You are on page 1of 29

Periodontology 2000, Vol. 74, 2017, 11–39 © 2017 John Wiley & Sons A/S.

& Sons A/S. Published by John Wiley & Sons Ltd


Printed in Singapore. All rights reserved PERIODONTOLOGY 2000

Interaction between endodontics


and periodontics
ILAN ROTSTEIN

Understanding the interaction between endodontics tubules extend from the pulp to the cemento–
and periodontics is of crucial importance to the clini- dentinal junction. They run a relatively straight
cian because of the challenges frequently encoun- course and range in size from 1 to 3 lm in diameter
tered in the assessment, diagnosis, treatment and (127). The diameter of the tubules decreases with age
prognosis of combined endodontic–periodontal dis- or in response to a chronic low-grade stimuli causing
eases. Etiologic factors (such as microorganisms), as apposition of highly mineralized peritubular dentin.
well as contributing factors (such as trauma, root The number of dentin tubules varies from approxi-
resorptions, perforations, fractures and dental mal- mately 8,000 at the cemento–dentinal junction to
formations), play a role in the development and pro- 57,000 per square millimeter at the pulpal end. In the
gression of such diseases. Treatment and prognosis of cervical area of the root there are about 15,000 denti-
endodontic–periodontal diseases vary, depending on nal tubules per square millimeter (102, 127).
the etiology, pathogenesis and correct recognition of When the cementum and enamel do not meet at
each specific condition. Therefore, understanding the the cemento–enamel junction, these tubules remain
interrelationship between endodontic and periodon- exposed, thus creating pathways of communication
tal diseases will enhance the clinician’s ability to between the pulp and the periodontal ligament.
establish the correct diagnosis, assess the prognosis Patients experiencing cervical dentin hypersensitivity
of the teeth involved and select a treatment plan are an example of such a phenomenon. Fluid and irri-
based on biological and clinical evidence. tants may flow through patent dentinal tubules and,
in the absence of an intact covering of enamel or
cementum, the pulp may be considered as exposed
Anatomical relationships to the oral environment via the gingival sulcus or
periodontal pocket. Experimental studies demon-
The dental pulp and the periodontium are connected strate that application of soluble material from bacte-
via three main avenues of communication: exposed rial plaque to exposed dentin can cause pulpal
dentinal tubules; small portals of exit; and the apical inflammation, indicating that dentinal tubules may
foramen. provide ready access between the periodontium and
the pulp (18).
Scanning electron microscopy studies have demon-
Exposed dentinal tubules
strated that exposure of dentin at the cemento–
Exposed dentinal tubules in areas devoid of cemen- enamel junction occurs in about 18% of teeth in gen-
tum may serve as viable communication pathways eral and in 25% of anterior teeth in particular (133). In
between the dental pulp and the periodontal liga- addition, the same tooth may have different
ment (Fig. 1). Exposure of dentinal tubules may occur cemento–enamel junction characteristics, presenting
as a result of developmental defects, disease, or peri- dentin exposure on one surface while the other sur-
odontal or surgical procedures. Coronal dentinal faces are covered with cementum (175). This area
tubules extend from the dentino–enamel junction to (the cemento-enamel junction) becomes important
the pulp and change their orientation substantially when assessing the progression of endodontic patho-
within the first half-millimeter zone beneath the gens, as well as the effect of root scaling and planing,
dentino–enamel junction (221). Radicular dentin on cementum integrity, trauma and bleaching-

11
Rotstein

induced pathosis (50, 161, 165, 165). Exposed dentinal associated with these types of canals is relatively low.
tubules may also be found in developmental grooves, Kirkham (100), studying 1,000 human teeth with
both palatogingival and apical (183). extensive periodontal disease, found that only 2% of
such canals were associated with the involved peri-
odontal pocket.
Small portals of exit
Other canal systems in the furcation of molars may
Small portals of exit are lateral and accessory canals. also be a direct pathway of communication between
They can be present anywhere along the length of the the pulp and the periodontium (68, 115). The inci-
root (Fig. 2). Their incidence and location have been dence of accessory canals may vary from 23% to 76%
well documented in both animal and human teeth (24, 44, 65). These accessory canals contain connec-
using a variety of methods, such as digital radiogra- tive tissue and blood vessels that connect the circula-
phy, dye perfusion, injection of impression materials, tory system of the pulp with that of the
microradiography, light microscopy and scanning periodontium. However, not all of these canals
electron microscopy (24, 44, 65, 68, 100, 115, 151, extend the full length from the pulp chamber to the
166). It is estimated that 30–40% of all teeth have floor of the furcation (65).
small canal systems, the majority of which are found Seltzer et al. (177) reported that pulpal inflamma-
in the apical third of the root. It was reported that tion may cause an inflammatory reaction in the inter-
17% of teeth presented muliple canal systems in the radicular periodontal tissues. The presence of these
apical third of the root, about 9% in the middle third patent small portals of exit is a potential pathway for
and fewer than 2% in the coronal third (44). However, the spread of microorganisms and their toxic by-pro-
it seems that the incidence of periodontal disease ducts from the pulp to the periodontal ligament and

A B

Fig. 1. (A) Scanning electron micro-


graph of open dentinal tubules. (B)
Higher magnification demonstrates
the absence of an odontoblastic
processes.

A B C

Fig. 2. Nonsurgical endodontic


treatment of a maxillary central inci-
sor with a lateral radiolucency. (A)
Preoperative radiograph showing a
previously treated canal with a
mesial lateral lesion. (B) The tooth
was retreated and the root canal
filled with thermoplasticized gutta-
percha. Note the lateral canal
extending toward the lesion. (C)
One-year recall shows evidence of
active healing.

12
Interaction between endodontics and periodontics

vice versa, resulting in an inflammatory process in disease will stimulate epithelial growth, thus affecting
the involved tissues (Fig. 3). the integrity of the periradicular tissues (137, 182).
The effect of periodontal inflammation on the pulp
is controversial and conflicting studies abound (2, 3,
Apical foramen
16, 37, 64, 123, 177, 202, 218). It has been suggested
The apical foramen is the principal route of commu- that periodontal disease has no effect on the pulp, at
nication between the pulp and the periodontium. least until it involves the apex (37). On the other
Microbial and inflammatory by-products may exit hand, several studies suggest that the effect of peri-
readily through the apical foramen to cause periradic- odontal disease on the pulp is degenerative in nature
ular pathoses. The apex is also a potential portal of and includes an increase in calcification, fibrosis and
entry of inflammatory by-products from deep peri- collagen resorption, as well as a direct inflammatory
odontal pockets to the pulp. Pulp inflammation or affect (107, 119). It appears that the pulp is usually
pulp necrosis extends into the periradicular tissues not severely affected by periodontal disease until
causing a local inflammatory response often associ- recession has opened a lateral or accessory canal to
ated with bone and root resorption (177). Endodontic the oral environment. At this stage, pathogens leaking
treatment aims to eliminate the intraradicular etio- from the oral cavity through the lateral or accessory
logic factors, thereby leading to healing of the canal into the pulp may cause a chronic inflamma-
affected periradicular tissues. tory reaction followed by pulp necrosis. However, as
long as the lateral or accessory canals are protected
by sound cementum, necrosis usually does not occur.
Disease relationships Additionally, if the microvasculature of the apical
foramen remains intact, the pulp will maintain its
When the pulp becomes infected, it elicits an inflam- vitality (107). The effect of periodontal treatment on
matory response in the periodontal ligament at the the pulp is similar during scaling, curettage or peri-
apical foramen and/or adjacent to openings of the odontal surgery, if accessory canals are severed and/
small portals of exit (176). Inflammatory by-products or opened to the oral environment. In such cases,
of pulpal origin may permeate through the apex or pathogenic invasion and secondary inflammation
through smaller canals in the apical third of the root- and necrosis of the pulp can occur.
canal system and exposed dentinal tubules, and trig-
ger an inflammatory vascular response in the peri- Etiology
odontium. Among those are living pathogens, such as
Live pathogens and infectious biofilms
certain strains of bacteria, fungi and viruses (12, 13,
39, 49, 69, 86, 90, 135, 142, 186, 190, 207, 211), as well The live pathogens encountered in a diseased pulp
as nonliving pathogens (52, 135, 136, 181, 198). Many and in periradicular tissues may include bacteria,
of these are similar pathogens encountered in peri- fungi and viruses (Figs 4–6). These pathogens and
odontal inflammatory disease. In certain cases, pulpal their by-products can affect the periodontium in a

A B

Fig. 3. Micrograph of a maxillary


lateral incisor with a necrotic pulp
associated with a lateral inflamma-
tory process in the periodontal liga-
ment. (A) Main canal, accessory
canal and the resultant inflamma-
tory response in the periodontal
ligament are evident. (B) Higher
magnification of the area shows
chronic inflammation with prolifer-
ating epithelium. Masson’s trichrome
stain.

13
Rotstein

A B C D E

F G H I J

Fig. 4. Periapical actinomyces infection. This case demon- (D) Colonies of Actinomyces in the lumen of the lesion are
strates the growth of bacteria beyond the apical foramen evident. (E) Higher magnification shows a large colony of
and its invasion of apical cementum and periapical tissues. Actinomyces. (F) Foamy macrophages attacking the bacte-
(A) Radiograph of a maxillary central incisor with a necro- ria. (G) Edge of the bacterial mega-colony showing the
tic pulp showing a large periapical lesion. (B) Nonsurgical absence of inflammatory cells unable to penetrate the
endodontic therapy was performed but the symptoms per- colony. (H) Higher magnification of the bacterial colony.
sisted. (C) Apical surgery was then performed. Photomi- (I) Center of the colony devoid of the inflammatory cells.
crograph shows part of the root with the attached lesion. (J) Viable bacteria within the apical cementum.

variety of ways and need to be eliminated during inflammation occurred in the apical tissues. Korzen
root-canal treatment. et al. (105) reported similar results and suggested that
pulpal infections are usually mixed infection by nat-
Bacteria. Bacteria play a crucial role in the formation ure. Collectively, these studies provide early key evi-
and progression of both endodontic and periodontal dence regarding the role of microorganisms in pulpal
diseases (4, 12, 158, 187). The periradicular tissues and periradicular diseases.
become involved when bacteria invade the pulp, Blo€ mlof et al. (20) created defects on root surfaces
causing either partial or total necrosis. Kakehashi of intentionally extracted monkey teeth with either
et al. (92), in a classic study, demonstrated the rela- open or mature apices. The canals were either
tionship between the presence of bacteria and dis- infected or filled with calcium hydroxide and the
eases in pulp and periradicular tissues. In their study, teeth were replanted back into their sockets. After
pulps of normal rats were exposed and left open to 20 weeks, marginal epithelial downgrowth was found
the oral environment. Consequently, pulp necrosis on the denuded dentin surfaces of the infected teeth,
ensued, followed by inflammation of periradicular tis- a finding indicative of the association between
sue and lesion formation. However, when the same infected pulpal tissue and periodontal pathoses. Jans-
procedure was performed on germ-free rats, not only son et al. (87) assessed the effect of endodontic
did the pulps remain vital and relatively noninflamed, pathogens on marginal periodontal wound healing of
the exposure sites showed evidence of dentin repair. denuded dentinal surfaces surrounded by healthy
Mo € ller et al. (129) confirmed these findings in mon- periodontal ligament. Their results showed that in
keys and reported that noninfected necrotic pulp tis- infected teeth, the defects were covered by 20% more
sue did not induce periradicular lesions or epithelium, while in noninfected teeth the defects
inflammatory reactions. Nonetheless, once the pulp showed coverage by only 10% more connective tissue.
became infected, periradicular lesions and They concluded that pathogens in necrotic root

14
Interaction between endodontics and periodontics

A B C

Fig. 5. Fungi in a persistent periapi-


cal lesion. (A) Radiograph of a maxil-
D E F lary lateral incisor with necrotic pulp
and periapical radiolucency. (B)
Immediate postoperative radiograph
of nonsurgical treatment. (C) At the
3-month recall the patient is still
symptomatic and the periapical
radiolucency is larger. (D) Transmis-
sion electron micrograph shows
growing hyphae of a fungus. (E)
Higher magnification of the hyphae
showing the cell wall. (F) Spores of
reproductive fungi.

endodontic treatment failures showed an increase, of


approximately threefold, in marginal bone loss com-
pared with patients without endodontic infection.
Additionally, the effect of endodontic infection on
periodontal probing depth and the presence of furca-
tion involvement in mandibular molars were investi-
gated (85). It was found that endodontic infection in
mandibular molars was associated with more attach-
ment loss in the furcal area. It was therefore sug-
gested that endodontic infection in molars associated
with periodontal disease might enhance periodontitis
progression by spreading pathogens through acces-
sory canals and dentinal tubules (85). However, when
the endodontic infection was treated successfully, the
periodontal vector disappeared, indicating that only
one infective vector was present.
Proteolytic bacteria predominate in the root-canal
flora, which changes, over time, to a more anaerobic
Fig. 6. Transmission electron micrograph of the nucleus of microbiota (55, 172, 196). Rupf et al. (167) studied the
a macrophage in a periapical lesion, suggesting a possible
profiles of periodontal pathogens in pulpal and peri-
viral infection.
odontal diseases associated with the same tooth.
Specific PCR methods were used to detect Aggregati-
canals may stimulate epithelial downgrowth along bacter actinomycetemcomitans, Tannerella forsythia,
denuded dentin surfaces with marginal communica- Eikenella corrodens, Fusobacterium nucleatum, Por-
tion and thus augment periodontal disease. Jansson phyromonas gingivalis, Prevotella intermedia and Tre-
et al. (88), in a retrospective radiographic 3-year ponema denticola. These pathogens were found in all
study, evaluated 175 endodontically treated single- endodontic samples and also in teeth with chronic
rooted teeth of 133 patients. Patients who were more apical periodontitis and chronic adult periodontitis. It
susceptible to periodontitis and exhibited evidence of therefore appears that periodontal pathogens

15
Rotstein

accompany endodontic infections and that variant to another with numerous intermediate L-
endododontic–periodontal interrelationships are a form transitional stages. This may occur either spon-
critical pathway for both diseases. taneously or by induction in a cyclic manner. Under
Spirochetes are another class of microorganism certain conditions, depending on host resistance fac-
associated with both endodontic and periodontal tors and bacterial virulence, the L-forms revert to
diseases. Spirochetes are usually found more fre- their original pathogenic bacterial form and may then
quently in subgingival plaque than in root canals. be responsible for acute exacerbation of chronic peri-
Several studies show a large diversity of oral tre- radicular lesions (185).
ponemes in subgingival biofilms of periodontal
pockets (27, 46, 94, 152). It has been proposed that Fungi (yeasts). The presence and prevalence of fungi
the presence or absence of oral spirochetes can be associated with endodontic disease is well docu-
used to differentiate between endodontic and peri- mented (190). Fungal colonization associated with
odontal abscesses (14). Currently, however, the radicular pathosis has been demonstrated in
presence of spirochetes in the root-canal system is untreated root caries (84, 217), dentinal tubules (41,
well documented and has been demonstrated by 179), failing root-canal treatments (128, 138, 147,
different identification techniques, such as dark- 197), apices of teeth with asymptomatic apical peri-
field microscopy, electron microscopy, biochemical odontitis (114) and periradicular tissues (204). Most
identification and molecular methods (38, 39, 90, studies reported that the prevalence of fungi in cul-
91, 130, 159, 189). tured root-canal systems varies and may reach up to
The differences, reported by various authors, in 26% in untreated root canals (14, 67, 84, 97) or 33% in
the incidence of spirochetes associated with root canals treated previously (84, 128, 197, 204, 210).
endodontic disease may be attributed to the differ- A few studies, however, have demonstrated an even
ent detection methods used. It has been demon- higher incidence of fungi, of up to 55% (140, 179). The
strated that the spirochete species most frequently predominant species of fungi recovered was Candida
found in root canals are T. denticola (159, 189) albicans (210). Candida albicans has been detected in
and Treponema maltophilum (91). The main viru- 21% of infected root canals using 18S rRNA-directed
lence factor of T. denticola includes surface- species-specific primers (14) and also showed ability
expressed proteins with cytotoxic activities such as to colonize canal walls and invade dentinal tubules
the major surface protein and the chymotrypsin- (188). Other species, such as Candida glabrata, Can-
like protease complex, extracellular or membrane- dida guillermondii and Candida inconspicua (210)
associated proteolytic and hydrolytic enzymes, and and Rhodotorula mucilaginosa (49), were also
metabolites (56). This microorganism possesses an detected.
array of virulence factors associated with periodon- Factors affecting colonization of the root canal by
tal disease and may also participate in the patho- fungi are not completely understood. It appears, how-
genesis of periradicular disease (159). The ever, that among the predisposing factors of this pro-
virulence factors of T. maltophilum have not yet cess are immunocompromising diseases such as
been fully elucidated. It has been proposed that cancer (41), certain intracanal medicaments (84),
the motility of T. maltophilum, caused by the rota- local and systemic antibiotics (122, 217) and previ-
tion of its periplasmic flagella, might contribute to ously unsuccessful endodontic therapy (191, 197).
its pathogenicity (81). This microorganism was also Reduction in the numbers of specific strains of bacte-
frequently isolated from patients with rapidly pro- ria in the root canal during endodontic treatment
gressive periodontitis (132). However, the exact may allow fungal overgrowth in the remaining low-
role of this microorganism in the progression of nutrient environment (191, 197). Another possibility
endododontic–periodontal disease requires further is that fungi may gain access to the root canal from
investigation. the oral cavity as a result of poor asepsis during
L-form bacteria, also known as L-phase bacteria, endodontic treatment or post-preparation proce-
were also suggested to have a role in periradicular dures. It has been reported that approximately 20% of
disease (185). These strains of bacteria lack cell walls. adult patients with periodontitis also harbor subgin-
It has been recognized that some strains of bacteria gival fungi (40, 192) and C. albicans is the most com-
can undergo morphological transition to their L-form mon species isolated (70). In addition, it has been
after exposure to certain agents, particularly penicillin demonstrated that the presence of fungi in root
(95). The L-form and the bacterium may appear indi- canals is directly associated with their presence in sal-
vidually or together and may transform from one iva and oral tissues (49, 126). These findings further

16
Interaction between endodontics and periodontics

stress the importance of using aseptic endodontic may be reponsible for rapid breakdown of such tis-
and periodontal techniques, maintaining the integrity sue. Absence of herpesvirus infection or viral reactiva-
of dental hard tissues and covering the tooth crown tion may be why some periradicular lesions remain
as soon as practical with a well-sealed permanent clinically stable for extended periods of time (168).
restoration in order to prevent reinfection.
Infectious biofilms. The majority of bacteria in virtu-
Viruses. There is increasing evidence suggesting that ally all natural ecosystems grow in biofilms and their
viruses play an important role in both endodontic growth in affected tissues is characterized by matrix-
and periodontal diseases. In patients with periodontal enclosed communities (32, 33, 154, 211). Biofilm
disease, herpes simplex virus is frequently detected in microcolonies are composed of approximately 15%
gingival crevicular fluid and in gingival biopsies of cells (by volume) embedded in 85% matrix material
periodontal lesions (28, 30, 193). Human cytomegalo- (34). They are bisected by ramifying water channels
virus has been observed in about 65% of periodontal that carry bulk fluid into the community by convec-
pocket samples and in about 85% of gingival tissue tive flow (43). The structural composition of biofilms
samples (28). Epstein–Barr virus type I has been indicates that these communities are regulated by
observed in more than 40% of periodontal pocket signals analogous to the hormones and pheromones
samples and in about 80% of gingival tissue samples that regulate many cellular eukaryotic communities
(28). Gingival herpesviruses are associated with (34).
increased occurrence of subgingival P. gingivalis, Biofilm formation has a developmental sequence
T. forsythia, P. intermedia, Prevotella nigrescens, that results in the formation of a mature community
T. denticola and A. actinomycetemcomitans, thus sug- of tower-shaped and mushroom-shaped micro-
gesting their role in overgrowth of periodontal patho- colonies, with some variation between species. The
genic bacteria. sequence of events usually involved is microbial sur-
The presence of viruses in the dental pulp was first face attachement, cell proliferation, matrix produc-
reported in a patient with AIDS (63). HIV DNA was tion and detachment (61). Biofilm formation and
also detected in periradicular lesions (51). However, it detachment are under the control of chemical signals
has not been established if HIV can directly cause that regulate and guide the formation of slime-
pulpal disease. Herpes simplex virus was also studied enclosed microcolonies and water channels (34). It
in relation to endodontic disease. It seems, however, has been stated that microbial biofilms constitute the
that unlike its possible role in periodontal disease most ‘defensive’ life strategy that can be adopted by
(193), herpes simplex virus does not play a significant prokaryotic cells (195). In very hostile environments,
role in endodontic disease (78, 156). On the other such as extreme heat, acidity or dryness, this station-
hand, other common species of herpes viruses may ary mode of growth is inherently defensive because
be involved in pulpal and periradicular diseases. It bacterial cells are not swept into areas where they can
has been suggested that human cytomegalovirus and be killed (34).
Epstein–Barr virus play a role in the pathogenesis of Infectious biofilms are difficult to detect using
symptomatic periradicular lesions (168, 169, 200). It routine diagnostic methods and are inherently toler-
seems that active infection may result in the produc- ant to host defenses and antibiotic therapies (61).
tion of an array of cytokines and chemokines with the In addition, biofilms facilitate the spread of antibi-
potential to induce immunosuppression or tissue otic resistence by promoting horizontal gene trans-
destruction (29). Activation of inflammatory cells in fer. They are also actively adapted to environmental
periradicular tissue by herpesvirus may impair the stresses, such as alteration in nutritional quality, cell
host defense mechanisms and give rise to overgrowth density, temperature, pH and osmolarity (141). Pro-
of bacteria, as seen in periodontal lesions. Her- longed starvation induces loss of cultivability under
pesvirus-mediated immune suppression may be standard conditions, while the microorganism
detrimental in periradicular infections as a result of remains metabolically active and structurally intact
already compromised host responses in the granulo- (155). This is considered the main reason for the
matous tissue (120). Alterations between prolonged low detection rate of biofilm infections by routine
periods of herpesvirus latency interrupted by periods culture methods. To date, however, the exact role of
of activation may explain some burst-like symp- biofilms in the interrelationship between endodon-
tomatic episodes of periradicular disease. Frequent tic and periodontal diseases has not been fully
reactivation of herpesvirus in periradicular tissues elucidated.

17
Rotstein

respond to stimuli by proliferating in an attempt


Nonliving pathogens
to wall off the irritants coming through the apical
Nonliving pathogens can be either extrinsic or intrin- foramen. Epithelium can be surrounded by
sic, depending on their origin and nature. chronic inflammation. This lesion is termed
epitheliated granuloma and, if not treated, the
Extrinsic. Foreign bodies are often found to be asso- epithelium will continue to proliferate in an
ciated with the inflammatory process of the periradic- attempt to wall off the source of irritation commu-
ular tissues (Figs 7 and 8). Although endodontic and nicating from the apical foramen. The term ‘bay’
periodontal diseases are primarily associated with the cyst has been introduced for the microscopic rep-
presence of microorganisms, the presence of certain resentation of this situation (182). This is a chronic
foreign substances in situ may explain some treat- inflammatory lesion that has an epithelial lining
ment failures. Possible culprits include substances surrounding the lumen, but the lumen has a direct
such as dentin and cementum chips (59, 83, 219), communication with the root-canal system
amalgam (103, 219), root-canal filling materials (59, through the apical foramen (Fig. 9). On the other
96, 103, 219), cellulose fibers from absorbent paper hand, a ‘true’ cyst is the completion of the epithe-
points (54, 103, 104), gingival retraction cords (57), lial proliferative lesion. It is a three-dimensional,
leguminous foods (125, 149) and calculus-like depos- epithelium-lined cavity with no communication
its (71). A foreign-body response may occur to any of between the lumen and the canal system (Fig. 10).
these substances and the clinical reaction may be When periapical lesions are studied in relation to
either acute or chronic. Therefore, clinically, such the root canal, a clear distinction between these
conditions may be either symptomatic or asymp- two entities should be made (137, 182). There has
tomatic. Microscopically, these lesions demonstrate been some confusion regarding the diagnosis
the presence of multinucleated giant cells surround- when lesions are studied only on curetted biopsy
ing the foreign material in a chronic inflammatory material. As the tooth is not attached to the lesion,
infiltrate. Mechanical or surgical removal of the for- orientation to the apex is lost. Therefore, the crite-
eign bodies is usually the treatment of choice. rion used for diagnosis of a cyst is a strip of epithe-
lium that appears to be lining a cavity. It is
Intrinsic. There are a number of nonliving intrinsic therefore apparent that curetting both a ‘bay’ cyst
pathogens. and a ‘true’ cyst could lead to the same micro-
 Epithelium. One of the normal components of the scopic diagnosis. A ‘bay’ cyst could be sectioned
lateral and apical periodontal ligament is the in such a way that it could resemble or give the
epithelial rests of Malassez. The term ‘rests’ is mis- appearance of a ‘true’ cyst. This distinction
leading as it evokes a vision of discrete islands of between a ‘bay’ cyst and a ‘true’ cyst is important
epithelial cells. It has been shown that these rests from the standpoint of healing. It may be that
are actually a fishnet-like, three-dimensional, ‘true’ cysts must be surgically removed, but ‘bay’
interconnected network of epithelial cells (178). In cysts that communicate with the root canal may
many periradicular lesions, epithelium is not pre- heal with nonsurgical root-canal treatment. As
sent and therefore is presumed to have been root-canal treatment can directly affect the lumen
destroyed (178). If the rests remain, they may of the ‘bay’ cyst, the environmental change may

A B C D

Fig. 7. Foreign-body particles in a periapical lesion. (A) particles in the presence of giant cells. (C) Higher magnifi-
Radiograph of a symptomatic maxillary central incisor cation of the foreign-body particles and giant cells. (D)
with a large periapical lesion. Endodontic treatment had Part of the foreign body. When illuminated under polar-
been carried out 27 years previously. (B) Apical surgery ized light it responded as vegetable matter. The diagnosis
was performed and apical tissue was submitted for histo- was parts of a paper point past the apical foramen.
logic analysis. The photomicrograph shows foreign-body

18
Interaction between endodontics and periodontics

A B C

D E F

Fig. 8. Multiple etiologic factors beyond the apical fora- material and necrotic muscle tissue (‘dead meat granu-
men associated with failing treatment. (A) Radiograph loma’). (D) A different area of the lesion showing necrotic
showing treatment failure in a maxillary second premolar. muscle with viable bacterial colonies. (E) Necrotic muscle
The tooth was treated by intentional replantation during tissue infected with bacteria and the presence of lentil
which the apical lesion was removed. (B) Photomicrograph beans (pulse granuloma). (F) One-year follow-up radio-
of the lesion showing the presence of foreign material. (C) graph. The tooth is asymptomatic, firm and bony healing
Higher magnification shows unidentified purple foreign is evident.

true cyst occurs over time. Valderhaug (209), in a


study carried out on monkeys, showed no cyst for-
mation until at least 6 months after the canal con-
tents became necrotic. Thus, the longer a lesion
was present, the greater the probability of becom-
ing a ‘true’ cyst. However, the incidence of ‘true’
cysts is probably less than 10% (137, 182). This
may explain the relatively high success rate of
nonsurgical root-canal treatment in teeth associ-
ated with periradicular lesions.
 Cholesterol. The presence of cholesterol crystals
in apical periodontitis is a common histopatho-
logical finding (19, 23, 139, 180, 208). With time,
the cholesterol crystals are dissolved and washed
away, leaving behind spaces as clefts. The
reported incidence of cholesterol clefts in peri-
radicular disease varies from 18% to 44% (23, 180,
208). It has been suggested that the crystals could
be formed from cholesterol released by disinte-
Fig. 9. Photomicrograph showing a bay cyst associated grating erythrocytes of stagnant blood vessels
with a root canal that opens directly into the lumen of the within the periradicular lesion (23), lymphocytes,
lesion. plasma cells and macrophages which die in great
numbers and disintegrate in chronic periradicular
bring about resolution of the lesion. The ‘true’ cyst lesions (208) or by the circulating plasma lipids
is independent of the root-canal system and (180). It is possible, however, that all of these fac-
therefore conventional root-canal treatment may tors may contribute to the accumulation, concen-
not have an effect on the ‘true’ cyst. The formation tration and crystallization of cholesterol in a
of a cyst and its progression from a bay cyst to a periradicular lesion (Fig. 11). It has been

19
Rotstein

Fig. 10. Photomicrograph of a true inflammatory cyst


stained with Masson’s trichrome showing a three-dimen-
sional epithelial-lined lesion with no connection to the
root-canal system and apical foramen.

suggested that accumulation of cholesterol crys- Fig. 11. Cholesterol clefts in a periapical lesion. (A) Pho-
tomicrograph, stained with Masson’s trichrome, of a cyst
tals in inflamed periradicular tissues in some cases
with a thick fibrous wall. Embedded in the wall is a large
might cause failure of endodontic treatment (136, collection of cholesterol clefts. (B) Higher magnification
139). It seems that the macrophages and the mult- showing empty clefts where cholesterol was dissolved dur-
inucleated giant cells that congregate around ing the histologic preparation.
cholesterol crystals are not efficient enough to
destroy the crystals completely. In addition, the
accumulation of macrophages and giant cells plasma cells engaged in active synthesis of
around the cholesterol clefts in the absence of immunoglobulins. The endoplasmic reticulum
other inflammatory cells, such as neurophils, lym- becomes greatly distended, thus producing large
phocytes and plasma cells, suggests that the homogeneous eosinophilic inclusions (35). The
cholesterol crystals induce a typical foreign-body incidence of Russell bodies, their production
reaction (136). mechanism, as well as their exact role in pulpal
 Russell bodies. Russell bodies can be found in inflammation have not yet been fully elucidated.
most inflamed tissues throughout the body,  Rushton hyaline bodies. The presence of Rushton
including the periradicular tissues (Fig. 12). These hyaline bodies is a feature unique to some odon-
are small, spherical accumulations of an eosino- togenic cysts and their frequency in cysts varies
philic substance found within or near plasma cells from 2.6% to 9.5% (6). Rushton hyaline bodies
and other lymphoid cells. The presence and usually appear either within the epithelial lining
occurrence of Russell bodies in oral tissues and or the cyst lumen (Fig. 13). They have a variety of
periradicular lesions is well documented (62, 112, morphological forms, including linear (straight or
121). Studies have indicated the presence of Rus- curved), irregular, rounded and polycyclic, or they
sell bodies in about 80% of periradicular lesions. may appear granular (6, 52). The exact nature of
Large intracellular and extracellular Russell bodies Rushton hyaline bodies is not fully understood. It
were found also in inflammatory pulpal tissue of has been suggested that they are keratinous in
carious primary teeth (198). It is hypothesized that nature (180), of hematogenous origin (82), a spe-
Russell bodies are caused by synthesis of excessive cialized secretory product of odontogenic epithe-
amounts of normal secretory protein in certain lium (131) or degenerated red blood cells (35).

20
Interaction between endodontics and periodontics

A A

B B

Fig. 12. (A) Photomicrograph of a periapical lesion show-


ing the presence of Russell bodies. (B) Transmission elec-
tron micrograph demonstrates the round, amorphous
shape of these structures.

Some authors suggest that Rushton hyaline bodies


are material left behind at the time of a previous
surgical operation (124). It is not yet clear why the
Rushton hyaline bodies form mostly within the
epithelium.
 Charcot–Leyden crystals. Charcot–Leyden crystals
are naturally occurring hexagonal bipyrimidal Fig. 13. (A) Photomicrograph showing Rushton bodies in
crystals derived from the intracellular granules of the epithelial lining of a periapical cyst. (B, C) Higher-mag-
eosinophils and basophils (1, 199, 214). Their pres- nification photomicrographs demonstrating pleumor-
phism of these bodies.
ence is most often associated with increased num-
bers of peripheral blood or tissue eosinophils in
parasitic, allergic, neoplastic and inflammatory after phagocytosis, Charcot–Leyden crystal pro-
diseases (1, 109, 199). Activated macrophages are tein begins to crystallize, forming discrete particles
reported to have an important role in the forma- that increase in volume and density over time.
tion of Charcot–Leyden crystals in several disease Ultimately, these crystals can be released via
processes (25, 47, 109). Charcot–Leyden crystals phagosomal exocytosis or by piercing through the
and damaged eosinophils, along with their gran- membrane of the phagosome and macrophage
ules, have been observed within macrophages (25, cytoplasm, becoming free in the stromal tissue.
47, 109). It has been proposed that after the Recent findings support the theory that activated
degranulation of eosinophils, Charcot–Leyden macrophages have a role in the formation of Char-
crystal protein can be phagocytized into acidified cot–Leyden crystals (181). In addition, the pres-
membrane-bound lysosomes (109). At some point ence of Charcot–Leyden crystals has been

21
Rotstein

detected within a periradicular lesion that failed has been found that nonsurgical endodontic treat-
to resolve after conventional endodontic treat- ment is a predictable procedure with excellent
ment (Fig. 14). Although the biological and patho- long-term prognosis (110, 162, 170). It is imperative
logical role of Charcot–Leyden crystals in to completely clean, shape and obdurate the canal
endodontic and periodontal disease is still not system well in order to enhance successful out-
fully understood, they may be attributed to some comes. Poor endodontic treatment allows canal
cases of treatment failure. reinfection, which may often lead to treatment
failure (148).
Contributing factors Endodontic failures can be treated either by ortho-
grade or by retrograde retreatment, with good success
Poor endodontic treatment
rates reported in both (17, 171). In the last 3 decades,
Correct endodontic procedures and techniques are retreatment techniques have improved dramatically
key factors for treatment success. When assessing as a result of use of the operating microscope and
the retention rate of endodontically treated teeth, it development of new armamentarium.

A B

C D

E F

Fig. 14. Charcot–Leyden crystals in a


periapical lesion. (A) Maxillary lat-
eral incisor with necrotic pulp and a
periapical lesion. (B) Nine months
after endodontic treatment the tooth
is still symptomatic and the lesion is
larger. (C) Apical surgery was carried
out and the lesion was analyzed by
G H
microscopy. The photomicrograph
stained with hematoxylin and eosin
shows only acute and chronic
inflammatory infiltrate. (D, F, H)
May–Grunwald–Giemsa stain reveals
the presence of Charcot–Leyden
crystals. (E, G) Polarized light
demonstrates refraction of the Char-
cot–Leyden crystals.

22
Interaction between endodontics and periodontics

(vi) endodontic retreatment should be considered for


Poor restoration
teeth with a coronal seal compromised for longer
Coronal leakage is an important cause of endodontic than 3 months (77). Taking these factors into consid-
treatment failure. Root canals may become recon- eration, many endodontic and periodontal complica-
taminated with microorganisms as a result of delay in tions can, and should, be prevented. A variety of
placement of a coronal restoration and fracture of the treatment modalities can be offered to the patient
coronal restoration and/or the tooth (174). Madison depending on the case (98, 163).
& Wilcox (117) found that exposure of root canals to
Trauma
the oral environment allowed coronal leakage to
occur, and in some cases, leakage occurred along the Trauma to teeth and alveolar bone may involve the
entire length of the root canal. Ray & Trope (153) pulp and the periodontal ligament. Both tissues can
reported that defective restorations and adequate be affected either directly or indirectly. Dental inju-
root-canal fillings had a higher incidence of failures ries may take many shapes but generally can be clas-
than did teeth with inadequate root-canal fillings and sified as enamel fractures, crown fractures without
adequate restorations. Teeth in which both the root- pulp involvement, crown fractures with pulp involve-
canal fillings and restorations were adequate had an ment, crown–root fracture, root fracture, luxation and
incidence of failure of only 9%, and teeth in which avulsion (8, 9, 11, 15, 48, 134, 205, 220). Treatment of
both root-canal fillings and restorations were defec- traumatic dental injuries varies depending on the
tive had an incidence of failure of about 82% (153). type of injury and this will determine pulpal and peri-
Saunders & Saunders (173) showed that coronal leak- odontal ligament healing prognosis (11).
age was a significant clinical problem in root-filled
Resorption
molars. In an in vitro study, they found that packing
excess gutta-percha and sealer over the floor of the Root resorption is a condition associated with either a
pulp chamber, after completion of root-canal filling, physiologic or a pathologic process resulting in a loss
did not provide a better seal of the root canals. It is of dentin, cementum and/or bone (7). Despite an
therefore recommended that excess gutta-percha fill- extensive literature, this complex process presents
ing should be removed to the level of the canal ori- some confusion, mainly because of the many classifi-
fices and that the floor of the pulp chamber should cations used. The following classification is therefore
be protected with a well-sealed restorative material suggested: noninfective root resorption and infective
(173). root resorption.
Coronal restoration is the primary barrier against
coronal leakage and bacterial contamination of the Noninfective root resorption. This process occurs as a
root-canal treatment. Accordingly, lack of coronal result of a tissular response to nonmicrobial stimuli
coverage following endodontic treatment can signifi- in the affected tissues. It includes transient root
cantly compromise tooth prognosis (170). Therefore, resorption, pressure-induced root resorption, chemi-
it is essential that the root-canal system be protected cal-induced root resorption, replacement resorption
by good endodontic obturation and a well-sealed and extracanal invasive root resorption.
coronal restoration. Nevertheless, even popular per-  Transient root resorption. Transient root resorp-
manent restorative materials may not always prevent tion, or remodeling resorption, is a reparative pro-
coronal leakage (216). Cemented full crowns (66, 215) cess that occurs in response to minor trama to
as well as dentin-bonded crowns (145) show leakage. teeth functioning normally. Microscopically, small
A systemic review of the literature (143) concluded areas of cemental and dentinal resorption
that the most significant factor enhancing long-term repaired by cementum are seen. This phe-
tooth survival is placement of a coronal restoration nomenon does not present a clinical problem and
after root-canal treatment. Heling et al. (77) indicated can only be viewed microscopically.
that: (i) post space preparation and cementation  Pressure-induced root resorption. The resorption
should be performed with rubber-dam isolation; (ii) of roots of a deciduous tooth by a permanent
the post space should be prepared with a heated tooth is a typical example of this type of root
plugger; (iii) a minimum of 3 mm of root-canal filling resorption. The deciduous root is resorbed with-
should remain in the preparation; (iv) the post space out infection, and usually without inflammation.
should be irrigated and dressed as during root-canal This process is genetically controlled but pressure
treatment; (v) leakproof restorations should be placed is also a factor. If a succedaneous tooth is not pre-
as soon as possible after endodontic treatment; and sent below a deciduous tooth, resorption is often

23
Rotstein

delayed or absent altogether. Tooth impaction which the teeth are replaced with bone varies
can also create pressure on roots, causing resorp- depending mainly on the metabolic rate of the
tion. Once the source of pressure is removed, the patient. In most cases, it may take years before the
resorptive process stops. Similarly, expanding root is completely resorbed. Clinically, replace-
lesions that exert pressure (e.g. tumors or cysts) ment root resorption is diagnosed when lack of
may cause root resorption. Removal of the lesion mobility of the ankylosed teeth is determined (10).
will arrest the resorptive process. This type of The teeth will also have a specific metallic sound
resorption is usually asymptomatic unless sec- upon percussion, and after a period of time will be
ondary infection occurs. Iatrogenic pressure, such in infraocclusion. Radiographically, absence of a
as excessive orthodontic movements, can also periodontal ligament space is evident and the
result in root resorption. Depending on their nat- ingrowth of bone into the root will present a char-
ure, these forces can cause blunting and areas of acteristic ‘moth-eaten’ appearance (203).
resorption along the root surfaces. The resorption  Extracanal invasive root resorption. Extracanal
will stop once the stimulus is removed. invasive root resorption is a relatively uncommon
 Chemical-induced root resorption. Certain chemi- form of root resorption (73, 193, 194). It is charac-
cals used in dentistry have the potential to cause terized by its cervical location and invasive nature.
root resorption. Clinical reports (58, 72, 76, 80, Invasion of the cervical region of the root is pre-
116, 161) have shown that intracoronal bleaching dominated by fibrovascular tissue derived from
with highly concentrated oxiding agents, such as the periodontal ligament. The process progres-
30–35% hydrogen peroxide, can induce root sively resorbs cementum, enamel and dentin and
resorption. The irritating chemical may diffuse later may involve the pulp space. There may be no
through the dentinal tubules, and, when com- signs or symptoms unless the resorption is associ-
bined with heat, is likely to cause necrosis of the ated with pulpal or periodontal infection. Second-
cementum, inflammation of the periodontal liga- ary bacterial invasion into the pulp or periodontal
ment and subsequently root resorption (116, 161, ligament space will cause inflammation of the tis-
164). The process is liable to be enhanced in the sues accompanied with pain. Frequently, how-
presence of bacteria (79). Previous traumatic ever, the resorptive defect is only detected by
injury and young age may act as predisposing fac- routine radiographic examination. Where the
tors (72). lesion is visible, the clinical features vary from a
 Replacement root resorption. Replacement root small defect at the gingival margin to a pink coro-
resorption, or ankylosis, occurs following exten- nal discoloration of the tooth crown (73). Radio-
sive necrosis of the periodontal ligament with for- graphically, the lesion varies from well-delineated
mation of bone onto a denuded area of the root to irregularly boarded radiolucencies. A character-
surface (203). This condition is most often seen as istic radiopaque line generally separates the image
a complication of luxation injuries, especially in of the lesion from that of the root canal because
avulsed teeth that have been out of their sockets the pulp remains protected by a thin layer of pre-
in dry conditions for several hours. Certain peri- dentin until late in the process (73). The etiology
odontal procedures were reported to induce of invasive cervical resorption is not fully under-
replacement root resorption (118). Potential for stood. It seems, however, that potential predispos-
replacement resorption was also associated with ing factors are traumatic injuries, orthodontic
periodontal wound healing (93). Granulation tis- treatment and intracoronal bleaching with highly
sue derived from bone or gingival connective tis- concentrated oxidizing agents (74, 161). Treat-
sue may induce root resorption and ankylosis. It ment of the condition presents clinical problems
seems that the inability to form connective tissue because the resorptive tissue is highly vascular
attachment on a denuded root surface is the cul- and the resulting hemorrhage may impede visual-
prit. The only cells within the periodontium that ization and compromise placement of a restora-
appear to have the capacity for doing so are the tion (75). Successful treatment relies upon the
periodontal ligament cells (22). In general, if less complete removal, or inactivation, of the resorp-
than 20% of the root surface is involved, reversal tive tissue. This is difficult to obtain in more
of the ankylosis can occur (10). If not, ankylosed advanced lesions characterized by a series of small
teeth are incorporated in the alveolar bone and channels often interconnecting with the periodon-
become part of the normal remodeling process of tal ligament apical to the main lesion. In most
bone. This is a gradual process, and the speed by cases, surgery is necessary to gain access to the

24
Interaction between endodontics and periodontics

resorptive defect and often may cause loss of bone The etiology of this type of root resorption is usu-
and periodontal attachement. Topical application ally trauma (212). Extreme heat was suggested as a
of a 90% aqueous solution of trichloracetic acid, possible cause for this type of resorption (206). There-
curettage and sealing of the defect proves success- fore, the clinician must use sufficient irrigating solu-
ful in many cases (75). It appears that 90% trichlo- tions when performing root scaling with ultrasonic
racetic acid has a softening effect on dental hard- devices, as well as when using cauterization during
tissues (113). Large defects associated with surgical procedures.
advanced stages of this condition have a poor Internal root resorption is usually asymptomatic
prognosis. and diagnosed during a routine radiographic exami-
Replacement root resorption and extracanal inva- nation. Early diagnosis is critical for the prognosis.
sive root resorption are usually classified separately in The radiographic appearance of the resorptive defect
the literature. However, on closer inspection they discloses a distorted outline of the root canal. A round
appear to be very similar. Histologically, the cemen- or an oval-shaped enlargement of the root-canal
tum and dentin are invaded and resorbed by nonin- space is usually found. In most cases, resorption of
flamed tissue. Later, a hard bone-like tissue is the adjacent bone does not occur unless large parts of
deposited on the resorbed dentin surface, leading to the pulp become infected. Histologically, pulpal gran-
ankylosis. ulation tissue associated with multinucleated giant
cells and coronal pulp necrosis is commonly found.
Infective root resorption. This process occurs as a When diagnosed at an early stage, endodontic treat-
result of a vascular response to microorganisms ment of such lesions is usually uneventful and the
invading the affected tissues. It may occur in both the prognosis is excellent (206).
pulp space and the periodontium and be located
Perforations
either within the root-canal space (internal resorp-
tion) or on the external root surface of the root (exter- Root perforations are undesirable clinical complica-
nal resorption). In the pulp, this process is associated tions that may lead to treatment failure. When root
with an inflammatory response that progresses until perforation occurs, communication between the
the pulp becomes necrotic. Usually, this is also root-canal system and either peri-radicular tissues
accompanied by periradicular inflammation. Practi- (including the periodontal ligament) or the oral cavity
cally, almost all teeth with apical periodontitis will may often worsen the prognosis of treatment. Root
exhibit a certain degree of root resorption (45), which perforations may result from extensive carious
can be located either on the apical or on the lateral lesions, resorption or operator error occurring during
aspects of the root but more frequently at the apex. root-canal instrumentation or post preparation (106,
During initial stages, the resorption cannot be 201).
detected radiographically; however, it is evident in Treatment prognosis of root perforations depends
histological sections. If allowed to progress, the on the size, location, time of diagnosis and treatment,
resorptive process can destroy the entire root. If degree of periodontal damage and the sealing ability
detected and treated early, the prognosis is good. and biocompatibility of the repair material (60). It has
Removal of the inflamed pulpal tissue and obturation been recognized that treatment success depends
of the root-canal system is the treatment of choice mainly on immediate sealing of the perforation and
(36). appropriate infection control. Several materials have
In some cases, an internal root-resorption process been recommended to seal root perforations, includ-
occurs as a result of activity of multinucleated giant ing, among others, mineral trioxide aggregate, Super
cells in an inflamed pulp. The origin of this condition EBA, Cavit, Intermediate Restorative Material, glass
is not fully understood but appears to be related to ionomer cements, composites and amalgam (42, 89,
chronic pulpal inflammation associated with an 111, 144, 157). At present, mineral trioxide aggregate
infected coronal pulp space (212). This resorption will is the material most widely used.
only take place in the presence of granulation tissue An excellent and conservative treatment modality
and if the odontoblastic layer and predentin are for perforations, root resorptions and certain root
affected or lost (203, 213). When confined to the root- fractures is controlled root extrusion (182). This pro-
canal space only, the implications on the periodontal cedure has a good prognosis and a low risk of relapse,
ligament are minimal. However, once the resorptive and its versatility has been demonstrated in multiple
defect perforates the dentin walls, periodontal com- clinical situations (31, 53, 194). It can be performed
plications will ensue. either immediately or over a period of a few weeks,

25
Rotstein

depending on each individual case. The goal of con- A


trolled root extrusion is to modify the soft tissues and
bone and it is therefore used to correct gingival dis-
crepancies and osseous defects of periodontally
involved teeth (194). It is also used in the manage-
ment of nonrestorable teeth.
The objective of forced eruption in prosthetically
compromised, endodontically treated teeth is to allow
restoration of the subcrestal defect by elevating the
defect to a point where access is no longer a problem
(182). In all cases, the epithelial attachment remains
B
at the level of the cemento–enamel junction. Forced
eruption also presents a good alternative to crown
lengthening because it prevents esthetic alterations
and unnecessary reduction of the bony support of
adjacent teeth.

Developmental malformation

Teeth with developmental malformations tend to fail


to respond to treatment when they are directly asso-
ciated with an invagination or a vertical developmen-
tal radicular groove (183). Such conditions can lead to
an untreatable periodontal condition. These grooves
C
usually begin in the central fossa of maxillary central
and lateral incisors crossing over the cingulum and
continuing apically down the root for varying dis-
tances. Such a groove is probably the result of an
attempt of the tooth germ to form another root. As
long as the epithelial attachment remains intact, the
periodontium remains healthy. However, once this
attachment is breached and the groove becomes con-
taminated, a self-sustaining infrabony pocket can
form along its entire lengh (31). This fissure-like chan-
nel provides a nidus for accomulation of bacterial D
biofilm and an avenue for the progression of peri-
odontal disease that may also affect the pulp. Radio-
graphically, the area of bone destruction follows the
course of the groove (5).
From the diagnosis standpoint, the patient may
present signs and symptoms of a periodontal condi-
tion or a variety of asymptomatic endodontic con-
ditions. If the condition is purely periodontal, it can
be diagnosed by visually following the groove to the Fig. 15. Primary endodontic disease in a mandibular first
molar with a necrotic pulp. (A) Abscess on the buccal
gingival margin and by probing the depth of the
aspect of the tooth is traced with a gutta-percha cone. (B)
pocket, which is usually tubular in form and local- Periapical radiograph showing radiolucency associated
ized to this one area, as opposed to a more gener- with the mesial and distal roots, as well as the furcation
alized periodontal problem. The tooth will respond area. (C) Radiograph taken 4 months after root-canal ther-
to pulp-testing procedures. Bone destruction that apy showing active bony healing. (D) Clinically, the buccal
periodontal defect has healed and probing is normal.
vertically follows the groove may be apparent radio-
(Courtesy of Dr Ziv Simon, Beverly Hills, CA, USA.)
graphically. If this condition is also associated with
an endodontic disease, the patient may present The prognosis of root-canal treatment in such cases
clinically with any of the spectrum of endodontic is guarded, being dependent on the apical extent of
symptoms. the groove. The clinician must look for the groove

26
Interaction between endodontics and periodontics

because it may have been altered by a previous access (ii) primary periodontal diseases; and (iii) combined
opening or restoration placed in the access cavity. diseases. The combined diseases include: (i) primary
The appearance of a teardrop-shaped area on the endodontic disease with secondary periodontal
radiograph should immediately arouse suspicion. The involvement; (ii) primary periodontal disease with
developmental groove may actually be visible on the secondary endodontic involvement; and (iii) true
radiograph. If so, it will appear as a dark vertical line. combined diseases.
This condition must be differentiated from a vertical This classification is based on theoretic pathways
fracture, which may give a similar radiographic explaining how these radiographic lesions are
appearance. formed. By understanding the pathogenesis, the clini-
Treatment consists of burring out the groove, cian can then suggest an appropriate course of treat-
placement of bone substitutes and surgical manage- ment and assess the prognosis. Once the lesions
ment of the soft tissues and underlying bone. progress to their final involvement, they give a similar
Recently, Emdogain was also suggested as a treat- radiographic picture and the differential diagnosis
ment adjunct (5). Radicular grooves are self-sustain- becomes more challenging.
ing infrabony pockets and therefore scaling and root
planing will not suffice. Although the acute nature of
Primary endodontic diseases
the problem may be alleviated initially, the source of
the chronic or acute inflammation must be eradi- An acute exacerbation of a chronic apical lesion in a
cated by a surgical approach. Occasionally, the tooth tooth with a necrotic pulp may occasionally drain
needs to be extracted because of poor prognosis. coronally through the periodontal ligament into the
gingival sulcus. This condition may mimic clinically
the presence of a periodontal abscess. In reality, it is a
Differential diagnosis sinus tract of pulpal origin that opens through the
periodontal ligament area. For diagnosis purposes, it
For differential diagnostic and treatment purposes is essential for the clinician to insert a gutta-percha
the so-called ‘endo–perio lesions’ are best classified cone, or another tracking instrument, into the sinus
as endodontic, periodontal or combined diseases tract and to take one or more radiographs to deter-
(163). These include: (i) primary endodontic diseases; mine the origin of the lesion. When the pocket is

A B

Fig. 16. Primary periodontal disease


in a mandibular second molar. The
patient was referred for endodontic
therapy. (A) Preoperative radiograph
C D showing periradicular radiolucency;
however, the tooth responded nor-
mally to pulp-sensitivity tests. The
referring dentist insisted that
endodontic therapy should be per-
formed. (B) Photomicrograph of the
pulp tissue removed during treat-
ment. Note the normal appearance
of the pulp. (C) Higher magnification
shows normal cellular components
as well as blood microvasculature.
(D) Postoperative radiograph. The
tooth was subsequently lost to peri-
odontal disease.

27
Rotstein

probed, it is narrow and lacks width. A similar situa-


A
tion occurs when drainage from the apex of a molar
tooth extends coronally into the furcation area. This
may also occur in the presence of lateral canals
extending from a necrotic pulp into the furcation area
(163).
Primary endodontic diseases usually heal following
root-canal treatment (Fig. 15). The sinus tract extend-
ing into the gingival sulcus or furcation area disap-
pears at an early stage once the affected necrotic pulp B
has been removed and the root canals are well
cleaned, shaped and obturated (163).

Primary periodontal diseases


These lesions are caused primarily by periodontal
pathogens. In this process, chronic marginal peri-
odontitis progresses apically along the root surface.
In most cases, pulp tests indicate a clinically normal C
pulpal reaction (Figs 16 and 17). There is frequently
an accumulation of plaque and calculus and the
pockets are wider.
The prognosis depends upon the stage of periodon-
tal disease and the efficacy of periodontal treatment.
The clinician must also be aware of the radiographic
appearance of periodontal disease associated with
developmental radicular anomalies (Fig. 18).

Combined diseases
D
Primary endodontic disease with secondary
periodontal involvement

If, after a period of time, a suppurating primary


endodontic disease remains untreated, it may then
become secondarily involved with marginal peri-
odontal breakdown (Figs 19 and 20). Plaque forms at
the gingival margin of the sinus tract and leads to
Fig. 17. Primary periodontal lesion simulating an
periodontitis. When plaque or calculus is present, the endodontic lesion. (A) Radiograph of the mandibular first
treatment and prognosis of the tooth are different molar showing periradicular radiolucency and periapical
from those of teeth involved with only primary resorption. (B, C) Buccal and lingual views of the affected
endodontic disease. The tooth now requires both tooth. Note gingival swelling and evidence of periodontal
disease. In addition, an occulsal filling is present close to
endodontic and periodontal treatments. If the
the pulp chamber. In spite of the clinical and radiographic
endodontic treatment is adequate, the prognosis picture, the pulp responded normally to vitality testing
depends on the severity of the marginal periodontal procedures, indicating that the radiolucency, resorption
damage and the efficacy of periodontal treatment. and gingival swelling are of periodontal origin. (D)
With endodontic treatment alone, only part of the Microphotograph stained with hematoxylin and eosin,
showing the floor of the pulp chamber and entrance to the
lesion will heal to the level of the secondary periodon-
mesial canal containing normal pulp tissue.
tal lesion. In general, healing of the tissues damaged
by suppuration from the pulp can be anticipated
(163). where pins or posts have been misplaced during
Primary endodontic lesions with secondary peri- coronal restoration (Fig. 21). Symptoms may be
odontal involvement may also occur as a result of acute, with periodontal abscess formation associated
root perforation during root-canal treatment, or with pain, swelling, pus exudate, pocket formation

28
Interaction between endodontics and periodontics

A B C

D E F

G H

Fig. 18. Primary periodontal disease in a maxillary sec- (C) Clinical view of the extracted tooth with the attached
ond premolar. (A) Radiograph showing alveolar bone lesion. Note a deep mesial radicular development
loss and a periapical lesion. Clinically, a deep, narrow groove. (D) Photomicrograph of the apex of the tooth
pocket was found on the mesial aspect of the root. with the attached lesion. (E, F) Higher magnification
There was no evidence of caries and the tooth shows the inflammatory lesion, cementum and dentin
responded normally to pulp-sensitivity tests. (B) Radio- resorption, and osteoclasts. (G, H) Histologic sections of
graph showing pocket tracking with gutta-percha cone the pulp chamber show uninflamed pulp, odontoblastic
to the apical area. It was decided to extract the tooth. layer and intact predentin.

A B C

Fig. 19. Primary endodontic disease with secondary peri- resolution of most of the periradicular lesion; however, a
odontal involvement in a mandibular first molar. (A) bony defect at the furcal area remained. Note that
Preoperative radiograph demonstrating an interradicular endodontic treatment alone did not yield complete heal-
defect extending to the apical region of the mesial root. ing of the defect. Periodontal treatment is necessary for
(B) Radiograph taken at completion of root-canal further healing of the furcal area and inflamed gingival
therapy. (C) One-year follow-up radiograph showing tissues.

and tooth mobility. A more chronic response may When the root perforation is situated close to the
sometimes occur without pain and involves the sud- alveolar crest, it may be possible to raise a flap and
den appearance of a pocket with bleeding on probing repair the defect with an appropriate filling mate-
or exudation of pus. rial. In deeper perforations, or in the roof of the

29
Rotstein

A B

C D

Fig. 20. Primary endodontic disease


with secondary periodontal involve-
ment. (A) Preoperative radiograph of
a symptomatic mandibular right
E F first molar. Pulp-sensitivity tests
were negative. Initial diagnosis of
periapical periodontitis caused by a
necrotic pulp was established. (B)
Endodontic treatment was per-
formed; however, a residual deep
pocket (7–9 mm) on the mesial root
remained. (C) Examination reveals a
large mucogingival defect with mini-
mal keratinization and absence of
attached gingiva. (D) Upon explo-
G ration, severe bone loss and abun-
dant calculus on the mesial root
were observed. (E) The root surface
was then debrided and the bony
defect cleaned. (F) The bony defect
was repaired with bone graft mate-
rial and a resorbable membrane. (G)
Two-month follow-up shows active
healing of the defect. (Courtesy of Dr
Ziv Simon, Beverly Hills, CA, USA).

fucation, immediate repair of the perforation has a


Primary periodontal disease with secondary
better prognosis than management of an infected
endodontic involvement
one. It has been shown that the use of mineral tri-
oxide aggregate in such cases may enhance cemen- The apical progression of a periodontal pocket may
tal healing following immediate perforation repair continue until the apical tissues are involved. In this
(150). case, the pulp may become necrotic as a result of
Root fractures may also present as primary infection entering via lateral canals or the apical fora-
endodontic lesions with secondary periodontal men (Fig. 22). In single-rooted teeth the prognosis is
involvement. These typically occur on root-treated usually poor. In molar teeth the prognosis may be
teeth, often with post and crowns. The signs may better. As not all roots may experience the same loss
range from a local deepening of a periodontal pocket, of supporting tissues, root resection can be consid-
to more acute periodontal abscess formation. Root ered as a treatment alternative.
fractures have also become an increasing problem The effect of progressive periodontitis on the vital-
with molar teeth that have been treated by root resec- ity of the pulp is controversial (2, 3, 107). If the blood
tion (108, 160). supply circulating through the apex is intact, the pulp

30
Interaction between endodontics and periodontics

A B
Fig. 21. Complications of certain
defects along the root may have sim-
ilar clinical appearance of primary
endodontic disease with secondary
periodontal involvement. (A) The
patient presented with a localized
deep pocket (traced with a gutta-
percha cone) associated with an
endodontically treated mandibular
second premolar. (B) Periapical
radiograph showing the gutta-
C D percha cone pointing to an area of
the root associated with a screw
post. (C) Subsequent abscess forma-
tion on the buccal aspect of the tooth
caused loss of the buccal plate. (D)
Surgical exploration confirmed the
presence of a vertical root fracture
and the tooth was extracted and
replaced with an implant. (Courtesy
of Dr Ziv Simon, Beverly Hills, CA,
USA).

has good prospects for survival. It was reported that teeth (Fig. 24). In molar teeth, root resection can be
pulpal changes resulting from periodontal disease are considered as a treatment alternative if not all roots
more likely to occur when the apical foramen is are severely involved. Sometimes, supplementary sur-
involved (107). In these cases, bacteria originating gical procedures are necessary (Fig. 25). In most
from the periodontal pocket are the source of root- cases, periradicular healing may be anticipated fol-
canal infection. A strong correlation between the lowing successful endodontic treatment. The peri-
presence of microorganisms in root canals and their odontal tissues, however, may not respond well to
presence in periodontal pockets of advanced peri- treatment and will depend on the severity of the com-
odontitis has been demonstrated (99, 101). Support bined disease.
for this concept has come from research in which cul- The radiographic appearance of combined
tured samples obtained from the pulp tissue and endodontic–periodontal disease may be similar to
radicular dentin of periodontally involved human that of a vertically fractured tooth. A fracture that has
teeth showed bacterial growth in 87% of the teeth invaded the pulp space, with resultant necrosis, may
(2, 3). also be labeled a true combined lesion and yet not be
The treatment of periodontal disease can also lead amenable to successful treatment. If a sinus tract is
to secondary endodontic involvement. Lateral canals present, it may be necessary to raise a flap to deter-
and dentinal tubules may be opened to the oral envi- mine the etiology of the lesion.
ronment by curettage, scaling or surgical flap proce-
dures. It is possible for a blood vessel within a lateral
canal to be severed by a curette and for microorgan- Prognosis
isms to be pushed into the area during treatment,
thus resulting in pulp inflammation and necrosis Treatment prognosis depends primarily on the diag-
(163). nosis of the specific endodontic and/or periodontal
disease. The main factors to consider for treatment
True combined diseases
decision-making are pulp vitality and type and extent
True combined endodontic–periodontal disease of the periodontal defect. Diagnosis of primary
occurs with less frequency. It arises when an endodontic disease and primary periodontal disease
endodontic disease progressing coronally joins with usually presents no clinical difficulty. In primary
an infected periodontal pocket progressing apically endodontic disease the pulp is infected and its vitality
(177, 184). The degree of attachment loss in this type is affected. On the other hand, in a tooth with primary
of lesion is invariably large and the prognosis guarded periodontal disease, pulp vitality is not affected and
(Fig. 23). This is particularly true in single-rooted the tooth is responsive to clinical pulp testing.

31
Rotstein

A A

Fig. 22. Primary periodontal disease with secondary


endodontic involvement in a maxillary premolar. (A)
Radiograph showing bone loss in one-third of the root and
separate periapical radiolucency. The crown was intact but
pulp-sensitivity tests were negative. (B) Radiograph taken Fig. 24. True combined endodontic–periodontal disease.
immediately after root-canal therapy showing sealer in the (A) Radiograph showing bone loss in two-thirds of the root
lateral canal that was exposed as a result of bone loss. with calculus present and separate periapical radiolu-
cency. (B) Clinical examination revealed coronal color
change of the tooth involved and pus exuding from the
gingival crevis. Pulp-sensitivity tests were negative.

primarily endodontic disease because of lack of evi-


dence of periodontitis, and there is soft-tissue healing
on clinical probing and bony healing on a recall
radiogragh, a valid retrospective diagnosis can then
be made. The degree of healing that has taken place
following root-canal treatment will determine the ret-
rospective classification. In the absence of adequate
Fig. 23. True combined endodontic–periodontal disease in healing, further periodontal treatment is indicated.
a mandibular first molar. Radiograph showing separate The prognosis and treatment of each endodontic–
progression of endodontic disease and periodontal disease. periodontal disease type vary. Primary endodontic
The tooth remained untreated and consequently the two disease should only be treated by endodontic therapy
lesions joined together.
with good prognosis. Primary periodontal disease
should only be treated by periodontal therapy. In this
However, primary endodontic disease with secondary case, the prognosis depends on the severity of the
periodontal involvement, primary periodontal disease periodontal disease and the patient response. Pri-
with secondary endodontic involvement, or true mary endodontic disease with secondary periodontal
combined diseases are clinically and radiographically involvement should first be treated with endodontic
very similar. If a lesion is diagnosed and treated as therapy. Treatment results should be evaluated in 2–

32
Interaction between endodontics and periodontics

A B

C D

Fig. 25. True combined endodontic–


periodontal diseases in a mandibular
first molar. (A) Preoperative radio-
graph showing periradicular radiolu-
cencies. Pulp-sensitivity tests were
negative. (B) Immediate postopera-
tive radiograph of nonsurgical
E endodontic treatment. (C) Six-
month follow-up radiograph show-
ing no evidence of healing. Gutta-
percha cone is inserted in the buccal
gingival sulcus. (D) Clinical pho-
tograph showing treatment of the
root surfaces and removal of the
periradicular lesion. (E) One-year
follow-up radiograph demonstrates
evidence of active healing.

3 months and only then should periodontal treat- showed 10% more connective tissue coverage than
ment be considered. This sequence of treatment infected teeth (86). The prognosis of primary peri-
allows sufficient time for initial tissue healing and odontal disease with secondary endodontic involve-
better assessment of the periodontal condition (26, ment and true combined diseases depends primarily
146). It also reduces the potential risk of introducing upon the severity of the periodontal disease and the
bacteria and their by-products during the initial heal- response of periodontal tissue to treatment.
ing phase. In this regard, it was suggested that aggres- True combined diseases usually have a guarded
sive removal of the periodontal ligament and prognosis. In general, assuming the endodontic
underlying cementum during interim endodontic therapy is adequate, what is of endodontic origin
therapy adversely affects periodontal healing (21). will heal. Thus, the long-term prognosis of com-
Areas of the roots that were not aggressively treated bined diseases rests with the efficacy of periodontal
showed unremarkable healing (21). Prognosis of pri- therapy.
mary endodontic disease with secondary periodontal
involvement depends primarily on the severity of
periodontal involvement, periodontal treatment and Acknowledgment
patient response.
Primary periodontal disease with secondary In memory of Dr James H. S. Simon (1934–2013), a
endodontic involvement and true combined great mentor, teacher and friend.
endodontic–periodontal diseases require both
endodontic and periodontal therapies. It has been
demonstrated that intrapulpal infection tends to pro- References
mote marginal epithelial downgrowth along a
denuded dentin surface (21). Additionally, experi- 1. Ackerman SJ, Corrette SE, Rosenberg HF, Bennett JC, Mas-
trianni DM, Nicholson-Weller A, Weller PF, Chin DT,
mentally induced periodontal defects in infected
Tenen DG. Molecular cloning and characterization of
teeth were associated with 20% more epithelial cells human eosinophils Charcot-Leyden crystal protein
than noninfected teeth (86). Noninfected teeth (lysophospholipase). J Immunol 1993: 150: 456–468.

33
Rotstein

2. Adriaens PA, De Boever JA, Loesche WJ. Bacterial invasion 20. Blomlo € f L, Lengheden A, Lindskog S. Endodontic infection
in root cementum and radicular dentin of periodontally and calcium hydroxide treatment. Effects on periodontal
diseased teeth in humans. A reservoir of periodontopathic healing in mature and immature replanted monkey teeth.
bacteria. J Periodontol 1988: 59: 222–230. J Clin Periodontol 1992: 19: 652–658.
3. Adriaens PA, Edwards CA, De Boever JA, Loesche WJ. 21. Blomlo € f LB, Lindskog S, Hammarstrom L. Influence of pul-
Ultrastructural observations on bacterial invasion in pal treatments on cell and tissue reactions in the marginal
cementum and radicular dentin of periodontally diseased periodontium. J Periodontol 1988: 59: 577–583.
human teeth. J Periodontol 1988: 59: 493–503. 22. Boyko GA, Melcher AH, Brunette DM. Formation of new
4. Ahmed GM, El-Baz AA, Hashem AA, Shalaan AK. Expres- periodontal ligament by periodontal ligament cells
sion levels of matrix metalloproteinase-9 and gram-nega- implanted in vivo after culture in vitro. J Periodontal Res
tive bacteria in symptomatic and asymptomatic periapical 1981: 16: 73–88.
lesions. J Endod 2013: 39: 444–448. 23. Browne RM. The origin of cholesterol in odontogenic cysts
5. Al-Hezaimi K, Naghshbandi J, Simon JHS, Rotstein I. Suc- in man. Arch Oral Biol 1971: 16: 107–113.
cessful treatment of a radicular groove by intentional 24. Burch JG, Hulen S. A study of the presence of accessory
replantation and Emdogain therapy: four years follow-up. foramina and the topography of molar furcations. Oral
Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2009: Surg Oral Med Oral Pathol 1974: 38: 451–455.
107: e82–e85. 25. Carson HJ, Buschmann RJ, Weisz-Carrington P, Choi YS.
6. Allison RT. Electron microscopic study of “Rushton” hya- Identification of Charcot-Leyden crystals by electron
line bodies in cyst linings. Br Dent J 1974: 137: 102–104. microscopy. Ultra Path 1992: 16: 403–411.
7. American Association of Endodontists. Glossary of 26. Chapple I, Lumley PJ. The periodontal-endodontic inter-
endodontic terms, 8th ed. Chicago: American Association face. Dent Update 1999: 26: 331–334.
of Endodontists, 2012. 27. Choi BK, Paster BJ, Dewhirst FE, Gobel UB. Diversity of
8. Andreasen FM. Pulpal healing after luxation injuries and cultivable and uncultivable oral spirochetes from a patient
root fracture in the permanent dentition. Endod Dent with severe destructive periodontitis. Infect Immun 1994:
Traumatol 1989: 5: 111–131. 62: 1889–1895.
9. Andreasen FM, Flugge E, Daugaard-Jensen J, Munksgaard 28. Contreras A, Nowzari H, Slots J. Herpesviruses in peri-
EC. Treatment of crown fractured incisors with laminate odontal pocket and gingival tissue specimens. Oral Micro-
veneer restorations. An experimental study. Endod Dent biol Immunol 2000: 15: 15–18.
Traumatol 1992: 8: 30–35. 29. Contreras A, Slots J. Herpesviruses in human periodontal
10. Andreasen JO. Periodontal healing after replantation of disease. J Periodontal Res 2000: 35: 3–16.
traumatically avulsed human teeth. Assessment by mobil- 30. Contreras A, Slots J. Typing of herpes simplex virus from
ity testing and radiography. Acta Odontol Scand 1975: 33: human periodontium. Oral Microbiol Immunol 2001: 16:
325–335. 63–64.
11. Andreasen JO, Andreasen FM, Skeie A, Hjorting-Hansen E, 31. Corr A-Faria P, Alcantara CE, Santos CR, Marques LS,
Schwartz O. Effect of treatment delay upon pulp and peri- Ramos-Jorge ML. Palatal radicular groove: clinical implica-
odontal healing of traumatic dental injuries. Dent Trauma- tions of early diagnosis and surgical sealing. J Indian Soc
tol 2002: 18: 116–128. Pedod Prev Dent 2011: 29: S92–S94.
12. Baumgartner JC. Microbiologic aspects of endodontic 32. Costerton JW, Lewandowski Z, Caldwell DE, Korber DR,
infections. J Calif Dent Assoc 2004: 32: 459–468. Lappin-Scott HM. Microbial biofilms. Annu Rev Microbiol
13. Baumgartner JC, Falkler WA Jr. Bacteria in the apical 1995: 49: 711–745.
5 mm of infected root canals. J Endod 1991: 17: 380–383. 33. Costerton JW, Stewart PS, Greenberg EP. Bacterial bio-
14. Baumgartner JC, Watts CM, Xia T. Occurrence of Candida films: a common cause of persistent infections. Science
albicans in infections of endodontic origin. J Endod 2000: 1999: 284: 1318–1322.
26: 695–698. 34. Costerton W, Veeh R, Shirtliff M, Pasmore M, Post C, Ehr-
15. Bechara B, McMahan CA, Nasseh I, Geha H, Hayek E, Kha- lich G. The application of biofilm science to the study and
wam G, Raad M, Noujeim M. Number of basis images control of chronic bacterial infections. J Clin Invest 2003:
effect on detection of root fractures in endodontically trea- 112: 1466–1477.
ted teeth using a cone beam computed tomography 35. Cotran RS, Kumar V, Collins T. Robbins’ pathologic basis of
machine: an in vitro study. Oral Surg Oral Med Oral Pathol disease, 6th ed. Philadelphia: WB Saunders, 1999.
Oral Radiol 2013: 115: 676–681. 36. Cvek M. Treatment of non-vital permanent incisors. II.
16. Bender IB, Seltzer S. The effect of periodontal disease on Effect on external root resorption in luxated teeth com-
the pulp. Oral Surg Oral Med Oral Pathol 1972: 33: 458– pared with the effect of root filling with gutta-percha.
474. Odontol Revy 1973: 24: 343–354.
17. Bergenholtz G, Lekholm U, Milthon R, Heden G, Odesjo B, 37. Czarnecki RT, Schilder H. A histological evaluation of the
Engstrom B. Retreatment of endodontic fillings. Scand J human pulp in teeth with varying degrees of periodontal
Dent Res 1979: 87: 217–224. disease. J Endod 1979: 5: 242–253.
18. Bergenholtz G, Lindhe J. Effect of soluble plaque factors 38. Dahle UR, Tronstad L, Olsen I. Observation of an unusu-
on inflammatory reactions in the dental pulp. Scand J Dent ally large spirochete in endodontic infection. Oral Micro-
Res 1975: 83: 153–158. biol Immunol 1993: 8: 251–253.
19. Bhaskar SN. Periapical lesion: types, incidence, and clini- 39. Dahle UR, Tronstad L, Olsen I. Characterization of new
cal features. Oral Surg Oral Med Oral Pathol 1966: 21: 657– periodontal and endodontic isolates of spirochetes. Eur J
671. Oral Sci 1996: 104: 41–47.

34
Interaction between endodontics and periodontics

40. Dahle n G, Wikstro € m M. Occurrence of enteric rods, 58. Friedman S, Rotstein I, Libfeld H, Stabholz A, Heling I.
staphylococci and Candida in subgingival samples. Oral Incidence of external root resorption and esthetic results
Microbiol Immunol 1995: 10: 42–46. in 58 bleached pulpless teeth. Endod Dent Traumatol
41. Damm DD, Neville BW, Geissler RH Jr, White DK, Drum- 1988: 4: 23–26.
mond JF, Ferretti GA. Dentinal candidiasis in cancer 59. Friend LA, Browne RM. Tissue reactions to some root fill-
patients. Oral Surg Oral Med Oral Pathol 1988: 65: 56–60. ing materials. Br Dent J 1968: 125: 291–298.
42. Dazey S, Senia ES. An in vitro comparison of the sealing 60. Fuss Z, Trope M. Root perforations: classification and
ability of materials placed in lateral root perforations. J treatment choices based on prognostic factors. Endod
Endod 1990: 16: 19–23. Dent Traumatol 1996: 12: 255–264.
43. de Beer D, Stoodley P, Lewandowski Z. Liquid flow in 61. Fux CA, Costerton JW, Stewart PS, Stoodley P. Survival
heterogeneous biofilms. Biotechnol Bioeng 1994: 44: 636– strategies of infectious biofilms. Trends Microbiol 2005: 13:
641. 34–40.
44. De Deus QD. Frequency, location, and direction of the lat- 62. Giardino L, Savoldi E, Pontieri F, Berutti E. Russel bodies
eral, secondary, and accessory canals. J Endod 1975: 1: in dental pulp of permanent human teeth. Oral Surg Oral
361–366. Med Oral Pathol Oral Radiol Endod 2004: 98: 760–764.
45. Delzangles B. Apical periodontitis and resorption of the 63. Glick M, Trope M, Pliskin ME. Detection of HIV in the den-
root canal wall. Endod Dent Traumatol 1988: 4: 273– tal pulp of a patient with AIDS. J Am Dent Assoc 1989: 119:
277. 649–650.
46. Dewhirst FE, Tamer MA, Ericson RE, Lau CN, Levanos VA, 64. Gold SI, Moskow BS. Periodontal repair of periapical
Boches SK, Galvin JL, Paster BJ. The diversity of periodon- lesions: the borderland between pulpal and periodontal
tal spirochetes by 16S rRNA analysis. Oral Microbiol disease. J Clin Periodontol 1987: 14: 251–256.
Immunol 2000: 15: 196–202. 65. Goldberg F, Massone EJ, Soares I, Bittencourt AZ. Acces-
47. Dvorak AM, Weller PF, Monahan-Earley RA, Letoumeau L, sory orifices: anatomical relationship between the pulp
Ackerman SJ. Ultrastructural localization of Charcot-Ley- chamber floor and the furcation. J Endod 1987: 13: 176–
den crystal protein (lysophospholipase) and peroxidase in 181.
macrophages, eosinophils, and extracellular matrix of the 66. Goldman M, Laosonthorn P, White RR. Microleakage – full
skin in the hypereosinophilic syndrome. Lab Invest 1990: crowns and the dental pulp. J Endod 1992: 18: 473–475.
62: 590–607. 67. Goldman M, Pearson AH. Post debridement bacterial flora
48. Edlund M, Nair MK, Nair UP. Detection of vertical root and antibiotic sensitivity. Oral Surg Oral Med Oral Pathol
fractures by using cone-beam computed tomography: a 1969: 28: 897–905.
clinical study. J Endod 2011: 37: 768–772. 68. Gutmann JL. Prevalence, location, and patency of acces-
49. Egan MW, Spratt DA, Ng YL, Lam JM, Moles DR, Gulabi- sory canals in the furcation region of permanent molars. J
vala K. Prevalence of yeasts in saliva and root canals of Periodontol 1978: 49: 21–26.
teeth associated with apical periodontitis. Int Endod J 69. Haapasalo M, Ranta H, Ranta K, Shah H. Black-pigmented
2002: 35: 321–329. Bacteroides spp. in human apical periodontitis. Infect
50. Ehnevid H, Jansson L, Lindskog S, Weintraub A, Blomlof L. Immun 1986: 53: 149–153.
Endodontic pathogens: propagation of infection through 70. Hannula J, Saarela M, Alaluusua S, Slots J, Asikainen S.
patent dentinal tubules in traumatized monkey teeth. Phenotypic and genotypic characterization of oral yeasts
Endod Dent Traumatol 1995: 11: 229–234. from Finland and the United States. Oral Microbiol Immu-
51. Elkins DA, Torabinejad M, Schmidt RE, Rossi JJ, Kettering nol 1997: 12: 358–365.
JD. Polymerase chain reaction detection of human 71. Harn WM, Chen YH, Yuan K, Chung CH, Huang PH. Cal-
immunodeficiency virus DNA in human periradicular culus-like deposit at apex of tooth with refractory apical
lesions. J Endod 1994: 20: 386–388. periodontitis. Endod Dent Traumatol 1998: 14: 237–240.
52. El-Labban NG. Electron microscopic investigation of hya- 72. Harrington GW, Natkin E. External resorption associated
line bodies in odontogenic cysts. J Oral Pathol 1979: 8: 81– with bleaching of pulpless teeth. J Endod 1979: 5: 344–348.
93. 73. Heithersay GS. Clinical, radiographic, and histopathologic
53. Emerich-Poplatek K, Sawicki L, Bodal M, Adamowitz-Kle- features of invasive cervical resorption. Quintessence Int
palska B. Forced eruption after crown/root fracture with a 1999: 30: 27–37.
simple and aesthetic method using the fractured crown. 74. Heithersay GS. Invasive cervical root resorption: an analy-
Dent Traumatol 1995: 21: 165–169. sis of potential predisposing factors. Quintessence Int 1999:
54. Engstro€ m B, Spangberg L. Wound healing after partial 30: 83–95.
pulpectomy. A histological study performed on contralat- 75. Heithersay GS. Treatment of invasive cervical resorption:
eral tooth pairs. Odontol Tidskr 1967: 75: 5–18. an analysis of results using topical application of trichlo-
55. Fabricius L, Dahle n G, Ohman AE, Mo € ller AJR. Predomi- racetic acid, curettage, and restoration. Quintessence Int
nant indigenous oral bacteria isolated from infected root 1999: 30: 96–110.
canals after varied times of closure. Scand J Dent Res 1982: 76. Heithersay GS, Dahlstrom SW, Marin PD. Incidence of
90: 134–144. invasive cervical resorption in bleached root-filled teeth.
56. Fenno JC, McBride BC. Virulence factors of oral tre- Aust Dent J 1994: 39: 82–87.
ponemes. Anaerobe 1998: 4: 1–17. 77. Heling I, Gorfil C, Slutzky H, Kopolovic K, Zalkind M,
57. Fine L, Mostofi R, Wiemann MR, Crinzi RA. Foreign body- Slutzky-Goldberg I. Endodontic failure caused by inade-
type reaction following crown cementation. J Periodontol quate restorative procedures: review and treatment rec-
1977: 48: 294–297. ommendations. J Prosthet Dent 2002: 87: 674–678.

35
Rotstein

78. Heling I, Morag-Hezroni M, Marva E, Hochman N, Zakay- 96. Keresztesi K, Kellner G. The biological effects of root filling
Rones Z, Morag A. Is herpes simplex virus associated with materials. Int Dent J 1966: 16: 222–231.
pulp/periapical inflammation? Oral Surg Oral Med Oral 97. Kessler S. Bacteriological examination of root canals. J
Pathol Oral Radiol Endod 2001: 91: 359–361. Dent Assoc S Afr 1972: 27: 9–13.
79. Heling I, Parson A, Rotstein I. Effect of bleaching agents on 98. Kinaia BM, Chogle SM, Kinaia AK, Goodis HE. Regenera-
dentin permeability to Streptococcus faecalis. J Endod tive therapy: a periodontal-endodontic perspective. Dent
1995: 21: 540–542. Clin North Am 2012: 56: 537–547.
80. Heller D, Skriber J, Lin LM. Effect of intracoronal bleaching 99. Kipioti A, Nakou M, Legakis N, Mitsis F. Microbiological
on external cervical root resorption. J Endod 1992: 18: 145– findings of infected root canals and adjacent periodontal
148. pockets in teeth with advanced periodontitis. Oral Surg
81. Heuner K, Grosse K, Schade R, Gobel UB. A flagellar gene Oral Med Oral Pathol 1984: 58: 213–220.
cluster from the oral spirochete Treponema maltophilum. 100. Kirkham DB. The location and incidence of accessory pul-
Microbiology 2000: 146: 497–507. pal canals in periodontal pockets. J Am Dent Assoc 1975:
82. Hodson JJ. Origin and nature of the cuticula dentis. Nature 91: 353–356.
1966: 209: 990–993. 101. Kobayashi T, Hayashi A, Yoshikawa R, Okuda K, Hara K.
83. Holland R, De Souza V, Nery MJ, de Mello W, Bernabe PF, The microbial flora from root canals and periodontal
Otoboni Filho JA. Tissue reactions following apical plug- pockets of non-vital teeth associated with advanced peri-
ging of the root canal with infected dentin chips. A histo- odontitis. Int Endod J 1990: 23: 100–106.
logic study in dogs’ teeth. Oral Surg Oral Med Oral Pathol 102. Kombayashi T, Nonomura G, Watanabe LG, Marshall GW
1980: 49: 366–369. Jr, Marshall SJ. Dentin tubule numerical density variations
84. Jackson FL, Halder AR. Incidence of yeasts in root canals below the CEJ. J Dent 2008: 36: 953–958.
during therapy. Br Dent J 1963: 115: 459–460. 103. Koppang HS, Koppang R, Stolen SO. Identification of com-
85. Jansson L, Ehnevid H. The influence of endodontic infec- mon foreign material in postendodontic granulomas and
tion on periodontal status in mandibular molars. J Peri- cysts. J Dent Assoc S Afr 1992: 47: 210–216.
odontol 1998: 69: 1392–1396. 104. Koppang HS, Koppang R, Solheim T, Aarnes H, Stolen SO.
86. Jansson L, Ehnevid H, Blomlo € f L, Weintraub A, Lindskog S. Cellulose fibers from endodontic paper points as an etio-
Endodontic pathogens in periodontal disease augmenta- logical factor in postendodontic periapical granulomas
tion. J Clin Periodontol 1995: 22: 598–602. and cysts. J Endod 1989: 15: 369–372.
87. Jansson L, Ehnevid J, Lindskog SF, Blomlo € f LB. 105. Korzen BH, Krakow AA, Green DB. Pulpal and periapical
Radiographic attachment in periodontitis-prone teeth tissue responses in conventional and monoinfected gnoto-
with endodontic infection. J Periodontol 1993: 64: 947– biotic rats. Oral Surg Oral Med Oral Pathol 1974: 37: 783–
953. 802.
88. Jansson L, Ehnevid H, Lindskog S, Blomlo € f L. The influ- 106. Kvinnsland I, Oswald RJ, Halse A, Gronningsaeter AG. A
ence of endodontic infection on progression of marginal clinical and roentgenological study of 55 cases of tooth
bone loss in periodontitis. J Clin Periodontol 1995: 22: 729– perforation. Int Endod J 1989: 22: 75–84.
734. 107. Langeland K, Rodrigues H, Dowden W. Periodontal dis-
89. Jew RC, Weine FS, Keene JJ, Smulson MH. A histologic ease, bacteria, and pulpal histopathology. Oral Surg Oral
evaluation of periodontal tissues adjacent to root perfora- Med Oral Pathol 1974: 37: 257–270.
tions filled with Cavit. Oral Surg Oral Med Oral Pathol 108. Langer B, Stein SD, Wagenberg B. An evaluation of root
1982: 54: 124–135. resections: a ten year study. J Periodontol 1981: 52: 719–
90. Jung IY, Choi BK, Kum KY, Roh BD, Lee SJ, Lee CY, Park 722.
DS. Molecular epidemiology and association of putative 109. Lao LM, Kumakiri M, Nakagawa K, Ishida H, Ishiguro K,
pathogens in root canal infection. J Endod 2000: 26: 599– Yanagihara M, Ueda K. The ultrastructural findings of
604. Charcot-Leyden crystals in stroma of mastocytoma. J Der-
91. Jung IY, Choi B, Kum KY, Yoo YJ, Yoon TC, Lee SJ, Lee CY. matol Sci 1998: 17: 198–204.
Identification of oral spirochetes at the species level and 110. Lazarski MP, Walker WA 3rd, Flores CM, Schindler WG,
their association with other bacteria in endodontic infec- Hargreaves KM. Epidemiological evaluation of the out-
tions. Oral Surg Oral Med Oral Pathol Oral Radiol Endod comes of nonsurgical root canal treatment in a large
2001: 92: 329–334. cohort of insured dental patients. J Endod 2001: 27: 791–
92. Kakehashi S, Stanley HR, Fitzgerald RJ. The effects of surgi- 796.
cal exposures of dental pulps in germ-free and conven- 111. Lee SJ, Monsef M, Torabinejad M. Sealing ability of a min-
tional laboratory rats. Oral Surg Oral Med Oral Pathol eral trioxide aggregate for repair of lateral root perfora-
1965: 20: 340–349. tions. J Endod 1993: 19: 541–544.
93. Karring T, Nyman S, Lindhe J, Sirirat M. Potentials for root 112. Leonard EP, Lunin M, Provenza DV. On the occurrence
resorption during periodontal wound healing. J Clin Peri- and morphology of Russell bodies in the dental granu-
odontol 1984: 11: 41–52. loma: an evaluation of seventy-nine specimens. Oral Surg
94. Kasuga Y, Ishihara K, Okuda K. Significance of detection of Oral Med Oral Pathol 1974: 38: 584–590.
Porphyromonas gingivalis, Bacteroides forsythus and Tre- 113. Lewinstein I, Rotstein I. Effect of trichloracetic acid on the
ponema denticola in periodontal pockets. Bull Tokyo Dent microhardness and surface morphology of human dentin
Coll 2000: 41: 109–117. and enamel. Endod Dent Traumatol 1992: 8: 16–20.
95. Kenny JF. Role of cell-wall-defective microbial variants in 114. Lomcali G, Sen BH, Cankaya H. Scanning electron micro-
human infections. South Med J 1978: 71: 180–190. scopic observations of apical root surfaces of teeth with

36
Interaction between endodontics and periodontics

apical periodontitis. Endod Dent Traumatol 1996: 12: 70– 135. Nair PN. Pathogenesis of apical periodontitis and the
76. causes of endodontic failures. Crit Rev Oral Biol Med 2004:
115. Lowman JV, Burke RS, Pellea GB. Patent accessory canals: 15: 348–381.
incidence in molar furcation region. Oral Surg Oral Med 136. Nair PN. Cholesterol as an aetiological agent in endodon-
Oral Pathol 1973: 36: 580–584. tic failures – a review. Aust Endod J 1999: 25: 19–26.
116. Madison S, Walton R. Cervical root resorption following 137. Nair PN, Pajarola G, Schroeder HE. Types and incidence of
bleaching of endodontically treated teeth. J Endod 1990: human periapical lesions obtained with extracted teeth.
16: 570–574. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 1996:
117. Madison S, Wilcox LR. An evaluation of coronal microleak- 81: 93–102.
age in endodontically treated teeth. Part III. In vivo study. 138. Nair PN, Sjogren U, Krey G, Kahnberg KE, Sundqvist G.
J Endod 1988: 14: 455–458. Intraradicular bacteria and fungi in root-filled, asymp-
118. Magnusson I, Claffey N, Bogle G, Garrett S, Egelberg J. tomatic human teeth with therapy-resistant periapical
Root reserption following periodontal flap procedures in lesions: a long-term light and electron microscopic follow-
monkeys. J Periodontal Res 1985: 20: 79–85. up study. J Endod 1990: 16: 580–588.
119. Mandi FA. Histological study of the pulp changes caused 139. Nair PN, Sjogren U, Schumacher E, Sundqvist G. Radicular
by periodontal disease. J Br Endod Soc 1972: 6: 80–82. cyst affecting a root-filled human tooth: a long-term post-
120. Marton IJ, Kiss C. Protective and destructive immune reac- treatment follow-up. Int Endod J 1993: 26: 225–233.
tions in apical periodontitis. Oral Microbiol Immunol 140. Najzar-Fleger D, Filipovic D, Prpic G, Kobler D. Candida in
2000: 15: 139–150. root canals in accordance with oral ecology. Int Endod J
121. Matthews JB. The immunoglobulin nature of Russell bod- 1992: 25: 40.
ies. Br J Exp Path 1983: 64: 331–335. 141. Novic RP. Autoinduction and signal transduction in the
122. Matusow RJ. Acute pulpal-alveolar cellulitis syndrome. III: regulation of staphylococcal virulence. Mol Microbiol
endodontic therapeutic factors and the resolution of a 2003: 48: 1429–1449.
Candida albicans infection. Oral Surg Oral Med Oral 142. Ozok AR, Persoon IF, Huse SM, Keijser BJ, Wesselink PR,
Pathol 1981: 52: 630–634. Crielaard W, Zaura E. Ecology of the microbiome of the
123. Mazur B, Massler M. Influence of periodontal disease on infected root canal system: a comparison between apical
the dental pulp. Oral Surg Oral Med Oral Pathol 1964: 17: and coronal root segments. Int Endod J 2012: 45: 530–541.
592–603. 143. Ng YL, Mann V, Gulabivala K. Tooth survival following
124. Medak H, Weinmann JP. Hyaline bodies in dental cysts. Br non-surgical root canal treatment: a systemic review of
Dent J 1960: 109: 312–317. the literature. Int Endod J 2010: 43: 171–189.
125. Mincer HH, McCoy JM, Turner JE. Pulse granuloma of the 144. Oynick J, Oynick T. Treatment of endodontic perforations.
alveolar ridge. Oral Surg Oral Med Oral Pathol 1979: 48: J Endod 1985: 11: 191–192.
126–130. 145. Patel S, Saunders WP, Burke FJ. Microleakage of dentin
126. Miranda TT, Vianna CR, Rodrigues L, Monteiro AS, Rosa CA, bonded crowns placed with different luting materials. Am
Correa A Jr. Diversity and frequency of yeasts from the dor- J Dent 1997: 10: 179–183.
sum of the tongue and necrotic root canals associated with 146. Paul BF, Hutter JW. The endodontic-periodontal contin-
primary apical periodontitis. Int Endod J 2009: 42: 839–844. uum revisited: new insights into etiology, diagnosis and
127. Mjor IA, Nordahl I. The density and branching of dentinal treatment. J Am Dent Assoc 1997: 128: 1541–1548.
tubules in human teeth. Arch Oral Biol 1996: 41: 401–412. 147. Peciuliene V, Reynaud AH, Balciuniene I, Haapasalo M.
128. Molander A, Reit C, Dahlen G, Kvist T. Microbiological sta- Isolation of yeasts and enteric bacteria in root-filled teeth
tus of root-filled teeth with apical periodontitis. Int Endod with chronic apical periodontitis. Int Endod J 2001: 34:
J 1998: 31: 1–7. 429–434.
129. Mo € ller 
AJR, Fabricius L, Dahle €
n G, Ohman AE, Heyden G. 148. Peters LB, Wesselink PR, Moorer WR. The fate and the role
Influence on periapical tissues of indigenous oral bacteria of bacteria left in root dentinal tubules. Int Endod J 1995:
and necrotic pulp tissue in monkeys. Scand J Dent Res 28: 95–99.
1981: 89: 475–484. 149. Philipsen HP, Reichart PA. Pulse or hyaline ring granu-
130. Molven O, Olsen I, Kerekes K. Scanning electron micro- loma. Review of the literatutre on etiopathogenesis of oral
scopy of bacteria in the apical part of root canals in per- and extraoral lesions. Clin Oral Investig 2010: 14: 121–128.
manent teeth with periapical lesions. Endod Dent 150. Pitt Ford TR, Torabinejad M, McKendry D, Hong CU, Kar-
Traumatol 1991: 7: 226–229. iyawasam SP. Use of mineral trioxide aggregate for repair
131. Morgan PR, Johnson NW. Histological, histochemical and of furcal perforations. Oral Surg Oral Med Oral Pathol
ultrastructural studies on the nature of hyaline bodies in 1995: 79: 756–763.
odontogenic cysts. J Oral Pathol 1974: 3: 127–147. 151. Poornima P, Subba Reddy VV. Comparison of digital
132. Moter A, Hoenig C, Choi BK, Riep B, Gobel UB. Molecular radiography, decalcification, and histologic sectioning in
epidemiology of oral treponemes associated with peri- the detection of accessory canals in furcation areas of
odontal disease. J Clin Microbiol 1998: 36: 1399–1403. human primary molars. J Indian Soc Pedod Prev Dent
133. Muller CJ, Van Wyk CW. The amelo-cemental junction. J 2008: 26: 49–52.
Dent Assoc S Afr 1984: 39: 799–803. 152. Pradhan-Palikhe P, Ma €ntyla
€ P, Paju S, Buhlin K, Pers-
134. Nair MK, Nair UDP, Grondahl HG, Webber RL, Wallace JA. son GR, Nieminen MS, Sinisalo J, Pussinen PJ. Subgin-
Detection of artificially induced vertical radicular fractures gival bacterial burden in relation to the clinical and
using tuned aperture computed tomography. Eur J Oral radiographic periodontal parameters. J Periodontol 2013:
Sci 2001: 109: 375–379. 84: 1809–1817.

37
Rotstein

153. Ray HA, Trope M. Periapical status of endodontically trea- 171. Salehrabi R, Rotstein I. Epidemiologic evaluation of the
ted teeth in relation to the technical quality of the root fill- outcomes of orthograde endodontic retreatment. J Endod
ing and the coronal restoration. Int Endod J 1995: 28: 12– 2010: 36: 790–792.
18. 172. Sato T, Yamaki K, Ishida N, Hashimoto K, Takeuchi Y,
154. Ray JM, Triplett RG. What is the role of biofilms in severe Shoji M, Sato E, Matsuyama J, Shimauchi H, Takahashi N.
head and neck infections. Oral Maxillofac Surg Clin North Cultivable anaerobic microbiota of infected root canals.
Am 2011: 23: 497–505. Int J Dent 2012: 609689: 1–5.
155. Rayner MG, Zhang Y, Gorry MC, Chen Y, Post JC, Ehrlich 173. Saunders WP, Saunders EM. Assessment of leakage in the
GD. Evidence of bacterial metabolic activity in culture- restored pulp chamber of endodontically treated multi-
negative otitis media with effusion. J Am Med Assoc 1998: rooted teeth. Int Endod J 1990: 23: 28–33.
279: 296–299. 174. Saunders WP, Saunders EM. Coronal leakage as a cause of
156. Rider CA, Rupkalvis R, Miller AS, Chen SY. Search for evi- failure in root-canal therapy: a review. Endod Dent Trau-
dence of three viral agents in radicular (periapical) cysts matol 1994: 10: 105–108.
with immunohistochemistry. Oral Surg Oral Med Oral 175. Schroeder HE, Scherle WF. Cemento-enamel junction-
Pathol Oral Radiol Endod 1995: 80: 87–91. revisited. J Periodontal Res 1988: 23: 53–59.
157. Roane JB, Benenati FW. Successful management of a per- 176. Seltzer S, Bender IB, Nazimov H, Sinai I. Pulpitis-induced
forated mandibular molar using amalgam and hydroxyla- interradicular periodontal changes in experimental ani-
patite. J Endod 1987: 13: 400–404. mals. J Periodontol 1967: 38: 124–129.
158. Ro^ cßas IN, Siqueira JF Jr. Characterization of microbiota of 177. Seltzer S, Bender IB, Ziontz M. The interrelationship of
root canal-treated teeth with post treatment disease. J Clin pulp and periodontal disease. Oral Surg Oral Med Oral
Microbiol 2012: 50: 1721–1724. Pathol 1963: 16: 1474–1490.
159. Ro^ cßas IN, Siqueira JF Jr, Santos KR, Coelho AM. “Red com- 178. Seltzer S, Soltanoff W, Bender IB. Epithelial proliferation
plex” (Bacteroides forsythus, Porphyromonas gingivalis, in periapical lesions. Oral Surg Oral Med Oral Pathol 1969:
and Treponema denticola) in endodontic infections: a 27: 111–121.
molecular approach. Oral Surg Oral Med Oral Pathol Oral 179. Sen BH, Piskin B, Demirci T. Observation of bacteria and
Radiol Endod 2001: 91: 468–471. fungi in infected root canals and dentinal tubules by SEM.
160. Ross IF, Thompson RH Jr. A long term study of root reten- Endod Dent Traumatol 1995: 11: 6–9.
tion in the treatment of maxillary molars with furcation 180. Shear M. Hyaline and granular bodies in dental cysts. Br
involvment. J Periodontol 1978: 49: 238–244. Dent J 1961: 110: 301–307.
161. Rotstein I, Friedman S, Mor C, Katznelson J, Sommer M, 181. Silver GK, Simon JH. Charcot-Leyden crystals within a
Bab I. Histological characterization of bleaching-induced periapical lesion. J Endod 2000: 26: 679–681.
external root resorption in dogs. J Endod 1991: 17: 436– 182. Simon JH. Incidence of periapical cysts in relation to the
441. root canal. J Endod 1980: 6: 845–848.
162. Rotstein I, Salehrabi R, Forrest JL. Endodontic treatment 183. Simon JHS. Root extrusion-rationale and techniques. Dent
outcome: survey of oral health care professionals. J Endod Clin North Am 1984: 28: 909–921.
2006: 32: 399–403. 184. Simon JH, Dogan H, Ceresa LM, Silver GK. The radicular
163. Rotstein I, Simon JHS. The endo-perio lesion: a critical groove: its potential clinical significance. J Endod 2000: 26:
appraisal of the disease condition. Endod Topics 2006: 13: 295–298.
34–56. 185. Simon JHS, Glick DH, Frank AL. The relationship of
164. Rotstein I, Torek Y, Lewinstein I. Effect of bleaching time endodontic-periodontic lesions. J Periodontol 1972: 43:
and temperature on the radicular penetration of hydrogen 202–208.
peroxide. Endod Dent Traumatol 1991: 7: 196–198. 186. Simon JHS, Hemple PL, Rotstein I, Salter PK. The possible
165. Rotstein I, Torek Y, Misgav R. Effect of cementum defects role of L-form bacteria in periapical disease. Endodontol-
on radicular penetration of 30% H2O2 during intracoronal ogy 1999: 11: 40–45.
bleaching. J Endod 1991: 17: 230–233. 187. Siqueira JF Jr, Alves FR, Ro ^ cßas IN. Pyrosequencing analysis
166. Rubach WC, Mitchell DF. Periodontal disease, accessory of the apical root canal microbiota. J Endod 2011: 37:
canals and pulp pathosis. J Periodontol 1965: 36: 1499–1503.
34–38. 188. Siqueira JF Jr, Ro ^ cßas IN. Diversity of endodontic micro-
167. Rupf S, Kannengiesser S, Merte K, Pfister W, Sigusch B, biota revisited. J Dent Res 2009: 88: 969–981.
Eschrich K. Comparison of profiles of key periodontal 189. Siqueira JF Jr, Ro^ cßas IN, Lopes HP, Elias CN, de Uzeda M.
pathogens in periodontium and endodontium. Endod Fungal infection of the radicular dentin. J Endod 2002: 28:
Dent Traumatol 2000: 16: 269–275. 770–773.
168. Sabeti M, Simon JH, Nowzari H, Slots J. Cytomegalovirus 190. Siqueira JF Jr, Ro ^cßas IN, Souto R, de Uzeda M, Colombo
and Epstein-Barr virus active infection in periapical AP. Checkerboard DNA-DNA hybridization analysis of
lesions of teeth with intact crowns. J Endod 2003: 29: 321– endodontic infections. Oral Surg Oral Med Oral Pathol
323. Oral Radiol Endod 2000: 89: 744–748.
169. Sabeti M, Slots J. Herpesviral-bacterial coinfection in peri- 191. Siqueira JF Jr, Sen BH. Fungi in endodontic infections.
apical pathosis. J Endod 2004: 30: 69–72. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2004:
170. Salehrabi R, Rotstein I. Endodontic treatment outcomes in 97: 632–641.
a large patient population in the USA: an epidemiological 192. Siren EK, Haapasalo MP, Ranta K, Salmi P, Kerosuo EN.
study. J Endod 2004: 30: 846–850. Microbiological findings and clinical treatment procedures

38
Interaction between endodontics and periodontics

in endodontic cases selected for microbiological investiga- 207. Trope M, Blanco L, Chivian N, Sigurdsson A. The role of
tion. Int Endod J 1997: 30: 91–95. endodontics after dental traumatic injuries. In: Cohen S,
193. Slots J, Rams TE, Listgarten MA. Yeasts, enteric rods and Hargreaves KM, editors. Pathways of the pulp. St. Louis:
pseudomonads in the subgingival flora of severe adult Mosby/Elsevier, 2006: 610–649.
periodontitis. Oral Microbiol Immunol 1988: 3: 47–52. 208. Trope M, Tronstad L, Rosenberg ES, Listgarten M. Dark-
194. Stein JM, Said Yekta S, Kleines M, Ok D, Kasaj A, Reichert field microscopy as a diagnostic aid in differentiating exu-
S, Schulz S, Scheithauer S. Failure to detect an association dates from endodontic and periodontal abscesses. J Endod
between aggressive periodontitis and the prevalence of 1988: 14: 35–38.
herpesviruses. J Clin Periodontol 2013: 40: 1–7. 209. Trott JR, Chebib F, Galindo Y. Factors related to choles-
195. Stevens BH, Levine RA. Forced eruption: a multidisci- terol formation in cysts and granulomas. J Can Dent Assoc
plinary approach for form, function, and biologic pre- 1973: 39: 550–555.
dictability. Compend Contin Educ Dent 1998: 19: 994–1010. 210. Valderhaug J. Histologic study of experimentally produced
196. Stoodley P, Sauer K, Davies DG, Costerton JW. Biofilms as intraoral odontogenic fistulae in monkeys. Int J Oral Surg
complex differentiated communities. Ann Rev Microbiol 1973: 2: 54–61.
2002: 56: 187–209. 211. Waltimo TM, Siren EK, Torkko HL, Olsen I, Haapasalo MP.
197. Sundqvist G. Ecology of the root canal flora. J Endod 1992: Fungi in therapy-resistant apical periodontitis. Int Endod J
18: 427–430. 1997: 30: 96–101.
198. Sundqvist G, Figdor D, Persson S, Sjogren U. Microbiologic 212. Wang J, Jiang Y, Chen W, Zhu C, Liang J. Bacterial flora
analysis of teeth with failed endodontic treatment and the and extraradicular biofilm associated with the apical seg-
outcome of conservative re-treatment. Oral Surg Oral Med ment of teeth with post-treatment apical periodontitis. J
Oral Pathol Oral Radiol Endod 1998: 85: 86–93. Endod 2012: 38: 954–959.
199. Tagger E, Tagger M, Sarnat H. Russell bodies in the pulp of 213. Wedenberg C, Lindskog S. Experimental internal resorp-
a primary tooth. Oral Surg Oral Med Oral Pathol Oral tion in monkey teeth. Endod Dent Traumatol 1985: 1: 221–
Radiol Endod 2000: 90: 365–368. 227.
200. Tanabe K, Takahashi K, Maeda M, Kimur I. Formation of 214. Wedenberg C, Zetterqvist L. Internal resorption in human
Charcot-Leyden crystals by human basophils in sputum teeth- a histological, scanning electron microscopic, and
and peripheral blood. Acta Med Okayama 1993: 47: 85–90. enzyme histochemical study. J Endod 1987: 13: 255–259.
201. Thomasini RL, Bonon SH, Durante P, Costa SC. Correla- 215. Weller PF, Bach D, Austen KF. Human eosinophil
tion of cytomegaloviru and human herpesvirus 7 with lysophospholipase: the sole protein component of Char-
CD3+ and CD3+ CD4+ cells in chronic periodontitis cot-Leyden crystals. J Immunol 1982: 128: 1346–1349.
patients. J Periodontol Res 2012: 47: 114–120. 216. White SN, Yu Z, Tom JF, Sangsurasak S. In vivo microleak-
202. Torabinejad M, Johnson JD. Procedural accidents. In: age of luting cements for cast crowns. J Prosthet Dent
Torabinejad M, Walton RE, Fouad AF, editors. Endodon- 1994: 71: 333–338.
tics. Principles and practice. St. Louis: Elsevier, 2015: 338– 217. Wilcox LR, Diaz-Arnold A. Coronal microleakage of perma-
354. nent lingual access restorations in endodontically treated
203. Torabinejad M, Kiger RD. A histologic evaluation of dental anterior teeth. J Endod 1989: 15: 584–587.
pulp tissue of a patient with periodontal disease. Oral Surg 218. Wilson MI, Hall J. Incidence of yeasts in root canals. J Br
Oral Med Oral Pathol 1985: 59: 198–200. Endod Soc 1968: 2: 56–59.
204. Tronstad L. Root resorption–etiology, terminology and 219. Wong R, Hirsch RS, Clarke NG. Endodontic effects of root
clinical manifestations. Endod Dent Traumatol 1988: 4: planing in humans. Endod Dent Traumatol 1989: 5: 193–
241–252. 196.
205. Tronstad L, Barnett F, Riso K, Slots J. Extraradicular 220. Yusuf H. The significance of the presence of foreign mate-
endodontic infections. Endod Dent Traumatol 1987: 3: 86– rial periapically as a cause of failure of root treatment. Oral
90. Surg Oral Med Oral Pathol 1982: 54: 566–574.
206. Trope M. Endodontic considerations in dental trauma. In: 221. Zachrisson BU, Jacobsen I. Long-term prognosis of 66 per-
Ingle JI, Bakland LK, Baumgartner JC, editors. Ingle’s manent anterior teeth with root fracture. Scand J Dent Res
endodontics. Ontario: BC Decker, 2008: 1330–1357. 1975: 83: 345–354.

39

You might also like