Glycation and oxidative stress in the failure of dental implants: a case series

Published online 2013 Jul 26

Davide PietropaoliEleonora OrtuMarco SeverinoIrma CiarrocchiRoberto Gatto, and Annalisa Monaco

Abstract

Background

The aim of this case series/control study is to investigate the presence of the Advanced Glycation End products (AGEs) and oxidative stress in periimplantitis.

The study group was composed of five dental implants, failed within 6 months after implantation, taken from 5 subjects (3 M/2 F) aged between 43–57 years and stored in isotonic liquid before freezing at -80°C, according to literature. All the implants had been placed using traditional submerged technique. The whole saliva was also collected using Salimetrics device and stored at -80°C, to assess molecular analysis. Two age-matched control groups were examined: they consisted of 5 subjects encountering dental extraction for chronic periodontal disease (2 M/3 F) and 5 healthy subjects (3 M/2 F) who needed extraction for dental trauma. Their whole saliva was collected with the same method. The implants and the tooth of control groups were processed to assess Western Blotting for identification of AGEs. The case/control whole saliva was used to perform ThioBarbituric Acid Reactive Substances (TBARS) for oxidative stress evaluation.

Findings

The Western Blotting analysis on periimplantitis and periodontal disease tissues showed marked increase of AGEs when compared to healthy control tissues. Also TBARS assay of whole saliva confirmed the expectations, showing higher oxidative stress levels in periimplantitis and periodontitis groups than in healthy group.

Conclusions

With the limitation of the sample size, these results showed that oxidative stress could be involved in the aetiology of periimplantitis. This hypothesis could lead to new therapeutic strategies in periimplantitis, using antioxidant approach in addition to conventional treatments.

Findings

To date it is commonly accepted that the failure of dental implants can be defined as the inability of tissue to establish or maintain osteointegration, caused by host response and opportunistic infection. In fact, the Sixth European Workshop on Periodontology in 2008 has confirmed that “peri-implant diseases are infectious in nature. Peri-implant mucositis describes an inflammatory lesion that resides in the mucosa, while periimplantitis also affects the supporting bone” [].

To date many studies suppose that oxidative stress plays and important role in the aetiology and severity of periodontal diseases, but there are no researches in this field for periimplantitis. We suppose that the same mechanisms are involved in the failure of dental implants.

Substantially, the periodontal bacteria promote the flogistic events that lead to an increase in intracellular production of physiological Reactive Oxygen Species (ROS). The latter are highly reactive compounds due to the presence of shell electrons with unpaired valence. The most relevant radicals are OH and H2O2. ROS are formed as natural products of normal oxygen metabolism and play important roles in cells signaling and homeostasis. However, during inflammation, ROS levels can dramatically increase. This may result in the increase of oxidizing conditions, thus leading to cell structures damage. Cumulatively, this is known as oxidative stress[]. Normally, cells defend themselves against ROS damage with enzymatic and non-enzymatic systems. Alpha-1-microglobulin, superoxide dismutases, catalases, lactoperoxidases, glutathione peroxidases and peroxiredoxins are considered an important enzymatic system. Moreover, small molecules such as ascorbic acid (vitamin C), tocopherol (vitamin E), uric acid and glutathione also play important roles as non-enzymatic systems [,].

Effects of ROS on cell metabolism are well documented. The effects of ROS are not limited to the apoptosis but also include positive actions, such as the induction of host defence genes [] and the mobilization of ion transport systems []. Platelets involved in wound repair and blood homeostasis release ROS to recruit additional platelets in the sites of injury. In addition, they provide a link to the adaptive immune system via the recruitment of leucocytes [].

ROS are implicated in the cellular activity as well as in variety of inflammatory responses; furthermore, they can potentially cause cellular injury by the damage of DNA, RNA, proteins, lipids, which contributes to ageing and other pathologies like cardiovascular diseases, neurodegeneration and periodontal diseases []. The free radical theory of aging assumes that there is a single basic cause of aging, modified by genetic and environmental factors, and postulates that free radical reactions are involved in aging and age-related disorders [,,].

Soory et al. claim that an increase in ROS can induce a hyper-inflammatory status in aggressive forms of periodontal diseases, leading to an unbalance of redox status, that results in a cellular damage [] .

In addition to ROS, the Advanced Glycation End products (AGEs) are another emerging marker of oxidative stress. The AGEs are heterogeneous products, that are constantly formed in physiological conditions, but significantly increase in hyperglycaemia and excessive oxidative stress [,]. Recent studies suppose that the AGEs are involved in a large number of systemic diseases, where the oxidative component is strong, such as in diabetes and hypertension [].

The activation of these pathways is not restricted to limited areas of the body; in fact, their signaling triggers systemic responses, which are also visible in teeth-supporting tissues. Indeed, it is documented that AGEs induce the activation of pro-inflammatory molecules such as TNFalfa, Interleukin 6, 10 (IL-6, IL-10) and C-Reactive Protein (CRP). These molecules are strongly involved in host response against periodontal pathogens and promote the osteoclast activation in periodontitis [].

The activation of these complexes leads to the interaction between AGEs and RAGE cellular receptors (found in many cell populations), which amplify the release of cytokines, metalloproteinases (MMPs) and ROS []. The pathogenetic action of these compounds, in fact, performs directly, damaging the tissues, or indirectly, through the binding of a specific receptor, called RAGE, which belongs to the family of immunoglobulins []. This receptor is physiologically but poorly present in many cells, while it is over-expressed in conditions such as diabetes, vasculopathy and cancer []. The AGE-RAGE bond produces a cascade of pro-inflammatory signaling with subsequent activation of redox-sensitive transcription factors, such as NF-kB []. This interaction produces a hyper-permeability at the level of endothelial cells and activates the VCAM-1 molecules, whereas on monocytes it contributes to chemotaxis and to the increase of cytokines, such as the Tumor Necrosis Factor (TNF), and interleukins IL-1 and IL-6 []. Collagen synthesis by fibroblasts is also reduced [].

From this basis, in this preliminary case series/control study we investigate the presence of AGEs and oxidative stress in periimplantitis, in order to deepen this poorly understood field.

Methods

This study was conducted in accordance with the Declaration of Helsinki. The Committee on Ethics in Science of the University of L’Aquila approved the study and written informed consent was obtained from each subject.

Due to the lack of data about this topic, we decided at first to elucidate the possibility that oxidative stress plays a role not only in periodontitis, but also in periimplantitis. From this basis, we needed to compare a group of people with failed dental implant with a first group of people with chronic periodontal disease and a second group of healthy people. So, three groups were examined: Group 1 (periimplantitis), Group 2 (chronic periodontal disease) and Group 3 (healthy subjects), for a total number of 15 patients.

According to literature, the peri-implant tissues were compared with periodontal tissues of healthy and chronic parodontopathic subjects, as suggested by Fritz et al. [].

The enrolled patients shared the following exclusion criteria: moderate/severe hypertension (according to American Heart Association), metabolic syndrome, alcoholism, history of antibiotics/anti-inflammatories/other medications consumed in the past 6 months, drugs, diabetes, vegetarian diet, autoimmune diseases, liver and kidney diseases, cancer, metastasis, osteoporosis, radiographic evidence of bone loss, hypovitaminosis D, dyslipidemia, smoking, history of systemic diseases.

For each group, moreover, there were specific inclusion criteria:

• Group 1 (periimplantitis) – dental implants, placed using traditional submerged technique and failed within 6 months after implantation.

• Group 2 (chronic periodontal disease) – age-matched patients who needed dental extraction for chronic periodontitis. Mean of probing depth ≥ 5 mm (six conventional sites) with RX evidence of alveolar bone loss.

• Group 3 (healthy subjects) – age-matched patients who needed dental extraction for dental trauma without history of periodontal diseases.

The same operator who had placed the implants removed them after diagnosis of periimplantitis in order to mantain intraoperator repeatability. As described by literature, different production lots were used in order to reduce the mechanical defects bias of the implants. Diagnosis of periimplantitis was made according to Lindhe et al. using repeated measures over time of probing depth (PD) of conventional six sites and RX [].

Implants collection method

Five dental implants, failed within 6 months after implantation (3 from mandible, 2 from maxilla), were taken from 5 subjects (3 M/2 F) aged between 43–57 years (average 49.6 ± 4.6). Peri-implant tissues adherent to the implant spires were conserved; the contamination of the implants by oral cavity was prevented. Samples were stored in Phosphate Saline Buffest (PBS) pH 7,4 (P5368; Sigma–Aldrich, St. Louis, MO, USA) before dry freezing at -80°C.

Tooth collection method

Both in the periodontopatic and the healthy group, dental extraction followed the same procedure. After local anesthesia (Mepivacaine 2% and adrenaline 1:100.000 – Scandonest 2% Ogna Laboratori Farmaceutici – Milan), teeth were extracted and then put in PBS solution before dry freezing at -80°C. We collected 5 teeth from as many subjects with chronic periodontal disease (2 M/3 F) aged between 45–53 years (average 49.2 ± 2.9) and 5 teeth from healthy subjects (3 M/2 F) aged between 37–51 years (average 45.0 ± 5.8).

Saliva collection method

Whole saliva of all enrolled patients was also collected to assess oxidative stress analysis. Saliva was collected before any oral operation, in the morning, after a vigorous rinsing with water, using Salimetrics® collection system (Salimetrics UK – Oral Swab – Swab Storage Tube) [].

Tissues sample processing: extraction of periodontal and peri-implant tissues

According to Takatsu et al. [], the same method for tissues extraction was used in the samples. Both implants and teeth were unfrozen by bain-marie at 37°C for 5 min. First, the samples were processed with scalpel to remove the small quantity of apical and coronal tissues into the Petri dish with PBS 1x. The periodontal ligament and the peri-implant tissues were so obtained, and then dry frozen at -80°C.

SDS-Page Electrophoresis and Western Blotting of extracted tissues

The tissues pellet were unfrozen using bain-marie at 37°C for 5 min. Three wash cycles with PBS 1x at 4000RPM/10 min were done. The cells lysation was assessed in ice for 20 min using 200 uL of standard RIPA buffer (R0278; Sigma–Aldrich, St. Louis, MO, USA). Every 5 minutes vortexing was done. Three cycles of freezing-unfreezing with liquid nitrogen and three cycles of sonication in ice were assessed (10 sec for 60 sec of recovery). Bradford method was used for the proteins quantification.

Standard SDS-Page and Western Blotting were done using monoclonal antibody for AGEs (AGE06B – Biologo.de). Silver stained was performed on Polyacrylamide gel.

Saliva analysis

According to producer (Salimetric® UK), the vials containing saliva were unfrozen and centrifuged at 6000RPM/10 min to obtain whole saliva for analysis.

Oxidative stress was measured as lipid peroxidation using specific colorimetric assay for ThioBarbituric Acid Reactive Substances – TBARS – (BioAssay USA, DTBA-10). The TBARS absorbance, for each sample, was measured three times and the mean value was taken as oxidative stress level, as suggested by the producer.

Results

The statistical analysis for oxidative stress was performed using software SPSS (Version 20.0 for Windows 7). Statistical difference in oxidative stress in terms of TBARS light absorbance between groups was analyzed by one-way ANOVA with post-hoc Bonferroni correction due to the small sample size, even though the Shapiro-Wilk test revealed a normal distribution of data. The level of significance was set at p < 0.05 for all tests. Results are expressed as mean and standard deviation (Table 1).

Table 1

Results of salivary TBARS assey

Groups Abs
Health


0,62 (0,22)a


Peri Implantitis


1,25 (0,11)b


Chronic Periodontitis 1,70 (0,12)c

Mean of absorbance (Abs) and standard deviation (SD) in groups. Different superscript letters indicates statistical significant difference (P < 0.05) between groups performing one way ANOVA with post-hoc Bonferroni.

The results showed statistically significant differences in each group. In particular, the chronic periodontal disease group showed higher oxidative stress than periimplantitis and healthy groups. Periimplantitis group compared to the healthy one had higher oxidative stress levels (p < 0,001). Chronic periodontal disease compared to periimplantitis showed higher oxidative stress (p = 0,002) as described in Figure 1. Chronic periodontal disease compared to health met the expectation, showing even higher (p < 0,0001) oxidative stress levels than compared to periimplantitis.

An external file that holds a picture, illustration, etc. Object name is 1756-0500-6-296-1.jpg

Box blot of salivary TBARS in the study groups.

Western Blotting analysis showed that AGEs are present in both tissues of periimplantitis and periodontopatic groups compared to healthy subject tissues (Figure 2).

An external file that holds a picture, illustration, etc. Object name is 1756-0500-6-296-2.jpg

Western Blotting image of analyzed tissues. A) Molecular weight; B) healthy subjects; C) periimplantitis; D) chronic periodontal disease.

Discussion

With the limitation of the sample size, in this preliminary study we highlighted the possible role of AGEs and oxidative stress in the genesis and progression of periimplantitis. From our data a clear picture emerges, in which oxidative stress and consequent AGEs production are possibly involved in the failure of dental implants. This hypothesis is strongly supported by recent literature, since a possible role of AGEs and oxidative stress was established in many oral inflammatory diseases [].

One of our recent reviews of literature also supports the hypothesis that glycation and oxidative stress are the possible common link with periodontal diseases in patients with metabolic syndrome, which is an example of systemic proinflammatory condition []. Minor pro oxidant conditions, too, like moderate smoking, unbalanced diet, mild hypertension, minor or undiagnosed hyperglycemia, can promote ROS production and irreversible accumulation in genetically predisposed patients []. Finally, genetic conditions probably play an important role in the failure of dental implants and would explain why it can manifest in some patients without apparent risk factors for periimplantitis. Many studies, in fact, support the hypothesis that the accumulation of ROS is influenced by genetic factors. Motohashi et al., for example, showed that the mutation of the gene Nrf2 promotes ROS accumulation [].

The ROS production induces formation of AGEs, which irreversibly accumulate in the peri-implant tissues, and this may induce tissue damage by both affecting the functional status of collagen fibers and promoting an increase of oxidative stress and inflammation. These effects are mediated by AGE-RAGE interaction. From its side, AGEs formation in the extracellular matrix may contribute to increase the production and release of ROS from peri-impant tissues cells and phagocytes, with subsequent induction of proinflammatory cytokines and metalloproteinases (MMPs), leading to osteoblast inactivation and consequent osteoclast activation [].

Our findings are in line with literature, since we found higher values of oxidation in periodontitis than in healthy people, and also add new data about the unexplored field of oxidation/glycation in periimplantitis. Our preliminary data, in fact, showed that periimplantitis is characterized by an increase in oxidative stress markers in saliva and higher levels of AGEs in periimplant tissues. These alterations are similar in severity to those of periodontitis, even though they don’t reach the same values. The quantity of oxidation in periimplantitis group, in fact, is less than in periodontitis group. This finding is coherent with the fact that periodontal disease is a chronic disorder, with an early beginning, a major duration and, probably, a systemic involvement, of which they may be the mirror.

Also the Western Blotting analysis of AGEs showed more glycation in periimplantitis than in healthy people. This finding supports and strengthens the previous data, confirming the involvement of the oxidizing system and the AGE-RAGE interaction in periimplantitis.

Based on the data reported, a potential innovative therapy of periimplantitis may be hypothesized, using drugs with antioxidant and anti-AGEs effects. Possible interventions against AGEs formation and AGE-RAGE mediated damage are numerous. The discovery of chemical agents that can inhibit glycation reactions may have potential therapeutic importance. Pyridoxamine, one of the neutral form of vitamin B, has been shown to inhibit AGEs formation by interfering with post Amadori oxidative reactions, and it is employed in clinical trial evaluating the efficacy of pyridoxamine in inhibiting the progression of proteinuria and hyperlipidemia in diabetic patients []. A further inhibitor of protein glycation is metformine, that has additional effects on AGEs accumulation by reducing oxidative stress []. Recently it has been reported that nifedipine and telmisartan, respectively a Ca2+ channel blocker and an angiotensin receptor antagonist, exert anti-oxidative and anti-AGE-RAGE axis properties [].

A further group of anti-AGE drugs may be that of modified tetracyclines with antioxidant effects. In addition to their antimicrobial action, these molecules are effective in combating oxidative stress in periodontal disease and have beneficial effects on systemic diseases driven by oxidative stress []. This is the case of doxycycline, whose efficacy in reducing periodontal inflammation has been associated with a reduction in level of glycated haemoglobin in diabetics [].

In addition to drugs therapy, also diet and life style can provide to reduce oxidative stress and AGEs formation. In fact, flavonoids, present in many foods like green tea, chocolate and fruit, have the capacity to reduce glycation []. Also vitamins C, E, P have antioxidant proprieties []. Therefore, it is clear how dietary factors may play an important role in the strategy against the oxidation.

Conclusion

This preliminary study, with the limitation of the sample size, identifies the periimplantitis as a multifactorial disease, in which glycation and oxidative stress play a role in terms of etiology and severity. This finding remarks the recent literature assumptions, that support a closer correlation between systemic conditions and oral health. The molecular pathways that are involved in periimplantitis are unknown, but probably oxidizing is involved in its etiology, as it is in the periodontitis.

These data suggest to the clinician a new strategy to prevent the failure of dental implants. Probably, in a not too distant future, the simple measurement of oxidative stress or AGEs in saliva, prior to dental implant surgery, could prevent implant failure. To this aim, future investigation on the presence of AGEs and oxidizing in the periimplantitis is needed, in order to develop prevention and treatment strategies for this disease. However, common and safe drugs or target diets could be used in order to prevent or treat the periimplantitis.

Competing interests

All authors declare that they have no competing interests.

Authors’ contributions

DP an AM contributed to the conception and design of the study, the analysis and interpretation of the data and drafted the manuscript. EO, MS and IC were involved in the interpretation of the data and contributed to the revision of the drafted manuscript. RG provided statistical analysis. All authors read and approved the final manuscript.

References

  • Lindhe J, Meyle J. Group D of European Workshop on Periodontology. Peri-implant diseases: Consensus Report of the Sixth European Workshop on Periodontology. J Clin Periodontol. 2008;35:282–285. [PubMed[]
  • Chen AF, Chen D-D, Daiber A, Faraci FM, Li H, Rembold CM, Laher I. Free radical biology of the cardiovascular system. Clin Sci. 2012;123:73–91. doi: 10.1042/CS20110562. [PubMed] [CrossRef[]
  • Pietropaoli D, Sgolastra F, Ciarrocchi I, Spadaro A, Masci C, Petrucci A, Cattaneo R, Monaco A. Oxidative stress status in the saliva of growing subjects as a potential pubertal biomarker. Eur J Inflamm. 2012;10:209–216. []
  • Rada B, Leto TL. Oxidative innate immune defenses by Nox/Duox family NADPH oxidases. Contrib Microbiol. 2008;15:164–187. [PMC free article] [PubMed[]
  • Tyml K. Critical role for oxidative stress, platelets, and coagulation in capillary blood flow impairment in sepsis. Microcirculation. 2011;18:152–162. doi: 10.1111/j.1549-8719.2010.00080.x. [PubMed] [CrossRef[]
  • Pietropaoli D, Tatone C, D’Alessandro AM, Monaco A. Possible involvement of advanced glycation end products in periodontal diseases. Int J Immunopathol Pharmacol. 2010;23:683–691. [PubMed[]
  • Soory M. Redox status in periodontal and systemic inflammatory conditions including associated neoplasias: antioxidants as adjunctive therapy? Infect Disord Drug Targets. 2009;9:415–427. doi: 10.2174/187152609788922582. [PubMed] [CrossRef[]
  • Pietropaoli D, Monaco A, Del Pinto R, Cifone MG, Marzo G, Giannoni M. Advanced glycation end products: possible link between metabolic syndrome and periodontal diseases. Int J Immunopathol Pharmacol. 2012;25:9–17. [PubMed[]
  • Sgolastra F, Severino M, Pietropaoli D, Gatto R, Monaco A. Effectiveness of periodontal treatment to improve metabolic control in patients with chronic periodontitis and type 2 diabetes: a meta-analysis of randomized clinical trials. J Periodontol. 2012;84(7):958–973. [PubMed[]
  • Ramasamy R, Yan SF, Schmidt AM. Receptor for AGE (RAGE): signaling mechanisms in the pathogenesis of diabetes and its complications. Ann N Y Acad Sci. 2011;1243:88–102. doi: 10.1111/j.1749-6632.2011.06320.x. [PMC free article] [PubMed] [CrossRef[]
  • Ramasamy R, Vannucci SJ, Yan SSD, Herold K, Yan SF, Schmidt AM. Advanced glycation end products and RAGE: a common thread in aging, diabetes, neurodegeneration, and inflammation. Glycobiology. 2005;15:16R–28R. doi: 10.1093/glycob/cwi053. [PubMed] [CrossRef[]
  • Schmidt AM, Stern DM. Hyperinsulinemia and vascular dysfunction: the role of nuclear factor-kappaB, yet again. Circ Res. 2000;87:722–724. doi: 10.1161/01.RES.87.9.722. [PubMed] [CrossRef[]
  • Vlassara H, Brownlee M, Manogue KR, Dinarello CA, Pasagian A. Cachectin/TNF and IL-1 induced by glucose-modified proteins: role in normal tissue remodeling. Science. 1988;240:1546–1548. doi: 10.1126/science.3259727. [PubMed] [CrossRef[]
  • Hollá LI, Kanková K, Fassmann A, Bucková D, Halabala T, Znojil V, Vanĕk J. Distribution of the receptor for advanced glycation end products gene polymorphisms in patients with chronic periodontitis: a preliminary study. J Periodontol. 2001;72:1742–1746. doi: 10.1902/jop.2001.72.12.1742. [PubMed] [CrossRef[]
  • Fritz ME, Braswell LD, Koth D, Jeffcoat M, Reddy M, Cotsonis G. Experimental peri-implantitis in consecutively placed, loaded root-form and plate-form implants in adult Macaca mulatta monkeys. J Periodontol. 1997;68:1131–1135. doi: 10.1902/jop.1997.68.11.1131. [PubMed] [CrossRef[]
  • Topkas E, Keith P, Dimeski G, Cooper-White J, Punyadeera C. Evaluation of saliva collection devices for the analysis of proteins. Clin Chim Acta. 2012;413:1066–1070. doi: 10.1016/j.cca.2012.02.020. [PubMed] [CrossRef[]
  • Takatsu M, Uyeno S, Komura J, Watanabe M, Ono T. Age-dependent alterations in mRNA level and promoter methylation of collagen alpha1(I) gene in human periodontal ligament. Mech Ageing Dev. 1999;110:37–48. doi: 10.1016/S0047-6374(99)00041-X. [PubMed] [CrossRef[]
  • Motohashi H, Kimura M, Fujita R, Inoue A, Pan X, Takayama M, Katsuoka F, Aburatani H, Bresnick EH, Yamamoto M. NF-E2 domination over Nrf2 promotes ROS accumulation and megakaryocytic maturation. Blood. 2010;115:677–686. doi: 10.1182/blood-2009-05-223107. [PMC free article] [PubMed] [CrossRef[]
  • Syngle A, Vohra K, Garg N, Kaur L, Chand P. Advanced glycation end-products inhibition improves endothelial dysfunction in rheumatoid arthritis. Int J Rheum Dis. 2012;15:45–55. [PubMed[]
  • Ishibashi Y, Matsui T, Takeuchi M, Yamagishi S-I. Beneficial effects of metformin and irbesartan on advanced glycation end products (AGEs)-RAGE-induced proximal tubular cell injury. Pharmacol Res. 2012;65:297–302. doi: 10.1016/j.phrs.2011.11.001. [PubMed] [CrossRef[]
  • Falcone C, Buzzi MP, Bozzini S, Boiocchi C, D’Angelo A, Schirinzi S, Esposito C, Torreggiani M, Choi J, Ochan Kilama M, Mancia G. Microalbuminuria and sRAGE in high-risk hypertensive patients treated with nifedipine/telmisartan combination treatment: a substudy of TALENT. Mediators Inflamm. 2012;2012:874149. [PMC free article] [PubMed[]
  • Soory M. A role for non-antimicrobial actions of tetracyclines in combating oxidative stress in periodontal and metabolic diseases: a literature review. Open Dent J. 2008;2:5–12. doi: 10.2174/1874210600802010005. [PMC free article] [PubMed] [CrossRef[]
  • Engebretson SP, Hey-Hadavi J. Sub-antimicrobial doxycycline for periodontitis reduces hemoglobin A1c in subjects with type 2 diabetes: a pilot study. Pharmacol Res. 2011;64:624–629. doi: 10.1016/j.phrs.2011.06.024. [PMC free article] [PubMed] [CrossRef[]
  • Calder PC, Ahluwalia N, Brouns F, Buetler T, Clement K, Cunningham K, Esposito K, Jönsson LS, Kolb H, Lansink M, Marcos A, Margioris A, Matusheski N, Nordmann H, O’Brien J, Pugliese G, Rizkalla S, Schalkwijk C, Tuomilehto J, Wärnberg J, Watzl B, Winklhofer-Roob BM. Dietary factors and low-grade inflammation in relation to overweight and obesity. Br J Nutr. 2011;106(Suppl 3):S5–78. [PubMed[]
Read More

Surface electromyography pattern of human swallowing

Published online 2008 Mar 26

Annalisa MonacoRuggero CattaneoAlessandro Spadaro, and Mario Giannoni

Abstract

Background

The physiology of swallowing is characterized by a complex and coordinated activation of many stomatognathic, pharyngeal, and laryngeal muscles. Kinetics and electromyographic studies have widely investigated the pharyngeal and laryngeal pattern of deglutition in order to point out the differences between normal and dysphagic people. In the dental field, muscular activation during swallowing is believed to be the cause of malocclusion.

Despite the clinical importance given to spontaneous swallowing, few physiologic works have studied stomatognathic muscular activation and mandibular movement during spontaneous saliva swallowing.

The aim of our study was to investigate the activity patterns of the mandibular elevator muscles (masseter and anterior temporalis muscles), the submental muscles, and the neck muscles (sternocleidomastoid muscles) in healthy people during spontaneous swallowing of saliva and to relate the muscular activities to mandibular movement.

Methods

The spontaneous swallowing of saliva of 111 healthy individuals was analyzed using surface electromyography (SEMG) and a computerized kinesiography of mandibular movement.

Results

Fifty-seven of 111 patients swallowed without occlusal contact (SNOC) and 54 individuals had occlusal contact (SOC). The sternocleidomastoid muscles showed a slight, but constant activation during swallowing. The SEMG of the submental and sternocleidomastoid muscles showed no differences between the two groups. The SEMG of the anterior temporalis and masseter muscles showed significant differences (p < 0.0001). The duration of swallowing was significantly higher in the SNOC subjects. Gender and age were not related to electromyographic activation. Healthy SOC and SNOC behaved in different ways.

Conclusion

The data suggest that there is not a single “normal” or “typical” pattern for spontaneous saliva swallowing. The polygraph seemed a valuable, simple, non-invasive and reliable tool to study the physiology of swallowing.

Background

The balanced muscular activity in the development of the face has been recognized for many years.

Many authors have stated that the tongue and its function, swallowing, is very important in obtaining muscular and skeletal equilibrium. The muscular forces could directly mold cranial and facial development and could affect teeth position [,].

The mandibular anchorage to the cranium is a primary physiologic event that allows the action of the tongue and of the suprahyoid muscles on the hyoid bone during all phases of swallowing. The hyoid bone moves upward during the first phase of swallowing; at the same time, the mandible moves upward too to reach occlusal contact. At the end of deglutition, the hyoid bone moves downward and the mandible, leaving occlusal contact, also moves downward and reaches its rest position [,]. Mandibular stabilization on the cranium requires an isometric contraction of the mandibular elevator muscles. This work could be obtained both with the occlusal contact and with the tongue interposition between the dental arches.

Moreover, during swallowing, the neck muscles help to stabilize the cranium on the thorax, counterbalancing the propulsive forces exerted by the tongue, pharynx, and larynx [].

In dental research and in orthodontics it has been accepted for long time that saliva swallowing could be divided into two groups: Typical and Atypical swallowing. Deglutition without occlusal contact is one of the characteristics of Atypical swallowing. “Atypical” swallowing has been considered one of the malocclusion causes and its correction was suggested by some Clinicians in order to solve the “malocclusion”. The idea that only “Typical” deglutition would be a correct deglutition induced to believed that “Atypical” deglutition is pathologic and need some kind of treatment. If malocclusion is a disease, its cause (Atypical deglutition) has to consider a pathological problem too. The “Typical” and the “Atypical” swallowing concept is based on spontaneous saliva swallowing occurring during the night and the day. These definitions, currently, didn’t concern the swallowing of solid or liquid boluses occurring during feeding. Swallowing difficulties during feeding could be considered a particular case in dental practice. “Atypical” swallowing, in this case, it is a restrictive definition used in dental field.

Surface electromyography (SEMG) has been widely used in recent years to study the physiology of swallowing. Recent studies have suggested that the SEMG graphic record is a valid and reliable support in identifying normal swallowing []. The rectified and filtered SEMG represents a non-invasive tool to assess certain aspects of the complex muscle activity involved in deglutition. The SEMG for swallowing is a simple and reliable, non-invasive screening method for evaluating swallowing with low levels of discomfort [,].

The electrodes placed on the anterior belly of the digastric muscle record the electrical activity from both the anterior digastric muscles and the suprahyoid area muscles (i.e., the geniohyoid and mylohyoid muscles). All these muscles fire during swallowing []. Swallowing is a frequent physiologic act. Vaiman stated that the rate of spontaneous swallowing of saliva in healthy individuals is once every 2 minutes and 15 seconds []. The strength, duration, and frequency of contraction of the tongue muscles during swallowing are related to the morphology of the bones and teeth of the stomatognathic system, like the open bite or other kinds of malocclusion []. EMG activation precedes the swallowing biomechanical events and it is strictly connected to these. The strongest relationship is between the elevation and the anterior displacement of the hyoid bone and the SEMG signal of the submental muscles []. The duration of swallowing ranges from 0.80 to 1.60 seconds, according to the measurement techniques [,]. The same temporal pattern is shown in the SEMG of the submental muscles. This normal duration is achieved by the age of 12 years and does not change significantly until 70 years of age. After this age, the duration of swallowing shows a significant increase [,,,]. Ertekin et al. [] suggested that spontaneous saliva swallowing differs from voluntarily dry swallowing both in neurological control and in sub mental muscles activation.

During isometric work, the SEMG values are related in a non-linear way to the force and to the work generated by the muscles [,]. This situation is similar to mandibular stabilization during swallowing.

Many SEMG works concerning both the stomatognathic and the neck muscular activity during clenching or mastication in healthy or temporomandibular disorders (TMD) patients exist in the literature []. Relatively few works, however, have studied the SEMG behaviour of the stomatognathic and neck muscles during swallowing [,].

Furthermore, these works have not simultaneously considered SEMG activity and mandibular movement and could not relate isometric activity of the stomatognathic and neck muscles with an objective analysis of the occlusal contact or of the tongue interposition between the dental arches during swallowing.

In recent years, computerized kinesiography has allowed objective recordings of mandibular movement during swallowing [,,].

At present, with polygraph equipment, it is possible to record mandibular movement and SEMG activity at the same time.

The aim of our study was to investigate the mandibular movement and the activity pattern of the mandibular elevator muscles (i.e., masseter and anterior temporalis muscles) and the submental and neck muscles (i.e., sternocleidomastoid muscles) in healthy individuals during the spontaneous swallowing of saliva. A further aim of the study was to investigate the relative frequency and polygraph differences of spontaneous saliva swallowing pattern, separating people who swallows with polygraph occlusal contact (SOC) from those who swallows without occlusal contact (SNOC).

The null hypothesis, is that the mean values of the rectified SEMG and the mean time of swallowing would be statistically the same.

Methods

One hundred eleven patients (71 females and 40 males), with an average age of 32.7 years (sd, 14.1), registered for an annual dental check-up at the Dentistry Department of the University of L’Aquila, were selected for the study. A clinical dental evaluation assured the presence of all dental elements. Complete or partial denture-wearers and patients with wide reconstructions or fixed prostheses in the posterior quadrant were excluded. A clinical dental evaluation excluded the presence of TMD symptoms. A clinical neurologic evaluation excluded the presence of neurologic swallowing disorders due to the central nervous system or to the peripheral cranial and cervical nerves. A clinical medical evaluation excluded the presence of salivary gland diseases. Subjects reported no difficulties in bolus (solid or liquid) swallowing during feeding.

The subjects were informed of the study procedure, but they ignored the study purpose. The Institutional Review Board approved the research with the understanding that the subjects would undergo electromyography and kinesiography for reasons independent of the research. Ethical approval was obtained by the University’s Review Board for Health Sciences Research Involving Human Subjects and all subjects signed informed consent prior to testing.

SEMG and kinesiographic measurements

The bilateral masseter, anterior temporalis, submental, and sternocleodomastoid muscles were recorded. Disposable silver/silver chloride bipolar surface electrodes (Duotrode; Myotronics-Noromed, Inc., Seattle WA, USA) with an interelectrode distance of 2.1 +/- 1.0 mm. and an impedance at 10.0 Hz max were used. Fifty ohms were placed on the bellies of the muscles parallel to the muscle fibers.

The same electrodes and cables were used for each patient.

The submental muscles were recorded to visualize activation of the muscle during swallowing.

The masseter and anterior temporalis muscles were recorded to show isometric activation during swallowing.

The sternocleidomastoid muscles were recorded to check the position of the head during swallowing and to evaluate isometric activity of the neck during swallowing. The myoelectric and kinesiographic signals were recorded using computerised equipment. This equipment allowed the recording of muscular activity and, at the same time, mandibular movement.

The SEMG was rectified and integrated. The SEMG data of each recorded muscle were expressed in microvolts.

Mandibular movement during swallowing were recorded by the kinesiograph of the K6-I Diagnostic System (Myotronics Research Inc., Seattle, WA, USA). The equipment consisted of an array of sensors placed on the subject’s head that provided information about the position of the mandible. When the mandible moved, changes in the magnetic flux of the small bar magnet fixed on the mandibular incisor teeth were detected. The kinesiograph was connected to a computerized system that records and displays spatial coordinates in the vertical and anteroposterior axes to the nearest 0.1 mm.

When swallowing took place, the muscular pattern activation and mandibular movements were displayed on the video at the same time, allowing the observers to distinguish the swallowing function from other mandibular movements (Fig (Fig1).1). The poligraphic gains used to display the SEMG and the kinesiographic values ere Vert/AP = 1 (each plotted square represented 1 mm of vertical or anteroposterior movement every second), Lateral = 1 (each plotted square represented 1 mm of lateral movement every second), EMG = 100 in processed mode (1 vertical division represented 100 microvolts of rectified SEMG and 1 horizontal division represented 1 second), set AG (represented the displayed muscles), and speed = 1 (represented the speed of recording 1 second for each horizontal division).

An external file that holds a picture, illustration, etc. Object name is 1472-6831-8-6-1.jpg

Polygraphy of swallowing: A = rectified SEMG during swallowing; A1 = kinesiography during swallowing, B = polygraphy in occlusal position; LTA (left anterior temporalis); RTA (right anterior temporalis); LMM (left masseter); RMM (right masseter); LSM (left sternocleidomastoid); RSM (right sternocleidomastoid); LDA (left submentalis); RDA (right submentalis). Ver (blue line): vertical component of mandibular movement; AP (red line): anteroposterior component of mandibular movement; Lat (green line): lateral component of mandibular movement.

Kinesiograph was used in order to study the jaw position during spontaneous saliva swallowing. The distance between the deglutition and the occlusal position pointed out if swallowing was done in occlusal position or without occlusal contact.

The different patterns of muscle activation and of mandibular movement allowed the observer to distinguish the swallowing event and to divide the sample into two groups: 1) swallowing with an occlusal contact (SOC) and 2) swallowing with no occlusal contact (SNOC; Figs. Figs.22 and and33).

An external file that holds a picture, illustration, etc. Object name is 1472-6831-8-6-2.jpg

Swallowing without tongue interposition between dental arches (SNOC).

An external file that holds a picture, illustration, etc. Object name is 1472-6831-8-6-3.jpg

Swallowing with occlusal contact (SOC).

Recording procedure

In order to assure spontaneous saliva swallowing the following recording protocol was used.

No information were given to subjects on study purpose. In other words study subjects didn’t have any knowledge about electromyography, kinesiography and the reasons why they were tested. When the recording session started subjects had to close their eyes and wait, with closed eyes, for the end of recording session.

Polygraph recording session started after the electrodes placement and the magnet alignment.

Polygraph traces run on computer screen, when the traces reached the screen end, they were automatically deleted if the operator didn’t recorder and save them. If the traces weren’t recorded, they started to run the screen again. Operator could delete the traces at every time, in this way he could avoid that spontaneous saliva swallowing occurring at the end of the screen.

When a “spontaneous” saliva swallowing took place the operator waited for some seconds after the swallowing, and then asked the patient to reach the occlusal position. The operator recorded and saved the data of the first spontaneous saliva swallowing. After this first step, the computer was reset and it started again for another recording of spontaneous saliva swallowing. Three spontaneous saliva swallowing were recorded for each subject. Sometimes the procedure took fifteen or more minutes, in other cases it took five or less minutes, according to the variable frequency of swallowing in healthy people. The total duration of polygraph session wasn’t recorded.

A “Controller” looked at the face of the subject during the whole recording session. This “controller” didn’t have information about polygraph tracing, and he informed, lifting up one hand, on muscles and mandible movement occurring during swallowing. In this way study subject couldn’t associate his swallowing with a voice signal.

When the third polygraph swallowing was recorded the study subject was asked to open the eyes and poligraph recording session was stopped.

Measurement of the duration of swallowing

The polygraph was plotted on graph paper. A researcher trained in polygraph without knowledge of the study design measured the swallowing time using a SEMG submental activation. The time was computed from the start to the end of the submental muscle activation during the 3D movements on the kinesiographic trace. The measurement was done in seconds. For statistical analysis, an average of three measurements was used for each patient.

Statistical analysis

A descriptive statistical analysis was done using the STATA package, both for the SEMG amplitude of each recorded muscle and for the duration of the swallowing.

A summary statistic was initially done on the entire sample.

Two comparison tests with independent samples (i.e., paired Student’s t tests) were used to compare the mean value of the rectified SEMG and the mean duration time of swallowing with the SOC and SNOC groups. The level of significance was set at p < 0.05.

Analysis of variance (ANOVA) was performed to establish the influence of gender, age, and swallowing patterns (SOC-SNOC) on the SEMG values and on the duration of swallowing.

Results

Table Table11 shows the swallowing rectified SEMG (mean and sd) of the entire sample (111 patients). The submental muscles showed the highest SEMG activation (LDA, 25.42; RDA, 26.06). Masseter (LMM, 17.05; RMM, 16.30), and the anterior temporalis muscles (LTA, 17.81; RTA, 17.69) were activated during swallowing, but had a lower SEMG value than the submental group. The sternocleidomastoid muscles (LSC, 6.70; RSC, 6.91) reached the lowest activation values.

Table 1

Swallowing rectified SEMG of entire sample study (111 patients)

LTA LMM RMM RTA LSC LDA RDA RSC
Means 17.81 17.05 16.30 17.69 6.70 25.42 26.06 6.91
SD 19.17 17.09 14.59 20.84 5.25 12.43 11.83 4.39
SE 1.82 1.62 1.38 1.97 0.49 1.18 1.12 0.41

Values are represented in microvolts.

SD = standard deviation;

SE = standard error;

LTA = Left Temporalis Anterior;

LMM = Left Masseter;

RMM = Right Masseter;

RTA = Right Temporalis Anterior;

LSC = Left Sternoscleidomastoid;

LDA = Left Digastric;

RDA = Right Digastric;

RSC = Right Sternocleidomastoid

Table Table22 shows the comparison test (i.e., paired Student’s t tests) on the mean value of the rectified SEMG in the SOC and SNOC groups.

Table 2

SEMG swallowing values in SOC and SNOC group

LTA LMM RMM RTA LSC LDA RDA RSC
SOC 54 54 54 54 54 54 54 54

Means 26.24 24.09 22.94 27.16 6.38 25.59 26.87 6.83
SD 21.74 21.74 17.9 25.52 5.14 13.08 11.91 3.84
SE 3.18 2.95 2.43 3.47 0.69 1.78 1.62 0.52

SNOC 57 57 57 57 57 57 57 57

Means 9.82 10.38 10.01 8.71 7.0 25.26 25.29 7.0
SD 8.46 5.75 5.69 8.26 5.39 11.9 11.8 4.88
SE 1.10 0.76 0.75 1.09 0.71 1.57 1.56 0.64

p 0.0001 0.0001 0.0001 0.0001 0.2 0.4 0.2 0.3

Values are represented in microvolts

SOC = Subjects in Occlusal Contact;

SNOC = Subjects Not in Occlusal Contact;

SD = standard deviation; SE = standard error;

LTA = Left Temporalis Anterior;

LMM = Left Masseter;

RMM = Right Masseter;

RTA = Right Temporalis Anterior;

LSC = Left Sternoscleidomastoid;

LDA = Left Digastric;

RDA = Right Digastric;

RSC = Right Sternocleidomastoid

In agreement with the polygraphic analysis, 54 patients swallowed with occlusal contact and 57 patients swallowed without occlusal contact.

No significant differences existed in the SEMG activity of the submental muscles between the SOC and SNOC groups (LDA, p = 0.4; RDA, p = 0.2).

No significant differences existed in the sternocleidomastoid muscles between the SOC and SNOC groups (LSC, p = 0.2; RSC, p = 0.3).

A highly significant difference (p = 0.0001) existed between the SOC and SNOC groups in all the SEMG activation of the stomatognatic muscles (LTA, RTA, LMM, RMM).

Table Table33 shows the comparison test (i.e., paired Student’s test) on the mean duration of swallowing in the SOC and SNOC groups.

Table 3

Swallowing duration in SOC and SNOC groups

SOC SNOC
Swallowing duration (Means) 1.47 1.33 *
SD 0.48 0.38
SE 0.5 0.05

p

Values are represented in seconds

SOC = Subjects in Occlusal Contact;

SNOC = Subjects Not in Occlusal Contact;

SD = standard deviation;

SE = standard error

* = p = 0.02

A significant difference (p = 0.02) existed in the duration of swallowing between the SOC (1.472 seconds) and SNOC (1.327 seconds) groups.

Tables Tables4,4,5,5,66 show the incidence on the muscles recorded by the SEMG of the swallowing patterns (SOC and SNOC), gender, and age.

Table 4

Analysis of variance (ANOVA): incidence of swallowing pattern on SEMG

Source Partial SS df MS F Prob > F
LTA 7472.91 1 7472.91 24.69 0.0000
RTA 9436.58 1 9436.58 26.81 0.0000
LMM 5209.63 1 5209.63 21.09 0.0000
RMM 4633.77 1 4633.77 26.86 0.0000
LSC 10.35 1 10.35 0.37 0.5428
RSC .77 1 .77 0.04 0.8426
LDA 3 1 3. 0.02 0.8898
RDA 68.54 1 68.54 0.49 0.4867

SS = Sum of Square

MS = Model of Sum of Square

F = Distribution of the Sample

Table 5

Analysis of variance (ANOVA): incidence of gender on SEMG

Source Partial SS df MS F Prob > F
LTA 585.86 1 585.86 1.60 0.2084
RTA 1719.49 1 1719.49 4.07 0.0462
LMM 1105.25 1 1105.25 3.88 0.0513
RMM 725.59 1 725.59 3.48 0.0647
LSC 30.88 1 30.88 1.12 0.2925
RSC 41.94 1 41.94 2.20 0.1409
LDA 390.35 1 390.35 2.56 0.1126
RDA 394.94 1 394.94 2.87 0.0932

SS = Sum of Square

MS = Model of Sum of Square

F = Distribution of the Sample

Table 6

Analysis of variance (ANOVA): incidence of age on SEMG

Source Partial SS df MS F Prob > F
LTA 8959.39 1 218.52 0.48 0.9938
RTA 14496.99 1 353.58 0.73 0.8577
LMM 9164.85 1 223.53 0.67 0.9146
RMM 6554.50 1 159.86 0.65 0.9284
LSC 898.64 1 21.92 0.71 0.8841
RSC 819.33 1 19.98 1.06 0.4083
LDA 8718.61 1 212.65 1.77 0.0182
RDA 5892.52 1 143.72 1.04 0.4312

SS = Sum of Square

MS = Model of Sum of Square

F = Distribution of the Sample

Gender and age did not explain the SEMG variability in all the muscles recorded.

The swallowing pattern (SOC and SNOC) accounted for the high statistical significance in the variability of the LTA, RTA, LMM, and RMM SEMG data, but was not able to account for the variability in the LSC, RSC, LDA, and RDA muscles.

Table Table77 shows the incidence of gender, age, and swallowing pattern (SOC and SNOC) on the duration of swallowing.

Table 7

Analysis of variance (ANOVA): incidence of gender, age, and swallowing pattern (SOC, SNOC) on duration of swallowing.

Source Partial SS df MS F Prob > F
sex 14.88 1 14.88 0.75 0.3898
age 904.81 41 22.07 1.18 0.2643
swpat 82.25 1 82.25 4.25 0.0416

SS = Sum of Square

MS = Model of Sum of Square

F = Distribution of the Sample

Swpat = Swallowing Pattern

Gender and age did not account for the variability in the duration of swallowing.

The swallowing pattern explained in a significant way (p = 0.04) the variability in the duration of swallowing.

Discussion

In 1953, Jankelson [] stated that the only physiologic occlusal contact during mastication occurs when swallowing takes place. Occlusal stability is needed to give a skeletal support to the muscular events related to swallowing []. Up to now, some orthodontic schools aim to obtain correct swallowing when the occlusal contact is reached during swallowing.

Nevertheless, recent findings [,,] have suggested that the interposition of the tongue between the dental arches during swallowing is a physiologic event.

Our data confirmed the data of Monaco [] on spontaneous saliva swallowing and Sadalla [] on voluntarily dry swallowing regarding the incidence of swallowing without an occlusal contact.

In our sample of healthy patients, 57 of 111 swallowed without any occlusal contact and 54 had occlusal contact. Sadalla, in a kinesiographic study, stated that 70% of dentate patients swallowed with the interposition of the tongue between the teeth arches.

According to Erkin et al. [] who suggested a different nervous control and peripheral muscular pattern of spontaneous and voluntarily swallows, the difference reported in Monaco and Sadalla studies could be due to different method and different swallow protocol used. Furthermore, Monaco studied the spontaneous saliva svallowing, Sadalla investigated the voluntarily dry swallowing. Nevertheless, swallowing without occlusal contact seems to be frequent in healthy people.

Monaco, moreover, suggested that, swallowing with the tongue interposition could be considered a normal pattern of muscular activation during spontaneous saliva swallowing. Monaco dividing the study sample into sub-diagnosis groups, pointed out that spontaneous saliva swallowing with an occlusal contact is more frequent in people who received a prosthetic treatment []. She noticed that the neuromuscular pattern requiring occlusal contact during deglutition could be unfavourable for the health of the stomatognathic system if some unbalanced muscular forces developed in the occlusion.

Logeman [] stated that spontaneous saliva swallowing contain about 1 ml of saliva; it’s very likely that such small amount of liquid could be swallowed by either occlusal or no-occlusal contact in healthy people.

The fact that a small amount of bolus (i.e. 1 ml saliva) can easily be swallowed by either occlusal or no occlusal contact doesn’t means that a preferred spontaneous pattern of swallowing doesn’t exist.

Dental practice and Orthodontics are facing, daily, on the muscular forces exerted by tongue, lips, cheeks, masseter, temporalis anterior etc. during spontaneous saliva swallowing on teeth and on neck muscles.

One of the signs of “Atypical swallowing” is the interposition between dental arches of the tongue. The effect of the interposition is the no-occlusal contact between the dental arches. This spontaneous swallowing pattern is considered one of the causes of malocclusion.

Our study suggests that in healthy people without objective or subjective swallowing disorders there are two different preferred polygraph patterns of spontaneous swallowing.

A previous kinesiographic work [] suggested that spontaneous saliva swallowing without occlusal contact it isn’t more frequently associated than spontaneous saliva swallowing with occlusal contact to Temporo Mandibular Disorder (TMD) or to teeth loss and prosthetic treatment. On the contrary, spontaneous saliva swallowing with occlusal contact seemed to be more frequently related to teeth loss and prosthetic treatment than spontaneous saliva swallowing without occlusal contact.

Monaco conclusions were based on kinesiographic study and couldn’t relate muscular activation or tip of tongue position during mandible movement.

Despite of the fact that dental practitioners assign considerable importance to the physiology of swallowing, [,,,], few works have studied the stomatognathic muscular pattern of this event [,].

The rectified SEMG of the submental muscles has been recently used to study the physiology of swallowing. The SEMG appears to be a reliable, simple, and noninvasive tool to analyse the physiologic events related to swallowing. In particular, the SEMG of the submental muscles seems to be related to the elevation of the hyoid bone and its return to a resting position.

In our study, the SOC and SNOC groups showed the same amplitude in the SEMG of the submental muscles, suggesting that the forces released by these muscles, despite the spontaneous saliva swallowing pattern, were comparable.

In agreement with other authors [,,,] the submental SEMG allows screening of some normal and non-physiologic characteristics of swallowing. Our study group was comprised of healthy subjects without TMD or swallowing disorders, therefore, the submental SEMG showed the physiologic activation of the muscles. The submental SEMG gave little information about the forces released on the neck and on the stomatognathic structures by muscular activation during swallowing.

The sternocleidomastoid muscles showed a slight, but constant activation during spontaneous saliva swallowing. The neck muscles contributed to stabilizing the head on the trunk during the isometric work done by the mandibular elevator muscles []. Our data suggest that the same activity takes place during spontaneous saliva swallowing and that the sternocleidomastoid muscles of the SOC and SNOC groups activated themselves in the same way, discharging on the neck and on the trunk the forces exerted during spontaneous saliva swallowing. This finding is noteworthy for the influence on the body posture exerted by the physiologic stomatognathic acts.

Our data confirm that during deglutition, the masseter and anterior temporalis muscles activate themselves at the same time with the submental and sternocleidomastoid muscles.

The rectified SEMG showed an increase of electrical potential from these muscles.

The rectified SEMG increased when the isometric contraction took place. An increase in the electrical potential during swallowing goes along with the mandibular stabilization obtained with the isometric contraction of the recorded muscles.

During isometric contraction, the increase of the myoelectrical potential is related in a non-linear way to the increase in the force generated by the muscles [,].

During spontaneous saliva swallowing this force is projected on the teeth and on the skeletal structure of the face, head, and neck.

The frequency [,] of swallowing during the day and night justifies the molding action of this function on the skeletal structure.

In our sample the SOC and SNOC groups behaved in a different way.

The activity of the masseter and the anterior temporalis muscles in the SOC group was significantly higher than in the SNOC group. The data in the present study confirmed the findings of Wilson [] and Moriniere [], who stated that the duration of swallowing in healthy adult people ranges from 0.80 to 1.60 seconds. In our sample, the duration range of swallowing in the SOC group was significantly higher than in the SNOC group, but the swallowing duration of both groups is included in 0.8–1.6 range. It is possible that this range consider spontaneous saliva and voluntarily dry swallowing.

Ertekin [] suggested that spontaneous saliva swallowing lasted less than 1 sec., and asserted that swallowing lasting more than 1 sec. could be driven by some kind of voluntarily action.

Swallowing duration in our sample lasted 1.327 sec. in NSOC and 1.472 in SOC. We can’t exclude that some voluntarily drive affected our data, but such drive would be randomly distributed over the entire sample, and it wouldn’t compromise the significant difference of swallowing duration between the SOC and SNOC group, and it wouldn’t invalidate the general significance of the results for dental practice and orthodontics in healthy individuals.

Moreover, gender and age were not related to the activation of the SEMG during swallowing, while the swallowing pattern was related to the SEMG and to the duration of swallowing. People who swallow with an occlusal contact had higher SEMG values of the masseter and anterior temporalis muscles and higher values for the duration of swallowing.

These data suggest that during swallowing with an occlusal contact, higher and more prolonged forces are released on the teeth and on the skeletal structure partly justifying Monaco’s findings. [] It is possible that people with a swallowing neuromuscular pattern of the SOC type and with an occlusal plane in disharmony with the muscular forces, have suffering involving the teeth and periodontium. In contrast, people with a swallowing neuromuscular pattern of SNOC type could use the tongue like a bite and could counterbalance a possible disharmony between the occlusal plane and the muscular forces. In this case, the teeth could change position, but would not suffer from a periodontal loss.

Further work is needed to support these considerations; in particular, it would be interesting to compare the polygraph pattern of spontaneous saliva swallowing and voluntarily dry swallowing in order to investigate the difference of this two kind of deglutition, and to analyze the symmetry of activation of the muscles during swallowing in order to detect the occlusal disharmony and to relate it with periodontal diseases or loss of teeth.

Conclusion

The polygraph seems to be a valuable, simple, non-invasive, and reliable tool to study the physiology of swallowing.

Even if SOC and SNOC behave in different ways, our data suggest that there is not a single normal or typical pattern for swallowing in healthy subjects without objective or subjective signs or symptoms of swallowing disorders. It is possible to suggest that the interposition of the tongue between the dental arches during swallowing could be a physiologic event similar to swallowing with an occlusal contact.

No significant differences existed in the SEMG activity of the submental muscles between the SOC and SNOC groups.

No significant differences existed in the sternocleidomastoid muscles between the SOC and SNOC groups.

A highly significant difference (p = 0.0001) existed between the SOC and SNOC groups in all the SEMG activation of the stomatognatic muscles (LTA; RTA, LMM, RMM).

This work allows describing polygraph of muscle activity and mandible movement during spontaneous saliva swallowing in healthy subjects, showing normative data for dental use.

Furthermore, this study could be clinically relevant suggesting objective data (swallowing duration and muscle activity pattern) in order to check dental therapy (prosthetic and orthodontic treatment).

Competing interests

The author(s) declare that they have no competing interests.

Authors’ contributions

MA provided the study design, the coordination of the data collection, the manuscript conception, the drafting, and review of the article. CR reviewed the data collection, provided the analysis and the interpretation of the data, and contributed to the manuscript conception. SA participated in the collection and interpretation of the data and contributed to the drafting of the manuscript. GM participated in the study design and significantly contributed to the review of the manuscript. All authors read and approved the final manuscript.

Pre-publication history

The pre-publication history for this paper can be accessed here:

http://www.biomedcentral.com/1472-6831/8/6/prepub

Acknowledgements

The authors wish to acknowledge Dr. Vittorio Serafini for his contribution to the study.

References

  • Lowe AA, Takada K. Associations between anterior temporal, masseter, and orbicularis oris muscle activity and craniofacial morphology in children. Am J Orthod. 1984;86:319–30. doi: 10.1016/0002-9416(84)90143-X. [PubMed] [CrossRef[]
  • Frohlik K, Thuer U, Ingervall B. Pressure from the tongue on the teeth in young adults. Angle Orthod. 1991;61:17–24. [PubMed[]
  • Wilson EM, Green JR. Coordinative organization of lingual propulsion during the normal adult swallow. Dysphagia. 2006;21:226–36. doi: 10.1007/s00455-006-9053-4. [PMC free article] [PubMed] [CrossRef[]
  • Clark GT, Browne PA, Nakano M, Yang Q. Co-activation of sternocleidomastoid muscles during maximum clenching. J Dent Res. 1993;72:1499–502. [PubMed[]
  • McConnel FM, Cerenko D, Jackson RT, Guffin TN., Jr Timing of major events of pharingeal swallowing. Arch Otolaryngol Head Neck Surg. 1988;114:1413–8. [PubMed[]
  • Crary MA, Carnaby Mann GD, Groher ME. Identification of swallowing events from sEMG signals obtained from healthy adults. Dysphagia. 2007;22:94–9. doi: 10.1007/s00455-006-9059-y. [PubMed] [CrossRef[]
  • Vaiman M, Eviatar E, Segal S. Evaluation of normal deglutition with the help of rectified surface electromyography records. Dysphagia. 2004;19:125–32. doi: 10.1007/s00455-003-0504-x. [PubMed] [CrossRef[]
  • Vaiman M, Eviatar E, Segal S. Surface electromyography studies of swallowing in normal subjects: a review of 440 adults. Report 2. Quantitative data: amplitude measures. Otolaryngol Head Neck Surg. 2004;131:773–80. doi: 10.1016/j.otohns.2004.03.014. [PubMed] [CrossRef[]
  • Widmalm SE, Lillie JH, Ash MM. Anatomical and electromyographic studies of the digastric muscles. J Oral Rehab. 1988;15:3–21. doi: 10.1111/j.1365-2842.1988.tb00142.x. [PubMed] [CrossRef[]
  • Vaiman M, Nahlieli O, Segal S, Eviatar E. Electromyography monitoring of patients with salivary gland diseases. Otolaryngol Head Neck Surg. 2005;133:869–73. doi: 10.1016/j.otohns.2005.05.044. [PubMed] [CrossRef[]
  • Crary MA, Carnaby Mann GD, Groher ME. Biomechanical correlates of surface electromyography signals obtained during swallowing by healthy adults. J Speech Lang Hear Res. 2006;49:186–93. [PubMed[]
  • Moriniere S, Beutter P, Boiron M. Sound component duration of healthy human pharyngoesophageal swallowing: a gender comparison study. Dysphagia. 2006;21:175–82. doi: 10.1007/s00455-006-9023-x. [PubMed] [CrossRef[]
  • Vaiman M, Eviatar E, Segal S. Surface electromyographic studies of swallowing in normal subjects: a review of 440 adults. Report 3. Qualitative data. Otolaryngol Head Neck Surg. 2004;131:977–85. doi: 10.1016/j.otohns.2004.03.015. [PubMed] [CrossRef[]
  • Vaiman M, Segal S, Eviatar E. Surface electromyographic studies of swallowing in normal children, age 4–12 years. Int J Pediatr Otorhinolaryngol. 2004;68:65–73. doi: 10.1016/j.ijporl.2003.09.014. [PubMed] [CrossRef[]
  • Ertekin C, Kiylioglu N, Tarlaci S, Turman AB, Secil Y, Aydogdu I. Voluntary and reflex influences on the initiation of swallowing reflex in man. Dysphagia. 2000;16:40. doi: 10.1007/s004550000041. [PubMed] [CrossRef[]
  • Lawrence JH, De Luca CJ. The myoelectrical signal versus force relationship in different human muscles. J Applied Physiol. 1983;54:1653–1659. [PubMed[]
  • Hagberg C, Agerberg G, Hagberg M. Regression analysis of electromyographic activity of masticatory muscles versus bite force. Scand J Dent Res. 1985;93:396–402. [PubMed[]
  • Ferrario VF, Sforza C, Zanotti G, Tartaglia GM. Maximal bite forces in healthy young adults as predicted by surface electromyography. J Dent. 2004;32:451–457. doi: 10.1016/j.jdent.2004.02.009. [PubMed] [CrossRef[]
  • Sadalla K, de Souza R, Mollo F, Compagnoni MA. Kinesiographic study of deglutition in dentate individuals and complete denture wearers. Cranio. 2007;25:37–41. [PubMed[]
  • Widmalm SE, Lee Y, McKay DC. Clinical use of qualitative electromyography in the evaluation of jaw muscle function: a practitioner’s guide. Cranio. 2007;25:63–73. [PubMed[]
  • Liu ZJ, Yamagata K, Kasahara Y, Ito G. Electromyographic examination of jaw muscles in relation to symptoms and occlusion of patients with temporomandibular joint disorders. J Oral Rehabil. 1999;26:33–47. doi: 10.1046/j.1365-2842.1999.00356.x. [PubMed] [CrossRef[]
  • Miralles R, Gutierrez C, Zucchino G, Cavada G, Carvajal R, Valenzuela S, Palazzi C. Body position and jaw posture effects on supra-and infrahyoid electromyographic activity in humans. Cranio. 2006;24:98–103. [PubMed[]
  • Ertekin C, Keshin A, Kijlioglu N, Kirazli Y, Yagiz A, Tarlaci S, Aydogdu I. The effect of head and neck positions on oropharyngeal swallowing: a clinical and electrophysiologic study. Arch Phys Med Rehabil Sept. 2001;82:1255–1260. doi: 10.1053/apmr.2001.25156. [PubMed] [CrossRef[]
  • Monaco A, Cattaneo R, Spadaro A, Marchetti E, Barone A. Prevalence of atypical swallowing: a kinesiographic study. Eur J Paediatr Dent. 2006;7:187–91. [PubMed[]
  • Nogueras J, Salsench J, Torrent J, Samsò J, Peraire M, Anglada JM. Study on the mandibular movement during swallowing. Cranio. 1991;9:322–327. [PubMed[]
  • Jankelson B. The physiology of the stomatoghnathic system. JADA. 1953;46:375–381. [PubMed[]
  • Jankelson B. Physiology of the Human Dental Occlusion. JADA. 1955;50:664–670. [PubMed[]
  • Logeman JA. Evaluation and treatment of swallowing disorders. Pro-ed. Austin, Texas. Second. 1998. []
  • Chi-Fishman G, Stone M, McCall GN. Lingual action in normal sequential swallowing. J Speech Lang Hear Res. 1998;41:771–85. [PubMed[]
Read More

Periodontal disease: the influence of metabolic syndrome

Published online 2012 Sep 25

Enrico MarchettiAnnalisa MonacoLaura ProcacciniStefano MummoloRoberto GattoStefano TetèAlberto BaldiniSimona Tecco, and Giuseppe Marzo

Abstract

Metabolic syndrome (MetS) is a cluster of cardiovascular risk factors that include obesity, impaired glucose tolerance or diabetes, hyperinsulinemia, hypertension, and dyslipidemia. Recently, more attention has been reserved to the correlation between periodontitis and systemic health. MetS is characterized by oxidative stress, a condition in which the equilibrium between the production and the inactivation of reactive oxygen species (ROS) becomes disrupted. ROS have an essential role in a variety of physiological systems, but under a condition of oxidative stress, they contribute to cellular dysfunction and damage. Oxidative stress may act as a common link to explain the relationship between each component of MetS and periodontitis. All those conditions show increased serum levels of products derived from oxidative damage, promoting a proinflammatory state. Moreover, adipocytokines, produced by the fat cells of fat tissue, might modulate the balance between oxidant and antioxidant activities. An increased caloric intake involves a higher metabolic activity, which results in an increased production of ROS, inducing insulin resistance. At the same time, obese patients require more insulin to maintain blood glucose homeostasis – a state known as hyperinsulinemia, a condition that can evolve into type 2 diabetes. Oxidation products can increase neutrophil adhesion and chemotaxis, thus favoring oxidative damage. Hyperglycemia and an oxidizing state promote the genesis of advanced glycation end-products, which could also be implicated in the degeneration and damage of periodontal tissue. Thus, MetS, the whole of interconnected factors, presents systemic and local manifestations, such as cardiovascular disease and periodontitis, related by a common factor known as oxidative stress.

Introduction

This review inquires into and tries to clarify the relationship between metabolic syndrome and periodontitis.

Periodontitis is a family of diseases that affect dental supporting tissues, caused by infections sustained by periodontal pathogens such as Porphyromonas gingivalis, Prevotella intermedia, Tannarella forsythia, and Aggregatibacter actinomycetmcomintans, which lead to soft and hard tissue destruction, dental mobility, and the loss of dental elements [].

Susceptibility to these diseases is highly variable and depends on host responses to periodontal pathogens. Although bacteria cause plaque-induced inflammatory periodontal disease, the progression and clinical characteristics of these diseases are influenced by both acquired and genetic factors that can modify susceptibility to infection [].

Periodontitis in the United States has a prevalence of 30% to 50% of the population, but only about 10% have severe forms. It tends to be more common in economically disadvantaged populations or regions. Its occurrence decreases with a higher standard of living. Individuals of Israeli, Yemenite, North-African, South Asian, or Mediterranean origin have a higher prevalence of periodontal disease than individuals from European areas [].

Periodontitis must be distinguished from gingivitis (properly, inflammation of the gum tissue), which is a term used to describe a non-destructive periodontal disease [].

The pathophysiological mechanism of gingivitis is in response to bacterial biofilms adhering to tooth surfaces. Epidemiologically, gingivitis is the most common form of periodontal disease. From a prognostic point of view, in the absence of treatment, gingivitis may progress to periodontitis, which is a destructive form of periodontal disease [], but in some sites or individuals, gingivitis never progresses to periodontitis [].

Data indicate that periodontitis is always preceded by gingivitis [], while gingivitis can be prevented through regular oral hygiene that includes daily brushing and flossing [,].

Periodontitis depends on host responses to periodontal pathogens.

The initial increased presence of neutrophils at the site is followed by the release of cytokines by neutrophils and macrophages; the chemical mediators released include tumor necrosis factor alpha (TNF-α), interleukin-1 (IL-1), and prostaglandins.

The inflammatory process includes the stimulation of fibroblasts by IL-1 and the secretion of matrix metalloproteinases (MMP), of which collagenase is the most prominent, by polymorphonuclear neutrophils. MMPs are responsible for increased collagen breakdown, and TNF-α is primarily responsible for increased osteoclast activity resulting in bone resorption.

T-lymphocytes secrete receptor activator of nuclear factor kappa-B ligand (RANKL), which is involved in osteoclast activity and, therefore, bone resorption [].

Periodontitis has also been associated with elevations in circulating levels of IL-6 and C-reactive protein (CRP). IL-6 is an important proinflammatory cytokine involved in the regulation of host response to tissue injury and infection. It is produced by a variety of cells, such as monocytes, fibroblasts, osteoblasts, and vascular endothelial cells, in response to inflammatory challenges. Moreover, it is widely accepted that IL-6 induces CRP production.

In addition, a significant overexpression of IL-21, IL-1β, IL-17, and IL-23p19 has been detected in tissues affected by periodontal disease compared with healthy gingival tissues. In particular, IL-21 is overexpressed in chronic periodontitis gingival tissues and is correlated with the clinical parameters of periodontal destruction and with proinflammatory cytokines [].

A negative modulatory role of IL-4 and IL-13 in osteotropic cytokine production could be a mechanism that plays an important inhibitory role in inflammation-induced periodontitis. In facts the activation of STAT6 by IL-4 and IL-13, through type 2 IL-4 receptors, seems to inhibit the production of IL-11 and leukemia inhibitory factor stimulated by IL-1β and TNF-α in human gingival fibroblasts [].

In addition, IL-10 and tumor growth factor-β1 (TGF-β1) are down-regulated in periodontal lesions. Generalized aggressive periodontitis subjects are characterized by a higher IL-1β/IL-10 ratio than are periodontally healthy subjects, suggesting an imbalance between pro- and anti-inflammatory cytokines in generalized aggressive periodontitis. IL-10 is also associated with periodontal health and seems to be a regulator of inflammation and alveolar bone loss in periodontal diseases. It might be involved in controlling the inflammatory process at periodontally healthy sites [].

Metabolic syndrome

Metabolic syndrome (MetS) is characterized by multiple disorders. Oxidative stress seems to have a great role in the ethiopathogenesis of MetS and is the common factor useful to explain the interconnection of all the components of MetS.

In this review we analyze singularly the overlapping factors of each component and the common ways of ethiopathogenesis Figure Figure11.

An external file that holds a picture, illustration, etc. Object name is 1743-7075-9-88-1.jpg

Flow chart that indicates the pathophysiological mechanism underlying the onset of Mets.

MetS is a pathology largely diffused in western countries. Its diagnostic criteria have only recently been better defined, even though they are still ambiguous, because these diseases, including obesity, hyperglycemia, hyperinsulinemia, and dyslipidemia, are all considered serious risk factors for heart diseases [,]. As MetS is characterized by numerous factors, it is very difficult to establish a correct diagnosis and an adequate therapy.

MetS was initially defined as Syndrome X by Reaven [] and then as the “deadly quartet,” referring to the synergy of its components, such as hyperinsulinemia, hypertension, hypertriglyceridemia, and visceral obesity. Subsequently, it was defined as insulin-resistance syndrome, since authors believed that insulin resistance was the dominant factor predisposing the occurrence of other symptoms.

Besides the risk factors already mentioned, other peculiarities that seem to be associated with MetS include hepatic steatosis, inflamed adipose tissue, enhanced clotting factor activity, endothelial dysfunction, inflammation, and obviously oxidative stress [].

Numerous definitions have been proposed for MetS, but the criteria proposed by the National Cholesterol Education Program (NCEP) Adult Treatment Panel III are actually the most used []. The American Heart Association describes MetS as a syndrome characterized by a group of metabolic risk factors in an individual []. These factors include the following:

Abdominal obesity, i.e., excessive fat tissue in and around the abdomen;

Atherogenic dyslipidemia, i.e., blood fat disorders, high triglycerides, low high-density lipoprotein cholesterol (HDL-C), and high low-protein lipoprotein cholesterol (LDL-C) that foster plaque buildup in the artery walls;

Elevated blood pressure;

Insulin resistance or glucose intolerance, i.e., the body cannot properly use insulin or blood sugar;

Prothrombotic state, e.g., high fibrinogen or plasminogen activator inhibitor-1 in the blood; and

Proinflammatory state, e.g., elevated CRP in the blood.

The American Heart Association and the National Heart, Lung, and Blood Institute recommend that MetS be identified as the presence of three or more of the following components:

Elevated waist circumference:

Men – equal to or greater than 40 inches (102 cm);

Women – equal to or greater than 35 inches (88 cm);

Elevated triglycerides: equal to or greater than 150 mg/dL;

Reduced HDL (“good”) cholesterol:

Men – less than 40 mg/dL;

Women – less than 50 mg/dL;

Elevated blood pressure: equal to or greater than 130/85 mm Hg;

Elevated fasting glucose: equal to or greater than 100 mg/dL.

However, some authors do not find as clinically useful MetS diagnosis as a defined entity. They consider definition criteria and clinical and laboratory parameters to be ambiguous and incomplete, and the cardiovascular risk factors associated with MetS the same as pathologies considered separately []. Moreover, they argue that total therapy does not differ from that for each single disease and that the hypothesis of insulin resistance as a common cause is uncertain [].

People with MetS are at increased risk of coronary heart disease or other diseases related to plaque buildup in the artery walls, such as stroke and peripheral vascular disease and type 2 diabetes. The preponderant, fundamental risk factors for this syndrome appear to be obesity and insulin resistance, but other conditions associated with the syndrome include physical inactivity, aging, hormonal imbalance, and genetic predisposition as seen in the handicapped and senior citizens.

The necessity of organizing the exact diagnosis of MetS arises from the need to determine patients at extreme risk of developing cardiovascular disease (CVD) and to establish an appropriate therapy and prevention []. The first goal of the clinical management of MetS is to reduce the major risks for CVD and type 2 diabetes by stopping smoking, stabilizing LDL and blood pressure parameters, maintaining glucose levels at the recommended values, reducing body weight (body mass index less than 25 kg/m2) through an adapted diet, and doing moderate-intensity physical activity for at least 30 minutes on most days of the week.

Oxidative stress and metabolic syndrome

As previously shown, the role of oxidative stress in the ethiopathogenesis of MetS and its characteristic signs is widely supported [].

Reactive oxygen species (ROS) are physiologically produced by the cellular metabolism. They are highly reactive species, able to exist independently, and are characterized by the presence of unpaired electrons that allow interaction with a great number of biomolecules, causing their oxidation [,]. Exogenous sources of ROS are smoking, ultraviolet light, heat, ultrasound, ozone, radiation, exhaust fumes, infection, excessive exercise, trauma, and drugs, whereas endogenous sources include products of metabolic pathways and products of immune and connective tissue cells [,]. Antioxidants are substances that can inhibit the action of oxidant species []. There is a physiological fine balance between oxidant activities and antioxidant defenses, but when this equilibrium is disrupted to the advantage of ROS, or to increased ROS activity or to want of antioxidant defenses, the result is oxidative stress []. In this condition ROS operate by creating an adequate environment for phagocytic vacuole and enzymatic digestion, and by mediating cellular signaling. An amplified activity of ROS implies a large spectrum of molecular and cellular damage, such as lipoxidation. This results in covalent binding with proteins, which alters their structure and function []. Some oxidized proteins are difficult to remove by cells and tend to accumulate with aging and in the presence of chronic diseases such as diabetes mellitus.

Several studies have demonstrated a real correlation between oxidative stress and MetS. In fact, in patients suffering from MetS, systemic oxidative stress seems to be more elevated than in healthy controls, and antioxidant defense seems to be decreased, as demonstrated by the diminished rate of Vit C, α-tochopherol, and superoxide dismutase activity in serum, and by increased lipoxidation []. Moreover, obesity is firmly related to oxidative stress–mediated endothelial damage []. An increased caloric excess not balanced by an elevated energy expense leads to an increase in the metabolism of Krébs cycle, generating a ROS excess []. Some HDL-C subfractions present antioxidant activity that is diminished in people suffering from MetS, and this reduction is correlated with systemic oxidative stress and insulin resistance. Furthermore, obese adults with MetS have an increased plasmatic rate of oxidized LDL-C compared with obese patients without this syndrome []. In addition, individuals suffering from MetS present an increased presence of serum lipid peroxide compared with healthy controls [].

Insulin resistance is a condition in which the normal amount of insulin is insufficient to obtain an adequate response from muscular and adipose tissues and from hepatic cells, and this leads to a severe hyperglycemia with deleterious systemic effects [], such as lower intracellular antioxidant defenses [,].

A great number of studies have focused on molecules that can reduce oxidative stress. Recently, for example, some studies have proposed tetracycline as useful in combating oxidative stress in periodontitis and metabolic disorders. In fact, in addition to its antimicrobial effect, it shows antioxidant, anti-inflammatory, proanabolic, immunomodulatory, angiogenetic, and antiapoptotic effects [,]. Decreased yields of oxidative stress were also obtained in the presence of minocycline, which demonstrates its potential role as an adjunctive therapeutic agent in an environment of oxidative stress, such as in periodontal disease and coexisting cardiometabolic pathologies []. Then, the beneficial systemic effect of the antioxidant tempol on apical periodontitis in both control and pathological rats with doxorubicin-elicited cardiomyopathy has also been evidenced []. In addition, a recent study focused on the antioxidant, anti-human immunodeficiency virus, anticarcinogenic, and anti-inflammatory properties of Gomisins G and J extracted from Schisandra chinensis, as it seems to inhibit Porphyromonas gingivalis[]. Furthermore, the proanthocyanidins (PAC), the most abundant flavonoids extracted from red cranberry fruits, have been reported to possess antimicrobial, antiadhesion, antioxidant, and anti-inflammatory properties. The PACs have variable pharmacological and nutraceutical benefits, including improvement of ischemic CVD, prevention of atherosclerosis, and antiarthritic, anticancer, and antimicrobial activities. A recent in vitro study has shown that cranberry PACs may be also potential therapeutic agents for the prevention and management of periodontitis [].

Thymoquinone has also demonstrated a variety of pharmacologic properties, including antihistaminic, antibacterial, antihypertensive, hypoglycemic, anti-inflammatory, and antioxidative activities. Through its anti-inflammatory and antioxidant properties, thymoquinone seems to play an important role in preventing periodontal diseases [].

Aso, S-nitrosoglutathione is a nitric oxide donor that seems to exert antioxidant, anti-inflammatory, and microbicidal actions, and has been demonstrated as a potential drug for the topical treatment of periodontitis []. Finally, because of its anti-inflammatory effects, a novel α-iso-cubebenol isolated from the dried fruit of S. chinensis is considered a novel therapeutic agent to ameliorate periodontitis [].

Hyperglycemia and periodontal diseases

Diabetes, a pathology that is extremely widespread, involves an adulterated homeostasis in the glucose metabolism. Two types of diabetes exist: type 1 diabetes and type 2 diabetes (T2D). Either typology of diabetes, if not controlled, is a risk factor for periodontitis.

The first type affects young people and sets in from childhood. The pathological mechanism is that β cells of Langerhans are unable to produce insulin. Consequently, glucose tends to accumulate in the tissues and the blood. The pathology develops in middle age.

T2D is the most common type and consists in an adulterated insulin cellular response. The onset of T2D is associated with the recruitment of proinflammatory cytokines that are involved in the onset of the disease and related complications such as dyslipidemia and atherosclerosis, contributing to the onset of microvascular and macrovascular complications [,].

Furthermore, patients with periodontitis show a higher risk of ketoacidosis, retinopathy, and neuropathy than do diabetic patients without periodontal disease. In addition, diabetic patients with neurological complications have severe gingivitis compared with diabetic individuals without this complication. Many studies demonstrate a biological relation between diabetes and periodontitis, as tested relations exist between glycated hemoglobin, a diabetic marker, and periodontal parameters, and between plasmatic lipid peroxide, an index of oxidative stress, and periodontal markers [,].

Regarding the mechanism involved in this association, it seems that prolonged hyperglycemia associated with diabetes causes the formation of advanced glycation end-products (AGE). AGEs are physiologically produced by the organism, but in conditions of hyperglycemia or augmented oxidative stress their presence is largely augmented. Exogenous sources of AGE are smoking and browned foods []. AGEs result from reversible glycation reactions, that is, the addition of sugars on the polypeptide chain of proteins, lipids, or nucleic acids. Whether an enzyme attends or not in the glycation reactions, we can have an enzymatic or nonenzymatic glycation []. Furthermore, more resistant proteins such as interstitial or vassal collagen can undergo a nonenzymatic glycation. Several cells, such as endothelial, muscular, and immune cells, possess specific AGE receptors, called RAGE. In fact, a confirmation of AGE activity against periodontal tissue is proved by an elevated presence of RAGE in the periodontal compartment [].

AGEs have deleterious effects for the organism and, being resistant to proteolytic digestion, are difficult to eliminate and tend to accumulate, expressing their harmful effect. AGE-degradation products are chiefly expelled through urine [].

AGEs are substances able to promote cytokine production by macrophages such as TNF-α and IL-6, and to stimulate hepatic secretion of acute-phase proteins such as CRP, fibrinogen, plasminogen activator/inhibitor, and serum amyloid A, also correlated with oral infection and cardiovascular diseases, especially in patients suffering from periodontal diseases [,]. AGEs also promote monocyte migration and increase endothelial permeability, fibroblasts, and muscular cell activity []. In addition, AGEs can bind collagen, which if modified alters basement membrane structure. This gives to an inhibition of oxygen diffusion and tissular oxygenation, diminished waste removal, altered immune mediators, and migration that causes an adulterate inflammatory response, which results in inadequate tissue recovery [].

AGEs also lead to the rapid expansion of energy in the respiratory polymorphonuclear neutrophils (PMN), and this causes increased damage to periodontal tissues [,] and some changes in bone metabolism, especially on repair, also reducing the production of the extracellular matrix [].

The course of the periodontal illness may be due to basic neutrophilic activity that results in an exaggerated immune response and an uncontrolled ROS production. These cause local and peripheral oxidative damage, both directly, through oxidative injury, and indirectly, by means of redox-sensitive gene transcription factors. Nuclear factor kappa B and activating protein 1 are activated, triggering an inflammatory mediator cascade and quick cellular aging [].

Diabetic patients with severe periodontitis have demonstrated a reduced chemotaxis of PMN compared with healthy subjects, or patients with less aggressive periodontitis, and an altered control of apoptosis, which is followed by an accumulation of PMNs in the periodontal tissues and therefore of MMPs and ROS []. The lack of glycemic control has also been associated with alterations in the oral mucosa barrier []. The presence of diabetes seems to increase inflammation in the periodontium, so much so that in the gingival crevicular fluid there are increased levels of PGE2 and IL1-β compared with parodontopatic normoglycemic patients. Moreover, the presence of diabetes mellitus prolongs the inflammatory response to P. gingivalis, causing an increase in the production of TNF-α [].

In synthesis, diabetes appears correlated to the onset of periodontal diseases, both through changes in the host immune response and through anomalies in the collagen metabolism. Recently, different molecules have been investigated for the treatment of diabetes.

Decursin (De), an active component of Angelica gigas, known to exert anticancer and neuroprotective effects, was recently studied for its antiobesity and antidiabetic potential. De treatment resulted in the inhibition of adipocyte differentiation and expression of fatty acid synthase. Administration of De along with a high-fat diet (HFD) significantly reduced the secretion of HFD-induced adipocytokines such as leptin, resistin, and IL-6 [].

Periodontal inflammation can later give rise to a systemic inflammatory state. A recent study suggested a concrete relationship between severe periodontitis and systemic oxidative stress with a reduction in antioxidant systemic defenses. Moreover, it seems that after a periodontal treatment, a transitory inflammatory acute response occurs with a concomitant increase of systemic oxidative markers []. In periodontal diabetic individuals suffering from T2D, there is an augmented rate of oxidative systemic markers and a diminished plasma small-molecule antioxidant capacity []. These patients also show reduced β-cell activity. Therefore, it seems that periodontitis enlarges oxidative stress–mediated dysfunction in β-cells of Langerhans []. Even in periodontitis, the mediated response of the host, in the presence of bacterial infections, results in an increased release of systemic inflammatory mediators. The toll-like receptors (TLR), the receptors for the Ig superfamily (TLR4), and RAGE are implicated in the intensification of the systemic immune response during chronic diseases, such as diabetes and periodontal disease []. Fibroblasts, macrophages, and epithelial cells present in the periodontium possess both RAGE and TLR4 []. It seems that hyperglycemia can promote the expression of these receptors in the periodontal district and increase the response to their ligands, favoring the accumulation of these ligands []. Several studies have shown how chemical and mechanical periodontal treatment may improve glycemic and lipidic control in time []. It seems that periodontal therapy leads to a reduction in the glycated hemoglobin (HbA1c) plasma rate and to a better lipidic profile [,].

Atheroma and periodontitis

Periodontitis also contributes to atherosclerosis and CVD [].

Periodontal pathogens draw lymphocytes in an attempt to stem infection through phagocytosis and killing, causing augmented production of ROS, which ends in a situation of oxidative stress. The presence of ROS promotes chemotaxis and recruits inflammatory mediators from the liver, causing periodontal destruction and favoring the making of endothelial atheromas.

Periodontal pathogens are also able to invade the endothelium and atheromas; in fact, oral infection, being a source of bacteria, is associated with CVD [].

There are several theories that attempt to explain the correlation between periodontitis and CVD [].

One theory is the bacteriological theory, according to which oral pathogens, such as P. gingivalis, invade the systemic circulation and by means of the virulence factors, such as fimbriae, are able to invade the atheromatous plaques. These bacteria can activate endothelial cells through TLR4 and induce apoptosis in these cells, disrupting the mechanisms of cell adhesion.

The inflammatory theory holds that in the course of periodontitis, gingival cells produce inflammatory mediators such as TNF-α, IL-6, PGE-2, and MMPs that locally promote tissue destruction and, once in the circulation, stimulate endothelial cells to produce other mediators such as monocyte chemotactic protein (MCP-1), macrophage colony stimulating factor (MCSF), intercellular adhesion molecule (I-CAM), vascular cell adhesion molecule (V-CAM), P-selectin, and E-selectin. These cytokines accelerate the formation of atheroma.

The third theory is the autoimmune theory, according to which antibodies against bacterial antigens may also react against endothelial protein, causing their destruction and therefore the arterial lesion [].

Systemic and local chronic inflammatory states, such as periodontitis and MetS, are characterized by an elevated presence of acute-phase proteins such as CRP and fibrinogen, which represent a decisive contribution to the insurgence of atherosclerosis and CVD []. Vice-versa, the reduction of periodontal inflammation through plaque control, systemic antibiotics, scaling, and root planning seems to decrease CRP levels in patients suffering from MetS, reducing the risk of CVD [].

Obesity and periodontal damage

The clear relationship between obesity and periodontitis is well documented []. Overweight is a clear risk factor for the onset of T2D and CVD, as well as for respiratory and pressure disorders, osteoarthritis, reproductive abnormality, hepatitis, and some types of cancer []. Adipocytes of fat tissue show the ability to secrete adipocytokines, which seem to be very important in controlling appetite and body weight. One of these cytokines is leptin, which shows a protective role against obesity []. In fact, a condition of clear obesity can be also defined as a situation of leptin resistance with a consequent leptinemia [].

Adipocytes can secrete other cytokines such as adiponectin and resistin. Serum adiponectin remains constant in normal condition but decreases in the presence of diabetes, obesity, insulin resistance, and CVD []. Resistin shows a great proinflammatory role, and from studies conducted on mice it seems associated with insulin resistance [,].

However, many researches affirm that the most important mediator related with obesity and insulin resistance is TNF-α, expressed plentifully in adipose tissue, in obese individuals with severe insulin resistance, and in neoplastic patients [,]. Both TNF-α and Il-6, secreted by adipose cells, seem to trouble intracellular signaling, cause insulin resistance, and stimulate hepatic production of phase acute-phase proteins such as CRP []. Recent murine studies show that the direct infusion of TNF-α or LPS, a TNF-α inductor, can cause a severe insulin resistance; by contrast, mice missing TNF-α or its receptor-codifying gene and submitted to a fatty diet appeared protected against insulin resistance compared with controls in the same condition having genes []. It seems that when insulin binds its tissular receptor, a receptor tyrosine residue on the cytoplasmatic portion is autophosphorylated. The tyrosine residue of the cytoplasmatic insulin-receptor substrate (IRS-1) binding insulin receptor is phosphorylated and causes an intracellular signaling, which leads to the membrane recruitment of intracellular transporter of glucose, GLUT-4, to trap glucose molecules. TNF-α seems to be able to phosphorylate a serine residue on IRS-1 and, subsequently, to inhibit phosphorylation of either the insulin receptor or IRS-1.

Phosphorylation on IRS-1 can also prevent IRS-1 Tyr phosphorylation on adipocytes [].

Other authors have supposed that TNF-α can interact with RNA transcription codifying for IRS-1 and GLUT 4, compromising their stability. It could also prevent Tyr phosphorylation of the insulin receptor, inducing hydrogen peroxide formation [,].

Regarding the relationship between periodontitis and obesity, recent studies have demonstrated how normal-weight persons who participate in sports and physical activity show a decreased incidence of periodontal disease []. Recent studies have also demonstrated how individuals with normal weight showed a lower prevalence of periodontitis, decreased plasma levels of inflammatory markers, and increased insulin sensitivity []. It is of note that during the development of periodontitis, in the attempt to remove causal agents, hyperinflammation is triggered, attracting numerous neutrophilic granulocytes, which, through the production of digestive enzymes during phagocytosis and killing, favor the production of ROS and the consequent constant release of inflammatory mediators with direct bacterial action damage on periodontal tissue. In addition, it seems that in people suffering from periodontitis, pathogen bacteria, endotoxins, and inflammatory mediators cause leucocytosis and increased lipidic metabolism, with an increase of cholesterol and hepatic triglycerides favoring the risk of CVD [,]. Furthermore, a systemic increase of oxidative stress caused by periodontal disease seems to promote LDL oxidation []. LDLs are cholesterol carriers with proinflammatory and pro-atherogenic activity. Periodontal treatment by scaling and root planning, and dental care instruction seem to reduce the circulating oxLDL level and, consequentially, the risk of the onset of CVD []. In addition, leptin seems to be involved in bone metabolism []. In recent murine studies, animals missing the leptin-codifying gene developed pronounced obesity. At the same time, mice with an adulterated leptin receptor presented overweight and T2D []. Moreover, children lacking leptin showed severe obesity, but when exogenous leptin was given, obesity decreased considerably. In periodontal individuals, serum leptin seems increased [,]. On the contrary, leptin in gingival fluid shows decreased levels, especially in the presence of aggressive and advanced periodontitis, advising of a protective leptin role, although this topic is still disputed [].

There are no relevant data about the rate of adiponectin in gingival sulcular fluid, but in vitro it has been shown to be able to inhibit osteoclast activity induced by LPS. Thus, it seems to have anti-inflammatory and protective action against the progression of periodontitis and a predictive role for T2D [].

In synthesis a recent systematic review confirmed a positive association between obesity and periodontal diseases across multiple studies and populations from around the world, but this relationship needs more investigation []. The relationship appears clarified when the treatment of obesity is also analyzed. According to a recent study by Lakkis et al. [] some parodontopathic obese patients who underwent bariatric surgery showed weight loss and fat-mass loss after surgery, and an improvement in inflammatory conditions with the reduction of circulating markers and hyperglycemia. In addition, it seems that patients who undergo bariatric surgery show an improved response to non-surgical periodontal therapy compared with obese parodontopathic patients not treated with bariatric surgery. These data contrast with those reported by a study conducted by Zouza et al. [], who showed that non-surgical periodontal treatment allows a reduction of all clinical parameters of periodontal inflammation, both in obese patients and in normal-weight subjects, arguing that obesity does not negatively affect the success of periodontal therapy. Some studies also try to magnify this relationship. A recent meta-analysis indicated statistically significant associations between periodontitis and body mass index: the category of obese subjects showed an odds ratio (OR) of 1.81 the category of overweight subjects showed an OR of 1.27, and obese and overweight combined showed an OR of 2.13. Although there is insufficient evidence to provide guidelines to clinicians on the clinical management of periodontitis in overweight and obese individuals [], these studies evidence a clear relationship.

Bariatric surgical procedures, including the laparoscopic adjustable gastric band (LAGB), are currently the only effective treatments for morbid obesity. The LAGB exerts its effects on satiety, reducing food intake and body weight by the modulation of both neural and hormonal responses, with the latter involving an elevation of meal-related levels of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) []. The brain, mainly the hypothalamus, is the organ responsible for maintaining the balance between food intake and energy expenditure by receiving peripheral signals from adipose tissue (adipose signals) and responding to them. Several neuropeptides are involved in this process, such as neuropeptide Y (NPY), which increases food consumption and decreases energy expenditure. NPY, acting through specific receptors, plays an important role in several physiological functions, including cardiovascular homeostasis and regulation of sympathetic nervous system activity. It mediates stress-induced obesity in adult male mice by activating its Y2 receptor (Y2R) in visceral adipose tissue. In fact, the expression of NPY is correlated with body weight changes, rather than with the presence of type 2 diabetes [].

Chemerin is a recently identified adipokine that is highly expressed in liver and adipose tissue and is associated with adiposity, insulin resistance, MetS risk factors, and degree of nonalcoholic fatty liver. Importantly, chemerin is thought to regulate adipogenesis and metabolic homeostasis in murine and human adipocytes. Additionally, it modulates the innate immune system through its binding to the orphan G-protein coupled receptor chemokine-like receptor 1. It also modulates chemotaxis of immature dendritic cells and macrophages. Recent studies have associated chemerin with several inflammatory markers in obesity and T2D. Thus, chemerin is considered as a candidate in linking inflammation to obesity-related diseases [].

Weight gain and the appearance of insulin resistance go hand in hand and are thought to be caused by an abnormal adipokine release by visceral fat. Adipocytes release retinol binding protein 4 (RBP4) and visfatin, and their plasma levels are elevated in individuals with abdominal obesity. The observation that RBP4 induces insulin resistance by interfering with insulin receptor substrate 1 adds to the list of plasma abnormalities that link obesity with the development of T2D. At levels found in vivo these adipokines interfere with insulin receptor substrate 1 regulation (short contact) and induce its degradation (prolonged contact) []. Another treatment for obesity consists of behavioral weight management interventions, which consistently produce 8% to 10% reductions in body weight, although most subjects regain weight after treatment ends. One strategy for extending the effects of behavioral interventions has been the provision of extended care, which is a viable and efficacious solution to addressing the long-term maintenance of lost weight. Given the chronic disease nature of obesity, extended care may be necessary for long-term health benefits [].

About the modulation of both neural and hormonal responses, periodontitis has also been associated with preterm birth. For many years, the incidence of preterm birth has not decreased in developed countries despite the promotion of public health programs. Many risk factors have been identified, including ethnicity, age, tobacco, and infection. However, the causes of almost 50% of preterm births remain unknown. As periodontal diseases are highly prevalent and negatively influence general health, worsening the incidence of CVD and diabetes, they have also been suspected to increase the rate of preterm birth. However, data on this topic remain contradictory. Physicians/obstetricians can identify women at risk of preterm birth and refer these patients to dentists for periodontal examination and treatment to limit adverse pregnancy outcomes [].

Hypertension and periodontitis

Hypertensives suffering from MetS show increased oxidative stress and compromised antioxidant activity in plasma and cells [,]. In addition, obesity and overweight are strictly related to hypertension. In fact, weight loss determines a diminished blood pressure independent from sodic diet []. Moreover, hyperglycemia and hypertension are strictly related. Hyperglycemia provokes an increased stimulation of a sympathetic nervous system that causes vasoconstriction and increased sodium reabsorption with consequent water attraction and insurgence of hypertension, which damages the endothelium integrity of vessels [].

Augmented endothelial permeability allows the passage of lipoproteins and platelet-derived growth factors (PDGF), which give rise to the proliferation of muscular smooth cells in the intima, which occludes vessel lumen and causes embolia, hypoxia, and consequent cellular death [].

It also seems that periodontitis can influence some types of hypertension []. Several studies have taken into consideration the relationship between hypertension and periodontitis, although an association between periodontal disease measures and incident hypertension in cohort studies has not yet been evidenced. In a sample of 31,543 participants of the Health Professionals’ Follow-Up Study, based on a prospective cohort of 40- to 75-year-old men at baseline, with no prior hypertension history and complete baseline information on oral health, an incidence of 10,828 cases of hypertension over 20 years of follow-up was identified, with no significant association between incident hypertension and periodontal disease [].

In a study population of 182 adults, a multivariate analysis showed no association between severe periodontitis and hypertension history (OR = 0.99; 95% CI: 0.40-2.48). Severe periodontitis was associated with high blood pressure, with an OR of 2.93 (95% CI: 1.25-6.84) after adjusting for age, gender, smoking, and binge drinking. This association was stronger when restricted to those with hypertension or taking antihypertensive medications (OR = 4.20; 95% CI: 1.28-13.80), suggesting that periodontitis may contribute to poor blood pressure control among older adults [].

Although statistical evidence is lacking, a clinical relation between high blood pressure and aggressive periodontitis has been deduced, as patients with poor oral hygiene have higher blood pressure problems than do healthy subjects with good oral hygiene condition []. Regarding the biological mechanism of this relationship, a recent study evaluated endothelial function in patients with periodontitis. Circulating levels of CRP and IL-6 were significantly higher in the periodontitis subjects with hypertension, than in the control group. Periodontal therapy seems to reduce serum concentrations of CRP and IL-6 [].

Metabolic syndrome and periodontitis

As already mentioned, MetS is a syndrome characterized by several signs that together seriously compromise the health of an individual. It is clear that the common denominator of the member pathologies of MetS is oxidative stress and the consequent hyperinflammation that primes chain interactions and leads to grave systemic complications, such as CVD, or local complications, such as periodontitis.

MetS allows a pro-oxidative state in periodontal tissue, altering antioxidant defense mechanisms. This adversely affects tissular response against bacterial plaque attack. On the contrary, periodontitis, being a great source of oxidative markers, promotes the onset of insulin resistance and MetS in a vicious circle []. Chronic inflammation during old age periodontitis causes increased neutrophil defense activity, which involves increased oxidative activity, resulting in peroxidation and oxidative stress. In fact, both MetS and periodontitis show increased serum rates of oxidative stress markers [,].

Regarding the oxidative stress markers found in periodontitis, individuals with periodontal disease exhibit a significant increase in the activities of oxidative stress markers. The increase in glutathione peroxidase may represent possible antioxidant compensation in detoxification reactions of organic peroxides produced during oxidative stress in gingival tissue. Since glutathione S-transferase (GST) has a direct role in the neutralization of hydroperoxides derived from the lipoperoxidation processes, increases in GST activities are probably related to the oxidative stress caused by the periodontal inflammatory process. GST comprises a group of enzymes that are also able to detoxify a variety of compounds, including xenobiotics derived from pathogenic microorganisms. Hence, increases in GST activities are excellent indicators of endogenous detoxification from exogenous sources. Myeloperoxidase activity in gingival tissue has shown a significant increase in patients with periodontal disease when compared with controls: this seems indicative of a chronic inflammatory process also reflected at a systemic level. A significant increase in oxidized glutathione (GSSG) concentrations has been detected in periodontitis patients, which is a clear biomarker of oxidative stress detected in inflammatory processes linked to periodontitis. Consistent with the results for GSSG, tissue lipoperoxidation, measured as thiobarbituric acid reactive substances, seems to increase in the gingival tissue of periodontitis []. Periodontal diseases seem related to pathologies and conditions characterized by high oxidative stress and by the presence of AGE, such as diabetes and physiologic aging. AGEs are able to favor chemotaxis and the production of proinflammatory mediators, to inhibit fibroblasts and osteoblasts, and to accelerate periodontal damage directly or binding their receptors RAGE []. Periodontitis is strictly correlated to hyperglycemia; in fact, it is also considered the sixth complication of diabetes mellitus []. Predialysis and hemodialysis in chronic kidney diseases are also associated with a higher prevalence of severe periodontitis compared with healthy individuals.

Chronic kidney failure is a clinical syndrome due to the slow, progressive, and irreversible loss of the glomerular filtration rate, and may be associated with several oral manifestations, such as xerostomia, uremic stomatitis, and periodontitis, diagnosed as clinical attachment loss.

Recent studies have shown an association between high levels of CRP and IL-6 and periodontitis, an association that decreases after periodontal treatment. Due to this association with the systemic inflammatory response, chronic periodontitis has recently been included as a nontraditional risk factor for chronic kidney failure [].

In synthesis, metabolic alterations related to MetS component diseases cause an augmented response to bacterial plaque, which favors periodontitis insurgence.

It has been pointed in many studies out how periodontal treatment can reduce inflammatory mediators related to endothelial and cardio-circulatory dysfunctions []. A very recent work reported a real relationship between periodontitis and MetS, especially in women, while abdominal obesity was the largest contributory factor in both genders []. On the contrary, another new work about MetS and periodontal diseases and caries did not find a strong association between MetS and periodontal infections [].

Odontologic management of metabolic patients

Dentists play a key role in the precocious diagnosis of MetS and its local complications such as periodontitis.

The diet

The capability of an aliment to raise the level of insulin in the blood, compared with a reference food such as glucose or with bread, is known as the glycemic index (GI) [,]. It seems that the greater the consumption of foods with higher GI, the wider will be the risk of developing insulin resistance, endothelial dysfunction, and CVD. In fact, taking foods with high GI results in the quick increase of glycemia and, consequently, in insulinemia in the attempt to restore glycemia; the secretion of insulin continues in spite of reestablished euglycemia, so as to conduce to a hypoglycemic state [].

Hypoglycemia causes the secretion of counter-regulatory hormones, such as corticosteroids, glucagon, and epinephrine, with the consequent reduction of lipolysis, glycogenolysis, fatty-acid release, and vasoconstriction that can bring hypertension []. Repeating this process may lead to β-cell damage and T2D. A poor diet of foods with high GI is basilar to reduce at least the risk factor. Several studies have demonstrated the relationship between the intake of foods with high GI and MetS []. It is ideal to consume nutrient-rich, high-fiber food such as fruits and vegetables to maintain good health [].

Good oral health

Maintaining good oral health is fundamental for individuals who suffer from MetS and have a tendency to develop CVD. Progressive loss of teeth produces a variation of diet, with an increased intake of foods with great caloric rate, saturated fats, trans fatty acids, and cholesterol, and less intake of fruits and vegetables rich in vitamins and fibers, folate, and potassium []. The use of partial or total removable prosthesis does not seem to cover the masticatory efficacy of natural teeth [], but fixed dental prosthetic devices and prosthetic overimplants seem to improve dietary practices []. Drugs used in patients suffering from MetS may give rise to oral collateral effects and could interact with drugs prescribed in dentistry [].

Sibutramine, used in weight control, may cause hypertension and tachycardia, and if associated with opioids, may provoke serotonin syndrome with confusion, palpitations, and loss of consciousness; the concomitant use of erythromycin and clarithromycin could lead to toxicity. Ace inhibitors, if prescribed in association with nonsteroidal anti-inflammatory agents, may be inhibited in their activity. Statin medications in association with erythromycin and clarithromycin could lead to renal failure []. As diabetes is a member of MetS, dentists may evaluate glycemic value before starting dental therapeutic measures [].

The oral complications of diabetes are candidiasis, xerostomia, burning mouth syndrome, gingivitis, oral acute infections, and, clearly, periodontal diseases – all diseases treated with dentistry []. Therefore, the role of dentists in the diagnosis, therapy, and management of metabolic patients is fundamental [], and an improvement of collaboration among dentists, cardiologists, endocrinologists, and dietists is needed to promote the multidisciplinary therapeutic approach to this syndrome.

Conclusions

In this review, we tried to explain how various pathologies associated with MetS can be related to and favor the onset of CVD and, especially, periodontitis. Oxidative stress seems to be the chief suspect in ethiopathogenesis of periodontal disease; for this, the use of drugs with antioxidative activity or anti-AGE is the subject of research. Chemical agents such as pyridoxamine, metformin, and nefedipine, with their antioxidative ability, could be used. Other anti-AGE drugs are modified tetracyclines, such as doxycycline, which have anti-inflammatory and antioxidative effects. However, this argument needs more clarity, and the search for answers goes encouraged. As we await more results, we can increase prevention in at-risk individuals by advising lifestyle changes and prescribing a balanced diet to control body weight, hyperlipidemia, and hypertension; advising a stop to smoking and the maintenance good oral hygiene in periodontal therapy; and finally, establishing pharmacological and eating control of diabetes.

The role of dentists in the diagnosis, therapy, and management of metabolic patients is fundamental, but an improvement of collaboration among dentists, cardiologists, endocrinologists, dietists, etc., is needed.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

EM coordinated the revision of literature; AM described main results; LP revised the literature; SM coordinated the introduction; RG coordinated the discussion paragraph; STetè revised the bibliography,; AB reviewed the literature suggested by the reviewers; STecco conceived the study, analyzed the data, coordinated the discussion, and draft the manuscript; GM conceived the study and coordinated the analysis of data. All authors read and approved the final manuscript.

Acknowledgements

The authors want to acknowledge the Department of Health Science of the University of L’Aquila and S.P.I. (Professional Editing Service; http://www.proof-editing.com) for the revision of the english language.

References

  • Andriankaja OM, Sreenivasa S, Dunford R, DeNardin E. Association beteen metabolic syndrome and periodontal disease. Aust Dent J. 2010;55:252–259. doi: 10.1111/j.1834-7819.2010.01231.x. [PubMed] [CrossRef[]
  • Armitage GC. Diagnosis of periodontal disease. J periodontology. 2003;74:1237–1247. [PubMed[]
  • Zadik Y, Bechor R, Shochat Z, Galor S. Ethnic origin and alveolar bone loss in Israeli adults. Refuat Hapeh Vehashinayim. 2008;25(2):19–22. [PubMed[]
  • The American Academy of Periodontology. Proceedings of the World Workshop in Clinical Periodontics. The American Academy of Periodontology, Chicago; 1989. pp. I/23–I/24. []
  • Ammons WF, Schectman LR, Page RC. Host tissue response in chronic periodontal disease. 1. The normal periodontium and clinical manifestations of dental and periodontal disease in the marmoset. J Periodontal Res. 1972;7(2):131–143. doi: 10.1111/j.1600-0765.1972.tb00638.x. [PubMed] [CrossRef[]
  • Page RC, Schroeder HE. Pathogenesis of inflammatory periodontal disease. A summary of current work. Laboratory investigation; a journal of technical methods and pathology. 1976;34(3):235–249. [PubMed[]
  • Sambunjak D, Nickerson JW, Poklepovic T, Johnson TM, Imai P, Tugwell P, Worthington HV. Flossing for the management of periodontal diseases and dental caries in adults. In Johnson, Trevor M. Cochrane Database Syst Rev. 2011;7(12):CD008829. [PubMed[]
  • Hasturk H, Nunn M, Warbington M, Van Dyke TE. Efficacy of a Fluoridated Hydrogen Peroxide-Based Mouthrinse for the Treatment of Gingivitis: A Randomized Clinical Trial. J Periodontol. 2004;75(1):57–65. doi: 10.1902/jop.2004.75.1.57. [PubMed] [CrossRef[]
  • Di Paola R, Briguglio F, Paterniti I, Mazzon E, Oteri G, Militi D, Cordasco G, Cuzzocrea S. Emerging role of PPAR-β/δ in inflammatory process associated to experimental periodontitis. Mediators Inflamm. 2011;7:87–159. [PMC free article] [PubMed[]
  • Dutzan N, Vernal R, Vaque JP, García-Sesnich J, Hernandez M, Abusleme L, Dezerega A, Gutkind JS, Gamonal J. Interleukin-21 Expression and its Association with Pro-Inflammatory Cytokines in Untreated Chronic Periodontitis Patients. J Periodontol. 2012;83(7):948–954. doi: 10.1902/jop.2011.110482. [PubMed] [CrossRef[]
  • Souza PP, Palmqvist P, Lundberg P, Lundgren I, Hänström L, Souza JA, Conaway HH, Lerner UH. Interleukin-4 and interleukin-13 inhibit the expression of leukemia inhibitory factor and interleukin-11 in fibroblasts. Mol Immunol. 2012;49(4):601–610. doi: 10.1016/j.molimm.2011.10.009. [PubMed] [CrossRef[]
  • Reaven GM. Banting lecture 1988. Role of insulin resistance in human disease. Diabetes. 1998;37:1595–1607. [PubMed[]
  • Abhishek G, Gupta V. Metabolic Syndrome: what are risks for humans? BioScience Trend. 2010;4:204–212. [PubMed[]
  • Aquilante CL, Kosmiski LA, Knusten SD, Zineh I. Relationship between plasma resistin concentrations, inflammatory chemokines and components of the Metabolic Syndrome in adults. Metabolism. 2008;5(Suppl 4):494–501. [PubMed[]
  • Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III). Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults. JAMA. 2001;285(19):2486–2497. doi: 10.1001/jama.285.19.2486. [PubMed] [CrossRef[]
  • Metabolic Syndrome.  ,  ;  . http://www.heart.org/HEARTORG/Conditions/More/MetabolicSyndrome/About-Metabolic-Syndrome_UCM_301920_Article.jsp[]
  • Bullon P, Morillo JM, Ramirez-Tortosa MC, Quiles JL, Newman HN, Battino M. Metabolic Syndrome and Periodontitis: Is Oxidative Stress a Common Link? J Dent Res. 2009;88:503–518. doi: 10.1177/0022034509337479. [PubMed] [CrossRef[]
  • Kahn R. Metabolic syndrome: what is the clinical usefulness? Lancet. 2008;371:1891–1893. [PubMed[]
  • Reaven GM. The metabolic syndrome: is this diagnosis necessary? Am J Clin Nutr. 2006;83:1237–1247. erratum in Am J Clin Nutr 2006, 84:1253. [PubMed[]
  • Ceriello A, Motz E. Is oxidative stress the pathogenic mechanism underlying insulin resistance, diabetes, and cardiovascular disease? The common soil hypothesis revisited. Arterioscler Thromb Vasc Biol. 2004;24(Suppl5):816–823. [PubMed[]
  • Iain LC C, Matthews JB. The role of reactive oxygen and antioxidant species in periodontal tissue destruction. Periodontol. 2007;43:160–232. doi: 10.1111/j.1600-0757.2006.00178.x. [PubMed] [CrossRef[]
  • Sgolastra F, Petrucci A, Gatto R, Monaco A. Efficacy of Er:YAG laser in the treatment of chronic periodontitis: systematic review and meta-analysis. Lasers Med Sci. 2011;27(3):661–673. [PubMed[]
  • Halliwell B, Gutteridge JMC, Cross CE. Free radicals, antioxidants and human disease: where are we now? J Lab Clin Med. 1992;119:598–617. [PubMed[]
  • Armutcu F, Ataymen M, Atmaca H, Gurel A. Oxidative stress markers, C-reactive protein and heat shock protein 70 levels in subjects with metabolic syndrome. Clin Chem Lab Med. 2008;46(Suppl 6):785–790. [PubMed[]
  • Perticone F, Ceravolo R, Candigliota M, Ventura G, Iacopino S, Sinopoli F, Mattioli PL. Obesity and body fat distribution induce endothelial dysfunction by oxidative stress: protective effect of vitamin C. Diabetes. 2001;50(Suppl 1):159–165. [PubMed[]
  • Maddux BA, See W, Lawrence JC Jr, Goldfine AL, Goldfine ID, Evans JL. Protection against oxidative stress-induced insulin resistance in rat L6 muscle cells by micromolar concentrations of α-lipoic acid. Diabetes. 2001;50:404–410. doi: 10.2337/diabetes.50.2.404. [PubMed] [CrossRef[]
  • Hansel B, Giral P, Nobecourt E, Chantepie S, Bruckert E, Chapman MJ. et al.Metabolic syndrome is associated with elevated oxidative stress and dysfunctional dense high-density lipoprotein particles displaying impaired antioxidative activity. J Clin Endocrinol Metab. 2004;89:4963–4971. doi: 10.1210/jc.2004-0305. [PubMed] [CrossRef[]
  • Sharma P, Mishra S, Ajmera P, Mathur S. Oxidative stress in metabolic syndrome. Indian J Clin Biochem. 2005;20:145–149. doi: 10.1007/BF02893061. [PMC free article] [PubMed] [CrossRef[]
  • Di Filippo C, Verza M, Coppola L, Rossi F, D’Amico M, Marfella R. Insulin resistance and postprandial hyperglycemia the bad companionsin natural history of diabetes: effects on health of vascular tree. Curr Diabetes Rev. 2007;3:268–273. doi: 10.2174/157339907782330012. [PubMed] [CrossRef[]
  • Tilg H, Moschen AR. Inflammatory mechanisms in the regulation of insulin resistance. Mol Med. 2008;14:222–231. [PMC free article] [PubMed[]
  • Bruce CR, Carey AL, Hawley JA, Febbraio MA. Intramuscular heat shock protein 72 and heme oxygenase-1 mRNA are reduced in patients with type 2 diabetes: evidence that insulin resistance is associated with a disturbed antioxidant defence mechanism. Diabetes. 2003;52:2338–2345. doi: 10.2337/diabetes.52.9.2338. [PubMed] [CrossRef[]
  • Koyama H, Shoji T, Yokoyama H, Motoyama K, Mori K, Fukumoto S. et al.Plasma level of endogenous secretory RAGE is associated with components of the metabolic syndrome and atherosclerosis. Arterioscler Thromb Vasc Biol. 2005;25:2587–2593. doi: 10.1161/01.ATV.0000190660.32863.cd. [PubMed] [CrossRef[]
  • Soory M. A Role for Non-Antimicrobial Actions of Tetracyclines in Combating Oxidative Stress in Periodontal and Metabolic Diseases: A Literature Review. The Open Dentistry Journal. 2008;2:5–12. doi: 10.2174/1874210600802010005. [PMC free article] [PubMed] [CrossRef[]
  • Sapadin AN, Fleischmajer R. Tetracyclines: Non antibiotic properties and their implications. J Am Acad Dermatol. 2006;54:258–265. doi: 10.1016/j.jaad.2005.10.004. [PubMed] [CrossRef[]
  • Tilakaratne A, Soory M. Osteoblastic responses to LPS, glucose-oxidised LDL and minocycline: therapeutic targets for periodontal and cardiometabolic diseases. Recent Pat Endocr Metab Immune Drug Discov. 2012;6(1):73–84. doi: 10.2174/187221412799015281. [PubMed] [CrossRef[]
  • Brilhante Wolle CF, de Aguiar Zollmann L, Etges A, Vitalis GS, Leite CE, Campos MM. Effects of the antioxidant agent tempol on periapical lesions in rats with doxorubicin-induced cardiomyopathy. J Endod. 2012;38(2):191–195. doi: 10.1016/j.joen.2011.11.007. [PubMed] [CrossRef[]
  • Ryu EY, Park SY, Kim SG, da Park J, Kang JS, Kim YH, Seetharaman R, Choi YW, Lee SJ. Anti-inflammatory effect of heme oxygenase-1 toward Porphyromonas gingivalis lipopolysaccharide in macrophages exposed to gomisins A, G, and J. J Med Food. 2011;14(12):1519–1526. doi: 10.1089/jmf.2011.1656. [PMC free article] [PubMed] [CrossRef[]
  • Feghali K, Feldman M, La VD, Santos J, Grenier D. Cranberry Proanthocyanidins: Natural Weapons against Periodontal Diseases. J Agric Food Chem. 2011;29: . [PubMed[]
  • Ozdemir H, Kara MI, Erciyas K, Ozer H, Ay S. Preventive effects of thymoquinone in a rat periodontitis model: a morphometric and histopathological study. J Periodontal Res. 2012;47(1):74–80. doi: 10.1111/j.1600-0765.2011.01406.x. [PubMed] [CrossRef[]
  • de Menezes AM, de Souza GF, Gomes AS, de Carvalho Leitão RF, de Albuquerque Ribeiro R, de Oliveira MG, de Castro Brito GA. S-nitrosoglutathione decreases inflammation and bone resorption in experimental periodontitis in rats. J Periodontol. 2012;83(4):514–521. doi: 10.1902/jop.2011.110332. [PubMed] [CrossRef[]
  • Park SY, da Park J, Kim YH, Kim Y, Choi YW, Lee SJ. Schisandra chinensis α-iso-cubebenol induces heme oxygenase-1 expression through PI3K/Akt and Nrf2 signaling and has anti-inflammatory activity in Porphyromonas gingivalis lipopolysaccharide-stimulated macrophages. Int Immunopharmacol. 2011;11(11):1907–1915. doi: 10.1016/j.intimp.2011.07.023. [PubMed] [CrossRef[]
  • Bjelland S, Bray P, Gupta N. et al.Dentists, diabetes and periodontitis. Austr Dent Journ. 2002;47:202–207. doi: 10.1111/j.1834-7819.2002.tb00329.x. [PubMed] [CrossRef[]
  • Mustapha IZ, Debrey S, Oladubu M, Ugarte R. Markers of systemic bacterial exposure in periodontal disease and cardiovascular disease risk: a systematic review and meta-analysis. J Periodontol. 2007;78(12):2289–2302. doi: 10.1902/jop.2007.070140. [PubMed] [CrossRef[]
  • Iwamoto Y, Nishimura F, Nakagawa M, Sugimoto H, Shikata K, Makino H. et al.The effect of antimicrobial periodontal treatment on circulating tumor necrosis factor-alpha and glycated hemoglobin level in patients with type 2 diabetes. J Periodontol. 2001;72:774–778. doi: 10.1902/jop.2001.72.6.774. [PubMed] [CrossRef[]
  • Grossi SG, Skrepcinski FB, DeCaro T, Robertson DC, Ho AW, Dunford RG. et al.Treatment of periodontal disease in diabetics reduces glycated hemoglobin. J Periodontol. 1997;68:713–719. doi: 10.1902/jop.1997.68.8.713. [PubMed] [CrossRef[]
  • Pietropaoli D, Tatone C, D’Alessandro, Monaco A. Possible involment of advanced glycation end products in periodontal diseases. B Int Journ of immunopathol and pharmacol. 2010;23(Suppl 3):683–691. [PubMed[]
  • Katz J, Bhattacharyya I, Farkhondeh-Kish F, Perez FM, Caudle RM, Heft MW. Expression of the receptor of advanced glycation end products in gingival tissues of type 2 diabetes patients with chronic periodontal disease: a study utilizing immunohistochemistry and RT-PCR. J Clin Periodontol. 2005;32:40–44. doi: 10.1111/j.1600-051X.2004.00623.x. [PubMed] [CrossRef[]
  • Bohlender JM, Franke S, Stein G, Wolf G. Advanced glycation end products and the kidney. Am J Physiol Renal Physiol. 2005;289:F645–F659. doi: 10.1152/ajprenal.00398.2004. [PubMed] [CrossRef[]
  • Wong RK, Pettit AI, Quinn PA. et al.Advanced glycation end products stimulate an enhanced neutrophil respiratory burst mediated through the activation of cytosolic phospholipase A2 and generation of arachidonic acid. Circulation. 2003;108:1858–1864. doi: 10.1161/01.CIR.0000089372.64585.3B. [PubMed] [CrossRef[]
  • Preshaw PM, Alba AL, Herrera D. et al.Periodontitis and diabetes: a two way relationship. Diabetologia. 2012;55:21–31. doi: 10.1007/s00125-011-2342-y. [PMC free article] [PubMed] [CrossRef[]
  • Allen EM, Mattews JB, O’Halloran DJ. et al.Oxidative and inflammatory status in type 2 diabetes patients with periodontitis. J Clin Peridontol. 2011;38:894–901. doi: 10.1111/j.1600-051X.2011.01764.x. [PubMed] [CrossRef[]
  • Amir J, Waite M. et al.The role of hyperglycemia in mechanisms of exacerbated inflammatory responses within the oral cavity. Cell Immunol. 2011;272:45–52. doi: 10.1016/j.cellimm.2011.09.008. [PMC free article] [PubMed] [CrossRef[]
  • Hwang JT, Kim SH, Hur HJ, Kim HJ, Park JH, Sung MJ, Yang HJ, Ryu SY, Kim YS, Cha MR, Kim MS, Kwon DY. Decursin, an Active Compound Isolated from Angelica gigas, Inhibits Fat Accumulation, Reduces Adipocytokine Secretion and Improves Glucose Tolerance in Mice Fed a High-Fat Diet. Phytother Res. 2012;26(5):633–638. doi: 10.1002/ptr.3612. [PubMed] [CrossRef[]
  • D’Aiuto F, Nibali L, Parkar M, Patel K. et al.Oxidative stress, systemic inflammation and severe periodontitis. J Dent Res. 2010;89:1241–1246. doi: 10.1177/0022034510375830. [PMC free article] [PubMed] [CrossRef[]
  • Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Are oxidative stress-activated signaling pathways mediators of insulin resistance and beta-cell dysfunction? Diabetes. 2003;52:1–8. doi: 10.2337/diabetes.52.1.1. [PubMed] [CrossRef[]
  • D’Aiuto F, Parkar M, Andreou G. et al.Periodontitis and systemic inflammation: control of the local infection is associated with a reduction in serum inflammatory markers. J Dent Res. 2004;83:156–160. doi: 10.1177/154405910408300214. [PubMed] [CrossRef[]
  • Behle JH, Sedaghatfar MH, Demmer RT. et al.Heterogeneity of systemic inflammatory responses to periodontal therapy. J Clin Periodontol. 2009;36:287–294. doi: 10.1111/j.1600-051X.2009.01382.x. [PMC free article] [PubMed] [CrossRef[]
  • D’Aiuto F, Nibali L, Parkar M, Suvan J, Tonetti MS. Short-term effects of intensive periodontal therapy on serum inflammatory markers and cholesterol. J Dent Res. 2005;84:269–273. doi: 10.1177/154405910508400312. [PubMed] [CrossRef[]
  • Offenbacher S, Beck JD, Moss K. et al.Results from the Periodontitis and Vascular Events (PAVE) Study: a pilot multicentered, randomized, controlled trial to study effects of periodontal therapy in a secondary prevention model of cardiovascular disease. J Periodontol. 2009;80:190–201. doi: 10.1902/jop.2009.080007. [PMC free article] [PubMed] [CrossRef[]
  • Navarro-Sanchez AB, Faria-Almeida R, Bascones-Martinez A. Effect of non-surgical periodontal therapy on clinical and immunological response and glycaemic control in type 2 diabetic patients with moderate periodontitis. J Clin Periodontol. 2007;34:835–843. doi: 10.1111/j.1600-051X.2007.01127.x. [PubMed] [CrossRef[]
  • Offenbacher S, Madianos PN, Champagne CM, Southerland JH, Paquette DW, Williams RC. et al.Periodontitis-atherosclerosis syndrome: an expanded model of pathogenesis. J Periodont Res. 1999;34:346–352. doi: 10.1111/j.1600-0765.1999.tb02264.x. [PubMed] [CrossRef[]
  • Libby P, Ridker P, Maseri A. Inflammation and atherosclerosis. Circulation. 2002;105:1135–1143. doi: 10.1161/hc0902.104353. [PubMed] [CrossRef[]
  • López NJ, Quintero A, Casanova PA, Ibieta CI, Baelum V, López R. Effects of Periodontal Therapy on Systemic Markers of Inflammation in Patients With Metabolic Syndrome: A Controlled Clinical Trial. J Periodontol. 2012;83(3):267–278. doi: 10.1902/jop.2011.110227. [PubMed] [CrossRef[]
  • Pischon N, Heng JP. et al.Obesity, inflammation and periodontal disease. J Dent Res. 2007;86(suppl. 5):400–409. [PubMed[]
  • El-Haschimi K, Pierroz DD, Hileman SM, Bjorbaek C, Flier JS. Two defects contribute to hypothalamic leptin resistance in mice with diet induced obesity. J Clin Invest. 2000;105:1827–1832. doi: 10.1172/JCI9842. [PMC free article] [PubMed] [CrossRef[]
  • Nishimura F, Murayama Y. Periodontal inflammation and Insulin resistance, lessons from obesity. J Dent Res. 2001;80((suppl.8):1690–1694. [PubMed[]
  • Hotamisligil GS, Amer P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-at in human obesity and insulin resistance. J Clin Invest. 1995;95:2409–2415. doi: 10.1172/JCI117936. [PMC free article] [PubMed] [CrossRef[]
  • Lundholm K, Holm G, Schersten T. Insulin resistance in patients with cancer. Cancer Res. 1997;38:4665–4670. [PubMed[]
  • Rotter V, Nagaev I, Smith U. Interleukin-6 (IL-6) induces insulin resistance in 3 T3-L1 adipocytes and is, like IL-8 and tumor necrosis factor-alpha, overexpressed in human fat cells from insulin-resistant subjects. J Biol Chem. 2003;278:45777–45784. doi: 10.1074/jbc.M301977200. [PubMed] [CrossRef[]
  • Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS. Protection from obesity-induced insulin resistance in mice lacking TNF-at function. Nature.  ;389:610–614. [PubMed[]
  • Rui L, Aguirre V, Kim JK, Shulman GI, Lee A, Corbould A. et al.Insulin/IGF-1 and TNF-ot stimulate phosphorylation of IRS-1 at inhibitory Ser307 via distinct pathways. J Clin Invest. 2001;107:181–189. doi: 10.1172/JCI10934. [PMC free article] [PubMed] [CrossRef[]
  • Hansen LL, Ikeda Y, Olsen GS, Busch AK, Mosthaaf L. Insulin signaling is inhibited by micromolar concentrations of H202. Evidence for a role of H202 in tumor necrosis factor a-mediated insulin resistance. J Biol Chem. 1999;274:25078–25084. doi: 10.1074/jbc.274.35.25078. [PubMed] [CrossRef[]
  • Merchant AT, Pitiphat W, Rimm EB, Joshipura K. Increased physical activity decreases periodontitis risk in men. Eur J Epidemiol. 2003;18:891–898. doi: 10.1023/A:1025622815579. [PubMed] [CrossRef[]
  • Pischon T, Hankinson SE, Hotamisligil GS, Rifai N, Rimm EB. Leisure-time physical activity and reduced plasma levels of obesityrelated inflammatory markers. Obes Res. 2003;11:1055–1064. doi: 10.1038/oby.2003.145. [PubMed] [CrossRef[]
  • Al-Zahrani MS, Borawski EA, Bissada NF. Periodontitis and three health-enhancing behaviors: maintaining normal weight, engaging in recommended level of exercise, and consuming a high-quality diet. J Periodontol. 2005;76:1362–1366. doi: 10.1902/jop.2005.76.8.1362. [PubMed] [CrossRef[]
  • McDevitt MJ, Wang HY, Knobelman C. et al.Interleukin-1 genetic association with periodontitis in clinical practice. J Periodontology. 2000;71:156–163. doi: 10.1902/jop.2000.71.2.156. [PubMed] [CrossRef[]
  • Bawadi HA, Khader YS, Haroun TF, Al-Omari M, Tayyem RF. The association between periodontal disease, physical activity and healthy diet among adults in Jordan. J Periodontal Res. 2011;46(1):74–81. doi: 10.1111/j.1600-0765.2010.01314.x. [PubMed] [CrossRef[]
  • Beck JD, Offenbacher S. Systemic effects of periodontitis: epidemiology of periodontal disease and cardiovascular disease. J Periodontol. 2005;76(Suppl11):2089–2100. [PubMed[]
  • Loos BG. Systemic markers of inflammation in periodontitis. J Periodontol. 2005;76(Suppl11):2106–2115. [PubMed[]
  • Tamaki N, Tomofuji T, Ekuni D, Yamanaka R, Morita M. Periodontal treatment decreases plasma oxidized LDL level and oxidative stress. Clin Oral Investig. 2011;15(6):953–958. doi: 10.1007/s00784-010-0458-y. [PubMed] [CrossRef[]
  • Furugen R, Hayashida H, Yamaguchi N, Yoshihara A, Ogawa H, Miyazaki H. et al.The relationship between periodontal condition and serum levels of resistin and adiponectin in elderly Japanese. J Clin Periodontol. 2008;43:556–562. [PubMed[]
  • Saito T, Yamaguchi N, Shimazaki Y, Hayashida H, Yonemoto K, Doi Y. et al.Serum levels of resistin and adiponectin in women with periodontitis: the Hisayama study. J Dent Res. 2008;87:319–322. doi: 10.1177/154405910808700416. [PubMed] [CrossRef[]
  • Bozkurt FY, Yetkin Ay Z, Sutcu R, Delibaş N, Demirel R. Gingival crevicular fluid leptin levels in periodontitis patients with long-term and heavy smoking. J Periodontol. 2006;77:634–640. doi: 10.1902/jop.2006.050277. [PubMed] [CrossRef[]
  • Chaffee BW, Weston SJ. Association between chronic periodontal disease and obesity: a systematic review an meta-analysis. J Periodontol. 2010;12:1708–1724. [PMC free article] [PubMed[]
  • Lakkis D, Bissada NF, Saber A, Khaitan L, Palomo L, Narendran S, Al-Zahrani MS. Response to Periodontal Therapy in Subjects Who Had Weight Loss Following Bariatric Surgery and Obese Counterparts: A Pilot Study. J Periodontol. 2011;83(6):684–689. [PubMed[]
  • Zouza EP, Barroso EM. et al.The role of obesity as a modifying factor in patients undergoing non-surgical periodontal therapy. J Periodontol. 2010;82:676–682. [PubMed[]
  • Suvan J, D’Aiuto F, Moles DR, Petrie A, Donos N. Association between overweight/obesity and periodontitis in adults. A systematic review. Obes Rev. 2011;12(5):e381–e404. doi: 10.1111/j.1467-789X.2010.00808.x. [PubMed] [CrossRef[]
  • Kampe J, Stefanidis A, Lockie SH, Brown WA, Dixon JB, Odoi A, Spencer SJ, Raven J, Oldfield BJ. Neural and humoral changes associated with the adjustable gastric band: insights from a rodent model. Int J Obes (Lond) 2012;27: . [PubMed[]
  • Baltazi M, Katsiki N, Savopoulos C, Iliadis F, Koliakos G, Hatzitolios AI. Plasma neuropeptide Y (NPY) and alpha-melanocyte stimulating hormone (a-MSH) levels in patients with or without hypertension and/or obesity: a pilot study. Am J Cardiovasc Dis. 2011;1(1):48–59. [PMC free article] [PubMed[]
  • Chu SH, Lee MK, Ahn KY, Im JA, Park MS, Lee DC, Jeon JY, Lee JW. Chemerin and adiponectin contribute reciprocally to metabolic syndrome. PLoS One. 2012;7(4):e34710. doi: 10.1371/journal.pone.0034710. [PMC free article] [PubMed] [CrossRef[]
  • Gerrits AJ, Gitz E, Koekman CA, Visseren FL, van Haeften TW, Akkerman JW. Induction of insulin resistance by the adipokines resistin, leptin, PAI-1 and RBP4 in human megakaryocytes. Haematologica. 2012;97(8):1149–1157. doi: 10.3324/haematol.2011.054916. [PMC free article] [PubMed] [CrossRef[]
  • Ross Middleton KM, Patidar SM, Perri MG. The impact of extended care on the long-term maintenance of weight loss: a systematic review and meta-analysis. Obes Rev. 2011;13(6):509–517. [PubMed[]
  • Huck O, Tenenbaum H, Davideau JL. Relationship between Periodontal Diseases and Preterm Birth: Recent Epidemiological and Biological Data. J Pregnancy. 2011;16:46–54. [PMC free article] [PubMed[]
  • Abdilla N, Tormo MC, Fabia MJ, Chaves FJ, Saez G, Redon J. Impact of the components of metabolic syndrome on oxidative stress and enzymatic antioxidant activity in essential hypertension. J Hum Hypertens. 2007;21(suppl.1):68–75. [PubMed[]
  • Chaves FJ, Mansego ML, Blesa S, Gonzalez-Albert V, Jimenez J, Tormos MC, Espinosa O, Giner V, Iradi A, Saez G, Redon J. Inadequate cytoplasmic antioxidant enzymes response contributes to the oxidative stress in human hypertension. Am J Hypertens. 2007;20(supp 1):62–69. [PubMed[]
  • Scott CL. Diagnosis, prevention, and intervention for the metabolic syndrome. Am J Cardiol. 2003;92:35i–42i. doi: 10.1016/S0002-9149(03)00507-1. [PubMed] [CrossRef[]
  • Arthur H, Friedlander DMD, Jane Weinreb MD. Metabolic syndrome: pathogenesis, medical care and dental implications. Clinical pactice. 2007;138:179–187. [PubMed[]
  • Angeli F, Verdecchia P, Pellegrino C, Pellegrino RG, Pellegrino G, Prosciutti L. et al.Association between periodontal disease and left ventricle mass in essential hypertension. Hypertension. 2003;41:488–492. doi: 10.1161/01.HYP.0000056525.17476.D7. [PubMed] [CrossRef[]
  • Rivas-Tumanyan S, Spiegelman D, Curhan GC, Forman JP, Joshipura KJ. Periodontal disease and Incidence of Hypertension in the Health Professionals Follow-Up Study. Am J Hypertens. 2012;25(7):770–776. doi: 10.1038/ajh.2012.32. [PMC free article] [PubMed] [CrossRef[]
  • Rivas-Tumanyan S, Campos M, Zevallos JC, Joshipura KJ. Periodontal Disease, Hypertension and Blood Pressure Among Older Adults in Puerto Rico. J Periodontol. 2012;: . [PMC free article] [PubMed[]
  • Higashi Y, Goto C, Jitsuiki D, Umemura T, Nishioka K, Hidaka T, Takemoto H, Nakamura S, Soga J, Chayama K, Yoshizumi M, Taguchi A. Periodontal infection is associated with endothelial dysfunction in healthy subjects and hypertensive patients. Hypertension. 2008;51(2):446–453. doi: 10.1161/HYPERTENSIONAHA.107.101535. [PubMed] [CrossRef[]
  • Chapple IL, Brock GR, Milaward MR. et al.Compromised GCF total antioxidant capacity in periodontitis. Cause or effect? J Clin Periodontol. 2007;34:103–110. [PubMed[]
  • Brock GR, Matthews JB, Butterworth CJ, Chapple ILC. Local and systemic antioxidant capacity in periodontitis and health. J Clin Periodontol. 2004;31:515–521. doi: 10.1111/j.1600-051X.2004.00509.x. [PubMed] [CrossRef[]
  • Montebugnoli L, Servidio D, Miaton RA, Prati C, Tricoci P, Melloni C. Poor oral health is associated with coronary heart disease and elevated systemic inflammatory and haemostatic factors. J Clin Periodontol. 2004;31:25–29. doi: 10.1111/j.0303-6979.2004.00432.x. [PubMed] [CrossRef[]
  • Borges I Jr, Moreira EA, Filho DW, de Oliveira TB, da Silva MB, Fröde TS. Proinflammatory and oxidative stress markers in patients with periodontal disease. Mediators Inflamm. 2007;4:57–94. [PMC free article] [PubMed[]
  • Alikhani M, Alikhani Z, Boyd C, MacLellan CM, Raptis M, Liu R. et al.Advanced glycation endproducts stimulate osteoblast apoptosis via the MAP kinase and cytosolic apoptotic pathways. Bone. 2007;40:345–353. doi: 10.1016/j.bone.2006.09.011. [PMC free article] [PubMed] [CrossRef[]
  • Vilela EM, Bastos JA, Fernandes N, Ferreira AP, Chaoubah A, Bastos MG. Treatment of chronic periodontitis decreases serum prohepcidin levels in patients with chronic kidney disease. Clinics (Sao Paulo) 2011;66:657–662. doi: 10.1590/S1807-59322011000400022. [PMC free article] [PubMed] [CrossRef[]
  • Tonetti MS, D Aiuto F, Nibali L, Donald A, Storry C, Parkar M. et al.Treatment of periodontitis and endothelial function. N Engl J Med. 2007;356:911–920. doi: 10.1056/NEJMoa063186. [PubMed] [CrossRef[]
  • Chaffee BW, Wetson SJ. Association between chronic periodontal disease and obesity: a systematic review and meta-analysis. J Periodontol. 2010;81:1708–1724. doi: 10.1902/jop.2010.100321. [PMC free article] [PubMed] [CrossRef[]
  • Timonen P, Niskanen M, Suominen-Taipale L. et al.Metabolic syndrome, periodontal infection and dental caries. J Dent Res. 2010;89:1068–1073. doi: 10.1177/0022034510376542. [PubMed] [CrossRef[]
  • Jenkins DJ, Wolever TM, Taylor RH. et al.Glycemix index of foods:a physiological basis for carbohydrate exachange. Am J Clin Nutr. 1981;34:362–366. [PubMed[]
  • Ludvig DS. The glycemic index: physiological mechanisms relating to obesity, diabetes and cardiovascular disease. JAMA. 2002;287:2414–2423. doi: 10.1001/jama.287.18.2414. [PubMed] [CrossRef[]
  • Janket S-J, Jones JA, Meurman JH, Baird AE, Van Dyke TE. Oral Infection, hyperglycemia and endothelial dysfunction. Oral Surg, Oral Med, Oral Pathol, Oral Radiol and Endodontology. 2008;105(suppl 2):173–179. [PMC free article] [PubMed[]
  • Brotman DJ. Effects of counterregulatory hormones in a high glycemic index diet. JAMA. 2002;14(6):288–695. [PubMed[]
  • Liu S, Manson JE. Dietary carbohydrates, physical inactivity, obesity, and the ‘metabolic syndrome’ as predictors of coronary heart disease. Curr Opin Lipidol. 2001;12:395–404. doi: 10.1097/00041433-200108000-00005. [PubMed] [CrossRef[]
  • Nowjack-Raymer RE, Sheiham A. Association of edentulism and diet and nutrition in US adults. J Dent Res. 2003;82:123–126. doi: 10.1177/154405910308200209. [PubMed] [CrossRef[]
  • Ritchie CS, Joshipura K, Hung HC, Douglass CW. Nutrition as a mediator in the relation between oral and systemic disease: associations between specific measures of adult oral health and nutrition outcomes. Crit Rev Oral Biol Med. 2002;13:291–300. doi: 10.1177/154411130201300306. [PubMed] [CrossRef[]
  • Krall E, Hayes C, Garcia R. How dentition status and masticatory function affect nutrient intake. JADA. 1998;129:1261–1269. [PubMed[]
  • Morais JA, Heydecke G, Pawliuk J, Lund JP, Feine JS. The effects of mandibular two-implant overdentures on nutrition in elderly edentulous individuals. J Dent Res. 2003;82:53–58. doi: 10.1177/154405910308200112. [PubMed] [CrossRef[]
  • Wynn RL, Meiller TF, Crossley HL. Drug information for dentistry: Oral medicine for medically-compromised patients and specific oral conditions. 9. Lexi-Comp, Hudson, Ohio; 2003. []
  • Burger W, Chemnitius JM, Kneissl GD, Rucker G. Low-dose aspirin for secondary cardiovascular prevention: cardiovascular risks after its perioperative withdrawal versus bleeding risks with its continuatio: review and meta-analysis. J Intern Med. 2005B;257:399–414. doi: 10.1111/j.1365-2796.2005.01477.x. [PubMed] [CrossRef[]
  • Vernillo AT. Diabetes mellitus: relevance to dental treatment. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2001;91:263–270. doi: 10.1067/moe.2001.114002. [PubMed] [CrossRef[]
  • Vernillo AT. Dental considerations for the treatment of patients with diabetes mellitus. JADA. 2003;134:24s–33s. [PubMed[]
Read More

Occurrence of Spontaneous Periodontal Disease in the SAMP1/YitFc Murine Model of Crohn Disease

Davide PietropaoliRita Del PintoDaniele CorridoniAlexander Rodriguez-Palacios, Gabriella Di StefanoAnnalisa MonacoAaron Weinberg, and Fabio Cominelli

Abstract

Background

Oral involvement is often associated with inflammatory bowel disease (IBD). Recent evidence suggests a high incidence of periodontal disease in patients with Crohn disease (CD). To the best of the authors’ knowledge, no animal model of IBD that displays associated periodontal disease was reported previously. The aim of this study is to investigate the occurrence and progression of periodontal disease in SAMP1/YitFc (SAMP) mice that spontaneously develop a CD-like ileitis. In addition, the temporal correlation between the onset and progression of periodontal disease and the onset of ileitis in SAMP mice was studied.

Methods

At different time points, SAMP and parental AKR/J (AKR) control mice were sacrificed, and mandibles were prepared for stereomicroscopy and histology. Terminal ilea were collected for histologic assessment of inflammation score. Periodontal status, i.e., alveolar bone loss (ABL) and alveolar bone crest, was examined by stereomicroscopy and histomorphometry, respectively.

Results

ABL increased in both strains with age. SAMP mice showed greater ABL compared with AKR mice by 12 weeks of age, with maximal differences observed at 27 weeks of age. AKR control mice did not show the same severity of periodontal disease. Interestingly, a strong positive correlation was found between ileitis severity and ABL in SAMP mice, independent of age.

Conclusions

The present results demonstrate the occurrence of periodontal disease in a mouse model of progressive CD-like ileitis. In addition, the severity of periodontitis strongly correlated with the severity of ileitis, independent of age, suggesting that common pathogenic mechanisms, such as abnormal immune response and dysbiosis, may be shared between these two phenotypes.

Keywords: Crohn disease, inflammatory bowel diseases, models, animal, periodontal diseases, periodontitis

Inflammatory bowel disease (IBD) is a chronic, relapsing and remitting disorder of the gastrointestinal tract, manifesting in two main clinical forms: 1) Crohn disease (CD) and 2) ulcerative colitis (UC). UC commonly involves the rectum but may also affect either part of or the entire colon in a continuous pattern, whereas CD primarily affects the ileum and colon but can also discontinuously involve any region of the gastrointestinal tract. It is widely accepted that IBD results from a dysregulated mucosal immune response to environmental factors in genetically susceptible hosts. However, the precise cause of the disease was not fully elucidated.,,

Although IBD primarily affects the gastrointestinal tract, both CD and UC share a number of extraintestinal manifestations generally related to inflammatory disease activity. These include musculoskeletal, dermatologic, hepatopancreatobiliary, ocular, renal, and pulmonary inflammation. Oral involvement is also common, usually in the form of aphthous stomatitis. Less commonly, patients may experience pyostomatitis vegetans and the more specific aspects of tag-like lesions, mucogingivitis, or “cobblestone” lesions., A higher prevalence of dental caries was also described in individuals with CD.,

Previous studies reported an association between the occurrence of periodontal disease in patients with CD, suggesting that the two inflammatory conditions may share common pathogenic pathways. Similar to IBD, periodontal disease is considered a multifactorial condition, wherein a chronic inflammatory response is triggered and maintained by a combination of genetic and environmental factors, including a prominent role for an abnormal microflora and impaired host responses against the soft and hard tissues surrounding the teeth. Cigarette use and metabolic impairment (as observed in diabetes mellitus) are other common determinants of periodontal disease.,, Mechanistic studies to investigate the relationship between CD and periodontitis relied heavily on the use of chemically induced mouse models of colitis, which are generated through administration of either dextran sodium sulfate (DSS) or trinitrobenzenesulfonic acid (TNBS) enemas to otherwise healthy mice., Although these chemically induced models can be used to understand specific mechanistic aspects of intestinal inflammation and periodontal disease pathogenesis, they do not fully represent the multifactorial and complex nature of these two immune-mediated diseases.

The goal of the present study is to investigate the presence and progression of periodontal disease in a well-characterized spontaneous mouse model of chronic intestinal inflammation. SAMP1/YitFc (SAMP) mice develop severe, chronic ileitis by 20 weeks of age without chemical, genetic, or immunologic manipulation. Furthermore, the resulting ileitis in these mice bears remarkable phenotypic similarities to human CD with regard to disease location, histologic features, response to conventional CD therapies, and, notably, the occurrence of extraintestinal manifestations.

In the present study, evidence is provided that SAMP mice develop a spontaneous periodontitis that temporally correlates with the onset, progression, and severity of ileitis. The present results provide new insight into the pathogenic association between CD and periodontitis and describe, to the best of the authors’ knowledge, for the first time a unique animal model of spontaneous periodontal disease.

MATERIALS AND METHODS

Animals

SAMP mice were bred at Case Western Reserve University, and parental AKR/J (AKR) control mice were purchased.§ All experimental mice were maintained under specific-pathogen-free conditions, fed standard laboratory chow, and kept on 12-hour light/dark cycles. All procedures were approved by the Case Western Reserve University Institutional Animal Care and Use Committee (animal protocol 2011-0196) and conducted in accordance with the Association for Assessment and Accreditation of Laboratory Animal Care guidelines.

Alveolar Bone Loss (ABL) and Ileal Histologic Analyses

SAMP and AKR mice (n = 20 per group) were euthanized by inhalation of CO2 at 4, 12, and 27 weeks of age for evaluation of ABL and ileal inflammation. Additionally, older SAMP mice of 30 to 60 weeks of age (n = 11) were sacrificed for ABL evaluation only, using the same method.

Briefly, mandibles were collected, and soft tissues and gingiva were removed after 10 minutes of boiling. Samples were then defleshed, treated overnight with 3% H2O2, cleaned, and stained with 1%methylene blue. ABL was evaluated using the trough morphometric approach because other methods, such as histomorphometry and microcomputer tomography, revealed similar results. Digital photographs were taken using stereomicroscopy on a custom-made stage holder with an unglued jaw to enhance the visualization of the cemento-enamel junction (CEJ) and bone levels, as described previously. ABL was defined by computer software as the area bordered by the CEJ, the alveolar bone crest, and the mesial and distal lines on the lingual side of the first (M1) and second (M2) mandibular molars. All measurements were performed in triplicate in a masked manner. In addition, to minimize interanimal and age-related variability, normalization by the mesio-distal axis of the teeth was performed. Data are expressed in square millimeters for ABL and millimeters for tooth axis.

For histologic evaluation of intestinal inflammation, ilea from experimental mice were removed, flushed of fecal contents, opened longitudinally, and placed in Bouin fixative for 24 hours, followed by 70% ethanol. Tissues were embedded in paraffin and stained with hematoxylin and eosin (H&E). Inflammation was evaluated by a trained pathologist (Prof. Wei Xin, Case Medical Center, Cleveland, OH) in a masked manner using an established and validated scoring system. The total inflammatory index for ileal specimens represents the sum of three individual indices: 1) active inflammation; 2) chronic inflammation; and 3) villous distortion.

Histologic Evaluation of the Periodontal Tissues

Four- and 30-week-old SAMP and AKR mice were euthanized by inhalation of CO2 for histologic evaluation of periodontal tissues. Mandibles were collected, fixed in 10% formalin for 24 hours, placed in 70% ethanol for 24 hours, decalcified by hydrochloric acid solution,# washed, dehydrated, and embedded in paraffin. Serial sections (five semi-serial sections of each hemimandible) with 5- µm thickness were cut and stained with H&E.

The same morphometric approach was used to evaluate the area of the alveolar bone crest and the hemiproximal and hemidistal coronal-to-apical cementum layer of the first and second mandibular molars, respectively. The cementum layer was measured on the hemidistal root of the first molar and the hemiproximal root of the second molar, and a total cementum area was calculated as a sum of the two. A trained pathologist (DP) evaluated each section in a masked manner, and all measurements were performed in triplicate.

Statistical Analyses

All experiments were performed at least twice, and statistical analysis was conducted using the collective data from replicated experiments. Data that revealed a normal distribution by the Shapiro-Wilk test were compared using the Student t test, one-way or two-way analysis of variance (ANOVA), linear regression, or receiver operating characteristic (ROC) analysis. Alternative nonparametric tests were used for data that were not normally distributed. The level of significance was set at P <0.05, and standard error of the means are shown in the figures. Statistical analysis and graph plots were performed using a statistical program.**

RESULTS

Stereomicroscopic results are shown in Figures 1 and and2.2. Stereomicroscopy analysis revealed more severe ABL in SAMP mice compared with AKR mice starting at 12 weeks of age (Fig. 1A), and the severity at each site is shown in the heat map (Fig. 1B). Total ABL of M1 and M2 of both sides (M1 ABL and M2 ABL, respectively) were analyzed individually and together according to age in both SAMP and AKR control mice, as shown in Figure 2A. M1 ABL increased in both strains according to age. Elevated levels were observed in SAMP mice compared with AKR mice beginning as early as 12 weeks of age (P = 0.04) and further increased at 27 weeks (P <0.001). No differences in ABL of M2 were found between the two strains or within strain at any age (Fig. 2A). Total ABL for the entire resorption area of M1 and M2 (M1+M2 ABL) was increased in SAMP mice versus AKR mice at 27 weeks (P <0.001) but not at 12 weeks of age. The same comparisons performed with normalized data (ratio of ABL [square millimeters] to teeth major axis [millimeters]) provided comparable results, and, as expected, teeth axis lengths were statistically similar between strains (Fig. 1D). Based on these results, M1 ABL was determined to have the highest level of discrimination between the two strains (versus M2 ABL or M1+M2 ABL) and was used for all subsequent analyses.

An external file that holds a picture, illustration, etc. Object name is nihms695841f1.jpg

A) Stereomicroscopy images showing ABL in SAMP and AKR mice at 4, 12, and 27 weeks of age. B) A heat map is a graphical way of displaying a table of numbers by using colors to represent numerical values. Specifically, low severity scores tend toward lighter colors, whereas high severity scores are represented by darker colors. The heat map herein shows single-tooth and total ABL, ABL ratios, and ileum score for each sample in both strains, thus summarizing the relations of periodontal disease to ileal inflammation severity. Each row represents samples from a single animal, with AKR mice clustered in the top half of the picture and SAMP mice in the bottom. Darker colors occur more frequently in the bottom half (SAMP mice), thus representing higher severity scores of both periodontal disease and ileitis in SAMP mice compared with AKR mice. C) Ileal sections from 27-week-old SAMP mice and AKR control mice (H&E; magnification ×10). SAMP mice develop spontaneous, transmural inflammation of the terminal ileum characterized by discontinuous inflammatory infiltrates, villous architecture alterations, and bowel wall thickening, with hypertrophy of the muscular layers, villous blunting and distortion, and infiltration of acute and chronic immune cells. A normal structure in uninflamed control AKR mice is also shown. D) Comparison of tooth axis of left/right M1 and M2 showed no statistical difference between strains (ANOVA), thus indicating similar tooth dimensions in the two strains. LF = left; RG = right.

An external file that holds a picture, illustration, etc. Object name is nihms695841f2.jpg

A) ANOVA of ABL on M1, M2, and M1+M2 according to age. M1 alone was able to discriminate the strains by 12 weeks of age, M1+M2 ABL only at 27 weeks, and M2 alone was not able to identify differences between strains. NS = not significant. B) Regression analysis of ABL in SAMP and AKR mice. SAMP mice showed a comparable ABL value in both sides of the mouth, suggesting a very reproducible model (R2 = 0.834); AKR did not show the same features. C) ROC analysis ofM1 ABL and M1 ABL ratio showed high sensitivity to differentiate between the groups, with SAMP AUC = 0.873 and AKR AUC = 0.932. SE = standard error. D) A positive significant correlation between ileal disease and periodontal disease was found in SAMP mice (P = 0.002; R2 = 0.432). The severity of periodontal disease was directly related to the severity of ileal damage.

Combined cluster analyses on right and left M1 ABL in SAMP and AKR mice clearly showed the segregation of samples into two groups according to strain (Fig. 2B), with SAMP mice showing more bone resorption compared with AKR mice. Regression analysis yielded a very reproducible correlation between right and left M1 ABL in SAMP mice (y = −0.03 + 1.04 × xR2 = 0.834; P <0.001), indicating that the same process occurs in both sides of the mouth, and is suggestive of either a systemic disease or a phenomenon driven by a systemic factor. In contrast, AKR mice tended to show a small correlation between left and right M1 ABL (M1 left, 0.39 mm2; M1 right, 0.47 mm2) and did not display a clear trend toward increased variability (Fig. 2B). To determine the discriminatory ability of M1 ABL and M1 ratio (M1 ABL [square millimeters] to M1 major axis [millimeters]) to differentiate between the two strains, sensitivity/specificity ROC curve analysis was conducted, indicating that both methods have a high sensitivity to differentiate the groups (AKR: area under the curve [AUC] = 0.932, 95% confidence interval (CI) = 0.87 to 0.99; SAMP: AUC = 0.873, 95% CI = 0.77 to 0.97) (Fig. 2C).

To test the hypothesis that the occurrence of periodontal disease correlates with the severity of intestinal inflammation in SAMP mice (Fig. 1C), a regression analysis was performed between the ileal histologic scores and M1 ABL. The results showed a significant positive correlation between the two parameters (y = 0.69 + 0.02 × xR2 = 0.432; P = 0.002), with periodontal disease severity increasing as the severity of ileal scores in SAMP mice also increased (Fig. 2D).

Histologic results are shown in Figure 3. Histologic evaluation of mandibles from SAMP mice showed the presence of periodontal disease (Fig. 3A). Evidence of alveolar bone resorption was observed in SAMP mice as early as 4 weeks, with gradual worsening over time. A significant increase in polymorphonuclear leukocyte (PMN) and mononuclear cell (MN) infiltration was noted in the alveolar bone crest over time in SAMP mice compared with AKR control mice (Fig. 3A). Alveolar bone morphology was not preserved, and mosaic-like remodeling was present in SAMP mice but not in AKR mice (Figs. 3A and 3C). Cementum layer and granuloma-like lesions with substantial infiltration of inflammatory cells was observed in SAMP mice at all ages but remarkably increased only at 30 weeks compared with 4 weeks (Figs. 3A through 3C). AKR mice did not show the same features in the aforementioned described structures. Computer-assisted morphometric analysis showed no difference for SAMP versus AKR mice in alveolar bone crest and cementum area at 4 weeks of age. In contrast, SAMP mice displayed lower alveolar bone crest area (P = 0.02) and increased cementum area compared with AKR control mice at 30 weeks of age (P = 0.001) (Fig. 3D).

An external file that holds a picture, illustration, etc. Object name is nihms695841f3.jpg

A) Sagittal mandibular sections of SAMP and AKR mice at 4 and 30 weeks of age. Selected areas of interest (black squares) are presented as magnified images below the corresponding section. No notable alveolar bone resorption or signs of inflammation were observed at 4 weeks in either strain. Thirty-week-old SAMP mice showed reduction in the thickness of the alveolar bone crest compared with AKR mice (filled arrows). Surprisingly, severe hypercementosis was found in SAMP mice but not in AKR mice (open arrows) (H&E; original magnification ×10). B) Higher magnification (×40) of the CEJ area shown in Fig. 3A reveals the major thickness of cementum layer (C). PMN and MN cells were present in the periodontal ligament (PDL) layer of SAMP mice. The alveolar bone crest (AB) was markedly reduced in SAMP mice but not in AKR mice at 30 weeks of age. Root dentin (D) appears normal. (H&E stain.) C) Hypercementosis in SAMP mice was accompanied by severe apical lesions (dotted circles) in the absence of apparent endodontic causes. Magnification of apical lesions is shown under the corresponding original image. Numerous inflammatory cells surround the apical region, and alveolar bone remodeling was visible between the roots (H&E; original magnification ×10). D) Ranked Kruskal-Wallis one-way ANOVA showed statistical differences in terms of alveolar bone crest area (ABC) (P = 0.02) and cementum area (P = 0.001) only at 30 weeks of age. NS = not significant.

DISCUSSION

This study demonstrates the spontaneous occurrence of periodontal disease in a mouse model of CD-like terminal ileitis, in the absence of any exogenous proinflammatory stimuli. The present results are consistent with data from clinical studies showing a high prevalence of periodontitis in patients with IBD. In addition, many case-control studies and case reports observed this relationship.,, Interestingly, periodontal disease and hypercementosis in SAMP mice occurs spontaneously, with similar alveolar bone resorption on both sides of the mouth, suggesting a systemic disease or phenomenon.

Moreover, the present study provides strong evidence of a correlation between the severity of both inflammatory disorders. Notably, although the data confirm the progression of periodontal disease with age in both SAMP and AKR mice as described previously, the association of periodontal disease with ileitis severity was found to be independent of age, a finding that is-consistent with human disease wherein severe periodontitis was reported in young patients with IBD.

To the best of the authors’ knowledge, this is the first study to evaluate the occurrence and severity of periodontal disease in a mouse model of spontaneous CD-like ileitis. A recent study described a mouse model of chemically induced periodontitis that uses prolonged oral administration of TNBS and DSS to induce a status of non-specific chronic inflammation and ABL. DSS is commonly used to chemically induce colitis in mice, and the same study describes how the administration of low doses of DSS is able to induce a chronic inflammatory state in both the intestine and periodontal tissue, suggesting an imbalance in the antioxidant functional reserve as a common link. However, the limitations of chemically induced models lie in the dose and time dependence of the clinical manifestations; consequently, these models do not represent the best system for evaluating shared pathogenic mechanisms between periodontal disease and intestinal lesions in IBD.

One possible common immunopathogenic mechanism may be a shared abnormal cytokine pattern and intercellular crosstalk that leads to T-helper 1 polarization and progressive tissue damage, which is exacerbated by coexistent predisposing genetic mutations, poor health conditions, malnutrition, and commensal bacterial load, and pathogenicity. In the 1980s, Van Dyke et al. first described the presence of a peculiar oral microflora in patients with IBD with concomitant periodontal disease that was mainly composed of motile Gram-negative rods, referred to as Wolinella genus. This study also observed an important defect in neutrophil chemotaxis in all patients with IBD with periodontal disease that was not associated with impaired phagocytic function in the same cells. It was proposed that this oral pathogen may play a role in IBD pathogenesis, possibly as a trigger for an infectious agent or as an immune response modifier.

The present results in the SAMP mouse model are consistent with this hypothesis. Previous in vivo and in vitro studies with the SAMP mice show that these mice have a reduced capacity for bacterial clearance, although their phagocytic ability is preserved. Although they do not harbor genetic mutations in the nucleotide-binding oligomerization domain containing 2 (NOD2) (which were associated with CD in a subset of patients), they do display abnormal immune responses and bacterial clearance in response to muramyl dipeptide stimulation, suggesting an underlying functional defect in the NOD2 molecular pathway. This immunologic dysfunction may contribute to the pathogenesis of both the observed intestinal and oral lesions. Moreover, SAMP mice also show a defect in intestinal permeability, demonstrated through ex vivo studies on portions of terminal ileum. Interestingly, the use of probiotics ameliorates the mucosal permeability defect in SAMP mice, further suggesting a role for the microbiota in the clinical course of chronic ileitis.

CONCLUSIONS

The SAMP mouse model of spontaneous CD-like ileitis represents a powerful tool for the study not only of the natural history of IBD but also of the associated periodontal disease. These two conditions likely share similar etiopathogenic features, and multiple pathogenic mechanisms that were identified as contributing to SAMP ileitis conceptually may also be involved in the development of SAMP periodontal disease. These findings suggest an intriguing pathogenic hypothesis linking the two conditions and provide a new tool for the development of novel therapeutic interventions for both diseases. Future research will aim at defining the role of cytokines and the function of the oral microbiota in SAMP mice and in humans with CD.

ACKNOWLEDGMENTS

This research was supported by National Institutes of Health/National Institute of Diabetes and Digestive and Kidney Diseases Grants P01DK091222, R01DK055812, and R01DK04219 (to FC). The authors thank Mitchell Guanzon, Dennis Guszka, Joshua Weber, and Lindsey Kaydo for technical assistance (Department of Medicine and Pathology, and Digestive Health Research Center, Case Western Reserve University, Cleveland, Ohio) and Kristen Arseneau (Department of Medicine and Pathology, and Digestive Health Research Center, Case Western Reserve University) and Theresa Pizarro (Department of Pathology, Case Western Reserve University School of Medicine) for critical revision of this manuscript.

Footnotes

§The Jackson Laboratory, Bar Harbor, ME.

Teklad, Harlan, Indianapolis, IN.

Image J v1.46r, National Institutes of Health, Bethesda, MD.

#Rapid Decalcifier, VWR International, Radnor, PA.

**The R packages “stat” and “ggplot” v.3.0.3, R Foundation for Statistical Computing, Vienna, Austria.

The authors report no conflicts of interest related to this study.

REFERENCES

1. Larsen S, Bendtzen K, Nielsen OH. Extraintestinal manifestations of inflammatory bowel disease: Epidemiology, diagnosis, and management. Ann Med. 2010;42:97–114. [PubMed[]
2. Su CG, Judge TA, Lichtenstein GR. Extraintestinal manifestations of inflammatory bowel disease. Gastroenterol Clin North Am. 2002;31:307–327. [PubMed[]
3. Danese S, Semeraro S, Papa A, et al. Extraintestinal manifestations in inflammatory bowel disease. World J Gastroenterol. 2005;11:7227–7236. [PMC free article] [PubMed[]
4. Urlep D, Mamula P, Baldassano R. Extraintestinal manifestations of inflammatory bowel disease. Minerva Gastroenterol Dietol. 2005;51:147–163. [PubMed[]
5. Lankarani KB, Sivandzadeh GR, Hassanpour S. Oral manifestation in inflammatory bowel disease: A review. World J Gastroenterol. 2013;19:8571–8579. [PMC free article] [PubMed[]
6. Lourencxo SV, Hussein TP, Bologna SB, Sipahi AM, Nico MM. Oral manifestations of inflammatory bowel disease: A review based on the observation of six cases. J Eur Acad Dermatol Venereol. 2010;24:204–207. [PubMed[]
7. Grössner-Schreiber B, Fetter T, Hedderich J, Kocher T, Schreiber S, Jepsen S. Prevalence of dental caries and periodontal disease in patients with inflammatory bowel disease: A case-control study. J Clin Periodontol. 2006;33:478–484. [PubMed[]
8. Sundh B, Emilson CG. Salivary and microbial conditions and dental health in patients with Crohn’s disease: A 3-year study. Oral Surg Oral Med Oral Pathol. 1989;67:286–290. [PubMed[]
9. Vavricka SR, Manser CN, Hediger S, et al. Periodontitis and gingivitis in inflammatory bowel disease: A casecontrol study. Inflamm Bowel Dis. 2013;19:2768–2777. [PubMed[]
10. Brandtzaeg P. Inflammatory bowel disease: Clinics and pathology. Do inflammatory bowel disease and periodontal disease have similar immunopathogeneses? Acta Odontol Scand. 2001;59:235–243. [PubMed[]
11. Oz HS, Chen T, Ebersole JL. A model for chronic mucosal inflammation in IBD and periodontitis. Dig Dis Sci. 2010;55:2194–2202. [PubMed[]
12. Van Dyke TE, Dowell VR, Jr, Offenbacher S, Snyder W, Hersh T. Potential role of microorganisms isolated from periodontal lesions in the pathogenesis of inflammatory bowel disease. Infect Immun. 1986;53:671–677. [PMC free article] [PubMed[]
13. Pietropaoli D, Monaco A, Del Pinto R, Cifone MG, Marzo G, Giannoni M. Advanced glycation end products: Possible link between metabolic syndrome and periodontal diseases. Int J Immunopathol Pharmacol. 2012;25:9–17. [PubMed[]
14. Maddi A, Scannapieco FA. Oral biofilms, oral and periodontal infections, and systemic disease. Am J Dent. 2013;26:249–254. [PubMed[]
15. Oz HS, Ebersole JL. A novel murine model for chronic inflammatory alveolar bone loss. J Periodontal Res. 2010;45:94–99. [PMC free article] [PubMed[]
16. Pizarro TT, Pastorelli L, Bamias G, et al. SAMP1/YitFc mouse strain: A spontaneous model of Crohn’s disease-like ileitis. Inflamm Bowel Dis. 2011;17:2566–2584. [PMC free article] [PubMed[]
17. Li CH, Amar S. Morphometric, histomorphometric, and microcomputed tomographic analysis of periodontal inflammatory lesions in a murine model. J Periodontol. 2007;78:1120–1128. [PubMed[]
18. Rivaldo EG, Padilha DM, Hugo FN, Hilgert JB, Rybu BR. Reproducibility of a hemi mandible positioning device and a method for measuring alveolar bone loss area in mice. J Oral Sci. 2007;49:13–17. [PubMed[]
19. Wang Q, Li H, Xie H, et al. 25-Hydroxyvitamin D3 attenuates experimental periodontitis through down-regulation of TLR4 and JAK1/STAT3 signaling in diabetic mice. J Steroid Biochem Mol Biol. 2013;135:43–50. [PubMed[]
20. Bamias G, Martin C, Mishina M, et al. Proinflammatory effects of TH2 cytokines in a murine model of chronic small intestinal inflammation. Gastroenterology. 2005;128:654–666. [PubMed[]
21. Shibata Y, Fujita S, Takahashi H, Yamaguchi A, Koji T. Assessment of decalcifying protocols for detection of specific RNA by non-radioactive in situ hybridization in calcified tissues. Histochem Cell Biol. 2000;113:153–159. [PubMed[]
22. Habashneh RA, Khader YS, Alhumouz MK, Jadallah K, Ajlouni Y. The association between inflammatory bowel disease and periodontitis among Jordanians: A case-control study. J Periodontal Res. 2012;47:293–298. [PubMed[]
23. Flemmig TF, Shanahan F, Miyasaki KT. Prevalence and severity of periodontal disease in patients with inflammatory bowel disease. J Clin Periodontol. 1991;18:690–697. [PubMed[]
24. Sigusch BW. Periodontitis as manifestation of Crohn’s disease in primary dentition: A case report. J Dent Child (Chic) 2004;71:193–196. [PubMed[]
25. Corridoni D, Kodani T, Rodriguez-Palacios A, et al. Dysregulated NOD2 predisposes SAMP1/YitFc mice to chronic intestinal inflammation. Proc Natl Acad Sci USA. 2013;110:16999–17004. [PMC free article] [PubMed[]
26. Corridoni D, Pastorelli L, Mattioli B, et al. Probiotic bacteria regulate intestinal epithelial permeability in experimental ileitis by a TNF-dependent mechanism. PLoS One. 2012;7:e42067. [PMC free article] [PubMed[]
Read More

Polyarteritis nodosa involving the hard palate: a case report

Published online 2013 Mar 18

Eleonora OrtuDavide PietropaoliMario BaldiGiuseppe MarzoMario Giannoni, and Annalisa Monaco

Abstract

Introduction

Polyarteritis nodosa is a rare disease resulting from blood vessel inflammation (vasculitis), causing damage to organ systems and featuring an extended range of possible symptoms. The cause of polyarteritis nodosa is unknown.

Case presentation

In the present report we describe the presentation and treatment of polyarteritis nodosa involving the hard palate in an 88-year-old Caucasian woman. Clinical and laboratory analyses showed stenosis of the greater palatine artery, which led to necrosis of the affected area. At one year after pharmacological treatment, the lesion has regressed completely.

Conclusions

We successfully treated a case of polyarteritis nodosa via a pharmacological approach, which we describe here.

Introduction

Polyarteritis nodosa (PAN), also known as Kussmaul disease or Kussmaul-Maier disease, is a vasculitis of the medium and/or small arteries that become swollen and damaged as a result of the attack by rogue immune cells, and the condition may be associated with various atypical presentations []. The annual incidence of PAN varies between five and nine cases per million []. Men are generally more affected than women in a 2:1 ratio, most frequently between the ages of 40 and 60 years. PAN is more common in people with hepatitis B infection []. PAN may affect multiple organs, including skin, kidneys, and gastrointestinal tract, as well as the peripheral and central nervous systems []. The inflammatory process causes necrosis of cells and structural components of the artery with aneurysms or stenosis formations []. PAN often can culminate in necrosis or hemorrhage of the affected organ. Arterial bifurcations are predilected by PAN []. This causes a characteristic pattern of aneurysms that Kussmaul and Maier, in their article from 1866, described as being like the apples on the branches of a tree or a wreath of roses []. There are no specific laboratory tests for the diagnosis of PAN. The American College of Rheumatology has established criteria for distinguishing PAN from other forms of vasculitis. In the diagnosis of PAN, at least three of the following ten criteria must be considered when radiographic or pathological diagnosis of vasculitis is made []: weight loss of 4kg or more; livedo reticularis; testicular pain/tenderness; myalgia or leg weakness/tenderness; mononeuropathy or polyneuropathy; diastolic blood pressure greater than 90mmHg; elevated blood urea nitrogen (BUN) or creatinine level unrelated to dehydration or obstruction; presence of hepatitis B surface antigen or antibody in serum; arteriogram demonstrating aneurysms or occlusions of the visceral arteries; biopsy of small-sized or medium-sized artery containing polymorphonuclear neutrophils.

Here, we report a rare case of PAN that featured necrosis of the hard palate as the main manifestation of the disease. The clinical evidence highlighted a greater palatine artery stenosis, which led to necrosis of the affected area.

Case presentation

An 88-year-old Caucasian woman, suffering from type 2 diabetes, hypertension (ambulatory blood pressure of 145/90mmHg) and Parkinson’s disease was referred to our clinic with a one-week history of a white area on her palate.

An oral examination revealed a white area, not raised, occupying more than half of the hard palate. Our patient did not report any pain but her temperature had risen to 38°C during the initial four-day period. A moderate weight loss (5kg), not caused by dietary restriction, was also reported. The results of laboratory tests were as follows: hemoglobin 9.5g/dL, white blood cell count 6,300 cells/L (neutrophils 55 percent, lymphocytes 37 percent, monocytes 5 percent, eosinophils 2 percent, basophils 1 percent), serum creatinine 1.6mg/dL, and erythrocyte sedimentation rate (ESR) 107mm/hour. The results of serum antigen tests for hepatitis B virus (HBV), hepatitis C virus (HCV) and parvovirus B19 were negative. A test for anti-neutrophil cytoplasmic antibodies (ANCAs) was also negative. On clinical examination, the affected area showed a well demarcated fibrinous border, matching the vascular zone affected by necrosis (Figure 1). An incisional biopsy of the peripheral margin of the zone was taken under local anesthesia.

An external file that holds a picture, illustration, etc. Object name is 1752-1947-7-79-1.jpg

Palatal lesion at presentation. On clinical examination, the affected area showed a well demarcated, fibrinous border, corresponding to the vascular zone affected by necrosis.

Microscopic study of histological sections revealed that while fibrinoid necrosis involved both lateral extremities of the vascular segment, and that the middle portion was relatively unaffected. The presence of inflammatory infiltrate with variable number of neutrophils, macrophages and scattered lymphocytes was also observed (Figure 2). All these histopathological findings were consistent with a diagnosis of PAN. Also, as suggested by the American College of Rheumatology guidelines, the diagnosis of PAN was made on the basis of six signs that our patient exhibited []. It was not possible to perform other studies due to the inability of our patient to move.

An external file that holds a picture, illustration, etc. Object name is 1752-1947-7-79-2.jpg

Histological section.

According to recent literature [] laboratory analysis was used to evaluate compete blood counts with potassium, vitamin D3 and calcium. Treatment was started urgently and our patient was treated with oral prednisone at a dosage of 1mg/kg/day, prescribed for three weeks, before being tapered by 5mg every 10 days to a dosage of 0.5mg/kg/day then by 2.5mg every 10 days until a dosage of 15mg/day was obtained and, finally, by 1mg every 10 days to the minimal effective dose or, when possible, until definitive withdrawal. She was also treated with aspirin and warfarin, added to her anti-hypertensive medications, and rinsing with saline solution three to four times at day to clean the affected area. Adjuvant treatment with vitamin D3, calcium and potassium to prevent corticosteroid-induced osteoporosis and cytidine 5′-diphosphocholine (500mg/day) as supportive treatment for Parkinson’s disease was also given.

After this treatment, the lesion showed improvement and had regressed completely at one year from treatment initiation (Figure 3).

An external file that holds a picture, illustration, etc. Object name is 1752-1947-7-79-3.jpg

The lesion two years from onset. At this time the area of interest is very well vascularized with no recurrences.

Discussion

Patients with PAN may present with various symptoms related to vascular involvement within a specific organ. Our patient’s case illustrates a rare and interesting manifestation of PAN with involvement of the hard palate. The authors do not know if the appearance of vasculitis was limited to the palate or whether in the past the pathology had involved other organs.

The pathogenesis of this unusual location is unknown, and to the best of our knowledge has not been described in literature to date.

PAN is a vasculitis that affects small and medium vessels and is characterized by periods of exacerbation and remission periods []. This case of PAN showed the involvement of the greater palatine artery, which caused the necrosis of the area of interest. A thorough differential diagnosis is needed to confirm a diagnosis PAN because other forms of vasculitis could lead to an incorrect diagnosis. No specific tests are known for diagnosis of PAN, but the American College of Rheumatology suggests 10 clinical criteria including blood profiles of antibodies and angiography and histology []. Patients may present with a variety of symptoms, the most common of which are fever (25 percent to 56 percent), neuropathy (77 percent to 92 percent) and weight loss (63 percent to 85 percent) []. The differential diagnosis should include other systemic autoimmune diseases (systemic lupus erythematosus, severe rheumatoid arthritis and others) or marked fever and weight loss in tumor cases []. Corticosteroids are usually the first-line treatment drugs and a response is usually noted within three months. Cyclophosphamide has shown a better outcome in patients with severe disease as well as those refractory to steroid treatment. Although there is no clear survival benefit when steroids are used in combination with cyclophosphamide, the relapse rate was reduced significantly [].

In our patient’s case, PAN was not treated with cyclophosphamide as use of corticosteroids significantly reduced the lesion, restoring proper blood circulation in the affected area. Our patient was also treated with anti-osteoporotic drugs to avoid consequent bone problems. The drugs already prescribed to our patient for the control of diabetes and Parkinson’s disease were not modified with regard to dosages. Only aspirin and warfarin were added to the anti-hypertensive medications.

Conclusions

The present report describes the successful treatment of PAN via a pharmacological approach. The clinical case presented is a very rare event and to the best of our knowledge has not been described in the literature previously. At two years after the appearance of the lesion, the area of interest is very well vascularized and there have been no recurrences. Routine blood tests and biopsies are essential for diagnosis and to avoid delays in treatment and possible organ damage.

Consent

Written informed consent was obtained from the patient for publication of this case report and any accompanying images. A copy of the written consent is available for review by the Editor-in-Chief of this journal.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

AM, MG and MB analyzed and interpreted the data from our patient regarding the blood tests and the disease. AM, GM, MG and MB made the diagnosis of PAN and established the treatment. EO and DP were the major contributors in writing the manuscript. All authors read and approved the final manuscript.

References

  • Jennette JC, Falk RJ, Bacon PA, Basu N, Cid MC, Ferrario F, Flores-Suarez LF, Gross WL, Guillevin L, Hagen EC, Hoffman GS, Jayne DR, Kallenberg CG, Lamprecht P, Langford CA, Luqmani RA, Mahr AD, Matteson EL, Merkel PA, Ozen S, Pusey CD, Rasmussen N, Rees AJ, Scott DG, Specks U, Stone JH, Takahashi K, Watts RA. 2012 revised international chapel hill consensus conference nomenclature of vasculitides. Arthritis Rheum. 2013;65:1–11. [PubMed[]
  • Erhardt A, Sagir A, Guillevin L, Neuen-Jacob E, Haussinger D. Successful treatment of hepatitis B virus associated polyarteritis nodosa with a combination of prednisolone, alpha-interferon and lamivudine. J Hepatol. 2000;33:677–683. doi: 10.1016/S0168-8278(00)80025-2. [PubMed] [CrossRef[]
  • Leib ES, Restivo C, Paulus HE. Immunosuppressive and corticosteroid therapy of polyarteritis nodosa. Am J Med. 1979;67:941–947. doi: 10.1016/0002-9343(79)90634-X. [PubMed] [CrossRef[]
  • Battula N, Tsapralis D, Morgan M, Mirza D. Spontaneous liver haemorrhage and haemobilia as initial presentation of undiagnosed polyarteritis nodosa. Ann R Coll Surg Engl. 2012;94:e163–e165. doi: 10.1308/003588412X13171221590737. [PMC free article] [PubMed] [CrossRef[]
  • Kermani TA, Ham EK, Camilleri MJ, Warrington KJ. Polyarteritis nodosa-like vasculitis in association with minocycline use: a single-center case series. Semin Arthritis Rheum. 2012;42:213–221. doi: 10.1016/j.semarthrit.2012.03.006. [PubMed] [CrossRef[]
  • Stroup SP, Herrera SR, Crain DS. Bilateral testicular infarction and orchiectomy as a complication of polyarteritis nodosa. Reviews in urology. 2007;9:235–238. [PMC free article] [PubMed[]
  • Ambrosio MR, Rocca BJ, Ginori A, Onorati M, Fabbri A, Carmellini M, Lazzi S, Tripodi S. Renal infarction due to polyarteritis nodosa in a patient with angioimmunoblastic T-cell lymphoma: a case report and a brief review of the literature. Diagn Pathol. 2012;7:50. doi: 10.1186/1746-1596-7-50. [PMC free article] [PubMed] [CrossRef[]
  • Savage CO, Harper L, Adu D. Primary systemic vasculitis. Lancet. 1997;349:553–558. doi: 10.1016/S0140-6736(97)80118-3. [PubMed] [CrossRef[]
  • Rao JK, Allen NB, Pincus T. Limitations of the 1990 American College of Rheumatology classification criteria in the diagnosis of vasculitis. Ann Intern Med. 1998;129:345–352. [PubMed[]
  • de Menthon M, Mahr A. Treating polyarteritis nodosa: current state of the art. Clin Exp Rheumatol. 2011;29(Suppl):S110–S116. [PubMed[]
  • Jennette JC, Falk RJ, Andrassy K, Bacon PA, Churg J, Gross WL, Hagen EC, Hoffman GS, Hunder GG, Kallenberg CG, Mccluskey RT, Sinico RA, Rees AJ, Van Es LA, Waldherr R, Wiik A. Nomenclature of systemic vasculitides. Proposal of an international consensus conference. Arthritis Rheum. 1994;37:187–192. doi: 10.1002/art.1780370206. [PubMed] [CrossRef[]
  • Stone JH. Polyarteritis nodosa. JAMA. 2002;288:1632–1639. doi: 10.1001/jama.288.13.1632. [PubMed] [CrossRef[]
Read More

Relationship between mastication and cognitive function in elderly in L’Aquila

Published online 2014 Apr 15

Stefano MummoloEleonora OrtuStefano NecozioneAnnalisa Monaco, and Giuseppe Marzo

Abstract

Patients with cognitive deficit have poor oral health and fewer teeth than cognitive normal elderly. The aim of the study was to investigate potential differences in masticatory function between elderly with dementia and those with normal cognitive function. Fifty-five patients (age >61; 82.05 ± 3.53) were enrolled in the study. Twenty-five subjects cognitively normal (10 females/15 males; 81.04 ± 4.89 years), were randomly selected and were assigned to Control Group. Thirty subjects (15 females/15 males; 83.16 ± 6.017 with cognitive impairments were randomly selected from hospitalized patients (Medically Assisted Residences RSA) and were assigned to Test Group. MMSE test, B-ADL and number of teeth were evaluated for each subject. The number of teeth in relation to levels of schooling is not resulted significative. In the cognitively impaired group 26 subjects had fewer than 20 teeth (86.6%); in the cognitively normal group 9 subjects had fewer than 20 teeth (36%). The correlation between number of teeth and age in both groups is significative (p<0.05). There is also a significative correlation between subjects with renal diseases and type II diabetes and number of teeth (p<0.05). Finally a significative correlation is present between number of teeth and sex of the patients (p<0.05) (Table 1). The results of the Wilcoxon’s test revealed a significative correlation between MMSE in the two groups (p<0.01). There is also a significative correlation between the two groups and the educational background (p<0.01). The results of the study shows a clear correlation between tooth loss and cognitive function in elderly of L’Aquila.

Introduction

Mastication has been shown to promote and preserve general health, especially (and it seems involved in maintenance of) the cognitive function of the brain, beyond its primary functions of food intake and digestion [,]. Research on aging and mastication have shown that the decrease of number of teeth and the impairment jaw muscle activity due to aging cause a reduction in sensory input activity to the central nervous system [,]. Functional magnetic resonance imaging and positron emission topography revealed that mastication increases cortical blood flow and widely activates various cortical areas of the somatosensory, supplementary motor, and insular cortices, as well as the striatum, thalamus and cerebellum [,]. Evidences suggest a possible relationship between mastication and brain function [,]. The World Health Organization (WHO 2006. Oral Health in Ageing Societies—integration of oral health and general health. WHO 2, 1-54) recognized the importance of oral health care and indicates a stringent need for research, training of caregivers, and development of policy regarding oral health care. Oral care should be actively provided to older persons in nursing homes and should furthermore not be limited to individuals retaining some teeth but extended to edentate persons as well []. Elderly people are at risk factors for cognitive impairment and for developing Alzheimer’s disease (AD), which is one of the most common subtypes of dementia []. Among the risk factors including ageing, illiteracy, lower level of education, lower socioeconomic status [], head trauma, genetic factors, cardiovascular risk factors, overweight, smoking, hypertension and diabetes mellitus, an inactive lifestyle, surprisingly, loss of teeth showed significant importance []. Mini-Mental State examination (MMSE) and the revised Hasegawa Dementia Rating Scale (HDS-R) has been used to evaluate higher brain function in standardized screening test. Recent data showed that masticatory ability and the number of natural teeth are related to cognitive function among the elderly without dementia [,].

The purpose of the present study was to investigate potential differences in masticatory function between elderly with dementia and those with normal cognitive function. In particular, we matched age and basic activities of daily living (B-ADL), masticatory function assessed in terms of the presence/absence of teeth, educational background, health general conditions (Blood pressure, diabetes, and renal diseases) and cognitive function (MMSE).

Material and methods

This study was conducted in accordance with the Declaration of Helsinki. The Committee on Ethics in Science of the University of L’Aquila, L’Aquila, Italy approved the study and informed consent was obtained from each subject or from kins or legal representatives. The study was conducted between June 2011 and June 2012. Fifty-five patients (age >61; 82.05 ± 3.53) were enrolled in the study. Twenty-five subjects cognitively normal (10 females/15 males; 81.04 ± 4.89 years), were randomly selected from the patients attending the Dental Unit of the University Of L’Aquila and were assigned to Control Group. Thirty subjects (15 females/15 males; 83.16 ± 6.017 with cognitive impairments were randomly selected from hospitalized patients (Medically Assisted Residences RSA) and were assigned to Test Group. All subjects were living in L’Aquila, Italy at the time of the survey. Exclusion criteria were disorders interfering with psychometric assessment (severe blindness, terminal illness). Cognitive impairment was evaluated using the Mini-Mental State Examination (MMSE). The MMSE is the most commonly used instrument to gauge the severity of dementia by assessing cognitive functions. It comprises tests on orientation, registration, short-term memory, language use, comprehension, and basic motor skills (Figure 1). The score ranges from 0-30. Patients are considered to be in a mild stage of the disease when scoring 20 points or above; in a moderate stage when scoring between 10 and 19; and in a severe stage when scoring 9 or less []. In this study we considered scores of 20 and less represent low cognitive ability, and scores of 21 and greater representing normal cognitive function (MMSE >20 or MMSE <20). B-ADL (BASIC ACTIVITIES OF DAILY LIVING) was rated on five items: walking, eating, excreting, bathing and dressing. For each of these activities there some questions to be answered (Figure 2). Later respondents were classified as either ‘dependent’ or ‘independent’ by themselves or their caregivers. Respondents who rated themselves as completely independent on all five items were defined as ‘independent’, whereas respondents who rated themselves as dependent on one or more of the five B-ADL items were ‘dependent’ []. Also, for each patients was evaluated the presence of chronic medical diseases such as hypertension, cardiac diseases, diabetes, renal diseases, respiratory tract diseases, cerebrovascular diseases, rheumatoid arthritis and hepatic diseases as suggested by the recent literature [,].

A dental examination was carried out by two dentists calibrated as to the techniques, with the dentist and the subject in a sitting position under artificial lighting. The number of teeth was recorded for each subject (<20 or >20) []. The remaining teeth were defined as healthy, carious or treated teeth (including crowned, inlay, and abutment teeth for bridge work), inclusive of completely erupted third molars. In the study were not considered teeth with chronic periodontitis; also were not considered partial oftotal removable prosthesis [].

Statistical analysis

The Student’s t-test was used as test of significance and correlation coefficient were performed with respect to age, gender, systemic diseases, educational background, MMSE, B-ADL and number of teeth. The level of significance was assumed to be p≤0.05 for all tests. Test group were compared to the control group using Wilcoxon rank sum tests. The level of significance was assumed to be p≤0.01.

Results

Table 1 summarizes the epidemiological and clinical characteristics of control and test groups. The mean age of subjects in the cognitively normal group was 81.04 years (s.d. 4.89 years), and in the cognitively impaired group 83.16 years (s.d. 6.017 years). There was no significant difference in age between the groups. The number of teeth in relation to levels of schooling is not resulted significative. In the cognitively impaired group 26 subjects had fewer than 20 teeth (86.6%); in the cognitively normal group 9 subjects had fewer than 20 teeth (36%). The correlation between number of teeth and age in both groups is significative (p<0.05). There is also a significative correlation between subjects with renal diseases and type II diabetes and number of teeth (p<0.05). Finally a significative correlation is present between number of teeth and sex of the patients (p<0.05). The results of the Wilcoxon’s test revealed a significative correlation between MMSE in the two groups (p<0.01). There is also a significative correlation between the two groups and the educational background (p<0.01).

Table 1

Epidemiological and clinical characteristics of the two groups

C.I. (Test Group) C.N. (Control Group)
Age 83.16 (± 6.017) 81.04 (± 4.89)
    Male 15 15
    Female 15 10
Educational Background
    <8 years 21 2
    >8 years 9 23
Number of teeth
    >20 4 16
    <20 26 9
Diabete (Type II)
    Yes 5 5
    No 25 20
Renal Diseases
    Yes 11 8
    No 19 17
MMSE
    >20 5 24
    <20 25 1
B-ADL
    Ind 1 11
    Dip 29 14

Discussion

Dysfunctional mastication affects cognitive function, and reduced mastication contributes to senile dementia, Alzheimer’s disease, and a declining quality of life in the elderly. In particular, the systemic effects of tooth loss are an epidemiologic risk factor for Alzheimer’s disease. In fact, missing teeth, due to dental caries and periodontal diseases are common in the elderly, and reduce their ability to masticate []. Particularly, the loss of teeth induces pathologic changes in the hippocampus and deficits in learning and memory. In our work we looked for a relationship between edentulism and dementia in elderly patients in L’Aquila. Following occlusion and age-matching, masticatory function was compared between cognitively impaired and cognitively normal elderly. The results indicated a close association between masticatory function and age in both groups. In the cognitively impaired group 26 subjects had fewer than 20 teeth (86.6%); in the cognitively normal group 9 subjects had fewer than 20 teeth (36%). Both age and tooth loss are associated with each other. Age and tooth loss are expected to have a complex relationship with oral health-related quality of life []. Multiple tooth loss and difficulty chewing food were found to correlate with significantly greater odds of cognitive impairment. The difference remained significant even after history of depression and mental illness were added to the analysis []. Also, persons with cognitive impairment may have poorer ability to maintain oral hygiene, which would increase the risk of dental caries and periodontal disease, the major causes of tooth loss and limited ability to chew hard food [,]. The results indicated also that the ability to chew is not be associated with the basic activities of daily living (B-ADL). Later we analyzed occlusion and pathologies (renal diseases and type II diabetes) in both groups. The results showed a relationship between number of teeth (<20) and subjects with these diseases (p<0.05). Theadults with diabetes are at higher risk of experiencing tooth loss and edentulism than are adults without diabetes. However, although the association between diabetes and periodontal disease is well established, health care professionals also need to recognize the risk of tooth loss and its effect on quality of life among people with diabetes [,]. Also periodontal infection and tooth loss contribute to chronic kidney disease []. Finally we compared number of teeth and gender (p<0.05). 15 males have less of 20 teeth, and 19 female have less of 20 teeth. This outcome seems to be the same from others outcomes. Infact, the prevalence of edentulism among the elderly Italian population studied was at the high end among Western countries, and higher in women than in men []. In women, tooth loss is correlated with aging, female events (pregnancies, menopausal status), and living alone. In men, aging and smoking are important determinants of edentulism, which is associated with the risk condition of hypoalbuminemia.

Difficulty in chewing was associated with dentition type [,]. The educational background is not correlated with the number of teeth but is correlated with the groups. For this reason, theanalysis shows that the patients with an elevated educational background have also greater cognitive ability. A recent review and meta-analysis demonstrates robust evidence that a high level education in early life is related with a significant reduction both in the prevalence and incidence of dementia, including Alzheimer’s disease and vascular dementia. These results are in accordance with the Cognitive Reserve hypothesis, which assumes some aspects of life experience such as education protects against the onset of dementia. Education also influences the course and outcome of the disease in terms the pattern of cognitive decline and underlying brain pathology. As a prevention strategy Ritchie suggests that increasing a population’s ability to use skills, knowledge, and experience (crystallized intelligence) as well as increasing vegetable consumption, and eliminating depression and diabetes would have a greater impact on the prevalence and incidence of dementia than modifying known genetic risk factors []. Subjects with dementia or cognitive impairments, by our data, obviously have an MMSE lower as suggested by literature [,]. The results of the study show a clear correlation between tooth loss and cognitive function. The clinical relevance of these results is evident. In the general population, and in those nursing facilities caring for persons with dementia in particular, attention and priority should be given to prevention of loss of masticatory function and treatment of oral impairments to stabilize or even improve cognition.

Disclosure of conflict of interest

None.

References

1. Nakata M. Masticatory function and its effects on general health. Int Dent J. 1998;48:540–548. [PubMed[]
2. Scherder E, Posthuma W, Bakker T, Vuijk PJ, Lobbezoo F. Functional status of masticatory system, executive function and episodic memory in older persons. J Oral Rehabil. 2008;35:324–336. [PubMed[]
3. Proschel PA, Raum J. Preconditions for estimation of masticatory forces from dynamic EMG and isometric bite force-activity relations of elevator muscles. Int J Prosthodont. 2001;14:563–569. [PubMed[]
4. Onozuka M, Fujita M, Watanabe K, Hirano Y, Niwa M, Nishiyama K, Saito S. Age-related changes in brain regional activity during chewing: a functional magnetic resonance imaging study. J Dent Res. 2003;82:657–660. [PubMed[]
5. Momose T, Nishikawa J, Watanabe T, Sasaki Y, Senda M, Kubota K, Sato Y, Funakoshi M, Minakuchi S. Effect of mastication on regional cerebral blood flow in humans examined by positron-emission tomography with (1)(5)O-labelled water and magnetic resonance imaging. Arch Oral Biol. 1997;42:57–61. [PubMed[]
6. Onozuka M, Fujita M, Watanabe K, Hirano Y, Niwa M, Nishiyama K, Saito S. Mapping brain region activity during chewing: a functional magnetic resonance imaging study. J Dent Res. 2002;81:743–746. [PubMed[]
7. Weijenberg RA, Scherder EJ, Lobbezoo F. Mastication for the mind–the relationship between mastication and cognition in ageing and dementia. Neurosci Biobehav Rev. 2011;35:483–497. [PubMed[]
8. Ono Y, Yamamoto T, Kubo KY, Onozuka M. Occlusion and brain function: mastication as a prevention of cognitive dysfunction. J Oral Rehabil. 2010;37:624–640. [PubMed[]
9. Kalaria RN, Maestre GE, Arizaga R, Friedland RP, Galasko D, Hall K, Luchsinger JA, Ogunniyi A, Perry EK, Potocnik F, Prince M, Stewart R, Wimo A, Zhang ZX, Antuono P World Federation of Neurology Dementia Research Group. Alzheimer’s disease and vascular dementia in developing countries: prevalence, management, and risk factors. Lancet Neurol. 2008;7:812–826. [PMC free article] [PubMed[]
10. Plassman BL, Havlik RJ, Steffens DC, Helms MJ, Newman TN, Drosdick D, Phillips C, Gau BA, Welsh-Bohmer KA, Burke JR, Guralnik JM, Breitner JC. Documented head injury in early adulthood and risk of Alzheimer’s disease and other dementias. Neurology. 2000;55:1158–1166. [PubMed[]
11. Cedazo-Minguez A. Apolipoprotein E and Alzheimer’s disease: molecular mechanisms and therapeutic opportunities. J Cell Mol Med. 2007;11:1227–1238. [PMC free article] [PubMed[]
12. Kramer AF, Colcombe SJ, McAuley E, Eriksen KI, Scalf P, Jerome GJ, Marquez DX, Elavsky S, Webb AG. Enhancing brain and cognitive function of older adults through fitness training. J Mol Neurosci. 2003;20:213–221. [PubMed[]
13. Fillit H, Nash DT, Rundek T, Zuckerman A. Cardiovascular risk factors and dementia. Am J Geriatr Pharmacother. 2008;6:100–118. [PubMed[]
14. Miura H, Yamasaki K, Kariyasu M, Miura K, Sumi Y. Relationship between cognitive function and mastication in elderly females. J Oral Rehabil. 2003;30:808–811. [PubMed[]
15. Kim JM, Stewart R, Prince M, Kim SW, Yang SJ, Shin IS, Yoon JS. Dental health, nutritional status and recent-onset dementia in a Korean community population. Int J Geriatr Psychiatry. 2007;22:850–855. [PubMed[]
16. Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12:189–198. [PubMed[]
17. Hindmarch I, Lehfeld H, de Jongh P, Erzigkeit H. The Bayer Activities of Daily Living Scale (B-ADL) Dement Geriatr Cogn Disord. 1998;9(Suppl 2):20–26. [PubMed[]
18. Moriya S, Tei K, Murata A, Yamazaki Y, Hata H, Muramatsu M, Kitagawa Y, Inoue N, Miura H. Associations between self-assessed masticatory ability and higher brain function among the elderly. J Oral Rehabil. 2011;38:746–753. [PubMed[]
19. Okamoto N, Morikawa M, Okamoto K, Habu N, Hazaki K, Harano A, Iwamoto J, Tomioka K, Saeki K, Kurumatani N. Tooth loss is associated with mild memory impairment in the elderly: the Fujiwara-kyo study. Brain Res. 2010;1349:68–75. [PubMed[]
20. Kondo K, Niino M, Shido K. A case-control study of Alzheimer’s disease in Japan–significance of life-styles. Dementia. 1994;5:314–326. [PubMed[]
21. Jain M, Kaira LS, Sikka G, Singh S, Gupta A, Sharma R, Sawla L, Mathur A. How do age and tooth loss affect oral health impacts and quality of life? A study comparing two state samples of gujarat and rajasthan. J Dent (Tehran) 2012;9:135–144. [PMC free article] [PubMed[]
22. Lexomboon D, Trulsson M, Wardh I, Parker MG. Chewing ability and tooth loss: association with cognitive impairment in an elderly population study. J Am Geriatr Soc. 2012;60:1951–1956. [PubMed[]
23. Marchetti E, Monaco A, Procaccini L, Mummolo S, Gatto R, Tete S, Baldini A, Tecco S, Marzo G. Periodontal disease: the influence of metabolic syndrome. Nutr Metab (Lond) 2012;9:88. [PMC free article] [PubMed[]
24. Marchetti E, Mummolo S, Di Mattia J, Casalena F, Di Martino S, Mattei A, Marzo G. Efficacy of essential oil mouthwash with and without alcohol: a 3-day plaque accumulation model. Trials. 2011;12:262. [PMC free article] [PubMed[]
25. Patel MH, Kumar JV, Moss ME. Diabetes and tooth loss: An analysis of data from the National Health and Nutrition Examination Survey, 2003-2004. J Am Dent Assoc. 2013;144:478–485. [PubMed[]
26. Pietropaoli D, Ortu E, Severino M, Ciarrocchi I, Gatto R, Monaco A. Glycation and oxidative stress in the failure of dental implants: a case series. BMC Res Notes. 2013;6:296. [PMC free article] [PubMed[]
27. Fisher MA, Taylor GW, Papapanou PN, Rahman M, Debanne SM. Clinical and serologic markers of periodontal infection and chronic kidney disease. J Periodontol. 2008;79:1670–1678. [PubMed[]
28. Musacchio E, Perissinotto E, Binotto P, Sartori L, Silva-Netto F, Zambon S, Manzato E, Corti MC, Baggio G, Crepaldi G. Tooth loss in the elderly and its association with nutritional status, socio-economic and lifestyle factors. Acta Odontol Scand. 2007;65:78–86. [PubMed[]
29. Fratiglioni L, Viitanen M, von Strauss E, Tontodonati V, Herlitz A, Winblad B. Very old women at highest risk of dementia and Alzheimer’s disease: incidence data from the Kungsholmen Project, Stockholm. Neurology. 1997;48:132–138. [PubMed[]
30. Ritchie K, Carriere I, Ritchie CW, Berr C, Artero S, Ancelin ML. Designing prevention programmes to reduce incidence of dementia: prospective cohort study of modifiable risk factors. BMJ. 2010;341:c3885. [PMC free article] [PubMed[]
31. Meng X, D’Arcy C. Education and dementia in the context of the cognitive reserve hypothesis: a systematic review with meta-analyses and qualitative analyses. PLoS One. 2012;7:e38268. [PMC free article] [PubMed[]
32. Letenneur L, Gilleron V, Commenges D, Helmer C, Orgogozo JM, Dartigues JF. Are sex and educational level independent predictors of dementia and Alzheimer’s disease? Incidence data from the PAQUID project. J Neurol Neurosurg Psychiatry. 1999;66:177–183. [PMC free article] [PubMed[]
33. Hock RS, Lee HB, Bienvenu OJ, Nestadt G, Samuels JF, Parisi JM, Costa PT Jr, Spira AP. Personality and Cognitive Decline in the Baltimore Epidemiologic Catchment Area Follow-up Study. Am J Geriatr Psychiatry. 2013 [Epub ahead of print] [PMC free article] [PubMed[]
34. Wolfsgruber S, Jessen F, Wiese B, Stein J, Bickel H, Mosch E, Weyerer S, Werle J, Pentzek M, Fuchs A, Kohler M, Bachmann C, Riedel-Heller SG, Scherer M, Maier W, Wagner M AgeCoDe study group. The CERAD Neuropsychological Assessment Battery Total Score Detects and Predicts Alzheimer Disease Dementia with High Diagnostic Accuracy. Am J Geriatr Psychiatry. 2013 [Epub ahead of print] [PubMed[]
Read More

Evaluation of Cervical Posture Following Rapid Maxillary Expansion: A Review of Literature

Published online 2014 Apr 4

Eleonora OrtuDavide PietropaoliMaurizio Ortu, Mario Giannoni, and Annalisa Monaco

Abstract

Objective:

The aim of this article is to elucidate the state of the art about how rapid maxillary expansion (RME) produces changes in natural head position (NHP) in terms of cephalometric angles or inclinometer data during the growth period, according to the available literature.

Methods:

Electronic search was done from January 2002 to June 2012 on PubMed, Cochrane and Scopus databases; in addition manual search was conducted as well. According to keywords, eleven papers were eligible for our purpose, but definitely six papers were selected in agreement with the inclusion/exclusion criteria.

Conclusions:

The current literature suggests that the potential relationship between RME and NHP is still unclear. In fact, although the change in the position of the head after the rapid palatal expansion is evident clinically, current orthodontic literature does not provide conclusive evidence about the nature of this relationship

INTRODUCTION

Rapid Maxillary Expansion (RME) – or Rapid Palatal Expansion (RPE) – was introduced in 1860s by Angell for the treatment of maxillary constriction and consisted of a shaft with tubular nuts that was rotated using a wrench made from a dime []. To date, new technologically advanced tools have been introduced, but the final aim remains the same. Recent literature suggests that RME is the most predictable tool for the correction of the posterior cross bite, due to deficiency of maxillary width, providing to expand the transverse dimension of the palate and maxillary dental arch []. RME produces the separation of the two maxillary halves using an orthopaedic approach introducing lateral forces on horizontal maxillary processes and a medial force, produced by a screw activated twice a day []. When the mid-palatal suture is kept expanded for a retention period, the space obtained by separating the two halves of maxilla is replaced by newly formed bone []. Several Authors report changes in facial morphology and head posture caused by RME []. According to these reports, recent literature suggests modification on Natural Head Position (NHP) during and after RME application. Investigating the possible impact of RME in terms of change in NHP is a great and actual topic that needs to be deepened, because more aspects remain unclear. At today, the gold standard for studying NHP changes is the cephalometric analysis, but, due to the lack of specific standardized protocols to this aim, it leads to inability to compare results of different papers, and more powerful systematic or meta-analytic considerations couldn’t be done. Different ways of evaluation of NHP do not allow a systematic review of literature or meta-analysis.

However, the influence of RME in NHP can be analyzed with reference to objective measurements from studies using comparable cephalometric analyses or inclinometer data.

METHODS

Research Strategy

The following electronic databases were selected for search from January 2002 to June 2012: PubMed, Cochrane and Scopus. The database queries were performed independently by two reviewers (EO and DP) according to key words listed in Table 11. Disagreement regarding inclusion was resolved by discussionTo avoid inappropriate exclusions, adjectives, nouns, plural and singular forms of all terms were used. No language restriction was applied.

Table 1.

Electronic databases used and search strategy.

Database Search Strategy
Key words/MeSH
Pubmed
http://www.ncbi.nlm.nih.gov/pubmed/
Rapid palatal expansion(s) OR
Rapid maxillary expansion(s) OR
RME OR
Scopus
www.scopus.com/home.url
Rapid maxillary expansion treatment(s) OR
Rapid palatal expansion treatment(s) OR
Cochrane
www.the cochranelibrary.com/view/0/index.html
RPE OR
Rapid palatal disjunction(s) OR
Rapid maxillary disjunction(s) OR
Rapid palatal expansion technique(s) OR
Rapid maxillary expansion technique(s) AND
Changes in head posture OR
Changes in natural head position OR
Changes in cervical posture

In addition, a manual search was performed in the following journals: European Journal of Orthodontics, American Journal of Orthodontics and Dentofacial Orthopedics, Angle Orthodontist and Australian Orthodontic Journal from January 2002 to June 2012.

The search resulted in a total of 11 articles and, subsequently, a final eligibility screening was conducted to verify the agreement with the inclusion and exclusion criteria listed below.

Inclusion Criteria

  • Randomized controlled trials (RCTs) and historical controls for clinical trials (HCTs) [];

  • Follow-up of at least 6 months after RME/RPE therapy;

  • Subjects during growth period (<16 years);

Exclusion Criteria

  • Studies involving patients with systemic diseases, psychosocial impairment, craniofacial abnormalities, or skeletal open bite;

  • Absence of complete data from the beginning of therapy to the end of the follow- up [,];

  • Studies without the use of lateral cervical radiography and inclinometer to evaluate the effects and changes of NHP after RME RPE [];

  • No data for the evaluation of inclusion /exclusion criteria.

A total of six articles were included according to both inclusion and exclusion criteria. These are summarized in Table 22 with notations regarding Authors, year of publication, sample size, treatments and cephalometric index or inclinometer index.

Table 2.

Characteristics of the selected studies. Cephalometric indexes – CVT: the upper part of the cervical spine. EVT: the lower part of the cervical spine. OPT: odontoid line. SN: anterior cranial base. PP: palatal plane. MP: mandibular plane. CVT/EVT: cervical lordosis angle. OPT/Ver: odontoid angle. EVT/Ver: lower cervical column angle. CVT/ Ver: upper cervical column angle. SN/Ver: the downward opening angle between the SN and true vertical lines. PP/Ver: the downward opening angle between the palatal and true vertical lines. MP/Ver: the downward opening angle between the mandibular and true vertical lines. SN/OPT: the downward opening angle between OPT and SN lines. SN/CVT: the downward opening angle between CVT and SN lines. PP/OPT: the downward opening angle between OPT and palatal lines. PP/CVT: the downward opening angle between CVT and palatal lines. MP/OPT: the downward opening angle between OPT and mandibular lines. MP/CVT: the downward opening angle between CVT and mandibular lines. OPT/NSL: the angle formed by the line tangent to the odontoid process (CV2tp) through cv2ip (the most inferior and posterior point on the corpus of the second cervical vertebra), and the nasion–sella line. CVT/NSL: the angle formed by the line tangent to the odontoid process through cv4ip (the most inferior and posterior point on the fourth cervical vertebra), and the nasion-sella line. OPT/CVT: the angle between the odontoid process tangent through cv2ip and the tangent to the odontoid process through cv4ip. NSL/VER: the angle between the nasion–sella line and true vertical. OPT/HOR: the angle between the line OPT and true horizontal. CVT/HOR: the angle between the line CVT and true horizontal. NSL/ML (cranium basis inclination related to the mandible). NSL/CVT (cranium inclination in relation to C2 and in relation to the cervical spine). Transversal roll and sagittal pitch are the units used in the inclinometer studies. RPE: rapid palatal expansion. RME: rapid maxillary expansion.

Study Sample Size Treatment Design Age Sex Type of Treatment Criteria of Study
Yagci et al.
[12]
38 23 patients (test)
15 patients (control)
10.1 ±1.1
9.7±1.4
20 F
18 M
RME Transversal roll
Sagittal pitch
Tecco et al.
[13]
45 23 patients (test)
22 patients (control)
8-15 years
8.1±2.0
45 F RPE CVT/EVT
OPT/Ver
CVT/Ver
EVT/Ver
SN/OPT
SN/CVT
PP/OPT
PP/CVT
MP/OPT
MP/CVT
Mc Guinness et al. [14] 79 43 patients (test)
36 patients (control)
10-16 years  49F
30M
RME OPT/NSL
CVT/NSL
OPT/CVT
NSL/VER
OPT/HOR
CVT/HOR
Tecco et al.
[15]
45 23 patients (test)
22 patients (control)
8-15 years
8.1±2.0
45 F RPE CVT/EVT
OPT/Ver
CVT/Ver
EVT/Ver
SN/OPT
SN/CVT
PP/OPT
PP/CVT
MP/OPT
MP/CVT
Yagci et al.
[16]
45 15 patients (test 1)
15 patients (test 2)
15 patients
(control)
9.6±1.3
9.5±1.5
9.8±1.6
22F
23M
RPE;
Conventional facemask therapy;
Modified facemask therapy
Transversal roll
Sagittal pitch
Kilinc et al.
[17]
35 18 patients (test)
17 patients
(control)
10.9
10.9
20 F
15M
RPE NSL/ML
NSL/CVT

RESULTS

Six studies were identified for the aim of our paper. In two studies, inclinometer was used for the evaluation of outcomes [,]; in the others, cephalometric indexes were used as described in Table 22 and in the Figs. (11-33) [,]. The inclinometer and a portable data logger were used to collect the dynamic head posture data. The inclinometer were placed on the eyeglesses’ arms. The right sensor was positioned parallel to the sagittal plane to determine changes in pitch and the left sensor was positioned vertical to the sagittal plane to determine changes in roll (transversal roll and sagittal pitch). Although an index-matched analysis could not be performed, due to the use of different indexes between studies, an evaluation by comparable measurements was conducted.

An external file that holds a picture, illustration, etc. Object name is TODENTJ-8-20_F1.jpg

Cephalometric indexes used in two articles [13, 15]. CVT: the upper part of the cervical spine. A line through cv2tg and cv4ip. EVT: the lower part of the cervical spine. A line through cv4ip and cv6ip. OPT: odontoid line. A line through cv2tg and cv2ip. SN: anterior cranial base. A line through point S and point N. PP: palatal plane. A line through posterior nasal spine (pns) and anterior nasal spine (ans). MP: mandibular plane. A tangent line to the inferior border of the mandible. Variables: CVT/EVT: cervical lordosis angle. The downward opening angle between the CVT and EVT line. OPT/Ver: odontoid angle. The downward opening angle between the OPT and true vertical lines. EVT/Ver: lower cervical column angle. The downward opening angle between the EVT and true vertical lines. CVT/ Ver: upper cervical column angle. The downward opening angle between the CVT and true vertical lines. SN/Ver: the downward opening angle between the SN and true vertical lines. PP/Ver: the downward opening angle between the palatal and true vertical lines. MP/Ver: the downward opening angle between the mandibular and true vertical lines. SN/OPT: the downward opening angle between OPT and SN lines. SN/CVT: the downward opening angle between CVT and SN lines. PP/OPT: the downward opening angle between OPT and palatal lines. PP/CVT: the downward opening angle between CVT and palatal lines. MP/OPT: the downward opening angle between OPT and mandibular lines. MP/CVT: the downward opening angle between CVT and mandibular lines.

An external file that holds a picture, illustration, etc. Object name is TODENTJ-8-20_F3.jpg

Craniocervical angulations used in the study of Kilinc et al [17]. Reference points and angular measurements. Reference points (Linder-Aronson, 1970): Hyoid (hy), the most postero-superior point on the body of the second cervical vertebra (cv2); cv2tg, the most postero-inferior point on the body of cv2; cv2ip, the most postero-inferior point on the body of cv2; cv4ip, the most antero-inferior point on the body of the fourth cervical vertebra (cv4ia); ad2, the intersection between a line from posterior nasal spine (pns) to the midpoint of a line joining basion (ba) and sella (s) and the posterior contour of the adenoid soft tissue shadow; ad1, the intersection between a line from pns to ba and the posterior contour of the adenoid soft tissue shadow; APW, the anterior pharyngeal wall along the line intersecting cv2ia and hy; PPW, the posterior pharyngeal wall along the line intersecting cv2ia and hy; APW′, the anterior pharyngeal wall along the line intersecting cv4ia and hy; PPW′, the posterior pharyngeal wall along the line intersecting cv4ia and hy. Angular measurements: 1-SNA, 2-SNB, 3-ANB, 4-U1 to NSL, 5-L1 to ML, 6-NSL/ML, 7-NSL/CVT; NSL, nasion sella line; ML, mandibular plane; NSL-CVT, the angle between line NSL and the line from cv4ip to cv2ip (cervical vertebra tangent).

Yagci et al. [] studied the changes in NHP after RME in 38 subjects. The treatment and the control groups consisted of 23 and 15 patients, respectively. The test subjects underwent RME treatment using full cap acrylic device. The midline expansion screw was activated twice a day for the first week and once a day thereafter, until the desired change in the transverse dimension was achieved. The appliance was left in place for one month after the active expansion period, and then it was used as a removable retention tool. An inclinometer and a portable data logger were used to collect the NHP data. The NHP recordings were made at the initial stage of appliance placement and at the end of the RME therapy, both in the treatment and in the control groups (mean observation period: 7-8 months). Subjects in the control group did not undergo any type of treatment during the observation period. The mean difference between initial and final NHP was 0.31° and this difference was not statistically significant.

Tecco et al. [] studied the long-term effects of RPE on nasopharyngeal airway size, head posture, and cervical curvature angle in children and adolescents with nasal obstruction using the RPE appliance. Twenty-three female patients aged 8 to 15 years were included in the test group, and 22 age- and sex-matched patients were included in the control group. The protocol was: 4 turns on the first day followed by 2 turns per day until the required expansion was achieved. Lateral cephalograms were used to measure the craniocervical angulation and the nasopharyngeal airway adequacy. The long-term evaluation was 12 months. One year after expansion, nasopharyngeal airway adequacy, measured by Pm-Ad 2, increased by 5.3 mm, the angles SN/OPT decreased by 5.1, the angle PP/OPT decreased by 4.36, and the angle MP/OPT decreased by 5.12 []. All of these results were statistically significant. These findings indicated a reduction in head elevation. The correlation coefficients indicated a significant correlation between nasopharyngeal airway size and craniocervical angulation. The authors concluded that an improvement of nasopharyngeal airway adequacy as a result of RPE is associated with a decrease in craniocervical angles and an increase in cervical curvature angle and head flexion (p<0.05).

Mc Guinness et al. [] studied the long-term effects of RME with a bonded-type appliance, in craniocervical angulation. The treatment protocol was of 2 turns per day for an average of 3 weeks. This study used lateral cephalograms to measure the craniocervical angulations. Patients from 10 to 16 years of age were included: 39 subjects in the test group and 36 patients in the control group. The long-term evaluation was completed 12 months later. One year after expansion of the palate, the angle NSL/VER decreased by 3.14 degrees, and the angles OPT/HOR by 2.13 degrees and CVT/HOR by 2.55 degrees []. All these results were statistically significant. These findings indicate a reduction in head elevation and, according to the authors, is consistent with improved nasal respiration.

Tecco et al. [] studied the changes in head posture after rapid maxillary expansion in 45 mouth breathing girls (8-15 years of age). The treatment group comprised 23 girls treated with RME appliance with a protocol of 4 turns on the first day followed by 2 turns per day until the required expansion was achieved, the control group comprised 22 girls followed about 8 months before beginning therapy and became untreated controls. Dental casts and lateral skull radiographs exposed in natural head position were obtained at the first visit and 6 months later for all subjects. In the girls under active treatment there was a statistically significant increase of pm-Ad 2 (p< 0001), a significant increase of the cervical lordosis angle (p< 0001), a flexion of the head (p<0001), and a decrease in the craniocervical angulation (p<0001). No significant changes were seen in the control group. RME is capable of increasing nasopharyngeal airway adequacy in girls, and consequently these results confirm an increase of head flexion.

Yagci et al. [] valuated the effects of varying force directions on the dynamic measurement of natural head position and orofacial airway dimensions of Class III patients (30 patients divided in two groups, conventional and modified facemask therapies) during maxillary orthopedic protraction compared with an untreated control group (15 patients). The mean age was about 10 years. Natural head position measurements, obtained trought the use of an inclinometer and cephalometric records were obtained from all subjects before and after treatment or the control period (approximately 1 year). Both treatment groups showed statistically significant changes in the sagittal measurements of natural head position and upper pharynx. In the control group, the only statistically significant change was an increased upper pharynx measurement (p=020). Statistically significant natural head position differences were found in the conventional (6.4° flexion) and the modified (5.7° flexion) facemask groups when compared with the controls. The modified facemask group also showed significant changes in aerial (p=.003) and total (p<.001) areas of the airway measurements compared with the control group. No statistically significant differences were observed between the 2 treatment groups. These findings suggest that modified and conventional facemask therapy with expansion have significant cranial flexion effects on the dynamic measurements of natural head position. Additionally, the modified facemask procedure showed significant effects on the orofacial airway dimensions compared with the initial values and the values of the untreated controls.

Kilinc et al. [] examined the effects of rapid palatal expansion (RPE) and maxillary prolonging headgear therapy in 18 patients with a skeletal Class III malocclusion and on upper airway dimensions compared with 19 patients of the untreated control group. In both groups the mean age was 11 years. Pre- and post-treatment cephalometric lines and angles was analyzed in both groups. The angles analyzed to evaluate the changes in the position of the head have been NSL/ML and NSL/CVT. The results showed that the first angle is increased considerably, as did the second. After treatment, the head was in a more extended position in relation to the cervical vertebrae, as demonstrated by a mean increase of 2.64 degrees in the NL/CVT angle.

DISCUSSION

This review of literature focused on changes produced by RME on head posture. Because of the great complexity of cranial anatomy and function, several measurement methods have different objectives and can complete each other to assess the real changes in NHP. Although the cephalometric radiographs in the selected studies were taken according to a standardized technique, the different structure superimpositions and image magnifications did not always allow accurate quantifications of the changes. However, also important was the lack of use of blinded examinations of the subjects and control. Many errors may result from the incorrect use of the head positioners for the cephalometric technique. These factors could influence the measured variable of head posture [,]. Tecco et al. analyzed only female subjects in these standardized studies because female have a particular vertebral curvature, with a partial reverse curvature more easily visible. Altough these findings confirm this hypothesis, it is very uncertain if these changes in posture head are clinically relevant [,]. Also, analyzing mouth breathers girls they excluded “patients with open bite” as well as those with “nasal allergic conditions or airway obstructions due to adenoids”. However, they did not report, how they gathered this information. In addition, these conditions are very often present in patients presenting with the need for maxillary expansion. A possible hypothesis for the role of RME in postural changes may be that the increase in palatal width results in enlargement of the pharyngeal airway space, improvement in respiratory function and flexion of the head on the cervical column, with an increase in the cervical curvature angle and a decrease in craniocervical angulation. In this mechanism, the muscular–neural network could play an important role []. There is also in the recent literature, an important case report in which the authors try to hypothesized that the changes in the palatal width obtained with RME appliances can involve other bone structures such as the tongue’s muscles and the suboccipital muscles. These structures are functionally bound and cooperate to manage the head and neck position, []. Solow et al. [] and, Solow and Greve [] observed a significant relationship between nasal airway adequacy and changes in posture head. Infact, the common interpretation of these results is that the mouth breathers have an extension of the head and this could an important compensation for nasal airway inadequacy []. Mc Guinness et al. [], used an historical control sample because it was not possible to obtain an untreated control group and exposed them to radiographs for comparisons. The recordings of the study of Kilinc et al. [] have limitations because were based on two dimensional cephalometric measurements, and not on three dimensional measurements. Finally, the authors in two studies [,] have found changes in the position of the head after the treatment of rapid palatal expansion before to continue the therapy with the advancement of the maxilla (subjects with a skeletal class III malocclusion). The studies that used the inclinometer showed a good reproducibility of recordings over time and a lower mean method error [,]. Yagci et al. [] used the inclinometer and the recordings were accurate. However, the measurements should be reproducible over long periods, and the accuracy of the recordings should not depend entirely on the operator’s skill. In this study both females and males were studied, but the sex differences were not determined. The increase in flexion of the head was found in both treatment groups []. Yagci et al. in the other study analyzed [] supported the hypothesis in which there is no statistically significant difference between RME and changes in natural head position in the test group and in the control group. The results suggested that dynamic measurement of NHP recorded by an inclinometer apparatus, which is most probably dictated by maintenance of visual axis, is not significantly affected by RME. However, this does not necessarily mean that the craniocervical relation is not affected by RME. Although the few studies that used an alternative technique did not consistently find differences between subjects and controls [,]. By analysis of the literature it appears to be strongly evident that of the different operational protocols were used in the different studies (Tab.II), both as regards the use of equipment, the methods of analysis (for example inclinometer or cephalometric angles), and the choice (sex, age) of the components of the two groups (test group and control group).

CONCLUSION

From the analysis of the recent scientific literature, it has been noted that RME causes not only dentoalveolar changes, but also craniocervical structure changes []. It has been hypothesized that, since the maxillary bones form a half of the nasal cavity’s structures, when the midpalatal suture is open, the nasal cavity’s lateral walls are also displaced apart, and its volume increases, and upper airway resistance decreases over time. Head posture had also been associated with respiratory function, and increased craniocervical angulation was observed as a functional response to facilitate oral breathing to compensate for nasal obstruction [,].

In conclusion, this review of literature suggests that the potential relationship between RME and head posture is still unclear. Although it is evident a change in the position of the head after the rapid palatal expansion, it cannot be quantified in an objective way since the few studies reported in the literature did not use the same evaluation method. Cephalometric angles, and different measurement techniques do not allow to quantify properly the actual change. There is a need for further well controlled long-term clinical trials using most precise methods to measure craniocervical anatomy and its function and to look at the data for a longer observation period. There is also a need of Long-term stability results in the Rapid palatal expansion [,].

An external file that holds a picture, illustration, etc. Object name is TODENTJ-8-20_F2.jpg

Craniocervical angulations used in the study of Mc Guinness at al. [14] OPT/NSL: the angle formed by the line tangent to the odontoid process (CV2tp) through cv2ip (the most inferior and posterior point on the corpus of the second cervical vertebra), and the nasion–sella line. CVT/NSL: the angle formed by the line tangent to the odontoid process through cv4ip (the most inferior and posterior point on the fourth cervical vertebra), and the nasion-sella line. OPT/CVT: the angle between the odontoid process tangent through cv2ip and the tangent to the odontoid process through cv4ip. NSL/VER: the angle between the nasion–sella line and true vertical. OPT/HOR: the angle between the line OPT and true horizontal. CVT/HOR: the angle between the line CVT and true horizontal.

ACKNOWLEDGEMENTS

Declared none.

CONFLICT OF INTEREST

The authors confirm that this article content has no conflicts of interest.

REFERENCES

1. Angell EH. Treatment of irregularity of the permanent or adult teeth. Dental Cosmos. 1860;1:540. []
2. Zuccati G, Casci S, Doldo T, Clauser C. Expansion of maxillary arches with crossbite: a systematic review of RCTs in the last 12 years. [Epub 2011/12/14] Eur J Orthod. 2011 [PubMed[]
3. Mundstock KS, Barreto G, Meloti AF, Araujo MA, dos Santos-Pinto A, Raveli DB. Rapid maxillary expansion with the Hyrax appliance: an occlusal radiographic evaluation study. World J Orthod. 2007;8(3):277–84. [PubMed[]
4. Franchi L, Baccetti T, Lione R, Fanucci E, Cozza P. Modifications of midpalatal sutural density induced by rapid maxillary expansion: a low-dose computed-tomography evaluation. Am J Orthod Dentofac Orthop [discussion 12A-13A] Epub 2010/04/07. 2010;137(4):486–8. [PubMed[]
5. Haas AJ. The treatment of maxillary deficiency by opening the midpalatal suture. Angle Orthod. 1965;35:200–17. [PubMed[]
6. Blackwelder WC. Showing a treatment is good because it is not bad: when does “noninferiority” imply effectiveness?. Control Clin Trials. 2002;23(1):52–4. [PubMed[]
7. Di Vece L, Faleri G, Picciotti M, Guido L, Giorgetti R. Does a transverse maxillary deficit affect the cervical vertebrae?. a pilot study. Am J Orthod Dentofac Orthop. 2010;137(4):515–9. [PubMed[]
8. Baratieri C, Alves MJr, de Souza MM, de Souza Araujo MT, Maia LC. Does rapid maxillary expansion have long-term effects on airway dimensions and breathing?. Am J Orthod Dentofac Orthop. 2011;40(2):46–56. [PubMed[]
9. Guijarro-Martinez R, Swennen GR. Cone-beam computerized tomography imaging and analysis of the upper airway: a systematic review of the literature. Int J Oral Maxillofac Surg. 2011;40(11):227–37. [PubMed[]
10. Domann CE, Kau CH, English JD, Xia JJ, Souccar NM, Lee RP. Cone beam computed tomography analysis of dentoalveolar changes immediately after maxillary expansion. Orthodontics. 2011;2(3):202–9. [PMC free article] [PubMed[]
11. Gamble J, Lagravere MO, Major PW, Heo G. New statistical method to analyze three-dimensional landmark configurations obtained with cone-beam CT: basic features and clinical application for rapid maxillary expansion. Korean J Radiol. 2012;13(2):126–35. [PMC free article] [PubMed[]
12. Yagci A, Uysal T, Usumez S, Orhan M. Rapid maxillary expansion effects on dynamic measurement of natural head position. Angle Orthod. 2011;81(5):850–5. [PubMed[]
13. Tecco S, Caputi S, Festa F. Evaluation of cervical posture following palatal expansion: a 12-month follow-up controlled study. Eur J Orthod. 2007;29(1):45–51. [PubMed[]
14. McGuinness NJ, McDonald JP. Changes in natural head position observed immediately and one year after rapid maxillary expansion. Eur J Orthod. 2006;28(2):126–34. [PubMed[]
15. Tecco S, Festa F, Tete S, Longhi V, D’Attilio M. Changes in head posture after rapid maxillary expansion in mouth-breathing girls: a controlled study. Angle Orthod. 2005;75(2):171–6. [PubMed[]
16. Yagci A, Uysal T, Usumez S, Orhan M. Effects of modified and conventional facemask therapies with expansion on dynamic measurement of natural head position in Class III patients. Am J Orthod Dentofac Orthop. 2011;140(5):e223–31. [PubMed[]
17. Kilinc AS, Arslan SG, Kama JD, Ozer T, Dari O. Effects on the sagittal pharyngeal dimensions of protraction and rapid palatal expansion in Class III malocclusion subjects. Eur J Orthod. 2008;30(1):61–6. [PubMed[]
18. Monaco A, Tepedino M, Sabetti L, Petrucci A, Sgolastra F. An adolescent treated with rapid maxillary expansion presenting with strabismus: a case report. J Med Case Rep. 2013;7(1):222. [PMC free article] [PubMed[]
19. Solow B, Siersbaek-Nielsen S, Greve E. Airway adequacy head posture. and craniofacial morphology. Am J Orthod. 1984;86(3):214–23. [PubMed[]
20. Sorensen H, Solow B, Greve E. Assessment of the nasopharyngeal airway. A rhinomanometric and radiographic study in children with adenoids. Acta Oto-Laryngol. 1980;89(3-4):227–32. [PubMed[]
21. Ricketts RM. Respiratory obstruction syndrome. Am J Orthod. 1968;54(7):495–507. [PubMed[]
22. Oliveira De Felippe NL, Da Silveira AC, Viana G, Kusnoto B, Smith B, Evans CA. Relationship between rapid maxillary expansion and nasal cavity size and airway resistance: short- and long-term effects. Am J Orthod Dentofac Orthop. 2008;134(3):370–82. [PubMed[]
23. Zuccati G, Casci S, Doldo T, Clauser C. Expansion of maxillary arches with crossbite: a systematic review of RCTs in the last 12 years. Eur J Orthod. 2013;35(1):29–37. [PubMed[]
Read More

Relationship between Periodontitis and Pre-Eclampsia: A Meta-Analysis

Published online 2013 Aug 19

Fabrizio Sgolastra, Ambra PetrucciMarco SeverinoRoberto Gatto, and Annalisa Monaco

Abstract

Background

Studies have suggested controversial results regarding a possible association between pre-eclampsia (PE) and periodontal disease (PD) and no meta-analysis has been performed to clarify this issue.

Methods

A literature search of electronic databases was performed for articles published through March 24, 2013, followed by a manual search of several dental and medical journals. The meta-analysis was conducted according to the recommendations of the Cochrane Collaboration and PRISMA. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated. Heterogeneity was assessed with the χ2-based Cochran Q test and I2 statistic. The level of significance was set at P <0.05.

Results

Fifteen studies were included, including three cohort and 12 case-control studies. A positive association was found between PE and PD (OR 2.17, 95% CI 1.38–3.41, P = 0.0008). However, a high and significant heterogeneity was found (χ2 = 62.42, P<0.00001, I2 = 75%). In most cases, subgroup analysis had low power to detect significant differences between PE and non-PE groups.

Conclusion

Based on the findings of the meta-analysis, PD appears to be a possible risk factor for PE. However, given the important differences in the definitions and diagnoses of PD and PE among the studies, as well as their lack of good methodological quality, future trials are needed to confirm the results of the present meta-analysis.

Introduction

Infection and inflammation continue to be at the forefront of etiologic theories as causative factors of adverse pregnancy outcomes, such as stillbirth and growth restriction, that affect many women each year. Previous studies have demonstrated a link between infection or inflammation and preterm birth, preeclampsia (PE), and other adverse outcomes thought to be secondary to poor placentation . The prevalence of PE, a multisystem disorder of unclear etiology that is exclusive to human pregnancy, ranges from 2% to 7% in developed countries. PE results in high maternal and neonatal morbidity and mortality rates, attributable to complications affecting different organs and systems. In emerging countries, the prevalence of PE is more than 10% , and the condition is the main cause of maternal death .

PE occurs usually after 20 weeks of gestation. It is characterized by an abnormal vascular response to placentation, manifesting as generalized vasospasm, activation of the coagulation system, and reduced organ perfusion affecting the kidney, liver, and brain . Two syndromes are included in the definition of PE: maternal, characterized by endothelial cell activation, perturbations in volume and blood pressure control, gradual maternal blood pressure elevation, proteinuria, and generalized edema; and fetal, manifested primarily by intrauterine growth restriction . Putative PE risk factors include advanced maternal age, multifetal pregnancies, maternal prepregnancy obesity, pregestational hypertension, renal disorders, and diabetes mellitus . In recent years, infection has been reported to be important in the pathogenesis of PE, both in terms of its initiation and its potentiation .

Several studies have suggested that periodontal disease (PD), a chronic inflammatory oral infection, may be associated with an increased risk for PE development . PD affects 20% to 50% of pregnant women, especially economically disadvantaged women . In this inflammatory pathology, the dental plaque – which is a biofilm predominated by Gram-negative anaerobic microorganisms – destroys the tooth-supporting tissues. Oral microorganisms initiate PD, but the periodontal breakdown is primarily mediated by the host inflammatory response . PD may burden pregnant women systemically with endotoxins, inflammatory cytokines, and oxidative stressors at the maternal-fetal interface . Thus, PD may be a vascular stressor that plays a role in the development of PE in pregnant women.

Contradictory findings exist regarding the relationship between PD and PE , and a previous systematic review did not clarify this possible association . Therefore, there is a need for a systemic assessment of the literature on the possible association between PD and PE. The aim of the present systematic review and meta-analysis was to assess the scientific evidence on the possible association between PD and PE.

Materials and Methods

The present meta-analysis was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis  (PRISMA) guidelines.

Search

The following databases were searched from their earliest records through March 24, 2013: MEDLINE, Cochrane Controlled Clinical Trial Register, Cochrane Database of Systematic Reviews, Database of Abstracts of Reviews of Effects, CINAHL, Science Direct, ISI Web of Knowledge, and SCOPUS. The following search algorithm was used to explore databases, by using Boolean operators and the asterisk symbol (*) as truncation: (“Periodontitis”[Mesh] OR “Chronic Periodontitis”[Mesh] OR “Periodontal Diseases”[Mesh] OR “Periodontal Pocket”[Mesh] OR “Periodontal Attachment Loss”[Mesh] OR “Tooth Mobility”[Mesh] OR periodontitis OR periodontal disease* OR periodontal pocket* OR attachment loss OR alveolar bone loss OR pocket depth OR clinical attachment level) AND (“Pre-Eclampsia”[Mesh] OR “Eclampsia”[Mesh] OR “Hypertension, Pregnancy-Induced”[Mesh] OR preeclampsia OR pre-eclampsia OR eclampsia OR gestosis OR pregnancy hypertension OR pregnancy hypertensive). In the CINAHL, SCOPUS, ISI Web of Knowledge, and Science Direct databases, the MeSH terms were not used.

In addition, a manual search was performed of issues of the last 15 years of the following journals: Journal of Periodontology, Journal of Clinical Periodontology, Journal of Dental Research, Journal of Periodontal Research, Periodontology 2000, Journal of Dentistry, Journal of the American Dental Association, Journal of Clinical Dentistry, Clinical Oral Investigations, Acta Obstetricia and Gynecologica, Journal of Obstetrics and Gynaecology, British Journal of Obstetrics and Gynecology, American Journal of Obstetrics and Gynecology, and Obstetrics and Gynecology. To be as inclusive as possible, no restrictions were applied with regard to the publication year of the studies or to language. The references of all selected full-text articles and related reviews were scanned.

Study Selection

Screening was performed independently by two blinded reviewers (FS and MS). Interreviewer reliability in the study selection process was determined by the Cohen κ test, assuming an acceptable threshold value of 0.61 . In case of disagreement on the inclusion or the exclusion of studies, this issue was discussed until consensus was reached by the reviewers who selected the studies (FS and MS).

Eligibility Criteria

The study selection process was performed by two blinded reviewers (RG and AM) in two phases. In the first phase, the studies were analyzed according to the following inclusion criteria (A): 1) cross-sectional, prospective cohort or case-control studies, 2) studies analyzing the association between PD and PE, 3) PD defined by clinical or radiographic parameters, 4) studies reporting clear definition of PD and PE, and 5) studies conducted on adult human subjects (age >18 years). Only studies that met all inclusion criteria in (A) were admitted to the second phase, which consisted of the analysis of the preselected studies according to the following exclusion criteria (B): 1) studies including patients with systemic disease, 2) studies that did not report adjustment for known confounder factors, 3) studies not reporting adequate data, 4) ancillary or duplicate studies, and 5) no outcome of interest.

Data Extraction

Data were collected by two independent reviewers (FS and RG). The following data were extracted from the included studies: year of publication, country, study design, demographic characteristics of participants, definition of PE and PD, and main findings. If data were presented both numerically (in tables or text) and graphically (in figures), only numeric data were considered for extraction. The reviewers cross-checked all extracted data. Disagreements were resolved by discussion until consensus was reached.

Risk of Bias

Assessment of risk of bias was performed according to the Newcastle-Ottawa Scale by two independent reviewers (FS and AP). The level of agreement between reviewers was 0.76.

Quantitative Analysis

Measure of effect size

Data were combined for meta-analysis with a statistical software (RevMan, Version 5, 2008, The Nordic Cochrane Center, The Cochrane Collaboration, Copenhagen, Denmark). For dichotomous data, the odds ratio (OR) and 95% confidence interval (CI) were calculated. Due to the expected interstudy heterogeneity, a random effect model was used. The pooled effect was considered significant if P was <0.05. Forest plots for each meta-analysis present the raw data, OR (displayed as blocks), and CIs (displayed as lines) for the chosen effect, the heterogeneity statistic (I2), total number of participants per group, and overall OR in the random effect model.

Subgroup analysis

Subgroup analysis was performed according to the type of study (cohort or case-control study), severity of PD (mild, moderate, or severe), definition of PD (defined by probing pocket depth [PPD] and/or clinical attachment level [CAL]) and security of PD diagnosis (defined according to the criteria suggested by Nibali et al. 2013 ).

Heterogeneity

Heterogeneity was assessed by the χ2-based Q-statistic method and I2 measurement, with significance indicated by P<0.1.

Publication bias

The publication bias was investigated by two methods. Visual detection was used to analyze the funnel plots . Quantitative analysis was performed by the regression asymmetry test  and the trim-and-fill method . Publication bias was assessed with an additional statistical software (Stata IC version 10.1, StataCorp, College Station, Texas).

Results

Search Results

A total of 348 articles were found through the electronic and manual searches. After removing duplicates, 275 articles were found (inter-reviewer agreement, κ = 0.78), including 84 in MEDLINE, 4 in Cochrane Controlled Clinical Trial Register, 69 in CINHAL, 55 in Science Direct, 9 in Scopus, and 55 in ISI Web of Knowledge. Then, 234 papers were excluded on the basis of the evaluation of the title and abstract, leaving 41 articles to be assessed for eligibility (κ = 0.84). Of these, 11 articles were excluded in the first phase of the selection process, for not satisfying one or more inclusion criteria (κ = 1) . Fifteen of the remaining 30  articles were further excluded (κ = 1). Finally, 15 studies  qualified for inclusion in the systematic review and meta-analysis (κ = 1). The list of the excluded studies and their reasons for exclusion are provided in Table 1. A PRISMA flowchart is provided in Figure 1.

An external file that holds a picture, illustration, etc. Object name is pone.0071387.g001.jpg

PRISMA Flowchart of the search strategy.

Table 1

List of excluded studies and reasons for exclusion.
Study Year of publication Criteria for exclusion Type of study
Swati et al.  2012 A.2 Case-control study
Abati et al.  2012 B.3 Case-control study
Boggess et al.  2012 A.3 Cross-sectional study
Cetin et al.  2012 A.1 Review
Hirano et al.  2012 B.5 Case-control study
Piscoya et al.  2012 A.3 Case-control study
Swati et al.  2012 A.2 Case-control study
Lopez-Jaramillo et al.  2011 A.1 Comment
Sayar et al.  2011 B.2 Case-control study
Matevosyan  2011 A.1 Meta-analysis
Kunnen et al.  2010 A.1 Systematic review
Horton et al.  2010 B.4 Cohort study
Nabet et al.  2010 B.5 Case-control
Vergnes  2008 A.1 Systematic review
Ruma et al.  2008 B.4 Cohort study
Conde-Agudelo et al.  2008 A.1 Systematic review
Dasanayake  2007 A.1 Comment
Canacki et al.  2007 B.4 Case-control
Cota et al.  2006 B.4 Case –control
Khader et al.  2006 B.3 Meta-analysis
Castaldi et al.  2006 B.5 Cross-sectional study
Boggess et al.  2006 B.4 Cross-sectional study
Meurman et al.  2006 B.2 Retrospective study
Contreras et al.  2006 B.2 Case-control study
Xiong et al.  2006 A.1 Systematic review
Oettinger-Barak et al.  2005 B.2 Case-control study

Description of Included Studies

The main characteristics of the included studies are described in Table 2. Three of 15 included studies were cohort studies, whereas the remaining 12 were case-control studies. The definitions of PE and PD varied greatly among the included studies. The sample size varied from 54 patients in the study of Chaparro et al.  to 1206 in the study of Siqueira et al. . Three studies  did not found a positive association between PD and PE, whereas the remaining studies reported a significant association.

Table 2

Characteristics of the included studies.
Author, Year Design Country (Race/ethnicity) Population Age (y) Definition of PE Definition of PD Findings
Canakci, 2007 C/C Erzurum, Turkey 20 Mild PE, 18 Severe PE, 21 Control Mild PE: 24.1±3.9; Severe PE: 23.6±4.2; Control: 24.7±4.5 PE: DBP ≥90 mmHg and PU (300 mg/24h US) and edema; Mild PE: BP ≥140/90 mmHg on ≥2 occasions 6h apart, w/or w/o PU; Severe PE: SBP ≥160 or DBP ≥110 mmHg on 2 occasions ≥6h apart and PU ≥5 g/24h US or ≥3 l on dipstick in ≥2 random clean-catch samples ≥4h apart BOP and ≥4 mm PPD on 1–15 sites (Mild PD) or ≥15 sites (Severe PD) There was a significant association between mild to severe PE and severe PD.
Chaparro, 2012 C/C Santiago, Chile 11 PE, 43 Control PE: 28.91±6.11; Control: 27.19±7.18 BP >140/90 mmHg and PU (300 mg/24h US) ≥4 teeth w/≥1 sites w/PPD ≥4 mm and CAL ≥3 and BOP Increased IL-6 levels in GCF (OR  = 1.06, P = 0.02, CI 95% 1.007–1.117) in early pregnancy were associated with increased PE risk.
Ha, 2011 C/C Seoul, Korea 16 PE, 48 Control PE: 32.69±5.30 (21–40); Control: 32.69±4.40 (21–40) BP >140/90 mmHg on 2 occasions and 1+ or more PU on a random US CAL ≥3.5 mm on 2–3 sites (Localized PD) or on ≥4 sites (Generalized PD) of different teeth PE could be associated with maternal PD.
Wang, 2012 C/C Niigata, Japan (Japanese) 13 PE, 106 Control SBP ≥140 or DBP ≥90 mmHg on 2 occasions and PU (≥300 mg/d) after 20 GW >60% of sites with CAL ≥3 mm Polymorphism and subgingival DNA level of A. actinomycetemcomitans were significantly associated with PE, independent of PD.
Srinivas, 2009 Co Philadelphia, PA, USA (AA) 876 Patients >140/90 mmHg with PU CAL ≥3 mm on ≥3 teeth No significant association was found between PD and PE.
Siqueira, 2008 C/C Belo Horizonte (Multiethnic) 164 PE, 1042 Control SBP >140 or DBP >90 mmHg on 2 occasions after 20 GW and 1+ or more PU ≥4 mm and CAL ≥3 mm at the same site in ≥4 teeth Maternal PD was observed to be a risk factor associated with PE.
Boggess, 2003 Co NC, USA (Multiethnic) 39 PE, 763 Control SBP >140 or DBP >90 mmHg on 2 occasions and 1+ or more PU PPD ≥4 mm on 1–15 teeth or BOP+ (Mild PD) PPD ≥4 mm on >15 teeth (Severe PD) Severe PD at delivery was associated with increased PE risk (adj OR 2.4, 95% CI 1.1–5.3).
Da Silva, 2012 C/C Recife, Brazil 284 PE, 290 Control SBP ≥140 or DBP ≥90 mmHg and PU ≥300 mg/24h or 2+ PU on dipsticks, developed after 20 GW ≥4 teeth with ≥1 sites with PPD ≥4 mm and AL ≥3 mm in the same site PD could be a risk factor for PE.
Shetty, 2010 C/C 30 PE, 100 Control SBP ≥140 or DBP ≥90 mmHg on >2 occasions 4h apart and 1+ or more PU by dipstick on random US CAL ≥3 mm and PPD ≥4 mm PD at enrollment (OR  = 5.78, 95% CI 2.41–13.89) and w/i 48h of delivery (OR  = 20.15, 95% CI 4.55–89.29) may be associated with increased PE risk.
Taghzouti, 2012 C/C Quebec, Canada (Multiethnic) 92 PE, 245 Control SBP ≥140 or DBP ≥90 mmHg and 1+ or more PU ≥4 sites with PPD ≥5 mm and CAL ≥3 mm at the same sites PD was not associated with PE (adj OR  = 1.13, 95% CI  = 0.59 to 2.17).
Kumar, 2012 Co New Delhi 35 PE, 305 Control PE: 22.32±2.75; Control: 22.32±2.79 SBP ≥140 or DBP ≥90 mmHg and PU ≥300 mg/24h or 2+ PU on dipsticks, developed after 20 GW CAL and PPD ≥4 mm in ≥1 sites PD was significantly associated with PE.
Canakci, 2004 C/C Erzurum, Turkey (Turkish) 41 PE, 41 Control PE: 25.9±5.9; Control: 25.8±5.8 SBP ≥140 or DBP ≥90 mmHg and PU ≥300 mg/24h or 2+ PU on dipsticks, on 2 occasions ≥6h apart if 24h US is unavailable ≥ 4 teeth with ≥1 sites with PPD ≥4 mm and BOP+ and CAL ≥3 mm at the same site No. of sites with PD ≥4 mm and CAL ≥3 mm was higher among PE patients than among controls (P<0.01).
Lohsoonthorn, 2009 C/C Bangkok, Thailand (Thai) 150 PE, 150 Control SBP ≥140 or DBP ≥90 mmHg and PU ≥30 mg/dl (or 1+ on a urine dipstick) on ≥2 random specimens collected ≥4h apart. ≥1 teeth (Mild PD) or ≥2 nonadjacent teeth (Moderate or Severe PD) with interproximal sites showing PPD ≥4 mm and CAL ≥4 mm (Mild or Moderate PD) or ≥5 mm (Severe PD) Severe clinical PD was not associated with an increased risk of PE (adj OR  = 0.92, 95% CI: 0.26–3.28).
Kunnen, 2007 C/C Groningen, The Netherlands (Caucasian) 17 PE, 35 Control PE: 29.5±5.1; Control: 31.7±4.2 DBP ≥90 mmHg on 2 occasions and PU ≥30 mg/dl (or 1+ on a urine dipstick) on ≥2 random specimens collected ≥4h apart. BOP and PPD ≥4 mm on 1 – 15 sites (Mild PD) or >15 sites (Severe PE) Severe PD was found in 82% of the PE patients and 37% of the control group (P = 0.009).
Politano, 2011 C/C São Paulo, Brazil 58 PE, 58 Control PE: 28.62±6.93; Control: 24.69±5.37 SBP ≥140 or DBP ≥90 mmHg after 20 GW and PU ≥300 mg ≥2 sites w/PPD ≥4 mm and CAL ≥4 mm and BOP There was an association between PD and PE (adj OR 3.73, 95% CI 1.32–10.58).
Legend: C/C, case-control study; Co, cohort study; AA, African-American; GW, gestational weeks; adj, adjusted; US, urine specimen; PD, periodontal disease; PE, pre-eclampsia; PU, proteinuria; BP, blood pressure; SBP, systolic blood pressure; DBP, diastolic blood pressure; GCF, gingival crevicular fluid.

Quality Analysis

None of the included studies reached the maximum score of the Newcastle Ottawa Scale (Table 3). Only two studies  gained the maximum score in the Selection outcome; nine studies  had the maximum score in the Comparability outcome; and all studies had a partial score in the Exposure outcome.

Table 3

Risk of bias in included studies.
Study Selection (Max 4 *) Comparability (Max 2 *) Exposure (Max 3 *)
Boggess et al., 2003 *** * **
Canakci et al., 2004 *** ** **
Canakci et al., 2007 *** ** **
Chaparro et al., 2012 *** * **
Ha et al., 2011 **** ** **
Kumar et al., 2012 *** * **
Kunnen et al., 2007 *** ** **
Lohsoonthorn et al., 2009 *** ** **
Politano et al., 2011 *** ** **
Shetty et al., 2010 *** ** **
Siqueira et al., 2008 *** * **
Moura da Silva et al., 2012 **** ** **
Srinivas et al., 2009 *** * **
Taghzouti et al., 2012 *** ** **
Wang et al., 2012 *** * **

Results of the Meta-analysis

The results of the meta-analysis showed that an increased risk for PE was present for patients with PD (OR 2.17, 95% CI 1.38–3.41, P = 0.0008; Figure 2); however, a high and significant heterogeneity was found (χ2 = 62.42, P<0.00001; I2 = 75%).

An external file that holds a picture, illustration, etc. Object name is pone.0071387.g002.jpg

Forest plot for the association between PD and PE.

Subgroup analysis

An analysis of the results according to study type (Figure 3) revealed an increased risk of PE in PD patients in the case-control (OR 2.16, 95% CI 1.29–3.63, P = 0.004) and in the cohort studies (OR 2.20; 95% CI 0.66–7.36, P = 0.20). However, this increased risk only remained significant in the case-control subgroup. Heterogeneity was significant in both subgroups (χ2 = 44.57, P<0.00001, I2 = 75% for case-control, and χ2 = 18.21, P = 0.0001, I2 = 89% for cohort studies).

An external file that holds a picture, illustration, etc. Object name is pone.0071387.g003.jpg

Forest plot for the subgroup analysis according to the type of study design.

When the meta-analysis results were analyzed according to the definition of PD, an increased risk of PE was observed in all subgroups (Figure 4). However, it was significant only in the subgroup in which PD was defined by PPD and CAL (OR 2.50, 95% CI 1.54–4.04, P = 0.0002). Heterogeneity was high and significant for the subgroups in which PD was defined by PPD and CAL (χ2 = 36.55, P<0.0001, I2 = 75%) and by CAL alone (χ2 = 11.26, P = 0.004; I2 = 82%), but not for those in which PD was defined by PPD alone (χ2 = 0.16, P = 0.69; I2 = 0%).

An external file that holds a picture, illustration, etc. Object name is pone.0071387.g004.jpg

Forest plot for the subgroup analysis according to the PD definition.

When the results were analyzed according to PD severity, no significant risk was observed for mild PD or for severe PD (Figure 5). Heterogeneity was high in the subgroup with mild PD (χ2 = 11.28, P = 0.01; I2 = 73%) and moderate in the subgroup with severe PD (χ2 = 4.73, P = 0.09; I2 = 58%). The analysis of results stratified according to the security of PD diagnosis showed that a higher risk of PE was present in both subgroups (Figure 6), but it was significant only in the subgroup with insecure diagnosis (OR 2.68; 95% CI 1.64–4.37, P<0.0001). Heterogeneity was high in the subgroup with secure diagnosis (χ2 = 31.26, P<0.00001; I2 = 87%) and moderate in the subgroup with unsecure diagnosis (χ2 = 22.19, P = 0.008; I2 = 59%).

An external file that holds a picture, illustration, etc. Object name is pone.0071387.g005.jpg

Forest plot for the subgroup analysis according to the PD severity.

An external file that holds a picture, illustration, etc. Object name is pone.0071387.g006.jpg

Forest plot for the subgroup analysis according to the PD diagnosis.

Publication bias

An inspection of the funnel plot seemed to reveal an asymmetry (Figure 7). However, the trim and fill analysis did not indicate any missing studies (OR 3.49, 95% CI, 2.24– 4.75, P = 0.2; Figure 8). Egger’s regression asymmetry test indicated that the differences between the original estimate and the adjusted effect were not significant.

An external file that holds a picture, illustration, etc. Object name is pone.0071387.g007.jpg

Funnel plot for the association between PD and PE.

An external file that holds a picture, illustration, etc. Object name is pone.0071387.g008.jpg

Trim and filled funnel plot for the association between PD and PE.

Discussion

The aim of the present meta-analysis was to assess the potential association between PE and PD. The results seemed to indicate that PD is a risk factor for PE. Important differences were observed when the results were stratified into subgroups. In particular, when we analyzed the results according to the type of study design, the case-control studies, despite suffering from more confounding biases compared to cohort studies, revealed a significantly greater OR of PE for PD. The meta-analysis of cohort studies did not reveal any significant differences. However, the meta-analyses of subgroups potentially suffered from low power, due to the small number of included studies in each subgroup.

When analyzed according to the definition of PD, the meta-analysis performed with studies analyzing PD by PPD and CAL showed significant differences, whereas the meta-analysis of studies that defined PD by PPD alone or CAL alone did not. The definition of PD by PPD and CAL may be more appropriate, but only a few studies were included in the CAL and PPD subgroups (three and two studies, respectively). No differences were detected in the subgroup analysis of PD severity. Subgroup analysis for the security of PD diagnosis revealed that the insecure diagnosis subgroup, but not the secure diagnosis subgroup, reported a higher and significant risk of PE. Overall, given the small number of studies included in each subgroup and the corresponding low power to detect differences, it was difficult to assess the influence of the severity, the definition, and the security of diagnosis of PD on the association between PD and PE.

The present meta-analysis had several limitations. First, although meta-analysis is a useful tool in epidemiology, important issues related to methodology may limit its benefits. Among observational study designs, the case-control approach is not the best design. Thus, evidence from these studies is likely to be less accurate and possibly more influenced by recall bias compared to that from cohort studies. Second, we could not analyze the influence of the methodological quality on the results of the meta-analysis. Third, the funnel plot of publication bias was asymmetrical, and publication bias could not be excluded. This finding suggests that we may have missed important unpublished studies with results that are inconsistent with our findings. Nevertheless, the trim and fill analysis indicated that no other adjunctive study was missed. Egger’s regression test revealed that the differences between the original and the adjusted analyses were not significant. The filled funnel plot showed that no additional unpublished study was needed. Fourth, although all of the included studies reported an adjusted analysis for important and known confounders, important differences were noted in the definitions of PD and PE. No general consensuses have been reached in the definition and diagnosis of PD . The heterogeneity in these definitions may have influenced the results and introduced a bias into the meta-analysis. Therefore, given the methodological shortcomings, future studies are needed to confirm our results.

Conclusions

Based on the findings of the meta-analysis, PD appears to be a possible risk factor for PE. However, the included studies demonstrated important differences in the definitions and diagnoses of PD and PE, and lacked good methodological quality. Therefore, future studies are needed to confirm the results of the present meta-analysis. These studies should have high methodological quality, with adjustment for known confounding factors, and should report a clear and secure diagnosis of PD.

Supporting Information

Checklist S1

PRISMA Checklist.

(DOC)

Funding Statement

The authors have no funding or support to report.

References

1. Gibbs RS, Romero R, Hillier SL, Eschenbach DA, Sweet RL (1992) A review of premature birth and subclinical infectionAm J Obstet Gynecol 166: 1515–1528. [PubMed[]
2. Gibbs RS (2001) The relationship between infections and adverse pregnancy outcomes: an overviewAnn Periodontol 6: 153–163. [PubMed[]
3. Goldenberg RL, Culhane JF, Iams JD, Romero R (2008) Epidemiology and causes of preterm birthLancet 371: 75–84. [PMC free article] [PubMed[]
4. Sibai B, Dekker G, Kupferminc M (2005) Pre-eclampsiaLancet 365: 785–799. [PubMed[]
5. Noris M, Perico N, Remuzzi G (2005) Mechanisms of disease: pre-eclampsiaNat Clin Pract Nephrol 1: 98–114. [PubMed[]
6. Darmochwal-Kolarz D, Rolinski J, Leszczynska-Gorzelak B, Oleszczuk J (2002) The expressions of intracellular cytokines in the lymphocytes of preeclamptic patients. Am J Reprod Immunol 48: 381– 386. [PubMed]
7. Von Dadelszen P, Magee LA (2002) Could an infectious trigger explain the differential maternal response to the shared placental pathology of preeclampsia and normotensive intrauterine growth restriction? Acta Obstet Gynecol Scand 81: 642–648. [PubMed[]
8. Roberts JM, Taylor RN, Musci TJ, Rodgers GM, Hubel CA, et al. (1989) Preeclampsia: an endothelial cell disorderAm J Obstet Gynecol 161: 1200–1204. [PubMed[]
9. Abenhaim HA, Kinch RA, Morin L, Benjamin A, Usher R (2007) Effect of prepregnancy body mass index categories on obstetrical and neonatal outcomesArch Gynecol Obstet 275: 39–43. [PubMed[]
10. Gilbert WM, Young AL, Danielsen B (2007) Pregnancy outcomes in women with chronic hypertension: a population-based studyJ Reprod Med 52: 1046–1051. [PubMed[]
11. Sibai BM, Ewell M, Levine RJ, Klebanoff MA, Esterlitz J, et al. (1997) Risk factors associated with preeclampsia in healthy nulliparous women. The Calcium for Preeclampsia Prevention (CPEP) Study GroupAm J Obstet Gynecol 177: 1003–1010. [PubMed[]
12. Sun Y, Yang H, Sun WJ (2008) Risk factors for pre-eclampsia in pregnant Chinese women with abnormal glucose metabolismInt J Gynaecol Obstet 101: 74–76. [PubMed[]
13. Trogstad LIS, Eskild A, Bruu AL, Jeansson S, Jenum PA (2001) Is preeclampsia an infectious disease? Acta Obstet Gynecol Scand 80: 1036–1038. [PubMed[]
14. Boggess KA, Lieff S, Murtha AP, Moss K, Beck J, et al. (2003) Maternal periodontal disease is associated with an increased risk for preeclampsiaObstet Gynecol 101: 227–231. [PubMed[]
15. Canakci V, Canakci CF, Canakci H, Canakci E, Cicek Y, et al. (2004) Periodontal disease as a risk factor for pre-eclampsia: a case-control studyAust N Z J Obstet Gynaecol 44: 568–573. [PubMed[]
16. Oettinger-Barak O, Barak S, Ohel G, Oettinger M, Kreutzer H, et al. (2005) Severe pregnancy complication (preeclampsia) is associated with greater periodontal destructionJ Periodontol 76: 134–137. [PubMed[]
17. Contreras A, Herrera JA, Soto JE, Arce RM, Jaramillo A, et al. (2006) Periodontitis is associated with preeclampsia in pregnant womenJ Periodontol 77: 182–188. [PubMed[]
18. Cota LOM, Guimaraes AN, Costa JE, Lorentz TCM, Costa FO (2006) Association between maternal periodontitis and an increased risk of preeclampsiaJ Periodontol 77: 2063–2069. [PubMed[]
19. Kunnen A, Blaauw J, van Doormaal JJ, van Pampus MG, van der Schans CP, et al. (2007) Women with a recent history of early-onset pre-eclampsia have a worse periodontal conditionJ Clin Periodontol 34: 202–207. [PubMed[]
20. Offenbacher S, Lieff S, Boggess KA, Murtha AP, Madianos PN, et al. (2001) Maternal periodontitis and prematurity. Part I: Obstetric outcome of prematurity and growth restrictionAnn Periodontol 6: 164–174. [PubMed[]
21. Xiong X, Buekens P, Fraser WD, Beck J, Offenbacher S (2006) Periodontal disease and adverse pregnancy outcomes: a systematic reviewBJOG 113: 135–143. [PubMed[]
22. Offenbacher S, Boggess KA, Murtha AP, Jared HL, Lieff S, et al. (2006) Progressive periodontal disease and risk of very preterm deliveryObstet Gynecol 107: 29–36. [PubMed[]
23. Pihlstrom BL, Michalowicz BS, Johnson NW (2005) Periodontal diseasesLancet 366: 1809–1820. [PubMed[]
24. Lohsoonthorn V, Kungsadalpipob K, Chanchareonsook P, Limpongsanurak S, Vanichjakvong O, et al. (2009) Maternal periodontal disease and risk of preeclampsia: a case-control studyAm J Hypertens 22: 457–463. [PubMed[]
25. Politano GT, Passini R, Nomura ML, Velloso L, Morari J, et al. (2011) Correlation between periodontal disease, inflammatory alterations and pre-eclampsiaJ Periodontal Res 46: 505–511. [PubMed[]
26. Taghzouti N, Xiong X, Gornitsky M, Chandad F, Voyer R, et al. (2012) Periodontal disease is not associated with preeclampsia in Canadian pregnant womenJ Periodontol 83: 871–877. [PMC free article] [PubMed[]
27. Kunnen A, van Doormaal JJ, Abbas F, Aarnoudse JG, van Pampus MG, et al. (2010) Periodontal disease and pre-eclampsia: a systematic reviewJ Clin Periodontol 37: 1075–1087. [PubMed[]
28. Moher D, Liberati A, Tetzlaff J, Altman DG (2009) Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statementAnn Intern Med 151: 264–269. [PubMed[]
29. Landis JR, Koch GG (1977) An application of hierarchical kappa-type statistics in the assessment of majority agreement among multiple observersBiometrics 33: 363–374. [PubMed[]
30. Landis JR, Koch GG (1977) The measurement of observer agreement for categorical dataBiometrics 33: 159–174. [PubMed[]
31. Nibali L, Tatarakis N, Needleman I, Tu YK, D’Aiuto F (2013) Association between metabolic syndrome and periodontitis: a systematic review and meta-analysisJ Clin Endocrinol Metab 98: 913–920. [PubMed[]
32. Sterne JA, Egger M (2001) Funnel plots for detecting bias in meta-analysis: guidelines on choice of axisJ Clin Epidemiol 54: 1046–1055. [PubMed[]
33. Egger M, Smith GD, Schneider M, Minder C (1997) Bias in meta-analysis detected by a simple, graphical testBMJ 315: 629–634. [PMC free article] [PubMed[]
34. Duval S, Tweedie R (2000) Trim and fill: a simple funnel plot-based method of testing and adjusting for publication bias in meta-analysisBiometrics 56: 455–463. [PubMed[]
35. Swati P, Thomas B, Kushtagi P (2012) Periodontal disease and pregnancy hypertension – a clinical correlation. J Periodontol In press.
36. Boggess KA, Berggren EK, Koskenoja V, Urlaub D, Lorenz C (2012) Severe preeclampsia and maternal self-report of oral health, hygiene, and dental care. J Periodontol. In press. [PMC free article] [PubMed]
37. Cetin I, Pileri P, Villa A, Calabrese S, Ottolenghi L, et al. (2012) Pathogenic mechanisms linking periodontal diseases with adverse pregnancy outcomesReprod Sci 19: 633–641. [PubMed[]
38. Piscoya MD, Ximenes RA, Silva GM, Jamelli SR, Coutinho SB (2012) Maternal periodontitis as a risk factor for prematurityPediatr Int 54: 68–75. [PubMed[]
39. López-Jaramillo P, Gómez-Arbeláez D, López-López J (2011) Periodontal disease and hypertension: the pre-eclampsia model in Hispanic populationJ Hypertens 29: 1020–1021. [PubMed[]
40. Matevosyan NR (2011) Periodontal disease and perinatal outcomesArch Gynecol Obstet 283: 675–686. [PubMed[]
41. Vergnes JN (2008) Studies suggest an association between maternal periodontal disease and pre-eclampsiaEvid Based Dent 9: 46–47. [PubMed[]
42. Conde-Agudelo A, Villar J, Lindheimer M (2008) Maternal infection and risk of preeclampsia: systematic review and metaanalysisAm J Obstet Gynecol 198: 7–22. [PubMed[]
43. Dasanayake AP (2007) Preeclampsia is not related to clinical periodontal disease in nonsmoking and nondrinking Jordanian womenJ Evid Based Dent Pract 7: 176–177. [PubMed[]
44. Xiong X, Buekens P, Fraser WD, Beck J, Offenbacher S (2006) Periodontal disease and adverse pregnancy outcomes: a systematic reviewBJOG 113: 135–143. [PubMed[]
45. Abati S, Villa A, Cetin I, Dessole S, Lugliè PF, et al… (2012) Lack of association between maternal periodontal status and adverse pregnancy outcomes: a multicentric epidemiologic study. J Matern Fetal Neonatal Med In press. [PubMed]
46. Hirano E, Sugita N, Kikuchi A, Shimada Y, Sasahara J, et al. (2012) The association of Aggregatibacter actinomycetemcomitans with preeclampsia in a subset of Japanese pregnant womenJ Clin Periodontol 39: 229–338. [PubMed[]
47. Swati P, Thomas B, Vahab SA, Kapaettu S, Kushtagi P (2012) Simultaneous detection of periodontal pathogens in subgingival plaque and placenta of women with hypertension in pregnancyArch Gynecol Obstet 285: 613–619. [PubMed[]
48. Sayar F, Hoseini MS, Abbaspour S (2011) Effect of periodontal disease on preeclampsiaIran J Public Health 40: 122–127. [PMC free article] [PubMed[]
49. Horton AL, Boggess KA, Moss KL, Beck J, Offenbacher S (2010) Periodontal disease, oxidative stress, and risk for preeclampsiaJ Periodontol 81: 199–204. [PMC free article] [PubMed[]
50. Nabet C, Lelong N, Colombier ML, Sixou M, Musset AM, et al. (2010) Maternal periodontitis and the causes of preterm birth: the case-control Epipap studyJ Clin Periodontol 37: 37–45. [PMC free article] [PubMed[]
51. Ruma M, Boggess K, Moss K, Jared H, Murtha A, et al. (2008) Maternal periodontal disease, systemic inflammation, and risk for preeclampsiaAm J Obstet Gynecol 198: 389–395. [PubMed[]
52. Canakci V, Yildirim A, Canakci CF, Eltas A, Cicek Y, et al. (2007) Total antioxidant capacity and antioxidant enzymes in serum, saliva, and gingival crevicular fluid of preeclamptic women with and without periodontal diseaseJ Periodontol 78: 1602–1611. [PubMed[]
53. Cota LO, Guimarães AN, Costa JE, Lorentz TC, Costa FO (2006) Association between maternal periodontitis and an increased risk of preeclampsiaJ Periodontol 77: 2063–2069. [PubMed[]
54. Khader YS, Jibreal M, Al-Omiri M, Amarin Z (2006) Lack of association between periodontal parameters and preeclampsiaJ Periodontol 77: 1681–1687. [PubMed[]
55. Castaldi JL, Bertin MS, Giménez F, Lede R (2006) Periodontal disease: Is it a risk factor for premature labor, low birth weight or preeclampsia? Rev Panam Salud Publica 19: 253–258. [PubMed[]
56. Boggess KA, Beck JD, Murtha AP, Moss K, Offenbacher S (2006) Maternal periodontal disease in early pregnancy and risk for a small-for-gestational-age infantAm J Obstet Gynecol 194: 1316–1322. [PubMed[]
57. Meurman JH, Furuholm J, Kaaja R, Rintamäki H, Tikkanen U (2006) Oral health in women with pregnancy and delivery complicationsClin Oral Investig 10: 96–101. [PubMed[]
58. Contreras A, Herrera JA, Soto JE, Arce RM, Jaramillo A, et al. (2006) Periodontitis is associated with preeclampsia in pregnant womenJ Periodontol 77: 182–188. [PubMed[]
59. Oettinger-Barak O, Barak S, Ohel G, Oettinger M, Kreutzer H, et al. (2005) Severe pregnancy complication (preeclampsia) is associated with greater periodontal destructionJ Periodontol 76: 134–137. [PubMed[]
60. Chaparro A, Sanz A, Quintero A, Inostroza C, Ramirez V, et al… (2012) Increased inflammatory biomarkers in early pregnancy is associated with the development of pre-eclampsia in patients with periodontitis: a case control study. J Periodontal Res In press. [PubMed]
61. Kumar A, Basra M, Begum N, Rani V, Prasad S, et al… (2012) Association of maternal periodontal health with adverse pregnancy outcome J Obstet Gynaecol Res In press.
62. Wang Y, Sugita N, Kikuchi A, Iwanaga R, Hirano E (2012) FcγRIIB-nt645+25A/G gene polymorphism and periodontitis in Japanese women with preeclampsiaInt J Immunogenet 39: 492–500. [PubMed[]
63. Moura da Silva G, Coutinho SB, Piscoya MD, Ximenes RA, Jamelli SR (2012) Periodontitis as a risk factor for preeclampsiaJ Periodontol 2012 83: 1388–1396. [PubMed[]
64. Ha JE, Oh KJ, Yang HJ, Jun JK, Jin BH (2011) Oral health behaviors, periodontal disease, and pathogens in preeclampsia: a case-control study in KoreaJ Periodontol 82: 1685–1692. [PubMed[]
65. Shetty M, Shetty PK, Ramesh A, Thomas B, Prabhu S, et al. (2010) Periodontal disease in pregnancy is a risk factor for preeclampsiaActa Obstet Gynecol Scand 89: 718–721. [PubMed[]
66. Srinivas SK, Sammel MD, Stamilio DM, Clothier B, Jeffcoat MK (2009) Periodontal disease and adverse pregnancy outcomes: is there an association? Am J Obstet Gynecol 200: 497–505. [PubMed[]
67. Siqueira FM, Cota LO, Costa JE, Haddad JP, Lana AM, et al. (2008) Maternal periodontitis as a potential risk variable for preeclampsia: a case-control studyJ Periodontol 79: 207–215. [PubMed[]
68. Canakci V, Canakci CF, Yildirim A, Ingec M, Eltas A (2007) Periodontal disease increases the risk of severe pre-eclampsia among pregnant womenJ Clin Periodontol 34: 639–645. [PubMed[]
69. Sgolastra F, Severino M, Petrucci A, Gatto R, Monaco A (2013). Effectiveness of metronidazole as an adjunct to scaling and root planing in the treatment of chronic periodontitis: a systematic review and meta-analysis. J Periodontal Res (Ahead of print). [PubMed]
70. Sgolastra F, Petrucci A, Severino M, Graziani F, Gatto R, Monaco A (2013) Adjunctive photodynamic therapy to non-surgical treatment of chronic periodontitis: a systematic review and meta-analysisJ Clin Periodontol 40: 514–526. [PubMed[]
71. Sgolastra F, Severino M, Petrucci A, Gatto R, Monaco A (2013). Nd:YAG laser as an adjunctive treatment to nonsurgical periodontal therapy: A meta-analysis. Lasers Med Sci (Ahead of print). [PubMed]
72. Sgolastra F, Gatto R, Petrucci A, Monaco A (2012) Effectiveness of systemic amoxicillin/metronidazole as adjunctive therapy to scaling and root planing in the treatment of chronic periodontitis: a systematic review and meta-analysisJ Periodontol 83: 1257–1269. [PubMed[]
73. Sgolastra F, Petrucci A, Gatto R, Monaco A (2012) Effectiveness of systemic amoxicillin/metronidazole as an adjunctive therapy to full-mouth scaling and root planing in the treatment of aggressive periodontitis: a systematic review and meta-analysisJ Periodontol 83: 731–743. [PubMed[]
74. Sgolastra F, Petrucci A, Gatto R, Giannoni M, Monaco A (2011) Long-term efficacy of subantimicrobial-dose doxycycline as an adjunctive treatment to scaling and root planing: a systematic review and meta-analysisJ Periodontol 82: 1570–1581. [PubMed[]
Read More

Dysregulation of the Descending Pain System in Temporomandibular Disorders Revealed by Low-Frequency Sensory Transcutaneous Electrical Nerve Stimulation: A Pupillometric Study

Published online 2015 Apr 23

Annalisa Monaco, Ruggero Cattaneo, Luca Mesin, Eleonora Ortu, Mario Giannoni, and Davide Pietropaoli

Abstract

Using computerized pupillometry, our previous research established that the autonomic nervous system (ANS) is dysregulated in patients suffering from temporomandibular disorders (TMDs), suggesting a potential role for ANS dysfunction in pain modulation and the etiology of TMD. However, pain modulation hypotheses for TMD are still lacking. The periaqueductal gray (PAG) is involved in the descending modulation of defensive behavior and pain through μ, κ, and δ opioid receptors. Transcutaneous electrical nerve stimulation (TENS) has been extensively used for pain relief, as low-frequency stimulation can activate µ receptors. Our aim was to use pupillometry to evaluate the effect of low-frequency TENS stimulation of μ receptors on opioid descending pathways in TMD patients. In accordance with the Research Diagnostic Criteria for TMD, 18 females with myogenous TMD and 18 matched-controls were enrolled. All subjects underwent subsequent pupillometric evaluations under dark and light conditions before, soon after (end of stimulation) and long after (recovery period) sensorial TENS. The overall statistics derived from the darkness condition revealed no significant differences in pupil size between cases and controls; indeed, TENS stimulation significantly reduced pupil size in both groups. Controls, but not TMD patients, displayed significant differences in pupil size before compared with after TENS. Under light conditions, TMD patients presented a smaller pupil size compared with controls; the pupil size was reduced only in the controls. Pupil size differences were found before and during TENS and before and after TENS in the controls only. Pupillometry revealed that stimulating the descending opioid pathway with low-frequency sensory TENS of the fifth and seventh pairs of cranial nerves affects the peripheral target. The TMD patients exhibited a different pattern of response to TENS stimulation compared with the controls, suggesting that impaired modulation of the descending pain system may be involved in TMD.

Introduction

Recent literature has suggested that TMD patients may suffer from dysfunction in the brain network that supports sensory, pain, emotional, and cognitive processes []. Some authors have focused on the dysregulation of the autonomic nervous system (ANS) in TMD patients [], suggesting that TMD could be the clinical manifestation of multisystem dysregulation [].

Among the structures that are involved in the brain network regulating the sensory, pain, emotional, and cognitive systems, the periaqueductal gray (PAG) has a key role. It receives afferent connections from cortical areas associated with cognition and motivation related to sensory and pain perception [], and it then projects the connections to the centers controlling the peripheral afferent inputs and couples autonomic reactions in a specific manner [,].

The role of the PAG in descending pain control via endogenous opioids is one of the most studied pathways [], and several findings suggest that chronic pain is promoted by an abnormal modulation of the descending endogenous pain system [,].

Transcutaneous electric nerve stimulation (TENS) has been used for a long time to relieve pain []. Its main effect is believed to be achieved through the modulation of descending inputs from the ventral-lateral PAG to the rostroventral medial medulla (RVM) [,]. In particular, low-frequency TENS is suggested to activate μ opioid receptors [].

Bilateral low-frequency TENS of the fifth and seventh cranial nerves has been suggested to treat TMD [] on the basis of the resulting pain relief [,] monitored by surface electromyography (EMG) of masticatory muscles and kinesiography of jaw movements [,]. However, despite the clinical relevance of TENS in TMD, the above-cited studies focused on clinical pain or jaw muscle effects. Therefore, no information can be drawn regarding the hypothesis of autonomous or central dysfunction in patients with TMD.

Recent findings promote the use of pupillometry to study the effects of opioids in opioid-maintained patients and healthy subjects []. Moreover, the dysregulation of the ANS has been reported in TMD [], obstructive sleep apnea syndrome [], and other autonomous disorders [] by pupillometry. For these reasons, pupillometry can be considered a non-invasive, sensitive and cost-effective tool for investigating, via pupil constriction (miosis), the ANS and opioid-related miosis [].

According to the above-mentioned literature, it is possible that the effects of low-frequency TENS that are mediated by the supraspinal center and the endogenous opioid path can be demonstrated at the pupil level (miosis). The aim of this study was to evaluate pupillometric indexes using low-frequency TENS to test the hypothesis that the opioid descending pathway is dysregulated in patients with TMD.

Materials and Methods

Subjects

This study was conducted in accordance with the Declaration of Helsinki. The Committee on Ethics in Science of the University of L’Aquila, L’Aquila, Italy, approved the study, and written informed consent was obtained from each subject and electronically stored as suggested by our institutional guidelines.

Inclusion/Exclusion Criteria

Eighteen Caucasian patients (mean age 26.5±5.3 years) who fulfilled the following criteria were included in the study group: female gender; age less than 40 years; myogenous TMD; pain duration longer than 3 months; presence of complete permanent dentition, with the possible exception of the third molars; normal occlusion. Patients were excluded from the study if they met one or more of the following criteria: presence of systemic or metabolic diseases; eye diseases or visual defects; history of local or general trauma; neurological or psychiatric disorders; muscular diseases; cervical pain; bruxism, as diagnosed by the presence of parafunctional facets and/or anamnesis of parafunctional tooth clenching and/or grinding; pregnancy; assumed use of anti-inflammatory, analgesic, anti-depressant, opioid, or myorelaxant drugs; smoking; fixed or removable prostheses; fixed restorations that affected the occlusal surfaces; and either previous or concurrent orthodontic or orthognathic treatment.

For comparison with previous literature [,,,], the diagnosis of myofascial-type TMD was provided after clinical examination by a trained clinician according to group 1a and 1b of the Research Diagnostic Criteria for TMD (RDC/TMD) [], in a blinded manner (RC).

The control group consisted of 18 age- and gender-matched Caucasian subjects scheduled for a routine checkup at the University Clinic, without signs or symptoms of TMD, who fulfilled the inclusion and exclusion criteria (mean age 25.1±5.7 years).

Each enrolled subject underwent the experimental protocol described in Fig 1.

An external file that holds a picture, illustration, etc. Object name is pone.0122826.g001.jpg

Diagram of the Experimental Protocol.

Before beginning the experimental session, each enrolled subject was prepared with TENS electrodes and connected to a turned-off TENS device. Subsequently, they underwent three experimental phases: Pre TENS stimulation (green); During TENS stimulation (red); and Post TENS stimulation (blue). Pupillometer application and its removal occurred after the acclimation period and at the end of the experimental session, respectively (green). Based on the literature, central nervous system stimulation was obtained by sensory stimulation of cranial nerves V and VII with low-frequency TENS for 21 minutes (red) []. To avoid operator interference, the pupillometer, electrodes and TENS device were removed only at the end of the experimental session.

Pupillometry

Pupillometry was performed with an infrared pupillometer (Oculus system, Inventis srl, Padova, Italy), which was composed of two infrared CCD cameras (resolution of 720×576 pixels, 256 gray levels) mounted on a light helmet (1.5 kg), with a sampling frequency of 25 frames/s. To stabilize accommodation, the subjects were asked to focus their eyes on the light point in the pupillometer [].

The assessment of pupil size was performed under light conditions, with the eyes illuminated using a yellow-green LED with a 740-nanometer wavelength, as well as under dark conditions that were obtained using only three infrared diodes with a wavelength of 880 nanometers. Pupillometric recordings were acquired in digital form and processed using the Tarjan algorithm designed to evaluate strongly connected components [] to obtain frame-by-frame measurements of the pupil area, expressed as the number of pixels covering it. A template was positioned on the computer screen to allow for the correction of the eye position to avoid errors in the alignment of the pupils, as previously described [].

Pupillometry was performed with the subjects in a horizontal supine position on a bed. Room temperature (21°C) and relative humidity (50%) remained constant. Any external or internal noise sources were excluded.

Before the pupillometric recording sessions, patients were invited to lie on the bed for clinical examination with their eyes open for at least 10 minutes to adapt to the temperature and humidity of the room, as well to reduce their anxiety (Fig 1). Then, the pupillometer was applied and maintained until the end of the recording session.

Recording Procedure

All recording procedures are described in the protocol diagram (Fig 1). Briefly, after 2 minutes of darkness under an infrared light condition, 1 minute of recording was obtained, followed by the application of yellow-green light for 2 minutes and subsequent recording for 1 minute; this procedure was applied before, during and after sensory TENS stimulation, for a total of 6 pupillometric recordings of 60 seconds each (Fig 1). The recording sessions were performed by an expert operator (RC) in a blinded manner.

Based on previous literature [], darkness and light adaptation was minimized, due to the short duration of infrared and yellow-green stimulation, and any potential, residual adaptive effect, if present, could be observed in both groups.

Stimulation Procedure

The method for sensory TENS was described previously []. Briefly, a J5 Myomonitor TENS Unit device (Myotronics-Noromed, Inc., Tukwila, WA, USA) with disposable electrodes (Myotrode SG Electrodes, Myotronics-Noromed, Inc., Tukwila, WA, USA) was used. This low-frequency neurostimulator generates a repetitive synchronous and bilateral stimulus delivered at 1.5-s intervals, with an adjustable amplitude of approximately 0–24 mA, a duration of 500 μs, and a frequency of 0.66 Hz. The two TENS electrodes were placed bilaterally over the cutaneous projection of the notch of the fifth pair of cranial nerves, which was located between the coronoid and condylar processes and was retrieved by manual palpation of the zone anterior to the tragus; a third grounding electrode was placed in the center of the back of the neck.

Based on the literature, central nervous system stimulation was obtained by sensory stimulation of cranial nerves V and VII with low-frequency TENS for 21 minutes []. The amplitude of TENS stimulation started at 0 mA, with the stimulator turned on and the rheostat, which controls the amplitude, positioned at 0. The amplitude of stimulation was progressively increased at a rate of 0.6 mA/s until the patients reported the sensation of pricking. Particular attention was paid to avoid reaching the threshold of motor stimulation; if any movement of the investigated muscles was observed, the patient was excluded from the study. One patient with TMD and 1 control subject were excluded because the amplitude reached the motor threshold of the seventh cranial nerve, as shown by the contraction of the masseter and temporal muscles.

The same operator (RC) applied the pupillometer and delivered the TENS according to the manufacturer’s guidelines.

Statistical Analysis

Statistical analysis was performed using STATA 10 (StataCorp LP, College Station, TX, USA) on average pupil sizes, computed on 60 seconds of recordings, as previously described []. Preliminary analysis of pupil size showed, as expected, a high correlation between the left and right pupil sizes, allowing the use of the left-right mean value to simplify the statistics. The ratio between pupil size in the presence of yellow-green light and that in darkness (herein referred to as the L/D ratio) was calculated. The Shapiro-Wilk test revealed a normal distribution of data. Within-group differences in the pupil size and L/D ratio were analyzed using a paired t test, while differences in pupil size between groups were analyzed using an unpaired t test. The level of significance was set at p = 0.05 for all tests. The results are expressed as the mean and standard deviation (SD).

Results

The size of the pupil in darkness (infrared light) was significantly reduced during sensory low-frequency TENS both in the control (overall mean of 7886.51 before TENS vs. 7434.33 during TENS; p = 0.02) and TMD (7599.88 vs. 7148.94; p = 0.001) groups (Table 1). After TENS, the reduction of pupil size remained significant in the control group (7886.51 vs. 7427.11; p = 0.003) but not in the TMD group (7599.88 vs. 7490.61; p = 0.082) because of the return of pupil size to that prior to TENS.

Table 1

Comparison between the Pupil Size in the Control and TMD Groups in Infrared Light.
Infrared pupil size Infrared pupil size
Control group TMD group
Before TENS 7886.51 (1231.0) 7599.88 (1254.6)
During TENS 7434.33 (1464.0)* 7148.94 (1983.3)*
After TENS 7427.11 (1168.6)** 7490.61 (1477.6)

The data are expressed as the mean and standard deviation (in parentheses). Asterisks indicate statistically significant differences.

*paired t test within groups between the pre-TENS and TENS conditions: p = 0.02 in the control group and p = 0.001 in the TMD group.

**paired t test within groups between the pre-TENS and post-TENS conditions in the control group: p = 0.003.

The unpaired t test did not reveal any significant difference in pupil size between the control and TMD groups before TENS (7886.51 vs. 7599.88; p = 0.24), during TENS (7434.33 vs. 7184.94; p = 0.11), or after TENS (7427.11 vs. 7490.61; p = 0.47). In the TENS condition, it is possible that the higher value of data dispersion in the TMD group (SD 1983.31) was responsible for the lack of significance.

Table 2 shows the statistics of pupil size under yellow-green light. The size of the pupils in the control group was significantly reduced during TENS compared with the size prior to TENS (3934.98 vs. 3523.55; p = 0.005), and the pupil size continued to decrease post-TENS (3934.98 vs. 3294.16; p = 0.005). The TMD group did not show significant differences in pupil size between the pre-TENS and TENS conditions (2911.51 vs. 2836.38: p = 0.21) or between the pre-TENS and post-TENS conditions (2911.51 vs. 2973.83: p = 0.34). The unpaired t test between groups showed that the pupil size in the TMD group was significantly lower than that in the controls in the pre-TENS (2911.51 vs. 3934.38: p = 0.001) and TENS (2836.38 vs. 3523.55: p = 0.02) conditions, but no significant differences between groups were found in the post-TENS condition (2973.83 vs. 3294.16: p = 0.14). It is possible that, as in the post-TENS condition in darkness, the higher dispersion of data in the TMD group was responsible for the lack of significance. Note that the dispersion of data in the control group was lower than that in the TMD group, and, in the post-TENS condition, it was approximately one-third of that in the TMD group (sd: 617.32 vs. 1978.37). The absolute value of size differed by 10% (control 3294.16 vs. TMD 2973.83), suggesting more homogeneous pupil behavior among the control subjects.

Table 2

Comparison between Pupil Size in the Control and TMD Groups in the Yellow-Green Light Condition.
Yellow-Green pupil size Yellow-Green pupil size
Control group TMD group
Before TENS 3934.38 (821.35) 2911.51 (1041.97)°
TENS 3523.55 (867.38)* 2836.38 (1184.54)°°
After TENS 3294.16 (617.32)** 2973.83 (1978.37)

The data are expressed as means and standard deviations (in parentheses). Asterisks and circles show the statistically significant differences.

*paired t test within groups between the pre-TENS and TENS conditions in the control group: p = 0.003.

**paired t test within groups between the pre-TENS and post-TENS conditions in the control group: p = 0.0005.

°unpaired t test between the groups before TENS: p = 0.001.

°°unpaired t test between the groups during TENS: p = 0.02.

Results of the L/D ratio within and between groups are shown in Table 3 in the pre-TENS, TENS, and post-TENS conditions. The ratio decreased significantly in the control group pre-TENS compared with TENS (0.504 vs. 0.478: p = 0.03) and pre-TENS compared with post-TENS (0.504 vs. 0.444: p = 0.01). The TMD group did not show statistically significant differences in this ratio during the within-group comparisons.

Table 3

Comparison of the Pupil Size and Yellow-Green/Infrared Light (L/D) Ratio between the Control and TMD Groups.
L/D ratio L/D ratio
Control group TMD group
Before TENS 0.504 (0.111) 0.370 (0.091)°
TENS 0.478 (0.096)* 0.387 (0.123)°°
After TENS 0.444 (0.063)** 0.402 (0.112)

The data are expressed as means and standard deviations (in parentheses). Asterisks and circles show the statistically significant differences.

*paired t test within groups between the pre-TENS and TENS conditions in the control group: p = 0.03.

**paired t test within groups between the pre-TENS and post-TENS conditions in the control group: p = 0.01.

°unpaired t test between the groups before TENS: p = 0.0002.

°°unpaired t test between the groups during TENS: p = 0.01.

The L/D ratio was significantly higher in the control group compared with the TMD group, both pre-TENS (0.504 vs. 0.370: p = 0.0002) and during TENS (0.478 vs. 0.387: p = 0.01). Post-TENS, the L/D ratio remained higher in the control group (0.444 vs. 0.402), but the difference between the two groups was not significant (p = 0.08). Note that the ratio in the TMD group trended toward the value of the control group, with a value less than that in the TENS and pre-TENS conditions.

Discussion

The data in our study can be summarized as follows:

  1. No significant difference was found in pupil size in darkness between the control and TMD groups.

  2. Significant within-group differences in pupil size in the darkness condition before TENS compared with during TENS in both the control and TMD groups were found; specifically, the pupil size was reduced during TENS.

  3. Significant differences in pupil size in the darkness condition were found pre- compared with post-TENS in the control group but not in the TMD group. In the control group, the pupil size was reduced post-TENS, with the same reduction obtained during TENS; in the TMD group, the size of the pupil post-TENS returned close to the pre-TENS value.

  4. Significant differences in pupil size in the light condition were found between the control and TMD groups; specifically, the TMD group showed a significantly smaller pupil size.

  5. Significant within-group differences in pupil size in the light condition were found pre-TENS compared with during TENS and pre-TENS compared with post-TENS in the control group but not in the TMD group; pupil size in the light condition was reduced only in the control group.

Pupil size is a peripheral effect of a central network that controls the balance between sympathetic and parasympathetic outflow. The ambient light and the physiological and pathological causes of sympathetic excitation and/or parasympathetic inhibition evoke pupil dilation, and, vice versa, causes inducing sympathetic inhibition and/or parasympathetic excitation evoke pupil constriction. For this reason, pupil dynamics have been studied in physiological conditions that are suspected to impair autonomic balance and enhance arousal and sympathetic drive [,], enabling the assessment of the relationship between pupil dilation and noradrenergic activity [,]. Some authors have focused on the locus coeruleus-noradrenergic neuromodulatory system [,], which is considered to be one of the main centers regulating arousal, vigilance, alertness [], pain, and sensory afferents, and it is likely involved in a variety of disorders, such as chronic pain and TMD, characterized by dysfunction of the noradrenergic arousal system [].

The locus coeruleus and the noradrenergic arousal/vigilance system receive opposing regulation by corticotropin-releasing factor and opioids [,]. Opioids reduce the activity of the locus coeruleus [,] and the diameter of the pupil [,]. In contrast, a wakefulness-promoting drug, likely acts on the locus coeruleus-noradrenergic system and induces pupil dilation [,] (Fig 2).

An external file that holds a picture, illustration, etc. Object name is pone.0122826.g002.jpg

The Edinger-Westphal Nucleus (EWN) Exerts a Tonic Activity on the Pupil Sphincter.

Larson [] proposed that in anesthetized patients, opioid action can be exerted, blocking the efferents of inhibitory neurons to the EWN. Translating this hypothesis to non-anesthetized patients, it could be suggested that opioids reduce the inhibition of interneurons, increasing the effects of excitatory afferents of the EWN. The final effect was opioid miosis. In the figure, excitatory components of the model are shown in red, while inhibitory components are shown in black.

Low-frequency TENS induces a release of endorphins from the ventral-lateral PAG and the activation of the opioid descending path to the RVM. Simultaneously, PAG fibers extend to the pericoeruleus region, in which opioids are released in the dendritic system from the locus coeruleus neurons []. For this reason, it is possible that low-frequency TENS induces a reduction in the firing frequency in the locus coeruleus, simultaneously reducing the state of arousal, the perception of pain, and the diameter (size) of the pupil.

According to the above data, in our study, the pupil size decreased with low-frequency sensory TENS in darkness, both in the control and TMD groups. Five minutes after the end of stimulation, the effect was still present in the control group, but not in the TMD group.

The dilation of the pupil in darkness is mediated by the adrenergic sympathetic branch of the ANS and is supplied by a nerve originating from Budge’s cilio-spinal center that excites the dilator muscles of the pupil. The action of the dilator muscles is counterbalanced by the cholinergic parasympathetic branch, which originates from the Edinger-Westphal nucleus and inhibits the dilation. Moreover, pupil dilation obtained by sphincter inhibition can be equal to 1/3 of the maximum physiological dilation [], and during mental or physical efforts, the pupil dilation may occur via central inhibition of the parasympathetic center [].

It is possible that in the darkness condition in TMD patients, the endorphin effect due to TENS is sufficient to stimulate the parasympathetic inhibition of the dilator muscle; however, after the stimulation, if the baseline condition is characterized by impaired parasympathetic control, the stimulation provided by TENS rapidly completes its action on the pupillary muscles.

In the light condition, the parasympathetic branch responds to light that activates the sphincter muscle of the iris, causing miosis. At the same time, the sphincter receives beta-adrenergic innervation that is sufficient to reduce and/or counterbalance the contraction [,].

In our study, the subjects in the control and TMD groups responded to light as evidenced by a reduction in pupil size, demonstrating the proper activation of the central parasympathetic pathway that controls this basic function. Compared with the control group, the TMD patients showed a significantly higher reduction of pupil size, as indicated by both the absolute values and the ratio between the pupil size in light and in darkness. This finding might reflect impaired counterbalance of adrenergic inhibition or hyperexcitation of parasympathetic effectors in the TMD group. The result that TENS in the light condition reduces the size of the pupil in the control group but has no effect in the TMD group could suggest the impairment of the adrenergic counterbalance system, given that the opioid path works as an inhibitor of the parasympathetic branch of pupil control [].

Taken together, the data from the present study show that TMD patients suffer from a type of impairment of the descending pathway that controls the sensory, pain, and autonomic output. It is possible that one of the mechanisms involved in this impairment can be addressed in the opioid descending pathway, considering that TMD patients have a weaker response to low-frequency sensory TENS than controls, with respect to modulating the inhibition of physiological parasympathetic excitation. This study focused on adrenergic dysfunction in TMD and confirmed previous studies in which TMD patients showed impairment in the recruitment of adrenergic activity in a weakly stressful task of muscle activation, such as forced voluntarily clenching [] and other cognitive or emotional tasks [].

It is probable that the impairments could be due to changes in the connectivity among the structures that belong to the distributed network of pain processing, such as the PAG, which is a node between cortical descending control and midbrain/spinal effectors [,,,] (Figs (Figs22 and and33).

An external file that holds a picture, illustration, etc. Object name is pone.0122826.g003.jpg

Mechanism of Endogenous Opioid Action Induced by LF TENS in Non-Anesthetized Subjects with Pupil Miosis.

The locus coeruleus-noradrenergic system (LC) has been suggested to be a key structure in arousal, and its firing frequency tone has been correlated with wakefulness, vigilance, and sensory and pain perception. The size of the pupil correlates with the activity of LC, and light increases the frequency of firing. The action of the LC excites the excitation pathway that directly induces pupil constriction and excites inhibitory interneurons in the inhibitory pathway, exciting the inhibitor of the inhibitor; the net result is a reduction of inhibition and, indirectly, an agonist effect of the excitatory pathway of pupil constriction. Opioids act on the inhibitory pathway, and TENS has been suggested to be activated via the ventral lateral PAG and the opioid pathway. According to Valentino [] and Curtis [], opioids reduce the activation of the LC, and corticotropin-releasing hormone (CRH) increases the firing rate. It is possible that low-frequency sensory TENS inhibits LC inhibitory interneurons, increasing the tone of the pupillary constrictor of the LC. TMD patients could suffer from an impairment of the inhibitory pathway stemming from the ventral lateral PAG. Consequently, pupil constriction in response to light is not counterbalanced by inhibition (smaller pupil size under yellow-green light), and low-frequency sensory TENS is unable to further induce pupil constriction. Red, excitation pathway; black, inhibitory pathway.

Confirmation of the involvement of the central descending pain pathway in future studies would explain the observation that therapies such as occlusal splint therapy, orthodontics, and physiotherapy yield short-term pain management success in only 75–80% of cases []. However, simple instruments are needed to test the impairment of the descending pathway. Konchak et al. [] demonstrated that low-frequency TENS of the fifth and seventh pairs of cranial nerves “relaxed” only 75–80% of TMD subjects who received the stimulation. The remaining 20–25% showed signs of irritation after TENS, namely, an increase in EMG values of the jaw elevator muscles at rest and a lack of interocclusal freeway space. This result disagrees with the activation of the opioid descending system and directs attention to the activation of the sympathetic pathway, perhaps due to the stimulation of the dorsolateral PAG root.

As in a previous study [], we used low-frequency TENS of the fifth and seventh pairs of cranial nerves to stimulate the descending control system and to examine the effect on the peripheral target. Beyond the absolute values of pupil size and the L/D ratio, it is noteworthy to mention that the data from TMD subjects are characterized by a higher standard deviation, an index of the dispersion of the values, compared with the controls. This higher dispersion of TMD data was particularly seen during and after TENS, while the dispersion was reduced in the control subjects. We speculate that low-frequency sensory TENS could aid in differentiating TMD patients with a higher impairment of the descending pain modulation pathway from those with a less impaired system. Unfortunately, we did not assess the response to TENS for the prediction of therapy. Thus, we were unable to determine whether different degrees of impairment, such as a lower light-darkness ratio or greater TENS reduction of the pupil size in the light condition, could predict the therapy outcome. Another limitation of our study is that we did not assess whether TENS induced a reduction in pain perception in the examined subjects; therefore, we could not directly assess the efficacy of our stimulation protocol on experimental pain. However, our goal did not concern the symptom “pain” and the efficacy of low-frequency TENS used in dentistry for its relief, and thus we did not take into account a specific study design to address the response to experimental pain. These topics are beyond the scope of the present work and are suggested for future studies.

Compared with the previous work [], TMD patients in this study exhibited a smaller pupil size in darkness. The average patient age in the present study was 5.32 years older than that in the previous investigation. It is possible that this age discrepancy is responsible for this difference; in fact, it is known that pupil size decreases with age [,]. Moreover, it is possible that the duration of the disorder influences the degree of dysregulation, which shifts from a high sympathetic tone (greater pupil size in darkness) in the early stage of TMD to the failure of sympathetic counterbalance (lower pupil size in darkness) with the progression of the disorder. We did not assess the TMD duration in our sample; however, to assess the possible effect of this variable, the duration of the disorder must be investigated in the future through designed studies.

Conclusion

The stimulation of the descending opioid pathway with low-frequency sensory TENS of the fifth and seventh pairs of cranial nerves and the evaluation of the effect on the peripheral target with a simple pupillometric parameter (pupil size) lead to the conclusion that TMD patients suffer from an impairment in the modulation of the descending pain system. Further studies are necessary to confirm this hypothesis and to relate the impairment of the descending pain system to the therapeutic outcome.

Funding Statement

The authors received no specific funding for this work.

Data Availability

Data are available by request due to ethical restrictions. Interested researchers who meet the criteria for access to confidential data may contact Ruggero Cattaneo (ti.orebil@gurtac). A disclosure form by applicant Scientist is mandatory.

References

1. Maixner W, Fillingim R, Booker D, Sigurdsson A. Sensitivity of patients with painful temporomandibular disorders to experimentally evoked painPain. 1995;63: 341–351. [PubMed[]
2. Maixner W, Fillingim R, Sigurdsson A, Kincaid S, Silva S. Sensitivity of patients with painful temporomandibular disorders to experimentally evoked pain: evidence for altered temporal summation of painPain. 1998;76: 71–81. [PubMed[]
3. Svensson P, List T, Hector G. Analysis of stimulus-evoked pain in patients with myofascial temporomandibular pain disordersPain. 2001;92: 399–409. [PubMed[]
4. Eliav E, Teich S, Nitzan D, El Raziq DA, Nahlieli O, Tal M, et al. Facial arthralgia and myalgia: can they be differentiated by trigeminal sensory assessment? Pain. 2003;104: 481–490. [PubMed[]
5. Ayesh EE, Jensen TS, Svensson P. Somatosensory function following painful repetitive electrical stimulation of the human temporomandibular joint and skinExp Brain Res. 2007;179: 415–425. [PubMed[]
6. Schmidt JE, Carlson CR. A controlled comparison of emotional reactivity and physiological response in masticatory muscle pain patientsJ Orofac Pain. 2009;23: 230–242. [PubMed[]
7. Nebel MB, Folger S, Tommerdahl M, Hollins M, McGlone F, Essick G. Temporomandibular disorder modifies cortical response to tactile stimulationJ Pain. 2010;11: 1083–1094. 10.1016/j.jpain.2010.02.021 [PMC free article] [PubMed] [CrossRef[]
8. Weissman-Fogel I, Moayedi M, Tenenbaum HC, Goldberg MB, Freeman BV, Davis KD. Abnormal cortical activity in patients with temporomandibular disorder evoked by cognitive and emotional tasksPain. 2011;152: 384–396. 10.1016/j.pain.2010.10.046 [PubMed] [CrossRef[]
9. Korszun A, Young EA, Singer K, Carlson NE, Brown MB, Crofford L. Basal circadian cortisol secretion in women with temporomandibular disordersJ Dent Res. 2002;81: 279–283. [PubMed[]
10. Nilsson AM, Dahlström L. Perceived symptoms of psychological distress and salivary cortisol levels in young women with muscular or disk-related temporomandibular disordersActa Odontol Scand. 2010;68: 284–288. 10.3109/00016357.2010.494620 [PubMed] [CrossRef[]
11. Maixner W, Greenspan JD, Dubner R, Bair E, Mulkey F, Miller V, et al. Potential autonomic risk factors for chronic TMD: descriptive data and empirically identified domains from OPPERA case-contrl studyJ Pain. 2011: 12(11 Suppl): T75–T91. 10.1016/j.jpain.2011.09.002 [PMC free article] [PubMed] [CrossRef[]
12. Light KC, Bragdon EE, Grewen KM, Brownley KA, Girdler SS, Maixner W. Adrenergic dysregulation and pain with and without acute beta-blockade in women with fibromyalgia and temporomandibular disorderJ Pain. 2009;10: 542–552. 10.1016/j.jpain.2008.12.006 [PMC free article] [PubMed] [CrossRef[]
13. Eze-Nliam CM, Quartana PJ, Quain AM, Smith MT. Nocturnal heart rate variability is lower in temporomandibular disorder patients than in healthy, pain-free individualsJ Orofac Pain. 2011;25: 232–239. [PubMed[]
14. Chen H, Nackley A, Miller V, Diatchenko L, Maixner W. Multisystem dysregulation in painful temporomandibular disordersJ Pain. 2013;14: 983–996. 10.1016/j.jpain.2013.03.011 [PMC free article] [PubMed] [CrossRef[]
15. Lumb BM. Inescapable and escapable pain is represented in distinct hypothalamic-midbrain circuits: specific roles for Adelta- and C-nociceptorsExp Physiol. 2002;87: 281–286. [PubMed[]
16. Ossipov MH. The perception and endogenous modulation of painScientifica. 2012;2012: 561761 10.6064/2012/561761 [PMC free article] [PubMed] [CrossRef[]
17. Kobayashi S. Organization of neural systems for aversive information processing: pain, error, and punishmentFront Neurosci. 2012;6: 136 10.3389/fnins.2012.00136 [PMC free article] [PubMed] [CrossRef[]
18. Bandler R, Keay KA, Floyd N, Price J. Central circuits mediating patterned autonomic activity during active vs. passive emotional copingBrain Res Bull. 2000;53: 95–104. [PubMed[]
19. Keay KA, Clement CI, Depaulis A, Bandler R. Different representations of inescapable noxious stimuli in the periaqueductal gray and upper cervical spinal cord of freely moving ratsNeurosci Lett. 2001;313: 17–20. [PubMed[]
20. Heinricher MM, Tavares I, Leith JL, Lumb BM. Descending control of nociception: specificity, recruitment and plasticityBrain Res Rev. 2009;60: 214–225. 10.1016/j.brainresrev.2008.12.009 [PMC free article] [PubMed] [CrossRef[]
21. Bruehl S, Burns JW, Chung OY, Chont M. Pain-related effects of trait anger expression: neural substrates and the role of endogenous opioid mechanismsNeurosci Biobehav Rev. 2009;33: 475–491. 10.1016/j.neubiorev.2008.12.003 [PMC free article] [PubMed] [CrossRef[]
22. Graeff FG. New perspective on the pathophysiology of panic: merging serotonin and opioids in the periaqueductal grayBraz J Med Biol Res. 2012;45: 366–375. [PMC free article] [PubMed[]
23. Staud R. Abnormal endogenous pain modulation is a shared characteristic of many chronic pain conditionsExpert Rev Neurother. 2012;12: 577–585. 10.1586/ern.12.41 [PMC free article] [PubMed] [CrossRef[]
24. Linnman C, Moulton EA, Barmettler G, Becerra L, Borsook D. Neuroimaging of the periaqueductal gray: state of the fieldNeuroimage. 2012;60: 505–522. 10.1016/j.neuroimage.2011.11.095 [PMC free article] [PubMed] [CrossRef[]
25. DeSantana JM, Walsh DM, Vance C, Rakel BA, Sluka KA. Effectiveness of transcutaneous electrical nerve stimulation for treatment of hyperalgesia and painCurr Rheumatol Rep. 2008;10: 492–499. [PMC free article] [PubMed[]
26. Johnson M, Martinson M. Efficacy of electrical nerve stimulation for chronic musculoskeletal pain: a meta-analysis of randomized controlled trialsPain. 2007;130: 157–165. [PubMed[]
27. Sluka KA, Walsh D. Transcutaneous electrical nerve stimulation: basic science mechanisms and clinical effectivenessJ Pain. 2003. April 4: 109–121. [PubMed[]
28. Ainsworth L, Budelier K, Clinesmith M, Fiedler A, Landstrom R, Leeper BJ, et al. Transcutaneous electrical nerve stimulation (TENS) reduces chronic hyperalgesia induced by muscle inflammationPain. 2006;120: 182–187. [PubMed[]
29. DeSantana JM, Da Silva LF, De Resende MA, Sluka KA. Transcutaneous electrical nerve stimulation at both high and low frequencies activates ventrolateral periaqueductal grey to decrease mechanical hyperalgesia in arthritic ratsNeuroscience. 2009;163: 1233–1241. 10.1016/j.neuroscience.2009.06.056 [PMC free article] [PubMed] [CrossRef[]
30. Sluka KA, Deacon M, Stibal A, Strissel S, Terpstra A. Spinal blockade of opioid receptors prevents the analgesia produced by TENS in arthritic ratsJ Pharmacol Exp Ther. 1999;289: 840–846. [PubMed[]
31. Kalra A, Urban MO, Sluka KA. Blockade of opioid receptors in rostral ventral medulla prevents antihyperalgesia produced by transcutaneous electrical nerve stimulation (TENS)J Pharmacol Exp Ther. 2001;298: 257–263. [PubMed[]
32. Radhakrishnan R, King EW, Dickman JK, Herold CA, Johnston NF, Spurgin ML, et al. Spinal 5-HT(2) and 5-HT(3) receptors mediate low, but not high, frequency TENS-induced antihyperalgesia in ratsPain. 2003;105: 205–213. [PMC free article] [PubMed[]
33. Jankelson B. Electronic control of muscle contraction—a new clinical era in occlusion and prosthodonticsSci Educ Bull. 1969;2: 29–31. [PubMed[]
34. Katch EM. Application of transcutaneous electrical nerve stimulation in dentistryAnesth Prog. 1986;33: 156–160. [PMC free article] [PubMed[]
35. Cooper BC. International College of Cranio-Mandibular Orthopedics (ICCMO). Temporomandibular disorders: A position paper of the International College of Cranio-Mandibular Orthopedics (ICCMO)Cranio. 2011;29: 237–244. [PubMed[]
36. Chipaila N, Sgolastra F, Spadaro A, Pietropaoli D, Masci C, Cattaneo R, et al. The effects of ULF-TENS stimulation on gnathology: the state of the artCranio. 2014;32: 118–130. [PubMed[]
37. Rodrigues D, Siriani AO, Bérzin F. Effect of conventional TENS on pain and electromyographic activity of masticatory muscles in TMD patientsBraz Oral Res. 2004;18: 290–295. [PubMed[]
38. Cooper BC, Kleinberg I. Establishment of a temporomandibular physiological state with neuromuscular orthosis treatment affects reduction of TMD symptoms in 313 patientsCranio. 2008;26: 104–117. [PubMed[]
39. Monaco A, Sgolastra F, Ciarrocchi I, Cattaneo R. Effects of transcutaneous electrical nervous stimulation on electromyographic and kinesiographic activity of patients with temporomandibular disorders: a placebo-controlled studyJ Electromyogr Kinesiol. 2012;22: 463–468. 10.1016/j.jelekin.2011.12.008 [PubMed] [CrossRef[]
40. Kamyszek G, Ketcham R, Garcia R, Radke J. Electromyographic evidence of reduced muscle activity when ULF-TENS is applied to the Vth and VIIth cranial nervesCranio. 2001;19: 162–168. [PubMed[]
41. Murillo R, Crucilla C, Schmittner J, Hotchkiss E, Pickworth WB. Pupillometry in the detection of concomitant drug use in opioid-maintained patientsMethods Find Exp Clin Pharmacol. 2004;26: 271–275. [PubMed[]
42. Fliegert F, Kurth B, Göhler K. The effects of tramadol on static and dynamic pupillometry in healthy subjects—the relationship between pharmacodynamics, pharmacokinetics and CYP2D6 metaboliser statusEur J Clin Pharmacol. 2005;61: 257–266. [PubMed[]
43. Matouskova O, Slanar O, Chytil L, Perlik F. Pupillometry in healthy volunteers as a biomarker of tramadol efficacyJ Clin Pharm Ther. 2011;36: 513–517. 10.1111/j.1365-2710.2010.01203.x [PubMed] [CrossRef[]
44. Monaco A, Cattaneo R, Mesin L, Ciarrocchi I, Sgolastra F, Pietropaoli D. Dysregulation of the autonomous nervous system in patients with temporomandibular disorder: a pupillometric studyPLOS ONE. 2012;7: e45424 10.1371/journal.pone.0045424 [PMC free article] [PubMed] [CrossRef[]
45. Monaco A, Cattaneo R, Mesin L, Fiorucci E, Pietropaoli D. Evaluation of autonomic nervous system in sleep apnea patients using pupillometry under occlusal stress: a pilot studyCranio. 2014;32: 139–147. [PubMed[]
46. Wilhelm B, Giedke H, Lüdtke H, Bittner E, Hofmann A, Wilhelm H. Daytime variations in central nervous system activation measured by a pupillographic sleepiness testJ Sleep Res. 2001;10: 1–7. [PubMed[]
47. Wilhelm B, Wilhelm H, Widmaier D, Ludtke H, Ruhle K. Therapy control in sleep apnea patients by means of a pupillographic sleepiness testSleep. 1998;21: 266. []
48. Yamamoto K, Kobayashi F, Hori R, Arita A, Sasanabe R, Shiomi T. Association between pupillometric sleepiness measures and sleep latency derived by MSLT in clinically sleepy patientsEnviron Health Prev Med. 2013;18: 361–367. 10.1007/s12199-013-0331-0 [PMC free article] [PubMed] [CrossRef[]
49. Bär KJ, Schulz S, Koschke M, Harzendorf C, Gayde S, Berg W, et al. Correlations between the autonomic modulation of heart rate, blood pressure and the pupillary light reflex in healthy subjectsJ Neurol Sci. 2009;279: 9–13. 10.1016/j.jns.2009.01.010 [PubMed] [CrossRef[]
50. Keivanidou A, Fotiou D, Arnaoutoglou C, Arnaoutoglou M, Fotiou F, Karlovasitou A. Evaluation of autonomic imbalance in patients with heart failure: a preliminary study of pupillomotor functionCardiol J. 2010;17: 65–72. [PubMed[]
51. Jain S, Siegle GJ, Gu C, Moore CG, Ivanco LS, Jennings JR, et al. Autonomic insufficiency in pupillary and cardiovascular systems in Parkinson’s diseaseParkinsonism Relat Disord. 2011;17: 119–122. 10.1016/j.parkreldis.2010.11.005 [PMC free article] [PubMed] [CrossRef[]
52. Muppidi S, Adams-Huet B, Tajzoy E, Scribner M, Blazek P, Spaeth EB, et al. Dynamic pupillometry as an autonomic testing toolClin Auton Res. 2013;23: 297–303. 10.1007/s10286-013-0209-7 [PubMed] [CrossRef[]
53. Gutstein HB, Akil H. Opioid analgesics In: Brunton LL, Lazo JS, Parker KL, editors. Goodman and Gilman’s the pharmacological basis of therapeutics. 11th ed. New York: McGraw-Hill; 2006. pp. 547–590. []
54. Verster JC, Veldhuijzen DS, Volkerts ER. Effects of an opioid (oxycodone/paracetamol) and an NSAID (bromfenac) on driving ability, memory functioning, psychomotor performance, pupil size, and moodClin J Pain. 2006;22: 499–504. [PubMed[]
55. Larson MD. Mechanism of opioid-induced pupillary effectsClin Neurophysiol. 2008;119: 1358–1364. 10.1016/j.clinph.2008.01.106 [PubMed] [CrossRef[]
56. Ichesco E, Quintero A, Clauw DJ, Peltier S, Sundgren PM, Gerstner GE, et al. Altered functional connectivity between the insula and the cingulate cortex in patients with temporomandibular disorder: a pilot studyHeadache. 2012;52: 441–454. 10.1111/j.1526-4610.2011.01998.x [PMC free article] [PubMed] [CrossRef[]
57. Younger JW, Shen YF, Goddard G, Mackey SC. Chronic myofascial temporomandibular pain is associated with neural abnormalities in the trigeminal and limbic systemsPain. 2010;149: 222–228. 10.1016/j.pain.2010.01.006 [PMC free article] [PubMed] [CrossRef[]
58. Dworkin SF, LeResche L. Research diagnostic criteria for temporomandibular disorders: review, criteria, examinations and specifications, critiqueJ Craniomandib Disord. 1992;6: 301–355. [PubMed[]
59. Tarjan R. Depth-first search and linear graph algorithmsSIAM J Comput. 1972;1: 146–160. []
60. Monaco A, Sgolastra F, Pietropaoli D, Giannoni M, Cattaneo R. Comparison between sensory and motor transcutaneous electrical nervous stimulation on electromyographic and kinesiographic activity of patients with temporomandibular disorder: a controlled clinical trialBMC Musculoskelet Disord. 2013;14: 168 10.1186/1471-2474-14-168 [PMC free article] [PubMed] [CrossRef[]
61. Hayashi N, Someya N, Fukuba Y. Effect of intensity of dynamic exercise on pupil diameter in humansJ Physiol Anthropol. 2010;29: 119–122. [PubMed[]
62. Ishigaki H, Miyao M, Ishihara S. Change of pupil size as a function of exerciseJ Hum Ergol. 1991;20: 61–66. [PubMed[]
63. Preuschoff K, ‘t Hart BM, Einhäuser W. Pupil dilation signals surprise: evidence for noradrenaline’s role in decision makingFront Neurosci. 2011;5: 115 10.3389/fnins.2011.00115 [PMC free article] [PubMed] [CrossRef[]
64. Gabay S, Pertzov Y, Henik A. Orienting of attention, pupil size, and the norepinephrine systemAtten Percept Psychophys. 2011;73: 123–129. 10.3758/s13414-010-0015-4 [PubMed] [CrossRef[]
65. Murphy PR, O’Connell RG, O’Sullivan M, Robertson IH, Balsters JH. Pupil diameter covaries with BOLD activity in human locus coeruleusHum Brain Mapp. 2014;35: 4140–4154. 10.1002/hbm.22466 [PMC free article] [PubMed] [CrossRef[]
66. Alnaes D, Sneve MH, Espeseth T, Endestad T, van de Pavert SHP, Laeng B. Pupil size signals mental effort deployed during multiple object tracking and predicts brain activity in the dorsal attention network and the locus coeruleus which represents one of the most important center of arousalJ. Vision. 2014;14: 1–20. [PubMed[]
67. Aston-Jones G. Brain structures and receptors involved in alertnessSleep Med. 2005;6(Suppl 1): S3–S7. [PubMed[]
68. Aston-Jones G, Cohen JD. Adaptive gain and the role of the locus coeruleus-norepinephrine system in optimal performanceJ Comp Neurol. 2005;493: 99–110. [PubMed[]
69. Stone EA, Lin Y, Sarfraz Y, Quartermain D. The role of the central noradrenergic system in behavioral inhibitionBrain Res Rev. 2011;67: 193–208. 10.1016/j.brainresrev.2011.02.002 [PMC free article] [PubMed] [CrossRef[]
70. Berridge CW, Schmeichel BE, España RA. Noradrenergic modulation of wakefulness/arousalSleep Med Rev. 2012;162: 187–197. [PMC free article] [PubMed[]
71. Berridge CW, Waterhouse BD. The locus coeruleus-noradrenergic system: modulation of behavioral state and state-dependent cognitive processesBrain Res Brain Res Rev. 2003;42: 33–84. [PubMed[]
72. Szabady E. Modulation of physiological reflexes by pain: role of the locus coeruleusFront Inegr Neurosci. 2012;6: 94. [PMC free article] [PubMed[]
73. Brightwell JJ, Taylor BK. Noradrenergic neurons in the locus coeruleus contribute to neuropathic painNeuroscience. 2009;160: 174–185. 10.1016/j.neuroscience.2009.02.023 [PMC free article] [PubMed] [CrossRef[]
74. Valentino RJ, Van Bockstaele E. Opposing regulation of the locus coeruleus by corticotropin-releasing factor and opioids. Potential for reciprocal interactions between stress and opioid sensitivityPsychopharmacology. 2001;158: 331–342. [PubMed[]
75. Curtis AL, Bello NT, Valentino RJ. Evidence for functional release of endogenous opioids in the locus ceruleus during stress terminationJ Neurosci. 2001;21: RC152 [PMC free article] [PubMed[]
76. Mazei-Robinson MS, Nestler EJ. Opiate-induced molecular and cellular plasticity of ventral tegmental area and locus coeruleus catecholamine neuronsCold Spring Harb Perspect Med. 2012;2: a012070 10.1101/cshperspect.a012070 [PMC free article] [PubMed] [CrossRef[]
77. Valentino RJ, Van Bockstaele E. Convergent regulation of locus coeruleus activity as an adaptive response to stressEur J Pharmacol. 2008;583: 194–203. 10.1016/j.ejphar.2007.11.062 [PMC free article] [PubMed] [CrossRef[]
78. Hou RH, Freeman C, Langley RW, Szabadi E, Bradshaw CM. Does modafinil activate the locus coeruleus in man? Comparison of modafinil and clonidine on arousal and autonomic functions in human volunteersPsychopharmacology. 2005;181: 537–549. [PubMed[]
79. Nikolaou A, Schiza SE, Giakoumaki SG, Roussos P, Siafakas N, Bitsios P. The 5-min pupillary alertness test is sensitive to modafinil: a placebo controlled study in patients with sleep apneaPsychopharmacology. 2008;196: 167–175. [PubMed[]
80. Ishimatsu M, Williams JT. Synchronous activity in locus coeruleus results from dendritic interactions in pericoerulear regionsJ Neurosci. 1996;16: 5196–5204. [PMC free article] [PubMed[]
81. Tachado SD, Akhtar RA, Abdel-Latif AA. Activation of beta-adrenergic receptors causes stimulation of cyclic AMP, inhibition of inositol trisphosphate, and relaxation of bovine iris sphincter smooth muscle. Biochemical and functional interactions between the cyclic AMP and calcium signalling systemsInvest Ophthalmol Vis Sci. 1989;30: 2232–2239. [PubMed[]
82. Steinhauer SR, Siegle GJ, Condray R, Pless M. Sympathetic and parasympathetic innervation of pupillary dilation during sustained processingInt J Psychophysiol. 2004;52: 77–86. [PubMed[]
83. Topalkara A, Karadas B, Toker MI, Kaya T, Durmus N, Turgut B. Relaxant effects of beta-adrenoceptor agonist formoterol and BRL 37344 on bovine iris sphincter and ciliary muscleEur J Pharmacol. 2006;548: 144–149. [PubMed[]
84. Patil PN, Weber PA. In vivo functional implications of isoproterenol-mediated relaxation of isolated human iris sphincterJ Ocul Pharmacol. 1991;7: 297–300. [PubMed[]
85. Ploner M, Lee MC, Wiech K, Bingel U, Tracey I. Flexible cerebral connectivity patterns subserve contextual modulations of painCereb Cortex. 2011;21: 719–726. 10.1093/cercor/bhq146 [PubMed] [CrossRef[]
86. Ploner M, Lee MC, Wiech K, Bingel U, Tracey I. Prestimulus functional connectivity determines pain perception in humansProc Natl Acad Sci U S A. 2010;107: 355–360. 10.1073/pnas.0906186106 [PMC free article] [PubMed] [CrossRef[]
87. Suvinen TI, Reade PC, Kemppainen P, Könönen M, Dworkin SF. Review of aetiological concepts of temporomandibular pain disorders: towards a biopsychosocial model for integration of physical disorder factors with psychological and psychosocial illness impact factorsEur J Pain. 2005;9: 613–633. [PubMed[]
88. Konchak PA, Thomas NR, Lanigan DT, Devon RM. Free way space measurement using mandibular kinesiograph and EMG before and after TENSAngle Orthod. 1988;58: 343–350. [PubMed[]

 

Read More

Graftless sinus augmentation technique with contextual placement of implants: a case report

Published online 2014 Dec 17

Nicolae ChipailaRoberta MariniGian Luca SfasciottiAlessandro CieloLaura Bonanome, and Annalisa Monaco

Abstract

Introduction

The positioning of implants in the jaw bones with contextual graftless lateral approach sinus lifting is finding an increasingly broad consensus in the literature.

Since the 1970s, various clinical research projects have been conducted on applications of biological and synthetic biomaterials in bone regenerative surgery, both in sinus lift procedures and in cystic cavity filling after cystectomy or in bone defects in regenerative periodontal surgery. Currently, we are finding that there is an increasing trend of clinicians aiming to adopt graftless techniques, with satisfactory results in terms of implant survival in the long term.

In our study, through a case report, we describe a variant of graftless sinus augmentation technique with contextual implant placement, emphasizing the role of the blood clot, combined with collagen sponges, as a natural scaffold and the osteogenic potential of the subantral membrane in guided bone regeneration, with reduced morbidity of the patient.

Case presentation

To describe the surgical technique, the clinical case of a 38-year-old Caucasian woman with a lateral posterior edentulism was selected. The rehabilitation was solved by a graftless sinus augmentation technique with a contextual implant placement.

For each implant, a resonance frequency analysis evaluation was reported as implant stability quotient values. The performance of the implant stability quotient values followed a gradual increase from time zero to the sixth month, as the clot was differentiated into osteoid tissue and then into bone tissue, due to the scaffold effect conferred by the equine collagen sponge. The stabilization phase took place between the fourth and the sixth month, according to the implant stability quotient values.

Conclusions

Our graftless sinus augmentation technique seems to be very predictable thanks to the osteoconductive principles on which it is based, and in association with the proper management of peri-implant soft tissue, so as to increase the amount of keratinized tissue, which could represent the new gold standard for this type of rehabilitation in the future.

Introduction

Maxillary sinus augmentation procedures’ background

Oral implantology in prosthetic rehabilitation is certainly one of the most important acquisitions for dentistry in the last sixty years.

Often, in the posterior region of maxilla, following the loss of teeth, there is a reduction in volume of the bone crest and this reduction is further exacerbated by increased pneumatization of the maxillary sinuses. Therefore, the presence of a residual bone crest volumetrically unsuitable for the insertion of endosseous implants has led to the definition of surgical procedures designed to restore a suitable bone volume for implant procedures.

The maxillary sinus augmentation procedure is currently considered a highly predictable and safe technique that allows the insertion of osseointegrated implants into the atrophic posterior maxilla [], where the pneumatization of Higmoro’s antrum on one side and the edentulous alveolar process resorption by another side, often compete to make implant anchorage prosthetic rehabilitation impossible [].

At the end of the 1960s, Linkow [] first referred to the possibility of introducing blade implants into the maxillary sinus, by partially lifting the Schneider membrane without tearing it.

In 1975, Tatum proposed to raise the sinus membrane by performing a modified Caldwell-Luc technique, then called the ‘inverted lateral window’, introducing, as graft, autologous bone taken from the rib [].

The first publication of this technique is due to Boyne and James [], who in 1980 reported 14 cases of autologous graft (iliac crest) with simultaneous insertion of blade implants. Branemark [] in 1984 reported 139 penetrating implants into the maxillary sinus and nasal cavity with a follow-up from two to ten years.

In 1986, Smiler and Holmes [] proposed a technique that involves the use of nonresorbable hydroxyapatite as a bone graft substitute.

Several grafts are currently used: autologous, homologous, heterologous or alloplastic. Moy [] and Smiler [] have published studies in which the different materials are compared.

In 1987, Misch [] proposed a maxillary atrophy classification that also takes into account the therapeutic solution, as well as the most recent classification proposed by Favero and Branemark [] in 1994: it takes into account the maxillary atrophy in its entirety.

There is currently no single protocol to follow when planning this type of surgical operation: some variables such as the crestal height, the separation between the walls and the sinus pneumatization, the state of the membrane or the type of residual bone in the crest, affect the surgical indication and the techniques that must be adopted.

Antroplastic techniques can be divided into: maxillary sinus augmentation procedures (or lateral access sinus lifts) and mini sinus lifts (or crestal access sinus lifts). The first are those most frequently indicated in large-volume bone regeneration, the second are indicated in lower-volume regeneration, taking advantage of the preparation of the implant site such as access to the maxillary sinus.

The lateral access technique with insertion of various types of graft has been well codified and showed a good success predictability of the grafts and implants inserted. In the last 30 years it has undergone numerous changes, aimed at reducing the overall invasive surgery, the intra- and postoperative complications, and the patient’s morbidity.

The excessive opening of the side window, the extraoral bone graft harvesting and the late implant insertion made the surgical operation invasive and expensive.

Initially, the surgical technique used for antrostomy access made use of rotary instruments and the most frequent intraoperative complication was the perforation of the sinus membrane, with percentages ranging between 20 and 30% for different authors [].

Lately, oral surgeons have begun to use a piezoelectric tool to perform the antrostomy and the percentage of perforations has significantly decreased [].

The crestal access, on the other hand, involves less surgical invasiveness and has an equally predictable success rate of endosseous implants.

The crestal approach sinus-lifting technique introduction has further increased the indications for contextual implant placement and alveolar ridge increase. In these cases, the technique requires dedicated instrumentation and remarkable operator sensitivity during the lifting of the membrane to avoid tearing. In any case, the indication to perform a crestal approach with simultaneous implant insertion appears to be limited to clinical situations with residual alveolar ridge height >5mm and where the requested vertical increase does not exceed 5mm [].

In addition to the surgical techniques, the interest of clinical research has focused particularly on the biomaterials used to perform the filling of the lifted maxillary sinus [], as the ability to generate bone in its interior was attributed mainly to the biomaterials’ intrinsic characteristics rather than the spontaneous healing capacity of the area [].

The biomaterials used nowadays can be divided into autogenous (derived from the same patient), allogenic (obtained from another human being), and xenogenic (derived from another animal species).

Autologous bone is the gold standard for its recognized inductive and conductive abilities and because of its intrinsic osteogenicity []. It, in fact, coming from the same patient, guarantees the complete absence of adverse immune response.

Mineralized or demineralized freeze-dried alloplastic bone grafts (FDBA, DFDBA), xenografts of bovine origin, sulphate and calcium phosphate, hydroxyapatite, and bioglass have been widely employed and scientifically evaluated in order to determine the formation of new bone within the maxillary sinus and to allow osseointegration of the implants.

All substitutes possess the ability to form bone even if there is a wide range of results from a histomorphometric point of view [].

Bone regeneration, in fact, follows valid principles [] independently of the type of graft used, according to which there is the possibility of new bone formation whenever it shall create a space that can be maintained, favoring growth factors input and avoiding infectious phenomena.

In this report, a graftless sinus augmentation technique with contextual implant placement is described: the use of a simple collagen sponge is able to stabilize the blood clot in the early stages of healing.

Blood clot and Schneiderian membrane osteogenic potential

The blood clot regenerative potential is currently a topic of much discussion in the literature and it is the research subject of many authors, both for its application in guided bone regeneration (GBR) and for its application in graftless sinus augmentation techniques [].

The blood clot revaluation, as the only filler in the maxillary sinus augmentation technique in an era in which the biological and synthetic biomaterials seem to have taken over, is mainly due to Lundgren, who observed, after a maxillary sinus cyst enucleation, that the cavity left by the membrane elevation after the cyst removal was filled with bone within three months without doing anything.

In this context, he decided to further investigate this phenomenon by developing a new surgical approach, which consisted of the following steps: to carve the bone window with a beveled incision and then to remove it completely, to raise the Schneiderian membrane without tearing it, to insert the implant fixtures and to relocate the bone operculum, allowing the blood clot to fill the free space between the sinusal membrane and basal bone. The histological analysis confirms the perfectly vital bone formation starting from the surface of the antral membrane, so assuming its osteogenic potential [].

Lambert et al. in 2010 [], compared, in a study on rabbits, different materials to be used as fillers in the function of subantral bone regeneration, including the blood clot, autogenous bone and bovine hydroxyapatite (BHA): all three space-fillers allowed bone formation. The authors emphasized that the blood clot is an excellent growth factor carrier, showing initially a faster and greater bone formation, but the increase in volume is significantly reduced at five weeks postoperatively, showing that the blood clot alone may not be able to provide an adequate resistance to the sinusal re-expansion.

These observations are in agreement with other authors [], who stress that the osteoinductive properties of the blood clot alone would, therefore, be limited primarily by the inability to maintain the created space.

In this context, in our report we have considered the importance of maintaining blood clot stability through the simultaneous insertion of the implant fixtures and the association of the collagen sponge, in order to facilitate the membrane repositioning maintenance in the long term.

The results, assessed clinically and radiographically, confirmed the osteoinductive effects of the blood clot, and did not find any limitations in relation to the difficulty of maintaining the created space, overcome by the strategy of our surgical technique.

Various studies have also been conducted on the Schneiderian membrane and on its osteogenic potential [].

Srouji et al. [] have demonstrated both in vitro and in vivo that the antral membrane contains osteoprogenitor cells able to proliferate and differentiate: the authors have thereby provided a biological background for the understanding of the clinical phenomenon observed in the surgical procedure.

Histologically, the Schneider membrane is composed of several layers: an epithelial lining, a richly vascularized lamina propria, and a deeper layer that covers the jawbone. This last layer is the interface with the underlying bone, and could be compared to the periosteal structure [].

In vitro, the osteoprogenitor cells in culture have been brought to secrete alkaline phosphatase, BMP-2, osteopontin, osteonectin, osteocalcin, and also to mineralize their extracellular matrix, as already demonstrated by Gruber et al. in 2004 [] with cells of porcine sinus mucosa. In vivo, the heterotopic implantation of membrane cells combined with an osteoconductive scaffold led to the formation of new trabecular bone.

In a subsequent report, Srouji et al. [] through an in vivo simulation of an animal model sinus lift, showed the osteogenic potential of Schneider’s membrane and its possible contribution to bone regeneration in sinus lift procedures.

Palma et al. [] in a study on primates, inserted Brånemark implants both with a smooth and an oxidized surface, with a contextual sinus lift with or without autogenous bone application and analyzed the results at six months by performing block sections, reporting these conclusions:

  1. the obtained bone augmentation amount was not significantly different between the group with an autogenous bone graft and the one without a graft;

  2. the use of surface-treated implants improved the bone-implant contact;

  3. new bone was evident between the Schneiderian membrane and the graftless implant sites, demonstrating therefore, the osteoinductive potential of the membrane.

These acquisitions bring further understanding to intimate biological mechanisms that are the basis of our study and confirm the trend to implement therapeutic strategies in which the body may be enabled to express the greatest potential for healing with the least possible external interference, increasing predictability and decreasing the potentially negative external variables.

Aim of the study

In this report, we present the implant-prosthetic rehabilitation of a maxillary posterior edentulism case (area 1.4 to 1.5) through a graftless sinus augmentation technique and contextual implant fixtures placement. The purpose of the study is to describe the surgical technique aimed at the edentulous site implant rehabilitation, enhancing the role of the blood clot as a biofiller between implant screw and sinus membrane, and stressing the Schneiderian membrane osteogenic potential.

Case presentation

Our patient (a Caucasian woman, aged 38), with no history of previous or current diseases, presented with a maxillary lateral edentulism in the area 1.4 to 1.5. The preliminary radiographic evaluation showed bone dimensions of about 6 to 8mm in the coronoapical direction and 4 to 6mm in the bucco-oral direction (Figure 1).

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig1_HTML.jpg

Preoperative orthopanoramic X-ray.

After the preliminary evaluation, the operation planning started. The rehabilitation of this maxillary edentulism was solved through the placement of two implant fixtures with the contextual sinus augmentation procedure (Figures 2,3,3,4,4,5,5,6,6,7,7,8,8,9,9,10,10,11,11,12,12,1313 and and14).14). The surgical operation was performed under local anesthesia, using mepivacaine (20mg/ml) with adrenaline (1:100,000), using the truncal technique at the infraorbital foramen and major palatine foramen. On the buccal side, a full-thickness paramarginal trapezial flap extended from element 1.3 to element 1.7 was performed. Subsequently, a lateral bony window with a Beaver 65 blade used as a surgical scalpel was made.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig2_HTML.jpg

Surgical procedure of sinus lift and implants placement.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig3_HTML.jpg

Surgical procedure of sinus lift and implants placement.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig4_HTML.jpg

Surgical procedure of sinus lift and implants placement.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig5_HTML.jpg

Surgical procedure of sinus lift and implants placement.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig6_HTML.jpg

Surgical procedure of sinus lift and implants placement.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig7_HTML.jpg

Surgical procedure of sinus lift and implants placement.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig8_HTML.jpg

Surgical procedure of sinus lift and implants placement.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig9_HTML.jpg

Surgical procedure of sinus lift and implants placement.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig10_HTML.jpg

Surgical procedure of sinus lift and implants placement.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig11_HTML.jpg

Surgical procedure of sinus lift and implants placement.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig12_HTML.jpg

Surgical procedure of sinus lift and implants placement.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig13_HTML.jpg

Surgical procedure of sinus lift and implants placement.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig14_HTML.jpg

Surgical procedure of sinus lift and implants placement.

The bone segment was removed by anatomical tweezers and stored in sterile physiological solution at 4°C. The sinusal membrane was detached by special retractors and raised so as to achieve a curtain effect. An equine collagen sponge was placed in the antral area between the sinus floor and the raised membrane, to protect the latter under the Prichard’s vane ending, during the surgical alveoli preparation.

The neo-alveoli for the implant fixtures placement were performed through the crestal area, with the implant kit drills.

The two implants, with a diameter of respectively 3.5mm (normal platform, NP) and 4.3mm (regular platform, RP), a length of 13mm and conical connection characteristics were placed in situ. The primary stability was measured by resonance frequency analysis (RFA).

The apical area of the fixtures was soaked by the blood clot, in association with an equine collagen sponge, in the sinus area between the sinus floor and the raised membrane. The bone segment mobilized to perform the bone window was repositioned, the surgical area was covered with a collagen sponge layer and the flap was sutured with a nonabsorbable polyfilament.

After monitoring our patient for half an hour, no hemorrhage signs or local and/or general suffering was observed, so she was discharged with a prescription for antibiotic therapy (amoxicillin + clavulanic acid cpr. 1g for oral administration every 12 h for six days), anti-inflammatory (sodium naproxen cpr. 275mg for oral administration every 12 h for three days) and oral antiseptic therapy (0.2% chlorhexidine + cetilpiridine chloride).

Recommendations for oral hygiene and feeding techniques were supplied with the aim of optimizing the postoperative course.

The suture removal on the seventh day showed a good healing of tissues, with a modest share of keratinized tissue differentiation.

The radiographic postoperative control at six months showed a conspicuous neo-bone apposition around the apical area of the implant fixture (Figure 15), in agreement with the indications of Lioubavina-Hack et al. [], according to whom the osteo-implant unit can be considered fully functional when, at X-ray examination, there is an increase in radiopacity surrounding the implant with progressive and gradual decreasing to the periphery.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig15_HTML.jpg

Radiographic postoperative control at six months.

With regard to the condition of the soft tissues, healing took place by first intention, with net retention of keratinized tissue (Figure 16).

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig16_HTML.jpg

Clinical aspect of the soft tissue at six months.

To establish the implant survival Albrektsson [] proposed the following criteria:

  1. the system immobility;

  2. the absence of peri-implant radiolucency;

  3. the absence of persistent inflammation signs or symptoms.

To clinically evaluate the implant stability and osseointegration, we adopted resonance frequency analysis (RFA), a noninvasive diagnostic technique used to determine the ability of an implant to be subjected to load.

RFA is, in fact, the only noninvasive method that can guide the selection of prosthetic times. Adequate implant stability in bone is crucial to allow, after the insertion of an implant, undisturbed healing with new bone formation.

The implant stability is divided into primary and secondary:

  • – the primary stability is a mechanical parameter that depends on the bone quantity and quality, the system geometry and the adopted technique [].

  • – the secondary stability can be considered as the stability increase as a result of the implant placement during the healing.

This increase is attributable to the bone formation and the remodeling process that occurs in the tissue-implant interface in the surrounding area.

For each implant RFA evaluation has been reported as implant stability quotient (ISQ) values. Measurements were performed by an ISQ device (Osstell ISQ, Osstell AB, Gothenburg, Sweden).

Both implants have shown sufficient primary stability associated with mean values of ISQ=54.22.

At the end of the second month, a higher stability than the threshold value of 57 ISQ was recorded in both implants, with an average value of 59.8 ISQ.

A gradual increase in ISQ values was observed until the third and fourth month, reaching an average value of 63.2 ISQ, after which the ISQ values appeared to level off in a straight line.

The gradual filling of the defect is, therefore, accompanied by an increase in the ISQ values (Table 1).

Table 1

Mean implant stability quotient values of the two fixtures from time zero to six months

Edentulous site Fixture dimension (mm) Soft tissues condition ISQ time 0 ISQ 2 months ISQ 3 months ISQ 4 months ISQ 5 months ISQ 6 months
1.4 3.5 – 13 Good 54 58 61 63 65 65
1.5 4.3 – 13 Good 56 61 62 63 64 64

ISQ, implant stability quotient.

The ISQ average values trend found in our study (Figure 17) does not seem to be very different from that described in the literature [] in reports that consider different clinical situations and graft applications, suggesting that the graft incorporation plays a marginal role on implant stability.

An external file that holds a picture, illustration, etc. Object name is 13256_2014_3036_Fig17_HTML.jpg

Performance of implant stability quotient values in the two sites from time zero to six months. The units of the vertical axis are ISQ (implant stability quotient) values. The unit of the horizontal axis is the time expressed in months (from time 0 to the 6th month).

On the basis of the recorded values, we can state that the stabilization phase took place between the fourth and sixth month.

Discussion

Nowadays, blood clot bone regeneration potential is increasingly corroborated by the literature []. The present report wants to underline how the blood clot, rich in autogenous growth factors, can act as a natural scaffold between the guide walls of the antral floor, implant screw and raised Schneiderian membrane and differentiate itself into bone-osteoid tissue, providing secondary stability, a necessary condition for osseointegration.

In our study, the Schneiderian membrane lifting created the conditions to allow the filling of the space delimited by its detachment with the stable clot (capable of turning into bone tissue). The space was procured and maintained by the implant placement while the use of a simple equine collagen sponge allowed the clot stabilization in the early stages of healing.

In relation to the results obtained in our study and the related literature, we can state that the grafting materials currently used as subantral space fillers seem to have a more mechanical function that is not purely biological, and this may explain why, in terms of volumetric stability, slow resorption grafts are more effective than autogenous bone [].

The graftless sinus augmentation technique with contextual implants placement is widely supported by the clinical experience of various authors and also by experimental studies [].

Srouji et al. in 2009 [], showed how the basal cell layer of Schneider’s membrane has a behavior associated to that of the periosteum, able to produce the osteoprogenitor cells and humoral factors necessary for bone regeneration (BMP2, osteonectin, osteocalcin and osteopontin), requiring only the presence of a stable blood clot.

Lundgren in his report [], pointed out that the bone deposition, despite the continuous bone remodeling, is the net result of the sinus mucosa elevation in the maxillary sinus augmentation without graft application, while in sites where the technique consisted of bone graft application, a situation of bone resorption predominates. At the end of this article, the author concludes that the simple elevation of the sinus membrane and the simultaneous implant placement resulted in bone formation and osseointegration of implant fixtures.

Many other authors have subsequently observed bone formation after the sinus lift without the use of bone grafts [].

In the context of guided tissue regeneration, various authors have firmly established the importance of blood clot, with its endogenous growth factors, in allowing bone tissue formation [].

The osteogenesis process always starts with the bone defect neovascularization, which is an indispensable condition of every osteogenetic event.

After the colonization of a bone defect by proliferating mesenchymal cells with osteogenic potential capacity, the bone regeneration depends on the influence of the systematically and locally produced bone, by inductor factors such as growth factors and hormones, and by the formation of an appropriate scaffold for the proliferation and differentiation of osteoprogenitor cells.

The first stage is realized during the first four to six weeks.

It is characterized by the formation of a clot and the vascular structures migration by the marrow spaces of the walls that surround the defect, in the space below the membrane, which is followed by the beginning of the osteoid tissue deposition. This tissue is also defined primary spongy tissue and is constituted by bone with interwoven fibers that, advancing, delimit and surround the newly formed vessels, and, merging between them, define the neoformed intertrabecular spaces.

The central part of the defect, not yet filled with regenerated tissue, is composed of loose connective tissue with collagen fibers without orientation, fibroblasts, macrophages, and vases.

The second stage occurs in the subsequent two to three months, during which the maturation of the cancellous bone advances and the formation of cortical bone begins.

The osteoid bone (primary cancellous bone) undergoes mineralization by osteoblast input and at its periphery a new cortical bone, consisting of parallel bundles of lamellar bone, begins the differentiaton.

The lamellar bone is deposited more slowly than the osteoid bone, which has rapidly filled the empty spaces in the first phase, and needs a stable surface on which the collagen fibrils can be deposited in parallel fibers.

The intertrabecular spaces are gradually reduced in volume up to the size of the Haversian canals and, with the neighboring concentric lamellae, they form the primary osteons.

The last phase, which is achieved after three to four months, is characterized by the cortical bone maturation and by the cortical and medullary bone remodeling, a phase that can continue even longer.

In this phase, various osteoclasts invade the remodeling area to eliminate the fibrous bone while the neo-osteoblasts deposit layers of mature lamellar bone that leads to a thinning of the connective tissue.

In the ‘ex novo’ bone tissue formation the platelets play a key role during the first phase of the healing process, when there is an initial deposit of fibrin and the formation of blood clot.

This phase is characterized by a significant activation of chemical signals mediated by cytokines and growth factors.

In fact, the posthemorrhagic clot formation process, through platelet aggregation and cell lysis, causes the release of coagulation cascade factors and growth factors, such as platelet-derived growth factor (PDGF), insulin-like growth factors (IGF 1, IGF 2) and vascular endothelial growth factor (VEGF) that are known for their activating effect on osteoblasts and osteoclasts, and transforming growth factor beta (TGF-β), which initiate the formation of bone tissue.

The osteoblastic precursors are responsible, after differentiation into osteoblasts, for the second phase of the healing process (enchondral and/or intramembranous ossification) through the synthesis of collagen and other extracellular matrix components.

A substrate or carrier suitable for the osteoinductive signal is also needed to support and guide the new bone formation. Sampath and Reddi [] in 1984 have shown that the type I cross-linked collagen is the most appropriate carrier to promote the activity of the osteoinductive signal.

The collagen is needed in the processes of tissue repair for its osteoblastic and angiogenic activity, and also for its hemostatic and debridement properties. The collagen bound to fibronectin promotes the anchoring of mesenchymal stem cells progenitors, which exerts its chemotactic action and allows differentiation into osteoblasts.

Vice versa, through the recruitment of monocytes/macrophages, both osteoblast activity and the angiogenesis process are stimulated at the healing site.

The hemostatic action is exerted as the collagen is able to activate the platelet membrane receptors, responsible for their aggregation and the lysis process. During the first week, the collagen is able to strengthen the fibrin action in the primary clot formation while in the second week it replaces the function of fibrin.

Collagen, also carrying out chemotactic monocytes/macrophages cell lines, promotes the formation of osteoclasts which, through their action in bone resorption, can attract, activate, and collaborate with osteoblasts in bone arrangement and remodeling.

The collagen sponge used in our clinical case offered, therefore, the natural substrate for proper regeneration of bone tissue, facilitating and promoting the physiological process of regeneration. Therefore the space containing the blood clot was created and maintained. This condition allowed the bone regeneration, through the formation of a healing pattern that provided for the migration of cells with an angiogenic and osteogenic potential from the medullary spaces of the adjacent bone tissue to the surgical site. The formation of an angiogenic front and the differentiation of perivascular cells into osteoblasts led to the deposition of extracellular matrix (substantially connective tissue) that was subsequently mineralized in osteoid/bone-like tissue.

The limitations of this technique are represented by clinical situations in which there is not sufficient bone volume in order to ensure the primary stability of the implant fixtures (crestal height <5 to 6mm), and in such cases, it is preferable to insert autologous grafts as fillers instead of synthetic biomaterials, as autologous bone is the gold standard, because it is rich in cytokines, growth factors, and so on, like the blood clot, with the advantage of ensuring an excellent scaffold graft in these circumstances.

On the other hand, the advantages of this technique are: a lower morbidity for the patient, because there is no involvement of a donor site, the lower cost of the procedure, because there is no bone substitutes or membrane application, and the same timing of prosthetic finalization when compared to the techniques that involve the use of bone substitutes.

Conclusions

Good patient compliance and the motivation for thorough oral hygiene at home has led to an excellent postoperative course. The clinical and radiographic follow-up at 0, 4 and 6 months and the excellent integration of the implant system in the osteo-mucosal context, allowed by proper soft tissue management, has consolidated the success of the surgical technique, inter alia with wide confirmation in the literature.

The application of growth factors in grafting biomaterials in order to improve the osteoinductive characteristics increasingly pushes clinicians and researchers to reevaluate the quality of the blood clot, pabulum rich in cytokines and bone morphogenetic proteins, autogenous and able to promote the differentiation bone, with significant biological advantages.

Our radiographic controls showed a continuous and significant peri-implant bone remodeling over time becoming more and more homogeneous.

The results obtained from our study are encouraging and comparable to those achieved with maxillary sinus elevation techniques with biomaterial application.

Our results, therefore, in agreement with those reported in the international literature, allow us to define the graftless sinus augmentation technique as a reliable and predictable surgical method.

Consent

Written informed consent was obtained from the patient for publication of this case report and any accompanying images. A copy of the written consent is available for review by the Editor-in-Chief of this journal.

Footnotes

Competing interests

The authors declare that they have no competing interests. The study was conducted without any financial assistance.

Authors’ contributions

NC performed the surgery. RM reviewed the final version of manuscript. GLS and LB participated in the study design and coordination of the team during the manuscript preparation. AC produced the clinical images and AM conceived the study and drafted the manuscript. All authors read and approved the final manuscript.

Contributor Information

Nicolae Chipaila, ti.orebil@aliapihc.ealocin.

Roberta Marini, ti.1amorinu@iniram.r.

Gian Luca Sfasciotti, ti.1amorinu@ittoicsafs.aculnaig.

Alessandro Cielo, ti.oohay@oleicordnassela.

Laura Bonanome, ti.oohay@emonanobarual.

Annalisa Monaco, ti.oohay@ocanomasilanna.

References

1. Jensen OT, Shulman LB, Block MS, Iacono VJ. Report of Sinus Consensus Conference of 1996. Int J Oral Maxillofac Implants. 1998;13:11–45. [PubMed[]
2. Stern A, Green J. Sinus lift procedures: an overview of current techniques. Dent Clin North Am. 2012;56:219–233. doi: 10.1016/j.cden.2011.09.003. [PubMed] [CrossRef[]
3. Tasoulis G, Yao SG, Fine JB. The maxillary sinus: challenges and treatments for implant placement. Compend Contin Educ Dent. 2011;32:10–14. [PubMed[]
4. He L, Chang X, Liu Y. Sinus floor elevation using osteotome technique without grafting materials: a 2-year retrospective study. Clin Oral Implants Res. 2013;Suppl A100:63–67. doi: 10.1111/j.1600-0501.2011.02376.x. [PubMed] [CrossRef[]
5. Chen TW, Chang HS, Leung KW, Lai YL, Kao SY. Implant placement immediately after the lateral approach of the trap door window procedure to create a maxillary sinus lift without bone grafting: a 2-year retrospective evaluation of 47 implants in 33 patients. J Oral Maxillofac Surg. 2007;65:2324–2328. doi: 10.1016/j.joms.2007.06.649. [PubMed] [CrossRef[]
6. Lundgren S, Andersson S, Gualini F, Sennerby L. Bone reformation with sinus membrane elevation: a new surgical technique for maxillary sinus floor augmentation. Clin Implant Dent Relat Res. 2004;6:165–173. doi: 10.1111/j.1708-8208.2004.tb00224.x. [PubMed] [CrossRef[]
7. Lungren S, Cricchio G, Palma VC, Salata LA, Sennerby L. Sinus membrane elevation and simultaneous insertion of dental implants: a new surgical technique in maxillary sinus floor augmentation. Periodontol. 2000;47:193–205. doi: 10.1111/j.1600-0757.2008.00264.x. [PubMed] [CrossRef[]
8. Linkow LI. Maxillary implants. A dynamic approach to oral implantology. North Haven, CT: Glarus; 1997. pp. 109–111. []
9. Tatum H. Maxillary and sinus implant reconstruction. Dent Clin North Am. 1986;30:207–229. [PubMed[]
10. Boyne PJ, James RA. Grafting of the maxillary sinus floor with the autogenous marrow and bone. J Oral Surg. 1980;38:613–616. [PubMed[]
11. Branemark PI, Adell R, Albrektsson T, Lekholm U, Lindstrom J, Rockler B. An experimental and clinical study of osseointegrated implants penetrating the nasal cavity and maxillary sinus. J Oral Maxillofac Surg. 1984;42:497–505. doi: 10.1016/0278-2391(84)90008-9. [PubMed] [CrossRef[]
12. Smiler DJ, Holmes RE. Sinus lift using porous hydroxyapatite: a preliminary clinical report. J Oral Implant. 1987;13:239–253. [PubMed[]
13. Moy PK, Lundgren S, Holmes RE. Maxillary sinus augmentation: histomorphometric analysis of graft materials for maxillary sinus floor augmentation. J Oral Maxillofac Surg. 1993;51:857–862. doi: 10.1016/S0278-2391(10)80103-X. [PubMed] [CrossRef[]
14. Smiler DJ, Johnson PW, Lozada JL, Misch C, Rosenlicht JR, Tatum OH, Wagner JR. Sinus lift grafts and endosseous implants. Dent Clin North Am. 1992;36:151–188. [PubMed[]
15. Misch CE. Maxillary sinus augmentation for endosteal implants: organized alternative treatment plans. Int J Oral Implant. 1987;4:49–58. [PubMed[]
16. Favero GA, Branemark PJ. Il rialzo del pavimento del seno mascellare e osseointegrazione. Luglio: Giornale di Stomatologia e Ortognatodonzia; 1994. []
17. Barone A, Santini S, Marconcini S, Giacomelli L, Gherlone E, Covani U. Osteotomy and membrane elevation during the maxillary sinus augmentation procedure. A comparative study: piezoelectric device vs. conventional rotative instrumentations. Clin Oral Implants Res. 2008;19:511–515. doi: 10.1111/j.1600-0501.2007.01498.x. [PubMed] [CrossRef[]
18. Becker ST, Terheyden H, Steinriede A, Behrens E, Springer I, Wiltfang J. Prospective observation of 41 perforations of the Schneiderian membrane during sinus floor elevation. Clin Oral Implants Res. 2008;19:1285–1289. doi: 10.1111/j.1600-0501.2008.01612.x. [PubMed] [CrossRef[]
19. Testori T, Wallace SS, Del Fabbro M, Taschieri S, Trisi P, Capelli M, Weinstein RL. Repair of large sinus membrane perforations using stabilized collagen barrier membranes: surgical techniques with histologic and radiographic evidence of success. Int J Perio Rest Dent. 2008;28:9–17. [PubMed[]
20. Pjetursson BE, Tan WC, Zwahlen M, Lang NP. A systematic review of the success of sinus floor elevation and survival of implants inserted in combination with sinus floor elevation. J Clin Periodontol. 2008;Suppl 8:216–240. doi: 10.1111/j.1600-051X.2008.01272.x. [PubMed] [CrossRef[]
21. Summers RB. The osteotomy technique: part III- less invasive methods of elevating the sinus floor. Comp Cont Ed Dent. 1994;15:698–708. [PubMed[]
22. Bruschi GB, Scipioni A, Calesini G, Bruschi E. Localized management of the sinus floor with simultaneous implant placement: a clinical report. Int J Oral Maxillofac Implants. 1998;13:219–226. [PubMed[]
23. Browaeys H, Bouvry P, De Bruyn H. A literature review on biomaterials in sinus augmentation procedures. Clin Impl Dent Rel Res. 2007;9:166–177. doi: 10.1111/j.1708-8208.2007.00050.x. [PubMed] [CrossRef[]
24. Burchardt H. The biology of bone graft repair. Clin Orthop Rel Res. 1983;174:28–40. [PubMed[]
25. Landi L, Pretel RW, Jr, Hakimi NM, Setayesh R. Maxillary sinus floor elevation using combination of DFDBA and bovine-derived porous hydroxyapatite: a preliminary histologic and histomorphometric report. Int J Perio Rest Dent. 2000;20:575–583. [PubMed[]
26. Froum SJ, Tarnow DP, Wallace SS, Rohrer MD, Cho SC. Sinus floor elevation using an organic bovine bone matrix (Osteograf/N) with and without autogenous bone: a clinical, histologic, radiographic and histomorphometric analysis. Part II of an ongoing prospective study. Int J Perio Rest Dent. 1998;18:528–543. [PubMed[]
27. Tadjoedin ES, de Lange GL, Lyaruu DM, Kuiper L, Burger EH. High concentrations of bioactive glass material (BioGran) versus autogenous bone for sinus floor elevation. Clin Oral Impl Res. 2002;13:428–436. doi: 10.1034/j.1600-0501.2002.130412.x. [PubMed] [CrossRef[]
28. Szabo G, Huys L, Coulthard P, Maiorna C, Gargiola U, Barabas J, Nemeth Z, Hrabak K, Suba Z. A prospective multicenter randomized clinical trial of autogenous bone versus beta-tricalcium phosphate graft alone for bilateral sinus elevation: histologic and histomorphometric evaluation. Int J Oral Maxillofac Implants. 2005;20:371–381. [PubMed[]
29. Dhalin C, Linde A, Gottlow J, Nyman S. Healing of bone defects by guided tissue regeneration. Plast Reconstr Surg. 1988;81:572–576. []
30. Smukler H, Barboza EP, Burliss C. A new approach to regeneration of surgically reduced alveolar ridges in dogs: a clinical and histologic study. Int J Oral Maxillofac Implants. 1995;10:537–551. [PubMed[]
31. Lundgren S, Andersson S, Sennerby L. Spontaneous bone formation in the maxillary sinus after removal of a cyst: coincidental or expected reaction? Clin Implant Dent Relat Res. 2003;5:78–81. doi: 10.1111/j.1708-8208.2003.tb00187.x. [PubMed] [CrossRef[]
32. Srouji S, Kizhner T, Ben David D, Riminucci M, Bianco P, Livne E. The Schneiderian membrane contains osteoprogenitor cells: in vivo and in vitro study. Calcif Tissue Int. 2009;84:138–145. doi: 10.1007/s00223-008-9202-x. [PubMed] [CrossRef[]
33. Srouji S, Ben-David D, Lotan R, Riminucci M, Livne E, Bianco P. The innate osteogenic potential of the maxillary sinus (Schneiderian) membrane: an ectopic tissue transplant model simulating sinus lifting. Int J Oral Maxillofac Surg. 2010;39:793–801. doi: 10.1016/j.ijom.2010.03.009. [PubMed] [CrossRef[]
34. Palma VC, Magro-Filho O, De Oliveria JA, Lundgren S, Salata LA, Sennerby L. Bone reformation and implant integration following maxillary sinus membrane elevation: an experimental study in primates. Clin Implant Dent Relat Res. 2006;8:11–24. doi: 10.2310/j.6480.2005.00026.x. [PubMed] [CrossRef[]
35. Hatano N, Sennerby L, Lundgren S. Maxillary sinus augmentation using membrane elevation and peripheral venous blood for implant supported rehabilitation of the atrophic posterior maxilla: case series. Clin Impl Dent Rel Res. 2007;9:150–155. doi: 10.1111/j.1708-8208.2007.00043.x. [PubMed] [CrossRef[]
36. Lambert F, Léonard A, Drion P, Sourice S, Layrolle P, Rompen E. Influence of space-filling materials in subantral bone augmentation: blood clot vs. autogenous bone chips vs. bovine hydroxyapatite. Clin Oral Impl Res. 2011;22:538–545. doi: 10.1111/j.1600-0501.2010.02069.x. [PubMed] [CrossRef[]
37. Xu H, Shimizu Y, Ooya K. Histomorphometric study of the stability of new formed bone after elevation of the floor of the maxillary sinus. Br J Oral Maxillofac Surg. 2005;43:493–499. doi: 10.1016/j.bjoms.2005.02.001. [PubMed] [CrossRef[]
38. Sun XJ, Zhang ZY, Wang SY, Gittens SA, Jiang XQ, Chou LL. Maxillary sinus floor elevation using a tissue-engineered bone complex with OsteoBoneTM and bMSCs in rabbits. Clin Oral Impl Res. 2008;19:804–813. doi: 10.1111/j.1600-0501.2008.01577.x. [PubMed] [CrossRef[]
39. Gruber R, Kandler B, Fuerst G, Fischer MB, Watzek G. Porcine sinus mucosa holds cells that respond to bone morphogenetic protein (BMP)-6 and BMP-7 with increased osteogenic differentiation in vitro. Clin Oral Implants Res. 2004;15:575–580. doi: 10.1111/j.1600-0501.2004.01062.x. [PubMed] [CrossRef[]
40. Lioubavina-Hack N, Lang NP, Karring T. Significance of primary stability for osteointegration of dental implants. Clin Oral Impl Res. 2006;17:244–250. doi: 10.1111/j.1600-0501.2005.01201.x. [PubMed] [CrossRef[]
41. Albrektsson T, Donos N, Working Group 1 Implant survival and complications. The Third EAO Consensus Conference. Clin Oral Implants Res. 2012;23:63–65. doi: 10.1111/j.1600-0501.2012.02557.x. [PubMed] [CrossRef[]
42. Meredith N. Assessment of implant stability as a prognostic determinant. Int J Prosthod. 1998;11:491–501. [PubMed[]
43. Aksoy U, Eratalay K, Tözüm TF. The possible association among bone density values, resonance frequency measurements, tactile sense, and histomorphometric evaluations of dental implant osteotomy sites: a preliminary study. Implant Dent. 2009;18:316–325. doi: 10.1097/ID.0b013e31819ecc12. [PubMed] [CrossRef[]
44. Riben C, Thor A. The maxillary sinus membrane elevation procedure: augmentation of bone around dental implants without grafts-a review of a surgical technique. Int J Dent. 2012;2012:105483. doi: 10.1155/2012/105483. [PMC free article] [PubMed] [CrossRef[]
45. Dikicier S, Dikicier E, Karacayli U. J Oral Implantol. 2012. Maxillary sinus augmentation and implant placement using venous blood without graft material: a case report. [PubMed[]
46. Ellegaard B, Kølsen-Petersen J, Baelum V. Implant therapy involving maxillary sinus lift in periodontally compromised patients. Clin Oral Implants Res. 1997;8:305–315. doi: 10.1034/j.1600-0501.1997.080409.x. [PubMed] [CrossRef[]
47. Haas R, Haidvogl D, Donath K, Watzek G. Freeze-dried homogeneous and heterogeneous bone for sinus augmentation in sheep. Part I: histological findings. Clin Oral Implants Res. 2002;13:396–404. doi: 10.1034/j.1600-0501.2002.130408.x. [PubMed] [CrossRef[]
48. Winter AA, Pollack AS, Odrich RB. Sinus⁄alveolar crest tenting (SACT): a new technique for implant placement in atrophic maxillary ridges without bone grafts or membranes. Int J Periodontics Restorative Dent. 2003;23:557–565. [PubMed[]
49. Dahlin C, Sennerby L, Lekholm U, Linde A, Nyman S. Generation of new bone around titanium implants by a membrane technique: an experimental study in rabbits. Int J Oral Maxillofac Implants. 1989;4:19–25. [PubMed[]
50. Jensen OT, Greer RO, Johnson L, Kassebaum D. Vertical guided bone-graft augmentation in a new canine mandibular model. Int J Oral Maxillofac Implants. 1995;10:335–344. [PubMed[]
51. Leghissa GC, Zaffe D, Assenza B, Botticelli AR. Guided bone regeneration using titanium grids: report of 10 cases. Clin Oral Implants Res. 1999;10:62–68. doi: 10.1034/j.1600-0501.1999.100108.x. [PubMed] [CrossRef[]
52. Lynch SE, Colvin RB, Antoniades HN. Growth factors in wound healing. Single and synergistic effects on partial thickness porcine skin wounds. J Clin Invest. 1989;84:640–646. doi: 10.1172/JCI114210. [PMC free article] [PubMed] [CrossRef[]
53. Smukler H, Capri D, Landi L. Harvesting bone in the recipient sites for ridge augmentation. Int J Periodontics Restorative Dent. 2008;28(4):411–419. [PubMed[]
54. Tal H, Pitaru S, Moses O, Kozlovsky A. Collagen gel and membrane in guided tissue regeneration in periodontal fenestration defects in dogs. J Clin Periodontol. 1996;23:1–6. doi: 10.1111/j.1600-051X.1996.tb00496.x. [PubMed] [CrossRef[]
55. Sampath TK, Reddi AH. Importance of geometry of the extracellular matrix in endochondral bone differentiation. J Cell Biol. 1984;98:2192–2197. doi: 10.1083/jcb.98.6.2192. [PMC free article] [PubMed] [CrossRef[]
Read More