Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Invited Review

M. Retzepi The effect of diabetes mellitus on


N. Donos
osseous healing

Authors’ affiliations: Key words: bone healing, diabetes mellitus, hyperglycaemia, insulin, ossification
M. Retzepi, N. Donos, Periodontology Unit,
Clinical Research Division, UCL Eastman Dental
Institute, London, UK Abstract: Diabetes mellitus and, in particular, type 1 diabetes has been associated with
impaired osseous wound healing properties. The scope of the present review is to discuss
Corresponding author:
Dr Maria Retzepi
the clinical evidence supporting a higher rate of complications during fracture healing in
Periodontology Unit diabetic patients and the histological evidence indicating impaired potential for
Clinical Research Division intramembranous and endochondral ossification in the presence of uncontrolled
UCL Eastman Dental Institute
256 Gray’s Inn Road experimental diabetes. The article further provides a synthesis of our current understanding
London of the plausible molecular mechanisms underlying the diabetic bone healing
WC1X 8LD, UK
Tel.: þ 44 207915 1135 pathophysiology and of the role of insulin treatment in promoting osseous healing in the
Fax: þ 44 207915 1137 diabetic status.
e-mail: m.retzepi@eastman.ucl.ac.uk

Diabetes mellitus is defined as a ‘‘group of or ‘‘diabetic osteopathy’’ (Bouillon 1991).


metabolic diseases characterised by hyper- The diabetic skeletal phenotype presents
glycaemia resulting from defects in insulin the following features:
secretion, insulin action or both’’ (The
Expert Committee on the Diagnosis and  diminished linear bone growth during
Classification of Diabetes Mellitus 1997). the pubertal growth spurt in adoles-
The vast majority of diabetic cases fall in cents with diabetes (Salerno et al.
one of the following categories: 1997);
 reduction of bone mineral density and
(a) Type 1 diabetes mellitus (T1DM), a
increased risk for occurrence of osteo-
condition characterised by absolute
penia and osteoporosis (Vestergaard
deficiency of insulin secretion, result-
2007);
ing from autoimmune destruction of
 increased fracture risk (Janghorbani
the insulin producing b cells of the
et al. 2007);
pancreas;
 poor osseous healing characteristics and
(b) Type 2 diabetes mellitus (T2DM), a
impaired bone regeneration potential
condition characterised by impaired
(Cozen 1972).
insulin function, i.e. resistance to in-
sulin action, combined with inade-
quate compensatory insulin secretion
response. The scope of this narrative review is to
Date:
Accepted 28 December 2009
present and synthesise the evidence emer-
Diabetes mellitus has been associated ging from preclinical studies, clinical trials
To cite this article:
Retzepi M, Donos N. The effect of diabetes mellitus on with the occurrence of a series of complica- and systematic reviews on the effect of
osseous healing. tions involving the skeletal system, collec- diabetes mellitus on the osseous healing
Clin. Oral Impl. Res. 21, 2010; 673–681.
doi: 10.1111/j.1600-0501.2010.01923.x tively referred to as ‘‘diabetic bone disease’’ process.

c 2010 John Wiley & Sons A/S


 673
Retzepi & Donos  The effect of diabetes mellitus on osseous healing

Diabetes effect on skeletal tion indices present unaltered or decreased Diabetes effect on osseous
tissue metabolism (Shires et al. 1981; Botolin et al. 2005; Hie healing: clinical studies
et al. 2007). In accordance with these
A recent meta-analysis has demonstrated observations, a series of studies have sug- Clinical studies have demonstrated that
that T1DM is clearly associated with de- gested normal or reduced osteoclastic ac- diabetic patients present significantly de-
creased bone density and with significantly tivity based on bone histomorphometry layed fracture healing, as the fracture callus
increased fracture risk, whereas, on the indices, including osteoclast number, ero- is slow to appear and mature (Cozen 1972;
contrary, although the relative risk for hip sion depth and erosion surface (Shires et al. Loder 1988; Bibbo et al. 2001). Specifi-
fracture is moderately increased in T2DM 1981; Verhaeghe et al. 1990; Hou et al. cally, Loder (1988) reported that the frac-
patients, the bone mineral density in the 1993). ture union time is prolonged by 87% in
hip and the spine presents increased (Ves- Taking into consideration that, in the non-displaced fractures in non-neuropathic
tergaard 2007). Because hyperinsulinaemia context of the bone remodelling process, diabetic patients compared with non-
is associated with increased bone mineral bidirectional signalling activity occurs be- diabetic patients. Furthermore, a retrospec-
density, it has been suggested that the tween osteoblasts and osteoclasts, these tive study addressing the treatment success
observed differential clinical presentation findings overall indicate that suppressed of calcaneus fractures, reported that the
may be accounted for by the discrepancy osteoblastic activity rather than increased presence of diabetes was associated with a
in systemic insulin concentration between osteoclastic activity accounts for the ob- relative risk of 3.4 for wound complications
the T1DM and the T2DM condition, i.e. served osteoporotic changes characterising (Folk et al. 1999). Three case series
insulinopenia vs. hyperinsulinaemia, re- the T1-diabetic bone phenotype (McCabe have reported increased incidence of infec-
spectively (Thrailkill et al. 2005). 2007). tious complications, delayed union, non-
It has been postulated that the decreased union, redislocation and pseudarthrosis
bone mineral density in T1DM patients is following elective arthrodesis in diabetic
aetiopathogenetically related to a sup- Impaired microarchitectural patients (Stuart & Morrey 1990; Papa
pressed bone turnover profile, which is quality of diabetic bone et al. 1993; Tisdel et al. 1995). It should
determined by the relative contributions be noted however that, up to date, there is a
of bone formation by osteoblasts and bone Further to the reduction in bone mineral lack of well-designed, large-scale, epide-
resorption by osteoclasts (McCabe 2007). density, detrimental changes in the bone miological studies reporting on the inci-
T1DM has been associated with decreased morphology, mineralisation and matrix dence of complications during the osseous
osteoblastic activity further to several stu- composition contribute to the observed healing process in the population of dia-
dies reporting reduced systemic levels of deterioration in the biomechanical proper- betic patients.
osteocalcin and insulin growth factor I ties of the diabetic bone, i.e. energy absorp-
(IGF-I) in T1DM patients (Pedrazzoni tion, torsional strength, angular deformity
et al. 1989; Olmos et al. 1994; Kemink (Dixit & Ekstrom 1980; Einhorn et al. Effect of experimental diabetes
et al. 2000) and in in vivo models of 1988). With regard to diabetic bone micro- on intramembranous
experimental diabetes (Shires et al. 1981; architectural morphology, poor trabecular ossification
Verhaeghe et al. 1990; Horcajada-Molteni connectivity, increased porosity and lower
et al. 2001; Botolin et al. 2005). In addi- bone spicule/marrow space ratio have been Deficient intramembranous bone forma-
tion, studies based on bone histomorpho- demonstrated via quantitative bone histo- tion in experimental diabetes has been
metric indices in untreated experimental morphometry in models of experimentally histologically and histomorphometrically
diabetes have consistently reported dimin- induced diabetes (Hou et al. 1993; Suzuki confirmed in various osteotomy and frac-
ished osteoid formation, combined with et al. 2003). Deficits in the mineralised ture models.
defective bone mineral apposition rate surface area may include decreased hydro- Santana et al. (2003) reported suppressed
(Goodman & Hori 1984; Verhaeghe et al. xyapatite crystal perfection, lower calcium- intramembranous bone formation, using a
1990). The observations of suppressed bone to-phosphate composition of the ash, as model of calvarial defects of 1, 1.6 and
formation and osteoblast recruitment and well as reduced ash content (Einhorn 2.1 mm in diameter, in streptozotocin-
activity in insulin-deficient models have et al. 1988). Furthermore, the collagenous induced experimental diabetes in mice.
been further confirmed in bone biopsy matrix content is reduced (Verhaeghe et al. Qualitative histological observations indi-
specimens from T1DM patients (Krakauer 1994). Spanheimer et al. (1988) reported a cated less developed marrow spaces and
et al. 1995). Contrary to bone formation, 48% reduction in collagen production by suppressed cellularity and vascularisation
markers of bone resorption display a non- bone, within 2 weeks following induction after 14 days of healing, whereas plani-
uniform systemic expression pattern in of experimental diabetes, which could not metric measurements demonstrated that
T1DM patients, as they may be increased be accounted for merely by undernutrition calvarial bone bridging was inhibited by
(Mathiassen et al. 1990; Olmos et al. (Umpierrez et al. 1989). The collagenous 40% in diabetic animals, irrespectively
1994), unaltered (Kemink et al. 2000) or matrix also features increased glycation- of the defect size. Follak et al. (2004a)
even decreased (Cloos et al. 1998). In induced cross-linking, which may further evaluated the effect of spontaneous experi-
animal models of untreated experimental deteriorate the diabetic bone quality (Saito mental diabetes on osseous healing of cra-
diabetes, the concentrations of bone resorp- et al. 2006). niotomy defects of various sizes in rats.

674 | Clin. Oral Impl. Res. 21, 2010 / 673–681 c 2010 John Wiley & Sons A/S

Retzepi & Donos  The effect of diabetes mellitus on osseous healing

The authors did not detect differences in associated with impaired intramembranous mineralised area in the diabetic animals
the bone healing process of small bone bone formation potential, mostly after the compared with healthy controls (Gandhi
defects (0.4 mm) between poorly controlled first and during the second week of osseous et al. 2005).
diabetic and healthy animals. In 0.8 mm healing. It is noteworthy that the type and The histological and histomorphometri-
defects, significantly decreased trabecular size of the osseous defect seem to consti- cal observations are in line with evidence of
bone volume was observed in poorly tute independent determinants of the ac- reduced biomechanical properties of the
controlled diabetes at 24 and 42 days tual impact of diabetes on the ossification fracture callus in uncontrolled experimen-
of healing. In 1.2 mm and larger defects process. tal diabetes (Macey et al. 1989). The
however, the poorly controlled diabetic diabetic femoral fracture callus presents
status correlated with severe mineralisa- delayed recovery of its structural properties
tion disorders during the first 2 weeks of Effect of experimental diabetes and material strength, and more specifi-
healing, as indicated by suppressed on endochondral ossification cally reduced shear stress, torque to failure
expression of dynamic histomorphometric and stiffness at 2, 4, 6 and 8 weeks
parameters of mineralisation, i.e. apposi- Qualitative histological studies on the ef- of healing (Macey et al. 1989; Funk
tion, formation and timing of mineralisa- fect of experimental diabetes on the frac- et al. 2000; Beam et al. 2002; Follak et al.
tion. Interestingly, in this study, the ture gap callus, a site where endochondral 2005).
osteotomy size was an independent vari- ossification occurs, demonstrated reduced In conclusion, experimental diabetes is
able in predicting successful osseous amounts of undifferentiated mesenchymal associated with reduced cellularity in
repair, irrespective of the diabetic meta- cells (MSCs) at 4 days of healing (Beam the fracture milieu during the first post-
bolic status. et al. 2002; Tyndall et al. 2003). Further- traumatic week and with subsequently
The periosteal callus represents an addi- more, at 4 and 7 days after the injury, the delayed and impaired cartilage differentia-
tional site featuring intramembranous ossi- diabetic gap callus featured reduced carti- tion and mineralisation, which account
fication during the fracture healing process. lage formation with less proliferating chon- for impaired histological and biomechani-
Histological reports on the effect of sponta- drocytes (Beam et al. 2002; Tyndall et al. cal healing parameters observed at the
neous experimental diabetes on periosteal 2003; Gandhi et al. 2005). Histological late stages of the endochondral ossification
callus formation in femoral fracture mod- observations in a similar fracture model, process.
els, indicated diminished osteoid deposi- at later healing periods, correlated the dia-
tion at 2 and 4 days of healing and more betic status with delayed differentiation of
immature appearance of the newly formed the callus at 2 weeks of healing, whereas, Effect of diabetes on
woven bone 7 days after the injury (Beam at 4 weeks, prominent hypertrophic chon- osseointegration
et al. 2002; Tyndall et al. 2003). drocytes undergoing ossification were still
Lu et al. (2003) have employed the present, thus indicating a delay in cellular Clinical studies
marrow ablation model in order to study differentiation (Follak et al. 2005). Evidence based on clinical trials addressing
the effect of streptozotocin-induced dia- Histomorphometric evaluation of the the effect of diabetes on the survival rates of
betes on intramembranous bone formation. diabetic gap callus at 2 and 4 weeks of titanium implants is equivocal at present.
The model involved removal of the trabe- healing, demonstrated significantly re- Among the controlled clinical studies
cular bone and marrow from the marrow duced dynamic parameters of mineralisa- available, some report an increased risk
cavity of the diaphyseal portion of the tibia, tion, including apposition, formation and for implant failure in diabetic patients
which was followed by a synchronised timing of mineralisation (Follak et al. compared with healthy controls with a
series of intramembranous bone repair 2005). These observations are in line with relative risk of 2.75 (95% confidence inter-
events during a period of 10 days. Histo- a previous report by the same group, which val: 1.46–5.18) (Morris et al. 2000; Moy
morphometric evaluation indicated similar evaluated histomorphometrically the effect et al. 2005), whereas others do not support
amounts of mesenchymal tissue on day 4 of spontaneous diabetes on the osseous an association between diabetes and im-
in diabetic and healthy animals, whereas healing process, using a model of 10 mm plant failure (Smith et al. 1992). Most of
decreased bone and marrow formation were circular osteotomies created in the distal the case-series studies on implant survival
observed after 6 and 10 days of healing in femur (Follak et al. 2004b). The authors in diabetic patients support a trend of
the diabetic group. reported that the poorly controlled diabetic increased early vs. late implant failure
The distraction osteogenesis model has state correlated with suppressed and re- (Mombelli & Cionca 2006), although there
also been used in order to study the effect of tarded mineralisation process, evidenced have also been reports of increased failure
experimental diabetes on intramembra- by significantly reduced apposition, forma- rate after functional loading, presumably
nous bone regeneration potential. Fowlkes tion and timing of the mineralisation up to associated to impaired bone remodelling
et al. (2008) reported that after 15 days of the 14th postoperative day. Histomorpho- potential (Shernoff et al. 1994; Fiorellini
healing, bone bridging was 19.3  12.5% metric evaluation of the diabetic gap callus et al. 2000). Furthermore, one study re-
in the diabetic animals vs. 52.4  6.2% in at late healing periods of 6 and 8 weeks, ported higher crestal bone loss during the
the control group. indicated incomplete bridging of the cor- first year of implant loading in T2DM
Summarising the available histological tice/fracture callus, diminished remodel- patients (Accursi 2000). Taking into
evidence, experimental diabetes has been ling activity and decreased percent consideration that the vast majority of

c 2010 John Wiley & Sons A/S


 675 | Clin. Oral Impl. Res. 21, 2010 / 673–681
Retzepi & Donos  The effect of diabetes mellitus on osseous healing

participants in these clinical trials were Molecular mechanisms the levels of critical growth factors, thus
well-controlled T2DM patients, it may be implicated in impaired diabetic disrupting the normal progression of the
concluded that implant placement is not bone healing inflammatory phase and osteogenesis, dur-
contraindicated in diabetic patients pre- ing the early osseous healing stages (Tyn-
senting good metabolic control (Mombelli The impaired osteoid matrix production dall et al. 2003; Gandhi et al. 2005). An
& Cionca 2006). and the reduced cellularity observed during initial report by Kawaguchi et al. (1994)
the early osseous healing period in uncon- indicated decreased expression of basic fi-
Preclinical studies trolled experimental diabetes may stem broblast growth factor (FGF), an established
Evidence of delayed peri-implant bone for- from deficits in the recruitment of MSCs mitogenic factor for MSCs during osseous
mation in uncontrolled experimental dia- in the osteoblastic lineage, impaired differ- healing, in the diabetic fracture callus at 1
betes is based on qualitative observations of entiation and proliferation of cells of the and 3 weeks following the injury. Reduced
more immature histological appearance of osteoblastic lineage and/or suppressed mRNA and protein expression of platelet-
the newly formed bone following place- osteoblastic cell activity (Weiss & Reddi derived growth factor (PDGF), another po-
ment of titanium implants in the femur 1980; McCabe 2007). tent mitogenic factor, have also been
(Nevins et al. 1998) or tibia (Giglio et al. The hypothesis of impaired commit- observed in femoral fractures of sponta-
2000) of diabetic rats. These results were ment of MSCs in the osteoblastic lineage neously diabetic rats during the first heal-
in accordance with previous reports of im- during osseous healing in uncontrolled dia- ing week and has been correlated with
paired new bone formation following place- betes is supported by evidence of sup- reduced cell proliferation rates during the
ment of hydroxyapatite-coated implants pressed expression of transcription factors early phases of endochondral bone forma-
in the tibiae of diabetic rats (Iyama et al. required for the acquisition of the osteo- tion (Tyndall et al. 2003). A recent study
1997). In accordance with the qualitative blastic phenotype. Specifically, suppressed by Gandhi et al. (2005) confirmed the
histological observations, significantly mRNA expression of the Dlx5 and Runx-2 reduction in PDGF, transforming growth
reduced bone-to-implant contact has been transcription factors, which control acqui- factor b1 (TGF-b1), IGF-I and vascular
reported in diabetic animals after 2, 3, 4, 8 sition of the osteoblastic phenotype, has endothelial growth factor protein expres-
and 12 weeks of healing following been observed at 4 and 6 days of intramem- sion levels at femoral fracture sites at 4
placement of titanium implants (Nevins branous bone healing in the marrow abla- and 7 days of healing, in poorly controlled
et al. 1998; Siqueira et al. 2003; Ottoni & tion model (Lu et al. 2003). These data spontaneously diabetic rodents. The results
Chopard 2004), although the figures indicate that the diminished bone forma- of various studies evaluating the efficacy of
were comparable to healthy controls at 12 tion observed in the diabetic status could be local delivery of growth factors in promot-
postoperative weeks in two other studies related to deficiencies in the differentiation ing osseous healing in uncontrolled dia-
(Gerritsen et al. 2000; Retzepi et al. 2010). of MSCs to osteoprogenitor cells and in betes support further the hypothesis of
The observed discrepancy may be attribu- the osteoblastic cell maturation (McCabe deficient production and/or action of
ted to different models of endosseous 2007). growth factors and hormones in the oss-
implantation, i.e. endochondral vs. intra- Reduced proliferation of osteoblastic eous healing process in uncontrolled dia-
membranous bones and/or to differences cells has been also suggested as a plausible betes. Thaller et al. (1995) reported that
in the implant surfaces employed, i.e. mechanism for impaired bone healing. delivery of IGF-I in critical size calvarial
smooth vs. rough. Furthermore, a reduc- This is supported by studies associating defects in streptozotocin-induced diabetic
tion in peri-implant bone density (Gerrit- the decreased cellularity commonly ob- rats enhanced the osseous healing process.
sen et al. 2000) and up to 50% reduction in served in the diabetic callus, with an up Santana & Trackman (2006) reported that
the amount of newly formed bone after the to 50% reduction in immunohistochem- exogenous application of 5 mg of rhFGF-2
first 2 weeks of healing (Takeshita et al. ical indices of the cell proliferation rate in promoted bone bridging by 2.6-fold in cal-
1998; Giglio et al. 2000; McCracken et al. the periosteal and in the gap callus at 2, 4 varial defects in streptozotocin-induced
2000; Siqueira et al. 2003; Ottoni & Cho- and 7 days of healing (Beam et al. 2002; diabetic mice after 14 days of healing.
pard 2004) have also been reported. With Gebauer et al. 2002; Tyndall et al. 2003; The implication of reduced activity of
regard to the effect of experimental diabetes Gandhi et al. 2005). In accordance with osteoblastic cells in the impaired bone
on the biomechanical properties of endoss- these immunohistochemical observations, healing potential in the presence of diabetes
eous implants, reduced removal torque gene expression profiling investigation has is supported by evidence of substantially
values have been observed at 8 and 12 indicated that uncontrolled diabetes fea- reduced mRNA expression of the specific
weeks postimplantation (Margonar et al. tured downregulation of pathways related bone matrix protein osteocalcin and of type
2003). to cell division, energy production and 1 collagen at 4 and 6 days of intramem-
It may be therefore concluded that, osteogenesis during the proliferative phase branous bone healing in the marrow abla-
although osseointegration is feasible in of intramembranous bone healing follow- tion model (Lu et al. 2003). This is in
uncontrolled experimental diabetes, this ing guided bone regeneration application accordance with previous reports of a 50–
condition is associated with impaired (Retzepi 2009). The underlying causes of 55% reduction in total collagen content
peri-implant bone formation, the aetio- the reduced cell proliferation rates remain, (Macey et al. 1989) and 54–70% decrease
pathologic features of which remain largely however, largely unknown. It has been in type X collagen content (Topping et al.
unknown. hypothesised that diabetes may suppress 1994) during the first 2 weeks of fracture

676 | Clin. Oral Impl. Res. 21, 2010 / 673–681 c 2010 John Wiley & Sons A/S

Retzepi & Donos  The effect of diabetes mellitus on osseous healing

healing in uncontrolled experimental dia- has been demonstrated that systemic in- treated diabetic rats with well-compen-
betes. sulin treatment improves the biomechani- sated metabolic status, were similar to
In conclusion, suppressed differentia- cal parameters in streptozotocin-induced non-diabetic controls and by 25% higher
tion, proliferation and/or bone forming experimental diabetes during the late stages compared with poorly controlled diabetic
capacity of osteoblastic cells during the of fracture healing (Herbsman et al. 1968; animals. These results are in accordance
critical early healing period have been im- Macey et al. 1989). Furthermore, Beam with a previous study reporting a 27%
plicated as plausible pathogenetic mechan- et al. (2002) showed that insulin therapy decrease in cell proliferation in a diabetic
isms underlying poor bone formation in normalised the biomechanical parameters, explant model, which was normalised fol-
uncontrolled diabetes. i.e. percent mechanical stiffness, torque to lowing insulin delivery (Weiss & Reddi
failure and ultimate shear stress in sponta- 1980). A more recent study confirmed
neously diabetic animals. In accordance these observations, by reporting enhanced
Insulin-mediated glycaemic with these observations, good metabolic cellular proliferation in well-controlled vs.
control reverses diabetic bone control achieved via systemic insulin de- poorly controlled diabetic animals, during
pathophysiology livery in spontaneously diabetic rats nor- the first 4 weeks of healing in a femoral
malised the histomorphometric indices of fracture model (Follak et al. 2005).
The results of clinical studies assessing the mineralisation and callus bone content,
relationship between diminished bone during later stages of fracture healing, com-
mineral density and glycaemic control in pared with diabetic animals with low me- Plausible pathogenetic links
T1DM are conflicting. Among the clinical tabolic control (Beam et al. 2002). These between diabetes and impaired
studies supporting an impact of T1DM reports are in accordance with evidence of osseous healing
on bone mineral density, several support improved bone bridging of osteotomy cal-
a correlation between decreased bone varial (Santana et al. 2003) and femoral Role of hyperglycaemia
mineral density and poor metabolic control (Follak et al. 2004b) defects in well- In vitro studies have reported that exposure
(Lettgen et al. 1995; Valerio et al. 2002; controlled diabetic animals via insulin of osteoblastic cells to increased glucose
Heap et al. 2004), whereas others did not treatment. Insulin-mediated metabolic concentrations reduces their differentiation
show an association (Gunczler et al. 1998). control has been also shown to improve potential, leads to irregular bone mineral
Various experimental studies have de- the histomorphometric parameters of nodule formation (Balint et al. 2001) and
monstrated that insulin treatment exerts a osseointegration of titanium implants in suppresses cell-growth rates (Terada et al.
preventive effect on the pathophysiology of experimental diabetes (Fiorellini et al. 1998). Suppressed Runx2 mRNA levels
osseous tissue observed in streptozotocin- 1999; Siqueira et al. 2003). have been observed following short-term
induced experimental diabetes (Hough Qualitative histological observations exposure to hyperglycaemic conditions,
et al. 1981; Hou et al. 1991; Suzuki et al. during the early stages of osseous healing, although not consistently observed in longer
2003) and in spontaneous experimental indicated similar intramembranous and en- term studies (Botolin et al. 2005; Botolin &
diabetes (Verhaeghe et al. 1997). The tim- dochondral bone formation potential in the McCabe 2007). In addition, prolonged expo-
ing of insulin treatment is considered cri- periosteal and gap callus in well-controlled sure of osteoblastic cells to hyperglycaemic
tical. Early initiation of insulin therapy in insulin-treated diabetic animals compared conditions is correlated with altered gene
experimental diabetes can prevent the ex- with non-diabetic controls at 2, 4 and 7 expression, characterised by decreased osteo-
pression of the diabetic skeletal phenotype, days of fracture healing. The authors sug- calcin, MMP-13, collagen-1 and c-jun
whereas, in later stages, insulin delivery gested that these findings support the view mRNA levels (Zayzafoon et al. 2002; Boto-
can only partly normalise parameters char- that insulin plays a critical role during the lin & McCabe 2006). Various potential
acterising the diabetic bone pathology early stages of fracture healing (Beam et al. pathogenetic mechanisms may account for
(Hough et al. 1981). It has been suggested 2002). Follak et al. (2005) further provided the responses of osteoblastic cells to pro-
that insulin normalises diabetic bone me- detailed histometrical data indicating longed hyperglycaemia, including modula-
tabolism by enhancing bone formation that, in spontaneously diabetic animals, tion of the redox state, increased activity of
and/or by preventing bone resorption (Dixit insulin-mediated strict glycaemic control the polyol-pathway, activation of the protein
& Ekstrom 1980; Haffner & Bauer 1993; normalised all dynamic parameters of kinase C pathway, and non-enzymatic gly-
Krakauer et al. 1995). mineralisation, apposition, osteoid forma- cosylation of key proteins, e.g. collagen type
Deficient osseous healing in experimen- tion and timing of mineralisation compared I and IGF-1 (McCabe 2007).
tal diabetes is also reversible via insulin with the poorly compensated diabetic ani- The accumulation of advanced glycation
treatment, because a series of in vivo stu- mals, during the first 4 weeks of femoral end-products (AGEs) in the osseous tissue
dies indicate that the histomorphometric, fracture healing. Beam et al. (2002) inves- as a result of non-enzymatic glycosylation
biomechanical and biochemical indices tigated the impact of insulin therapy on the has been implicated in the pathogenesis of
during osseous healing in experimentally cellular proliferation rates within the frac- diminished bone formation, in a fracture-
diabetic animals with insulin-mediated, ture callus in experimental diabetes. They healing model in experimental diabetes
good metabolic control are comparable showed that, during the first week of (Santana et al. 2003). AGEs may directly
to healthy controls (Goodman & Hori fracture healing, the cellular proliferation modulate the growth, differentiation and
1984; Hou et al. 1993). Specifically, it values in the fracture callus of insulin- activity of osteoblastic cells and these effects

c 2010 John Wiley & Sons A/S


 677 | Clin. Oral Impl. Res. 21, 2010 / 673–681
Retzepi & Donos  The effect of diabetes mellitus on osseous healing

are possibly mediated via their interaction bone pathophysiology are hyperglycaemia further increased the callus content and
with specific osteoblastic receptors (RAGEs) and/or hypoinsulinaemia (McCabe 2007). mineralisation, significantly improving the
(McCarthy et al. 2001; Santana et al. 2003; fracture gap bridging. The authors suggested
Schwartz 2003; Kume et al. 2005). One Hypoinsulinaemia that insulin plays a critical role in directly
possible pathway is that AGE–RAGE inter- Insulin appears to exert a direct anabolic promoting the fracture healing potential and
actions may result in generation of reactive effect on the bone formation process during that impaired diabetic osseous healing may
oxygen intermediates and/or nuclear factor osseous healing in vivo (Cornish et al. be associated with reduced local insulin
k B (NFkB) activation in osteoblastic cells 1996). In vitro studies have indicated that levels, as a result of decreased systemic
(Lalla et al. 1998; Kislinger et al. 1999; insulin exerts direct anabolic effects on insulin levels in the diabetic state.
McCarthy et al. 2001). Another plausible osteoblastic cells, evidenced by increased
mechanism is that AGEs modification of proliferation rates (Hashizume & Yamagu-
type-I collagen impairs integrin-mediated chi 1993), collagen synthesis (Craig et al. Conclusions
adhesion of osteoblasts on the extracellular 1989), alkaline phosphatase production
matrix and therefore compromises their (Canalis 1983) and glucose uptake (Hahn Diabetes mellitus has been consistently
normal bone-forming activity (McCarthy et al. 1988; Ituarte et al. 1989). Possible associated with deficient metabolism of
et al. 2001; 2004). direct actions of insulin on bone cells may the skeletal tissue, which, especially in
include mitogenic stimulation of bone- T1DM, is primarily related to suppressed
Hypercalciuria, neuropathy and angiopathy forming cells, coupled with inhibition of osteoblastic activity and reduced bone for-
Hypercalciuria resulting from increased apoptosis (Thrailkill et al. 2005). In addi- mation potential, irrespectively of the type
glycosuria has been proposed as a mechan- tion to its direct effects on cells of the of bone, the location and the mechanical
ism for diabetic osteopathy, although the osseous tissue, insulin may also synergis- loading characteristics. Clinical and in vivo
results are not consistent among different tically enhance the response of bone cells studies have further established that im-
studies and the effect cannot be accounted to other anabolic growth factors for osseous paired intramembranous and endochondral
for by the magnitude of the changes ob- tissue, such as IGF-I and PTH (Conover ossification constitute dominant pathophy-
served (Carnevale et al. 2004). Altered et al. 1996; Suzuki et al. 2003). It may be siological traits characterising diabetic bone
systemic excretion profile of calcium- therefore suggested that, because insulin disease. Nonetheless, well-designed, epide-
regulating hormones, such as parathormon constitutes a well-documented anabolic miological studies reporting on the preva-
and 25-hydroxyvitamin D has been further factor for osseous tissue, T1DM-associated lence, incidence and type of osseous
suggested to play a role in diabetic bone insulinopenia may, at least partly, account healing complications in large populations
phenotype (Alrefai et al. 2002), although for the observed bone loss and impaired of diabetic patients are warranted. Sys-
this mechanism may not account for the osseous healing potential. temic insulin treatment can promote the
regional pattern of the changes observed Although it has been documented that intramembranous and endochondral oss-
(Ahmad et al. 2004). abnormal bone repair in experimental dia- eous healing potential in diabetes, which
Neuropathy and angiopathy have been betes is insulin dependent because the may be associated with its direct mito-
suggested as the main features behind local deficient osseous healing process is re- genic, stimulatory and anti-apoptotic
alterations of diabetic bone metabolism. versed by insulin treatment, it is at present effects on osteoblasts and with its synergis-
Peripheral neuropathy is considered as an unclear whether this effect is primarily due tic effect with other growth factors.
independent risk factor for bone mineral to the insulin deficiency or to the hyper- Several plausible molecular mechanisms
density reduction among type 1 diabetics glycaemia characterising the diabetic status have been implicated in the impaired dia-
(Piepkorn et al. 1997; Rix et al. 1999), as it (Thrailkill et al. 2005). It has been advo- betic bone healing and are mainly related to
may exert direct effects on bone through cated that the relative contribution of in- suppressed differentiation, proliferation
neurotransmitter action on bone cells sulinopenia may be more significant than and/or bone forming capacity of osteoblas-
(Bjurholm et al. 1992; Lerner 2002). Alter- moderate hyperglycaemia, as normalisa- tic cells during the critical early phases of
natively, diabetic osteopenia may result tion of diabetic fracture parameters can be the osseous healing process. The currently
from disturbed vasoregulation caused by achieved with insulin treatment, despite available evidence seems to support that
neuropathy (Rajbhandari et al. 2002). suboptimal metabolic control (Gooch the main mechanisms underlying diabetic
Although increased bone resorption has et al. 2000). Gandhi et al. (2005) evaluated bone pathophysiology are hyperglycaemia
been observed in the presence of abnormal the direct effects of local insulin delivery on and/or hypoinsulinaemia in the case of
blood flow in an experimental T2DM diabetic bone repair, in the absence of T1DM. Future studies should be directed
model (Laroche 2002), the osteoporotic systemic metabolic alterations. Intrame- at dissecting the roles of hyperglycaemia
changes preceded the onset of microvascu- dullary placed pellets of sustained insulin and hypoinsulinaemia on the pathogenesis
lar changes, thus indicating that microan- release at femoral fracture sites in sponta- of the impaired bone healing in the diabetic
giopathy did not play a major role in the neously diabetic rats normalised cellular status and at implementing the acquired
pathogenesis of osteopenia (Amir et al. proliferation in the periosteal and gap knowledge for the design of novel tissue
2002). In conclusion, the currently avail- callus, as well as chondrogenesis, within engineering and drug delivery therapeutic
able evidence seems to support that the the early diabetic fracture callus. During strategies for the reconstruction of osseous
main mechanisms underlying diabetic later healing stages, insulin treatment defects in diabetic patients.

678 | Clin. Oral Impl. Res. 21, 2010 / 673–681 c 2010 John Wiley & Sons A/S

Retzepi & Donos  The effect of diabetes mellitus on osseous healing

References

Accursi, G.E. (2000) Treatment outcomes with Conover, C.A., Lee, P.D., Riggs, B.L. & Powell, Gandhi, A., Beam, H.A., O’Connor, J.P., Parsons,
osseointegrated Brånemark implants in diabetic D.R. (1996) Insulin-like growth factor-binding J.R. & Lin, S.S. (2005) The effects of local insulin
patients: a retrospective study. Thesis, University protein-1 expression in cultured human bone delivery on diabetic fracture healing. Bone 37:
of Toronto, Toronto, ON. cells: regulation by insulin and glucocorticoid. 482–490.
Ahmad, T., Ugarph-Morawski, A., Li, J., Bilevi- Endocrinology 137: 3295–3301. Gebauer, G.P., Lin, S.S., Beam, H.A., Vieira, P. &
ciute-Ljungar, I., Finn, A., Ostenson, C.G. & Cornish, J., Callon, K.E. & Reid, I.R. (1996) Insulin Parsons, J.R. (2002) Low-intensity pulsed ultra-
Kreicbergs, A. (2004) Bone and joint neuropathy increases histomorphometric indices of bone for- sound increases the fracture callus strength in
in rats with type-2 diabetes. Regulatory Peptides mation in vivo. Calcified Tissue International 59: diabetic BB Wistar rats but does not affect cellular
119: 61–67. 492–495. proliferation. Journal of Orthopaedic Research
Alrefai, H., Allababidi, H., Levy, S. & Levy, J. Cozen, L. (1972) Does diabetes delay fracture heal- 20: 587–592.
(2002) The endocrine system in diabetes mellitus. ing? Clinical Orthopaedics and Related Research Gerritsen, M., Lutterman, J.A. & Jansen, J.A. (2000)
Endocrine 18: 105–119. 82: 134–140. Wound healing around bone-anchored percuta-
Amir, G., Rosenmann, E., Sherman, Y., Greenfeld, Craig, R.G., Rowe, D.W., Petersen, D.N. & Kream, neous devices in experimental diabetes mellitus.
Z., Ne’eman, Z. & Cohen, A.M. (2002) Osteo- B.E. (1989) Insulin increases the steady state level Journal of Biomedical Materials Research 53:
porosis in the Cohen diabetic rat: correlation of alpha-1(I) procollagen mRNA in the osteoblast- 702–709.
between histomorphometric changes in bone and rich segment of fetal rat calvaria. Endocrinology Giglio, M.J., Giannunzio, G., Olmedo, D. &
microangiopathy. Laboratory Investigation 82: 125: 1430–1437. Guglielmotti, M.B. (2000) Histomorphometric
1399–1405. Dixit, P.K. & Ekstrom, R.A. (1980) Decreased study of bone healing around laminar implants
Balint, E., Szabo, P., Marshall, C.F. & Sprague, S.M. breaking strength of diabetic rat bone and its in experimental diabetes. Implant Dentistry 9:
(2001) Glucose-induced inhibition of in vitro bone improvement by insulin treatment. Calcified 143–149.
mineralization. Bone 28: 21–28. Tissue International 32: 195–199. Gooch, H.L., Hale, J.E., Fujioka, H., Balian, G. &
Beam, H.A., Parsons, J.R. & Lin, S.S. (2002) The Einhorn, T.A., Boskey, A.L., Gundberg, C.M., Vig- Hurwitz, S.R. (2000) Alterations of cartilage and
effects of blood glucose control upon fracture orita, V.J., Devlin, V.J. & Beyer, M.M. (1988) The collagen expression during fracture healing in
healing in the BB Wistar rat with diabetes melli- mineral and mechanical properties of bone in experimental diabetes. Connective Tissue Re-
tus. Journal of Orthopaedic Research 20: 1210– chronic experimental diabetes. Journal of Ortho- search 41: 81–91.
1216. paedic Research 6: 317–323. Goodman, W.G. & Hori, M.T. (1984) Diminished
Bibbo, C., Lin, S.S., Beam, H.A. & Behrens, F.F. Fiorellini, J.P., Chen, P.K., Nevins, M. & Nevins, bone formation in experimental diabetes. Rela-
(2001) Complications of ankle fractures in dia- M.L. (2000) A retrospective study of dental im- tionship to osteoid maturation and mineraliza-
betic patients. The Orthopedic Clinics of North plants in diabetic patients. International Journal tion. Diabetes 33: 825–831.
America 32: 113–133. of Periodontics and Restorative Dentistry 20: Gunczler, P., Lanes, R., Paz-Martinez, V., Martins,
Bjurholm, A., Kreicbergs, A., Schultzberg, M. & 366–373. R., Esaa, S., Colmenares, V. & Weisinger, J.R.
Lerner, U.H. (1992) Neuroendocrine regulation Fiorellini, J.P., Nevins, M.L., Norkin, A., Weber, H.P. (1998) Decreased lumbar spine bone mass and low
of cyclic AMP formation in osteoblastic cell lines & Karimbux, N.Y. (1999) The effect of insulin bone turnover in children and adolescents with
(UMR-106-01, ROS 17/2.8, MC3T3-E1, and therapy on osseointegration in a diabetic rat model. insulin dependent diabetes mellitus followed long-
Saos-2) and primary bone cells. Journal of Bone Clinical Oral Implants Research 10: 362–368. itudinally. Journal of Pediatric Endocrinology and
and Mineral Research 7: 1011–1019. Folk, J.W., Starr, A.J. & Early, J.S. (1999) Early Metabolism 11: 413–419.
Botolin, S., Faugere, M.C., Malluche, H., Orth, M., wound complications of operative treatment of Haffner, S.M. & Bauer, R.L. (1993) The association
Meyer, R. & McCabe, L.R. (2005) Increased bone calcaneus fractures: analysis of 190 fractures. of obesity and glucose and insulin concentrations
adiposity and peroxisomal proliferator-activated Journal of Orthopaedic Trauma 13: 369–372. with bone density in premenopausal and postme-
receptor-gamma2 expression in type I diabetic Follak, N., Kloting, I. & Merk, H. (2005) Influence nopausal women. Metabolism 42: 735–738.
mice. Endocrinology 146: 3622–3631. of diabetic metabolic state on fracture heating in Hahn, T.J., Westbrook, S.L., Sullivan, T.L., Good-
Botolin, S. & McCabe, L.R. (2006) Chronic hyper- spontaneously diabetic rats. Diabetes-Metabo- man, W.G. & Halstead, L.R. (1988) Glucose
glycemia modulates osteoblast gene expression lism Research and Reviews 21: 288–296. transport in osteoblast-enriched bone explants:
through osmotic and non-osmotic pathways. Jour- Follak, N., Kloting, I., Wolf, E. & Merk, H. (2004a) characterization and insulin regulation. Journal
nal of Cellular Biochemistry 99: 411–424. Histomorphometric evaluation of the influence of of Bone and Mineral Research 3: 359–365.
Botolin, S. & McCabe, L.R. (2007) Bone loss and the diabetic metabolic state on bone defect healing Hashizume, M. & Yamaguchi, M. (1993) Stimula-
increased bone adiposity in spontaneous and phar- depending on the defect size in spontaneously tory effect of beta-alanyl-L-histidinato zinc on cell
macologically induced diabetic mice. Endocrinol- diabetic BB/OK rats. Bone 35: 144–152. proliferation is dependent on protein synthesis in
ogy 148: 198–205. Follak, N., Kloting, I., Wolf, E. & Merk, H. (2004b) osteoblastic MC3T3-E1 cells. Molecular and Cel-
Bouillon, R. (1991) Diabetic bone disease. Calcified Improving metabolic control reverses the histo- lular Biochemistry 122: 59–64.
Tissue International 49: 155–160. morphometric and biomechanical abnormalities Heap, J., Murray, M.A., Miller, S.C., Jalili, T. &
Canalis, E. (1983) Effect of hormones and growth of an experimentally induced bone defect in spon- Moyer-Mileur, L.J. (2004) Alterations in bone
factors on alkaline phosphatase activity and col- taneously diabetic rats. Calcified Tissue Interna- characteristics associated with glycemic control
lagen synthesis in cultured rat calvariae. Metabo- tional 74: 551–560. in adolescents with type 1 diabetes mellitus. The
lism 32: 14–20. Fowlkes, J.L., Bunn, R.C., Liu, L., Wahl, E.C., Journal of Pediatrics 144: 56–62.
Carnevale, V., Romagnoli, E. & D’Erasmo, E. Coleman, H.N., Cockrell, G.E., Perrien, D.S., Herbsman, H., Powers, J.C., Hirschman, A. &
(2004) Skeletal involvement in patients with dia- Lumpkin, C.K. Jr & Thrailkill, K.M. (2008) Shaftan, G.W. (1968) Retardation of fracture heal-
betes mellitus. Diabetes/Metabolism Research Runt-related transcription factor 2 (RUNX2) and ing in experimental diabetes. The Journal of Sur-
and Reviews 20: 196–204. RUNX2-related osteogenic genes are down-regu- gical Research 8: 424–431.
Cloos, C., Wahl, P., Hasslacher, C., Traber, L., lated throughout osteogenesis in type 1 diabetes Hie, M., Shimono, M., Fujii, K. & Tsukamoto, I.
Kistner, M., Jurkuhn, K. & Schmidt-Gayk, H. mellitus. Endocrinology 149: 1697–1704. (2007) Increased cathepsin K and tartrateresistant
(1998) Urinary glycosylated, free and total pyridi- Funk, J.R., Hale, J.E., Carmines, D., Gooch, H.L. & acid phosphatase expression in bone of streptozo-
noline and free and total deoxypyridinoline in Hurwitz, S.R. (2000) Biomechanical evaluation of tocin-induced diabetic rats. Bone 41: 1045–1050.
diabetes mellitus. Clinical Endocrinology 48: early fracture healing in normal and diabetic rats. Horcajada-Molteni, M.N., Chanteranne, B., Lebec-
317–323. Journal of Orthopaedic Research 18: 126–132. que, P., Davicco, M.J., Coxam, V., Young, A. &

c 2010 John Wiley & Sons A/S


 679 | Clin. Oral Impl. Res. 21, 2010 / 673–681
Retzepi & Donos  The effect of diabetes mellitus on osseous healing

Barlet, J.P. (2001) Amylin and bone metabolism implications for the pathogenesis of accelerated Moy, P.K., Medina, D., Shetty, V. & Aghaloo, T.L.
in streptozotocin-induced diabetic rats. Journal of periodontal disease in diabetes. Annals of Perio- (2005) Dental implant failure rates and associated
Bone and Mineral Research 16: 958–965. dontology 3: 13–19. risk factors. The International Journal of Oral &
Hou, J.C., Zernicke, R.F. & Barnard, R.J. (1991) Laroche, M. (2002) Intraosseous circulation from Maxillofacial Implants 20: 569–577.
Experimental diabetes, insulin treatment, and physiology to disease. Joint Bone Spine 69: 262– Nevins, M.L., Karimbux, N.Y., Weber, H.P.,
femoral neck morphology and biomechanics in 269. Giannobile, W.V. & Fiorellini, J.P. (1998)
rats. Clinical Orthopaedics and Related Research Lerner, U.H. (2002) Neuropeptidergic regulation of Wound healing around endosseous implants
264: 278–285. bone resorption and bone formation. Journal of in experimental diabetes. The International Jour-
Hou, J.C., Zernicke, R.F. & Barnard, R.J. (1993) Musculoskeletal and Neuronal Interactions 2: nal of Oral & Maxillofacial Implants 13: 620–
Effects of severe diabetes and insulin on the 440–447. 629.
femoral neck of the immature rat. Journal of Lettgen, B., Hauffa, B., Mohlmann, C., Jeken, C. & Olmos, J.M., Perez-Castrillon, J.L., Garcia, M.T.,
Orthopaedic Research 11: 263–271. Reiners, C. (1995) Bone mineral density in chil- Garrido, J.C., Amado, J.A. & Gonzalez-Macias, J.
Hough, S., Avioli, L.V., Teitelbaum, S.L. & Fallon, dren and adolescents with juvenile diabetes: se- (1994) Bone densitometry and biochemical bone
M.D. (1981) Alkaline phosphatase activity in lective measurement of bone mineral density of remodeling markers in type 1 diabetes mellitus.
chronic streptozotocin-induced insulin deficiency trabecular and cortical bone using peripheral quan- Bone and Mineral 26: 1–8.
in the rat: effect of insulin replacement. Metabo- titative computed tomography. Hormone Re- Ottoni, C.E. & Chopard, R.P. (2004) Histomorpho-
lism 30: 1190–1194. search 43: 173–175. metric evaluation of new bone formation in dia-
Ituarte, E.A., Halstead, L.R., Iida-Klein, A., Ituarte, Loder, R.T. (1988) The influence of diabetes melli- betic rats submitted to insertion of temporary
H.G. & Hahn, T.J. (1989) Glucose transport tus on the healing of closed fractures. Clinical implants. Brazilian Dental Journal 15: 87–92.
system in UMR-106-01 osteoblastic osteosar- Orthopaedics and Related Research 232: 210– Papa, J., Myerson, M. & Girard, P. (1993) Salvage,
coma cells: regulation by insulin. Calcified Tissue 216. with arthrodesis, in intractable diabetic neuro-
International 45: 27–33. Lu, H., Kraut, D., Gerstenfeld, L.C. & Graves, D.T. pathic arthropathy of the foot and ankle. The
Iyama, S., Takeshita, F., Ayukawa, Y., Kido, M.A., (2003) Diabetes interferes with the bone forma- Journal of Bone and Joint Surgery. American
Suetsugu, T. & Tanaka, T. (1997) A study of the tion by affecting the expression of transcription volume 75: 1056–1066.
regional distribution of bone formed around hy- factors that regulate osteoblast differentiation. Pedrazzoni, M., Ciotti, G., Pioli, G., Girasole, G.,
droxyapatite implants in the tibiae of streptozoto- Endocrinology 144: 346–352. Davoli, L., Palummeri, E. & Passeri, M. (1989)
cin-induced diabetic rats using multiple Macey, L.R., Kana, S.M., Jingushi, S., Terek, R.M., Osteocalcin levels in diabetic subjects. Calcified
fluorescent labeling and confocal laser scanning Borretos, J. & Bolander, M.E. (1989) Defects of Tissue International 45: 331–336.
microscopy. Journal of Periodontology 68: 1169– early fracture-healing in experimental diabetes. Piepkorn, B., Kann, P., Forst, T., Andreas, J., Pfutz-
1175. The Journal of Bone and Joint Surgery. American ner, A. & Beyer, J. (1997) Bone mineral density
Janghorbani, M., Van Dam, R.M., Willett, W.C. & volume 71: 722–733. and bone metabolism in diabetes mellitus. Hor-
Hu, F.B. (2007) Systematic review of type 1 and Margonar, R., Sakakura, C.E., Holzhausen, M., mone and Metabolic Research 29: 584–591.
type 2 diabetes mellitus and risk of fracture. Pepato, M.T., Alba, R.C. & Marcantonio, E. Rajbhandari, S.M., Jenkins, R.C., Davies, C. &
American Journal of Epidemiology 166: 495–505. (2003) The influence of diabetes mellitus and Tesfaye, S. (2002) Charcot neuroarthropathy in
Kawaguchi, H., Kurokawa, T., Hanada, K., Hiyama, insulin therapy on biomechanical retention diabetes mellitus. Diabetologia 45: 1085–1096.
Y., Tamura, M., Ogata, E. & Matsumoto, T. around dental implants: a study in rabbits. Retzepi, M. (2009) The effect of experimental dia-
(1994) Stimulation of fracture repair by recombi- Implant Dentistry 12: 333–339. betes on Guided Bone Regeneration. PhD thesis,
nant human basic fibroblast growth factor in Mathiassen, B., Nielsen, S., Ditzel, J. & Rodbro, P. Eastman Dental Institute, University College
normal and streptozotocin-diabetic rats. Endocri- (1990) Long-term bone loss in insulin dependent London.
nology 135: 774–781. diabetes mellitus. Journal of Internal Medicine Retzepi, M., Lewis, M.P. & Donos, N. (2010) Effect
Kemink, S.A., Hermus, A.R., Swinkels, L.M., Lut- 227: 325–327. of diabetes and metabolic control on de novo bone
terman, J.A. & Smals, A.G. (2000) Osteopenia in McCabe, L.R. (2007) Understanding the pathology formation following Guided Bone Regeneration.
insulin-dependent diabetes mellitus; prevalence and mechanisms of type I diabetic bone loss. Clinical Oral Implants Research 21: 71–79.
and aspects of pathophysiology. Journal of Endo- Journal of Cellular Biochemistry 102: 1343– Rix, M., Andreassen, H. & Eskildsen, P. (1999)
crinological Investigation 23: 295–303. 1357. Impact of peripheral neuropathy on bone density
Kislinger, T., Fu, C., Huber, B., Qu, W., Taguchi, McCarthy, A.D., Etcheverry, S.B., Bruzzone, L., in patients with type 1 diabetes. Diabetes Care
A., Du, Y.S., Hofmann, M., Yan, S.F., Pischets- Lettieri, G., Barrio, D.A. & Cortizo, A.M. (2001) 22: 827–831.
rieder, M., Stern, D. & Schmidt, A.M. (1999) Non-enzymatic glycosylation of a type I collagen Saito, M., Fujii, K., Mori, Y. & Marumo, K. (2006)
N(epsilon)-(carboxymethyl)lysine adducts of pro- matrix: effects on osteoblastic development and Role of collagen enzymatic and glycation induced
teins are ligands for receptor for advanced glyca- oxidative stress. BMC Cell Biology 2: 16. cross-links as a determinant of bone quality in
tion end products that activate cell signaling McCarthy, A.D., Uemura, T., Etcheverry, S.B. & spontaneously diabetic WBN/Kob rats. Osteo-
pathways and modulate gene expression. The Cortizo, A.M. (2004) Advanced glycation end- porosis International 17: 1514–1523.
Journal of Biological Chemistry 274: 31740– products interfere with integrin-mediated osteo- Salerno, M., Argenziano, A., Di Maio, S., Gasparini,
31749. blastic attachment to a type-I collagen matrix. N., Formicola, S., De Filippo, G. & Tenore, A.
Krakauer, J.C., McKenna, M.J., Buderer, N.F., Rao, The International Journal of Biochemistry and (1997) Pubertal growth, sexual maturation, and
D.S., Whitehouse, F.W. & Parfitt, A.M. (1995) Cell Biology 36: 840–848. final height in children with IDDM. Effects of age
Bone loss and bone turnover in diabetes. Diabetes McCracken, M., Lemons, J.E., Rahemtulla, F., at onset and metabolic control. Diabetes Care 20:
44: 775–782. Prince, C.W. & Feldman, D. (2000) Bone re- 721–724.
Kume, S., Kato, S., Yamagishi, S., Inagaki, Y., Ueda, sponse to titanium alloy implants placed in dia- Santana, R.B. & Trackman, P.C. (2006) Controlled
S., Arima, N., Okawa, T., Kojiro, M. & Nagata, betic rats. The International Journal of Oral & release of fibroblast growth factor 2 stimulates
K. (2005) Advanced glycation end-products at- Maxillofacial Implants 15: 345–354. bone healing in an animal model of diabetes
tenuate human mesenchymal stem cells and pre- Mombelli, A. & Cionca, N. (2006) Systemic dis- mellitus. The International Journal of Oral &
vent cognate differentiation into adipose tissue, eases affecting osseointegration therapy. Clinical Maxillofacial Implants 21: 711–718.
cartilage, and bone. Journal of Bone and Mineral Oral Implants Research 17 (Suppl. 2): 97–103. Santana, R.B., Xu, L., Chase, H.B., Amar, S.,
Research 20: 1647–1658. Morris, H.F., Ochi, S. & Winkler, S. (2000) Implant Graves, D.T. & Trackman, P.C. (2003) A role
Lalla, E., Lamster, I.B. & Schmidt, A.M. (1998) survival in patients with type 2 diabetes: place- for advanced glycation end products in diminished
Enhanced interaction of advanced glycation end ment to 36 months. Annals of Periodontology 5: bone healing in type 1 diabetes. Diabetes 52:
products with their cellular receptor RAGE: 157–165. 1502–1510.

680 | Clin. Oral Impl. Res. 21, 2010 / 673–681 c 2010 John Wiley & Sons A/S

Retzepi & Donos  The effect of diabetes mellitus on osseous healing

Schwartz, A.V. (2003) Diabetes mellitus: does it image processing study. Journal of Periodontology Valerio, G., del Puente, A., Esposito-del Puente, A.,
affect bone? Calcified Tissue International 73: 69: 314–320. Buono, P., Mozzillo, E. & Franzese, A. (2002) The
515–519. Terada, M., Inaba, M., Yano, Y., Hasuma, T., lumbar bone mineral density is affected by long-
Shernoff, A.F., Colwell, J.A. & Bingham, S.F. (1994) Nishizawa, Y., Morii, H. & Otani, S. (1998) term poor metabolic control in adolescents with
Implants for type II diabetic patients: interim Growth-inhibitory effect of a high glucose con- type 1 diabetes mellitus. Hormone Research 58:
report. VA implants in diabetes study group. centration on osteoblast-like cells. Bone 22: 17– 266–272.
Implant Dentistry 3: 183–185. 23. Verhaeghe, J., Suiker, A.M., Einhorn, T.A.,
Shires, R., Teitelbaum, S.L., Bergfeld, M.A., Fallon, Thaller, S.R., Lee, T.J., Armstrong, M., Tesluk, H. Geusens, P., Visser, W.J., Van Herck, E., Van
M.D., Slatopolsky, E. & Avioli, L.V. (1981) The & Stern, J.S. (1995) Effect of insulin-like growth Bree, R., Magitsky, S. & Bouillon, R. (1994)
effect of streptozotocin-induced chronic diabetes factor type 1 on critical-size defects in diabetic Brittle bones in spontaneously diabetic female
mellitus on bone and mineral homeostasis in the rats. Journal of Craniofacial Surgery 6: 218– rats cannot be predicted by bone mineral measure-
rat. The Journal of Laboratory and Clinical Med- 223. ments: studies in diabetic and ovariectomized
icine 97: 231–240. Thrailkill, K.M., Lumpkin, C.K. Jr, Bunn, R.C., rats. Journal of Bone and Mineral Research 9:
Siqueira, J.T., Cavalher-Machado, S.C., Arana-Cha- Kemp, S.F. & Fowlkes, J.L. (2005) Is insulin an 1657–1667.
vez, V.E. & Sannomiya, P. (2003) Bone formation anabolic agent in bone? Dissecting the diabetic Verhaeghe, J., Van Herck, E., Van Bree, R., Moer-
around titanium implants in the rat tibia: role of bone for clues. American Journal of Physiology, mans, K. & Bouillon, R. (1997) Decreased osteo-
insulin. Implant Dentistry 12: 242–251. Endocrinology and Metabolism 289: E735–E745. blast activity in spontaneously diabetic rats.
Smith, R.A., Berger, R. & Dodson, T.B. (1992) Risk The Expert Committee on the Diagnosis and Clas- In vivo studies on the pathogenesis. Endocrine
factors associated with dental implants in healthy sification of Diabetes Mellitus. (1997) Report of 7: 165–175.
and medically compromised patients. The Inter- the Expert Committee on the Diagnosis and Verhaeghe, J., Van Herck, E., Visser, W.J., Suiker,
national Journal of Oral & Maxillofacial Im- Classification of Diabetes Mellitus. Diabetes A.M., Thomasset, M., Einhorn, T.A., Faierman,
plants 7: 367–372. Care 20: 1183–1197. E. & Bouillon, R. (1990) Bone and mineral meta-
Spanheimer, R.G., Umpierrez, G.E. & Stumpf, V. Tisdel, C.L., Marcus, R.E. & Heiple, K.G. (1995) bolism in BB rats with long-term diabetes. De-
(1988) Decreased collagen production in diabetic Triple arthrodesis for diabetic peritalar neuroar- creased bone turnover and osteoporosis. Diabetes
rats. Diabetes 37: 371–376. thropathy. Foot Ankle International 16: 332– 39: 477–482.
Stuart, M.J. & Morrey, B.F. (1990) Arthrodesis of 338. Vestergaard, P. (2007) Discrepancies in bone mineral
the diabetic neuropathic ankle joint. Clinical Topping, R.E., Bolander, M.E. & Balian, G. (1994) density and fracture risk in patients with type 1
Orthopaedics and Related Research 253: 209– Type X collagen in fracture callus and the effects and type 2 diabetes – a meta-analysis. Osteoporo-
211. of experimental diabetes. Clinical Orthopaedics sis International 18: 427–444.
Suzuki, K., Miyakoshi, N., Tsuchida, T., Kasu- and Related Research 308: 220–228. Weiss, R.E. & Reddi, A.H. (1980) Influence
kawa, Y., Sato, K. & Itoi, E. (2003) Effects of Tyndall, W.A., Beam, H.A., Zarro, C., O’Connor, of experimental diabetes and insulin on matrix
combined treatment of insulin and human para- J.P. & Lin, S.S. (2003) Decreased platelet derived induced cartilage and bone differentiation.
thyroid hormone(1-34) on cancellous bone mass growth factor expression during fracture healing American Journal of Physiology 238: E200–
and structure in streptozotocin-induced diabetic in diabetic animals. Clinical Orthopaedics and E207.
rats. Bone 33: 108–114. Related Research 408: 319–330. Zayzafoon, M., Botolin, S. & McCabe, L.R. (2002)
Takeshita, F., Murai, K., Iyama, S., Ayukawa, Y. & Umpierrez, G.E., Zlatev, T. & Spanheimer, R.G. P38 and activating transcription factor-2 involve-
Suetsugu, T. (1998) Uncontrolled diabetes hinders (1989) Correction of altered collagen metabolism ment in osteoblast osmotic response to elevated
bone formation around titanium implants in rat in diabetic animals with insulin therapy. Matrix extracellular glucose. The Journal of Biological
tibiae. A light and fluorescence microscopy, and 9: 336–342. Chemistry 277: 37212–37218.

c 2010 John Wiley & Sons A/S


 681 | Clin. Oral Impl. Res. 21, 2010 / 673–681

You might also like