You are on page 1of 7

Regenerative Endodontics

Survival of the Apical Papilla and Its Resident


Stem Cells in a Case of Advanced Pulpal Necrosis
and Apical Periodontitis
Vanessa Chrepa, DDS, MS,*† Brandon Pitcher, DDS,† Michael A. Henry, DDS, PhD,†‡
and Anibal Diogenes, DDS, PhD†

Abstract
Introduction: Apical papilla represents a source of an Key Words
enriched mesenchymal stem cell (MSC) population Apical papilla survival, apical periodontitis, characterization, periapical inflammation,
(stem cells of the apical papilla [SCAPs]) that modulates regenerative endodontics, stem cell, stem cells of the apical papilla
root development and may participate in regenerative
endodontic procedures in immature teeth with pulp ne-
crosis. The characteristics and phenotype of this tissue in
the presence of inflammation are largely unknown. The
T he apical papilla con-
sists of the apical portion
of the dental papilla and,
Significance
The apical papilla may survive despite intense
purpose of this study was to characterize a human apical inflammatory infiltrate following pulp necrosis. In
in conjunction with the
papilla sample that was isolated from an immature this report, SCAPs maintained their stemness and
Hertwig epithelial root
tooth with pulp necrosis and apical periodontitis. expressed increased osteogenic and angiogenesis
sheath, is responsible for
Methods: Inflamed periapical tissue that included potential. Regenerative strategies should focus on
root development (1). Stem
part of the apical papilla (apical papilla clinical sample promoting the continued survival, recruitment, and
cells of the apical papilla
[CS]) was collected from an immature mandibular pre- differentiation of these cells to achieve predictable
(SCAPs) have been shown
molar previously diagnosed with pulp necrosis and api- guided endodontic repair and regeneration.
to have great proliferation
cal periodontitis during an apexification procedure. and differentiation poten-
Harvested cells from this tissue (SCAP CS) were tial in addition to high motility (2). Studies have highlighted the potential role of SCAPs
compared with inflamed periapical progenitor cells and the apical papilla in the continuation of root development and regeneration of the
(IPAPCs) and normal SCAP (SCAP-RP89) in flow cytom- pulp-dentin complex (1, 3). Notably, in a pilot experiment, surgically removing the
etry and quantitative osteogenesis experiments. Part of apical papilla resulted in the arrest of root development even in the presence of
the issue was further processed for immunohistochem- intact pulp (1). Huang et al (3) further showed that SCAPs have the ability to differen-
istry and compared with apical papilla and coronal tiate into odontoblastlike cells and lead to de novo dental pulp regeneration in vivo.
pulp sections from normal immature teeth as well as in- These findings suggest the importance of maintaining the vitality of the apical papilla
flamed periapical tissues from mature teeth. Results: in immature teeth as a source of stem cells that contribute to and regulate root devel-
Similar to SCAP-RP89, 96.6% of the SCAP CS coex- opment.
pressed the MSC markers CD73, CD90, and CD105, In regenerative endodontic procedures (REPs), evoked bleeding from the periap-
whereas only 66.3% of IPAPCs coexpressed all markers. ical tissues has been shown to lead to a significant influx of mesenchymal stem cells
The SCAP CS showed a significantly greater mineraliza- (MSCs) in the root canal system of both immature and mature teeth (4, 5). Using
tion potential than both SCAP-RP89 and IPAPCs. Finally, this step as a method to introduce stem cells into the root canals is a significant
immunohistochemical analysis revealed moderate infil- concept in regenerative endodontics because it provides access to the most readily
tration of cells expressing the inflammatory markers available sources of MSCs (ie, apical papilla, periodontal ligament, alveolar bone,
CD45/68 in the apical papilla CS and prominent CD24, and inflamed periapical tissues) for potential dental pulp regeneration (6). In imma-
CD105, and von Willebrand factor expression. Conclu- ture teeth, the apical papilla represents an enriched pool of MSCs in direct contact with
sions: Under inflammatory conditions, human apical the tooth apex (2, 7). Even with the odontogenic differentiation potential of SCAPs, REPs
papilla was found moderately inflamed with retained do not always result in the formation of dentin and pulplike tissue (8–10). The root
SCAP vitality and stemness and increased osteogenic canal microenvironment including pulp status and infection control regimens seems
and angiogenesis potential. (J Endod 2016;-:1–7) to affect the regeneration phenotype (8, 11–16). REPs in teeth with pulp necrosis
and harboring bacteria in the root canal system have been associated with

From the *Department of Endodontics, University of Washington, Seattle, Washington; †Department of Endodontics, University of Texas Health Science Center at
San Antonio, San Antonio, Texas; and ‡University of Colorado School of Dental Medicine, Aurora, Colorado.
Address requests for reprints to Dr Anibal Diogenes, Department of Endodontics, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive,
San Antonio, TX 78229. E-mail address: Diogenes@uthscsa.edu
0099-2399/$ - see front matter
Copyright ª 2016 American Association of Endodontists.
http://dx.doi.org/10.1016/j.joen.2016.09.024

JOE — Volume -, Number -, - 2016 Survival of the Apical Papilla 1


Regenerative Endodontics
cementum or bonelike tissue formation, whereas dentin formation was cedure. We hypothesized that apical papilla and its resident cells
noted in teeth with irreversible pulpitis (11, 12). Importantly, retain their vitality under chronic inflammatory conditions.
prolonged infection and periapical inflammation may seriously affect
the survival of the apical papilla or alter its properties, which is
considered crucial for the continuation of root development in Materials and Methods
immature teeth that undergo regenerative endodontic treatment (1). Case Report
Despite the importance of the latter, the effect of pulp necrosis and peri- A 9-year-old female presented to the Graduate Endodontics Clinic at
apical inflammation on the survival of human apical papilla is largely the University of Texas Health Science Center at San Antonio, San Antonio,
unknown. TX, for evaluation and treatment of tooth #28. The patient reported tooth
Studies have identified unique properties and characteristics sensitivity in the past, approximately 1 year before the appointment, pro-
of the human apical papilla and SCAPs; however, the included sam- duced by hot/cold drinks, and her complaint at the time of the visit was
ples were from human immature teeth with normal pulp and peri- sensitivity on biting. Tooth #28 presented clinically with extensive
apical tissues (2, 3, 17, 18). These studies showed the distinct occlusal caries, normal probing depths and mobility, and normal soft tis-
histologic and immunohistochemical properties of apical papilla sues. Radiographically, tooth #28 presented with a carious lesion extend-
compared with dental pulp as well as characterized SCAP cell ing into the pulp chamber, immature apex, and periapical radiolucency
lines in terms of gene expression, stemness, and differentiation (Fig. 1A). Pulp sensibility tests with Endo-Ice (Coltene/Whaledent Inc,
potential (2, 3, 17, 18). Nonetheless, it is very likely that apical Cuyahoga, OH) and an electric pulp test (Electric Pulp Tester; Sybro-
papilla and SCAPs present differently in pulp necrosis and nEndo, Glendora, CA) were performed, and negative pulp responses
periapical inflammation (1). Hypoxia and inflammation have were noted. Periradicular tests revealed that tooth #28 was sensitive to
been shown to affect SCAP proliferation and differentiation in cul- percussion but not sensitive to palpation. Based on the clinical tests,
ture (19, 20). Tobias Duarte et al (21) evaluated the histopatho- the pulpal diagnosis was determined as pulp necrosis and the periradic-
logical conditions of dental pulp and apical papilla after inducing ular diagnosis as symptomatic apical periodontitis. Both the guardian
endodontic infection in a rat model and found that apical papilla and patient were informed of the clinical findings and presented the treat-
remained vital after 90 days of infection yet slightly or moderately ment options of regenerative endodontic treatment, apexification with a
inflamed. Despite the current evidence, to date, no study has eval- mineral trioxide aggregate (MTA) plug, extraction, and no treatment. The
uated human apical papilla from an immature tooth with pulp ne- guardian and patient elected MTA apexification and signed the informed
crosis and apical periodontitis. The purpose of this study was to consent. After adequate anesthesia, the tooth was isolated with a rubber
report for the first time the characterization of a human apical dam, and access was made into the root canal system. Pulp necrosis was
papilla sample harvested through the canal system of an immature confirmed clinically upon access. For the chemomechanical prepara-
tooth with pulp necrosis being treated with an apexification pro- tion, Hedstrom files (H-files, SybronEndo) were used to file the canal

Figure 1. Case report radiograph and clinical photographs. (A) A periapical radiograph of tooth #28. (B) Periapical tissue was engaged to a H-file after removal
from the root canal of tooth #28. (C) Tissue was immediately sectioned into 2 pieces, both containing part of the apical papilla (green arrow) and part of the
inflamed periapical tissue (black arrow). The yellow lines represent the junction between the 2 distinct tissues.

2 Chrepa et al. JOE — Volume -, Number -, - 2016


Regenerative Endodontics
walls along with 6% sodium hypochlorite irrigation. During the mechan- Flow Cytometry
ical preparation, tissue from the periapical region was removed with a Single-cell suspensions of SCAP CS and IPAPCs (106 cells/mL) were
size 25 H-file placed 2 mm beyond the apex. The tissue was engaged incubated in Stain Buffer (FBS; BD Biosciences, San Jose, CA) concomi-
with the H-file and removed from the canal system while still attached tantly with fluorescent-labeled antibodies and processed for multicolor
to the file (Fig. 1B). After careful inspection with the use of 10 magni- flow cytometry analysis as previously described (4). The following anti-
fication, we confirmed that this tissue included part of the apical papilla bodies were included: PE-CY7 mouse antihuman CD73 (561258, BD Bio-
and part of the inflamed periapical tissues (Fig. 1B). With the guardian’s sciences), APC mouse antihuman CD90 (clone 5E10, BD Biosciences),
consent, this tissue, which would have otherwise been discarded, was PerCP-CY5.5 mouse antihuman CD105 (clone 266RUO, BD Biosciences),
collected immediately in cold Hank’s balanced salt solution (Invitrogen, and APC-H7 mouse antihuman CD45 (clone 2D1BD, Biosciences). Gating
Carlsbad, CA) for processing. At the end of the first appointment, was performed on CD45-negative cells and included doublet discrimina-
Ca(OH)2 was placed as the interim intracanal medication. The patient tion for single cells and scatter gate for gating out small debris. For each cell
presented asymptomatic at the second visit, and MTA apexification was line, the percentage of cells expressing each marker and the percentage of
completed. The guardian was informed that the tooth prognosis was the cells coexpressing CD73, CD90, and CD105 was recorded. Results
favorable, and subsequent follow-ups were scheduled. were compared with previously published flow cytometry data including
the same MSC markers for the cell line RP-89 (SCAP-RP89) (18).
Sample Collection and Processing
The apical papilla clinical sample (CS) was transferred immediately Alizarin Red Staining and Osteogenesis Quantification
to the laboratory where it was sectioned into 2 pieces (Fig. 1C). One piece A previously characterized SCAP cell line (SCAP-RP89), SCAP CS,
was processed for immunohistochemistry and confocal microscopy as and IPAPC cell lines (5th passage for each) were cultured in 24-well
previously described in order to detect antigens in the tissue specific plates containing either basal medium (control) or osteogenesis induc-
for stem cells and inflammatory cells (18). The other piece was used tion medium (StemPro Osteogenesis Differentiation Kit; Life Technolo-
to generate a SCAP culture of the CS and contained the apical papilla gies, Grand Island, NY) for 3 weeks (n = 6/cell line). Qualitative and
(Fig. 1C, green arrow), which was clearly distinct from its attached apical quantitative analysis of mineralization was then performed using the
tissue (Fig. 1C, black arrow). The apical papilla was carefully dissected Osteogenesis Quantitation Kit (Millipore, Darmstadt, Germany)
from the rest of the tissue with a sterile 15C blade, separating it at the level following the manufacture’s protocol. Images of the alizarin red staining
of junction between the 2 distinct tissues (Fig. 1C, yellow lines). The were taken under light microscopy.
dissected apical papilla tissue was minced, subjected to enzymatic diges-
tion, and processed for cell culture as described previously (16). Har- Immunohistochemistry and Laser Confocal Microscopy
vested cells were cultured at 37 C and 5% CO2 in basal culture Tissue slices from the apical papilla CS, apical papilla (normal pulp),
medium composed of alpha-minimum essential medium (Gibco, Grand coronal pulp (normal pulp), and inflamed periapical lesions were pro-
Island, NY) supplemented with 10% fetal bovine, L-glutamine (Gemini, cessed for immunohistochemistry as previously described (22). Immuno-
West Sacramento, CA), penicillin (100 U/mL), streptomycin (100 mg/ histochemical analysis included the primary antibodies CD105 (mouse
mL), and amphotericin B (Sigma-Aldrich, St Louis, MO). Single-cell col- antihuman, M3527, 1:500 dilution; Dako, Glostrup, Denmark), CD24
onies were transferred to 10-cm culture dishes, maintained, and cultured (mouse antihuman, 555426, 1:500 dilution; BD Biosciences), CD45
until 80% confluency. Cells in the 5th passage were used for flow cy- (mouse antihuman, M0701, 1:200 dilution; Dako), and CD68 (mouse
tometry and differentiation experiments. antihuman, M0876, 1:200 dilution; Dako) combined with the endothelial
For immunohistochemical processing, the tissue (ie, apical papilla cell marker von Willebrand factor (vWF) antibody (rabbit antihuman,
CS) was rinsed 3 times (5 minutes each) in 0.1 mol/L phosphate buffer A0082, 1:4,000 dilution; Dako). Goat antimouse Alexa Fluor 488 and
(PB) followed by fixation in 4% paraformaldehyde in 0.1 mol/L PB and goat antirabbit Alexa Fluor 568 (both antibodies at 1:100 dilution; Molec-
then stored at 20 C for 24 hours in 0.1 mol/L PB with 30% sucrose. ular Probes, Eugene, OR) were used as secondary antibodies along with
The tissue was then serially sectioned with a cryostat (30-mm sections); the nuclear stain TO-PRO-3 (1:5000 dilution, Molecular Probes). Control
sections were collected onto glass slides and stored at 20 C until ready preparations consisted of tissue slices that lacked both primary and sec-
for staining. Sections from the apical papilla CS tissue sample that was ondary antibodies and were stained only with TO-PRO-3 and tissues
processed for immunohistochemistry included part of the inflamed tis- stained with secondary antibodies but lacked primary antibodies. Immu-
sue and part of the apical papilla (Fig. 1C, black and green arrows, noreactivity was visualized with a Nikon C1 Si laser confocal imaging
respectively). To better assess the phenotype of these cells (ie, SCAP system equipped with a 90i Nikon microscope (Nikon, Melville, NY).
CS) and their source tissue (ie, apical papilla CS), additional cell lines
and tissue slides previously collected, processed, and stored in our de-
Statistical Analysis
partment’s tissue bank were included in the analysis. The cell lines
Intragroup differences in osteogenic differentiation were calculated
included were inflamed periapical progenitor cells (IPAPCs) previously
for each cell line (ie, control vs osteogenic) using a Wilcoxon signed rank
harvested from a periradicular lesion of an adult patient and a character-
test and intercomparison differences in osteogenic differentiation among
ized SCAP cell line (ie, SCAP-RP89) from a young donor (18). These cell
all cell lines with the Kruskal-Wallis test and the Tukey post hoc multiple
lines were used in the flow cytometry and differentiation experiments
comparison test. For each cell line, control groups served as a reference
along with SCAP CS. The tissue slides included apical papilla and coronal
and were set to 100%. Data were summarized as percent change in osteo-
pulp sections from normal immature teeth (n = 2) as well as biopsies of
genesis quantification relative to controls. Statistical analysis was per-
inflamed periapical tissues collected during apicoectomies in adult pa-
formed with Graph Pad Prism 5.0 (Graph Pad, La Jolla, CA) at a = 0.05.
tients and diagnosed as cysts or granulomas (n = 5) (4). These tissue
slices were processed for immunohistochemistry along with apical
papilla CS slides. The Institutional Review Board of the University of Texas Results
Health Science Center at San Antonio had approved the collection of these SCAPs isolated from a CS of apical papilla surrounded by inflamed
tissues and cell lines for previous studies (4, 16, 18). periapical tissues (SCAP CS) coexpressed CD73 and CD90 while being

JOE — Volume -, Number -, - 2016 Survival of the Apical Papilla 3


Regenerative Endodontics
negative for CD45 in >99% of all cells. Of these cells, 96.6% also ex- 97.8% of the cell population) (18). IPAPCs were found negative for
pressed the MSC marker CD105 (Fig. 2A). MSC marker expression in the marker CD45 in >99% of the cells. Of the CD45-negative cells,
SCAP CS was similar to the coexpression of the same markers in the 91.8% coexpressed CD73 and CD90, and of these cells, 66.3% also
SCAP-RP89 cell line (ie, CD73, CD90, and CD105 coexpressed in expressed the marker CD105 (Fig. 2B).

Figure 2. Scatter plots showing the expression of MSC markers in the SCAP CS and IPAPC cell lines by multicolor flow cytometry analysis. After doublet discrim-
ination, CD45-negative cells were analyzed for CD73, CD90, and CD105 using gates based on the unstained control. (A) The SCAP CS. Plots a, b, and c show single
expression of CD73, CD90, and CD105, respectively, and plot d shows coexpression of all 3 markers (96.6%). (B) IPAPCs. Plots e, f, and g show single expression
of CD73, CD90, and CD105, respectively, and plot h shows coexpression of all 3 markers for this cell line (66.3%). SSC, side scatter.

4 Chrepa et al. JOE — Volume -, Number -, - 2016


Regenerative Endodontics

Figure 3. Quantitative osteogenesis of the cell line SCAP CS, SCAP-RP89, and IPAPCs. Cells were cultured for 3 weeks in either control or osteogenic medium. (A)
Alizarin red staining shows the robust mineralization potential of all cell lines compared with their respective control. (B) Quantification of osteogenesis confirms
the significantly greater mineralization of all cell lines compared with their control (all P values <.05). SCAP CS expressed significantly greater mineralization
compared with both SCAP-RP89 and IPAPCs (P < .001). *P < .05. **P < .01. ***P < .001. n.s: not significant.

All 3 cell lines showed a robust mineralization potential when regeneration outcome. Studies have evaluated the effect of various
induced in osteogenic media compared with their controls (Fig. 3A stress microenvironments on SCAPs including hypoxia, serum
and B). SCAP CS showed a more than 3-fold increase in mineralization deprivation, and inflammation; yet, these conditions were induced in
relative to the control group (P < .001), and it was significantly greater culture (19, 20, 23). More importantly, the survival of the apical
than the mineralization found in the SCAP-RP89 and IPAPCs groups papilla after pulpal necrosis and apical periodontitis has never been
(P < .001). There was no significant difference in mineralization be- reported. Here, for the first time, we characterized a human apical
tween the SCAP-RP89 cell line and the IPAPC group (Fig. 3B). papilla sample that was isolated from an immature tooth with pulp
Confocal microscopic examination of the sections from apical necrosis and apical periodontitis. Data from this report provide
papilla CS, apical papilla (normal pulp), coronal pulp (normal evidence that apical papilla tissues are capable of surviving pulpal
pulp), and inflamed periapical tissue specimens showed specific stain- necrosis despite moderate inflammatory cell infiltration and that
ing for CD105 and vWF in all specimens (Fig. 4A–D). CD105 was endodontic infection and inflammation may alter the differentiation
colocalized with the endothelial factor vWF in vascular structures in potential of SCAPs in humans.
apical papilla (normal pulp), coronal pulp, and periapical lesions In this study, SCAPs were isolated from the clinical sample of in-
(Fig. 4B–D). In apical papilla CS, CD105 was also colocalized with flamed apical papilla (SCAP CS) and processed for flow cytometry to
vWF; however, VWF expression was distributed throughout the tissue evaluate MSC marker expression. Flow cytometry analysis showed a
specimen outside the vascular compartment (Fig. 4A). The distribution 96.6% coexpression of the MSC markers CD73, CD90, and CD105.
pattern of vWF in the apical papilla CS was shown in all stained slides, These data were very similar to the MSC expression of SCAPs under
and control preparations confirmed that the staining was specific. Cells normal conditions (18), which implies that SCAPs very likely retain their
in the apical papilla (normal pulp) and apical papilla CS expressed the viability and stemness even in an inflammatory environment. Notably, a
MSC marker CD24 prominently, whereas prominent CD24 staining was lower percentage of CD73- and CD90-positive IPAPCs (66.3%) coex-
not evident in coronal pulp or periapical lesion specimens (Fig. 4E–H). pressed the marker CD105. Because a significant influx of cells express-
Expression of the inflammatory markers CD45 and CD68 was promi- ing CD73 and CD105 is introduced in the root canals during REPs (5), it
nent in the periapical lesion specimen, moderate in the apical papilla is possible that SCAPs participate more actively in this process than
CS, and mostly absent in the apical papilla (normal pulp) and coronal IPAPCs, at least in immature teeth.
pulp (normal pulp) specimens (Fig. 4I–L). Osteogenesis quantification was further performed to assess the
differentiation potential of the SCAP CS compared with a characterized
SCAP cell line (SCAP-RP89) and IPAPCs. Notably, the SCAP CS expressed
Discussion a significantly higher differentiation potential than SCAP-RP89 and
In immature teeth, the apical papilla represents an enriched pool IPAPCs. These data agree with a previous report that short-term expo-
of stem cells that play a significant role in root development and are sure of SCAPs to proinflammatory cytokines induced greater cell prolif-
likely to participate in regenerative procedures (1, 7, 17). Studies eration and mineralization potential than the control group (19).
have thoroughly characterized the human apical papilla and its Nevertheless, they also reported that longer exposure to inflammation
residing stem cells under normal conditions (17, 18). Nevertheless, had the opposite outcomes (19). In another study, histopathological
REPs aim to achieve dental pulp regeneration in root canals under analysis of apical papilla of rat immature teeth during inflammation re-
conditions of pulp necrosis, infection, and periapical inflammation. vealed that apical papilla survived pulp infection and periapical inflam-
These conditions may alter the characteristics and phenotype of mation for at least 90 days while being moderately inflamed (21).
apical papilla and SCAPs, which may affect, among other factors, the Confocal microscopic analysis in this study showed that the apical

JOE — Volume -, Number -, - 2016 Survival of the Apical Papilla 5


Regenerative Endodontics

Figure 4. Immunohistochemistry and confocal microscopy showing the expression of the MSC markers CD24 and CD105 and the inflammatory markers CD45/
CD68 along with the endothelial vWF) in the tissues. (A, E, and I) The apical papilla CS (ie, inflamed), (B, F, and J) normal apical papilla, (C, G, and K) normal
coronal pulp, and (D, H, and L) periapical lesion. (A–D) Arrows show the coexpression of CD105 (red) and vWF (green) in all samples. vWF appears localized in
the vascular structures in all tissues except for the (A) apical papilla CS where it appears distributed throughout the tissue specimen outside the vascular compart-
ment. (E–H) CD24 (red) was prominently expressed in the (E and F) apical papilla samples, but there was no expression noted in the (G and H) coronal pulp or
periapical tissue sections. (I–L) CD45/68 expression (red) was moderate in the (I) apical papilla CS, prominent in the (L) periapical lesion, and mostly absent in
the (J and K) normal pulp tissue samples. Nuclei are identified by TO-PRO-3 staining (blue).

papilla CS expressed markers for immune cells (CD45 and CD68 immu- expression exclusively in the apical papilla CS and normal apical
nofluorescence), showing an inflammatory infiltrate into the apical papilla that was lacking in the coronal pulp and inflamed periapical
papilla. However, the expression of these immune markers was signif- tissues. Moreover, inflammatory conditions in the apical papilla CS
icantly less prominent than that seen in periapical lesions. Although the seemed to not affect the expression of this MSC marker. Another
patient reported discomfort with the tooth for approximately 1 year, it important finding from this analysis is the robust difference in the
was not possible to determine for how long the apical papilla had been distribution pattern of the endothelial factor vWF in inflamed
exposed to inflammatory conditions before treatment. This represents a apical papilla compared with all other samples. The vWF
limitation of this study. Further research evaluating the effects of factors immunofluorescent signal was widespread throughout the apical
such as the exposure time of human apical papilla and stem cells to papilla CS rather than compartmentalized within vascular structures
endodontic infection and its subsequent inflammatory response is as seen in all other tissue specimens. A possible explanation of this
warranted. finding is that certain SCAP populations may have been subjected to
In addition to assessing inflammatory markers, confocal micro- endothelial transdifferentiation under the inflammatory conditions
scopic analysis included the MSC markers CD24 and CD105 combined (23). This phenomenon has been extensively described for dental
with the endothelial marker vWF. This analysis assessed apical papilla MSCs as their response to tissue injury, inflammation, or other stress
and coronal pulp specimens from normal teeth and periapical lesion microenvironments (20, 23–26). Moreover, the apical papilla has
specimens along with the inflamed apical papilla (apical papilla CS) been previously shown to be a relatively avascular tissue (27), and
in order to assess differences in the immunophenotype among these the presence of endothelial cell markers throughout the apical papilla
tissues. The expression of CD105 in the inflamed apical papilla sample may suggest an active process of angiogenesis. This process may be trig-
as well as in normal apical papilla confirmed the flow cytometry results gered by the resident SCAPs and their release of angiogenic factors such
that SCAPs maintain MSC marker expression under inflammatory con- as vascular endothelial growth factor upon exposure to the hypoxic
ditions. Studies have suggested the possibility of MSC marker CD24 to environment of a necrotic pulp (20).
be a specific marker for SCAP because it is highly expressed in SCAP In conclusion, under inflammatory conditions, human apical
populations and not detected in other MSC lines (2, 23). Results papilla was found inflamed with retained SCAP vitality and stemness
from our immunohistochemical analysis confirmed prominent CD24 and increased osteogenic differentiation and induction of angiogenesis.

6 Chrepa et al. JOE — Volume -, Number -, - 2016


Regenerative Endodontics
Future studies are warranted to confirm these findings and further 12. Shimizu E, Ricucci D, Albert J, et al. Clinical, radiographic, and histological obser-
explore the mechanisms of these environment-mediated changes on vation of a human immature permanent tooth with chronic apical abscess after
revitalization treatment. J Endod 2013;39:1078–83.
human apical papilla and SCAPs as well as their effect on dental pulp 13. Althumairy RI, Teixeira FB, Diogenes A. Effect of dentin conditioning with intracanal
regeneration outcome. medicaments on survival of stem cells of apical papilla. J Endod 2014;40:521–5.
14. Galler KM, D’Souza RN, Federlin M, et al. Dentin conditioning codetermines cell fate
Acknowledgments in regenerative endodontics. J Endod 2011;37:1536–41.
15. Ruparel NB, Teixeira FB, Ferraz CC, Diogenes A. Direct effect of intracanal medica-
The authors deny any conflicts of interest related to this study. ments on survival of stem cells of the apical papilla. J Endod 2012;38:1372–5.
16. Trevino EG, Patwardhan AN, Henry MA, et al. Effect of irrigants on the survival of
human stem cells of the apical papilla in a platelet-rich plasma scaffold in human
References root tips. J Endod 2011;37:1109–15.
1. Huang GT, Sonoyama W, Liu Y, et al. The hidden treasure in apical papilla: the po- 17. Sonoyama W, Liu Y, Yamaza T, et al. Characterization of the apical papilla and its
tential role in pulp/dentin regeneration and bioroot engineering. J Endod 2008;34: residing stem cells from human immature permanent teeth: a pilot study.
645–51. J Endod 2008;34:166–71.
2. Sonoyama W, Liu Y, Fang D, et al. Mesenchymal stem cell-mediated functional tooth 18. Ruparel NB, de Almeida JF, Henry MA, Diogenes A. Characterization of a stem cell of
regeneration in swine. PLoS One 2006;1:e79. apical papilla cell line: effect of passage on cellular phenotype. J Endod 2013;39:
3. Huang GT, Yamaza T, Shea LD, et al. Stem/progenitor cell-mediated de novo regen- 357–63.
eration of dental pulp with newly deposited continuous layer of dentin in an in vivo 19. Liu C, Xiong H, Chen K, et al. Long-term exposure to pro-inflammatory cytokines
model. Tissue Eng Part A 2010;16:605–15. inhibits the osteogenic/dentinogenic differentiation of stem cells from the apical
4. Chrepa V, Henry MA, Daniel BJ, Diogenes A. Delivery of apical mesenchymal stem papilla. Int Endod J 2016;49:950–9.
cells into root canals of mature teeth. J Dent Res 2015;94:1653–9. 20. Vanacker J, Viswanath A, De Berdt P, et al. Hypoxia modulates the differentiation
5. Lovelace TW, Henry MA, Hargreaves KM, Diogenes A. Evaluation of the delivery of potential of stem cells of the apical papilla. J Endod 2014;40:1410–8.
mesenchymal stem cells into the root canal space of necrotic immature teeth after 21. Tobias Duarte PC, Gomes-Filho JE, Ervolino E, et al. Histopathological condition of
clinical regenerative endodontic procedure. J Endod 2011;37:133–8. the remaining tissues after endodontic infection of rat immature teeth. J Endod
6. Hargreaves KM, Diogenes A, Teixeira FB. Treatment options: biological basis of 2014;40:538–42.
regenerative endodontic procedures. J Endod 2013;39:S30–43. 22. Henry MA, Luo S, Levinson SR. Unmyelinated nerve fibers in the human dental pulp
7. Huang GT, Garcia-Godoy F. Missing concepts in de novo pulp regeneration. J Dent express markers for myelinated fibers and show sodium channel accumulations.
Res 2014;93:717–24. BMC Neurosci 2012;13:29.
8. Cao Y, Song M, Kim E, et al. Pulp-dentin regeneration: current state and future pros- 23. Bakopoulou A, Kritis A, Andreadis D, et al. Angiogenic potential and secretome of
pects. J Dent Res 2015;94:1544–51. human apical papilla mesenchymal stem cells in various stress microenvironments.
9. Wang X, Thibodeau B, Trope M, et al. Histologic characterization of regenerated Stem Cells Dev 2015;24:2496–512.
tissues in canal space after the revitalization/revascularization procedure of imma- 24. Aranha AM, Zhang Z, Neiva KG, et al. Hypoxia enhances the angiogenic potential of
ture dog teeth with apical periodontitis. J Endod 2010;36:56–63. human dental pulp cells. J Endod 2010;36:1633–7.
10. Yamauchi N, Nagaoka H, Yamauchi S, et al. Immunohistological characterization of 25. Bento LW, Zhang Z, Imai A, et al. Endothelial differentiation of SHED requires MEK1/
newly formed tissues after regenerative procedure in immature dog teeth. J Endod ERK signaling. J Dent Res 2013;92:51–7.
2011;37:1636–41. 26. Sakai VT, Zhang Z, Dong Z, et al. SHED differentiate into functional odontoblasts and
11. Shimizu E, Jong G, Partridge N, et al. Histologic observation of a human immature endothelium. J Dent Res 2010;89:791–6.
permanent tooth with irreversible pulpitis after revascularization/regeneration pro- 27. Diogenes A, Henry MA, Teixeira FB, Hargreaves KM. An update on clinical regener-
cedure. J Endod 2012;38:1293–7. ative endodontics. Endod Topics 2013;28:2–23.

JOE — Volume -, Number -, - 2016 Survival of the Apical Papilla 7

You might also like