Koo TH, Jun HO, Bae S-K, Kim S-R, Moon C-P, Jeong S-K, et al. Porphyromonas gingivalis, periodontal pathogen, lipopolysaccharide induces angiogenesis via extracellular signal-regulated kinase 1/2 activation in human vascular endothelial cells. Arch Pharm Res. 2007;30:34–42.PubMedCrossRef Koo TH, Jun HO, Bae S-K, Kim S-R, Moon C-P, Jeong S-K, et al. Porphyromonas gingivalis, periodontal pathogen, lipopolysaccharide induces angiogenesis via extracellular signal-regulated kinase 1/2 activation in human vascular endothelial cells. Arch Pharm Res. 2007;30:34–42.PubMedCrossRef
14.
Zurück zum Zitat Matarese G, Isola G, Anastasi GP, Favaloro A, Milardi D, Vermiglio G, et al. Immunohistochemical analysis of TGF-β1 and VEGF in gingival and periodontal tissues: a role of these biomarkers in the pathogenesis of scleroderma and periodontal disease. Int J Mol Med. 2012;30:502–8.PubMedCrossRef Matarese G, Isola G, Anastasi GP, Favaloro A, Milardi D, Vermiglio G, et al. Immunohistochemical analysis of TGF-β1 and VEGF in gingival and periodontal tissues: a role of these biomarkers in the pathogenesis of scleroderma and periodontal disease. Int J Mol Med. 2012;30:502–8.PubMedCrossRef
15.
Zurück zum Zitat Xu W, Zhou W, Wang H, Liang S. Roles of Porphyromonas gingivalis and its virulence factors in periodontitis. Adv Protein Chem Struct Biol. 2020;120:45–84.PubMedPubMedCentralCrossRef Xu W, Zhou W, Wang H, Liang S. Roles of Porphyromonas gingivalis and its virulence factors in periodontitis. Adv Protein Chem Struct Biol. 2020;120:45–84.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Falcao A, Bullón P. A review of the influence of periodontal treatment in systemic diseases. Periodontol 2000. 2019;79:117–28.PubMedCrossRef Falcao A, Bullón P. A review of the influence of periodontal treatment in systemic diseases. Periodontol 2000. 2019;79:117–28.PubMedCrossRef
17.
Zurück zum Zitat Wang Q, Zhou X, Huang D. Role for Porphyromonas gingivalis in the progression of atherosclerosis. Med Hypotheses. 2009;72:71–3.PubMedCrossRef Wang Q, Zhou X, Huang D. Role for Porphyromonas gingivalis in the progression of atherosclerosis. Med Hypotheses. 2009;72:71–3.PubMedCrossRef
21.
Zurück zum Zitat Palasubramaniam J, Wang X, Peter K. Myocardial infarction-from atherosclerosis to thrombosis. Arterioscler Thromb Vasc Biol. 2019;39:e176–85.PubMedCrossRef Palasubramaniam J, Wang X, Peter K. Myocardial infarction-from atherosclerosis to thrombosis. Arterioscler Thromb Vasc Biol. 2019;39:e176–85.PubMedCrossRef
22.
Zurück zum Zitat Hoogeveen RC, Morrison A, Boerwinkle E, Miles JS, Rhodes CE, Sharrett AR, et al. Plasma MCP-1 level and risk for peripheral arterial disease and incident coronary heart disease: atherosclerosis risk in Communities study. Atherosclerosis. 2005;183:301–7.PubMedCrossRef Hoogeveen RC, Morrison A, Boerwinkle E, Miles JS, Rhodes CE, Sharrett AR, et al. Plasma MCP-1 level and risk for peripheral arterial disease and incident coronary heart disease: atherosclerosis risk in Communities study. Atherosclerosis. 2005;183:301–7.PubMedCrossRef
23.
Zurück zum Zitat Yu X-H, Fu Y-C, Zhang D-W, Yin K, Tang C-K. Foam cells in atherosclerosis. Clin Chim Acta. 2013;424:245–52.PubMedCrossRef Yu X-H, Fu Y-C, Zhang D-W, Yin K, Tang C-K. Foam cells in atherosclerosis. Clin Chim Acta. 2013;424:245–52.PubMedCrossRef
24.
Zurück zum Zitat Flynn MC, Pernes G, Lee MKS, Nagareddy PR, Murphy AJ. Monocytes, Macrophages, and metabolic disease in atherosclerosis. Front Pharmacol. 2019;10:666.PubMedPubMedCentralCrossRef Flynn MC, Pernes G, Lee MKS, Nagareddy PR, Murphy AJ. Monocytes, Macrophages, and metabolic disease in atherosclerosis. Front Pharmacol. 2019;10:666.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Gerszten RE, Tager AM. The monocyte in atherosclerosis–should I stay or should I go now? N Engl J Med. 2012;366:1734–6.PubMedCrossRef Gerszten RE, Tager AM. The monocyte in atherosclerosis–should I stay or should I go now? N Engl J Med. 2012;366:1734–6.PubMedCrossRef
26.
Zurück zum Zitat Wintergerst ES, Jelk J, Rahner C, Asmis R. Apoptosis induced by oxidized low density lipoprotein in human monocyte-derived macrophages involves CD36 and activation of caspase-3. Eur J Biochem. 2000;267:6050–9.PubMedCrossRef Wintergerst ES, Jelk J, Rahner C, Asmis R. Apoptosis induced by oxidized low density lipoprotein in human monocyte-derived macrophages involves CD36 and activation of caspase-3. Eur J Biochem. 2000;267:6050–9.PubMedCrossRef
28.
Zurück zum Zitat Javadifar A, Rastgoo S, Banach M, Jamialahmadi T, Johnston TP, Sahebkar A. Foam cells as therapeutic targets in atherosclerosis with a focus on the Regulatory Roles of non-coding RNAs. Int J Mol Sci. 2021;22. Javadifar A, Rastgoo S, Banach M, Jamialahmadi T, Johnston TP, Sahebkar A. Foam cells as therapeutic targets in atherosclerosis with a focus on the Regulatory Roles of non-coding RNAs. Int J Mol Sci. 2021;22.
30.
Zurück zum Zitat Hussain M, Stover CM, Dupont A. P. gingivalis in Periodontal Disease and atherosclerosis - scenes of action for antimicrobial peptides and complement. Front Immunol. 2015;6:45.PubMedPubMedCentralCrossRef Hussain M, Stover CM, Dupont A. P. gingivalis in Periodontal Disease and atherosclerosis - scenes of action for antimicrobial peptides and complement. Front Immunol. 2015;6:45.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Li C, Yu R, Ding Y. Association between Porphyromonas Gingivalis and systemic diseases: focus on T cells-mediated adaptive immunity. Front Cell Infect Microbiol. 2022;12:1026457.PubMedPubMedCentralCrossRef Li C, Yu R, Ding Y. Association between Porphyromonas Gingivalis and systemic diseases: focus on T cells-mediated adaptive immunity. Front Cell Infect Microbiol. 2022;12:1026457.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Lönn J, Ljunggren S, Klarström-Engström K, Demirel I, Bengtsson T, Karlsson H. Lipoprotein modifications by gingipains of Porphyromonas gingivalis. J Periodontal Res. 2018;53:403–13.PubMedPubMedCentralCrossRef Lönn J, Ljunggren S, Klarström-Engström K, Demirel I, Bengtsson T, Karlsson H. Lipoprotein modifications by gingipains of Porphyromonas gingivalis. J Periodontal Res. 2018;53:403–13.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Kurita-Ochiai T, Yamamoto M. Periodontal pathogens and atherosclerosis: implications of inflammation and oxidative modification of LDL. Biomed Res Int. 2014;2014:595981.PubMedPubMedCentralCrossRef Kurita-Ochiai T, Yamamoto M. Periodontal pathogens and atherosclerosis: implications of inflammation and oxidative modification of LDL. Biomed Res Int. 2014;2014:595981.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Mougeot J-LC, Stevens CB, Paster BJ, Brennan MT, Lockhart PB, Mougeot FKB. Porphyromonas gingivalis is the most abundant species detected in coronary and femoral arteries. J Oral Microbiol. 2017;9:1281562.PubMedPubMedCentralCrossRef Mougeot J-LC, Stevens CB, Paster BJ, Brennan MT, Lockhart PB, Mougeot FKB. Porphyromonas gingivalis is the most abundant species detected in coronary and femoral arteries. J Oral Microbiol. 2017;9:1281562.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Qi M, Miyakawa H, Kuramitsu HK. Porphyromonas gingivalis induces murine macrophage foam cell formation. Microb Pathog. 2003;35:259–67.PubMedCrossRef Qi M, Miyakawa H, Kuramitsu HK. Porphyromonas gingivalis induces murine macrophage foam cell formation. Microb Pathog. 2003;35:259–67.PubMedCrossRef
36.
Zurück zum Zitat Miyakawa H, Honma K, Qi M, Kuramitsu HK. Interaction of Porphyromonas gingivalis with low-density lipoproteins: implications for a role for periodontitis in atherosclerosis. J Periodontal Res. 2004;39:1–9.PubMedCrossRef Miyakawa H, Honma K, Qi M, Kuramitsu HK. Interaction of Porphyromonas gingivalis with low-density lipoproteins: implications for a role for periodontitis in atherosclerosis. J Periodontal Res. 2004;39:1–9.PubMedCrossRef
37.
Zurück zum Zitat Giacona MB, Papapanou PN, Lamster IB, Rong LL, D’Agati VD, Schmidt AM, et al. Porphyromonas gingivalis induces its uptake by human macrophages and promotes foam cell formation in vitro. FEMS Microbiol Lett. 2004;241:95–101.PubMedCrossRef Giacona MB, Papapanou PN, Lamster IB, Rong LL, D’Agati VD, Schmidt AM, et al. Porphyromonas gingivalis induces its uptake by human macrophages and promotes foam cell formation in vitro. FEMS Microbiol Lett. 2004;241:95–101.PubMedCrossRef
38.
Zurück zum Zitat Page MJ, Moher D, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD et al. PRISMA 2020 explanation and elaboration: updated guidance and exemplars for reporting systematic reviews. BMJ. 2021;372. Page MJ, Moher D, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD et al. PRISMA 2020 explanation and elaboration: updated guidance and exemplars for reporting systematic reviews. BMJ. 2021;372.
39.
Zurück zum Zitat Farshidfar N, Amiri MA, Firoozi P, Hamedani S, Ajami S, Tayebi L. The adjunctive effect of autologous platelet concentrates on orthodontic tooth movement: a systematic review and meta-analysis of current randomized controlled trials. Int Orthod. 2022;20:100596.PubMedCrossRef Farshidfar N, Amiri MA, Firoozi P, Hamedani S, Ajami S, Tayebi L. The adjunctive effect of autologous platelet concentrates on orthodontic tooth movement: a systematic review and meta-analysis of current randomized controlled trials. Int Orthod. 2022;20:100596.PubMedCrossRef
40.
Zurück zum Zitat Sheth VH, Shah NP, Jain R, Bhanushali N, Bhatnagar V. Development and validation of a risk-of-bias tool for assessing in vitro studies conducted in dentistry: the QUIN. J Prosthet Dent. 2022. Sheth VH, Shah NP, Jain R, Bhanushali N, Bhatnagar V. Development and validation of a risk-of-bias tool for assessing in vitro studies conducted in dentistry: the QUIN. J Prosthet Dent. 2022.
41.
Zurück zum Zitat Yang Y, He X, Xia S, Liu F, Luo L. Porphyromonas gingivalis facilitated the foam cell formation via lysosomal integral membrane protein 2 (LIMP2). J Periodontal Res. 2021;56:265–74.PubMedCrossRef Yang Y, He X, Xia S, Liu F, Luo L. Porphyromonas gingivalis facilitated the foam cell formation via lysosomal integral membrane protein 2 (LIMP2). J Periodontal Res. 2021;56:265–74.PubMedCrossRef
42.
Zurück zum Zitat Shaik-Dasthagirisaheb YB, Huang N, Baer MT, Gibson FC III. Role of M y D 88‐dependent and M y D 88‐independent signaling in P orphyromonas gingivalis‐elicited macrophage foam cell formation. Mol Oral Microbiol. 2013;28:28–39.PubMedCrossRef Shaik-Dasthagirisaheb YB, Huang N, Baer MT, Gibson FC III. Role of M y D 88‐dependent and M y D 88‐independent signaling in P orphyromonas gingivalis‐elicited macrophage foam cell formation. Mol Oral Microbiol. 2013;28:28–39.PubMedCrossRef
43.
Zurück zum Zitat Shaik-Dasthagirisaheb YB, Mekasha S, He X, Gibson FC 3rd, Ingalls RR. Signaling events in pathogen-induced macrophage foam cell formation. Pathog Dis. 2016;74. Shaik-Dasthagirisaheb YB, Mekasha S, He X, Gibson FC 3rd, Ingalls RR. Signaling events in pathogen-induced macrophage foam cell formation. Pathog Dis. 2016;74.
44.
Zurück zum Zitat Gupta N, Goswami R, Alharbi MO, Biswas D, Rahaman SO. TRPV4 is a regulator in P. gingivalis lipopolysaccharide-induced exacerbation of macrophage foam cell formation. Physiol Rep. 2019;7:e14069.PubMedPubMedCentralCrossRef Gupta N, Goswami R, Alharbi MO, Biswas D, Rahaman SO. TRPV4 is a regulator in P. gingivalis lipopolysaccharide-induced exacerbation of macrophage foam cell formation. Physiol Rep. 2019;7:e14069.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Kuramitsu HK, Kang I, Qi M. Interactions of Porphyromonas gingivalis with host cells: implications for cardiovascular diseases. J Periodontol. 2003;74:85–9.PubMedCrossRef Kuramitsu HK, Kang I, Qi M. Interactions of Porphyromonas gingivalis with host cells: implications for cardiovascular diseases. J Periodontol. 2003;74:85–9.PubMedCrossRef
46.
Zurück zum Zitat Liang D-Y, Liu F, Chen J-X, He X-L, Zhou Y-L, Ge B-X, et al. Porphyromonas gingivalis infected macrophages upregulate CD36 expression via ERK/NF-κB pathway. Cell Signal. 2016;28:1292–303.PubMedCrossRef Liang D-Y, Liu F, Chen J-X, He X-L, Zhou Y-L, Ge B-X, et al. Porphyromonas gingivalis infected macrophages upregulate CD36 expression via ERK/NF-κB pathway. Cell Signal. 2016;28:1292–303.PubMedCrossRef
47.
Zurück zum Zitat Li X-Y, Wang C, Xiang X-R, Chen F-C, Yang C-M, Wu J. Porphyromonas gingivalis lipopolysaccharide increases lipid accumulation by affecting CD36 and ATP-binding cassette transporter A1 in macrophages. Oncol Rep. 2013;30:1329–36.PubMedCrossRef Li X-Y, Wang C, Xiang X-R, Chen F-C, Yang C-M, Wu J. Porphyromonas gingivalis lipopolysaccharide increases lipid accumulation by affecting CD36 and ATP-binding cassette transporter A1 in macrophages. Oncol Rep. 2013;30:1329–36.PubMedCrossRef
48.
Zurück zum Zitat Kim H-J, Cha GS, Kim H-J, Kwon E-Y, Lee J-Y, Choi J, et al. Porphyromonas gingivalis accelerates atherosclerosis through oxidation of high-density lipoprotein. J Periodontal Implant Sci. 2018;48:60–8.PubMedPubMedCentralCrossRef Kim H-J, Cha GS, Kim H-J, Kwon E-Y, Lee J-Y, Choi J, et al. Porphyromonas gingivalis accelerates atherosclerosis through oxidation of high-density lipoprotein. J Periodontal Implant Sci. 2018;48:60–8.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Xu S, Liu B, Yin M, Koroleva M, Mastrangelo M, Ture S, et al. A novel TRPV4-specific agonist inhibits monocyte adhesion and atherosclerosis. Oncotarget. 2016;7:37622–35.PubMedPubMedCentralCrossRef Xu S, Liu B, Yin M, Koroleva M, Mastrangelo M, Ture S, et al. A novel TRPV4-specific agonist inhibits monocyte adhesion and atherosclerosis. Oncotarget. 2016;7:37622–35.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Shaik-Dasthagirisaheb YB, Huang N, Baer MT, Gibson FC 3. rd. role of MyD88-dependent and MyD88-independent signaling in Porphyromonas gingivalis-elicited macrophage foam cell formation. Mol Oral Microbiol. 2013;28:28–39. Shaik-Dasthagirisaheb YB, Huang N, Baer MT, Gibson FC 3. rd. role of MyD88-dependent and MyD88-independent signaling in Porphyromonas gingivalis-elicited macrophage foam cell formation. Mol Oral Microbiol. 2013;28:28–39.
51.
Zurück zum Zitat Goswami R, Merth M, Sharma S, Alharbi MO, Aranda-Espinoza H, Zhu X, et al. TRPV4 calcium-permeable channel is a novel regulator of oxidized LDL-induced macrophage foam cell formation. Free Radic Biol Med. 2017;110:142–50.PubMedCrossRef Goswami R, Merth M, Sharma S, Alharbi MO, Aranda-Espinoza H, Zhu X, et al. TRPV4 calcium-permeable channel is a novel regulator of oxidized LDL-induced macrophage foam cell formation. Free Radic Biol Med. 2017;110:142–50.PubMedCrossRef
52.
Zurück zum Zitat Moore KJ, Freeman MW. Scavenger receptors in atherosclerosis: beyond lipid uptake. Arterioscler Thromb Vasc Biol. 2006;26:1702–11.PubMedCrossRef Moore KJ, Freeman MW. Scavenger receptors in atherosclerosis: beyond lipid uptake. Arterioscler Thromb Vasc Biol. 2006;26:1702–11.PubMedCrossRef
53.
Zurück zum Zitat Tontonoz P, Nagy L, Alvarez JG, Thomazy VA, Evans RM. PPARgamma promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell. 1998;93:241–52.PubMedCrossRef Tontonoz P, Nagy L, Alvarez JG, Thomazy VA, Evans RM. PPARgamma promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell. 1998;93:241–52.PubMedCrossRef
54.
Zurück zum Zitat Nagy L, Tontonoz P, Alvarez JG, Chen H, Evans RM. Oxidized LDL regulates macrophage gene expression through ligand activation of PPARgamma. Cell. 1998;93:229–40.PubMedCrossRef Nagy L, Tontonoz P, Alvarez JG, Chen H, Evans RM. Oxidized LDL regulates macrophage gene expression through ligand activation of PPARgamma. Cell. 1998;93:229–40.PubMedCrossRef
55.
Zurück zum Zitat Luo Y, Tanigawa K, Kawashima A, Ishido Y, Ishii N, Suzuki K. The function of peroxisome proliferator-activated receptors PPAR-γ and PPAR-δ in Mycobacterium leprae-induced foam cell formation in host macrophages. PLoS Negl Trop Dis. 2020;14:e0008850.PubMedPubMedCentralCrossRef Luo Y, Tanigawa K, Kawashima A, Ishido Y, Ishii N, Suzuki K. The function of peroxisome proliferator-activated receptors PPAR-γ and PPAR-δ in Mycobacterium leprae-induced foam cell formation in host macrophages. PLoS Negl Trop Dis. 2020;14:e0008850.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Zhang DX, Mendoza SA, Bubolz AH, Mizuno A, Ge Z-D, Li R et al. Transient receptor potential vanilloid type 4-deficient mice exhibit impaired endothelium-dependent relaxation induced by acetylcholine in vitro and in vivo. Hypertens (Dallas, Tex 1979). 2009;53:532–8. Zhang DX, Mendoza SA, Bubolz AH, Mizuno A, Ge Z-D, Li R et al. Transient receptor potential vanilloid type 4-deficient mice exhibit impaired endothelium-dependent relaxation induced by acetylcholine in vitro and in vivo. Hypertens (Dallas, Tex 1979). 2009;53:532–8.
57.
Zurück zum Zitat Ye L, Kleiner S, Wu J, Sah R, Gupta RK, Banks AS, et al. TRPV4 is a regulator of adipose oxidative metabolism, inflammation, and energy homeostasis. Cell. 2012;151:96–110.PubMedPubMedCentralCrossRef Ye L, Kleiner S, Wu J, Sah R, Gupta RK, Banks AS, et al. TRPV4 is a regulator of adipose oxidative metabolism, inflammation, and energy homeostasis. Cell. 2012;151:96–110.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Du J, Wang X, Li J, Guo J, Liu L, Yan D, et al. Increasing TRPV4 expression restores flow-induced dilation impaired in mesenteric arteries with aging. Sci Rep. 2016;6:22780.PubMedPubMedCentralCrossRef Du J, Wang X, Li J, Guo J, Liu L, Yan D, et al. Increasing TRPV4 expression restores flow-induced dilation impaired in mesenteric arteries with aging. Sci Rep. 2016;6:22780.PubMedPubMedCentralCrossRef
59.
60.
Zurück zum Zitat Jotwani R, Cutler CW. Fimbriated Porphyromonas gingivalis is more efficient than fimbria-deficient P. gingivalis in entering human dendritic cells in vitro and induces an inflammatory Th1 effector response. Infect Immun. 2004;72:1725–32.PubMedPubMedCentralCrossRef Jotwani R, Cutler CW. Fimbriated Porphyromonas gingivalis is more efficient than fimbria-deficient P. gingivalis in entering human dendritic cells in vitro and induces an inflammatory Th1 effector response. Infect Immun. 2004;72:1725–32.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Weinberg A, Belton CM, Park Y, Lamont RJ. Role of fimbriae in Porphyromonas gingivalis invasion of gingival epithelial cells. Infect Immun. 1997;65:313–6.PubMedPubMedCentralCrossRef Weinberg A, Belton CM, Park Y, Lamont RJ. Role of fimbriae in Porphyromonas gingivalis invasion of gingival epithelial cells. Infect Immun. 1997;65:313–6.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Malek R, Fisher JG, Caleca A, Stinson M, van Oss CJ, Lee JY, et al. Inactivation of the Porphyromonas gingivalis fimA gene blocks periodontal damage in gnotobiotic rats. J Bacteriol. 1994;176:1052–9.PubMedPubMedCentralCrossRef Malek R, Fisher JG, Caleca A, Stinson M, van Oss CJ, Lee JY, et al. Inactivation of the Porphyromonas gingivalis fimA gene blocks periodontal damage in gnotobiotic rats. J Bacteriol. 1994;176:1052–9.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Hanazawa S, Murakami Y, Hirose K, Amano S, Ohmori Y, Higuchi H, et al. Bacteroides (Porphyromonas) gingivalis fimbriae activate mouse peritoneal macrophages and induce gene expression and production of interleukin-1. Infect Immun. 1991;59:1972–7.PubMedPubMedCentralCrossRef Hanazawa S, Murakami Y, Hirose K, Amano S, Ohmori Y, Higuchi H, et al. Bacteroides (Porphyromonas) gingivalis fimbriae activate mouse peritoneal macrophages and induce gene expression and production of interleukin-1. Infect Immun. 1991;59:1972–7.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Takeshita A, Murakami Y, Yamashita Y, Ishida M, Fujisawa S, Kitano S, et al. Porphyromonas gingivalis fimbriae use beta2 integrin (CD11/CD18) on mouse peritoneal macrophages as a cellular receptor, and the CD18 beta chain plays a functional role in fimbrial signaling. Infect Immun. 1998;66:4056–60.PubMedPubMedCentralCrossRef Takeshita A, Murakami Y, Yamashita Y, Ishida M, Fujisawa S, Kitano S, et al. Porphyromonas gingivalis fimbriae use beta2 integrin (CD11/CD18) on mouse peritoneal macrophages as a cellular receptor, and the CD18 beta chain plays a functional role in fimbrial signaling. Infect Immun. 1998;66:4056–60.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Hiramine H, Watanabe K, Hamada N, Umemoto T. Porphyromonas gingivalis 67-kDa fimbriae induced cytokine production and osteoclast differentiation utilizing TLR2. FEMS Microbiol Lett. 2003;229:49–55.PubMedCrossRef Hiramine H, Watanabe K, Hamada N, Umemoto T. Porphyromonas gingivalis 67-kDa fimbriae induced cytokine production and osteoclast differentiation utilizing TLR2. FEMS Microbiol Lett. 2003;229:49–55.PubMedCrossRef
66.
Zurück zum Zitat Hajishengallis G, Martin M, Sojar HT, Sharma A, Schifferle RE, DeNardin E, et al. Dependence of bacterial protein adhesins on toll-like receptors for proinflammatory cytokine induction. Clin Diagn Lab Immunol. 2002;9:403–11.PubMedPubMedCentral Hajishengallis G, Martin M, Sojar HT, Sharma A, Schifferle RE, DeNardin E, et al. Dependence of bacterial protein adhesins on toll-like receptors for proinflammatory cytokine induction. Clin Diagn Lab Immunol. 2002;9:403–11.PubMedPubMedCentral
67.
Zurück zum Zitat Chen T, Nakayama K, Belliveau L, Duncan MJ. Porphyromonas gingivalis gingipains and adhesion to epithelial cells. Infect Immun. 2001;69:3048–56.PubMedPubMedCentralCrossRef Chen T, Nakayama K, Belliveau L, Duncan MJ. Porphyromonas gingivalis gingipains and adhesion to epithelial cells. Infect Immun. 2001;69:3048–56.PubMedPubMedCentralCrossRef
68.
71.
Zurück zum Zitat Fleetwood AJ, Lee MKS, Singleton W, Achuthan A, Lee M-C, O’Brien-Simpson NM, et al. Metabolic remodeling, inflammasome activation, and pyroptosis in macrophages stimulated by Porphyromonas gingivalis and its outer membrane vesicles. Front Cell Infect Microbiol. 2017;7:351.PubMedPubMedCentralCrossRef Fleetwood AJ, Lee MKS, Singleton W, Achuthan A, Lee M-C, O’Brien-Simpson NM, et al. Metabolic remodeling, inflammasome activation, and pyroptosis in macrophages stimulated by Porphyromonas gingivalis and its outer membrane vesicles. Front Cell Infect Microbiol. 2017;7:351.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Zhang J, Xie M, Huang X, Chen G, Yin Y, Lu X, et al. The effects of porphyromonas gingivalis on atherosclerosis-related cells. Front Immunol. 2021;12:766560.PubMedPubMedCentralCrossRef Zhang J, Xie M, Huang X, Chen G, Yin Y, Lu X, et al. The effects of porphyromonas gingivalis on atherosclerosis-related cells. Front Immunol. 2021;12:766560.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Zardawi F, Gul S, Abdulkareem A, Sha A, Yates J. Association between periodontal disease and atherosclerotic cardiovascular diseases: revisited. Front Cardiovasc Med. 2021;7:625579.PubMedPubMedCentralCrossRef Zardawi F, Gul S, Abdulkareem A, Sha A, Yates J. Association between periodontal disease and atherosclerotic cardiovascular diseases: revisited. Front Cardiovasc Med. 2021;7:625579.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Isola G, Polizzi A, Iorio-Siciliano V, Alibrandi A, Ramaglia L, Leonardi R. Effectiveness of a nutraceutical agent in the non-surgical periodontal therapy: a randomized, controlled clinical trial. Clin Oral Investig. 2021;25:1035–45.PubMedCrossRef Isola G, Polizzi A, Iorio-Siciliano V, Alibrandi A, Ramaglia L, Leonardi R. Effectiveness of a nutraceutical agent in the non-surgical periodontal therapy: a randomized, controlled clinical trial. Clin Oral Investig. 2021;25:1035–45.PubMedCrossRef
75.
Zurück zum Zitat Moghaddam A, Ranjbar R, Yazdanian M, Tahmasebi E, Alam M, Abbasi K et al. The current antimicrobial and antibiofilm activities of synthetic/herbal/biomaterials in dental application. Biomed Res Int. 2022;2022. Moghaddam A, Ranjbar R, Yazdanian M, Tahmasebi E, Alam M, Abbasi K et al. The current antimicrobial and antibiofilm activities of synthetic/herbal/biomaterials in dental application. Biomed Res Int. 2022;2022.
76.
Zurück zum Zitat Motallaei MN, Yazdanian M, Tebyanian H, Tahmasebi E, Alam M, Abbasi K et al. The current strategies in controlling oral diseases by herbal and chemical materials. Evidence-Based Complement Altern Med. 2021;2021. Motallaei MN, Yazdanian M, Tebyanian H, Tahmasebi E, Alam M, Abbasi K et al. The current strategies in controlling oral diseases by herbal and chemical materials. Evidence-Based Complement Altern Med. 2021;2021.
77.
Zurück zum Zitat Yazdanian M, Rostamzadeh P, Alam M, Abbasi K, Tahmasebi E, Tebyaniyan H, et al. Evaluation of antimicrobial and cytotoxic effects of Echinacea and Arctium extracts and Zataria essential oil. AMB Express. 2022;12:1–13.CrossRef Yazdanian M, Rostamzadeh P, Alam M, Abbasi K, Tahmasebi E, Tebyaniyan H, et al. Evaluation of antimicrobial and cytotoxic effects of Echinacea and Arctium extracts and Zataria essential oil. AMB Express. 2022;12:1–13.CrossRef
78.
Zurück zum Zitat Karobari MI, Siddharthan S, Adil AH, Khan MM, Venugopal A, Rokaya D et al. Modifiable and non-modifiable risk factors affecting oral and periodontal health and quality of life in south asia. Open Dent J. 2022;16. Karobari MI, Siddharthan S, Adil AH, Khan MM, Venugopal A, Rokaya D et al. Modifiable and non-modifiable risk factors affecting oral and periodontal health and quality of life in south asia. Open Dent J. 2022;16.
79.
Zurück zum Zitat Isola G, Santonocito S, Distefano A, Polizzi A, Vaccaro M, Raciti G, et al. Impact of periodontitis on gingival crevicular fluid miRNAs profiles associated with cardiovascular disease risk. J Periodontal Res. 2023;58:165–74.PubMedCrossRef Isola G, Santonocito S, Distefano A, Polizzi A, Vaccaro M, Raciti G, et al. Impact of periodontitis on gingival crevicular fluid miRNAs profiles associated with cardiovascular disease risk. J Periodontal Res. 2023;58:165–74.PubMedCrossRef
80.
Zurück zum Zitat Gui Y, Zheng H, Cao RY. Foam cells in atherosclerosis: novel insights into its origins, consequences, and molecular mechanisms. Front Cardiovasc Med. 2022;:842. Gui Y, Zheng H, Cao RY. Foam cells in atherosclerosis: novel insights into its origins, consequences, and molecular mechanisms. Front Cardiovasc Med. 2022;:842.
Metadaten
Titel
A systematic review of the impact of Porphyromonas gingivalis on foam cell formation: Implications for the role of periodontitis in atherosclerosis
verfasst von
Saeed Afzoon
Mohammad Amin Amiri
Mostafa Mohebbi
Shahram Hamedani
Nima Farshidfar
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Oral Health / Ausgabe 1/2023
Elektronische ISSN: 1472-6831
DOI
https://doi.org/10.1186/s12903-023-03183-9

Weitere Artikel der Ausgabe 1/2023

BMC Oral Health 1/2023 Zur Ausgabe

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Klinik aktuell Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

Endlich: Zi zeigt, mit welchen PVS Praxen zufrieden sind

IT für Ärzte Nachrichten

Darauf haben viele Praxen gewartet: Das Zi hat eine Liste von Praxisverwaltungssystemen veröffentlicht, die von Nutzern positiv bewertet werden. Eine gute Grundlage für wechselwillige Ärztinnen und Psychotherapeuten.

Parodontalbehandlung verbessert Prognose bei Katheterablation

19.04.2024 Vorhofflimmern Nachrichten

Werden Personen mit Vorhofflimmern in der Blanking-Periode nach einer Katheterablation gegen eine bestehende Parodontitis behandelt, verbessert dies die Erfolgsaussichten. Dafür sprechen die Resultate einer prospektiven Untersuchung.

Newsletter

Bestellen Sie unseren kostenlosen Newsletter Update Zahnmedizin und bleiben Sie gut informiert – ganz bequem per eMail.