You are on page 1of 15

Diabetic wound healing in soft and hard oral

tissues

KANG I. KO, ANTON SCULEAN, and DANA T. GRAVES


PHILADELPHIA; AND BERN, SWITZERLAND

There is significant interest in understanding the cellular mechanisms responsible for


expedited healing response in various oral tissues and how they are impacted by
systemic diseases. Depending upon the types of oral tissue, wound healing may
occur by predominantly re-eptihelialization, by re-epithelialization with substantial
new connective tissue formation, or by a a combination of both plus new bone for-
mation. As a result, the cells involved differ and are impacted by systemic diaseses
in various ways. Diabetes mellitus is a prevalent metabolic disorder that impairs bar-
rier function and healing responses throughout the human body. In the oral cavity,
diabetes is a known risk factor for exacerbated periodontal disease and delayed
wound healing, which includes both soft and hard tissue components. Here, we
review the mechanisms of diabetic oral wound healing, particularly on impaired
keratinocyte proliferation and migration, altered level of inflammation, and
reduced formation of new connective tissue and bone. In particular, diabetes inhib-
its the expression of mitogenic growth factors whereas that of pro-inflammatory
cytokines is elevated through epigenetic mechanisms. Moreover, hyperglycemia
and oxidative stress induced by diabetes prevents the expansion of mesengenic
cells that are involved in both soft and hard tissue oral wounds. A better understand-
ing of how diabetes influences the healing processes is crucial for the prevention
and treatment of diabetes-associated oral complications. (Translational Research
2021; 236:72 86)

INTRODUCTION keratinized oral mucosa such as attached gingiva.1 The


The oral cavity contains a complex mixture of tissue attached gingiva contains dense connective tissue and
types and cells. Mucosal surfaces that cover muscle is found on the palate and masticatory gingiva that sur-
and/or small amounts of loose connective tissue rounds dentition over alveolar bone. Healing of oral
include the soft palate, nasopharynx, and buccal mucosa covering loose connective tissue and muscle is
mucosa that extends through the floor of the mouth to distinct from healing of attached gingiva that covers
the tongue (Fig 1). These oral tissues are non-kerati- alveolar bone. Epithelial coverage of excisional
nized, contain large amounts of elastin, and harbor epi- wounds in the former is rapid and does not depend
thelium that expresses different keratins compared to upon stromal healing, while for the latter, epithelial
closure coincides with substantial new connective tis-
sue formation, which occurs more slowly. In addition,
From the Department of Periodontics, School of Dental Medicine,
wounds that cover muscle and loose connective tissue
University of Pennsylvania, Philadelphia, 19104; Department of
Periodontology, School of Dental Medicine, University of Bern, heal by epithelial coverage plus contraction while those
Freiburgstrasse 7, CH-3010, Bern, Switzerland. that cover bone lack a significant contraction compo-
Submitted for Publication March 29, 2021; received submitted May nent.2 The oral mucosal epithelium is thicker than the
6, 2021; accepted for publication May 6, 2021. epidermis in skin.3 Unlike skin wounds, oral mucosal
Reprint requests: Dana T. Graves, Department of Periodontics, wounds lack dermal appendages such as hair follicles,
School of Dental Medicine, University of Pennsylvania, Philadel- sweat glands and sebacious glands. This is significant
phia, 19104 e-mail: dtgraves@upenn.edu.
since the appendages contain stem cells that promote
1931-5244/$ - see front matter
the healing process, which may be particularly impor-
Ó 2021 Elsevier Inc. All rights reserved.
tant in healing of connective tissue as described below.
https://doi.org/10.1016/j.trsl.2021.05.001

72
Translational Research
Volume 236 KO et al 73

Fig 1. Re-epithelialization in diabetic oral wounds. Left, location of different types of oral mucosa. Right,
graphical diagram of diabetic impact on epithelialization in oral wounds. Diabetes causes hyperglycemia and
production of advanced glycation end-products (AGEs) and reactive oxygen species (ROS). This in turn
increases neutrophil recruitment and redues microbial diversity. Moreover, salivary content for pro-healing fac-
tors (EGF and histatin) is reduced, whereas those that cause destruction (MMP2 and MMP9) are enhanced.
Another mechanism is FOXO1-mediated overexpression of MMP9 by keratinocytes. The overall outcome is
persistent inflammation and an environment that is conducive for keratinocyte apoptosis, coupled with reduced
keratinocyte proliferation and migration.

RE-EPITHELIALIZATION IN ORAL WOUNDS nonkeratinized oral mucosal compared to similar der-


Re-epithelialization of nonkeratinized tissue is faster mal wounds.1 In addition to growth factors, saliva con-
in oral wounds than in corresponding dermal wounds, tains histatins, which are antimicrobial peptides and
whereas re-epithelialization in keratinized attached have been proposed to promote keratinocyte migration
gingiva is not necessarily faster. Upon wounding, epi- in oral wounds.8 There is some evidence that the bio-
thelial keratinocytes at the wound edge proliferate and logically active site of histatins that stimulates kerati-
migrate to cover the wound surface. It has been pro- nocyte migration is distinct from the antimicrobial
posed that non-keratinized oral mucosa has a higher domain. It should be noted however, that saliva appears
proliferation rate than skin epidermis, which may to influence large mucosal wounds to a greater extent
account for the more rapid re-epithelialization.4 In than small mucosal wounds.9 Moreover, saliva alone is
addition, wounds in nonkeratinized tissue appear to unlikely to account for the differences in rates of heal-
have fewer leukocytes such as neutrophils and mast ing between skin and nonkeratinized oral mucosal
cells that may lead to less inflammation and more rapid wounds given that there are several intrinsic differen-
healing.5 Growth factors that induce keratinocyte ces in the keratinocytes from oral mucosa and skin.10,11
migration are produced in the early phases of healing, Acute inflammation is necessary to initiate the
including epidermal growth factor (EGF), heparin- recruitment of cells to the wound site and to induce the
binding EGF-like growth factor, transforming growth expression of factors that lead to repair. Although tran-
factor-b (TGF-b), and fibroblast growth factor sient inflammation is needed for normal healing to
(FGF).2,6 It has been proposed that saliva contains occur, prolonged inflammation limits the repair pro-
growth factors that promote oral wound healing. In a cess. When oral tongue wounds were compared to skin
tongue mucosal wound healing model, removal of sub- wounds, the expression of pro-inflammatory MMPs,
mandibular salivary glands reduced the salivary EGF peptidases, cytokines and chemokines was less in oral
content and impaired healing.6,7 Saliva also contains wounds, which may reflect differences intrinsic to cells
other biologically active growth factors including in oral mucosa.12 However, the temporal sequence of
nerve growth factor, TGF-b, insulin-like growth fac- factors induced by wounding was similar in mucosal
tor-1 and FGF.6 Furthermore, oral mucosal keratino- and dermal wounds.12 Reduced cytokine and chemo-
cytes upregulate host response genes more quickly and kine expression may facilitate rapid healing in non-ker-
to a greater extent than dermal keratinocytes, which atinized mucosal wounds compared to dermal wounds.
may contribute to the more rapid healing of An early event in wound healing is the development of
a hypoxic environment due to disruption of blood flow.
Translational Research
74 KO et al October 2021

Mucosal wounds produce less hypoxia inducible fac- MMP-9 and knockdown of the transcription factor,
tor-alpha than similar skin wounds.13 These results FOXO122. Interestingly, MMP-2 and MMP-9 are ele-
suggest that the level of hypoxia in mucosal wounds is vated in the saliva of diabetic patients,23 which may
less than that in skin and that the response of oral impair re-epithelialization.
mucosal wounds of nonkeratinized tissue is milder. Diabetes delays both the early and late stages of oral
DiPietro and colleagues have suggested that the differ- wound healing.24 Diabetic wounds have increased
ential responses to hypoxia may contribute to differen- inflammation as noted by greater numbers of neutro-
ces in rates of healing.13 phils.24,25 Re-epithelialization of gingival wounds is
enhanced by TNF inhibitors in diabetic animals but not
in normal animals, indicating that the increased levels
of TNF in the diabetic oral wounds is problematic
THE IMPACT OF DIABETES ON RE-EPITHELIALIZATION whereas the normal level of TNF does not inhibit the
OF ORAL WOUNDS healing process.26 In skin wounds, diabetes reduces the
Type 1 diabetes mellitus (T1DM) is caused by a loss of conversion from a M1 macrophage phenotype to a M2
beta cells in the pancreas, most frequently due to autoim- macrophage phenotype, which may contribute to pro-
mune etiology, and results in an insufficient amount of longed inflammation.27 Diabetic skin wounds also
insulin production. Type 2 diabetes mellitus (T2DM) is have a greater senescence-associated secretory pheno-
due to insulin resistance, caused by an inadequate cellular type (SASP) with increased inflammatory cytokine
response to normal levels of insulin signaling, combined expression (eg IL-1a, IL-6, IL-8, and CXCL2) and
with beta cell failure to provide a compensatory increase MMP expression.28 Moreover, aged mice have a simi-
in insulin.14 Oral mucosal wounds heal more slowly in lar increase in the SASP phenotype as diabetic mice,
diabetic animals compared to normoglycemic controls in suggesting that there may be similar mechanisms that
both T1DM and T2DM models.15 Contributing factors delay healing with both aging and diabetes.28
have consistently been shown reflect the direct effect of Mucosal wounds must deal with the influence of
high glucose levels, as well as indirect effects caused by bacteria since the oral cavity has a higher microbial
increased inflammation, high levels of advanced glyca- load than skin. Bacterial colonization impairs healing
tion endproducts (AGEs), and increased formation of by inducing prolonged inflammation, which intereferes
reactive oxygen species (ROS).16,15 These factors inhibit with transition to the next phase of healing, matura-
migration of oral and dermal keratinocytes in vitro.17,30 tion.29 Diabetic skin wounds have an impaired host
Oral keratinocytes in diabetic wounds in vivo have response to bacteria and an increased level of ROS that
increased levels of inflammatory cytokines such as TNF reduces microbial diversity and increases bacterial col-
and IL-1b and reduced levels of growth factors such as onization.30 Bacteria can also have direct effects on
FGF-2 and TGF-b.15 The combination of reduced growth keratinocytes by stimulating apoptosis, reducing
factor expression by keratinocytes and increased expres- migration and decreasing proliferation.31 Bacteria such
sion of inflammatory mediators may negatively impact as P. gingivalis and F. nucleatum can invade oral kera-
keratinocyte migration. tinocytes to interfere with re-epithelialization by down
Saliva has several constituents that are thought to regulating genes that promote proliferation and integ-
directly promote re-epithelialization, and diabetes has rins needed for cell migration.31 The characteristics of
been shown to alter saliva in a way that may be detri- diabetic oral wound healing by re-epithelilization are
mental to re-epithelialization. Diabetes reduces EGF summarized in Fig 1.
levels in the saliva of humans and mice,18,19 which
may contribute to reduced oral wound healing. This
conclusion is supported by evidence that supplementa-
tion with EGF in the drinking water improves healing RE-EPITHELIALIZATION, DIABETES AND FOXO1
of diabetic oral wounds but has little effect on healing As discussed above, diabetes negatively impacts kerati-
of non-diabetic oral wounds.6 Furthermore, diabetic nocyte proliferation and migration to interfere with re-
children have reduced levels of salivary antimicrobial epithelialization.32 Diabetic conditions impair keratino-
peptides including histatins.20 The reduced levels of cyte function, which include high glucose levels,
histatins may negatively impact keratinocyte migration increased formation of AGEs and ROS. Elevated ROS
and lessen the effectiveness of the antimicrobial levels have been shown to interfere with mucosal heal-
defense in diabetic individuals. In the skin, reduced ing33 and increase inflammation in gingival epithelium.34
migration is linked to high levels of MMP-9 in diabetic The epithelium of diabetic individuals has greater expres-
humans and animal models.21 In vitro, keratinocyte sion of RAGE, which increases inflammation and wor-
migration in high glucose is rescued by antibody to sens healing responses.35,36 A unifying explanation that
Translational Research
Volume 236 KO et al 75

links high glucose, AGEs and ROS formation to diabetes- gingiva has a denser connective tissue compartment,
impaired re-epithelialization is the transcription factor which contains fibroblasts, leukocytes, endothelial and
FOXO1. Under normal conditions, FOXO1 improves re- other cells. Upon wounding, the coordinated healing
epithelialization by protecting keratinocytes from oxida- response often results in complete regeneration of
tive stress and by inducing expression of TGF-b, both of injured gingiva, which contrasts with skin wounds that
which are needed for an appropriate healing response.37 heal by fibrosis.40-42 Fibroblasts are the principal cell
In diabetic conditions, high glucose or AGEs cause an type of connective tissues and are largely responsible
alteration in FOXO1 binding to the promoter regions of for stromal healing by producing extracellular matrix
specific genes, most likely due to epigenetic modifica- proteins such as collagen and proteoglycans. It has
tions, that result in a diminished capacity to protect been proposed that the unique regenerative response of
against oxidative stress and induce TGF b1 transcription gingival wounds is attributed to the intrinsic properties
is significantly reduced.32 These molecular changes are of gingival fibroblasts that are distinct from skin fibro-
observed in vivo and in vitro and in both skin and oral blasts. Cultured gingival fibroblasts are more prolifer-
mucosal keratinocytes. On a molecular level, diabetic ative and express reduced levels of TGF-b1, a pro-
conditions interfere with FOXO1 induced transcription fibrogenic cytokine, compared to skin fibroblasts.43
by reducing FOXO1 binding to consensus response ele- Importantly, gingival wounds have higher fibroblast
ments on gene promoters such as TGF b1 and tissue density and increased connective tissue fill compared
inhibitor of matrix metalloproteinases 1. In contrast, high to skin wounds,42 which is associated with reduced lev-
glucose and AGEs increase FOXO1-promoter interac- els of TGF-b1 and elevated levels of anti-fibrogenic
tions leading to overexpression of factors that reduce re- TGF-b3 in oral wounds.44-47 In a similar vein, fetal
epithelialization. More specifically, in a microenviron- wounds that heal without scarring also exhibit low lev-
ment with high glucose/AGE levels, FOXO1 induces els of TGF-b1 and high TGF-b3 levels.47,48 Moreover,
expression of MMP9, serpin peptidase inhibitor clade B it is proposed that fibroblasts may down regulate
member 2, chemokine CCL20 and IL-36g.17,22,32 Inter- immune responses in gingival wounds to facilitate res-
estingly, the negative effect of high glucose on keratino- olution of inflammation and promote healing.11,42 Ani-
cyte migration is improved by addition of specific mal studies have shown that gingival fibroblasts are
inhibitors for MMP9, or CCL20 or IL-36g or by reducing derived from neural crest origin (Wnt1-lineage) and
FOXO1 activity.22 The role of FOXO1 in inducing exces- can differentiate into tri-lineage mesenchymal cells in
sive production of these factors is demonstrated in vivo vitro,49 which contrasts with mesoderm-derived skin
by lineage specific FOXO1 deletion in keratinocytes that fibroblasts.49,50 Gingival fibroblasts cultured ex vivo
reduces high levels of MMP9, serpin peptidase inhibitor are known as gingival mesenchymal stromal cells
clade B member 2, CCL20 and IL-36g expression.22 It (GMSCs). GMSCs exhibit a pro-healing capacity by
should be noted that while high levels of these factors their anti-inflammatory properties rather than by
interfere with keratinocyte re-epithelialization, their total directly differentiating into mesengenic cells.51 55 It is
absence is also problematic. For example, the total speculated that gingival fibroblasts may exhibit similar
absence of MMP9 is detrimental to keratinocyte migra- immunomodulatory function in vivo, which remains to
tion,38 as is absence of IL-36 g.39 Thus, impaired re-epi- be proven. In vitro culture conditions drastically shift
thelialization in diabetes is due to overexpression of the fibroblast transcriptome.56,57 Thus the phenotypic
factors that normally promote an appropriate healing and functional similarities of gingival fibroblasts and
response at physiological concentrations. The expression GMSCs may not necessarily align in the context of
of FOXO1 provides a mechanistic explanation for how wound healing in vivo. It has been shown however
diabetic conditions can lead to overexpression of these that gingival fibroblasts express toll-like receptors
factors as shown in Fig 1. and can secrete pro-inflammatory cytokines such as
IL-6 and IL-8 when exposed to bacterial products in
vitro and in vivo.58-61 Therefore, gingival fibro-
CONNECTIVE TISSUE HEALING IN GINGIVAL blasts, in addition to their traditional role in produc-
WOUNDS ing extracellular matrix, may exert polarizing
As discussed above, keratinized mucosa (e.g. effects on inflammation depending on physiological
attached gingiva) covers the hard palate and alveolar or pathologic condition.
bone, and its healing differs from that of nonkerati- Inflammation in gingival and mucosal wounds is
nized mucosa by relying on substantial connective tis- more transient than in skin wounds.11,42 Initial inflam-
sue healing. Table I summarizes the findings from mation is characterized by neutrophil infiltration which
studies that investigate specific mucosal sites. Attached is necessary to limit bacterial invasion into the wound
Translational Research
76 KO et al October 2021

Table I. Wound healing characteristics in oral mucosa over loose connective tissue or bone
Mucosal healing over loose connective tissue
Mucosal type (model) Healing characteristics Reference
2
Buccal mucosa (mouse) Reduced collagen deposition and TGFb1 compared to skin healing
11
Buccal mucosa (human) Increased epithelial migration and proliferation, rapid resolution of
inflammation
63
Buccal mucosa (human) Increased NETosis and anti-microbial activity
5
Pharyngeal mucosa (human) Fewer numbers of immune cells in healthy and healed mucosa com-
pared to skin
7,10,13
Tongue (mouse) Enhanced mucosal healing by salivary EGF, and reduced apoptosis
and hypoxic response compared to skin wounds
45
Tongue (mouse) Reduced inflammation
74
Tongue (mouse) Reduced angiogenesis versus skin wounds
Mucosal healing over bone
Mucosal type (model) Healing characteristics Reference
42
Palatal gingiva (pig) Reduced inflammation, contraction and fibroblast density, improved
healing compared to skin wounds
54
Palatal gingiva (mouse) Reduced inflammation and improved healing by GMSC application
78
Palatal gingiva (mouse) Keratinocyte-derived VEGF controls angiogenesis in oral wounds
49,50
Palatal/marginal gingivae Presence of neural-crest-derived gingival MSCs
(mouse)
61
Marginal gingiva (human) Expression of IL-6 and IL-8 by gingival fibroblasts
66
Marginal gingiva (human, DEL-1 mediated resolution of inflammation in periodontitis
mouse)
69
Marginal gingiva (mouse) Homeostatic defense against masticatory stress by IL-17 driven
mechanism
70
Marginal gingiva (mouse) Protection by gamma-delta T cell-derived amphiregulin in periodon-
titis model

site via secretion of proinflammatory cytokines, phago- to excessive angiogenesis of skin wounds that is
cytosis and NETosis.62,63 Clearance of neutrophils accompanied by inflammation. Inflammatory signals
occurs via apoptosis,64 which may be targeted by bac- such as IL-1b and TNF promote neovasculariza-
teria to prolong their lifespan and inflammation.65 tion75,76 and are linked to aberrant angiogenesis in
Another mechanism of neutrophil clearance is DEL-1- rheumatoid arthritis.77 Moreover, oral keratinocytes
mediated efferocytosis by macrophages, which in turn express reduced levels of VEGF compared to skin ker-
reprograms macrophages toward a pro-healing pheno- atinocytes,74 which may be advantageous. VEGF
type to facilitate gingival regeneration.66 Furthermore, expression by the oral keratinocytes is FOXO1-depen-
lipid mediators such as resolvins and lipoxins have dent, and its deletion impairs angiogenesis and wound
shown to have a pro-resolving effect in gingival inflam- healing parameters in gingiva.78
mation from rabbit models.67,68 Lymphocytes have
also been shown to play an important role in shaping
immunity in gingival injury. In mice, induction of
mechanical damage to gingiva, either by mastication or DIABETIC IMPACT ON CELLS INVOLVED IN
external abrasion, stimulates gingival epithelial cells to CONNECTIVE TISSUE HEALING
secrete IL-6, which in turn leads to expansion of Th17 Diabetes negatively affects multiple cell types that
cells to produce defensins and neutrophil chemoattrac- participate in connective tissue wound healing of
tants.69 Moreover, gingival gd T cells have been shown mucosal surfaces as summarized in Table II. In both
to produce amphiregulin to enhance wound healing in T1DM and T2DM mouse models, new connective tis-
a periodontitis model.70 It is well established that unre- sue fill in palatal gingival wounds is reduced compared
solved inflammation results in impaired wound healing to normoglycemic control groups.79 This is attributed
and scarring.71,72 Therefore, transient inflammation to reduced fibroblast numbers in healing wounds and
may partially contribute to rapid gingival wound heal- downregulation of pro-collagen I and III expression.80
ing. Mechanistically, high glucose levels promote apoptosis
Oral wounds have reduced angiogenic response and reduce proliferation of gingival fibroblasts,79,81
when compared to skin wounds,73,74 which may be due which is due to the impact of greater oxidative stress,
Translational Research
Volume 236 KO et al 77

Table II. Diabetes-induced alterations in wound healing


Cell or Tissue Effect of Diabetes Reference
33,34
Epithelium Elevated ROS
35
Epithelium Increased expression of receptor for AGE
22
Epithelium Alteration in downstream genes induced by FOXO1
17,78
Epithelium Reduced growth factor expression
15,17
Epithelium Increased cytokine expression
17,79
Epithelium Reduced re-epithelialization
6,20,23
Saliva Reduced growth factor and increased inflammatory proteins
79
Connective tissue Increased fibroblast apoptosis
15,79
Connective tissue Increased cytokine expression and inflammation
102
Connective tissue Reduce growth factor expression
79
Connective tissue Reduced fibroblast proliferation
79
Connective tissue Increased FOXO1 activity
35
Connective tissue Increased expression of receptor for AGE
78
Connective tissue Reduced angiogenesis
78
Connective tissue Reduced production of connective tissue matrix
81
Gingiva Increased inflammation and ROS
111,136
Alveolar bone Increased bone resorption and osteoclasts
111,132,134,144-146
Alveolar bone Reduced bone formation
129,147
Alveolar bone Increased inflammation
153,154,165
Alveolar bone Increased osteonecrosis
123,124
Mandibular bone Reduced bone formation
123
Mandibular bone Increased postoperative infection risk

excessive activation of FOXO1 and high levels of cas- region of TNF and MCP-1 to promote persistent
pase-3 expression.80,82 High glucose levels also cause inflammation through over-activation of NF-kB.91,92
abnormal expression of MMP-1 from human gingival Moreover, diabetes and a high glucose environment
fibroblasts in vitro.83,84 Moreover, AGEs upregulate impair phagocytic activity of macrophages that is nec-
proinflammatory cytokine expression such as IL-6 and essary for clearance of apoptotic bodies and bacte-
IL-8 in human gingival fibroblasts.83,84 Human GMSCs ria.93,94 Diabetes has also shown to prime neutrophils
treatment improves wound healing in diabetic murine to undergo NETosis and cause aberrant inflammation
models,85,86 but it has been shown that GMSCs isolated in mice and humans,95,96 which also may potentiate
from diabetic patients exhibit reduced proliferative gingival inflammation.97 Furthermore, metabolic
capacity compared to those from nondiabetic changes in obesity and T2DM have been linked to
patients.87 Similarly, bone marrow-derived MSCs iso- reduced production of lipid mediators that are impor-
lated from T2DM patients exhibit reduced immuno- tant for resolving inflammation in diabetic wounds.98
modulatory capacity in suppressing T cell Overall, the impact of diabetes on multiple immune
proliferation.88 Therefore, diabetes affects gingival cell types is detrimental for wound healing by interfer-
fibroblasts by reducing their numbers through ing with their function and promoting persistent inflam-
enhanced apoptosis, increases catabolic events in mation. This subsequently sets the stage for sub-
wounds through upregulation of MMP-1 and down- optimal connective tissue wound healing.
regulation of TIMP-1, and increases expression of Diabetes can have an opposite effects on angiogene-
inflammatory cytokines, which altogether lead to sub- sis depending on various anatomic locations. In dia-
optimal connective tissue fill in gingival wounds. betic retinopathy and nephropathy, angiogenesis is
Diabetes impairs immune cell function and promotes excessive and subsequently leads to vascular edema
inflammation to delay gingival wound healing. It is that is linked to organ damage.99,100 In diabetic skin
well established that diabetic wounds present with per- wounds and chronic foot ulcers, angiogenesis is
sistent inflammation,81,89 particularly with high levels reduced compared to normoglycemic animals or sub-
of pro-inflammatory cytokine TNF. Inhibition of TNF jects, which is associated with delayed healing and
can reverse diabetes-induced apoptosis of fibroblasts reduced antimicrobial activity.99,101 In buccal mucosal
and mesenchymal stem cells in skin and osseous wounds of diabetic rats, new blood vessels are more
wounds.26,90 A major source of TNF is macrophage/ permeable, and the numbers are significantly reduced
monocyte population, and high glucose has been compared to normoglycemic wounds.15,102 It has been
shown to cause epigenetic changes in the promoter shown that TNF inhibition can reverse diminshed new
Translational Research
78 KO et al October 2021

and animal dermal wounds demonstrate that VEGF is


produced by keratinocytes during the healing process.
The transcription factor FOXO1 activates wound heal-
ing responses in keratinocytes including the production
of TGF-b1 to promote connective tissue healing in oral
wounds.17 FOXO1 ablation in keratinocytes diminishes
TGF-b1 expression, which reduces CTGF expression by
cells in connective tissue.105 In type 1 diabetic oral
wounds, the production of TGF-b1 is reduced. This may
be explained by substantially reduced binding of
FOXO1 to the TGF-b1 promoter caused by high levels
of glucose or AGEs, which reduce TGF-b1 transcrip-
tional activity.17 Lineage specific ablation of FOXO1
reduces VEGF-A expression in keratinocytes in mucosal
wounds and delays wound closure and with reduced
Fig 2. Connective tissue healing in diabetic oral wounds. Gingiva is angiogenesis.77,107
composed of a thick layer of lamina propria overlying bone. Diabe-
tes-mediated hyperglycemia and ROS are the major driving force for
directly inducing fibroblast apoptosis and inhibiting proliferation.
Moreover, fibroblasts express enhanced levels of pro-inflammatory
MOLECULAR MECHANISMS INVOLVED IN IMPAIRED
cytokines IL-6, IL-8 and MMP1 in diabetic gingival wounds. Diabe-
tes also interferes with a phagocytic function of macrophages and DIABETIC BONE HEALING
their polarization from pro-inflammatory (M1) to pro-resolving (M2) Dental and periodontal problems are exacerbated by
type. Diabetes-induced epigenetic changes may prevent resolution of
diabetes,109 often resulting in tooth extraction, bone aug-
inflammation. The promoter region of TGFb1 in keratinocytes are
methylated by high glucose whereas that of TNF in macrophages are mentation and restoration with dental implants. These
acetylated, resulting in reduced anti-inflammatory TGFb1 expression treatment modalities require an appropriate osseous
and over-expression of pro-inflammatory TNF. The overall outcome healing reponse that involves concerted activity of
is persistent inflammation, which reduces stromal healing outcome. osteoclasts, osteoblasts and mesenchymal stem cells
(MSCs). It is well established that diabetes has a detri-
mental impact on bone formation by affecting these cell
blood vessel formation in diabetic osseous wounds by types. Diabetes uncouples bone remodeling by increas-
restoring VEGF expression,103 providing a link between ing osteoclast numbers and inhibiting expansion of peri-
persistent inflammation and reduced angiogenesis. odontal fibroblasts and osteoblasts in an experimental
Because reduced angiogenesis in gingival wounds can periodontitis model.110,111 This is linked to a negative
lead to delayed healing,78 diabetes-inhibited neovascu- effect of high glucose on the proliferation and osteo-
larization may represent an important pathogenic mech- genic differentiation of progenitor cells.112 The molecu-
anism for altered gingival wound healing. Taken as a lar mechanism by which diabetes affects osteoblast
whole, it is apparent that diabetes dysregulates the progenitors and osteoblasts has been investigated exten-
expression of a number of different factors to impair sively in orthopedic injury models. The early phase of
the wound healing process in various cell types as diabetic bone healing is characterized by impaired oste-
shown in Fig 2. oid matrix production and reduced cellularity, which
may be caused by defective MSC recruitment, prolifera-
tion and impaired osteogenic differentiation.113,114 Dia-
betes further suppresses mRNA expression of the Dlx5
KERATINOCYTES AND CONNECTIVE TISSUE HEALING and Runx-2 transcription factors, which are responsible
Crosstalk between keratinocytes and fibroblasts is for the expression of osteoblastic phenotype.115 It also
needed for normal connective tissue healing.104 Kerati- suggests that the low rate of bone formation observed in
nocytes produce growth factors such as TGF-b, CTGF diabetic animals may be related to deficiencies in differ-
and VEGFA to stimulate connective tissue heal- entiation of MSCs to osteoprogenitor cells and in the
ing.17,105-107 TGF-b can directly and indirectly stimulate osteoblastic cell maturation.114 Studies in fracture heal-
connective tissue formation; the latter is mediated by ing support this hypothesis as diabetes results in
connective tissue growth factor (CTGF), also known as impaired bone formation.116 Moreover, diabetes leads to
cellular communication network factor-2.105 Applica- aberrant activation of NF-kB and enhances inflamma-
tion of CTGF in vivo stimulates fibroblast proliferation tion due in part to the loss of immunomodulatory
and deposition of collagen.108 Studies examining human MSCs.117 In turn, persistent inflammation characterized
Translational Research
Volume 236 KO et al 79

by high levels of TNF reduces MSC expansion through with local application of enamel matrix derivative that
upregulation of FOXO190 and suppression of IHH118 to contains a milleu of growth factors.133,134 A recent pro-
ultimately impair healing in diabetic fractures. In the spective study in human subjects showed that treatment
oral cavity, animal studies have recently identified of periodontal bony defects with enamel matrix deriva-
genetically traceable MSC populations by a-SMA, Axin2 tive, which contains a mixture of undefined growth fac-
and Lepr expression,119-121 which reside in the periodon- tors, in diabetic patients, can achieve a regenerative
tal ligament space between the tooth surface and bone outcome that is comparable to non-diabetic patients
and participate in bone healing after injury. These ani- after 3 years.135
mal models may be crucial in understanding intraoral Diabetes has a significant impact on inflammatory
MSC and osteoprogenitor behavior necessary for bone responses. Pathways linked to innate and adaptive
healing and how diabetes alters it in vivo. immune responses are generally altered in poorly con-
Guided bone regeneration (GBR) is a surgical proce- trolled diabetics compared to healthy controls. Pro-
dure that restores deficient bone volume with the use longed inflammation is detrimental to bone formation,
bone graft. A key element is a barrier membrane that in part, because NF-kB activation in response to inflam-
separates the bone graft from the soft tissue to exclude matory stimuli blocks the expression of bone matrix pro-
soft tissue infiltration that can interfere with bone for- teins.136 Diabetes also interferes with proper immune
mation.122 The effect of experimental diabetes on GBR functions that are necessary for pathogen recognition
has been investigated by means of histology and gene and removal.137-139 These findings may explain
expression analyses. GBR can sufficiently regenerate increased risk for infectious complications following
bone even in diabetic rats, but the quantity of bone GBR treatment in patients with uncontrolled diabetes.
formed is less than that formed in normoglycemic ani- Comparable findings were also reported by other authors
mals.123 Investigation of gene expression profiling has evaluating the healing of bone defects in animals140 and
shown that poorly controlled diabetes downregulates humans.141 The regulation of osteoblasts and osteoblast
pathways related to cell division, energy production precursors is complex and involves timely expression of
and osteogenesis during the proliferative phase of intra- genes encoding pro-inflammatory cytokines (Il1a, Il1b,
membranous bone healing after bone augmentation Il-6, TNF), chemokines (Ccl20, Cxcl1, Cxcl2, Cxcl10),
treatment.124 Although detailed mechanisms responsi- chemokine receptors (Ccr2, Ccr5, Ccr6) and cell adhe-
ble for the reduced cell proliferation rates are still not sion molecules (Vcam1 and Icam1) that are needed for
completely elucidated, it has been suggested that diabe- migration and activation of these cells.142,143 Prolonged
tes may suppress the expression of growth factors inflammation disrupts the level of expression and timing
needed for osteogenesis during the early phase of bone of these factors.
healing.125,126 In support of this, diabetes reduces Taken together, the available data suggest that sup-
expression of basic FGF, an important mitogenic factor pressed differentiation, proliferation and/or bone form-
for MSCs during bone healing.127 Animal studies indi- ing capacity of osteoblastic cells during the critical
cate that experimentally induced type 1 diabetes dimin- early healing period appear to play an important role in
ishes the osteoprogenitor population in bone marrow, the pathogenic mechanism underlying poor bone for-
possibly due to increased apoptosis via oxidative stress, mation in diabetes. In particular, poorly controlled dia-
which in turn may impair osteoblastogenesis, bone for- betes appears to result in a higher and dysfunctional
mation, and bone healing.128 Experimental studies inflammatory response during the early phase of bone
have also demonstrated reduced mRNA and protein healing, which in turn may be responsible for the
expression of platelet derived growth factor, TGF b1, increased risk of infection in subjects with uncontrolled
IGF-I and VEGF in femoral fractures of diabetic rats diabetes.
during early osseous healing, which correlates with
reduced cell proliferation.125,126 Diabetes increases
TNF in the oral cavity that prolongs detrimental
inflammation. FGF-2, TGFb-1, bone morphogenetic IMPACT OF DIABETES ON TOOTH EXTRACTION
protein-2, and BMP-6 are suppressed during periodon- The healing of bone in extraction sockets following
tal bone formation, which is rescued when inflamma- tooth removal in diabetes has been evaluated in a num-
tion is reduced with a TNF inhibitor.129 ber of animal and human studies. Using a T1DM rat
The exogenous delivery of IGF-I or rhFGF-2 model, alveolar bone remodeling following tooth
enhanced the healing of calvarial and periodontal extraction revealed that the quantity and rate of alveo-
defects in mice and rats.130-132 Preclinical studies have lar bone formation was significantly lower in the dia-
demonstrated that diabetic rats exhibit reduced peri- betic group compared to controls.144 After a healing
odontal bone regeneration, which can be partly rescued period of 10 days, diabetic extraction sockets contained
Translational Research
80 KO et al October 2021

thin and scanty collagen fibers whereas control groups surgery extraction.152 There is a strong association
had thick collagen fibers that formed a pretrabecular between diabetes and the incidence of MRONJ;153-157
scaffold along the trabeculae.145 Interestingly, blood even though the exact pathogenetic mechanisms
vessel formation in the extraction sockets of diabetic remain unclear, several experimental studies have pro-
animals was comparable to that of healthy controls or posed converging pathways. Much like diabetes-medi-
insulin-treated animals. In humans with T2DM, bone ated delay in extraction socket healing,158
healing of the extraction socket was delayed during the bisphosphonate treatment delays bone formation in
early stages of healing, and in diabetic pigs, the delayed extraction sockets in rats.159 There are a number of
healing was associated with decreased osteogenic differ- potential mechanisms that may play a role including
entiation of MSCs.146 Although substantial bone forma- diabetes-reduced coupled bone formation, and the
tion still occurred in diabetic animals, there was impact of diabetes on increased osteocyte and osteo-
incomplete mineralization in the center of the socket blast apoptosis.160,161 Another possible converging
compared to that of control groups which were mechanism is altered immune system. Animal and
completely filled with newly formed bone.146 The obser- human studies have demonstrated that bisphosphonate
vations made in animals were in line with those made in treatment impairs neutrophil and macrophage chemo-
humans. Compared to nondiabetic control group, T2DM taxis and function to promote immunosuppression.162-
164
patients had delayed bone fill in the extraction sockets at Studies in mice have also demonstrated a pro-
multiple time points post surgery146 and 54.7% had inflammatory response by diabetes and zolendronate
defective healing manifested by inadequate bone forma- treatment in macrophages, which was responsible for
tion. In diabetic mice, the extraction sockets consistently MRONJ-like healing after tooth extraction.165,166 The
have a higher inflammatory profile, with more M1 mac- etiology for either disease is multifactorial and com-
rophages and TNF-a expression and less M2 macro- plex, and further studies may identify pathways that
phages and PPARg expression compared to non- can be targeted to prevent MRONJ incidents in diabetic
diabetic controls.147 patients. The impact of diabetes on intraoral bone heal-
While several studies point to insufficient bone for- ing as discussed above is summarized in Fig 3.
mation after dental extraction in diabetic patients, not
all reports reach this conclusion.148-151 For example it
has been reported that diabetics have increased neutro-
phil recruitment following tooth extraction, but bone CONCLUSIONS AND FUTURE DIRECTIONS
healing in diabetic and normal subjects were similar.149 Diabetes negatively impacts several aspects of oral
Taken as a whole, the overall assessment of healing and dermal wound healing. When directly compared,
following tooth extraction in diabetic patients is slower the effects of diabetes on both are relatively consistent.
compared to non-diabetic patients, especially in early Diabetes alters the rate of healing by reducing keratino-
healing phases.151 The available evidence suggests that cyte migration, the production of growth factors by a
a) in poorly controlled T1DM, bone formation is inhib- number of different cell types including keratinocytes,
ited, resulting in delayed healing and deficient bone interferes with a number of cellular functions such as
formation; and b) tooth extractions can be performed cellular proliferation and differentiation and the
in well controlled diabetic patients, without major post- expression of genes that are essential for forming soft
operative complications. connective tissue and bone matrix. Some of the cellular
dysregulation that occurs may be due to alterations in
the gene targets of transcription factors such as
FOXO1 or over activation of others such as NF-kB.
DIABETES AS A COMORBIDITY FOR MRONJ These changes lead to difficulty in down regulating
In the oral cavity, osseous wounds such as extraction inflammation, increasing oxidative stress and enhanc-
socket or exogenous bone grafts eventually heal even ing apoptosis. In the skin these changes may lead to
under diabetic condition, albeit at a delayed rate. How- biofilm formation in healing wounds that delays the
ever, major differences exist in bone healing for nondi- repair process and in the mouth, a biofilm may form on
abetic and diabetic patients that suffer from surgically exposed bone that prevents healing. In oral
medication-related osteonecrosis of the jaw (MRONJ). wounds that require substantial connective tissue heal-
MRONJ may be localized and easily treated or may ing, diabetes exerts its deterimental impact on multiple
become widespread and develop into a serious clinical cell types such as gingival fibroblasts, endothelial cells
condition characterized by progressive destruction of and leukocytes through persistent inflammation and
the jawbones. It occurs in patients on anti-resorptive aberrant cell apoptosis. Impaired healing by diabetes
medications that have undergone oral or periodontal can be rescued by controlling inflammation or by
Translational Research
Volume 236 KO et al 81

Fig 3. Diabetic impact on various cases of intraoral bone healing. In the oral cavity, bone healing takes place
under different scenarios such as, left to right, periodontal bone regeneration, guided bone regeneration, healing
after dental extraction, or pathological healing such as MRONJ. Diabetes significantly delays bone healing in
each of these processes. In periodontal bone healing, diabetes enhances osteoclastogenesis while reducing the
number of osteoblasts and periodontal fibroblasts. This is accompanied by persistent inflammation and reduced
growth factor expression. In guided bone regeneration, which restores the hard tissue volume necessary for den-
tal implant placement, diabetes has shown to inhibit genes associated with cell division and osteogenesis. In
exodontia, diabetes delays socket healing through elevation of pro-inflammatory cytokines such as TNF, which
may indirectly suppress proper expansion of osteogenic stem cells. Diabetes is a significant co-morbidity for
MRONJ, and although exact pathogenesis is unclear, experimental studies have shown enhanced apoptosis, per-
sistent inflammation and improper recruitment of leukocytes as a converging mechanism.

genetic inhibition of FOXO1 in experimental models, Despite these limitations, animal studies have contrib-
thus targetting these processes and/or transcription fac- uted significantly to understanding some detrimental
tors with monoclonal antibody or small molecule effects of diabetes on oral wound healing, and clinical
inhibitor may be a viable method to treat diabetic oral efforts to supplement growth factors to improve heal-
wounds in humans. Similarly, diabetic intraoral bone ing process are active underway. Future studies may
healing is characterized by reduced expression of test a translational value of recombinant growth factor
growth factors that are necessary for bone formation, therapy, anti-inflammatory drugs and/or small mole-
therefore the use of recombinant growth factors is an cule inhibitors of transcription factors that are impli-
attractive therapeutic option for diabetic patients. cated in impaired diabetic healing.
Given the difficulty in resolving inflammation in dia-
betic patients, an optimal treatment may include a ther-
apeutic approach that facilitates resolution of
inflammation and delivers a growth factor to diabetic
wounds. ACKNOWLEDGMENTS
A mechanistic understanding of oral wound healing Conflict of interest: All authors have read the jour-
is largely based on animal studies. These studies are nal’s authorship policy on disclosure of potential con-
advantageous by targetting specific genes or factors flict interest.
that may be implicated in different stages of the healing All authors have read the journal’s authorship agree-
process. However, they may not fully translate to ment and the manuscript has been reviewed by and
humans as diabetes itself is a multifactorial disease that approved by all listed authors.
is often accompanied by other metabolic conditions. This study was supported by NIH grants to K.I.K.
Moreover, certain oral diseases in humans such as (K08-DE027129, R01-DE030415) and D.T.G.
MRONJ may not be fully replicated in mouse models. (R01DE019108).
Translational Research
82 KO et al October 2021

REFERENCES 21. Muller M, Trocme C, Lardy B, et al. Matrix metalloproteinases


and diabetic foot ulcers: the ratio of MMP-1 to TIMP-1 is a pre-
1. Glim JE, van Egmond M, Niessen FB, Everts V, Beelen RH. dictor of wound healing. Diabet Med 2008;25:419–26.
Detrimental dermal wound healing: what can we learn from the 22. Zhang C, Lim J, Jeon HH, et al. FOXO1 deletion in keratino-
oral mucosa? Wound Repair Regen 2013;21:648–60. cytes improves diabetic wound healing through MMP9 regula-
2. Angelov N, Moutsopoulos N, Jeong MJ, et al. Aberrant muco- tion. Sci Rep 2017;7:10565.
sal wound repair in the absence of secretory leukocyte protease 23. Kuehl MN, Rodriguez H, Burkhardt BR, Alman AC. Tumor
inhibitor. Thromb Haemost 2004;92:288–97. necrosis factor-alpha, matrix-metalloproteinases 8 and 9
3. Winning TA, Townsend GC. Oral mucosal embryology and levels in the saliva are associated with increased hemoglo-
histology. Clin Dermatol 2000;18:499–511. bin A1c in Type 1 diabetes subjects. PLoS One 2015;10:
4. Sculean A, Gruber R, Bosshardt DD. Soft tissue wound healing e0125320.
around teeth and dental implants. J Clin Periodontol 2014;41 24. Alzoubi F, Joseph B, Andersson L. Healing of soft tissue lacera-
(Suppl 15):S6–22. tions in diabetic-induced rats. Dent Traumatol 2017;33:438–43.
5. Glim JE, Beelen RH, Niessen FB, Everts V, Ulrich MM. The 25. Wang Y, Graves DT. Keratinocyte function in normal and dia-
number of immune cells is lower in healthy oral mucosa com- betic wounds and modulation by FOXO1. J Diabetes Res
pared to skin and does not increase after scarring. Arch Oral 2020;2020:3714704.
Biol 2015;60:272–81. 26. Siqueira MF, Li J, Chehab L, et al. Impaired wound healing in
6. Nagy A, Nagashima H, Cha S, et al. Reduced oral wound heal- mouse models of diabetes is mediated by TNF-alpha dysregula-
ing in the NOD mouse model for type 1 autoimmune diabetes tion and associated with enhanced activation of forkhead box
and its reversal by epidermal growth factor supplementation. O1 (FOXO1). Diabetologia 2010;53:378–88.
Diabetes 2001;50:2100–4. 27. Al-Mulla F, Leibovich SJ, Francis IM, Bitar MS. Impaired
7. Noguchi S, Ohba Y, Oka T. Effect of salivary epidermal growth TGF-beta signaling and a defect in resolution of inflammation
factor on wound healing of tongue in mice. Am J Physiol contribute to delayed wound healing in a female rat model of
1991;260:E620–5. type 2 diabetes. Mol Biosyst 2011;7:3006–20.
8. Torres P, Castro M, Reyes M, Torres VA. Histatins, wound 28. Wilkinson HN, Clowes C, Banyard KL, et al. Elevated local
healing, and cell migration. Oral Dis 2018;24:1150–60. senescence in diabetic wound healing is linked to pathological
9. Bodner L, Kaffe I, Cohen Z, Dayan D. Long-term effect of repair via CXCR2. J Invest Dermatol 2019;139:1171–81, e1176.
desalivation on extraction wound healing: a densitometric study 29. Guo S, Dipietro LA. Factors affecting wound healing. J Dent
in rats. Dentomaxillofac Radiol 1993;22:195–8. Res 2010;89:219–29.
10. Johnson A, Francis M, DiPietro LA. Differential apoptosis in 30. Kim JH, Ruegger PR, Lebig EG, et al. High levels of oxidative
mucosal and dermal wound healing. Adv Wound Care (New stress create a microenvironment that significantly decreases
Rochelle) 2014;3:751–61. the diversity of the microbiota in diabetic chronic wounds and
11. Iglesias-Bartolome R, Uchiyama A, Molinolo AA, et al. Tran- promotes biofilm formation. Front Cell Infect Microbiol
scriptional signature primes human oral mucosa for rapid 2020;10:259.
wound healing. Sci Transl Med 2018;10(451):eaap8798. 31. Bhattacharya R, Xu F, Dong G, et al. Effect of bacteria on the
https://doi.org/10.1126/scitranslmed.aap8798. wound healing behavior of oral epithelial cells. PLoS One
12. Chen L, Arbieva ZH, Guo S, et al. Positional differences in the 2014;9:e89475.
wound transcriptome of skin and oral mucosa. BMC Genomics 32. Zhang C, Ponugoti B, Tian C, et al. FOXO1 differentially regu-
2010;11:471. lates both normal and diabetic wound healing. J Cell Biol
13. Chen L, Gajendrareddy PK, DiPietro LA. Differential expres- 2015;209:289–303.
sion of HIF-1alpha in skin and mucosal wounds. J Dent Res 33. Coskun S, Gulec EG, Balabanli B, Acarturk F. Effects of epi-
2012;91:871–6. dermal growth factor on lipid peroxidation and nitric oxide lev-
14. American Diabetes. A. Diagnosis and classification of diabetes els in oral mucosal ulcer healing: a time-course study. Surg
mellitus. Diabetes Care 2013;36(Suppl 1):S67–74. Today 2007;37:570–4.
15. Brizeno LA, Assreuy AM, Alves AP, et al. Delayed healing of 34. Huang X, Yang X, Ni J, et al. Hyperglucose contributes to peri-
oral mucosa in a diabetic rat model: Implication of TNF-alpha, odontitis: involvement of the NLRP3 pathway by engaging the
IL-1beta and FGF-2. Life Sci 2016;155:36–47. innate immunity of oral gingival epithelium. J Periodontol
16. Lan CC, Liu IH, Fang AH, Wen CH, Wu CS. Hyperglycaemic 2015;86:327–35.
conditions decrease cultured keratinocyte mobility: implica- 35. Katz J, Bhattacharyya I, Farkhondeh-Kish F, et al. Expression
tions for impaired wound healing in patients with diabetes. Br J of the receptor of advanced glycation end products in gingival
Dermatol 2008;159:1103–15. tissues of type 2 diabetes patients with chronic periodontal dis-
17. Xu F, Othman B, Lim J, et al. Foxo1 inhibits diabetic mucosal ease: a study utilizing immunohistochemistry and RT-PCR. J
wound healing but enhances healing of normoglycemic Clin Periodontol 2005;32:40–4.
wounds. Diabetes 2015;64:243–56. 36. Shaikh-Kader A, Houreld NN, Rajendran NK, Abrahamse H.
18. Oxford GE, Tayari L, Barfoot MD, et al. Salivary EGF levels The link between advanced glycation end products and apopto-
reduced in diabetic patients. J Diabetes Complications sis in delayed wound healing. Cell Biochem Funct
2000;14:140–5. 2019;37:432–42.
19. Robinson CP, Cornelius J, Bounous DE, et al. Characterization 37. Ponugoti B, Xu F, Zhang C, et al. FOXO1 promotes wound
of the changing lymphocyte populations and cytokine expres- healing through the up-regulation of TGF-beta1 and prevention
sion in the exocrine tissues of autoimmune NOD mice. Autoim- of oxidative stress. J Cell Biol 2013;203:327–43.
munity 1998;27:29–44. 38. Bullard KM, Lund L, Mudgett JS, et al. Impaired wound con-
20. Cabras T, Pisano E, Mastinu A, et al. Alterations of the salivary traction in stromelysin-1-deficient mice. Ann Surg
secretory peptidome profile in children affected by type 1 dia- 1999;230:260–5.
betes. Mol Cell Proteomics 2010;9:2099–108.
Translational Research
Volume 236 KO et al 83

39. Jiang Z, Liu Y, Li C, et al. IL-36gamma induced by the TLR3- 57. Kim T, Echeagaray OH, Wang BJ, et al. In situ transcriptome
SLUG-VDR axis promotes wound healing via REG3A. J Invest characteristics are lost following culture adaptation of adult
Dermatol 2017;137:2620–9. cardiac stem cells. Sci Rep 2018;8:12060.
40. Griffin MF, desJardins-Park HE, Mascharak S, Borrelli MR, 58. Bautista-Hernandez LA, Gomez-Olivares JL, Buentello-
Longaker MT. Understanding the impact of fibroblast heteroge- Volante B, Lucio Bautista-de, Fibroblasts VM. The Unknown
neity on skin fibrosis. Dis Model Mech 2020;13(6): Sentinels Eliciting Immune Responses Against Microorgan-
dmm044164. https://doi.org/10.1242/dmm.044164. isms. Eur J Microbiol Immunol (Bp) 2017;7:151–7.
41. desJardins-Park HE, Mascharak S, Chinta MS, Wan DC, Long- 59. Bartold PM, Haynes DR. Interleukin-6 production by human
aker MT. The spectrum of scarring in craniofacial wound gingival fibroblasts. J Periodontal Res 1991;26:339–45.
repair. Front Physiol 2019;10:322. 60. Herath TD, Darveau RP, Seneviratne CJ, et al. Tetra- and
42. Mak K, Manji A, Gallant-Behm C, et al. Scarless healing of penta-acylated lipid A structures of Porphyromonas gingivalis
oral mucosa is characterized by faster resolution of inflamma- LPS differentially activate TLR4-mediated NF-kappaB signal
tion and control of myofibroblast action compared to skin transduction cascade and immuno-inflammatory response in
wounds in the red Duroc pig model. J Dermatol Sci human gingival fibroblasts. PLoS One 2013;8:e58496.
2009;56:168–80. 61. Dongari-Bagtzoglou AI, Ebersole JL. Increased presence of
43. Mah W, Jiang G, Olver D, et al. Human gingival fibroblasts dis- interleukin-6 (IL-6) and IL-8 secreting fibroblast subpopula-
play a non-fibrotic phenotype distinct from skin fibroblasts in tions in adult periodontitis. J Periodontol 1998;69:899–910.
three-dimensional cultures. PLoS One 2014;9:e90715. 62. Phillipson M, Kubes P. The healing power of neutrophils.
44. Schrementi ME, Ferreira AM, Zender C, DiPietro LA. Site-spe- Trends Immunol 2019;40:635–47.
cific production of TGF-beta in oral mucosal and cutaneous 63. Mohanty T, Sjogren J, Kahn F, et al. A novel mechanism for
wounds. Wound Repair Regen 2008;16:80–6. NETosis provides antimicrobial defense at the oral mucosa.
45. Szpaderska AM, Zuckerman JD, DiPietro LA. Differential Blood 2015;126:2128–37.
injury responses in oral mucosal and cutaneous wounds. J Dent 64. de Oliveira S, Rosowski EE, Huttenlocher A. Neutrophil migra-
Res 2003;82:621–6. tion in infection and wound repair: going forward in reverse.
46. Eslami A, Gallant-Behm CL, Hart DA, et al. Expression of Nat Rev Immunol 2016;16:378–91.
integrin alphavbeta6 and TGF-beta in scarless vs scar-forming 65. Murray DA, Wilton JM. Lipopolysaccharide from the peri-
wound healing. J Histochem Cytochem 2009;57:543–57. odontal pathogen Porphyromonas gingivalis prevents apoptosis
47. Occleston NL, O'Kane S, Laverty HG, et al. Discovery and of HL60-derived neutrophils in vitro. Infect Immun
development of avotermin (recombinant human transforming 2003;71:7232–5.
growth factor beta 3): a new class of prophylactic therapeutic 66. Kourtzelis I, Li X, Mitroulis I, et al. DEL-1 promotes macro-
for the improvement of scarring. Wound Repair Regen 2011;19 phage efferocytosis and clearance of inflammation. Nat Immu-
(Suppl 1):s38–48. nol 2019;20:40–9.
48. Lichtman MK, Otero-Vinas M, Falanga V. Transforming 67. Serhan CN, Jain A, Marleau S, et al. Reduced inflammation and
growth factor beta (TGF-beta) isoforms in wound healing and tissue damage in transgenic rabbits overexpressing 15-lipoxy-
fibrosis. Wound Repair Regen 2016;24:215–22. genase and endogenous anti-inflammatory lipid mediators. J
49. Xu X, Chen C, Akiyama K, et al. Gingivae contain neural- Immunol 2003;171:6856–65.
crest- and mesoderm-derived mesenchymal stem cells. J Dent 68. Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation:
Res 2013;92:825–32. dual anti-inflammatory and pro-resolution lipid mediators. Nat
50. Isaac J, Nassif A, Asselin A, et al. Involvement of neural crest Rev Immunol 2008;8:349–61.
and paraxial mesoderm in oral mucosal development and heal- 69. Dutzan N, Abusleme L, Bridgeman H, et al. On-going mechan-
ing. Biomaterials 2018;172:41–53. ical damage from mastication drives homeostatic Th17 cell
51. Zhang QZ, Su WR, Shi SH, et al. Human gingiva-derived responses at the oral barrier. Immunity 2017;46:133–47.
mesenchymal stem cells elicit polarization of m2 macro- 70. Krishnan S, Prise IE, Wemyss K, et al. Amphiregulin-produc-
phages and enhance cutaneous wound healing. Stem Cells ing gammadelta T cells are vital for safeguarding oral barrier
2010;28:1856–68. immune homeostasis. Proc Natl Acad Sci U S A
52. Zhang Q, Shi S, Liu Y, et al. Mesenchymal stem cells derived 2018;115:10738–43.
from human gingiva are capable of immunomodulatory func- 71. Rockey DC, Bell PD, Hill JA. Fibrosis a common pathway to
tions and ameliorate inflammation-related tissue destruction in organ injury and failure. N Engl J Med 2015;372:1138–49.
experimental colitis. J Immunol 2009;183:7787–98. 72. Wang ZC, Zhao WY, Cao Y, et al. The roles of inflammation in
53. Gu Y, Shi S. Transplantation of gingiva-derived mesenchymal keloid and hypertrophic scars. Front Immunol 2020;11:603187.
stem cells ameliorates collagen-induced arthritis. Arthritis Res 73. DiPietro LA. Angiogenesis and wound repair: when enough is
Ther 2016;18:262. enough. J Leukoc Biol 2016;100:979–84.
54. Kou X, Xu X, Chen C, et al. The Fas/Fap-1/Cav-1 complex reg- 74. Szpaderska AM, Walsh CG, Steinberg MJ, DiPietro LA. Dis-
ulates IL-1RA secretion in mesenchymal stem cells to acceler- tinct patterns of angiogenesis in oral and skin wounds. J Dent
ate wound healing. Sci Transl Med 2018;10(432):eaai8524. Res 2005;84:309–14.
https://doi.org/10.1126/scitranslmed.aai8524. 75. Fahey E, Doyle SL. IL-1 family cytokine regulation of vascular
55. Ahangar P, Mills SJ, Smith LE, Gronthos S, Cowin AJ. Human permeability and angiogenesis. Front Immunol 2019;10:1426.
gingival fibroblast secretome accelerates wound healing 76. Sainson RC, Johnston DA, Chu HC, et al. TNF primes endothe-
through anti-inflammatory and pro-angiogenic mechanisms. lial cells for angiogenic sprouting by inducing a tip cell pheno-
NPJ Regen Med 2020;5:24. type. Blood 2008;111:4997–5007.
56. Walmsley GG, Rinkevich Y, Hu MS, et al. Live fibroblast har- 77. Elshabrawy HA, Chen Z, Volin MV, et al. The pathogenic role
vest reveals surface marker shift in vitro. Tissue Eng Part C of angiogenesis in rheumatoid arthritis. Angiogenesis
Methods 2015;21:314–21. 2015;18:433–48.
Translational Research
84 KO et al October 2021

78. Jeon HH, Yu Q, Lu Y, et al. FOXO1 regulates VEGFA expres- 96. Fadini GP, Menegazzo L, Rigato M, et al. NETosis delays dia-
sion and promotes angiogenesis in healing wounds. J Pathol betic wound healing in mice and humans. Diabetes
2018;245:258–64. 2016;65:1061–71.
79. Desta T, Li J, Chino T, Graves DT. Altered fibroblast prolifera- 97. Vitkov L, Minnich B, Knopf J, et al. NETs Are Double-Edged
tion and apoptosis in diabetic gingival wounds. J Dent Res Swords with the Potential to Aggravate or Resolve Periodontal
2010;89:609–14. Inflammation. Cells 2020;9.
80. Liu R, Desta T, He H, Graves DT. Diabetes alters the response 98. Hellmann J, Tang Y, Spite M. Proresolving lipid mediators and
to bacteria by enhancing fibroblast apoptosis. Endocrinology diabetic wound healing. Curr Opin Endocrinol Diabetes Obes
2004;145:2997–3003. 2012;19:104–8.
81. Kido D, Mizutani K, Takeda K, et al. Impact of diabetes on gin- 99. Simons M. Angiogenesis, arteriogenesis, and diabetes: para-
gival wound healing via oxidative stress. PLoS One 2017;12: digm reassessed? J Am Coll Cardiol 2005;46:835–7.
e0189601. 100. Cooper ME, Vranes D, Youssef S, et al. Increased renal expres-
82. Buranasin P, Mizutani K, Iwasaki K, et al. High glucose- sion of vascular endothelial growth factor (VEGF) and its
induced oxidative stress impairs proliferation and migration of receptor VEGFR-2 in experimental diabetes. Diabetes
human gingival fibroblasts. PLoS One 2018;13:e0201855. 1999;48:2229–39.
83. Irwin CR, Myrillas TT, Traynor P, Leadbetter N, Cawston TE. 101. Veith AP, Henderson K, Spencer A, Sligar AD, Baker AB.
The role of soluble interleukin (IL)-6 receptor in mediating the Therapeutic strategies for enhancing angiogenesis in wound
effects of IL-6 on matrix metalloproteinase-1 and tissue inhibi- healing. Adv Drug Deliv Rev 2019;146:97–125.
tor of metalloproteinase-1 expression by gingival fibroblasts. J 102. Yamano S, Kuo WP, Sukotjo C. Downregulated gene expres-
Periodontol 2002;73:741–7. sion of TGF-betas in diabetic oral wound healing. J Craniomax-
84. Lew JH, Naruishi K, Kajiura Y, et al. High glucose-mediated illofac Surg 2013;41:e42–8.
cytokine regulation in gingival fibroblasts and THP-1 macro- 103. Lim JC, Ko KI, Mattos M, et al. TNFalpha contributes to diabe-
phage: a possible mechanism of severe periodontitis with dia- tes impaired angiogenesis in fracture healing. Bone
betes. Cell Physiol Biochem 2018;50:973–86. 2017;99:26–38.
85. Shi Q, Qian Z, Liu D, et al. GMSC-derived exosomes com- 104. Werner S, Krieg T, Smola H. Keratinocyte-fibroblast inter-
bined with a chitosan/silk hydrogel sponge accelerates wound actions in wound healing. J Invest Dermatol 2007;127:998–
healing in a diabetic rat skin defect model. Front Physiol 1008.
2017;8:904. 105. Zhang C, Lim J, Liu J, et al. FOXO1 expression in keratino-
86. Zhang W, Zhou L, Dang J, et al. Human gingiva-derived mes- cytes promotes connective tissue healing. Sci Rep
enchymal stem cells ameliorate streptozoticin-induced T1DM 2017;7:42834.
in mice via Suppression of T effector cells and up-regulating 106. Shephard P, Martin G, Smola-Hess S, et al. Myofibroblast dif-
treg subsets. Sci Rep 2017;7:15249. ferentiation is induced in keratinocyte-fibroblast co-cultures
87. Assem M, Kamal S, Sabry D, Soliman N, Aly RM. Preclinical and is antagonistically regulated by endogenous transforming
assessment of the proliferation capacity of gingival and peri- growth factor-beta and interleukin-1. Am J Pathol
odontal ligament stem cells from diabetic patients. Open 2004;164:2055–66.
Access Maced J Med Sci 2018;6:254–9. 107. Johnson KE, Wilgus TA. Vascular endothelial growth factor
88. Kizilay Mancini O, Shum-Tim D, Stochaj U, Correa JA, Col- and angiogenesis in the regulation of cutaneous wound repair.
megna I. Age, atherosclerosis and type 2 diabetes reduce Adv Wound Care (New Rochelle) 2014;3:647–61.
human mesenchymal stromal cell-mediated T-cell suppression. 108. Alfaro MP, Deskins DL, Wallus M, et al. A physiological role
Stem Cell Res Ther 2015;6:140. for connective tissue growth factor in early wound healing. Lab
89. Graves DT, Liu R, Alikhani M, Al-Mashat H, Trackman PC. Invest 2013;93:81–95.
Diabetes-enhanced inflammation and apoptosis impact on 109. Taylor GW, Manz MC, Borgnakke WS. Diabetes, periodontal
periodontal pathology. J Dent Res 2006;85:15–21. diseases, dental caries, and tooth loss: a review of the literature.
90. Ko KI, Coimbra LS, Tian C, et al. Diabetes reduces mesenchy- Compend Contin Educ Dent 2004;25:179–84, 186-178, 190;
mal stem cells in fracture healing through a TNFalpha-medi- quiz 192.
ated mechanism. Diabetologia 2015;58:633 642. 110. Andriankaja OM, Galicia J, Dong G, et al. Gene expression
91. Miao F, Gonzalo IG, Lanting L, Natarajan R. In vivo chromatin dynamics during diabetic periodontitis. J Dent Res 2012;91:
remodeling events leading to inflammatory gene transcription 1160–5.
under diabetic conditions. J Biol Chem 2004;279:18091–7. 111. Liu R, Bal HS, Desta T, et al. Diabetes enhances periodontal
92. El-Osta A, Brasacchio D, Yao D, et al. Transient high glucose bone loss through enhanced resorption and diminished bone
causes persistent epigenetic changes and altered gene expres- formation. J Dent Res 2006;85:510–4.
sion during subsequent normoglycemia. J Exp Med 112. Kim HS, Park JW, Yeo SI, Choi BJ, Suh JY. Effects of high
2008;205:2409–17. glucose on cellular activity of periodontal ligament cells in
93. Pavlou S, Lindsay J, Ingram R, Xu H, Chen M. Sustained high vitro. Diabetes Res Clin Pract 2006;74:41–7.
glucose exposure sensitizes macrophage responses to cytokine 113. Weiss RE, Reddi AH. Influence of experimental diabetes and
stimuli but reduces their phagocytic activity. BMC Immunol insulin on matrix-induced cartilage and bone differentiation.
2018;19:24. Am J Physiol 1980;238:E200–7.
94. Lecube A, Pachon G, Petriz J, Hernandez C, Simo R. Phagocytic 114. McCabe LR. Understanding the pathology and mechanisms of
activity is impaired in type 2 diabetes mellitus and increases after type I diabetic bone loss. J Cell Biochem 2007;102:1343–57.
metabolic improvement. PLoS One 2011;6:e23366. 115. Lu H, Kraut D, Gerstenfeld LC, Graves DT. Diabetes interferes
95. Wong SL, Demers M, Martinod K, et al. Diabetes primes neu- with the bone formation by affecting the expression of tran-
trophils to undergo NETosis, which impairs wound healing. scription factors that regulate osteoblast differentiation. Endo-
Nat Med 2015;21:815–9. crinology 2003;144:346–52.
Translational Research
Volume 236 KO et al 85

116. Huang Z, Pei X, Graves DT. The interrelationship between dia- invasive surgical technique with enamel matrix derivative
betes, IL-17 and bone loss. Curr Osteoporos Rep 2020;18:23–31. under 3-year observation: A prospective cohort study. J Perio-
117. Ko KI, Syverson AL, Kralik RM, et al. Diabetes-induced NF- dontol 2020. https://doi.org/10.1002/JPER.20-0590.
kappaB dysregulation in skeletal stem cells prevents resolution 136. Pacios S, Xiao W, Mattos M, et al. Osteoblast lineage cells play
of inflammation. Diabetes 2019;68:2095–106. an essential role in periodontal bone loss through activation of
118. Tevlin R, Seo EY, Marecic O, et al. Pharmacological rescue of nuclear factor-kappa B. Sci Rep 2015;5:16694.
diabetic skeletal stem cell niches. Sci Transl Med 2017;9(372): 137. Stegenga ME, van der Crabben SN, Blumer RM, et al. Hyper-
eaag2809. https://doi.org/10.1126/scitranslmed.aag2809. glycemia enhances coagulation and reduces neutrophil degran-
119. Zhang D, Zhang S, Wang J, et al. LepR-Expressing stem cells ulation, whereas hyperinsulinemia inhibits fibrinolysis during
are essential for alveolar bone regeneration. J Dent Res human endotoxemia. Blood 2008;112:82–9.
2020;99:1279–86. 138. Hair PS, Echague CG, Rohn RD, et al. Hyperglycemic condi-
120. Roguljic H, Matthews BG, Yang W, et al. In vivo identification tions inhibit C3-mediated immunologic control of Staphylococ-
of periodontal progenitor cells. J Dent Res 2013;92:709–15. cus aureus. J Transl Med 2012;10:35.
121. Yuan X, Pei X, Zhao Y, et al. A Wnt-responsive PDL popula- 139. Martinez N, Ketheesan N, Martens GW, et al. Defects in early
tion effectuates extraction socket healing. J Dent Res cell recruitment contribute to the increased susceptibility to
2018;97:803–9. respiratory Klebsiella pneumoniae infection in diabetic mice.
122. Elgali I, Omar O, Dahlin C, Thomsen P. Guided bone regenera- Microbes Infect 2016;18:649–55.
tion: materials and biological mechanisms revisited. Eur J Oral 140. Lee RSB, Hamlet SM, Ivanovski S. The influence of titanium
Sci 2017;125:315 337. surface characteristics on macrophage phenotype polarization
123. Retzepi M, Lewis MP, Donos N. Effect of diabetes and meta- during osseous healing in type I diabetic rats: a pilot study.
bolic control on de novo bone formation following guided bone Clin Oral Implants Res 2017;28:e159–68.
regeneration. Clin Oral Implants Res 2010;21:71–9. 141. Sun M, Yang J, Wang J, et al. TNF-alpha is upregulated in
124. Retzepi M, Calciolari E, Wall I, Lewis MP, Donos N. The T2DM patients with fracture and promotes the apoptosis of
effect of experimental diabetes and glycaemic control on osteoblast cells in vitro in the presence of high glucose. Cyto-
guided bone regeneration: histology and gene expression analy- kine 2016;80:35–42.
ses. Clin Oral Implants Res 2018;29:139–54. 142. Tanaka Y, Morimoto I, Nakano Y, et al. Osteoblasts are regu-
125. Tyndall WA, Beam HA, Zarro C, O'Connor JP, Lin SS. lated by the cellular adhesion through ICAM-1 and VCAM-1. J
Decreased platelet derived growth factor expression during Bone Miner Res 1995;10:1462–9.
fracture healing in diabetic animals. Clin Orthop Relat Res 143. Yellowley C. CXCL12/CXCR4 signaling and other recruitment
2003:319–30. and homing pathways in fracture repair. Bonekey Rep
126. Gandhi A, Beam HA, O'Connor JP, Parsons JR, Lin SS. The 2013;2:300.
effects of local insulin delivery on diabetic fracture healing. 144. Fang Y, Wang LP, Du FL, Liu WJ, Ren GL. Effects of insulin-
Bone 2005;37:482–90. like growth factor I on alveolar bone remodeling in diabetic
127. Kawaguchi H, Kurokawa T, Hanada K, et al. Stimulation of rats. J Periodontal Res 2013;48:144–50.
fracture repair by recombinant human basic fibroblast growth 145. Devlin H, Garland H, Sloan P. Healing of tooth extraction sock-
factor in normal and streptozotocin-diabetic rats. Endocrinol- ets in experimental diabetes mellitus. J Oral Maxillofac Surg
ogy 1994;135:774–81. 1996;54:1087–91.
128. Weinberg E, Maymon T, Moses O, Weinreb M. Streptozotocin- 146. Zhang S, Song S, Wang S, et al. Type 2 diabetes affects postex-
induced diabetes in rats diminishes the size of the osteoprogenitor traction socket healing and influences first-stage implant sur-
pool in bone marrow. Diabetes Res Clin Pract 2014;103:35–41. gery: A study based on clinical and animal evidence. Clin
129. Pacios S, Kang J, Galicia J, et al. Diabetes aggravates periodon- Implant Dent Relat Res 2019;21:436–45.
titis by limiting repair through enhanced inflammation. FASEB 147. Shen X, Shen X, Li B, et al. Abnormal macrophage polarization
J 2012;26:1423–30. impedes the healing of diabetes-associated tooth sockets. Bone
130. Thaller SR, Lee TJ, Armstrong M, Tesluk H, Stern JS. Effect of 2021;143:115618.
insulin-like growth factor type 1 on critical-size defects in dia- 148. Aronovich S, Skope LW, Kelly JP, Kyriakides TC. The rela-
betic rats. J Craniofac Surg 1995;6:218–23. tionship of glycemic control to the outcomes of dental extrac-
131. Santana RB, Trackman PC. Controlled release of fibroblast growth tions. J Oral Maxillofac Surg 2010;68:2955–61.
factor 2 stimulates bone healing in an animal model of diabetes 149. Fernandes KS, Glick M, de Souza MS, Kokron CM, Gallottini
mellitus. Int J Oral Maxillofac Implants 2006;21:711–8. M. Association between immunologic parameters, glycemic
132. Bizenjima T, Seshima F, Ishizuka Y, et al. Fibroblast growth control, and postextraction complications in patients with type
factor-2 promotes healing of surgically created periodontal 2 diabetes. J Am Dent Assoc 2015;146:592–9.
defects in rats with early, streptozotocin-induced diabetes via 150. Huang S, Dang H, Huynh W, Sambrook PJ, Goss AN. The
increasing cell proliferation and regulating angiogenesis. J Clin healing of dental extraction sockets in patients with Type 2 dia-
Periodontol 2015;42:62–71. betes on oral hypoglycaemics: a prospective cohort. Aust Dent
133. Wyganowska-Swiatkowska M, Urbaniak P, Nohawica MM, J 2013;58:89–93.
Kotwicka M, Jankun J. Enamel matrix proteins exhibit growth 151. Gadicherla S, Smriti K, Roy S, et al. Comparison of
factor activity: A review of evidence at the cellular and molecu- Extraction Socket Healing in Non-Diabetic, Prediabetic,
lar levels. Exp Ther Med 2015;9:2025–33. and Type 2 Diabetic Patients. Clin Cosmet Investig Dent
134. Takeda K, Mizutani K, Matsuura T, et al. Periodontal regenera- 2020;12:291–6.
tive effect of enamel matrix derivative in diabetes. PLoS One 152. Ruggiero SL, Dodson TB, Fantasia J, et al. American associa-
2018;13:e0207201. tion of oral and maxillofacial surgeons position paper on medi-
135. Mizutani K, Shioyama H, Matsuura T, et al. Periodontal regen- cation-related osteonecrosis of the jaw 2014 update. J Oral
erative therapy in patients with type 2 diabetes using minimally Maxillofac Surg 2014;72:1938–56.
Translational Research
86 KO et al October 2021

153. Khamaisi M, Regev E, Yarom N, et al. Possible association 160. Ikebuchi Y, Aoki S, Honma M, et al. Coupling of bone resorp-
between diabetes and bisphosphonate-related jaw osteonecro- tion and formation by RANKL reverse signalling. Nature
sis. J Clin Endocrinol Metab 2007;92:1172–5. 2018;561:195–200.
154. Molcho S, Peer A, Berg T, Futerman B, Khamaisi M. Diabetes 161. Gangoiti MV, Anbinder PS, Cortizo AM, McCarthy AD. Mor-
microvascular disease and the risk for bisphosphonate-related phological changes induced by advanced glycation endproducts
osteonecrosis of the jaw: a single center study. J Clin Endocri- in osteoblastic cells: effects of co-incubation with alendronate.
nol Metab 2013;98:E1807–12. Acta Histochem 2013;115:649–57.
155. Vidal-Real C, Perez-Sayans M, Suarez-Penaranda JM, Gandara- 162. Hoefert S, Schmitz I, Weichert F, Gaspar M, Eufinger H. Mac-
Rey JM, Garcia-Garcia A. Osteonecrosis of the jaws in 194 rophages and bisphosphonate-related osteonecrosis of the jaw
patients who have undergone intravenous bisphosphonate ther- (BRONJ): evidence of local immunosuppression of macro-
apy in Spain. Med Oral Patol Oral Cir Bucal 2015;20:e267–72. phages in contrast to other infectious jaw diseases. Clin Oral
156. Barasch A, Cunha-Cruz J, Curro FA, et al. Risk factors for Investig 2015;19:497–508.
osteonecrosis of the jaws: a case-control study from the CON- 163. Kalyan S, Chandrasekaran V, Quabius ES, Lindhorst TK,
DOR Dental PBRN. Tex Dent J 2013;130:299–307. Kabelitz D. Neutrophil uptake of nitrogen-bisphosphonates
157. Watters AL, Hansen HJ, Williams T, et al. Intravenous leads to the suppression of human peripheral blood gammadelta
bisphosphonate-related osteonecrosis of the jaw: long-term fol- T cells. Cell Mol Life Sci 2014;71:2335–46.
low-up of 109 patients. Oral Surg Oral Med Oral Pathol Oral 164. Kuiper JW, Forster C, Sun C, Peel S, Glogauer M. Zoledronate
Radiol 2013;115:192–200. and pamidronate depress neutrophil functions and survival in
158. Shyng YC, Devlin H, Sloan P. The effect of streptozotocin- mice. Br J Pharmacol 2012;165:532–9.
induced experimental diabetes mellitus on calvarial defect heal- 165. Zhang Q, Yu W, Lee S, et al. Bisphosphonate induces osteo-
ing and bone turnover in the rat. Int J Oral Maxillofac Surg necrosis of the jaw in diabetic mice via NLRP3/caspase-1-
2001;30:70–4. dependent IL-1beta mechanism. J Bone Miner Res
159. Hikita H, Miyazawa K, Tabuchi M, Kimura M, Goto S. 2015;30:2300–12.
Bisphosphonate administration prior to tooth extraction delays 166. Yang X, Xu X, Chen J, et al. Zoledronic acid regulates the syn-
initial healing of the extraction socket in rats. J Bone Miner thesis and secretion of IL-1beta through Histone methylation in
Metab 2009;27:663–72. macrophages. Cell Death Discov 2020;6:47.

You might also like