Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Oct 1.
Published in final edited form as: Transl Res. 2021 May 13;236:72–86. doi: 10.1016/j.trsl.2021.05.001

Diabetic Wound Healing in Soft and Hard Oral Tissues

Kang I Ko 1, Anton Sculean 2, Dana T Graves 1
PMCID: PMC8554709  NIHMSID: NIHMS1704475  PMID: 33992825

Abstract

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 formation. 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 barrier 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 inhibits 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 understanding of how diabetes influences the healing processes is crucial for the prevention and treatment of diabetes-associated oral complications.

Keywords: Diabetes, Advanced Glycation End-products, Bone, High Glucose, Epithelium, Mucosa, Repair, Oral Wound Healing, Oxidative Stress, Inflammation, Connective Tissue, Fibroblast, Angiogenesis, Osteogenesis, Mesenchymal Stem Cells, Regeneration

Introduction

The oral cavity contains a complex mixture of tissue types and cells. Mucosal surfaces that cover muscle and/or small amounts of loose connective tissue include the soft palate, nasopharynx, and buccal mucosa that extends through the floor of the mouth to the tongue (Figure 1). These oral tissues are non-keratinized, contain large amounts of elastin, and harbor epithelium that express different keratins compared to keratinized oral mucosa such as attached gingiva1. The attached gingiva contains dense connective tissue and is found on the palate and masticatory gingiva that surrounds dentition over alveolar bone. Healing of oral mucosa covering loose connective tissue and muscle is distinct from healing of attached gingiva that covers alveolar bone. Epithelial coverage of excisional wounds in the former is rapid and does not depend upon stromal healing, while for the latter, epithelial closure coincides with substantial new connective tissue formation, which occurs more slowly. In addition, wounds that cover muscle and loose connective tissue heal by epithelial coverage plus contraction while those that cover bone lack a significant contraction component2. The oral mucosal epithelium is thicker than the epidermis in skin3. Unlike skin wounds, oral mucosal wounds lack dermal appendages such as hair follicles, sweat glands and sebacious glands. This is significant since the appendages contain stem cells that promote the healing process, which may be particularly important in healing of connective tissue as described below.

Figure 1. Re-epithelialization in diabetic oral wounds.

Figure 1.

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-produce (AGEs) and reactive oxygen species (ROS). This in tum increases recruitment of neutrophil recruitment and redues microbial diversity. oareover, salivary content for pro-healing factors (EGF end 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 anti migration

Re-epithelialization in oral wounds

Re-epithelialization of nonkeratinized tissue is faster in oral wounds than in corresponding dermal wounds, whereas re-epithelialization in keratinized attached gingiva is not necessarily faster. Upon wounding, epithelial keratinocytes at the wound edge proliferate and migrate to cover the wound surface. It has been proposed that non-keratinized oral mucosa has a higher proliferation rate than skin epidermis, which may account for the more rapid re-epithelialization4. In addition, wounds in nonkeratinized tissue appear to have fewer leukocytes such as neutrophils and mast cells that may lead to less inflammation and more rapid healing5. Growth factors that induce keratinocyte migration are produced in the early phases of healing, including epidermal growth factor (EGF), heparin-binding EGF-like growth factor (HB-EGF), transforming growth factor-β (TGF-β), and fibroblast growth factor (FGF)2,6. It has been proposed that saliva contains growth factors that promote oral wound healing. In a tongue mucosal wound healing model, removal of submandibular salivary glands reduced the salivary EGF content and impaired healing6,7. Saliva also contains other biologically active growth factors including nerve growth factor (NGF), TGF-β, insulin-like growth factor-1 (IGF-1) and fibroblast growth factor (FGF)6. Furthermore, oral mucosal keratinocytes upregulate host response genes more quickly and to a greater extent than dermal keratinocytes, which may contribute to the more rapid healing of nonkeratinized oral mucosal compared to similar dermal wounds1. In addition to growth factors, saliva contains histatins, which are antimicrobial peptides and have been proposed to promote keratinocyte migration in oral wounds8. There is some evidence that the biologically active site of histatins that stimulates keratinocyte migration is distinct from the antimicrobial domain. It should be noted however, that saliva appears to influence large mucosal wounds to a greater extent than small mucosal wounds9. Moreover, saliva alone is unlikely to account for the differences in rates of healing between skin and nonkeratinized oral mucosal wounds given that there are several intrinsic differences in the keratinocytes from oral mucosa and skin10,11.

Acute inflammation is necessary to initiate the recruitment of cells to the wound site and to induce the expression of factors that lead to repair. Although transient inflammation is needed for normal healing to occur, prolonged inflammation limits the repair process. When oral tongue wounds were compared to skin wounds, the expression of pro-inflammatory MMPs, peptidases, cytokines and chemokines was less in the oral wounds, which may reflect differences intrinsic to cells in oral mucosa12. However, the temporal sequence of factors induced by wounding was similar in mucosal and dermal wounds12. Reduced cytokine and chemokine expression may facilitate rapid healing in non-keratinized mucosal wounds compared to dermal wounds. An early event in wound healing is the development of a hypoxic environment due to disruption of blood flow. Mucosal wounds produce less hypoxia inducible factor-alpha (HIF-1a) than similar skin wounds13. These results suggest that the level of hypoxia in mucosal wounds is less than that in skin and that the response of oral mucosal wounds of nonkeratinized tissue is milder. DiPietro and colleagues have suggested that the differential responses to hypoxia may contribute to differences in rates of healing13.

The Impact of Diabetes on Re-Epithelialization of Oral Wounds

Type 1 diabetes mellitus (T1DM) is caused by a loss of beta cells in the pancreas, most frequently due to autoimmune etiology, and results in an insufficient amount of insulin production. Type 2 diabetes mellitus (T2DM) is due to insulin resistance, caused by an inadequate cellular response to normal levels of insulin signaling, combined with beta cell failure to provide a compensatory increase in insulin14. Oral mucosal wounds heal more slowly in diabetic animals compared to normoglycemic controls in both T1DM and T2DM models1515. Contributing factors have consistently been shown reflect the direct effect of high glucose levels, as well as indirect effects caused by increased inflammation, high levels of advanced glycation endproducts (AGEs), and increased formation of reactive oxygen species (ROS)16,15. These factors inhibit migration of oral and dermal keratinocytes in vitro17,30. Oral keratinocytes in diabetic wounds in vivo have increased levels of inflammatory cytokines such as TNF and IL-1β and reduced levels of growth factors such as FGF-2 and TGF-β15,15. The combination of reduced growth factor expression by keratinocytes and increased expression of inflammatory mediators may negatively impact keratinocyte migration.

Saliva has several constituents that are thought to directly promote re-epithelialization, and diabetes has been shown to alter saliva in a way that may be detrimental to re-epithelialization. Diabetes reduces EGF levels in the saliva of humans and mice18,19, which may contribute to reduced oral wound healing. This conclusion is supported by evidence that supplementation with EGF in the drinking water improves healing of diabetic oral wounds but has little effect on healing of non-diabetic oral wounds6. Furthermore, diabetic children have reduced levels of salivary antimicrobial peptides including histatins20. The reduced levels of histatins may negatively impact keratinocyte migration and lessen the effectiveness of the antimicrobial defense in diabetic individuals. In the skin, reduced migration is linked to high levels of MMP-9 in diabetic humans and animal models21. In vitro, keratinocyte migration in high glucose is rescued by antibody to MMP-9 and knockdown of the transcription factor, FOXO122. Interestingly, MMP-2 and MMP-9 are elevated in the saliva of diabetic patients23, which may impair re-epithelialization.

Diabetes delays both the early and late stages of oral wound healing24. Diabetic wounds have increased inflammation as noted by greater numbers of neutrophils24,25. 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 whereas the normal level of TNF does not inhibit the healing process26.In skin wounds, diabetes reduces the conversion from a M1 macrophage phenotype to a M2 macrophage phenotype, which may contribute to prolonged inflammation27. Diabetic skin wounds also have a greater senescence-associated secretory phenotype (SASP) with increased inflammatory cytokine expression (e.g. IL-1a, IL-6, IL-8, and CXCL2) and MMP expression28. Moreover, aged mice have a similar increase in the SASP phenotype as diabetic mice, suggesting that there may be similar mechanisms that delay healing with both aging and diabetes28.

Mucosal wounds must deal with the influence of bacteria since the oral cavity has a higher microbial load than skin. Bacterial colonization impairs healing by inducing prolonged inflammation, which intereferes with transition to the next phase of healing, maturation29. Diabetic skin wounds have an impaired host response to bacteria and an increased level of ROS that reduces microbial diversity and increases bacterial colonization30. Bacteria can also have direct effects on keratinocytes by stimulating apoptosis, reducing migration and decreasing proliferation31. Bacteria such as P. gingivalis and F. nucleatum can invade oral keratinocytes to interfere with re-epithelialization by down regulating genes that promote proliferation and integrins needed for cell migration31. The characteristics of diabetic oral wound healing by re-epithelilization are summarized in Figure 1.

Re-epithelialization Diabetes and FOXO1

As discussed above, diabetes negatively impacts keratinocyte proliferation and migration to interfere with re-epithelialization32. Diabetic conditions impair keratinocyte function, which include high glucose levels, increased formation of advanced glycation end products (AGEs) and reactive oxygen species (ROS). Elevated ROS levels have been shown to interfere with mucosal healing33 and increase inflammation in gingival epithelium34. The epithelium of diabetic individuals has greater expression of RAGE, which increases inflammation and worsens healing responses35,36. A unifying explanation that links high glucose, AGEs and ROS formation to diabetes-impaired re-epithelialization is the transcription factor FOXO1. Under normal conditions, FOXO1 improves re-epithelialization by protecting keratinocytes from oxidative stress and by inducing expression of TGF-β, both of which are needed for an appropriate healing response37. In diabetic conditions, high glucose or AGES cause an alteration in FOXO1 binding to the promoter regions of specific genes, most likely due to epigenetic modifications, that result in a diminished capacity to protect against oxidative stress and induce TGF-β1 transcription is significantly reduced32. These molecular changes are observed in vivo and in vitro and in both skin and oral mucosal keratinocytes. On a molecular level, diabetic conditions interfere with FOXO1 induced transcription by reducing FOXO1 binding to consensus response elements on gene promoters such as TGF-β1 and tissue inhibitor of matrix metalloproteinases 1 (TIMP1). In contrast, high glucose and AGEs increase FOXO1-promoter interactions leading to overexpression of factors that reduce re-epithelialization. More specifically, in a microenvironment with high glucose/AGE levels, FOXO1 induces expression of MMP9, serpin peptidase inhibitor clade B member 2 (SerpinB2), chemokine CCL20 and IL-36γ17,22,32. Interestingly, the negative effect of high glucose on keratinocyte migration is improved by addition of specific inhibitors for MMP9, or CCL20 or IL-36γ or by reducing FOXO1 activity22. The role of FOXO1 in inducing excessive production of these factors is demonstrated in vivo by lineage specific FOXO1 deletion in keratinocytes that reduces high levels of MMP9, SerpinB2, CCL20 and IL-36γ expression22. It should be noted that while high levels of these factors interfere with keratinocyte re-epithelialization, their total absence is also problematic. For example, the total absence of MMP9 is detrimental to keratinocyte migration38 as is absence of IL-36γ39.Thus, impaired re-epithelialization in diabetes is due to overexpression of factors that normally promote an appropriate healing response at physiological concentrations. The expression of FOXO1 provides a mechanistic explanation for how diabetic conditions can lead to overexpression of these factors as shown in Figure 1.

Connective Tissue Healing in Gingival Wounds

As discussed above, keratinized mucosa (eg. attached gingiva) covers the hard palate and alveolar bone, and its healing differs from that of nonkeratinized mucosa by relying on substantial connective tissue healing. Table 1 summarizes the findings from studies that investigate specific mucosal sites. Attached gingiva has a denser connective tissue compartment, which is home to contains fibroblasts, leukocytes, endothelial and other cells cells. Upon wounding, the coordinated healing response often results in complete regeneration of injured gingiva, which contrasts with skin wounds that heal by fibrosis4042. Fibroblasts are the principal cell type of connective tissues and are largely responsible for stromal healing by producing extracellular matrix (ECM) proteins such as collagen and proteoglycans. It has been proposed that the unique regenerative response of gingival wounds is attributed to the intrinsic properties of gingival fibroblasts that are distinct from skin fibroblasts. Cultured gingival fibroblasts are more proliferative and express reduced levels of TGF-β1, a pro-fibrogenic cytokine, compared to skin fibroblasts43. Importantly, gingival wounds have higher fibroblast density and increased connective tissue fill compared to skin wounds42, which is associated with reduced levels of TGF-β1 and elevated levels of anti-fibrogenic TGF-β3 in oral wounds4447. In a similar vein, fetal wounds that heal without scarring also exhibit low levels of TGF-β1 and high TGF-β3 levels47,48. Moreover, it is proposed that fibroblasts may down regulate immune responses in gingival wounds to facilitate resolution of inflammation and promote healing 11,42. Animal studies have shown that gingival fibroblasts are derived from neural crest origin (Wnt1-lineage) and can differentiate into tri-lineage mesenchymal cells in vitro49, which contrasts with mesoderm-derived skin fibroblasts49,50. Gingival fibroblasts cultured ex vivo are known as gingival mesenchymal stromal cells (GMSCs). GMSCs exhibit a pro-healing capacity by their anti-inflammatory properties rather than by directly differentiating into mesengenic cells5155. It is speculated that gingival fibroblasts may exhibit similar immunomodulatory function in vivo, which remains to be proven. In vitro culture conditions drastically shift the fibroblast transcriptome56,57. Thus the phenotypic and functional similarities of gingival fibroblasts and GMSCs may not necessarily align in the context of wound healing in vivo. It has been shown however that gingival fibroblasts express toll-like receptors and can secrete pro-inflammatory cytokines such as IL-6 and IL-8 when exposed to bacterial products in vitro and in vivo5861. Therefore, gingival fibroblasts, in addition to their traditional role in producing ECM, may exert polarizing effects on inflammation depending on physiological or pathologic condition.

Table 1:

Wound healing characteristics in oral mucosa over loose connective tissue or bone

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

Inflammation in gingival and mucosal wounds is more transient than in skin wounds11,42. Initial inflammation is characterized by neutrophil infiltration which is necessary to limit bacterial invasion into the wound site via secretion of proinflammatory cytokines, phagocytosis and NETosis62,63. Clearance of neutrophils occurs via apoptosis64, which may be targeted by bacteria to prolong their lifespan and prolong inflammation65. Another mechanism of neutrophil clearance is DEL-1-mediated efferocytosis by macrophages, which in turn reprograms macrophages toward a pro-healing phenotype to facilitate gingival regeneration66. Furthermore, lipid mediators such as resolvins and lipoxins have shown to have a pro-resolving effect in gingival inflammation from rabbit models67,68. Lymphocyte population has 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 external abrasion, stimulates gingival epithelial cells to secrete IL-6, which in turn leads to expansion of Th17 cells to produce defensins and neutrophil chemoattractants69. Moreover, gingival γδ T cells have been shown to produce amphiregulin to enhance wound healing in a periodontitis model70. It is well established that unresolved inflammation results in impaired wound healing and scarring71,72. Therefore, transit inflammation may partially contribute to gingival wound healing.

Oral wounds have reduced angiogenic response when compared to skin wounds73,74, which may be due to excessive angiogenesis of skin wounds that is accompanied by inflammation. Inflammatory signals such as IL-1β and TNF promote neovascularization75,76 and are linked to aberrant angiogenesis in rheumatoid arthritis77. Moreover, oral keratinocytes express reduced levels of VEGF compared to skin keratinocytes74, which may be advantageous. VEGF expression by the oral keratinocytes is FOXO1-dependent, and its deletion impairs angiogenesis and wound healing parameters in gingiva78.

Diabetic Impact on Cells Involved in Connective Tissue Healing

Diabetes negatively affects multiple cell types that participate in connective tissue wound healing in mucosal surfaces as summarized in Table 2. In both T1DM and T2DM mouse models, new connective tissue fill in palatal gingival wounds is reduced compared to normoglycemic control groups79. This is attributed to reduced fibroblast numbers in healing wounds and downregulation of pro-collagen I and III expression80. Mechanistically, high glucose levels promote apoptosis and reduce proliferation of gingival fibroblasts79,81, which mechanistically is due to the impact of greater oxidative stress, excessive activation of FOXO1 and high levels of caspase-3 expression80,82. High glucose levels also cause abnormal expression of MMP-1 from human gingival fibroblasts in vitro83,84. Moreover, AGEs upregulate proinflammatory cytokine expression such as IL-6 and IL-8 in human gingival fibroblasts83,84. Human GMSCs treatment improves wound healing in diabetic murine models85,86, but it has been shown that GMSCs isolated from diabetic patients exhibit reduced proliferative capacity compared to those from nondiabetic patients87. Similarly, bone marrow derived MSCs isolated from T2DM patients exhibit reduced immunomodulatory capacity in suppressing T cell proliferation88. Therefore, diabetes affects gingival fibroblasts by reducing their numbers through enhanced apoptosis, increases catabolic events in wounds through upregulation of MMP-1 and down-regulation of TIMP-1, and increases expression of inflammatory cytokines, which altogether lead to suboptimal connective tissue fill in gingival wounds.

Table 2:

Diabetes-induced alterations in wound healing.

Cell or Tissue Effect of Diabetes Reference
Epithelium Elevated ROS 33, 34
Epithelium Increased expression of receptor for AGE 35
Epithelium Alteration in downstream genes induced by FOXO1 22
Epithelium Reduced growth factor expression 17, 78
Epithelium Increased cytokine expression 15, 17
Epithelium Reduced re-epithelialization 17, 79
Saliva Reduced growth factor and increased inflammatory proteins 6, 20, 23
Connective tissue Increased fibroblast apoptosis 79
Connective tissue Increased cytokine expression and inflammation 15, 79
Connective tissue Reduce growth factor expression 102
Connective tissue Reduced fibroblast proliferation 79
Connective tissue Increased FOXO1 activity 79
Connective tissue Increased expression of receptor for AGE 35
Connective tissue Reduced angiogenesis 78
Connective tissue Reduced production of connective tissue matrix 78
Gingiva Increased inflammation and ROS 81
Alveolar bone Increased bone resorption and osteoclasts 111, 136
Alveolar bone Reduced bone formation 111, 132, 134, 144146
Alveolar bone Increased inflammation 129, 147
Alveolar bone Increased osteonecrosis 153, 154, 165
Mandibular bone Reduced bone formation 123, 124
Mandibular bone Increased postoperative infection risk 123

Diabetes impairs immune cell function and promotes inflammation to delay gingival wound healing. It is well established that diabetic wounds present with persistent inflammation81,89, particularly with high levels of the pro-inflammatory cytokine TNF. Inhibition of TNF can reverse diabetes-induced apoptosis of fibroblasts and mesenchymal stem cells in skin and osseous wounds26,90. A major source of TNF is macrophage/monocyte population, and high glucose has been shown to cause epigenetic changes in the promoter region of TNF and MCP-1 to promote persistent inflammation through over-activation of NF-kB91,92. Moreover, diabetes and a high glucose environment impair phagocytic activity of macrophages that is necessary for clearance of apoptotic bodies and bacteria93,94. Diabetes has also shown to prime neutrophils to undergo NETosis and cause aberrant inflammation in mice and humans95,96, which also may potentiate gingival inflammation97. Furthermore, metabolic changes in obesity and T2DM have been linked to reduced production of lipid mediators that are important for resolving inflammation in diabetic wounds98. Overall, diabetic impact on multiple immune cell types is detrimental for wound healing by interfering with their function and promoting persistent inflammation. This subsequently sets the stage for sub-optimal connective tissue wound healing.

Diabetes can have an opposite effect on angiogenesis depending on various anatomic locations. In diabetic retinopathy and nephropathy, angiogenesis is excessive, and subsequent vascular edema is linked to organ damage99,100. In diabetic skin wounds and chronic foot ulcers, angiogenesis is minimal, which is associated with delayed healing and reduced antimicrobial activity99,101. In buccal mucosal wounds of diabetic rats, new blood vessels are more permeable and the numbers are significantly reduced compared to normoglycemic wounds15,102. It has been shown that TNF inhibition can reverse the number of new blood vessel formation in diabetic osseous wound by restoring VEGF expression103, providing a link between persistent inflammation and reduced angiogenesis. Because reduced angiogenesis in gingival wounds can lead to delayed healing78, diabetes-mediated reduction in neovascularization may represent an important pathogenic mechanism to alter gingival wound healing. Taken as a whole, it is apparent that diabetes dysregulates the expression of a number of different factors to impair the wound healing process in various cell types as shown in Figure 2.

Figure 2. Connective tissue healing in diabetic oral wounds.

Figure 2.

Gingiva is composed of a thick layer of lamina propria overlaying bone. Diabetes-mediated hyperglycemia and ROS are the major driving force for directly induced fibroblast apoptosis and inhibiting proliferation. Moreover, fibroblasts express enhanced levels of pro-inflammatory cytokines IL-6, IL-8 sad MMP-1 in diabetic gingival wounds. Diabetes also interferes with a phagocytic function of macrophages and ttuedir polarisation from pro-inflammatory (MI) to pro-resolving (M2) type. Diabetes-induced epigenetic changes prevent resolution of inflammation. The promoter region of TGFb1 in kerartinocytes are methylated by high glucose whereas that of ITNF in marcphages are acetylated, resulting in reduced anti-inflammatory TGFb1 expression and over-expression of pro-inflammatory TNF. The overall outcome is persistent inflammation, which reduces stromal healing outcome.

Keratinocytes and Connective Tissue Healing

Crosstalk between keratinocytes and fibroblasts is needed for normal connective tissue healing104. Keratinocytes produce growth factors such as TGF-β, CTGF and VEGFA to stimulate connective tissue healing17,105107. TGF-β can directly and indirectly stimulate connective tissue formation; the latter is mediated by connective tissue growth factor (CTGF), also known as cellular communication network factor-2 (CCN-2)105. Application of CTGF in vivo stimulates fibroblast proliferation and deposition of collagen108. Studies examining human and animal dermal wounds demonstrate that VEGF is produced by keratinocytes during the healing process. The transcription factor FOXO1 activates wound healing responses in keratinocytes including the production of TGF-β1 to promote connective tissue healing in oral wounds17. FOXO1 ablation in keratinocytes diminishes TGF-β1 expression, which reduces CTGF expression by cells in connective tissue105. In type 1 diabetic oral wounds, the production of TGF-β1 is reduced. This may be explained by substantially reduced binding of FOXO1 to the TGF-β1 promoter caused by high levels of glucose or AGEs, which reduce TGF-β1 transcriptional activity17. Lineage specific ablation of FOXO1 reduces VEGF-A expression in keratinocytes in mucosal wounds and delays wound closure and with reduced angiogenesis77,107.

Molecular Mechanisms Involved in Impaired Diabetic Bone Healing

Dental and periodontal problems are exacerbated by diabetes109, often resulting in tooth extraction, bone augmentation and restoration with dental implants. These treatment modalities require an appropriate osseous healing reponse that involves concerted activity of osteoclasts, osteoblasts and mesenchymal stem cells (MSCs). It is well established that diabetes has a detrimental impact on bone formation by affecting these cell types. Diabetes uncouples bone remodeling by increasing osteoclast numbers and inhibiting expansion of periodontal fibroblasts and osteoblasts in an experimental periodontitis model110,111. This is linked to a negative effect of high glucose on the proliferation and osteogenic differentiation of progenitor cells112. The molecular mechanism by which diabetes affects osteoblast progenitors and osteoblasts has been investigated extensively in orthopedic injury models. The early phase of diabetic bone healing is characterized by impaired osteoid matrix production and reduced cellularity, which may be caused by defective MSC recruitment, proliferation and impaired osteogenic differentiation113,114. Diabetes further suppresses mRNA expression of the Dlx5 and Runx-2 transcription factors, which are responsible for the expression of the osteoblastic phenotype115, suggesting that the low rate of bone formation observed in diabetic animals may be related to deficiencies in the differentiation of MSCs to osteoprogenitor cells and in the osteoblastic cell maturation114. Studies in fracture healing support this hypothesis as diabetes results in impaired bone formation116. Moreover, diabetes leads to aberrant activation of NF-kB and enhances inflammation due in part to the loss of immunomodulatory MSCs117. In turn, persistent inflammation characterized by high levels of TNF reduces MSC expansion through upregulation of FOXO190 and suppression of IHH118 to ultimately impair healing in diabetic fractures. In the oral cavity, animal studies have recently identified genetically traceable MSC populations by a-SMA, Axin2 and Lepr expression119121, which reside in the periodontal ligament space between the tooth surface and bone and participate in bone healing after injury. These animal models may be crucial in understanding intraoral MSC and osteoprogenitor behavior necessary for bone healing and how diabetes alters it in vivo.

Guided bone regeneration (GBR) is a surgical procedure that restores deficient bone volume with the use bone graft. A key element is a barrier membrane that separates the bone graft from the soft tissue to exclude soft tissue infiltration that can interfere with bone formation122. The effect of experimental diabetes on GBR has been investigated by means of histology and gene expression analyses. GBR can sufficiently regenerate bone even in diabetic rats, but the quantity of bone formed is less than that formed in normoglycemic animals123. Investigation of gene expression profiling has shown that poorly controlled diabetes downregulates pathways related to cell division, energy production and osteogenesis during the proliferative phase of intramembranous bone healing after bone augmentation treatment124. Although detailed mechanisms responsible for the reduced cell proliferation rates are still not completely elucidated, it has been suggested that diabetes may suppress the expression of growth factors needed for osteogenesis during the early phase of bone healing125,126. In support of this diabetes reduces expression of basic fibroblast growth factor (FGF), an important mitogenic factor for MSCs during bone healing127. Animal studies indicate that experimentally induced type 1 diabetes diminishes the osteoprogenitor population in bone marrow, possibly due to increased apoptosis via oxidative stress, which in turn, may impair osteoblastogenesis, bone formation, and bone healing128. Experimental studies have also demonstrated reduced mRNA and protein expression of platelet derived growth factor (PDGF), TGF-β1, IGF-I and VEGF in femoral fractures of diabetic rats during early osseous healing correlates with reduced cell proliferation125,126. Diabetes increases TNF in the oral cavity that prolongs detrimental inflammation. FGF-2, TGFβ-1, bone morphogenetic protein-2 (BMP-2), and BMP-6 are suppressed during periodontal bone foramtion, which is rescued when inflammation is reduced with a TNF inhibitor129.

The exogenous delivery of IGF-I or rhFGF-2 enhanced the healing of calvarial and periodontal defects in mice and rats130132. Preclinical studies have demonstrated that diabetic rats exhibit reduced periodontal bone regeneration, which can be partly rescued with local application of enamel matrix derivative (EMD) that contains a milleu of growth factors133,134. A recent prospective study in human subjects showed that treatment of periodontal bony defects with enamel matrix derivative, which contains a mixture of undefined growth factors, in diabetic patients, can achieve a regenerative outcome that is comparable to non-diabetic patients after 3 years135.

Diabetes has a significant impact on inflammatory responses. Pathways linked to innate and adaptive immune responses are generally altered in poorly controlled diabetics compared to healthy controls. Prolonged inflammation is detrimental to bone formation, in part, because NF-kB activation in response to inflammatory stimuli blocks the expression of bone matrix proteins136. Diabetes also interferes with proper immune functions that are necessary for pathogen recognition and removal137139. These findings may explain increased risk for infectious complications following GBR treatment in patients with uncontrolled diabetes. Comparable findings were also reported by other authors evaluating the healing of bone defects in animals140 and humans141. The regulation of osteoblasts and osteoblast precursors is complex and involves the timed expression of genes encoding pro-inflammatory cytokines (Il1α, Il1β, Il-6, TNF), chemokines (Ccl20, Cxcl1, Cxcl2, Cxcl10), chemokine receptors (Ccr2, Ccr5, Ccr6) and cell adhesion molecules (Vcam1 and Icam1) that are needed for migration and activation of these cells142,143. Prolonged inflammation disrupts the level of expression and timing of these factors.

Taken together, the available data suggest that suppressed differentiation, proliferation and/or bone forming capacity of osteoblastic cells during the critical early healing period appear to play an important role in the pathogenic mechanism underlying poor bone formation in diabetes. In particular, poorly controlled diabetes appears to result in a higher and dysfunctional inflammatory response during the early phase of bone healing which in turn, may be resposnible for the increased risk of infection in subjects with uncontrolled diabetes.

Impact of Diabetes on Tooth Extraction

The healing of bone in extraction sockets following tooth removal in diabetes has been evaluated in a number of animal and human studies. Using T1DM rat model, alveolar bone remodeling following tooth extraction evealed that the quantity and rate of alveolar bone formation was significantly lower in diabetic group compared to controls144. After a healing period of 10 days, diabetic sockets contained thin and scanty collagen fibers whereas in control groups there were thick collagen fibers that formed a pretrabecular scaffold along the trabeculae145. Interestingly, there was no evidence of diabetic microangiopathy in the extraction sockets of diabetic animals compared with healthy controls or with insulin-treated animals. Healing of in humans with T2DM showed a deficit in the early stages of healing and in diabetic pigs revealed delayed healing and decreased osteogenic differentiation of MSCs146. Although substantial bone formation still occurred in diabetic animals there was incomplete mineralization in the center of the socket compared to that of control groups which were completely filled with newly formed bone146. The observations made in animals were in line with those made in humans. Compared to nondiabetic control group, T2DM patients had delayed bone fill in the extraction sockets at multiple time points post surgery146 and 54.7% had defective healing manifested by inadequate bone formation. In diabetic mice the extraction sockets consistently have a higher inflammatory profile, with more M1 macrophages and TNF-α expression and less M2 macrophages and PPARγ expression compared to non-diabetic controls147.

While several studies point to deficits in bone formation after dental extraction in diabetic patients not all reports reach this conclusion148151. For example it has been reported that diabetics have increased neutrophil recruitment following tooth extraction but bone healing in diabetic and normal subjects were similar149. Taken as a whole the overall assessment of healing following tooth extraction in diabetic patients is slower compared to non-diabetic ones, especially in the initial phases151. The available evidence suggests that a) in poorly controlled T1DM bone formation is inhibited, resulting in delayed healing and deficient bone formation; and b) tooth extractions can be performed in well controlled diabetic patients, without major postoperative complications.

Diabetes as a Comorbidity for MRONJ

In the oral cavity, osseous wounds such as extraction socket or exogenous bone grafts eventually heal even under diabetic condition, albeit at a delayed rate. However, major differences exist in bone healing for nondiabetic and diabetic patients that suffer from medication-related osteonecrosis of the jaw (MRONJ). MRONJ may be localized and easily treated or may become widespread and develop into a serious clinical condition characterized by the progressive destruction of the jawbones. It occurs in patients on anti-resorptive medications that have undergone oral or periodontal surgery extraction152. There is a strong association between diabetes and the incidence of MRONJ153157; even though the exact pathogenetic mechanisms remain unclear, several experimental studies have proposed converging pathways. Much like diabetes-mediated delay in extraction socket healing158, bisphosphonate treatment delays bone formation in extraction sockets in rats159. There are a number of potential mechanisms that may play a role including diabetes-reduced coupled bone formation, and the impact of diabetes on increased osteocyte and osteoblast apoptosis160,161. Another possible converging mechanism is altered immune system. Animal and human studies have demonstrated that bisphosphonate treatment impairs neutrophil and macrophage chemotaxis and function to promote immunosuppression162164. Studies in mice have also demonstrated a pro-inflammatory response by diabetes and zolendronate treatment in macrophages, which was responsible for MRONJ-like healing after tooth extraction165,166. The etiology for either disease is multifactorial and complex, and further studies may identify pathways that can be targeted to prevent MRONJ incidents in diabetic patients. The impact of diabetes on intraoral bone healing as discussed above is summarized in Figure 3.

Figure 3. Diabetic impact on various cases of intraoral bone healing.

Figure 3.

In the oral cavity, bone healing takes place under different scenarios inch as. left 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 its and periodnotal fibroblasts This is accompanied by persistent inflammation and reduced growth factor expression. In guided bone regeneration, which, restores the hard tissue volume necessary for dental implant placement, diabetes has shown to inhtbit 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 shoun enhanced apoptosis, persistent inflammation and improper recruitment of leukocytes as a converging mechanism.

Conclusions and future directions

Diabetes negatively impacts several aspects of oral and dermal wound healing. When directly compared, the effects of diabetes on both are relatively consistent. Diabetes alters the rate of healing by reducing keratinocyte migration, the production of growth factors by a number of different cell types including keratinocytes, interferes with a number of cellular functions such as cellular proliferation and differentiation and the expression of genes that are essential for forming soft 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. These changes lead to difficulty in down regulating inflammation, increasing oxidative stress and enhancing apoptosis. In the skin these changes may lead to biofilm formation in healing wounds that stopped the repair process and in the mouth, a biofilm may form on surgically exposed bone that prevents healing. In oral wounds that require substantial connective tissue healing, diabetes exerts its deterimental impact on multiple cell types such as gingival fibroblasts, endothelial cells and leukocytes through persistent inflammation and aberrant cell apoptosis. Impaired healing by diabetes can be rescued by controlling inflammation or by genetic inhibition of FOXO1 in experimental models, thus targetting these processes and/or transcription factors with monoclonal antibody or small molecule inhibitor may be a viable method to treat diabetic oral wounds in humans. Similarly, diabetic intraoral bone healing is characterized by reduced expression of growth factors that are necessary for bone formation, therefore the use of recombinant growth factors is an attractive therapeutic option for diabetic patients. Given the difficulty that diabetic patients have in resolving inflammation the optimal treatment may include a therapeutic that facilitates resolution and delivers a growth factor to diabetic wounds.

A mechanistic understanding of oral wound healing is largely based on animal studies. These studies are advantageous by targetting specific genes or factors that may be implicated in different stages of the healing process. However, they may not fully translate to human models as diabetes itself is a multifactorial disease that is often accompanied by other metabolic conditions. Moreover, certain oral diseases in humans such as MRONJ may not be fully replicated in mouse models. Despite these limitations, animal studies have contributed significantly to understanding some detreimental effects of diabetes on oral wound healing, and clinical efforts to supplement growth factors to improve healing process are active underway. Future studies may test a translational value of recombinant growth factor therapy, anti-inflammatory drugs and/or small molecule inhibitors of transcription factors that are implicated in diabetic healing. Any other future studies?

Acknowledgments

Conflict of interest: All authors have read the journal’s authorship policy on disclosure of potential conflict interest.

All authors have read the journal’s authorship agreement and the manuscript has been reviewed by and approved by all listed authors.

This study was supported by NIH grants to K.I.K. (K08-DE027129, R01-DE030415) and D.T.G. (R01DE019108).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Glim JE, van Egmond M, Niessen FB, Everts V & Beelen RH Detrimental dermal wound healing: what can we learn from the oral mucosa? Wound Repair Regen 21, 648–660 (2013). [DOI] [PubMed] [Google Scholar]
  • 2.Angelov N, Moutsopoulos N, Jeong MJ, et al. Aberrant mucosal wound repair in the absence of secretory leukocyte protease inhibitor. Thromb Haemost 92, 288–297 (2004). [DOI] [PubMed] [Google Scholar]
  • 3.Winning TA & Townsend GC Oral mucosal embryology and histology. Clin Dermatol 18, 499–511 (2000). [DOI] [PubMed] [Google Scholar]
  • 4.Sculean A, Gruber R & Bosshardt DD Soft tissue wound healing around teeth and dental implants. J Clin Periodontol 41 Suppl 15, S6–22 (2014). [DOI] [PubMed] [Google Scholar]
  • 5.Glim JE, Beelen RH, Niessen FB, Everts V & Ulrich MM The number of immune cells is lower in healthy oral mucosa compared to skin and does not increase after scarring. Arch Oral Biol 60, 272–281 (2015). [DOI] [PubMed] [Google Scholar]
  • 6.Nagy A, Nagashima H, Cha S, et al. Reduced oral wound healing in the NOD mouse model for type 1 autoimmune diabetes and its reversal by epidermal growth factor supplementation. Diabetes 50, 2100–2104 (2001). [DOI] [PubMed] [Google Scholar]
  • 7.Noguchi S, Ohba Y & Oka T Effect of salivary epidermal growth factor on wound healing of tongue in mice. Am J Physiol 260, E620–625 (1991). [DOI] [PubMed] [Google Scholar]
  • 8.Torres P, Castro M, Reyes M & Torres VA Histatins, wound healing, and cell migration. Oral Dis 24, 1150–1160 (2018). [DOI] [PubMed] [Google Scholar]
  • 9.Bodner L, Kaffe I, Cohen Z & Dayan D Long-term effect of desalivation on extraction wound healing: a densitometric study in rats. Dentomaxillofac Radiol 22, 195–198 (1993). [DOI] [PubMed] [Google Scholar]
  • 10.Johnson A, Francis M & DiPietro LA Differential Apoptosis in Mucosal and Dermal Wound Healing. Adv Wound Care (New Rochelle) 3, 751–761 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Iglesias-Bartolome R, Uchiyama A, Molinolo AA, et al. Transcriptional signature primes human oral mucosa for rapid wound healing. Sci Transl Med 10(2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chen L, Arbieva ZH, Guo S, et al. Positional differences in the wound transcriptome of skin and oral mucosa. BMC Genomics 11, 471 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Chen L, Gajendrareddy PK & DiPietro LA Differential expression of HIF-1alpha in skin and mucosal wounds. J Dent Res 91, 871–876 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.American Diabetes, A. Diagnosis and classification of diabetes mellitus. Diabetes Care 36 Suppl 1, S67–74 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Brizeno LA, Assreuy AM, Alves AP, et al. Delayed healing of oral mucosaina diabetic rat model: Implication of TNF-alpha, IL-1beta and FGF-2. Life Sci 155, 36–47 (2016). [DOI] [PubMed] [Google Scholar]
  • 16.Lan CC, Liu IH, Fang AH, Wen CH & Wu CS Hyperglycaemic conditions decrease cultured keratinocyte mobility: implications for impaired wound healing in patients with diabetes. Br J Dermatol 159, 1103–1115 (2008). [DOI] [PubMed] [Google Scholar]
  • 17.Xu F, Othman B, Lim J, et al. Foxo1 inhibits diabetic mucosal wound healing but enhances healing of normoglycemic wounds. Diabetes 64, 243–256 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Oxford GE, Tayari L, Barfoot MD, et al. Salivary EGF levels reduced in diabetic patients. J Diabetes Complications 14, 140–145 (2000). [DOI] [PubMed] [Google Scholar]
  • 19.Robinson CP, Cornelius J, Bounous DE, et al. Characterization of the changing lymphocyte populations and cytokine expression in the exocrine tissues of autoimmune NOD mice. Autoimmunity 27, 29–44 (1998). [DOI] [PubMed] [Google Scholar]
  • 20.Cabras T, Pisano E, Mastinu A, et al. Alterations of the salivary secretory peptidome profile in children affected by type 1 diabetes. Mol Cell Proteomics 9, 2099–2108 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 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 predictor of wound healing. Diabet Med 25, 419–426 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhang C, Lim J, Jeon HH, et al. FOXO1 deletion in keratinocytes improves diabetic wound healing through MMP9 regulation. Sci Rep 7, 10565 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kuehl MN, Rodriguez H, Burkhardt BR & Alman AC Tumor Necros is Factor-alpha, Matrix-Metalloproteinases 8 and 9 Levels in the Saliva Are Associated with Increased Hemoglobin A1c in Type 1 Diabetes Subjects. PLoS One 10, e0125320 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Alzoubi F, Joseph B & Andersson L Healing of soft tissue lacerations in diabetic-induced rats. Dent Traumatol 33, 438–443 (2017). [DOI] [PubMed] [Google Scholar]
  • 25.Wang Y & Graves DT Keratinocyte Function in Normal and Diabetic Wounds and Modulation by FOXO1. J Diabetes Res 2020, 3714704 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Siqueira MF, Li J, Chehab L, et al. Impaired wound healing in mouse models of diabetes is mediated by TNF-alpha dysregulation and associated with enhanced activation of forkhead box O1 (FOXO1). Diabetologia 53, 378–388 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Al-Mulla F, Leibovich SJ, Francis IM & Bitar MS Impaired TGF-beta signaling and a defect in resolution of inflammation contribute to delayed wound healing in a female rat model of type 2 diabetes. Mol Biosyst 7, 3006–3020 (2011). [DOI] [PubMed] [Google Scholar]
  • 28.Wilkinson HN, Clowes C, Banyard KL, et al. Elevated Local Senescence in Diabetic Wound Healing Is Linked to Pathological Repair via CXCR2. J Invest Dermatol 139, 1171–1181 e1176 (2019). [DOI] [PubMed] [Google Scholar]
  • 29.Guo S & Dipietro LA Factors affecting wound healing. J Dent Res 89, 219–229 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kim JH, Ruegger PR, Lebig EG, et al. High Levels of Oxidative Stress Create a Microenvironment That Significantly Decreases the Diversity of the Microbiota in Diabetic Chronic Wounds and Promotes Biofilm Formation. Front Cell Infect Microbiol 10, 259 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bhattacharya R, Xu F, Dong G, et al. Effect of bacteria on the wound healing behavior of oral epithelial cells. PLoS One 9, e89475 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zhang C, Ponugoti B, Tian C, et al. FOXO1 differentially regulates both normal and diabetic wound healing. J Cell Biol 209, 289–303 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Coskun S, Gulec EG, Balabanli B & Acarturk F Effects of epidermal growth factor on lipid peroxidation and nitric oxide levels in oral mucosal ulcer healing: a time-course study. Surg Today 37, 570–574 (2007). [DOI] [PubMed] [Google Scholar]
  • 34.Huang X, Yang X, Ni J, et al. Hyperglucose contributes to periodontitis: involvement of the NLRP3 pathway by engaging the innate immunity of oral gingival epithelium. J Periodontol 86, 327–335 (2015). [DOI] [PubMed] [Google Scholar]
  • 35.Katz J, Bhattacharyya I, Farkhondeh-Kish F, et al. Expression of the receptor of advanced glycation end products in gingival tissues of type 2 diabetes patients with chronic periodontal disease: a study utilizing immunohistochemistry and RT-PCR. J Clin Periodontol 32, 40–44 (2005). [DOI] [PubMed] [Google Scholar]
  • 36.Shaikh-Kader A, Houreld NN, Rajendran NK & Abrahamse H The link between advanced glycation end products and apoptosis in delayed wound healing. Cell Biochem Funct 37, 432–442 (2019). [DOI] [PubMed] [Google Scholar]
  • 37.Ponugoti B, Xu F, Zhang C, et al. FOXO1 promotes wound healing through the up-regulation of TGF-beta1 and prevention of oxidative stress. J Cell Biol 203, 327–343 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bullard KM, Lund L, Mudgett JS, et al. Impaired wound contraction in stromelysin-1-deficient mice. Ann Surg 230, 260–265 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Jiang Z, Liu Y, Li C, et al. IL-36gamma Induced by the TLR3-SLUG-VDR Axis Promotes Wound Healing via REG3A. J Invest Dermatol 137, 2620–2629 (2017). [DOI] [PubMed] [Google Scholar]
  • 40.Griffin MF, desJardins-Park HE, Mascharak S, Borrelli MR & Longaker MT Understanding the impact of fibroblast heterogeneity on skin fibrosis. Dis Model Mech 13(2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.desJardins-Park HE, Mascharak S, Chinta MS, Wan DC & Longaker MT The Spectrum of Scarring in Craniofacial Wound Repair. Front Physiol 10, 322 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Mak K, Manji A, Gallant-Behm C, et al. Scarless healing of oral mucosa is characterized by faster resolution of inflammation and control of myofibroblast action compared to skin wounds in the red Duroc pig model. J Dermatol Sci 56, 168–180 (2009). [DOI] [PubMed] [Google Scholar]
  • 43.Mah W, Jiang G, Olver D, et al. Human gingival fibroblasts display a non-fibrotic phenotype distinct from skin fibroblasts in three-dimensional cultures. PLoS One 9, e90715 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Schrementi ME, Ferreira AM, Zender C & DiPietro LA Site-specific production of TGF-beta in oral mucosal and cutaneous wounds. Wound Repair Regen 16, 80–86 (2008). [DOI] [PubMed] [Google Scholar]
  • 45.Szpaderska AM, Zuckerman JD & DiPietro LA Differential injury responses in oral mucosal and cutaneous wounds. J Dent Res 82, 621–626 (2003). [DOI] [PubMed] [Google Scholar]
  • 46.Eslami A, Gallant-Behm CL, Hart DA, et al. Expression of integrin alphavbeta6 and TGF-beta in scarless vs scar-forming wound healing. J Histochem Cytochem 57, 543–557 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Occleston NL, O'Kane S, Laverty HG, et al. Discovery and development of avotermin (recombinant human transforming growth factor beta 3): a new class of prophylactic therapeutic for the improvement of scarring. Wound Repair Regen 19 Suppl 1, s38–48 (2011). [DOI] [PubMed] [Google Scholar]
  • 48.Lichtman MK, Otero-Vinas M & Falanga V Transforming growth factor beta (TGF-beta) isoforms in wound healing and fibrosis. Wound Repair Regen 24, 215–222 (2016). [DOI] [PubMed] [Google Scholar]
  • 49.Xu X, Chen C, Akiyama K, et al. Gingivae contain neural-crest- and mesoderm-derived mesenchymal stem cells. J Dent Res 92, 825–832 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Isaac J, Nassif A, Asselin A, et al. Involvement of neural crest and paraxial mesoderm in oral mucosal development and healing. Biomaterials 172, 41–53 (2018). [DOI] [PubMed] [Google Scholar]
  • 51.Zhang QZ, Su WR, Shi SH, et al. Human gingiva-derived mesenchymal stem cells elicit polarization of m2 macrophages and enhance cutaneous wound healing. Stem Cells 28, 1856–1868 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhang Q, Shi S, Liu Y, et al. Mesenchymal stem cells derived from human gingiva are capable of immunomodulatory functions and ameliorate inflammation-related tissue destruction in experimental colitis. J Immunol 183, 7787–7798 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gu Y & Shi S Transplantation of gingiva-derived mesenchymal stem cells ameliorates collagen-induced arthritis. Arthritis Res Ther 18, 262 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Kou X, Xu X, Chen C, et al. The Fas/Fap-1/Cav-1 complex regulates IL-1RA secretion in mesenchymal stem cells to accelerate wound healing. Sci Transl Med 10(2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Ahangar P, Mills SJ, Smith LE, Gronthos S & Cowin AJ Human gingival fibroblast secretome accelerates wound healing through anti-inflammatory and proangiogenic mechanisms. NPJ Regen Med 5, 24 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Walmsley GG, Rinkevich Y, Hu MS, et al. Live fibroblast harvest reveals surface marker shift in vitro. Tissue Eng Part C Methods 21, 314–321 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kim T, Echeagaray OH, Wang BJ, et al. In situ transcriptome characteristics are lost following culture adaptation of adult cardiac stem cells. Sci Rep 8, 12060 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bautista-Hernandez LA, Gomez-Olivares JL, Buentello-Volante B & Bautista-de Lucio VM Fibroblasts: The Unknown Sentinels Eliciting Immune Responses Against Microorganisms. Eur J Microbiol Immunol (Bp) 7, 151–157 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Bartold PM & Haynes DR Interleukin-6 production by human gingival fibroblasts. J Periodontal Res 26, 339–345 (1991). [DOI] [PubMed] [Google Scholar]
  • 60.Herath TD, Darveau RP, Seneviratne CJ, et al. Tetra- and penta-acylated lipid A structures of Porphyromonas gingivalis LPS differentially activate TLR4-mediated NF-kappaB signal transduction cascade and immuno-inflammatory response in human gingival fibroblasts. PLoS One 8, e58496 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Dongari-Bagtzoglou AI & Ebersole JL Increased presence of interleukin-6 (IL-6) and IL-8 secreting fibroblast subpopulations in adult periodontitis. J Periodontol 69, 899–910 (1998). [DOI] [PubMed] [Google Scholar]
  • 62.Phillipson M & Kubes P The Healing Power of Neutrophils. Trends Immunol 40, 635–647 (2019). [DOI] [PubMed] [Google Scholar]
  • 63.Mohanty T, Sjogren J, Kahn F, et al. A novel mechanism for NETosis provides antimicrobial defense at the oral mucosa. Blood 126, 2128–2137 (2015). [DOI] [PubMed] [Google Scholar]
  • 64.deOliveira S, Rosowski EE & Huttenlocher A Neutrophil migration in infection and wound repair: going forward in reverse. Nat Rev Immunol 16, 378–391 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Murray DA & Wilton JM Lipopolysaccharide from the periodontal pathogen Porphyromonas gingivalis prevents apoptosis of HL60-derived neutrophils in vitro. Infect Immun 71, 7232–7235 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kourtzelis I, Li X, Mitroulis I, et al. DEL-1 promotes macrophage efferocytosis and clearance of inflammation. Nat Immunol 20, 40–49 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Serhan CN, Jain A, Marleau S, et al. Reduced inflammation and tissue damage in transgenic rabbits overexpressing 15-lipoxygenase and endogenous anti-inflammatory lipid mediators. J Immunol 171, 6856–6865 (2003). [DOI] [PubMed] [Google Scholar]
  • 68.Serhan CN, Chiang N & Van Dyke TE Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol 8, 349–361 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Dutzan N, Abusleme L, Bridgeman H, et al. On-going Mechanical Damage from Mastication Drives Homeostatic Th17 Cell Responses at the Oral Barrier. Immunity 46, 133–147 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Krishnan S, Prise IE, Wemyss K, et al. Amphiregulin-producing gammadelta T cells are vital for safeguarding oral barrier immune homeostasis. Proc Natl Acad Sci U S A 115, 10738–10743 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rockey DC, Bell PD & Hill JA Fibrosis--a common pathway to organ injury and failure. N Engl J Med 372, 1138–1149 (2015). [DOI] [PubMed] [Google Scholar]
  • 72.Wang ZC, Zhao WY, Cao Y, et al. The Roles of Inflammation in Keloid and Hypertrophic Scars. Front Immunol 11, 603187 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.DiPietro LA Angiogenesis and wound repair: when enough is enough. J Leukoc Biol 100, 979–984 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Szpaderska AM, Walsh CG, Steinberg MJ & DiPietro LA Distinct patterns of angiogenesis in oral and skin wounds. J Dent Res 84, 309–314 (2005). [DOI] [PubMed] [Google Scholar]
  • 75.Fahey E & Doyle SL IL-1 Family Cytokine Regulation of Vascular Permeability and Angiogenesis. Front Immunol 10, 1426 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Sainson RC, Johnston DA, Chu HC, et al. TNF primes endothelial cells for angiogenic sprouting by inducing a tip cell phenotype. Blood 111, 4997–5007 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Elshabrawy HA, Chen Z, Volin MV, et al. The pathogenic role of angiogenesis in rheumatoid arthritis. Angiogenesis 18, 433–448 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Jeon HH, Yu Q, Lu Y, et al. FOXO1 regulates VEGFA expression and promotes angiogenesis in healing wounds. J Pathol 245, 258–264 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Desta T, Li J, Chino T & Graves DT Altered fibroblast proliferation and apoptosis in diabetic gingival wounds. J Dent Res 89, 609–614 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Liu R, Desta T, He H & Graves DT Diabetes alters the response to bacteria by enhancing fibroblast apoptosis. Endocrinology 145, 2997–3003 (2004). [DOI] [PubMed] [Google Scholar]
  • 81.Kido D, Mizutani K, Takeda K, et al. Impact of diabetes on gingival wound healing via oxidative stress. PLoS One 12, e0189601 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Buranasin P, Mizutani K, Iwasaki K, et al. High glucose-induced oxidative stress impairs proliferation and migration of human gingival fibroblasts. PLoS One 13, e0201855 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Irwin CR, Myrillas TT, Traynor P, Leadbetter N & Cawston TE The role of soluble interleukin (IL)-6 receptor in mediating the effects of IL-6 on matrix metalloproteinase-1 and tissue inhibitor of metalloproteinase-1 expression by gingival fibroblasts. J Periodontol 73, 741–747 (2002). [DOI] [PubMed] [Google Scholar]
  • 84.Lew JH, Naruishi K, Kajiura Y, et al. High Glucose-Mediated Cytokine Regulation in Gingival Fibroblasts and THP-1 Macrophage: a Possible Mechanism of Severe Periodontitis with Diabetes. Cell Physiol Biochem 50, 973–986 (2018). [DOI] [PubMed] [Google Scholar]
  • 85.Shi Q, Qian Z, Liu D, et al. GMSC-Derived Exosomes Combined with a Chitosan/Silk Hydrogel Sponge Accelerates Wound Healing in a Diabetic Rat Skin Defect Model. Front Physiol 8, 904 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zhang W, Zhou L, Dang J, et al. Human Gingiva-Derived Mesenchymal Stem Cells Ameliorate Streptozoticin-induced T1DM in mice via Suppression of T effector cells and Up-regulating Treg Subsets. Sci Rep 7, 15249 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Assem M, Kamal S, Sabry D, Soliman N & Aly RM Preclinic alAssessment of the Proliferation Capacity of Gingival and Periodontal Ligament Stem Cells from Diabetic Patients. Open Access Maced J Med Sci 6, 254–259 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kizilay Mancini O, Shum-Tim D, Stochaj U, Correa JA & Colmegna I Age, atherosclerosis and type 2 diabetes reduce human mesenchymal stromal cell-mediated T-cell suppression. Stem Cell Res Ther 6, 140 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Graves DT, Liu R, Alikhani M, Al-Mashat H & Trackman PC Diabetes-enhanced inflammation and apoptosis--impact on periodontal pathology. J Dent Res 85, 15–21 (2006). [DOI] [PubMed] [Google Scholar]
  • 90.Ko KI, Coimbra LS, Tian C, et al. Diabetes reduces mesenchymal stem cells in fracture healing through a TNFalpha-mediated mechanism. Diabetologia 58, 633–642 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Miao F, Gonzalo IG, Lanting L & Natarajan R In vivo chromatin remodeling events leading to inflammatory gene transcription under diabetic conditions. J Biol Chem 279, 18091–18097 (2004). [DOI] [PubMed] [Google Scholar]
  • 92.El-Osta A, Brasacchio D, Yao D, et al. Transient high glucose causes persistent epigenetic changes and altered gene expression during subsequent normoglycemia. J Exp Med 205, 2409–2417 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Pavlou S, Lindsay J, Ingram R, Xu H & Chen M Sustained high glucose exposure sensitizes macrophage responses to cytokine stimuli but reduces their phagocytic activity. BMC Immunol 19, 24 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Lecube A, Pachon G, Petriz J, Hernandez C & Simo R Phagocytic activity is impaired in type 2 diabetes mellitus and increases after metabolic improvement. PLoS One 6, e23366 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wong SL, Demers M, Martinod K, et al. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat Med 21, 815–819 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Fadini GP, Menegazzo L, Rigato M, et al. NETosis Delays Diabetic Wound Healing in Mice and Humans. Diabetes 65, 1061–1071 (2016). [DOI] [PubMed] [Google Scholar]
  • 97.Vitkov L, Minnich B, Knopf J, et al. NETs Are Double-Edged Swords with the Potential to Aggravate or Resolve Periodontal Inflammation. Cells 9(2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Hellmann J, Tang Y & Spite M Proresolving lipid mediators and diabetic wound healing. Curr Opin Endocrinol Diabetes Obes 19, 104–108 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Simons M Angiogenesis, arteriogenesis, and diabetes: paradigm reassessed? J Am Coll Cardiol 46, 835–837 (2005). [DOI] [PubMed] [Google Scholar]
  • 100.Cooper ME, Vranes D, Youssef S, et al. Increased renal expression of vascular endothelial growth factor (VEGF) and its receptor VEGFR-2 in experimental diabetes. Diabetes 48, 2229–2239 (1999). [DOI] [PubMed] [Google Scholar]
  • 101.Veith AP, Henderson K, Spencer A, Sligar AD & Baker AB Therapeutic strategies for enhancing angiogenesis in wound healing. Adv Drug Deliv Rev 146, 97–125 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Yamano S, Kuo WP & Sukotjo C Downregulated gene expression of TGF-betas in diabetic oral wound healing. J Craniomaxillofac Surg 41, e42–48 (2013). [DOI] [PubMed] [Google Scholar]
  • 103.Lim JC, Ko KI, Mattos M, et al. TNFalpha contributes to diabetes impaired angiogenesis in fracture healing. Bone 99, 26–38 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Werner S, Krieg T & Smola H Keratinocyte-fibroblast interactions in wound healing. J Invest Dermatol 127, 998–1008 (2007). [DOI] [PubMed] [Google Scholar]
  • 105.Zhang C, Lim J, Liu J, et al. FOXO1 expression in keratinocytes promotes connective tissue healing. Sci Rep 7, 42834 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Shephard P, Martin G, Smola-Hess S, et al. Myofibroblast differentiation is induced in keratinocyte-fibroblast co-cultures and is antagonistically regulated by endogenous transforming growth factor-beta and interleukin-1. Am J Pathol 164, 2055–2066 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Johnson KE & Wilgus TA Vascular Endothelial Growth Factor and Angiogenesis in the Regulation of Cutaneous Wound Repair. Adv Wound Care (New Rochelle) 3, 647–661 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Alfaro MP, Deskins DL, Wallus M, et al. A physiological role for connective tissue growth factor in early wound healing. Lab Invest 93, 81–95 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Taylor GW, Manz MC & Borgnakke WS Diabetes, periodontal diseases, dental caries, and tooth loss: a review of the literature. Compend Contin Educ Dent 25, 179–184, 186–178, 190; quiz 192 (2004). [PubMed] [Google Scholar]
  • 110.Andriankaja OM, Galicia J, Dong G, et al. Gene expression dynamics during diabetic periodontitis. J Dent Res 91, 1160–1165 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Liu R, Bal HS, Desta T, et al. Diabetes enhances periodontal bone loss through enhanced resorption and diminished bone formation. J Dent Res 85, 510–514 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kim HS, Park JW, Yeo SI, Choi BJ & Suh JY Effects of high glucose on cellular activity of periodontal ligament cells in vitro. Diabetes Res Clin Pract 74, 41–47 (2006). [DOI] [PubMed] [Google Scholar]
  • 113.Weiss RE & Reddi AH Influence of experimental diabetes and insulin on matrix-induced cartilage and bone differentiation. Am J Physiol 238, E200–207 (1980). [DOI] [PubMed] [Google Scholar]
  • 114.McCabe LR Understanding the pathology and mechanisms of type I diabetic bone loss. J Cell Biochem 102, 1343–1357 (2007). [DOI] [PubMed] [Google Scholar]
  • 115.Lu H, Kraut D, Gerstenfeld LC & Graves DT Diabetes interferes with the bone formation by affecting the expression of transcription factors that regulate osteoblast differentiation. Endocrinology 144, 346–352 (2003). [DOI] [PubMed] [Google Scholar]
  • 116.Huang Z, Pei X & Graves DT The Interrelationship Between Diabetes, IL-17 and Bone Loss. Curr Osteoporos Rep 18, 23–31 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Ko KI, Syverson AL, Kralik RM, et al. Diabetes-Induced NF-kappaB Dysregulation in Skeletal Stem Cells Prevents Resolution of Inflammation. Diabetes 68, 2095–2106 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Tevlin R, Seo EY, Marecic O, et al. Pharmacological rescue of diabetic skeletal stem cell niches. Sci Transl Med 9(2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Zhang D, Zhang S, Wang J, et al. LepR-Expressing Stem Cells Are Essential for Alveolar Bone Regeneration. J Dent Res 99, 1279–1286 (2020). [DOI] [PubMed] [Google Scholar]
  • 120.Roguljic H, Matthews BG, Yang W, et al. In vivo identification of periodontal progenitor cells. J Dent Res 92, 709–715 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Yuan X, Pei X, Zhao Y, et al. A Wnt-Responsive PDL Population Effectuates Extraction Socket Healing. J Dent Res 97, 803–809 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Elgali I, Omar O, Dahlin C & Thomsen P Guided bone regeneration: materials and biological mechanisms revisited. Eur J Oral Sci 125, 315–337 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Retzepi M, Lewis MP & Donos N Effect of diabetes and metabolic control on de novo bone formation following guided bone regeneration. Clin Oral Implants Res 21, 71–79 (2010). [DOI] [PubMed] [Google Scholar]
  • 124.Retzepi M, Calciolari E, Wall I, Lewis MP & Donos N The effect of experimental diabetes and glycaemic control on guided bone regeneration: histology and gene expression analyses. Clin Oral Implants Res 29, 139–154 (2018). [DOI] [PubMed] [Google Scholar]
  • 125.Tyndall WA, Beam HA, Zarro C, O'Connor JP & Lin SS Decreased platelet derived growth factor expression during fracture healing in diabetic animals. Clin Orthop Relat Res, 319–330 (2003). [DOI] [PubMed] [Google Scholar]
  • 126.Gandhi A, Beam HA, O'Connor JP, Parsons JR & Lin SS The effects of local insulin delivery on diabetic fracture healing. Bone 37, 482–490 (2005). [DOI] [PubMed] [Google Scholar]
  • 127.Kawaguchi H, Kurokawa T, Hanada K, et al. Stimulation of fracture repair by recombinant human basic fibroblast growth factor in normal and streptozotocin-diabetic rats. Endocrinology 135, 774–781 (1994). [DOI] [PubMed] [Google Scholar]
  • 128.Weinberg E, Maymon T, Moses O & Weinreb M Streptozotocin-induced diabetes in rats diminishes the size of the osteoprogenitor pool in bone marrow. Diabetes Res Clin Pract 103, 35–41 (2014). [DOI] [PubMed] [Google Scholar]
  • 129.Pacios S, Kang J, Galicia J, et al. Diabetes aggravates periodontitis by limiting repair through enhanced inflammation. FASEB J 26, 1423–1430 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Thaller SR, Lee TJ, Armstrong M, Tesluk H & Stern JS Effect of insulin-like growth factor type 1 on critical-size defects in diabetic rats. J Craniofac Surg 6, 218–223 (1995). [DOI] [PubMed] [Google Scholar]
  • 131.Santana RB & Trackman PC Controlled release of fibroblast growth factor 2 stimulates bone healing in an animal model of diabetes mellitus. Int J Oral Maxillofac Implants 21, 711–718 (2006). [PubMed] [Google Scholar]
  • 132.Bizenjima T, Seshima F, Ishizuka Y, et al. Fibroblast growth factor-2 promotes healing of surgically created periodontal defects in rats with early, streptozotocin-induced diabetes via increasing cell proliferation and regulating angiogenesis. J Clin Periodontol 42, 62–71 (2015). [DOI] [PubMed] [Google Scholar]
  • 133.Wyganowska-Swiatkowska M, Urbaniak P, Nohawica MM, Kotwicka M & Jankun J Enamel matrix proteins exhibit growth factor activity: A review of evidence at the cellular and molecular levels. Exp Ther Med 9, 2025–2033 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Takeda K, Mizutani K, Matsuura T, et al. Periodontal regenerative effect of enamel matrix derivative in diabetes. PLoS One 13, e0207201 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Mizutani K, Shioyama H, Matsuura T, et al. Periodontal regenerative therapy in patients with type 2 diabetes using minimally invasive surgical technique with enamel matrix derivative under 3-year observation: A prospective cohort study. J Periodontol (2020). [DOI] [PubMed] [Google Scholar]
  • 136.Pacios S, Xiao W, Mattos M, et al. Osteoblast Lineage Cells Play an Essential Role in Periodontal Bone Loss Through Activation of Nuclear Factor-Kappa B. Sci Rep 5, 16694 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Stegenga ME, vander Crabben SN, Blumer RM, et al. Hyperglycemia enhances coagulation and reduces neutrophil degranulation, whereas hyperinsulinemia inhibits fibrinolysis during human endotoxemia. Blood 112, 82–89 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Hair PS, Echague CG, Rohn RD, et al. Hyperglycemic conditions inhibit C3-mediated immunologic control of Staphylococcus aureus. J Transl Med 10, 35 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Martinez N, Ketheesan N, Martens GW, et al. Defects in early cell recruitment contribute to the increased susceptibility to respiratory Klebsiella pneumoniae infection in diabetic mice. Microbes Infect 18, 649–655 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Lee RSB, Hamlet SM & Ivanovski S The influence of titanium surface characteristics on macrophage phenotype polarization during osseous healing in type I diabetic rats: a pilot study. Clin Oral Implants Res 28, e159–e168 (2017). [DOI] [PubMed] [Google Scholar]
  • 141.Sun M, Yang J, Wang J, et al. TNF-alpha is upregulated in T2DM patients with fracture and promotes the apoptosis of osteoblast cells in vitro in the presence of high glucose. Cytokine 80, 35–42 (2016). [DOI] [PubMed] [Google Scholar]
  • 142.Tanaka Y, Morimoto I, Nakano Y, et al. Osteoblasts are regulated by the cellular adhesion through ICAM-1 and VCAM-1. J Bone Miner Res 10, 1462–1469 (1995). [DOI] [PubMed] [Google Scholar]
  • 143.Yellowley C CXCL12/CXCR4 signaling and other recruitment and homing pathways in fracture repair. Bonekey Rep 2, 300 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Fang Y, Wang LP, Du FL, Liu WJ & Ren GL Effects of insulin-like growth factor I on alveolar bone remodeling in diabetic rats. J Periodontal Res 48, 144–150 (2013). [DOI] [PubMed] [Google Scholar]
  • 145.Devlin H, Garland H & Sloan P Healing of tooth extraction sockets in experimental diabetes mellitus. J Oral Maxillofac Surg 54, 1087–1091 (1996). [DOI] [PubMed] [Google Scholar]
  • 146.Zhang S, Song S, Wang S, et al. Type 2 diabetes affects postextraction socket healing and influences first-stage implant surgery: A study based on clinical and animal evidence. Clin Implant Dent Relat Res 21, 436–445 (2019). [DOI] [PubMed] [Google Scholar]
  • 147.Shen X, Shen X, Li B, et al. Abnormal macrophage polarization impedes the healing of diabetes-associated tooth sockets. Bone 143, 115618 (2021). [DOI] [PubMed] [Google Scholar]
  • 148.Aronovich S, Skope LW, Kelly JP & Kyriakides TC The relationship of glycemic control to the outcomes of dental extractions. J Oral Maxillofac Surg 68, 2955–2961 (2010). [DOI] [PubMed] [Google Scholar]
  • 149.Fernandes KS, Glick M, deSouza MS, Kokron CM. & Gallottini M Association between immunologic parameters, glycemic control, and postextraction complications in patients with type 2 diabetes. J Am Dent Assoc 146, 592–599 (2015). [DOI] [PubMed] [Google Scholar]
  • 150.Huang S, Dang H, Huynh W, Sambrook PJ & Goss AN The healing of dental extraction sockets in patients with Type 2 diabetes on oral hypoglycaemics: a prospective cohort. Aust Dent J 58, 89–93 (2013). [DOI] [PubMed] [Google Scholar]
  • 151.Gadicherla S, Smriti K, Roy S, et al. Comparison of Extraction Socket Healing in Non-Diabetic, Prediabetic, and Type 2 Diabetic Patients. Clin Cosmet Investig Dent 12, 291–296 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Ruggiero SL, Dodson TB, Fantasia J, et al. American Association of Oral and Maxillofacial Surgeons position paper on medication-related osteonecrosis of the jaw--2014 update. J Oral Maxillofac Surg 72, 1938–1956 (2014). [DOI] [PubMed] [Google Scholar]
  • 153.Khamaisi M, Regev E, Yarom N, et al. Possible association between diabetes and bisphosphonate-related jaw osteonecrosis. J Clin Endocrinol Metab 92, 1172–1175 (2007). [DOI] [PubMed] [Google Scholar]
  • 154.Molcho S, Peer A, Berg T, Futerman B & Khamaisi M Diabetes microvascular disease and the risk for bisphosphonate-related osteonecrosis of the jaw: a single center study. J Clin Endocrinol Metab 98, E1807–1812 (2013). [DOI] [PubMed] [Google Scholar]
  • 155.Vidal-Real C, Perez-Sayans M, Suarez-Penaranda JM, Gandara-Rey JM & Garcia-Garcia A Osteonecrosis of the jaws in 194 patients who have undergone intravenous bisphosphonate therapy in Spain. Med Oral Patol Oral Cir Bucal 20, e267–272 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Barasch A, Cunha-Cruz J, Curro FA, et al. Risk factors for osteonecrosis of the jaws: a case-control study from the CONDOR Dental PBRN. Tex Dent J 130, 299–307 (2013). [PubMed] [Google Scholar]
  • 157.Watters AL, Hansen HJ, Williams T, et al. Intravenous bisphosphonate-related osteonecrosis of the jaw: long-term follow-up of 109 patients. Oral Surg Oral Med Oral Pathol Oral Radiol 115, 192–200 (2013). [DOI] [PubMed] [Google Scholar]
  • 158.Shyng YC, Devlin H & Sloan P The effect of streptozotocin-induced experimental diabetes mellitus on calvarial defect healing and bone turnover in the rat. Int J Oral Maxillofac Surg 30, 70–74 (2001). [DOI] [PubMed] [Google Scholar]
  • 159.Hikita H, Miyazawa K, Tabuchi M, Kimura M & Goto S Bisphosphonate administration prior to tooth extraction delays initial healing of the extraction socket in rats. J Bone Miner Metab 27, 663–672 (2009). [DOI] [PubMed] [Google Scholar]
  • 160.Ikebuchi Y, Aoki S, Honma M, et al. Coupling of bone resorption and formation by RANKL reverse signalling. Nature 561, 195–200 (2018). [DOI] [PubMed] [Google Scholar]
  • 161.Gangoiti MV, Anbinder PS, Cortizo AM & McCarthy AD Morphological changes induced by advanced glycation endproducts in osteoblastic cells: effects of co-incubation with alendronate. Acta Histochem 115, 649–657 (2013). [DOI] [PubMed] [Google Scholar]
  • 162.Hoefert S, Schmitz I, Weichert F, Gaspar M & Eufinger H Macrophages and bisphosphonate-related osteonecrosis of the jaw (BRONJ): evidence of local immunosuppression of macrophages in contrast to other infectious jaw diseases. Clin Oral Investig 19, 497–508 (2015). [DOI] [PubMed] [Google Scholar]
  • 163.Kalyan S, Chandrasekaran V, Quabius ES, Lindhorst TK & Kabelitz D Neutrophil uptake of nitrogen-bisphosphonates leads to the suppression of human peripheral blood gammadelta T cells. Cell Mol Life Sci 71, 2335–2346 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Kuiper JW, Forster C, Sun C, Peel S & Glogauer M Zoledronate and pamidronate depress neutrophil functions and survival in mice. Br J Pharmacol 165, 532–539 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Zhang Q, Yu W, Lee S, et al. Bisphosphonate Induces Osteonecrosis of the Jaw in Diabetic Mice via NLRP3/Caspase-1-Dependent IL-1beta Mechanism. J Bone Miner Res 30, 2300–2312 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Yang X, Xu X, Chen J, et al. Zoledronic acid regulates the synthesis and secretion of IL-1beta through Histone methylation in macrophages. Cell Death Discov 6, 47 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES