Open Access

Interaction between apical periodontitis and systemic disease (Review)

  • Authors:
    • Lanxiang Ye
    • Li Cao
    • Wencheng Song
    • Chen Yang
    • Qingming Tang
    • Zhenglin Yuan
  • View Affiliations

  • Published online on: May 30, 2023     https://doi.org/10.3892/ijmm.2023.5263
  • Article Number: 60
  • Copyright: © Ye et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Apical periodontitis is an oral common inflammatory disease initiated by infection of pulp chamber and is characterized by destruction and resorption of the periapical bone. As a local infection, pathogens and their products in periapical tissues, as well as inflammatory cytokines produced in periapical lesions, enter the blood circulation, triggering systemic immune responses and leading to the pathogenesis of various types of systemic disease. Therefore, apical periodontitis might be associated with systemic disease rather than solely simple local oral disease. In addition, the existence of a hyperinflammatory state in certain patients with chronic inflammation‑related disorder may affect the progression or prognosis of apical periodontitis. However, the association and potential mechanisms between apical periodontitis and systemic diseases remain unclear. An in‑depth understanding of the association between apical periodontitis and systemic disease will be useful for both dentists and physicians to eliminate the possible risk factors and promote the healing of apical periodontitis and systemic disease. Thus, the aim of the present review is to introduce the potential relationship between apical periodontitis and systemic disease.

1. Introduction

Apical periodontitis (AP) is a type of chronic oral inflammatory disease characterized by destruction and absorption of alveolar bone in the periapical tissue (1,2). It is usually caused by severe caries, pulp lesions or periodontal diseases. As a type of highly prevalent oral disease, 52% of the adult population worldwide was reported to have at least one tooth with AP according to a systematic review and meta-analysis in 2021 (3). Untreated teeth with AP can lead to tooth loss, jaw osteomyelitis and systemic disease associated with mortality (4-6).

In recent years, the association between AP and systemic disease has attracted attention, leading to the concept of endodontic medicine (7). As a local infection (8), previous studies have shown that microbes and toxins in periapical lesions have access to the bloodstream from the root canal system during or following endodontic therapy of teeth (9-11). Furthermore, AP modulates the systemic immune response by modifying the levels of inflammatory cytokines, such as C-reactive protein (CRP), tumor necrosis factor (TNF)-α, IL-6 and IL-1 (12-14). The aforementioned findings suggested that periapical inflammation is important for maintaining the health of the whole body. Furthermore, the long-term persistence of chronic inflammatory disease is implicated in systemic immune dysregulation and altered inflammatory factors in the circulation (15). Hence, chronic inflammation-associated disorder affects the status of AP, which is dependent on the antagonistic balance between the pathogenic microorganism and host immune response (16). To the best of our knowledge, however, evidence of the direct relationship between AP and systemic disease is lacking.

Over the past decades, there has been growing evidence on epidemiology (Table I) and mechanistic studies suggesting a potential link between periapical disease and systemic disease (17-19). However, the available studies (18-20) analyzing the relationship between AP and systemic disease are not comprehensive. The present review summarizes the association between AP and systemic diseases, including metabolic, autoimmune and cardiovascular disease (CVD), adverse pregnancy outcome (APO), as well as psychiatric disorder (Fig. 1). Further attention and understanding of the association between AP and systemic disease will help dentists and physicians develop innovative therapeutic proposals and promote effective healing of AP and systemic disease.

Table I

Epidemiological evidence of the association between AP and systemic disease.

Table I

Epidemiological evidence of the association between AP and systemic disease.

Systemic diseaseStudy designMain findingsAssociation reported(Refs.)
DM Cross-sectionalPatients with DM have a higher susceptibility to APYes(31,35,34,68)
Patients with long-term DM are predisposed to AP compared with short-term DM.Yes(36)
Periapical status/prevalence of AP is correlated positively with HbA1c levels in patients with T2DMYes(32,33)
DM decreases the success of RCT in teeth with APYes(54,66)
T2DM does not influence the prognosis of APNo(70)
Case-controlDM does not increase the levels of bone resorption mediators in periapical lesionsNo(57)
T2DM is independently associated with significantly greater prevalence of AP. Use of metformin or statins decreases the prevalence of AP in DMYes(29)
DM has a negative effect on the prognosis of APYes(61-63)
CohortMetformin or statins promote healing of APYes(40)
DM is a risk factor for tooth extraction after NSRCTYes(58)
Patients with T2DM exhibit significantly less periapical healing compared with patients without DMYes(67)
PilotAP does not affect the glycemic control or inflammatory levels in patients with T2DMNo(83)
Osteoporosis Cross-sectionalMarginal association exists between bone mineral density and periapical radiolucencyYes(87)
Prevalence of AP is significantly higher in osteoporotic patients. The use of BPs decreases prevalence of APYes(92)
RD Cross-sectionalPatients with RD are predisposed to AP compared with control groupYes(116)
RD does not increase the prevalence of RCT compared with control groupNo(119)
RD does not alter the periapical histological features or the incidence of APNo(121,122)
Amyloid protein is almost 5 times more frequent in periapical lesions of patients with RD than in controlsYes(125)
Autoimmune hepatitis/nephritisCase-controlPatients with periapical radiolucency have higher prevalence of cirrhosis-associated complicationsYes(138)
Cross-sectionalLiver transplant candidates have significantly higher prevalence of radiographic periapical lesionsYes(137)
ESRD is significantly associated with APYes(146,147)
IBDRetrospective clinicalPatients with IBD taking immunomodulators have a higher prevalence of AP. Patients with IBD and AP have larger periapical lesions than healthy subjectsYes(161)
Case-controlPatients with IBDs have higher prevalence of APYes(162)
IBD is associated with higher prevalence of RFT and higher percentage of RFT with APYes(165)
CVD Cross-sectionalAP and endodontic burden (AP and/or RCT) are associated with CABYes(195,205)
ALEO is associated with systemic inflammatory burden and cardiovascular riskYes(196)
Endodontic lesions are independently associated with CAD and ACSYes(200)
Patients with AP have a 2.79 times higher risk of developing CADYes(201)
There is no statistically significant positive correlation between AAP and CADNo(202)
Periapical lesions and root fillings are significantly associated with a first MIYes(209)
CAP is moderately involved in plasma biomarker changes in hypertensive patientsYes(207)
AP has a potential association between endodontic infection and CVDYes(216)
CohortIn patients aged ≤40 years, ALEOs are significantly associated with the age at CHD diagnosisYes(199)
Young adults with AP are more likely to have early endothelial dysfunctionYes(215)
Adverse pregnancy outcomeCase-controlPregnant people with AP are more likely to have LBWPBYes(225,226)
Maternal AP is significantly associated with occurrence of PEYes(227)
There is no association between maternal CAP and LBWPBNo(231)

[i] HbA1c, glycated hemoglobin; T2DM, type 2 diabetes mellitus; NSRCT, non-surgical root canal treatment; ALEO, apical lesion of endodontic origin; CAD, coronary artery disease; CHD, coronary heart disease; ACS, acute coronary syndrome; AAP, asymptomatic apical periodontitis; MI, myocardial infarction; RFT, root-filled teeth; PE, preeclampsia; BP, bisphosphonate; ESRD, end-stage renal disease; LBWPB, low birth weight and preterm birth; RD, rheumatoid disease; CAP, chronic apical periodontitis.

2. Metabolic disease

Diabetes mellitus (DM)

Globally, DM is a chronic metabolic disease characterized by hyperglycemia owing to insulin resistance and/or a deficiency in secretion of insulin or both (21). As one of the most common types of metabolic disease, it is estimated that the prevalence of DM for all age groups worldwide will rise from 2.8% in 2000 to 4.4% in 2030 and may affect ≥693 million people in 2045 (22,23). Diabetes is accompanied by various complications (24). It is considered that oral complications in patients with diabetes, such as AP, could affect quality of life (25). As the prevalence of diabetes increases, it may lead to increasing consequences of the complications of DM, such as enhancing the inflammatory response in apical tissue and accelerating alveolar bone loss (26). There have been numerous studies on the possible association between DM and AP (27-30). Notably, there may be a bidirectional association between DM and AP (Fig 2). DM may increase the prevalence of AP, accelerate the progression of AP and undermine the efficacy of root canal treatment (RCT). However, AP can affect insulin signaling pathways or reduce insulin sensitivity, leading to increased insulin requirements and blood glucose levels.

DM affects the prevalence, progression and prognosis of AP

Epidemiological studies (31-35) confirm that DM is positively associated with the prevalence of AP, especially in DM patients with poor glycemic control. Segura-Egea et al (31) performed a retrospective cohort study in which 70 participants were divided into patients with type (T)2 DM and control group. Patients with DM had a higher prevalence of AP compared with controls. In a hospital-based population, Saleh et al (34) indicated that patients with DM are ≥3 times more likely to exhibit AP compared with subjects without DM. Moreover, the association between AP and DM is more pronounced in patients with poor glycemic control and long-term diabetes (33,32,36) which is consistent with other investigations (35,30,47-39). Conversely, the use of metformin and statins decreases prevalence and promote the healing of AP in diabetic patients (29,40).

In addition to affecting the prevalence of AP, DM accelerates the progression of AP. In animal studies, high sugar intake and hyperglycemic states increase susceptibility to oral infection as a result of the disruption of leukocyte function, which is also supported by a greater and unbalanced presence of pathogenic microorganisms in root canals in periapical lesions in diabetes (41,42). In a rat model of combined AP and DM, increased inflammatory infiltration, higher levels of proinflammatory cytokines (IL-17, IL-23, IL-6 and TNF-α) and larger lesion size are observed in periapical tissue (41,43). Cintra et al (44) hypothesized that this might be partly due to enhanced systemic inflammation of DM, which contributes to a greater destruction of the alveolar bone in the periapical lesions. For example, IL-17, as a destructive inflammatory cytokine, serves a critical role in the resorption of periapical bone (45,46). Moreover, TNF-α could also promote osteoclast differentiation and maturation by activating the NF-κB signaling pathway to trigger a decrease in osteoblast function and apoptosis, resulting in an imbalance in osteoblast-osteolysis coupling (47). Moreover, the imbalance of calcium homeostasis associated with insulin deficiency leads to bone loss (48,49). Metformin, a classical diabetic prescription drug, improves hyperglycemic conditions and enhances osteogenesis by regulating glucose metabolism, promoting cell migration and increasing angiogenesis (50,51). In addition, metformin could activate the proliferation and osteogenic differentiation of dental pulp and periodontal ligament stem cells, which serve an important role in the repair and mineralization of oral-associated bone defects (52,53). In animal studies, intramuscular injection of metformin in rats with AP is effective in decreasing the size of periapical lesions via suppression of the NF-κB signaling pathway and diminishing the hypoxia-induced apoptosis of osteoblasts (54,55). In addition, intracanal metformin decreases expression of inducible nitric oxide synthase and nitric oxide to inhibit monocyte migration in periapical tissue (56). The aforementioned results confirm the positive effect of DM on the progression of AP. By contrast, Sarmento et al (57) found no significant differences in bone resorption mediators between patients with DM and normoglycemic patients with AP; this may be because the sample population was too small and the DM group comprised well-controlled individuals with a mean glycated hemoglobin (HbA1c) <7%.

Previous evidence has suggested that diabetes is a key preoperative factor in evaluating the prognosis of RCT and is a common cause for tooth extraction following non-surgical RCT (58-60). The accumulation of late glycosylated products in DM prolong the treatment time of RCT and increased the rate of infection in the oral cavity (60). In a retrospective case-control study, Uğur Aydın et al (61) assessed changes of fractal dimension and concluded that DM has a negative effect on the healing progress of periapical lesions after RCT. In the light of clinical and radiographic healing outcomes, the success rate of RCT is significantly lower in patients with DM or poor glycemic control due to impairment of the angiogenic process in DM (62,63), which is in accordance with other clinical studies (32,64-67). Similar results have been observed in patients with type 1 diabetes (68). Conversely, other studies suggest that DM does not affect the healing outcome of RCT (31,69-71). Different epidemiological methodologies may contribute to the different outcomes, including tooth type, radiographic method, assessment criteria and follow-up time. The healing of RCT in DM is a continuous process and could be affected by various factors, such as time to assess prognosis, status of general health, and control of oral reinfections. The outcomes of cross-sectional studies should be evaluated with care and more prospective studies with longer follow-up time, larger sample size and stricter control of confounding factors are required.

AP may affect insulin requirements and blood glucose status in DM

Inflammation is involved in the pathogenesis of DM, as inflammatory states could decrease insulin sensitivity (72). Chronic infection of AP could cause aggravated and dysregulated inflammatory response, which may result in poor glycemic control and increased insulin requirements (49,73,74). Similarly, animal experiments have showed that AP could alter insulin signaling and increase blood glucose concentrations by elevating serum inflammatory cytokines and activating the adaptive immune system (74-77). Maternal alterations in inflammation and insulin signaling pathways caused by AP may directly affect insulin resistance in adult offspring (78). In the rat model of DM, mean platelet count could be elevated in the presence of both AP and periodontitis (79). In addition, alterations in antioxidant status may be one of the potential mechanisms by which AP affects the pathogenesis of DM (80,81). AP could enhance systemic effects of DM, as shown by decreased serum albumin levels and increased uric acid concentrations in DM rats with AP (80).

The efficacy of RCT in diabetes is unknown. In a case report, a patient with DM noticed a rapid increase in insulin demand after an exacerbation of the endodontic-periodontic lesion of a tooth, while the requirement for insulin decreased suddenly within one day of the root canal preparation (82). By contrast, a prospective clinical study with a 1-year follow-up period by Arya et al (67) suggested that endodontic treatment does not improve the levels of HbA1c in patients with DM. Furthermore, a pilot investigation indicated that AP does not affect levels of inflammatory markers or glycemic control in patients with T2DM (83). The limited sample size and uncontrolled confounding variables may partly explain the findings of the pilot study. Moreover, the administration of metformin in some participants with combined DM and AP may be a confounding factor influence blood glucose levels, masking the contribution of AP to blood glucose. In addition, both periapical healing and HbA1c levels in diabetic patients vary over time. Hence, it is critical to determine a consistent and appropriate time to assess levels of HbA1c following endodontic therapy in DM patients with AP.

In summary, there is a two-way association between DM and AP (Fig. 2). DM affects the initiation, progression and prognosis after endodontic treatment of AP. AP might affect insulin sensitivity, increase blood glucose concentrations, as well as enhance the systemic effects of DM. However, it is unclear whether endodontic treatment helps to improve insulin resistance and HbA1c levels in patients with DM and more longitudinal studies are needed.

Osteoporosis may lead to increased AP incidence

Osteoporosis is a common metabolic bone disorder in post-menopausal patients with estrogen deficiency and manifests as increased bone fragility due to bone loss and microarchitectural deterioration of bone tissue (84,85). Osteoporosis decreases total skeletal mass, including jawbone (86-88). Bone changes in osteoporosis are correlated with decrease of alveolar bone density and height of the alveolar ridge and loss of teeth (89-91).

Similarly, López-López et al (87) observed a marginal correlation between osteoporosis and radiolucent periapical lesions in post-menopausal patients. In addition, emerging evidence indicates that the prevalence of AP is higher in osteoporotic patients (92). Bisphosphonates, a type of anti-resorptive medication, inhibit the progression of AP in osteoporotic patients (92-96).

Estrogen deficiency serves a key role in the development of AP in osteoporosis

Systemic factors of bone remodeling in osteoporosis, such as estrogen, modify the balance of periapical bone metabolism. In ovariectomized animal models, estrogen deficiency serves a key role in the pathogenesis of periapical lesion by upregulating expression of members of the NLRP3/caspase-1/IL-1β axis and RANKL (95,97,98), leading to increased periapical bone resorption. The disturbed systemic inflammatory state in patients with osteoporosis is hypothesized to be the main cause of the imbalance between periapical osteoclasts and osteoblasts, which induces osteoclast apoptosis (99,100). Lucisano et al (101) found oral microorganisms in the saliva as well as greater periapical bone loss in ovariectomized mice compared with a control group, which indicated that decreased estrogen aggravates development of periapical lesions by altering the microbiota in saliva. Similarly, Gomes-Filho et al (102) simulated the effects of estrogen by administering raloxifene (RLX) in ovariectomized mice and found that RLX was able to inhibit the production of local regulators of osteoclastogenesis and angiogenesis induced by estrogen deficiency during the development of AP.

Follicle-stimulating hormone (FSH), a hormone that increases with the decrease of estrogen levels, is an independent risk factor for periapical inflammation. FSH could exacerbate bone loss in periapical lesions via directly coupling FSH receptor on the surface of osteoclasts and elevated secretion of inflammatory cytokines (103). FSH inhibitor leuprorelin has a protective effect against periapical bone loss of the experimental periapical lesions in ovariectomized rats (104).

In general, osteoporosis is considered to show a unidirectional association with AP. Osteoporosis increases incidence of AP and estrogen linking these diseases. To the best of our knowledge, however, evidence for this conclusion is limited. More robust epidemiological evidence is required to corroborate this conclusion.

3. Autoimmune disease (AD)

AP is a chronic inflammatory disease involving a variety of immunocompetent cells, similar to the autoimmune system, such as such as T and B cells, macrophages and immunoglobulins synthesized by plasma cells (105,106). Given the similar immunocompetent cells, it is hypothesized that there is an associate the AP with AD. Previous studies have linked types of AD to AP, such as rheumatoid and inflammatory bowel disease (IBD) and autoimmune hepatitis/nephritis (107-109).

In addition, the use of immunosuppressive agents, a conventional option for the treatment of AD, is associated with AP. It is been hypothesized that the use of immunosuppressive agents leads to weakened resistance as a consequence of decreased systemic leukocyte count, thus increasing the risk of opportunistic oral infection and susceptibility to AP (110,111). Conversely, Yamasaki et al (112) conducted an animal study and suggested that the long-term use of immunosuppressive agents prior to pulpal exposure significantly inhibits inflammatory expansion of AP. By contrast, Waterman et al (113) and Teixeira et al (114) showed that use of immunosuppressive agents does not exacerbate or reduce the periapical inflammatory destruction.

The aforementioned contradictory views suggest a complex and uncertain role of the systemic immune response in AP. The pathological process of AP is determined by the balance between host immunity and virulence of the pathogen (111-113) but this dynamic balance is difficult to define. The differing findings may be related to the dose and duration of immunosuppressive agents used, as well as species or virulence of the invading bacteria.

Rheumatoid disease (RD) is associated with a higher prevalence of AP

Rheumatoid disease (RD) is a common autoimmune disease with a global prevalence of 0.24% in 2010 (115). A cross-sectional study conducted by Karataş et al (116) suggested that individuals with RD had at least two times higher risk of AP than controls. Rotstein and Katz (117) reported a similar tendency of patients with RD to develop AP based on the integrated data of hospital cases. Similarly, Oh et al (118) reported a clinical case of rapid destruction of periapical bone during the endodontic therapy in a patient with RD taking azathioprine. Conversely, Jalali et al (119) found no correlation between periapical rarefying osteitis and RD, which might be due to the restrictive diagnostic criteria of AP reducing the actual detection rate of periapical osteitis.

Periapical alveolar bone may be a bony target of RD

AP and RD are types of chronic inflammation involving similar inflammation markers, such as TNF-α, IL-6 and IL-17, which promote bone destruction by activating the NF-κB signaling pathway (120). In 1975, Malmström et al (121,122) performed a series of biopsies on periapical tissue from patients with RD and found no histological differences in periapical lesions between patients with RD and controls. However, they subsequently identified IgG, as well as other free rheumatoid factors, in periapical tissue from patients with RD (123,124). Furthermore, amyloid protein was almost five times more frequent in periapical lesions of patients with RD than in controls (125). Foxo3a, a representative protein that serves an osteolytic role in rheumatoid disease, is found in periapical tissue (126). Therefore, it is hypothesized that the periapical alveolar bone might be a bony target of RD.

Overall, most current studies (123-126) suggest a positive association between RD and AP and the osteolytic destruction of RD could also damage the periapical alveolar bone. To the best of our knowledge, however, there are few convincing studies focusing on the correlation between these diseases and the effect of AP on RD.

Autoimmune hepatitis/nephritis

Autoimmune hepatitis/nephritis is a chronic progressive inflammatory disease mediated by a dysfunctional immune response. Their etiology is obscure but higher susceptibility may be associated with genetics, medication and persistent infection (127-129). In the later stages of progression, severe autoimmune hepatitis/nephritis develops into end-stage liver/renal disease (ESRD), requiring liver/renal transplantation (130).

Autoimmune hepatitis/nephritis confers higher susceptibility to AP

Assessment of the oral health of patients with liver disease revealed that the patients with liver disease exhibit a higher frequency of oral infection (131-133), particularly in liver transplant candidates (134-136). Castellanos-Cosano et al (137) performed a descriptive cross-sectional study that found that the prevalence of AP in liver transplant candidates is higher than in controls, while the frequency of root-filled teeth (RFT) was lower. Furthermore, an epidemiological study by Grønkjær et al (138) confirmed that nearly half of patients with liver cirrhosis exhibited periapical radiolucency. These findings were confirmed by an animal study, in which increased periapical bone loss, inflammatory cell infiltration and expression of inflammatory factors in the periapical tissue were be observed in a rat model of liver fibrosis with AP (139).

Oral disease is also prominent in patients with kidney disease. Children with purpura nephritis are more likely to develop periapical infection (140,141). In patients with ESRD, oral hygiene status is worse than that of healthy individuals (142,143). Additionally, the prevalence of oral disease is associated with severity of the renal failure and worsens with length of dialysis treatment (144,145). A clinal study conducted by Buhlin et al (146) found that more than half of patients with ESRD have at least one tooth with periapical inflammation. In 2017, Khalighinejad et al (147) provided epidemiological evidence of the association between AP and ESRD and indicated that the incidence of AP is notably higher in patients with ESRD and number of AP teeth in patients with ESRD is significantly correlated with urea serum levels.

The aforementioned findings suggest that patients with autoimmune hepatitis/nephritis are at greater risk of AP compared with healthy individuals. The susceptibility of patients with autoimmune hepatitis/nephropathy to periapical inflammation may be partly attributable to liver/renal failure, loss of detoxification and accumulation of harmful metabolites, such as Urea, creatinine, uric acid, resulting in a systemic hyperinflammatory state and immunosuppression (130,148).

AP induces autoimmune hepatitis/nephritis

Bacterial colonies in AP (149) may be a source of infection in patients with autoimmune liver/nephropathy (140). To an extent, bacterial infections of oral origin may influence the morbidity and mortality of patients with autoimmune liver/nephropathy (150). The oral cavity is a well-known source of pathogens while the liver and kidney are key target organs for oral bacteria (151-153) Kojima et al (153) identified viable bacteria in hepatocytes of mice injected with Streptococcus pyogenes. In addition, Ogura et al (152) detected outer membrane antigens of Hemophilus influenzae (a common microorganism in AP) and specific antibodies in glomerular mesangium and serum from patients with nephritis. Moreover, there is a case report of root canal treatment of a child coincidentally inducing Henoch-Schönlein purpura nephritis (154).

The aforementioned data indicate that AP might serve a pathogenic role in autoimmune hepatitis/nephritis. The microorganisms within periapical tissue have the potential to colonize the liver/kidney either directly or by forming immune complexes, triggering a misdirected autoimmune response (152,154).

In brief, there is an interaction between AP and autoimmune hepatitis/nephritis. Extensive epidemiological evidence (137,138,146,147) suggests that patients with autoimmune hepatitis/nephritis are more likely to exhibit AP. Conversely, as an infectious source, periapical inflammation may trigger autoimmune hepatitis/nephritis. Therefore, early and thorough treatment of AP is important. In certain cases, endodontic treatment could be combined with antibiotics, especially in children who have a high propensity to develop purpura nephritis. In addition, it is essential to follow up children with Henoch Schönlein Purpura after endodontic treatment.

Inflammatory bowel disease (IBD)

IBDs are chronic recurrent autoimmune diseases characterized by diffuse inflammation of the intestinal mucosa and include ulcerative colitis (UC) and Crohn's disease (CD), which affect any section of digestive tract (155,156). The connections between IBD and oral health (157), particularly with respect to periodontitis and dental caries, are recognized (158-160). The relationship between AP and IBDs has also gained attention (109).

IBD is correlated with the onset and worsening of AP

In 2017, Piras et al (161) showed that patients with IBD are associated with a higher incidence of AP and higher periapical index (PAI) score, especially in female patients with IBD. Similar results were also observed by Poyato-Borrego et al (162), who confirmed that patients with IBD have nearly six times higher risk of developing AP than healthy individuals. In addition, a case-control study suggested that the number of RFT in patients with IBD is almost four times higher than in controls (163). This may explain why patients with IBD require more dental treatment (164). However, there is no significant difference in prevalence of AP between patients with IBD and controls (165), which might be attributed to the small sample size and uncontrolled confounding factors. The increased susceptibility of patients with IBD to AP might be partially attributable to an impaired immune system, which is the primary characteristic of autoimmune disease (109,166).

In addition to increased incidence, it is hypothesized that the impaired immune system of patients with IBD has a detrimental impact on the progression and prognosis of AP (107,167). CD is a T helper (Th) 1 type of immune disease, whereas UC is defined as a Th2 type of immune disease (168). Both types of immune response are associated with the pathological process of AP. For example, activation of osteoclasts in the Th1 lymphocyte response is implicated in the progression of periapical bone destruction, while the Th2 lymphocyte response is associated with the healing of AP following RCT (169-171). Accordingly, IBD may be associated with the onset and worsening of AP.

Microorganisms from the oral cavity link AP to IBD

From a microscopic perspective, microorganisms originating from the oral cavity may serve a role in the link between AP and IBD (151,172). The oral cavity is the start of the gastrointestinal tract (173). Oral bacteria that enter the gut during swallowing could stimulate intestinal pathogens (necrotrophy) and create novel phenotypical bacterial virulence genes by increasing bacterial virulence (151,174). On the other hand, oral bacteria might be directly linked with digestive disease by altering intestinal flora (175). An animal study in rats by Tavares et al (176) demonstrated that AP can elevate the intestinal leptin levels in metabolic disorders by modulating gut microbiota. Dysbiosis of the intestinal microbiota is a common feature of intestinal diseases, including IBD (177). Recently, Gan et al (178) established an AP model in rat infected by Porphyromonas gingivalis and found that AP changed the diversity of intestinal microbiota and P. gingivalis was not detected in the collected stools. The aforementioned results suggest that periapical inflammation might alter the intestinal flora through multiple pathways (178). Similarly, Kojima et al (153) found a significantly higher detection rate of Streptococcus mutans in patients with IBD compared with that in controls in a clinical study. In addition, it was observed that the injection of S. mutans into mice is more likely to cause IBD than oral administration (153).

Biological medications (BMs) are advocated for treatment of patients with IBD and AP

Immunosuppressive agents are generally used to treat IBD in the clinic. However, use of immunosuppressive agents increases the risk of oral opportunistic infections and bone marrow suppression, thus increasing the susceptibility to AP or promoting further deterioration of periapical lesions (118,179). BMs are recombinant human proteins that target inflammatory factors, such as anti-TNF agents (180). Existing studies have shown that patients with IBDs and AP exhibit faster and better healing with few complications in endodontic treatment combined with anti-TNF therapy (109,181). Theoretically, anti-TNF agents block not only the direct stimulatory effect of TNF-α on osteoclasts, but also the negative effect on osteoblast activity and differentiation, which relieves destruction of periapical tissue and stimulates regeneration of supporting tissue (182). Accordingly, it is hypothesized that patients with IBD and severe treatment-resistant AP and with elevated levels of circulating TNF-α may benefit from anti-TNF treatment.

Current studies (107,161,163) suggest an association between IBD and AP. IBD shows a positive correlation with onset and progression of AP (161-163). In turn, microorganisms in AP lead to the dysbiosis and immune imbalance of the intestinal flora via multiple pathways (176,178). Conventional immunosuppressive agents may not be appropriate for patients with IBD and AP (179). Instead, targeted treatment with BMs to promote healing of systemic disease and AP is favored (180).

4. CVD

CVD, particularly cerebrovascular and ischemic heart disease, are primary causes of mortality and disability worldwide. An estimated 17.7 million deaths could be attributed to CVD, accounting for 31% of all deaths worldwide in 2019 (183), which is a major threat to human life (184,185). Therefore, it is key to determine the risk factors associated with CVD. The relationship between AP and CVD has been investigated for several decades and numerous reviews or meta-analyses have been published (186-189). Most studies have found a weak correlation between AP and CVD (186-191), with only a few disagree (192,193).

Patients with CVD are at higher risk of AP

A systematic review with meta-analysis in 2022 suggested a weak positive association between AP and CVD (194). Atherosclerosis is the main pathological basis of CVD and is highly correlated with the prevalence of AP, as shown by epidemiological studies (195,196). Additionally, Conti et al (197) confirmed that atherosclerosis increases the inflammatory reaction and size of periapical lesions. As one of the most common types of CVD, coronary artery disease (CAD) is associated with a higher prevalence of periapical inflammation (198-200). In comparison with non-CAD subjects, patients with CAD have a nearly threefold increase in the risk of AP (201), which is consistent with another study detected AP in 42.6% of patients with CAD and in 40.1% of non-CAD controls (202). Hypertension, a recognized primary risk factor for CVD, is also linked with a high prevalence and increased radiographic area of AP (203), and the prevalence of AP in hypertensive patients is notably decreased following treatment with angiotensin II receptor blockers (204).

AP contributes to progression of CVD

Gan et al (178) established a model of AP by inoculating P. gingivalis into the pulpal cavity of apolipoprotein E-deficient mice and found that AP exacerbated formation of atherosclerotic plaque in the aortic arch. Consistently, Conti et al (197) demonstrated that AP could promote the progression of atherosclerosis by increasing triglyceride levels and the thickness of the carotid artery intima tunic. Furthermore, endodontic therapy is hypothesized to be an effective approach to reverse the inflammatory effect of AP on atherosclerosis (205). In patients with hypertension combined with AP, AP increases levels of serum inflammatory markers such as IL-6, CRP and fibrinogen, disturbs the balance of the oxidative state and impairs cardiac function (203,206,207). Messing et al (208) found a positive association between AP and a single nucleotide polymorphism in potassium channel subfamily K member 3 (KCNK3), a gene that increases susceptibility to hypertension, which suggested that AP and hypertension may share a common genetic variation. AP is linked with an increased risk of first myocardial infarction (209).

Potential mechanisms linking AP and CVD

Numerous underlying mechanisms have been proposed for the association between AP and CVD (Fig. 3). Bacteria and/or their products may translocate from periapical tissue to various parts of the body via systemic circulation, especially areas with pre-existing cardiovascular lesions, and accelerate the progression of CVD. For example, DNA of P. endodontalis in monocytes (210) and elevated levels of anti-P. endodontalis IgG are found in peripheral circulation of patients with AP (211). In addition, Kazanci et al (212) reported an 8-year-old boy was admitted to hospital for acute cerebral infarct; cerebrospinal fluid cultures yielded Streptococcus oralis from alveolar abscess of the left maxillary first premolar. These findings are in accordance with a previous study that found bacteria and/or their products in circulation trigger low-grade systemic inflammation and contribute to the formation of plaques or thrombi (178).

Inflammatory response (197), oxidative stress (203) and endothelial dysfunction (213) are potential mechanisms underlying the interaction between AP and CVD. Inflammatory mediators serve a crucial role in the initiation and progression of both AP and CVD (196). Systemic inflammation, which is amplified by CVDs, also acts on periapical tissues by intensifying the inflammatory reaction and increasing bone resorption in periapical lesions (197). In addition, inflammatory mediators mediate endothelial dysfunction (214). Previous studies have demonstrated that patients with AP exhibit increased lipid levels and serum inflammatory mediators (TNF-α, IL-1, IL-6, IL-2 and asymmetric dimethylarginine), which implies early endothelial dysfunction (196,197,213,215). Moreover, Chauhan et al (216) observed impaired flow-mediated dilatation and endothelial flow reserve, as well as greater carotid intima-media thickness, in individuals with AP compared with controls in a cross-sectional study. However, successful local endodontic therapy does not ameliorate early endothelial dysfunction (215).

In terms of oxidative stress, reactive oxygen species (ROS) are by-products of cellular metabolism involved in the pathological process of both AP and atherosclerosis (217). During pulp infection, bacterial motifs bind toll-like receptors (TLRs) on the surface of phagocytes and promote synthesis and release of ROS (218). Furthermore, the dysbiosis of intestinal flora due to AP may be a source of accumulated ROS in the body (96,176). Excess ROS production directly damages the vascular endothelium and activates multiple components of the immune system, such as increasing the inflammatory cytokine expression and amplifying the neutrophil recruitment (219), which was confirmed by a study demonstrating that AP could disrupt the oxidative status and impair cardiodynamics in hypertensive rats (203). Moreover, antioxidant agent tempol exerts a prevent effect on the establishment of AP in rats with doxorubicin-induced cardiomyopathy (220).

Previous studies (194,195,197) suggest an association between AP and CVD. To the best of our knowledge, however, detailed epidemiological studies on the mechanism underlying their interaction are lacking, especially for the effect of CVD on AP. CVD is a highly complex disease with various risk factors so more well-designed animal and epidemiological studies with strict control of confounding factors are essential to clarify the causal association between these two diseases.

5. APO

APOs are harmful to both pregnant people and newborn infants and related with early spontaneous abortion to an extent (221,222). In particular, the alteration of hormone levels during pregnancy increases vascular permeability and accelerates the spread of inflammation (223). Maternal infection is more likely to cross the placental barrier, leading to APOs (224).

Epidemiological evidence of the association between AP and APO

Harjunmaa et al (225) found a significant association between prevalence of AP and preterm delivery and fetal growth restriction in a cross-sectional study in Malawi. Leal et al (226) performed a case-control study involving 63 pregnant people and found that pregnant people with AP were nearly 5 times more likely to undergo preterm labor and to deliver a low-birth weight (LBW) newborn than those without AP. In addition to the adverse effects on fetal intrauterine growth, pregnant patients with AP exhibit greater risk of developing preeclampsia compared with those without AP (227). To an extent, the likelihood of APO varies with size of the periapical lesion (226). By contrast, Bain et al (228,229) hypothesized that AP contributes to a higher birthweight and a longer pregnancy, which might be related to gestational diabetes caused by insulin resistance (228-230).

Shah et al (231) recently conducted a cross-sectional study and found a protective association between AP and APOs, which is contrary to previous results (226,227). This result is biologically implausible, as the invasion of oral pathogens in periapical periodontitis is intrinsically harmful to the body and it can trigger a series of resistance behaviors of the body. This implausible outcome may be caused by sample selection bias, excessive loss to follow-up and uncontrolled confounding factors. Additionally, defining PAI≥3 as a diagnostic criterion for AP may underestimate the incidence of AP in the sample population (232).

Potential underlying mechanisms of AP and APO

To verify the relationship between AP and APO, Ao et al (233) created an AP model in rat infected with P. gingivalis and found that AP was associated with LBW and preterm birth (LBWPB). Immunohistochemistry and PCR showed P. gingivalis localization in placental tissue. Consistent with these results, Doyle et al (234) detected higher levels of Fusobacterium nucleatum, a representative specialized anaerobic bacteria in oral cavity, in amniotic fluid of premature infants.

The aforementioned results suggest that bacteria from periapical lesions could enter the circulation and induce temporary bacteremia, together with increased vascular permeability due to changes in hormone levels during pregnancy, which allow bacteria in the bloodstream to easily cross the placental barrier and induce maternal and fetal inflammatory reactions and immune responses. TNF-α, IL-1β and IL-6 induce premature labor, while IL-10 and vascular endothelial growth factor prevent LBW (235-238). Theoretically, maternal infection with P. gingivalis may inhibit expression of IL-10, thereby promoting intrauterine growth restriction; therefore, AP may induce LBWPB (239).

To the best of our knowledge, studies about the association between AP and APOs are lacking. The majority of studies show that AP is positively associated with APO. However, more studies are required to investigate the association between AP and APO and determine whether it is necessary to take preventive measures against AP in pregnant people.

6. Psychiatric disorder

With the rapid pace and increasing pressure of modern society, psychiatric disorders are becoming increasingly prominent, it is estimated the lifetime prevalence rates were 22.5% for any non-substance abuse mental disorder in the United States (240). Excessive stress, persistent negative emotion and severe sleep disorder affect psychiatric health to varying degrees. On one hand, psychiatric disorders affect the balance of endocrine hormone and immune response, which decreases host resistance and increases susceptibility to chronic disease (241). On the other hand, objective causes such as difficulties in maintaining oral hygiene confer susceptibility to AP. Negative mental state is involved in the underlying mechanism of various types of oral disease, such as periodontitis, oral mucosal ulcers and bruxism (241-243). For example, Maes (244) hypothesized that severe depression is accompanied by hypersecretion of adrenaline, disordered systemic immune response and activation of acute inflammatory markers, which lead to decreased host resistance and allow oral pathogens to trigger periapical lesions. Furthermore, in the dental clinic, excessive mental stress and disturbed sleep schedule are common in asymptomatic patients with AP who typically seek emergency dental treatment for acute toothache (245).

Wang et al (246) found that P. gingivalis induces a depressive phenotype. The potential mechanism may be that the free lipopolysaccharide (LPS) in the blood crosses the blood-brain barrier and downregulates expression of neurotrophic factor receptor p75 in astrocytes via the LPS/TLR4 signaling pathway (246). Furthermore, Martínez et al (247) detected the translocation of Fusobacterium nucleatum in in the frontal cortex and hypothesized that F. nucleatum might link oral disease to depression by inducing neuroinflammation (248). Transfer of oral bacteria across the blood-brain barrier may underlie the association between oral disease and psychiatric disorder.

Mental stress is associated with severity of AP

Mental stress is a state of physical or psychological tension caused by adverse mental or emotional stimuli that interferes with normal physiological functions of the body (249). Studies (250,251) have shown that the levels of catecholamines, cortisol hormones and inflammatory factors in the body are closely associated with mental stress Excessive stress stimulates secretion of catecholamines by exciting the sympathetic nervous system and activating the hypothalamic-pituitary-adrenocortical axis, promoting the release of cortisol (250,252). Cortisol regulates immunity by decreasing the number and activity of immune cells and inhibiting secretion of cytokines (253). In a survey, Haug and Marthinussen (245) found that a sudden onset of acute toothache in patients with chronic AP is typically associated with greater stress at home or work; higher levels of cortisol and inflammatory factors in saliva may be the direct cause of acute episodes of AP (254). Similarly, exogenous chronic stress exacerbates the pathological process of AP by increasing periapical bone resorption and release of inflammatory cytokines (255,256). In addition, the application of adrenergic blockers is effective in reducing the number of osteoclasts in the periapical region (257). Thus, mental stress induces imbalance between the secretion of cortisol and inflammatory factors in saliva, which is strongly correlated with the severity of AP (254).

Sleep disorder affects progression of AP by altering hormone levels

Sleep disorder is a common key clinical feature of psychiatric disorder (258,259). Sleep disorder is a key risk factor for oral cancer, periodontitis and adolescent maxillofacial development (260-262). Sleep disorder affects rhythmic expression of various proinflammatory cytokines and endocrine hormones, including cortisol and melatonin (263). Intraperitoneal injection or sealing of cortisol in the root canal in an AP rat model causes damage to periapical tissue and increases resorption of periapical bone (254,264). Melatonin is an important hormone with anti-inflammatory and antioxidant activity that is secreted by the pineal gland during the night (265). Melatonin suppresses oral inflammation and decreases loss of alveolar bone (266). Tavares et al (267) found that melatonin is a feasible adjuvant to promote healing of periapical bone in AP by increasing the insulin sensitivity and decreasing plasma concentrations of inflammatory cytokines. In addition, Sarıtekin et al (268) demonstrated that systemic injection of melatonin could significantly inhibit the resorption of periapical alveolar bone and decrease plasma expression of inflammatory factors in rats with AP. Therefore, it is hypothesized that sleep disturbance may be a risk factor in exacerbating the inflammatory response of periapical tissue in AP.

Studies suggest (253,254) that the association between psychiatric disorder and AP is primarily mediated by disruption of hormone levels and the imbalance of host immunoreaction. In clinical practice, doctors should attach importance to the oral condition of patients with psychiatric disorder and provide necessary oral health education. However, due to the lack of epidemiological data and limited animal studies, the association between AP and psychiatric disorder is unclear. Therefore, more studies are needed to investigate the interaction between AP and psychiatric disorder.

7. Conclusion

An increasing number of studies (18-20) have investigated the association between AP and systemic disease, including animal experiments, clinical trials and epidemiological investigations. Therefore, in the diagnosis and treatment of AP, dentists should consider systemic disease and medications to assess patient condition, develop a therapeutic plan and evaluate prognosis. Physicians should be aware of the periapical status of patients with systemic disease to minimize the negative impact of AP. For example, physicians should raise awareness of oral health in patients with systemic disease associated with AP, focus on the possible adverse effects of specific drugs, such as immunosuppressants like methotrexate and cyclophosphamide, on the periapical tissue. Furthermore, physicians should collaborate with outpatient dentists for regular follow-up to better assess patient condition and oral status. As AP may be one of the etiologies or clinical manifestations of certain types of systemic disease, focusing on the interaction between AP and systemic disease will help dentists and physicians seek reasonable therapeutic options to promote the healing of AP and systemic disease as effectively as possible.

However, the majority of current studies (18,19,27) only provide the evidence of the association between AP and systemic diseases rather than a strict causal association. Associations between AP and systemic diseases are complex, which may be bidirectional or unidirectional. In addition, the association is susceptible to multiple confounding factors. It is difficult to exclude confounding factors to establish a strict causal association, as shown by a review by Segura-Egea et al (27). Establishment of strict causality should analyze and achieve the Hill's criteria (strength of the association, biological rationality, temporal relationship, dose-response gradient, consistency of the association). To the best of our knowledge, there are few studies (67,82) that have investigated whether RCT could decrease the epidemiological incidence or attenuate clinical symptoms of systemic disease. Therefore, clinical trials are needed to confirm AP as a potential risk factor for systemic disease. Furthermore, confounding factors and bias should be controlled as much as possible during the design process of the correlation studies, which will help to elucidate the correlation and the underlying mechanisms about the association between AP and systemic disease more convincingly.

Availability of data and materials

Not applicable.

Authors' contributions

LY, LC, WS and CY wrote and revised the manuscript and constructed table and figures. QT and ZY conceived the study and revised the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

AP

apical periodontitis

T2DM

type 2 diabetes mellitus

RCT

root canal treatment

CVD

cardiovascular disease

HbA1c

glycated hemoglobin

CAD

coronary artery disease

CRP

C-reactive protein

ROS

reactive oxygen species

IBD

inflammatory bowel disease

CD

Crohn's disease

UC

ulcerative colitis

PAI

periapical index

RFT

root-filled teeth

RD

rheumatoid disease

RLX

raloxifene

FSH

follicle-stimulating hormone

ESRD

end-stage renal disease

TLR

toll-like receptor

LBWPB

low birth weight and preterm birth

APO

adverse pregnancy outcome

Acknowledgments

Not applicable.

Funding

The present study was supported by National Natural Science Foundation of China (grant no. 82000500), Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration (grant no. 2021kqhm004) and Scientific Research Program of Hubei Provincial Health Commission (grant no. WJ2021Q059).

References

1 

Yamasaki M, Kumazawa M, Kohsaka T, Nakamura H and Kameyama Y: Pulpal and periapical tissue reactions after experimental pulpal exposure in rats. J Endod. 20:13–17. 1994. View Article : Google Scholar : PubMed/NCBI

2 

Kakehashi S, Stanley HR and Fitzgerald RJ: The effects of surgical exposures of dental pulps in germ-free and conventional laboratory rats. Oral Surg Oral Med Oral Pathol. 20:340–349. 1965. View Article : Google Scholar : PubMed/NCBI

3 

Tibúrcio-Machado CS, Michelon C, Zanatta FB, Gomes MS, Marin JA and Bier CA: The global prevalence of apical periodontitis: A systematic review and meta-analysis. Int Endod J. 54:712–735. 2021. View Article : Google Scholar

4 

Bahrami G, Vaeth M, Kirkevang LL, Wenzel A and Isidor F: Risk factors for tooth loss in an adult population: A radiographic study. J Clin Periodontol. 35:1059–1065. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Gomes MS, Hugo FN, Hilgert JB, Sant'Ana Filho M, Padilha DM, Simonsick EM, Ferrucci L and Reynolds MA: Apical periodontitis and incident cardiovascular events in the baltimore longitudinal study of ageing. Int Endod J. 49:334–342. 2016. View Article : Google Scholar

6 

Kierdorf U, Olsen MT, Kahle P, Ludolphy C and Kierdorf H: Dental pulp exposure, periapical inflammation and suppurative osteomyelitis of the jaws in juvenile Baltic grey seals (Halichoerus grypus grypus) from the late 19th century. PLoS One. 14:e02154012019. View Article : Google Scholar : PubMed/NCBI

7 

Cintra LTA, Gomes MS, da Silva CC, Faria FD, Benetti F, Cosme-Silva L, Samuel RO, Pinheiro TN, Estrela C, González AC and Segura-Egea JJ: Evolution of endodontic medicine: A critical narrative review of the interrelationship between endodontics and systemic pathological conditions. Odontology. 109:741–769. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Li X, Kolltveit KM, Tronstad L and Olsen I: Systemic diseases caused by oral infection. Clin Microbiol Rev. 13:547–558. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Savarrio L, Mackenzie D, Riggio M, Saunders WP and Bagg J: Detection of bacteraemias during non-surgicalroot canal treatment. J Dent. 33:293–303. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Baumgartner JC, Heggers JP and Harrison JW: The incidence of bacteremias related to endodontic procedures. I. Nonsurgical endodontics. J Endod. 2:135–140. 1976. View Article : Google Scholar : PubMed/NCBI

11 

Debelian GJ, Olsen I and Tronstad L: Bacteremia in conjunction with endodontic therapy. Endod Dent Traumatol. 11:142–149. 1995. View Article : Google Scholar : PubMed/NCBI

12 

Gomes MS, Blattner TC, Sant'Ana Filho M, Grecca FS, Hugo FN, Fouad AF and Reynolds MA: Can apical periodontitis modify systemic levels of inflammatory markers? A systematic review and meta-analysis. J Endod. 39:1205–1217. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Samuel RO, Gomes-Filho JE, Azuma MM, Sumida DH, de Oliveira SHP, Chiba FY, Bomfim SRM, Ciarlini PC, Narciso LG and Cintra LTA: Endodontic infections increase leukocyte and lymphocyte levels in the blood. Clin Oral Investig. 22:1395–1401. 2018. View Article : Google Scholar

14 

Poornima L, Ravishankar P, Abbott PV, Subbiya A and PradeepKumar AR: Impact of root canal treatment on high-sensitivity C-reactive protein levels in systemically healthy adults with apical periodontitis-a preliminary prospective, longitudinal interventional study. Int Endod J. 54:501–508. 2021. View Article : Google Scholar

15 

Chen Y, Liu S and Leng SX: Chronic low-grade inflammatory phenotype (CLIP) and senescent immune dysregulation. Clin Ther. 41:400–409. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Chan LT, Zhong S, Naqvi AR, Self-Fordham J, Nares S, Bair E and Khan AA: MicroRNAs: New insights into the pathogenesis of endodontic periapical disease. J Endod. 39:1498–1503. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Sasaki H, Hirai K, Martins CM, Furusho H, Battaglino R and Hashimoto K: Interrelationship between periapical lesion and systemic metabolic disorders. Curr Pharm Des. 22:2204–2215. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Segura-Egea JJ, Martín-González J and Castellanos-Cosano L: Endodontic medicine: Connections between apical periodontitis and systemic diseases. Int Endod J. 48:933–951. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Khalighinejad N, Aminoshariae MR, Aminoshariae A, Kulild JC, Mickel A and Fouad AF: Association between systemic diseases and apical periodontitis. J Endod. 42:1427–1434. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Cotti E and Mercuro G: Apical periodontitis and cardiovascular diseases: Previous findings and ongoing research. Int Endod J. 48:926–932. 2015. View Article : Google Scholar : PubMed/NCBI

21 

American Diabetes Association: Diagnosis and classification of diabetes mellitus. Diabetes Care. 36(Suppl 1): S67–S74. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Wild S, Roglic G, Green A, Sicree R and King H: Global prevalence of diabetes: Estimates for the year 2000 and projections for 2030. Diabetes Care. 27:1047–1053. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Cho NH, Shaw JE, Karuranga S, Huang Y, da Rocha Fernandes JD, Ohlrogge AW and Malanda B: IDF diabetes atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res Clin Pract. 138:271–281. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Velasco-Ortega E, Delgado-Ruiz RA and López-López J: Dentistry and diabetes: The influence of diabetes in oral diseases and dental treatments. J Diabetes Res. 2016:60731902016. View Article : Google Scholar

25 

Haag DG, Peres KG, Balasubramanian M and Brennan DS: Oral conditions and health-related quality of life: A systematic review. J Dent Res. 96:864–874. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Rohani B: Oral manifestations in patients with diabetes mellitus. World J Diabetes. 10:485–489. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Segura-Egea JJ, Cabanillas-Balsera D, Jiménez-Sánchez MC and Martín-González J: Endodontics and diabetes: Association versus causation. Int Endod J. 52:790–802. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Tibúrcio-Machado CDS, Bello MDC, Maier J, Wolle CFB and Bier CAS: Influence of diabetes in the development of apical periodontitis: A critical literature review of human studies. J Endod. 43:370–376. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Yip N, Liu C, Wu D and Fouad AF: The association of apical periodontitis and type 2 diabetes mellitus: A large hospital network cross-sectional case-controlled study. J Am Dent Assoc. 152:434–443. 2021. View Article : Google Scholar : PubMed/NCBI

30 

Pérez-Losada FDL, Estrugo-Devesa A, Castellanos-Cosano L, Segura-Egea JJ, López-López J and Velasco-Ortega E: Apical periodontitis and diabetes mellitus type 2: A systematic review and meta-analysis. J Clin Med. 9:5402020. View Article : Google Scholar : PubMed/NCBI

31 

Segura-Egea JJ, Jiménez-Pinzón A, Ríos-Santos JV, Velasco-Ortega E, Cisneros-Cabello R and Poyato-Ferrera M: High prevalence of apical periodontitis amongst type 2 diabetic patients. Int Endod J. 38:564–569. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Smadi L: Apical periodontitis and endodontic treatment in patients with type II diabetes mellitus: Comparative cross-sectional survey. J Contemp Dent Pract. 18:358–362. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Sánchez-Domínguez B, López-López J, Jané-Salas E, Castellanos-Cosano L, Velasco-Ortega E and Segura-Egea JJ: Glycated hemoglobin levels and prevalence of apical periodontitis in type 2 diabetic patients. J Endod. 41:601–606. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Saleh W, Xue W and Katz J: Diabetes mellitus and periapical abscess: A cross-sectional study. J Endod. 46:1605–1609. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Sisli SN: Evaluation of the relationship between type II diabetes mellitus and the prevalence of apical periodontitis in root-filled teeth using cone beam computed tomography: An observational cross-sectional study. Med Princ Pract. 28:533–538. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Mesgarani A, Haghanifar S, Eshkevari N, Ehsani M, Khafri S, Nafarzade S and Damankesh Z: Frequency of odontogenic periradicular lesions in diabetic patients. Caspian J Intern Med. 5:22–25. 2014.PubMed/NCBI

37 

Kohsaka T, Kumazawa M, Yamasaki M and Nakamura H: Periapical lesions in rats with streptozotocin-induced diabetes. J Endod. 22:418–421. 1996. View Article : Google Scholar : PubMed/NCBI

38 

Al-Nazhan SA, Alsaeed SA, Al-Attas HA, Dohaithem AJ, Al-Serhan MS and Al-Maflehi NS: Prevalence of apical periodontitis and quality of root canal treatment in an adult Saudi population. Saudi Med J. 38:413–421. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Falk H, Hugoson A and Thorstensson H: Number of teeth, prevalence of caries and periapical lesions in insulin-dependent diabetics. Scand J Dent Res. 97:198–206. 1989.PubMed/NCBI

40 

Alghofaily M, Tordik P, Romberg E, Martinho F and Fouad AF: Healing of apical periodontitis after nonsurgical root canal treatment: The role of statin intake. J Endod. 44:1355–1360. 2018. View Article : Google Scholar : PubMed/NCBI

41 

De la Torre-Luna R, Domínguez-Pérez RA, Guillén-Nepita AL, Ayala-Herrera JL, Martínez-Martínez RE, Romero-Ayala ME, Pérez-Serrano RM and Vázquez-Garcidueñas MS: Prevalence of Candida albicans in primary endodontic infections associated with a higher frequency of apical periodontitis in type two diabetes mellitus patients. Eur J Clin Microbiol Infect Dis. 39:131–138. 2020. View Article : Google Scholar

42 

Graves DT, Corrêa JD and Silva TA: The oral microbiota is modified by systemic diseases. J Dent Res. 98:148–156. 2019. View Article : Google Scholar :

43 

Samuel RO, Ervolino E, de Azevedo Queiroz ÍO, Azuma MM, Ferreira GT and Cintra LTA: Th1/Th2/Th17/Treg balance in apical periodontitis of normoglycemic and diabetic rats. J Endod. 45:1009–1015. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Cintra LTA, da Silva Facundo AC, Prieto AKC, Sumida DH, Narciso LG, Mogami Bomfim SR, Oliveira e Silva C, Dezan-Júnior E and Gomes-Filho JE: Blood profile and histology in oral infections associated with diabetes. J Endod. 40:1139–1144. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Azuma MM, Gomes-Filho JE, Prieto AKC, Samuel RO, de Lima VMF, Sumida DH, Ervolino E and Cintra LTA: Diabetes increases interleukin-17 levels in periapical, hepatic, and renal tissues in rats. Arch Oral Biol. 83:230–235. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Hasiakos S, Gwack Y, Kang M and Nishimura I: Calcium signaling in T cells and chronic inflammatory disorders of the oral cavity. J Dent Res. 100:693–699. 2021. View Article : Google Scholar : PubMed/NCBI

47 

Wu Y, Yang Y, Wang L, Chen Y, Han X, Sun L, Chen H and Chen Q: Effect of Bifidobacterium on osteoclasts: TNF-α/NF-κB inflammatory signal pathway-mediated mechanism. Front Endocrinol (Lausanne). 14:11092962023. View Article : Google Scholar

48 

Iwama A, Nishigaki N, Nakamura K, Imaizumi I, Shibata N, Yamasaki M, Nakamura H, Kameyama Y and Kapila Y: The effect of high sugar intake on the development of periradicular lesions in rats with type 2 diabetes. J Dent Res. 82:322–325. 2003. View Article : Google Scholar : PubMed/NCBI

49 

Cintra LTA, Samuel RO, Azuma MM, Ribeiro CP, Narciso LG, de Lima VM, Sumida DH, Coclete GA, Dezan-Júnior E and Gomes-Filho JE: Apical periodontitis and periodontal disease increase serum IL-17 levels in normoglycemic and diabetic rats. Clin Oral Investig. 18:2123–2128. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Bahrambeigi S, Yousefi B, Rahimi M and Shafiei-Irannejad V: Metformin; an old antidiabetic drug with new potentials in bone disorders. Biomed Pharmacother. 109:1593–1601. 2019. View Article : Google Scholar

51 

Zhu M, Zhao Z, Xu HHK, Dai Z, Yu K, Xiao L, Schneider A, Weir MD, Oates TW, Bai Y and Zhang K: Effects of metformin delivery via biomaterials on bone and dental tissue engineering. Int J Mol Sci. 23:159052022. View Article : Google Scholar : PubMed/NCBI

52 

Wang S, Xia Y, Ma T, Weir MD, Ren K, Reynolds MA, Shu Y, Cheng L, Schneider A and Xu HHK: Novel metformin-containing resin promotes odontogenic differentiation and mineral synthesis of dental pulp stem cells. Drug Deliv Transl Res. 9:85–96. 2019. View Article : Google Scholar

53 

Zhang R, Liang Q, Kang W and Ge S: Metformin facilitates the proliferation, migration, and osteogenic differentiation of periodontal ligament stem cells in vitro. Cell Biol Int. 44:70–79. 2020. View Article : Google Scholar

54 

Liu L, Zhang C, Hu Y and Peng B: Protective effect of metformin on periapical lesions in rats by decreasing the ratio of receptor activator of nuclear factor kappa B ligand/osteoprotegerin. J Endod. 38:943–947. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Lai EHH, Yang CN, Lin SK, Wang HW, Kok SH, Hong CY, Su IH, Yang H and Chang JZC: Metformin ameliorates periapical lesions through suppression of hypoxia-induced apoptosis of osteoblasts. J Endod. 44:1817–1825. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Wang HW, Lai EHH, Yang CN, Lin SK, Hong CY, Yang H, Chang JZC and Kok SH: Intracanal metformin promotes healing of apical periodontitis via suppressing inducible nitric oxide synthase expression and monocyte recruitment. J Endod. 46:65–73. 2020. View Article : Google Scholar

57 

Sarmento EB, Gomes CC, Pires FR, Pinto LC, Antunes LAA and Armada L: Immunoexpression of bone resorption biomarkers in apical periodontitis in diabetics and normoglycaemics. Int Endod J. 53:1025–1032. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Wang CH, Chueh LH, Chen SC, Feng YC, Hsiao CK and Chiang CP: Impact of diabetes mellitus, hypertension, and coronary artery disease on tooth extraction after nonsurgical endodontic treatment. J Endod. 37:1–5. 2011. View Article : Google Scholar

59 

Mindiola MJ, Mickel AK, Sami C, Jones JJ, Lalumandier JA and Nelson SS: Endodontic treatment in an American Indian population: A 10-year retrospective study. J Endod. 32:828–832. 2006. View Article : Google Scholar : PubMed/NCBI

60 

Lima SMF, Grisi DC, Kogawa EM, Franco OL, Peixoto VC, Gonçalves-Júnior JF, Arruda MP and Rezende TM: Diabetes mellitus and inflammatory pulpal and periapical disease: A review. Int Endod J. 46:700–709. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Uğur Aydın Z, Ocak MG, Bayrak S, Göller Bulut D and Orhan K: The effect of type 2 diabetes mellitus on changes in the fractal dimension of periapical lesion in teeth after root canal treatment: A fractal analysis study. Int Endod J. 54:181–189. 2021. View Article : Google Scholar

62 

Rudranaik S, Nayak M and Babshet M: Periapical healing outcome following single visit endodontic treatment in patients with type 2 diabetes mellitus. J Clin Exp Dent. 8:e498–e504. 2016.PubMed/NCBI

63 

Martinho JP, Coelho A, Oliveiros B, Pires S, Abrantes AM, Paulo S, Carvalho AC, Carrilho E, Paula A, Carvalho L, et al: Impairment of the angiogenic process may contribute to lower success rate of root canal treatments in diabetes mellitus. Int Endod J. 54:1687–1698. 2021. View Article : Google Scholar : PubMed/NCBI

64 

Britto LR, Katz J, Guelmann M and Heft M: Periradicular radiographic assessment in diabetic and control individuals. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 96:449–452. 2003. View Article : Google Scholar : PubMed/NCBI

65 

Fouad AF and Burleson J: The effect of diabetes mellitus on endodontic treatment outcome: Data from an electronic patient record. J Am Dent Assoc. 134(43-51): 117–118. 2003. View Article : Google Scholar

66 

Laukkanen E, Vehkalahti MM and Kotiranta AK: Impact of systemic diseases and tooth-based factors on outcome of root canal treatment. Int Endod J. 52:1417–1426. 2019. View Article : Google Scholar : PubMed/NCBI

67 

Arya S, Duhan J, Tewari S, Sangwan P, Ghalaut V and Aggarwal S: Healing of apical periodontitis after nonsurgical treatment in patients with type 2 diabetes. J Endod. 43:1623–1627. 2017. View Article : Google Scholar : PubMed/NCBI

68 

Limeira FIR, Arantes DC, de Souza Oliveira C, de Melo DP, Magalhães CS and Bento PM: Root canal treatment and apical periodontitis in a brazilian population with type 1 diabetes mellitus: A cross-sectional paired study. J Endod. 46:756–762. 2020. View Article : Google Scholar : PubMed/NCBI

69 

Doyle SL, Hodges JS, Pesun IJ, Baisden MK and Bowles WR: Factors affecting outcomes for single-tooth implants and endodontic restorations. J Endod. 33:399–402. 2007. View Article : Google Scholar : PubMed/NCBI

70 

Marotta PS, Fontes TV, Armada L, Lima KC, Rôças IN and Siqueira JF Jr: Type 2 diabetes mellitus and the prevalence of apical periodontitis and endodontic treatment in an adult Brazilian population. J Endod. 38:297–300. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Ng YL, Mann V and Gulabivala K: A prospective study of the factors affecting outcomes of nonsurgical root canal treatment: Part 1: Periapical health. Int Endod J. 44:583–609. 2011. View Article : Google Scholar : PubMed/NCBI

72 

Bloomgarden ZT: Inflammation and insulin resistance. Diabetes Care. 26:1619–1623. 2003. View Article : Google Scholar : PubMed/NCBI

73 

Cintra LTA, Samuel RO, Facundo ACS, Prieto AK, Sumida DH, Bomfim SR, Souza JC, Dezan-Júnior E and Gomes-Filho JE: Relationships between oral infections and blood glucose concentrations or HbA1c levels in normal and diabetic rats. Int Endod J. 47:228–237. 2014. View Article : Google Scholar

74 

Astolphi RD, Curbete MM, Colombo NH, Shirakashi DJ, Chiba FY, Prieto AK, Cintra LT, Bomfim SR, Ervolino E and Sumida DH: Periapical lesions decrease insulin signal and cause insulin resistance. J Endod. 39:648–652. 2013. View Article : Google Scholar : PubMed/NCBI

75 

Astolphi RD, Curbete MM, Chiba FY, Cintra LT, Ervolino E, da Mota MS, Antoniali C, Garbin CA and Sumida DH: Periapical lesions decrease insulin signaling in rat skeletal muscle. J Endod. 41:1305–1310. 2015. View Article : Google Scholar : PubMed/NCBI

76 

Pereira RF, de Oliveira da Mota MS, de Lima Coutinho Mattera MS, Tsosura TV, Chiba FY, Garbin CA, Ervolino E, Cintra LT, Okamoto MM, Machado UF and Sumida DH: Periapical lesions decrease Akt serine phosphorylation and plasma membrane GLUT4 content in rat skeletal muscle. Clin Oral Investig. 20:1625–1630. 2016. View Article : Google Scholar

77 

Felipe Pereira R, Willian Lattari Tessarin G, Yamamoto Chiba F, Sara de Lima Coutinho Mattera M, Gomes Pereira A, Verônica Saori Tsosura T, Gustavo Balera Brito V, Akira Fujii, de Oliveira R, Ervolino E, Helena Penha, de Oliveira S, et al: Apical periodontitis promotes insulin resistance and alters adaptive immunity markers in rats. Saudi Dent J. 33:979–986. 2021. View Article : Google Scholar : PubMed/NCBI

78 

Tsosura TVS, Chiba FY, Mattera MSLC, Pereira RF, Cintra LTA, Conti LC, Santos RMD, Mateus JHP, Garbin CAS and Sumida DH: Maternal apical periodontitis is associated with insulin resistance in adult offspring. Int Endod J. 52:1040–1050. 2019. View Article : Google Scholar : PubMed/NCBI

79 

Ferreira LL: Diabetic rats present high mean platelet count in the presence of oral infections. Braz Dent J. 28:548–551. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Prieto AKC, Gomes-Filho JE, Azuma MM, Sivieri-Araújo G, Narciso LG, Souza JC, Ciarlini PC and Cintra LTA: Influence of apical periodontitis on stress oxidative parameters in diabetic rats. J Endod. 43:1651–1656. 2017. View Article : Google Scholar : PubMed/NCBI

81 

Barcelos RCS, Rosa HZ, Roversi K, Tibúrcio-Machado CDS, Inchaki PT, Burger ME and Bier CAS: Apical periodontitis induces changes on oxidative stress parameters and increases Na+/K+-ATPase activity in adult rats. Arch Oral Biol. 118:1048492020. View Article : Google Scholar

82 

Schulze A, Schönauer M and Busse M: Sudden improvement of insulin sensitivity related to an endodontic treatment. J Periodontol. 78:2380–2384. 2007. View Article : Google Scholar : PubMed/NCBI

83 

Stys LPA, Böttcher DE, Scarparo RK, Gonçalves Waltrick SB, de Figueiredo JAP, Gomes MS and Campos MM: Serum levels of inflammatory markers and HbA1c in patients with type 2 diabetes and apical periodontitis: Preliminary findings. Aust Endod J. 48:105–115. 2022. View Article : Google Scholar

84 

No authors listed. Consensus development conference: Diagnosis, prophylaxis, and treatment of osteoporosis. Am J Med. 94:646–650. 1993. View Article : Google Scholar : PubMed/NCBI

85 

NIH Consensus Development Panel on Osteoporosis Prevention, Diagnosis, and Therapy: Osteoporosis prevention, diagnosis, and therapy. JAMA. 285:785–795. 2001. View Article : Google Scholar

86 

Gilles JA, Carnes DL, Dallas MR, Holt SC and Bonewald LF: Oral bone loss is increased in ovariectomized rats. J Endod. 23:419–422. 1997. View Article : Google Scholar : PubMed/NCBI

87 

López-López J, Castellanos-Cosano L, Estrugo-Devesa A, Gómez-Vaquero C, Velasco-Ortega E and Segura-Egea JJ: Radiolucent periapical lesions and bone mineral density in post-menopausal women. Gerodontology. 32:195–201. 2015. View Article : Google Scholar

88 

Tounta TS: Diagnosis of osteoporosis in dental patients. J Frailty Sarcopenia Falls. 2:21–27. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Payne JB, Reinhardt RA, Nummikoski PV and Patil KD: Longitudinal alveolar bone loss in postmenopausal osteoporotic/osteopenic women. Osteoporos Int. 10:34–40. 1999. View Article : Google Scholar : PubMed/NCBI

90 

Brennan RM, Genco RJ, Hovey KM, Trevisan M and Wactawski-Wende J: Clinical attachment loss, systemic bone density, and subgingival calculus in postmenopausal women. J Periodontol. 78:2104–2111. 2007. View Article : Google Scholar : PubMed/NCBI

91 

Dervis E: Oral implications of osteoporosis. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 100:349–356. 2005. View Article : Google Scholar : PubMed/NCBI

92 

Katz J and Rotstein I: Prevalence of periapical lesions in patients with osteoporosis. J Endod. 47:234–238. 2021. View Article : Google Scholar

93 

Wayama MT, Yoshimura H, Ohba S, Yoshida H, Matsuda S, Kobayashi J, Kobayashi M, Gomes Filho JE and Sano K: Diminished progression of periapical lesions with zoledronic acid in ovariectomized rats. J Endod. 41:2002–2007. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Ikeda M, Karakawa A, Takizawa H, Azetsu Y, Sakai N, Chatani M, Suzuki N and Takami M: Effects of anti-receptor activator of nuclear factor kappa B ligand antibody and zoledronic acid on periapical lesion development in mice. J Endod. 48:632–640. 2022. View Article : Google Scholar : PubMed/NCBI

95 

Xiong H, Peng B, Wei L, Zhang X and Wang L: Effect of an estrogen-deficient state and alendronate therapy on bone loss resulting from experimental periapical lesions in rats. J Endod. 33:1304–1308. 2007. View Article : Google Scholar : PubMed/NCBI

96 

Silva RAB, Sousa-Pereira AP, Lucisano MP, Romualdo PC, Paula-Silva FWG, Consolaro A, Silva LAB and Nelson-Filho P: Alendronate inhibits osteocyte apoptosis and inflammation via IL-6, inhibiting bone resorption in periapical lesions of ovariectomized rats. Int Endod J. 53:84–96. 2020. View Article : Google Scholar

97 

Guan X, Guan Y, Shi C, Zhu X, He Y, Wei Z, Yang J and Hou T: Estrogen deficiency aggravates apical periodontitis by regulating NLRP3/caspase-1/IL-1β axis. Am J Transl Res. 12:660–671. 2020.

98 

Zhang X, Peng B, Fan M, Bian Z and Chen Z: The effect of estrogen deficiency on receptor activator of nuclear factor kappa B ligand and osteoprotegerin synthesis in periapical lesions induced in rats. J Endod. 33:1053–1056. 2007. View Article : Google Scholar : PubMed/NCBI

99 

Romualdo PC, Lucisano MP, Paula-Silva FWG, Leoni GB, Sousa-Neto MD, Silva RAB, Silva LAB and Nelson-Filho P: Ovariectomy exacerbates apical periodontitis in rats with an increase in expression of proinflammatory cytokines and matrix metalloproteinases. J Endod. 44:780–785. 2018. View Article : Google Scholar : PubMed/NCBI

100 

Brasil SC, Santos RMM, Fernandes A, Alves FR, Pires FR, Siqueira JF Jr and Armada L: Influence of oestrogen deficiency on the development of apical periodontitis. Int Endod J. 50:161–166. 2017. View Article : Google Scholar

101 

Lucisano MP, da Silva RAB, de Sousa Pereira AP, Romualdo PC, Feres M, de Queiroz AM, Nelson-Filho P and da Silva LAB: Alteration of the oral microbiota may be a responsible factor, along with estrogen deficiency, by the development of larger periapical lesions. Clin Oral Investig. 25:3651–3662. 2021. View Article : Google Scholar

102 

Gomes-Filho JE, Wayama MT, Dornelles RCM, Ervolino E, Yamanari GH, Lodi CS, Sivieri-Araújo G, Dezan-Júnior E and Cintra LT: Raloxifene modulates regulators of osteoclastogenesis and angiogenesis in an oestrogen deficiency periapical lesion model. Int Endod J. 48:1059–1068. 2015. View Article : Google Scholar

103 

Qian H, Guan X and Bian Z: FSH aggravates bone loss in ovariectomised rats with experimental periapical periodontitis. Mol Med Rep. 14:2997–3006. 2016. View Article : Google Scholar : PubMed/NCBI

104 

Liu S, Cheng Y, Xu W and Bian Z: Protective effects of follicle-stimulating hormone inhibitor on alveolar bone loss resulting from experimental periapical lesions in ovariectomized rats. J Endod. 36:658–663. 2010. View Article : Google Scholar : PubMed/NCBI

105 

Braz-Silva PH, Bergamini ML, Mardegan AP, De Rosa CS, Hasseus B and Jonasson P: Inflammatory profile of chronic apical periodontitis: A literature review. Acta Odontol Scand. 77:173–180. 2019. View Article : Google Scholar

106 

Matsuo T, Ebisu S, Shimabukuro Y, Ohtake T and Okada H: Quantitative analysis of immunocompetent cells in human periapical lesions: Correlations with clinical findings of the involved teeth. J Endod. 18:497–500. 1992. View Article : Google Scholar : PubMed/NCBI

107 

Guerrero-Gironés J, Ros-Valverde A, Pecci-Lloret MP, Rodríguez-Lozano FJ and Pecci-Lloret MR: Association between pulpal-periapical pathology and autoimmune diseases: A systematic review. J Clin Med. 10:48862021. View Article : Google Scholar : PubMed/NCBI

108 

Karataş E, Kul A and Tepecik E: Association of ankylosing spondylitis with radiographically and clinically diagnosed apical periodontitis: A cross-sectional study. Dent Med Probl. 57:171–175. 2020. View Article : Google Scholar

109 

Barta Z: Apical periodontitis in patients with inflammatory bowel disease: A puppet master? Inflamm Bowel Dis. 26:280–282. 2020. View Article : Google Scholar

110 

Nakamura K, Yamasaki M, Nishigaki N, Iwama A, Imaizumi I, Nakamura H and Kameyama Y: Effect of methotrexate-induced neutropenia on pulpal inflammation in rats. J Endod. 28:287–290. 2002. View Article : Google Scholar : PubMed/NCBI

111 

Yoshinari N, Kameyama Y, Aoyama Y, Nishiyama H and Noguchi T: Effect of long-term methotrexate-induced neutropenia on experimental periodontal lesion in rats. J Periodontal Res. 29:393–400. 1994. View Article : Google Scholar : PubMed/NCBI

112 

Yamasaki M, Kumazawa M, Kohsaka T and Nakamura H: Effect of methotrexate-induced neutropenia on rat periapical lesion. Oral Surg Oral Med Oral Pathol. 77:655–661. 1994. View Article : Google Scholar : PubMed/NCBI

113 

Waterman PA Jr, Torabinejad M, McMillan PJ and Kettering JD: Development of periradicular lesions in immunosuppressed rats. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 85:720–725. 1998. View Article : Google Scholar : PubMed/NCBI

114 

Teixeira FB, Gomes BP, Ferraz CC, Souza-Filho FJ and Zaia AA: Radiographic analysis of the development of periapical lesions in normal rats, sialoadenectomized rats and sialoadenectomized-immunosuppressed rats. Endod Dent Traumatol. 16:154–157. 2000. View Article : Google Scholar

115 

Wasserman A: Rheumatoid arthritis: Common questions about diagnosis and management. Am Fam Physician. 97:455–462. 2018.PubMed/NCBI

116 

Karataş E, Kul A and Tepecik E: Association between rheumatoid arthritis and apical periodontitis: A cross-sectional study. Eur Endod J. 5:155–158. 2020.

117 

Rotstein I and Katz J: Prevalence of periapical abscesses in patients with rheumatoid arthritis. A cross sectional study. Am J Dent. 34:211–214. 2021.PubMed/NCBI

118 

Oh WM, Hwang IN, Son HH and Hwang YC: Rapid periapical bone destruction during endodontic treatment of a patient with rheumatoid arthritis. J Endod. 34:1261–1263. 2008. View Article : Google Scholar : PubMed/NCBI

119 

Jalali P, Glickman GN, Schneiderman ED and Schweitzer JL: Prevalence of periapical rarefying osteitis in patients with rheumatoid arthritis. J Endod. 43:1093–1096. 2017. View Article : Google Scholar : PubMed/NCBI

120 

Cotti E, Schirru E, Acquas E and Usai P: An overview on biologic medications and their possible role in apical periodontitis. J Endod. 40:1902–1911. 2014. View Article : Google Scholar : PubMed/NCBI

121 

Malmström M: Immunoglobulin clases IgG, IgM, IgA and complement component C3 in dental periapical lesions of patients with rheumatoid disease. Scand J Rheumatol. 4:57–64. 1975. View Article : Google Scholar : PubMed/NCBI

122 

Malmström M and Calonius PE: Teeth loss and the inflammation of teeth-supporting tissues in rheumatoid disease. Scand J Rheumatol. 4:49–55. 1975. View Article : Google Scholar : PubMed/NCBI

123 

Malmström M and Jokinen EJ: Free rheumatoid factor in dental periapical lesions and gingivae of patients with rheumatoid disease. Scand J Rheumatol. 4:121–124. 1975. View Article : Google Scholar : PubMed/NCBI

124 

Malmström M and Natvig JB: IgG rheumatoid factor in dental periapical lesions of patients with rheumatoid disease. Scand J Rheumatol. 4:177–185. 1975. View Article : Google Scholar : PubMed/NCBI

125 

Malmström M and Husby G: Occurrence of amyloid in the teeth-supporting tissues of patients with rheumatoid diseases. An immunohistochemical study. Scand J Rheumatol. 4:186–192. 1975. View Article : Google Scholar : PubMed/NCBI

126 

Ishii K, Hatori K, Takeichi O, Makino K, Himi K, Komiya H and Ogiso B: Expression of the Forkhead box transcription factor Foxo3a in human periapical granulomas. J Oral Sci. 60:479–483. 2018. View Article : Google Scholar : PubMed/NCBI

127 

Vergani D and Mieli-Vergani G: Autoimmune manifestations in viral hepatitis. Semin Immunopathol. 35:73–85. 2013. View Article : Google Scholar

128 

Bjørklund G, Pivin M, Hangan T, Yurkovskaya O and Pivina L: Autoimmune polyendocrine syndrome type 1: Clinical manifestations, pathogenetic features, and management approach. Autoimmun Rev. 21:1031352022. View Article : Google Scholar : PubMed/NCBI

129 

Joyce E, Glasner P, Ranganathan S and Swiatecka-Urban A: Tubulointerstitial nephritis: Diagnosis, treatment, and monitoring. Pediatr Nephrol. 32:577–587. 2017. View Article : Google Scholar

130 

Cohen G: Immune dysfunction in Uremia 2020. Toxins (Basel). 12. pp. 4392020, View Article : Google Scholar

131 

Grønkjær LL and Vilstrup H: Oral health in patients with liver cirrhosis. Eur J Gastroenterol Hepatol. 27:834–839. 2015. View Article : Google Scholar : PubMed/NCBI

132 

Coates EA, Brennan D, Logan RM, Goss AN, Scopacasa B, Spencer AJ and Gorkic E: Hepatitis C infection and associated oral health problems. Aust Dent J. 45:108–114. 2000. View Article : Google Scholar : PubMed/NCBI

133 

Lins L, Bittencourt PL, Evangelista MA, Lins R, Codes L, Cavalcanti AR, Paraná R and Bastos J: Oral health profile of cirrhotic patients awaiting liver transplantation in the Brazilian Northeast. Transplant Proc. 43:1319–1321. 2011. View Article : Google Scholar : PubMed/NCBI

134 

Barbero P, Garzino Demo MG, Milanesio M and Ottobrelli A: The dental assessment of the patient waiting for a liver transplant. Minerva Stomatol. 45:431–439. 1996.In Italian. PubMed/NCBI

135 

Guggenheimer J, Eghtesad B and Stock DJ: Dental management of the (solid) organ transplant patient. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 95:383–389. 2003. View Article : Google Scholar : PubMed/NCBI

136 

Helenius-Hietala J, Meurman JH, Höckerstedt K, Lindqvist C and Isoniemi H: Effect of the aetiology and severity of liver disease on oral health and dental treatment prior to transplantation. Transpl Int. 25:158–165. 2012. View Article : Google Scholar

137 

Castellanos-Cosano L, Machuca-Portillo G, Segura-Sampedro JJ, Torres-Lagares D, López-López J, Velasco-Ortega E and Segura-Egea JJ: Prevalence of apical periodontitis and frequency of root canal treatments in liver transplant candidates. Med Oral Patol Oral Cir Bucal. 18:e773–e779. 2013. View Article : Google Scholar : PubMed/NCBI

138 

Grønkjær LL, Holmstrup P, Schou S, Schwartz K, Kongstad J, Jepsen P and Vilstrup H: Presence and consequence of tooth periapical radiolucency in patients with cirrhosis. Hepat Med. 8:97–103. 2016. View Article : Google Scholar : PubMed/NCBI

139 

Cantiga-Silva C, Estrela C, Segura-Egea JJ, Azevedo JP, de Oliveira PHC, Cardoso CBM, Pinheiro TN, Ervolino E, Sivieri-Araújo G and Cintra LTA: Inflammatory profile of apical periodontitis associated with liver fibrosis in rats: Histological and immunohistochemical analysis. Int Endod J. 54:1353–1361. 2021. View Article : Google Scholar : PubMed/NCBI

140 

Echavarría-García AC, Pozos-Guillén A, Tejeda-Nava F, Flores Arriaga JC and Garrocho-Rangel A: Oral management of children with Henoch-Schönlein purpura and associated glomerulonephritis: A scoping review. Eur J Paediatr Dent. 19:134–138. 2018.

141 

Inoue CN, Nagasaka T, Matsutani S, Ishidoya M, Homma R and Chiba Y: Efficacy of early dental and ENT therapy in preventing nephropathy in pediatric Henoch-Schönlein purpura. Clin Rheumatol. 27:1489–1496. 2008. View Article : Google Scholar : PubMed/NCBI

142 

Al-Wahadni A and Al-Omari MA: Dental diseases in a Jordanian population on renal dialysis. Quintessence Int. 34:343–347. 2003.PubMed/NCBI

143 

Souza CM, Braosi APR, Luczyszyn SM, Casagrande RW, Pecoits-Filho R, Riella MC, Ignácio SA and Trevilatto PC: Oral health in Brazilian patients with chronic renal disease. Rev Med Chil. 136:741–746. 2008. View Article : Google Scholar : PubMed/NCBI

144 

Bayraktar G, Kurtulus I, Duraduryan A, Cintan S, Kazancioglu R, Yildiz A, Bural C, Bozfakioglu S, Besler M, Trablus S and Issever H: Dental and periodontal findings in hemodialysis patients. Oral Dis. 13:393–397. 2007. View Article : Google Scholar : PubMed/NCBI

145 

Sobrado Marinho JS, Tomás Carmona I, Loureiro A, Limeres Posse J, García Caballero L and Diz Dios P: Oral health status in patients with moderate-severe and terminal renal failure. Med Oral Patol Oral Cir Bucal. 12:E305–E310. 2007.PubMed/NCBI

146 

Buhlin K, Bárány P, Heimbürger O, Stenvinkel P and Gustafsson A: Oral health and pro-inflammatory status in end-stage renal disease patients. Oral Health Prev Dent. 5:235–244. 2007.PubMed/NCBI

147 

Khalighinejad N, Aminoshariae A, Kulild JC, Sahly K and Mickel A: Association of end-stage renal disease with radiographically and clinically diagnosed apical periodontitis: A hospital-based study. J Endod. 43:1438–1441. 2017. View Article : Google Scholar : PubMed/NCBI

148 

Caly WR and Strauss E: A prospective study of bacterial infections in patients with cirrhosis. J Hepatol. 18:353–358. 1993. View Article : Google Scholar : PubMed/NCBI

149 

Nair PN: Apical periodontitis: A dynamic encounter between root canal infection and host response. Periodontol. 2000(13): 121–148. 1997. View Article : Google Scholar

150 

Mizutani K, Mikami R, Gohda T, Gotoh H, Aoyama N, Matsuura T, Kido D, Takeda K, Izumi Y, Sasaki Y and Iwata T: Poor oral hygiene and dental caries predict high mortality rate in hemodialysis: A 3-year cohort study. Sci Rep. 10:218722020. View Article : Google Scholar : PubMed/NCBI

151 

Olsen I and Yamazaki K: Can oral bacteria affect the microbiome of the gut? J Oral Microbiol. 11:15864222019. View Article : Google Scholar : PubMed/NCBI

152 

Ogura Y, Suzuki S, Shirakawa T, Masuda M, Nakamura H, Iijima K and Yoshikawa N: Haemophilus parainfluenzae antigen and antibody in children with IgA nephropathy and Henoch-Schönlein nephritis. Am J Kidney Dis. 36:47–52. 2000. View Article : Google Scholar : PubMed/NCBI

153 

Kojima A, Nakano K, Wada K, Takahashi H, Katayama K, Yoneda M, Higurashi T, Nomura R, Hokamura K, Muranaka Y, et al: Infection of specific strains of Streptococcus mutans, oral bacteria, confers a risk of ulcerative colitis. Sci Rep. 2:3322012. View Article : Google Scholar : PubMed/NCBI

154 

Tahmassebi JF and Paterson SA: Development of acute Henoch-Schönlein purpura subsequent to endodontic treatment. Int J Paediatr Dent. 17:217–222. 2007. View Article : Google Scholar : PubMed/NCBI

155 

Papageorgiou SN, Hagner M, Nogueira AVB, Franke A, Jäger A and Deschner J: Inflammatory bowel disease and oral health: Systematic review and a meta-analysis. J Clin Periodontol. 44:382–393. 2017. View Article : Google Scholar : PubMed/NCBI

156 

Baumgart DC and Sandborn WJ: Crohn's disease. Lancet. 380:1590–1605. 2012. View Article : Google Scholar : PubMed/NCBI

157 

Kalmar JR: Crohn's disease: Orofacial considerations and disease pathogenesis. Periodontol. 2000(6): 101–115. 1994. View Article : Google Scholar

158 

She YY, Kong XB, Ge YP, Liu ZY, Chen JY, Jiang JW, Jiang HB and Fang SL: Periodontitis and inflammatory bowel disease: A meta-analysis. BMC Oral Health. 20:672020. View Article : Google Scholar : PubMed/NCBI

159 

Zhang Y, Qiao D, Chen R, Zhu F, Gong J and Yan F: The association between periodontitis and inflammatory bowel disease: A systematic review and meta-analysis. Biomed Res Int. 2021:66924202021.PubMed/NCBI

160 

Tan CXW, Brand HS, Kalender B, De Boer NKH, Forouzanfar T and de Visscher JGAM: Dental and periodontal disease in patients with inflammatory bowel disease. Clin Oral Investig. 25:5273–5280. 2021. View Article : Google Scholar : PubMed/NCBI

161 

Piras V, Usai P, Mezzena S, Susnik M, Ideo F, Schirru E and Cotti E: Prevalence of apical periodontitis in patients with inflammatory bowel diseases: A retrospective clinical study. J Endod. 43:389–394. 2017. View Article : Google Scholar : PubMed/NCBI

162 

Poyato-Borrego M, Segura-Egea JJ, Martín-González J, Jiménez-Sánchez MC, Cabanillas-Balsera D, Areal-Quecuty V and Segura-Sampedro JJ: Prevalence of endodontic infection in patients with Crohn’s disease and ulcerative colitis. Med Oral Patol Oral Cir Bucal. 26:e208–e215. 2021. View Article : Google Scholar

163 

Poyato-Borrego M, Segura-Sampedro JJ, Martín-González J, Torres-Domínguez Y, Velasco-Ortega E and Segura-Egea JJ: High prevalence of apical periodontitis in patients with inflammatory bowel disease: An age- and gender-matched case-control study. Inflamm Bowel Dis. 26:273–279. 2020. View Article : Google Scholar

164 

Johannsen A, Fored MC, Håkansson J, Ekbom A and Gustafsson A: Consumption of dental treatment in patients with inflammatory bowel disease, a register study. PLoS One. 10:e01340012015. View Article : Google Scholar : PubMed/NCBI

165 

Segura-Sampedro JJ, Jiménez-Giménez C, Jane-Salas E, Cabanillas-Balsera D, Martín-González J, Segura-Egea JJ and López-López J: Periapical and endodontic status of patients with inflammatory bowel disease: Age- and sex-matched case-control study. Int Endod J. 55:748–757. 2022. View Article : Google Scholar : PubMed/NCBI

166 

Lu Y, Li X, Liu S, Zhang Y and Zhang D: Toll-like receptors and inflammatory bowel disease. Front Immunol. 9:722018. View Article : Google Scholar : PubMed/NCBI

167 

Ng YL, Mann V, Rahbaran S, Lewsey J and Gulabivala K: Outcome of primary root canal treatment: systematic review of the literature-part 2. Influence of clinical factors. Int Endod J. 41:6–31. 2008. View Article : Google Scholar

168 

Bamias G, Martin C, Mishina M, Ross WG, Rivera-Nieves J, Marini M and Cominelli F: Proinflammatory effects of TH2 cytokines in a murine model of chronic small intestinal inflammation. Gastroenterology. 128:654–666. 2005. View Article : Google Scholar : PubMed/NCBI

169 

Fukada SY, Silva TA, Garlet GP, Rosa AL, da Silva JS and Cunha FQ: Factors involved in the T helper type 1 and type 2 cell commitment and osteoclast regulation in inflammatory apical diseases. Oral Microbiol Immunol. 24:25–31. 2009. View Article : Google Scholar : PubMed/NCBI

170 

Stashenko P, Wang CY, Tani-Ishii N and Yu SM: Pathogenesis of induced rat periapical lesions. Oral Surg Oral Med Oral Pathol. 78:494–502. 1994. View Article : Google Scholar : PubMed/NCBI

171 

Wei L, Xu M and Xiong H: An update of knowledge on the regulatory role of Treg cells in apical periodontitis. Oral Dis. 27:1356–1365. 2021. View Article : Google Scholar

172 

Van Dyke TE, Dowell VR Jr, Offenbacher S, Snyder W and Hersh T: Potential role of microorganisms isolated from periodontal lesions in the pathogenesis of inflammatory bowel disease. Infect Immun. 53:671–677. 1986. View Article : Google Scholar : PubMed/NCBI

173 

Chi AC, Neville BW, Krayer JW and Gonsalves WC: Oral manifestations of systemic disease. Am Fam Physician. 82:1381–1388. 2010.PubMed/NCBI

174 

Rodriguez Herrero E, Boon N, Pauwels M, Bernaerts K, Slomka V, Quirynen M and Teughels W: Necrotrophic growth of periodontopathogens is a novel virulence factor in oral biofilms. Sci Rep. 7:11072017. View Article : Google Scholar : PubMed/NCBI

175 

Read E, Curtis MA and Neves JF: The role of oral bacteria in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 18:731–742. 2021. View Article : Google Scholar : PubMed/NCBI

176 

Tavares CO, Rost FL, Silva RBM, Dagnino AP, Adami B, Schirmer H, de Figueiredo JAP, Souto AA, Maito FDM and Campos MM: Cross talk between apical periodontitis and metabolic disorders: Experimental evidence on the role of intestinal adipokines and akkermansia muciniphila. J Endod. 45:174–180. 2019. View Article : Google Scholar : PubMed/NCBI

177 

Carding S, Verbeke K, Vipond DT, Corfe BM and Owen LJ: Dysbiosis of the gut microbiota in disease. Microb Ecol Health Dis. 26:261912015.PubMed/NCBI

178 

Gan G, Lu B, Zhang R, Luo Y, Chen S, Lei H, Li Y, Cai Z and Huang X: Chronic apical periodontitis exacerbates atherosclerosis in apolipoprotein E-deficient mice and leads to changes in the diversity of gut microbiota. Int Endod J. 55:152–163. 2022. View Article : Google Scholar

179 

Ideo F, Niazi S, Mezzena S, Mannocci F and Cotti E: Prevalence of apical periodontitis in patients with autoimmune diseases under immunomodulators: A retrospective cohort study. J Endod. 48:722–729. 2022. View Article : Google Scholar : PubMed/NCBI

180 

Ardizzone S and Bianchi Porro G: Biologic therapy for inflammatory bowel disease. Drugs. 65:2253–2286. 2005. View Article : Google Scholar : PubMed/NCBI

181 

Cotti E, Mezzena S, Schirru E, Ottonello O, Mura M, Ideo F, Susnik M and Usai P: Healing of apical periodontitis in patients with inflammatory bowel diseases and under anti-tumor necrosis factor alpha therapy. J Endod. 44:1777–1782. 2018. View Article : Google Scholar : PubMed/NCBI

182 

Ritchlin CT, Haas-Smith SA, Li P, Hicks DG and Schwarz EM: Mechanisms of TNF-alpha- and RANKL-mediated osteoclastogenesis and bone resorption in psoriatic arthritis. J Clin Invest. 111:821–831. 2003. View Article : Google Scholar : PubMed/NCBI

183 

Joseph P, Leong D, McKee M, Anand SS, Schwalm JD, Teo K, Mente A and Yusuf S: Reducing the global burden of cardiovascular disease, part 1: The epidemiology and risk factors. Circ Res. 121:677–694. 2017. View Article : Google Scholar : PubMed/NCBI

184 

Andersson C and Vasan RS: Epidemiology of cardiovascular disease in young individuals. Nat Rev Cardiol. 15:230–240. 2018. View Article : Google Scholar

185 

Prabhakaran D, Jeemon P and Roy A: Cardiovascular diseases in india: current epidemiology and future directions. Circulation. 133:1605–1620. 2016. View Article : Google Scholar : PubMed/NCBI

186 

Cotti E, Dessì C, Piras A and Mercuro G: Can a chronic dental infection be considered a cause of cardiovascular disease? A review of the literature. Int J Cardiol. 148:4–10. 2011. View Article : Google Scholar

187 

Berlin-Broner Y, Febbraio M and Levin L: Association between apical periodontitis and cardiovascular diseases: A systematic review of the literature. Int Endod J. 50:847–859. 2017. View Article : Google Scholar

188 

González Navarro B, Pintó Sala X and Jané Salas E: Relationship between cardiovascular disease and dental pathology. Systematic review. Med Clin (Barc). 149:211–216. 2017.In English, Spanish. View Article : Google Scholar : PubMed/NCBI

189 

Koletsi D, Iliadi A, Tzanetakis GN, Vavuranakis M and Eliades T: Cardiovascular disease and chronic endodontic infection. Is there an association? A systematic review and meta-analysis. Int J Environ Res Public Health. 18:91112021. View Article : Google Scholar : PubMed/NCBI

190 

Garg P and Chaman C: Apical periodontitis-is it accountable for cardiovascular diseases? J Clin Diagn Res. 10:ZE08–ZE12. 2016.PubMed/NCBI

191 

Jakovljevic A, Duncan HF, Nagendrababu V, Jacimovic J, Milasin J and Dummer PMH: Association between cardiovascular diseases and apical periodontitis: An umbrella review. Int Endod J. 53:1374–1386. 2020. View Article : Google Scholar : PubMed/NCBI

192 

Aloutaibi YA, Alkarim AS, Qumri EM, Almansour LA and Alghamdi FT: Chronic endodontic infections and cardiovascular diseases: Does the evidence support an independent association? Cureus. 13:e198642021.

193 

Jiménez-Sánchez MC, Cabanillas-Balsera D, Areal-Quecuty V, Velasco-Ortega E, Martín-González J and Segura-Egea JJ: Cardiovascular diseases and apical periodontitis: Association not always implies causality. Med Oral Patol Oral Cir Bucal. 25:e652–e659. 2020. View Article : Google Scholar : PubMed/NCBI

194 

Noites R, Teixeira M, Cavero-Redondo I, Alvarez-Bueno C and Ribeiro F: Apical periodontitis and cardiovascular disease in adults: A systematic review with meta-analysis. Rev Cardiovasc Med. 23:1002022. View Article : Google Scholar : PubMed/NCBI

195 

Leão TSS, Tomasi GH, Conzatti LP, Marrone LCP, Reynolds MA and Gomes MS: Oral inflammatory burden and carotid atherosclerosis among stroke patients. J Endod. 48:597–605. 2022. View Article : Google Scholar : PubMed/NCBI

196 

Garrido M, Cárdenas AM, Astorga J, Quinlan F, Valdés M, Chaparro A, Carvajal P, Pussinen P, Huamán-Chipana P, Jalil JE and Hernández M: Elevated systemic inflammatory burden and cardiovascular risk in young adults with endodontic apical lesions. J Endod. 45:111–115. 2019. View Article : Google Scholar : PubMed/NCBI

197 

Conti LC, Segura-Egea JJ, Cardoso CBM, Benetti F, Azuma MM, Oliveira PHC, Bomfim SRM and Cintra LTA: Relationship between apical periodontitis and atherosclerosis in rats: Lipid profile and histological study. Int Endod J. 53:1387–1397. 2020. View Article : Google Scholar : PubMed/NCBI

198 

Caplan DJ: Chronic apical periodontitis is more common in subjects with coronary artery disease. J Evid Based Dent Pract. 14:149–150. 2014. View Article : Google Scholar : PubMed/NCBI

199 

Caplan DJ, Chasen JB, Krall EA, Cai J, Kang S, Garcia RI, Offenbacher S and Beck JD: Lesions of endodontic origin and risk of coronary heart disease. J Dent Res. 85:9962006. View Article : Google Scholar : PubMed/NCBI

200 

Liljestrand JM, Mäntylä P, Paju S, Buhlin K, Kopra KA, Persson GR, Hernandez M, Nieminen MS, Sinisalo J, Tjäderhane L and Pussinen PJ: Association of endodontic lesions with coronary artery disease. J Dent Res. 95:1358–1365. 2016. View Article : Google Scholar : PubMed/NCBI

201 

Costa THR, de Figueiredo Neto JA, de Oliveira AE, Lopes e Maia Mde F and de Almeida AL: Association between chronic apical periodontitis and coronary artery disease. J Endod. 40:164–167. 2014. View Article : Google Scholar : PubMed/NCBI

202 

Paloma de Oliveira B, Câmara AC and Aguiar CM: Prevalence of asymptomatic apical periodontitis and its association with coronary artery disease in a Brazilian subpopulation. Acta Stomatol Croat. 51:106–112. 2017. View Article : Google Scholar : PubMed/NCBI

203 

Milojevic Samanovic A, Jakovljevic V, Vasovic M, Mitrovic S, Rankovic M, Mihajlovic K, Bolevich S and Zivkovic V: Cardiac, biochemical and histopathological analysis reveals impaired heart function in hypertensive rats with apical periodontitis. Int Endod J. 54:1581–1596. 2021. View Article : Google Scholar : PubMed/NCBI

204 

Katz J and Rotstein I: Prevalence of periapical abscesses in patients with hypertension: A cross-sectional study of a large hospital population. J Endod. 47:1070–1074. 2021. View Article : Google Scholar : PubMed/NCBI

205 

Petersen J, Glaßl EM, Nasseri P, Crismani A, Luger AK, Schoenherr E, Bertl K and Glodny B: The association of chronic apical periodontitis and endodontic therapy with atherosclerosis. Clin Oral Investig. 18:1813–1823. 2014. View Article : Google Scholar :

206 

Vidal F, Fontes TV, Marques TVF and Gonçalves LS: Association between apical periodontitis lesions and plasmatic levels of C-reactive protein, interleukin 6 and fibrinogen in hypertensive patients. Int Endod J. 49:1107–1115. 2016. View Article : Google Scholar

207 

Rashmi N, Galhotra V, Goel P, Rajguru JP, Jha SK and Kulkarni K: Assessment of C-reactive proteins, cytokines, and plasma protein levels in hypertensive patients with apical periodontitis. J Contemp Dent Pract. 18:516–521. 2017. View Article : Google Scholar : PubMed/NCBI

208 

Messing M, Souza LCD, Cavalla F, Kookal KK, Rizzo G, Walji M, Silva R and Letra A: Investigating potential correlations between endodontic pathology and cardiovascular diseases using epidemiological and genetic approaches. J Endod. 45:104–110. 2019. View Article : Google Scholar : PubMed/NCBI

209 

Sebring D, Buhlin K, Norhammar A, Rydén L, Jonasson P; EndoReCo; Lund H and Kvist T: Endodontic inflammatory disease: A risk indicator for a first myocardial infarction. Int Endod J. 55:6–17. 2022. View Article : Google Scholar

210 

Bordagaray MJ, Fernández A, Garrido M, Astorga J, Hoare A and Hernández M: Systemic and extraradicular bacterial translocation in apical periodontitis. Front Cell Infect Microbiol. 11:6499252021. View Article : Google Scholar : PubMed/NCBI

211 

Jiménez C, Garrido M, Pussinen P, Bordagaray MJ, Fernández A, Vega C, Chaparro A, Hoare A and Hernández M: Systemic burden and cardiovascular risk to Porphyromonas species in apical periodontitis. Clin Oral Investig. 26:993–1001. 2022. View Article : Google Scholar

212 

Kazanci E, Oguz KK, Gurgey A and Topçu M: Streptococcus oralis as a risk factor for middle cerebral artery thrombosis. J Child Neurol. 20:611–613. 2005. View Article : Google Scholar : PubMed/NCBI

213 

Theofilis P, Sagris M, Oikonomou E, Antonopoulos AS, Siasos G, Tsioufis C and Tousoulis D: Inflammatory mechanisms contributing to endothelial dysfunction. Biomedicines. 9:7812021. View Article : Google Scholar : PubMed/NCBI

214 

Cotti E, Dessì C, Piras A, Flore G, Deidda M, Madeddu C, Zedda A, Longu G and Mercuro G: Association of endodontic infection with detection of an initial lesion to the cardiovascular system. J Endod. 37:1624–1629. 2011. View Article : Google Scholar : PubMed/NCBI

215 

Bergandi L, Giuggia B, Alovisi M, Comba A, Silvagno F, Maule M, Aldieri E, Scotti N, Scacciatella P, Conrotto F, et al: Endothelial dysfunction marker variation in young adults with chronic apical periodontitis before and after endodontic treatment. J Endod. 45:500–506. 2019. View Article : Google Scholar : PubMed/NCBI

216 

Chauhan N, Mittal S, Tewari S, Sen J and Laller K: Association of apical periodontitis with cardiovascular disease via noninvasive assessment of endothelial function and subclinical atherosclerosis. J Endod. 45:681–690. 2019. View Article : Google Scholar : PubMed/NCBI

217 

Hernández-Ríos P, Pussinen PJ, Vernal R and Hernández M: Oxidative stress in the local and systemic events of apical periodontitis. Front Physiol. 8:8692017. View Article : Google Scholar : PubMed/NCBI

218 

Chapple IL: Reactive oxygen species and antioxidants in inflammatory diseases. J Clin Periodontol. 24:287–296. 1997. View Article : Google Scholar : PubMed/NCBI

219 

Tucker MA, Fox BM, Seigler N, Rodriguez-Miguelez P, Looney J, Thomas J, McKie KT, Forseen C, Davison GW and Harris RA: Endothelial dysfunction in cystic fibrosis: Role of oxidative stress. Oxid Med Cell Longev. 2019:16296382019. View Article : Google Scholar : PubMed/NCBI

220 

Brilhante Wolle CF, de Aguiar Zollmann L, Etges A, Vitalis GS, Leite CE and Campos MM: Effects of the antioxidant agent tempol on periapical lesions in rats with doxorubicin-induced cardiomyopathy. J Endod. 38:191–195. 2012. View Article : Google Scholar : PubMed/NCBI

221 

Vailhé B, Dietl J, Kapp M, Toth B and Arck P: Increased blood vessel density in decidua parietalis is associated with spontaneous human first trimester abortion. Hum Reprod. 14:1628–1634. 1999. View Article : Google Scholar : PubMed/NCBI

222 

Zygmunt M, Herr F, Münstedt K, Lang U and Liang OD: Angiogenesis and vasculogenesis in pregnancy. Eur J Obstet Gynecol Reprod Biol. 110(Suppl 1): pp. S10–S18. 2003, View Article : Google Scholar

223 

Plaisier M: Decidualisation and angiogenesis. Best Pract Res Clin Obstet Gynaecol. 25:259–271. 2011. View Article : Google Scholar

224 

Bobetsis YA, Graziani F, Gürsoy M and Madianos PN: Periodontal disease and adverse pregnancy outcomes. Periodontol. 2000(83): 154–174. 2020. View Article : Google Scholar

225 

Harjunmaa U, Järnstedt J, Alho L, Dewey KG, Cheung YB, Deitchler M, Ashorn U, Maleta K, Klein NJ and Ashorn P: Association between maternal dental periapical infections and pregnancy outcomes: results from a cross-sectional study in Malawi. Trop Med Int Health. 20:1549–1558. 2015. View Article : Google Scholar : PubMed/NCBI

226 

Leal ASM, de Oliveira AEF, Brito LMO, Lopes FF, Rodrigues VP, Lima KF and de Araújo Martins IC: Association between chronic apical periodontitis and low-birth-weight preterm births. J Endod. 41:353–357. 2015. View Article : Google Scholar : PubMed/NCBI

227 

Khalighinejad N, Aminoshariae A, Kulild JC and Mickel A: Apical periodontitis, a predictor variable for preeclampsia: A case-control study. J Endod. 43:1611–1614. 2017. View Article : Google Scholar : PubMed/NCBI

228 

Bain JL, Lester SR, Henry WD, Naftel JP and Johnson RB: Effects of induced periapical abscesses on rat pregnancy outcomes. Arch Oral Biol. 54:162–171. 2009. View Article : Google Scholar

229 

Bain JL, Lester SR, Henry WD, Pongetti JL, Blackman ME and Johnson RB: Association between maternal periapical lesions and brain inflammation in rat pups. Arch Oral Biol. 58:266–271. 2013. View Article : Google Scholar

230 

Kc K, Shakya S and Zhang H: Gestational diabetes mellitus and macrosomia: A literature review. Ann Nutr Metab. 66(Suppl 2): S14–S20. 2015. View Article : Google Scholar

231 

Shah H, Nisar N, Hassan A and Butt S: Association between maternal chronic apical periodontitis (CAP) and low birth weight preterm birth (LBWPT). J Pak Med Assoc. 72:436–439. 2022.PubMed/NCBI

232 

Orstavik D, Kerekes K and Eriksen HM: The periapical index: A scoring system for radiographic assessment of apical periodontitis. Endod Dent Traumatol. 2:20–34. 1986. View Article : Google Scholar : PubMed/NCBI

233 

Ao M, Miyauchi M, Furusho H, Inubushi T, Kitagawa M, Nagasaki A, Sakamoto S, Kozai K and Takata T: Dental Infection of Porphyromonas gingivalis induces preterm birth in mice. PLoS One. 10:e01372492015. View Article : Google Scholar : PubMed/NCBI

234 

Doyle RM, Alber DG, Jones HE, Harris K, Fitzgerald F, Peebles D and Klein N: Term and preterm labour are associated with distinct microbial community structures in placental membranes which are independent of mode of delivery. Placenta. 35:1099–1101. 2014. View Article : Google Scholar : PubMed/NCBI

235 

Lin D, Smith MA, Champagne C, Elter J, Beck J and Offenbacher S: Porphyromonas gingivalis infection during pregnancy increases maternal tumor necrosis factor alpha, suppresses maternal interleukin-10, and enhances fetal growth restriction and resorption in mice. Infect Immun. 71:5156–5162. 2003. View Article : Google Scholar : PubMed/NCBI

236 

Robertson SA, Care AS and Skinner RJ: Interleukin 10 regulates inflammatory cytokine synthesis to protect against lipopolysaccharide-induced abortion and fetal growth restriction in mice. Biol Reprod. 76:738–748. 2007. View Article : Google Scholar : PubMed/NCBI

237 

Coultas L, Chawengsaksophak K and Rossant J: Endothelial cells and VEGF in vascular development. Nature. 438:937–945. 2005. View Article : Google Scholar : PubMed/NCBI

238 

Awasthi S and Pandey M: Association of TLR4 and TNF-a gene polymorphisms and TLR4 mRNA levels in preterm birth in a Northern Indian population. Indian Pediatr. 56:202–204. 2019. View Article : Google Scholar : PubMed/NCBI

239 

Jakovljevic A, Sljivancanin Jakovljevic T, Duncan HF, Nagendrababu V, Jacimovic J, Aminoshariae A, Milasin J and Dummer PMH: The association between apical periodontitis and adverse pregnancy outcomes: A systematic review. Int Endod J. 54:1527–1537. 2021. View Article : Google Scholar : PubMed/NCBI

240 

Regier DA, Farmer ME, Rae DS, Locke BZ, Keith SJ, Judd LL and Goodwin FK: Comorbidity of mental disorders with alcohol and other drug abuse. Results from the epidemiologic catchment area (ECA) study. JAMA. 264:2511–2518. 1990. View Article : Google Scholar : PubMed/NCBI

241 

Sundararajan S, Muthukumar S and Rao SR: Relationship between depression and chronic periodontitis. J Indian Soc Periodontol. 19:294–296. 2015. View Article : Google Scholar : PubMed/NCBI

242 

Chen P, Yao H, Su W, He Y, Cheng K, Wang Y, Peng W and Li P: Sleep deprivation worsened oral ulcers and delayed healing process in an experimental rat model. Life Sci. 232:1165942019. View Article : Google Scholar : PubMed/NCBI

243 

Lavigne GJ, Khoury S, Abe S, Yamaguchi T and Raphael K: Bruxism physiology and pathology: An overview for clinicians. J Oral Rehabil. 35:476–494. 2008. View Article : Google Scholar : PubMed/NCBI

244 

Maes M: Evidence for an immune response in major depression: A review and hypothesis. Prog Neuropsychopharmacol Biol Psychiatry. 19:11–38. 1995. View Article : Google Scholar : PubMed/NCBI

245 

Haug SR and Marthinussen MC: Acute dental pain and salivary biomarkers for stress and inflammation in patients with pulpal or periapical inflammation. J Oral Facial Pain Headache. 33:227–233. 2019. View Article : Google Scholar

246 

Wang YX, Kang XN, Cao Y, Zheng DX, Lu YM, Pang CF, Wang Z, Cheng B and Peng Y: Porphyromonas gingivalis induces depression via downregulating p75NTR-mediated BDNF maturation in astrocytes. Brain Behav Immun. 81:523–534. 2019. View Article : Google Scholar : PubMed/NCBI

247 

Martínez M, Martín-Hernández D, Virto L, MacDowell KS, Montero E, González-Bris Á, Marín MJ, Ambrosio N, Herrera D, Leza JC, et al: Periodontal diseases and depression: A pre-clinical in vivo study. J Clin Periodontol. 48:503–527. 2021. View Article : Google Scholar : PubMed/NCBI

248 

Martín-Hernández D, Martínez M, Robledo-Montaña J, Muñoz-López M, Virto L, Ambrosio N, Marín MJ, Montero E, Herrera D, Sanz M, et al: Neuroinflammation related to the blood-brain barrier and sphingosine-1-phosphate in a pre-clinical model of periodontal diseases and depression in rats. J Clin Periodontol. 50:642–656. 2023. View Article : Google Scholar : PubMed/NCBI

249 

Saei Ghare Naz M, Ramezani Tehrani F, Behroozi Lak T, Mohammadzadeh F, Nasiri M, Kholosi Badr F and Ozgoli G: Quality of life and emotional states of depression, anxiety and stress in adolescents with polycystic ovary syndrome: A cross-sectional study. Psychol Res Behav Manag. 13:203–209. 2020. View Article : Google Scholar : PubMed/NCBI

250 

Montoro J, Mullol J, Jáuregui I, Dávila I, Ferrer M, Bartra J, del Cuvillo A, Sastre J and Valero A: Stress and allergy. J Investig Allergol Clin Immunol. 19(Suppl 1): S40–S47. 2009.

251 

Eisenhofer G, Lambie DG and Johnson RH: Beta-adrenoceptor responsiveness and plasma catecholamines as determinants of cardiovascular reactivity to mental stress. Clin Sci (Lond). 69:483–492. 1985. View Article : Google Scholar : PubMed/NCBI

252 

Haug SR and Heyeraas KJ: Modulation of dental inflammation by the sympathetic nervous system. J Dent Res. 85:488–495. 2006. View Article : Google Scholar : PubMed/NCBI

253 

Esteban MA, Rodríguez A, Ayala AG and Meseguer J: Effects of high doses of cortisol on innate cellular immune response of seabream (Sparus aurata L.). Gen Comp Endocrinol. 137:89–98. 2004. View Article : Google Scholar : PubMed/NCBI

254 

do Nascimento IV, Rodrigues MIDQ, Isaias PHC, Barros-Silva PG, Sousa FB, Nunes Alves APN and Mota MRL: Chronic systemic corticosteroid therapy influences the development of pulp necrosis and experimental apical periodontitis, exacerbating the inflammatory process and bone resorption in rats. Int Endod J. 55:646–659. 2022. View Article : Google Scholar : PubMed/NCBI

255 

Gomes ESB, Farias LC, Silveira LH, Jesus CÍ, Rocha RGD, Ramos GV, Magalhães HTAT, Brito-Júnior M, Santos SHS, Jham BC, et al: Conditioned fear stress increases bone resorption in apical periodontitislesions in Wistar male rats. Arch Oral Biol. 97:35–41. 2019. View Article : Google Scholar

256 

Minhoto GB, Khoury RD, Orozco EIF, Prado RF and Valera MC: Effect of chronic unpredictable stress on the progression of experimental apical periodontitis in rats. Int Endod J. 54:1342–1352. 2021. View Article : Google Scholar : PubMed/NCBI

257 

Khoury RD, Prado RFD, Matos FDS, Meireles BR, Cardoso FGDR, Oliveira LD, Carvalho CAT and Valera MC: The influence of adrenergic blockade in rats with apical periodontitis under chronic stress conditions. Arch Oral Biol. 110:1045902020. View Article : Google Scholar

258 

Van Someren EJW: Brain mechanisms of insomnia: New perspectives on causes and consequences. Physiol Rev. 101:995–1046. 2021. View Article : Google Scholar

259 

Scott J, Kallestad H, Vedaa O, Sivertsen B and Etain B: Sleep disturbances and first onset of major mental disorders in adolescence and early adulthood: A systematic review and meta-analysis. Sleep Med Rev. 57:1014292021. View Article : Google Scholar : PubMed/NCBI

260 

Park JS, Jeong Y, Jung J, Ryu JJ, Lim HK, Jung SK and Song IS: Shift work sleep disorder is closely associated with an increased risk for periodontal disease. J Clin Periodontol. 48:1066–1075. 2021. View Article : Google Scholar : PubMed/NCBI

261 

Fang HF, Miao NF, Chen CD, Sithole T and Chung MH: Risk of cancer in patients with insomnia, parasomnia, and obstructive sleep apnea: A nationwide nested case-control study. J Cancer. 6:1140–1147. 2015. View Article : Google Scholar : PubMed/NCBI

262 

Chen LL: Advances in circadian rhythms in oral maxillofacial tissues and oral-related diseases. Zhonghua Kou Qiang Yi Xue Za Zhi. 57:481–489. 2022.In Chinese. PubMed/NCBI

263 

Lange T, Dimitrov S and Born J: Effects of sleep and circadian rhythm on the human immune system. Ann N Y Acad Sci. 1193:48–59. 2010. View Article : Google Scholar : PubMed/NCBI

264 

Chu FCS, Leung WK, Tsang PCS, Chow TW and Samaranayake LP: Identification of cultivable microorganisms from root canals with apical periodontitis following two-visit endodontic treatment with antibiotics/steroid or calcium hydroxide dressings. J Endod. 32:17–23. 2006. View Article : Google Scholar : PubMed/NCBI

265 

Brzezinski A: Melatonin in humans. N Engl J Med. 336:186–195. 1997. View Article : Google Scholar : PubMed/NCBI

266 

Reiter RJ, Rosales-Corral SA, Liu XY, Acuna-Castroviejo D, Escames G and Tan DX: Melatonin in the oral cavity: Physiological and pathological implications. J Periodontal Res. 50:9–17. 2015. View Article : Google Scholar

267 

Tavares BS, Tsosura TVS, Mattera MSLC, Santelli JO, Belardi BE, Chiba FY, Cintra LTA, Silva CC and Matsushita DH: Effects of melatonin on insulin signaling and inflammatory pathways of rats with apical periodontitis. Int Endod J. 54:926–940. 2021. View Article : Google Scholar : PubMed/NCBI

268 

Sarıtekin E, Üreyen Kaya B, Aşcı H and Özmen Ö: Anti-inflammatory and antiresorptive functions of melatonin on experimentally induced periapical lesions. Int Endod J. 52:1466–1478. 2019. View Article : Google Scholar

Related Articles

Journal Cover

July-2023
Volume 52 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ye L, Cao L, Song W, Yang C, Tang Q and Yuan Z: Interaction between apical periodontitis and systemic disease (Review). Int J Mol Med 52: 60, 2023
APA
Ye, L., Cao, L., Song, W., Yang, C., Tang, Q., & Yuan, Z. (2023). Interaction between apical periodontitis and systemic disease (Review). International Journal of Molecular Medicine, 52, 60. https://doi.org/10.3892/ijmm.2023.5263
MLA
Ye, L., Cao, L., Song, W., Yang, C., Tang, Q., Yuan, Z."Interaction between apical periodontitis and systemic disease (Review)". International Journal of Molecular Medicine 52.1 (2023): 60.
Chicago
Ye, L., Cao, L., Song, W., Yang, C., Tang, Q., Yuan, Z."Interaction between apical periodontitis and systemic disease (Review)". International Journal of Molecular Medicine 52, no. 1 (2023): 60. https://doi.org/10.3892/ijmm.2023.5263