U.S. Department of Veterans Affairs

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

U.S.

Department of Veterans Affairs


Public Access Author manuscript
Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.
Published in final edited form as:
Osteoarthritis Cartilage. 2015 June ; 23(6): 841–850. doi:10.1016/j.joca.2015.03.031.
VA Author Manuscript

The adverse effects of diabetes on osteoarthritis: update on


clinical evidence and molecular mechanisms
K.B. King†,‡,1 and A.K. Rosenthal§,‖,*
†Department of Orthopaedics, University of Colorado School of Medicine, Aurora, CO, USA
‡SurgicalService, Orthopaedic Service, Eastern Colorado Health Care System, Veterans Affairs,
Denver, CO, USA
§Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
‖Medicine Service, Rheumatology Service, The Clement J. Zablocki Medical Center, Veterans
Affairs, Milwaukee, WI, USA
VA Author Manuscript

SUMMARY
Projected increases in the prevalence of both diabetes mellitus (DM) and osteoarthritis (OA)
ensure their common co-existence. In an era of increasing attention to personalized medicine,
understanding the influence of common comorbidities such as DM should result in improved care
of patients with OA. In this narrative review, we summarize the literature addressing the
interactions between DM and OA spanning the years from 1962 to 2014. We separated studies
depending on whether they investigated clinical populations, animal models, or cells and tissues.
The clinical literature addressing the influence of DM on OA and its therapeutic outcomes
suggests that DM may augment the development and severity of OA and that DM increases risks
associated with joint replacement surgery. The few high quality studies using animal models also
support an adverse effect of DM on OA. We review strengths and weaknesses of the common
rodent models of DM. The heterogeneous literature derived from studies of articular cells and
tissues also supports the existence of biochemical and biomechanical changes in articular tissues
VA Author Manuscript

in DM, and begins to characterize molecular mechanisms activated in diabetic-like environs which
may contribute to OA. Increasing evidence from the clinic and the laboratory supports an adverse
effect of DM on the development, severity, and therapeutic outcomes for OA. To understand the
mechanisms through which DM contributes to OA, further studies are clearly necessary. Future
studies of DM-influenced mechanisms may shed light on general mechanisms of OA pathogenesis
and result in more specific and effective therapies for all OA patients.

*
Address correspondence and reprint requests to: A.K. Rosenthal, Zablocki VA Medical Center, 5000 W. National Avenue,
Milwaukee, WI 53295-1000, USA. Tel: 1-(414)-955-7027; Fax: 1-(414)-955-6205.
1Department of Orthopaedics, University of Colorado School of Medicine, 12800 E. 19th Avenue, RC1 North, Room 2101, Mail Stop
8343, Aurora, CO 80045, USA. Tel: 1-(303)-724-1596; Fax: 1-(303)-724-0394.
Conflicts of interest
No authors have any conflicts of interest related to this work.
Author contributions
Both authors contributed substantially to the conception and design of this work, participated actively in writing the manuscript, and
approved the final version. Both Dr. King (Karen.King@UCDenver.edu) and Dr. Rosenthal (arosenthal@mcw.edu) take full
responsibility for the integrity of the work as a whole.
King and Rosenthal Page 2

Keywords
Osteoarthritis; Diabetes; Hyperglycemia

Introduction
VA Author Manuscript

Diabetes mellitus (DM) and osteoarthritis (OA) are common diseases that are predicted to
increase in prevalence in the US and worldwide1,2. Because of the resultant rise in the
coexistence of these two diseases and a burgeoning body of evidence suggesting that DM
may adversely affect articular tissues and exacerbate OA, it seems an opportune time to
review literature in this area and consider ways in which these diseases interact. The ultimate
goal of this field of study is to understand OA in the diabetic patient in order to individualize
therapies and prognosis for patients having both of these common diseases.

DM is defined by metabolic abnormalities resulting from dysfunction in the glucose-


handling machinery of the body. It occurs in two major forms. Type 1 DM (T1DM) is
caused by reduction in quantities of insulin which regulates glucose metabolism. It is the
most common form of DM in children and may have an autoimmune or post-infectious
etiology. Although type 2 DM (T2DM) may occur in children, it comprises the vast majority
VA Author Manuscript

of adult-onset DM. T2DM typically involves insulin resistance at the cellular level. The
metabolic abnormalities of DM are shared by all forms of the disease; effective reduction of
insulin activity causes prolonged hyperglycemia which leads to osmotic and oxidative stress
and results in damage to the kidneys, eyes, nerves, and other tissues. By the year 2035, the
number of adults with DM worldwide is estimated to be 592 million1.

Like DM, OA is also rising in prevalence and affects millions2. It is typically defined as a
degenerative articular process characterized by eroded articular cartilage, altered
subchondral and peri-cartilagenous bone, mild to moderate synovial inflammation, and pain.
While cartilage damage is the final common endpoint in OA, primary abnormalities of other
tissues such as tendon, bone, or muscle may contribute to or even cause OA. DM, especially
T2DM, and OA share many epidemiologic features. They are both complex diseases with
considerable clinical heterogeneity and multifactorial etiologies involving interactions of
genetic and environmental factors. They also share common risk factors. Aging is a risk
VA Author Manuscript

factor for both T2DM and OA. The prevalence in the US of DM is 3.3/1000 among
individuals aged 18–44, and rises to 15.4/1000 for ages 65–793. Similarly, OA dramatically
increases with age, affecting 13.5% of adults 25 years and older and 33.6% of those over the
age of 654. Another important risk factor for both diseases is obesity. The association of OA
with obesity is well-supported5, and obesity occurs in the majority of people with T2DM6.

OA and DM frequently co-exist simply by chance due to their high prevalence and shared
risk factors. Nearly half (47.3%) of patients with DM have some form of arthritis7. The
presence of co-morbid conditions typically increases the care needs of individual patients,
decreases the effectiveness of care, and escalates health–related costs. In addition,
therapeutic strategies that emphasize personalized medicine and take into account co-morbid
conditions may result in improved outcomes for patients with OA8. The development of OA
may also complicate DM. While not the focus of this review, there is increasing evidence

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 3

that OA adds to the burden of cardiovascular disease, which is higher than average in DM
patients9.

For this narrative review, we have searched the literature using combinations of the
following key words: “osteoarthritis”, “diabetes”, “hyperglycemia”, “articular cartilage”,
“bone”, “tendon” and “ligament”. We critically assess the literature exploring the effects of
VA Author Manuscript

DM on both OA and its management strategies. The few clinical studies addressing this
issue strongly support an association between DM and OA. While animal studies are also
quite sparse and suffer from some methodological weaknesses, they also suggest that DM
may accelerate OA progression. Further, current evidence derived from basic science studies
suggests that DM may adversely affect the homeostasis and repair of articular tissues. These
research studies give credibility to the hypothesis that DM has clinically relevant effects on
OA and warrant further study.

Clinical studies
Population-based studies of DM and OA
There are only a handful of studies that directly address the effects of DM on OA. Two
cross-sectional population-based studies using questionnaires found that DM was
VA Author Manuscript

significantly more common in an OA cohort than a non–OA cohort10,11. In contrast, another


study found no increase in DM among OA patients; however, the study subjects’ healthier
lifestyle may have influenced the results12. The observation that specific features of OA may
be more common with DM – such as increased enthesophytes or more severe cartilage
degeneration – supports an interaction between these diseases13,14.

Multiple studies have used rates of arthroplasty as a surrogate for OA severity in examining
the role of DM in OA. In a cohort of patients undergoing arthroplasty for OA, no
relationship was found between T2DM and bilateral OA or generalized OA after adjustment
for age, sex, BMI, and other confounders for OA15. However, the study design based on
cohort analysis of arthroplasty patients limits the conclusions. This is due to two issues; 1) a
bias effect since OA severe enough to require arthroplasty eliminates potential study subjects
with early stage OA, and 2) a survivor effect since uncontrolled diabetes may have been a
cause to deter study subjects from arthroplasty.
VA Author Manuscript

To avoid these effects, two more recent studies examine a cohort of DM patients with non-
DM patients as control16,17. The first study used the Veterans Affairs Health Administration
(VHA) national administrative database, and DM was defined as having two DM-related in-
or out-patient visits or being prescribed anti-diabetic medication16. The cohorts included
450,000 with DM and over 3 million without DM. Rates of total joint arthroplasty were
increased in patients with DM. The statistically significant odds ratios ranged from 1.2
(primary total knee) to 3.4 (revision total hip). When these data were stratified by age, the
highest risk ratios for each joint were found in the youngest age group studied (46–55 years
of age). The major advantage of this study was its size, while the major disadvantage was
that data on body mass index (BMI) were not available. Overall, if one uses arthroplasty as a
surrogate for severe OA, this study suggests that DM has a negative effect on OA by either
increasing its prevalence or by increasing the rate of progression16.

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 4

A similar but smaller study17, analyzed retrospective data from a prospective study with data
collected over 20 years from a regional hospital in Northern Italy. Of the 927 subjects that
began the prospective study, 69 met criteria for T2DM, defined as having a fasting blood
glucose level over 126 mg/dL or a clinical diagnosis with use of anti-DM medication. The
authors found a significant and positive association of DM with arthroplasty; relative risk
VA Author Manuscript

was about two and remained significant in models that controlled for age, sex, and BMI.
Using a subset of the same population (347 total, 43 with T2DM), the authors found worse
functional measures in the T2DM group. In particular, the pain variable was significantly
higher in both the KOOS (Knee Injury and Osteoarthritis Outcome Score) and the WOMAC
(Western Ontario and McMaster Universities Osteoarthritis Index) questionnaires. In
addition, knee synovitis was significantly more common in people with DM. The major
advantages of this study were that known risk factors for OA, particularly BMI, were
controlled in the statistical models and that the characteristics of OA (pain, function, and
synovitis) were further probed. The disadvantage of this study was its small population size.

Despite the differences in approach, these two population-based studies both found that DM
doubles the risk for arthroplasty16,17. The VHA database was large enough to determine that
persons with DM have arthroplasty at younger ages16; while the smaller single hospital
study identified that the hazard risk associated with DM remained after adjustment for other
VA Author Manuscript

predictors for OA17. Obviously, arthroplasty is not a perfect outcome measure in OA, and
additional clinical studies comparing individual outcome measures such as pain, function, or
structural damage will be necessary in the future.

Other clinical studies of DM and OA


Several recent studies began to approach the role of DM in contributing to important
outcome measures in OA, including OA progression and pain. The effect of DM on OA
progression was recently studied in a cohort of 559 subjects with well-characterized knee
OA18. At baseline, 6.6% of the participants had T2DM. In men with T2DM, annualized joint
space narrowing was statistically significantly higher than in non-diabetic men matched for
BMI, age, hypertension and dyslipidemia. Of all the components of the metabolic syndrome,
only DM was identified as an independent risk factor for knee OA progression. In a study of
530 subjects with radiographic hand OA, primary analysis showed no clear association
between pain and DM19. Subset analysis, however, showed higher pain scores in patients
VA Author Manuscript

with DM who had erosive OA defined as radiographic evidence of more than one IP joint
with erosions. The impact of these conclusions is uncertain since the field does not have
standard classification criteria for erosive OA. Certainly, further validation of these findings
in OA is warranted.

Effect of DM on OA clinical care


The impact of DM on clinical care has been studied frequently, with particular attention paid
to the effects of DM on arthroplasty outcomes. Arthroplasty patients with DM sampled from
the Nationwide Inpatient Sample database have increased postsurgical death20,21. In
contrast, older patients in the Medicare database had no increased risk of postsurgical death
with DM22,23. Several studies have identified decreased functional outcomes for arthroplasty
performed on patients with DM24,25. The most commonly reported DM-associated

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 5

complication is the increased rate of infection21,22,25–29. DM may also increase the need for
revision arthroplasty25,30,31. Some arthroplasty outcomes may pertain to metabolic effects
on joint tissues such as defective bone healing, while others reflect general risks of DM
associated with major surgical procedures. All of these effects can add considerable cost to
clinical care20,21,32.
VA Author Manuscript

Effect of DM on human articular tissues


Studies of tissues from patients demonstrate that changes capable of promoting OA occur in
articular tissues in patients with DM. For example, ankle cartilage from DM patients was
softer with lower stiffness indices and larger permeability parameters than cartilage from
non-diabetic patients33. Despite the limitations of this study – including the use of frozen
tissues, the focus on a joint infrequently involved in OA, and the significant age difference of
control subjects – the observed changes in biomechanical properties of articular cartilage
might contribute to the development of OA. Advanced glycation endproducts (AGE) may
accumulate prematurely in diabetic tissues and affect biomechanics34–37. A single, small
study using human tissue showed increased levels of the AGE pentosidine in bone from
patients with DM at the time of joint replacement, but no significant changes in pentosidine
levels in cartilage38. Tendon abnormalities may also contribute to OA. DM patients
demonstrate biochemical and biomechanical changes in tendons that include decreased
VA Author Manuscript

rupture threshold and disordered collagen fibrils39. There is also mounting evidence that
bone healing is defective in DM patients40–46. Increased rates of non-healing microfractures
in patients with DM may alter bone mechanics and promote OA, and they may also
contribute to poor arthroplasty outcomes. Multiple clinical studies demonstrate increased
fracture risk in post-menopausal women with T2DM which is not simply linked to low bone
mineral density on clinical densitometry41,42. This paradox may be explained by changes in
bone microarchitecture, including increased cortical porosity43, excess mineralization, and
reduced subchondral bone heterogeneity44 with DM. Patients with DM have reduced serum
biomarkers of bone turnover as well as elevated sclerostin levels45,46. Such changes could
lead to increased bone stiffness which could accelerate OA.

Animal studies
The presence of multiple comorbidities and other inherent challenges in designing clinical
VA Author Manuscript

studies to delineate mechanisms responsible for increased OA in DM patients have logically


led to the use of animal models of these diseases. While primate and rabbit models of DM
have been described47–49, rodents are most commonly used to replicate human DM50,51.
Very few studies directly address the severity or progression of OA in diabetic animals.

Animal models of T1DM


In T1DM, insulin production is absent or significantly impaired52. Genetic models of T1DM
such as the Akita mouse (C57BL/6-Ins2Akita) are available on several background
strains53,54. A non-genetic and very common method of modeling T1DM is to chemically
damage the pancreatic islets with single or multiple dose regimens of streptozotocin
(STZ)55–57. There are a few rat models of T1DM including the LEW.1AR1/-iddm50 and the
BBDP rat58 both of which involve spontaneous autoimmune pancreatic damage.

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 6

Animal models of T2DM


In order to model human T2DM, the animal should have hyperglycemia non-fasting blood
glucose >250 mg/dL50,51,59,60, insulin insensitivity, and preferably additional characteristics
such as polyuria and polydipsia49,50. Further information on protocols for characterizing
DM in rodents can be found in the literature61,62. However, as with models of T1DM, no
VA Author Manuscript

mouse model perfectly mirrors human T2DM disease. The relative contributions of obesity
to acceleration of OA in DM are particularly important in T2DM and its models. While
isolating the biomechanical effects of obesity from the effects of DM is itself a challenge, in
addition, obesity has well-known metabolic consequences that add further complexity to the
situation. These include increased production of inflammatory cytokines such as IL-1,
TNFα, and adipokines (e.g., adiponectin, leptin, and resistin)63 that promote inflammation
and may accelerate OA. Because obesity and T2DM almost always co-exist in human
T2DM, animal studies may be particularly useful in separating the pathological
contributions derived from these different mechanisms.

A popular model of T2DM is the diet-induced obesity model (DIO) which attempts to
simulate human obesity-induced T2DM but often results in only modestly increased glucose
levels51,64–66. The db/db mouse, ob/ob mouse, and fa/fa rat have monogenetic defects that
disrupt leptin signaling resulting in hyperphagia and morbid obesity67. This leptin signaling
VA Author Manuscript

defect is rare in human T2DM60,64,67, and T2DM models created using polygenic mutations
are better models of human T2DM. The KK.Cg-Ay/J mouse was developed by crossing a
spontaneously diabetic strain (KK) with the yellow obese strain (Ay) resulting in a mature-
onset T2DM phenotype68. Both DM and control normal glycemic siblings are obese69. The
NONcNZO10/LtJ mouse (NcZ10) is a polygenic model of T2DM with onset of DM in
adulthood. The NcZ10 model requires a chow content of 10–11% fat for higher penetrance
(90–100%)59. The TALLYHO/JngJ mouse (TH) has polygenic and maturity-onset diabetes
and has high penetrance in males59,70. A short list of commonly available rodent models of
T1DM and T2DM is presented in Table I. Readers are encouraged to review additional
details in the references provided.

Animal studies investigating a link between DM and OA


There is an unfortunate paucity of quality research studies in animal models of DM relevant
VA Author Manuscript

to OA. A recent study using STZ-induced T1DM in mice suggested the presence of cartilage
damage after 8 weeks of hyperglycemia, and showed elevated levels of circulating AGEs73.
Both abnormalities were ameliorated by the use of the DM drug pioglitazone. The authors
concluded that this drug response implicated PPARγ in this effect, but it was unclear
whether this was related to improvement of hyperglycemia or PPARγ inhibition. One
valuable study applied the DIO model to the C57Bl/6 strain and made careful measures of
physiological data and histological OA outcomes74. This study combined a high fat diet
(60% by calorie) with the meniscal ligament injury model to induce OA. Higher OA scores
(increased joint degeneration) were found in the mouse group receiving both high-fat diet
and ligament injury. However, since hyperglycemia was not established until the last month
of the experiment, it is unclear whether the hyperadiposity75 or the rising hyperglycemia was
the driving factor for greater OA progression. None the less, this is a valuable study

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 7

demonstrating acceleration of joint degeneration in association with metabolic changes


typically seen in DM patients.

Effects of DM and hyperglycemia on articular cartilage


Biomechanical properties of cartilage are highly influenced by the composition of the
VA Author Manuscript

extracellular matrix (ECM), and there is some evidence that metabolic abnormalities
associated with DM alter cartilage ECM. Early studies in animal models of DM have shown
that decreased collagen production76 and increased proteoglycan catabolism77 occurs in DM
cartilage. Increased proteoglycan catabolism in DM animals has also been observed in non-
articular connective tissues78.

Effects of DM and hyperglycemia on bone


Studies showing delayed fracture healing in animals with DM support the ample clinical
data associating DM with bone abnormalities. The STZ mouse model demonstrates that poor
diabetic fracture healing is related to premature resorption of cartilage at the fracture
callus79, and that this was due to high levels of the inflammatory cytokine TNFα in mice
with DM80.

Effects of DM and hyperglycemia on tendons and ligaments


VA Author Manuscript

Animal models of DM fairly consistently show histologic and biochemical abnormalities in


tendons and ligaments, as well as less well-characterized biomechanical changes, such as
lower Young’s modulus and increased intra-substance failure81–83. Further, several studies
identify delayed tendon healing after injury in the presence of DM84–87. These types of
tendon and ligament abnormalities are known to promote OA88.

Effects of DM and hyperglycemia on synovium


The synovium of T1DM rats is abnormal and contains fibrotic tissue with higher amounts of
type I collagen and lower quantities of types III and V collagen89. Synovial pathology is a
significant contributing factor to OA; early inflammatory changes in the synovium may
cause damage that then creates long-term production of catabolic mediators90.

Potential mechanisms
VA Author Manuscript

Basic science studies have also identified some potential mechanisms linked to DM-
influenced end-organ joint damage. Mediators such as hyperglycemia, AGEs, sorbitol,
adipokines, and cytokines act through oxidative, osmotic, and inflammatory mechanisms to
produce tissue damage (Table II). Further complexity is added by the participation of similar
molecular participants in multiple catabolic pathways and the contributions of similar
metabolic derangements in obesity.

An increasing recognition of a key role for inflammation in both OA98 and DM provides an
important mechanistic link between these two conditions. Significant synovitis occurs in OA
and may be exacerbated by the increased levels of inflammatory cytokines, adipokines, and
prostaglandins seen in DM tissues63. Signaling through pathways of innate immunity, such
as toll-like receptors, also may produce inflammation in both DM92 and OA99.

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 8

In a hyperglycemic environment, there is increased production of reactive oxygen species


contributing to tissue damage. Cellular transport of glucose becomes critical and may
contribute to excess oxidative stress. One group found that human OA chondrocytes from
donors aged ≥66 years cultured in high glucose are unable to decrease GLUT1 protein or
reduce glucose transport activity in comparison with normal chondrocytes from young
donors (age 28–35 years)94,100. They also found that high glucose conditions favored
VA Author Manuscript

production of oxidants and promoted matrix catabolism which would accelerate OA.
However, age and media osmolarity were not controlled in those experiments101. The effects
of high glucose may be associated with impaired function of ATP-sensitive K+ channels
which couple GLUT to intracellular ATP/ADP levels and membrane potential102,103.

The AGE/RAGE system also plays a role in DM end-organ damage through induction of
inflammation and/or increased oxidative stress. Collagen has an extraordinary low turnover
in many connective tissues and as such is prone to modification by AGEs. The formation of
AGEs is accelerated by high tissue levels of glucose38. AGEs signal through RAGE
(receptor for AGEs) and other receptors to produce various deleterious effects on
chondrocytes including inflammation and cytokine-mediated catabolism104–106 and have
been postulated to play a role in end-organ damage in DM38. Further, AGE mediated cross-
linking of collagen can alter a tissue’s biomechanical properties as shown for cartilage and
VA Author Manuscript

tendon35–37. Cross-linking by AGEs may also inhibit ECM turnover by restricting access to
proteolytic sites106. On the other hand, a recent study in dogs suggests that an artificial
increase in AGE levels alone using repeated ribose injections did not accelerate OA in a mild
injury model107, but we know little about the effect of AGEs in the context of the diabetic
milieu. Thus, whether or how AGE’s play a significant role in OA remains unclear.

In the polyol pathway, aldose reductase converts glucose to sorbitol and galactose to
galactitol. This mechanism is activated in DM with excess polyols leading to cellular
osmotic stress108. Although not yet directly linked to OA, there is some evidence that this
pathway is activated in DM in intervertebral disc cartilage and enhances matrix catabolism
via p38 MAPK activation109.

Although not covered in detail in this review, additional DM-relevant pathways have been
proposed. For example, there is considerable evidence that adipokines may induce
VA Author Manuscript

inflammation and have adverse effects on cartilage75,110 and tissue healing111. Because
altered adipokine levels are seen in obesity in both the absence and presence of diabetes112,
the contribution of adipokines to OA in obese patients with DM will require further study.
Alteration in angiogenesis, autophagy, and apoptosis are also associated with end-organ
damage in OA58,113–116. Insulin receptors are present on chondrocytes100, and thus, excess
insulin as seen in T2DM patients may also damage cartilage. In one study, PPARγ
downregulation was shown to occur in articular chondrocytes exposed to high glucose
media, but methodologic challenges warrant confirmation of this finding73. Whether one or
more pathways are involved, which pathway is most relevant, and how molecular mediators
intersect multiple pathways will require additional study.

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 9

Conclusions
In summary, increasing evidence from the clinic and the laboratory supports an adverse
effect of DM on the development, severity, and therapeutic outcomes for OA. The etiology
and clinical manifestations of OA are complex, and currently we know little about how the
multiple mechanisms altered in DM may affect OA that originates from different causes.
VA Author Manuscript

Further, the clinical impact of DM on OA may be underestimated by the high prevalence of


undiagnosed diabetes3. A deeper understanding of OA in the setting of DM could result in
significant improvements in clinical care. For example, reducing OA-related joint pain may
allow DM patients to perform the exercise necessary for cardiovascular health. A full
appreciation of these disease interactions may also reduce the increased medical costs
associated with arthroplasty and other surgeries in patients with comorbid DM20,21,32. To
understand the mechanisms through which DM contributes to OA, further work is clearly
necessary. Future studies of DM-influenced mechanisms may shed light on the general
mechanisms of OA pathogenesis and result in more specific and effective therapies for all
OA patients.

Acknowledgments
We would like to thank the VA Research Service for research space and support (AKR, 101BX000812), an OREF/
VA Author Manuscript

Goldberg Arthritis Research Grant (Bucknell and King), and Dr Frank Beier for his thoughtful comments and
suggestions on this review.

References
1. Guariguata L, Whiting DR, Hambleton I, Beagley J, Linnenkamp U, Shaw JE. Global estimates of
diabetes prevalence for 2013 and projections for 2035. Diabetes Res Clin Pract. 2014; 103:137–49.
[PubMed: 24630390]
2. Vos T, Flaxman AD, Naghavi M, Lozano R, Michaud C, Ezzati M, et al. Years lived with disability
(YLDs) for 1160 sequelae of 289 diseases and injuries 1990–2010: a systematic analysis for the
Global Burden of Disease Study. Lancet. 2010; 2012(380):2163–96.
3. CDC. National Diabetes Statistics Report: Estimates of Diabetes and its Burden in the United States.
US Department of Health and Human Services; 2014.
4. Lawrence RC, Felson DT, Helmick CG, Arnold LM, Choi H, Deyo RA, et al. Estimates of the
prevalence of arthritis and other rheumatic conditions in the United States. Part II. Arthritis Rheum.
2008; 58:26–35. [PubMed: 18163497]
VA Author Manuscript

5. Visser, AW., de Mutsert, R., le Cessie, S., den Heijer, M., Rosendaal, FR., Kloppenburg, M. The
relative contribution of mechanical stress and systemic processes in different types of osteoarthritis:
the NEO study. Ann Rheum Dis. 2014 May 20. pii. annrheumdis 2013-205012, http://dx.doi.org/
10.113/annrheumdis-2013-205012, (In press, epub ahead of print)
6. Teodoro JS, Varela AT, Rolo AP, Palmeira CM. High-fat and obesogenic diets: current and future
strategies to fight obesity and diabetes. Genes Nutr. 2014; 9:406. [PubMed: 24842072]
7. CDC. Prevalence of doctor-diagnosed arthritis and arthritis-attributable activity limitation – United
States, 2010–2012. MMWR Morb Mortal Wkly Rep. 2013; 62:869–73. [PubMed: 24196662]
8. Gierisch JM, Myers ER, Schmit KM, McCrory DC, Coeytaux RR, Crowley MJ, et al. Prioritization
of patient-centered comparative effectiveness research for osteoarthritis. Ann Intern Med. 2014;
160:836–41. [PubMed: 24821227]
9. Rahman MM, Kopec JA, Anis AH, Cibere J, Goldsmith CH. Risk of cardiovascular disease in
patients with osteoarthritis: a prospective longitudinal study. Arthritis Care Res (Hoboken). 2013;
65:1951–8. [PubMed: 23925995]

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 10

10. Hart DJ, Doyle DV, Spector TD. Association between metabolic factors and knee osteoarthritis in
women: the Chingford study. J Rheumatol. 1995; 22:1118–23. [PubMed: 7674240]
11. Dahaghin S, Bierma-Zeinstra SMA, Koes BW, Hazes JMW, Pols HAP. Do metabolic factors add to
the effect of over-weight on hand osteoarthritis? The Rotterdam study. Ann Rheum Dis. 2007;
66:916–20. [PubMed: 17314121]
12. Frey MI, Barrett-Connor E, Sledge PA, Schneider DL, Weisman MH. The effect of noninsulin
dependent diabetes mellitus on the prevalence of clinical osteoarthritis. A population based study. J
VA Author Manuscript

Rheumatol. 1996; 23:716–22. [PubMed: 8730133]


13. Gibson N, Guermazi A, Clancy M, Niu J, Grayson P, Aliabadi P, et al. Relation of hand
enthesophytes with knee enthesopathy: is osteoarthritis related to a systemic enthesopathy? J
Rheumatol. 2012; 39:359–64. [PubMed: 22174209]
14. Jungmann PM, Kraus MS, Alizai H, Nardo L, Baum T, Nevitt MC, et al. Association of metabolic
risk factors with cartilage degradation assessed by T2 relaxation time at the knee: data from the
osteoarthritis initiative. Arthritis Care Res (Hoboken). 2013; 65:1942–50. [PubMed: 23926027]
15. Sturmer T, Brenner H, Brenner RE, Gunther KP. Non-insulin dependent diabetes mellitus
(NIDDM) and patterns of osteoarthritis. The Ulm osteoarthritis study. Scand J Rheumatol. 2001;
30:169–71. [PubMed: 11469529]
16. King KB, Findley TW, Williams AE, Bucknell AL. Veterans with diabetes receive arthroplasty
more frequently and at a younger age. Clin Orthop Relat Res. 2013; 471:3049–54. [PubMed:
23649224]
17. Schett G, Kleyer A, Perricone C, Sahinbegovic E, Iagnocco A, Zwerina J, et al. Diabetes is an
independent predictor for severe osteoarthritis: results from a longitudinal cohort study. Diabetes
Care. 2013; 36:403–9. [PubMed: 23002084]
VA Author Manuscript

18. Eymard F, Parsons C, Edwards MH, Petit-Dop F, Reginster JY, Bruyère O, et al. Diabetes is a risk
factor for knee osteoarthritis progression. Osteoarthritis Cartilage. 2015 S1063-4584(15)00026-6,
In press.
19. Magnusson K, Hagen KB, Østerås N, Nordsletten L, Natvig B, Haugen IK. Diabetes is associated
with increased hand pain in erosive hand osteoarthritis: data from a population-based study.
Arthritis Care Res. 2015; 67:187–95.
20. Bolognesi MP, Marchant MH Jr, Viens NA, Cook C, Pietrobon R, Vail TP. The impact of diabetes
on perioperative patient outcomes after total hip and total knee arthroplasty in the United States. J
Arthroplasty. 2008; 23:92–8. [PubMed: 18722309]
21. Marchant MH Jr, Viens NA, Cook C, Vail TP, Bolognesi MP. The impact of glycemic control and
diabetes mellitus on perioperative outcomes after total joint arthroplasty. J Bone Joint Surg Am.
2009; 91:1621–9. [PubMed: 19571084]
22. Bozic KJ, Lau E, Kurtz S, Ong K, Berry DJ. Patient-related risk factors for postoperative mortality
and periprosthetic joint infection in medicare patients undergoing TKA. Clin Orthop Relat Res.
2012; 470:130–7. [PubMed: 21874391]
23. Bozic KJ, Lau E, Kurtz S, Ong K, Rubash H, Vail TP, et al. Patient-related risk factors for
VA Author Manuscript

periprosthetic joint infection and postoperative mortality following total hip arthroplasty in
medicare patients. J Bone Joint Surg Am. 2012; 94:794–800. [PubMed: 22552668]
24. Fisher DA, Dierckman B, Watts MR, Davis K. Looks good but feels bad: factors that contribute to
poor results after total knee arthroplasty. J Arthroplasty. 2007; 22:39–42.
25. Meding JB, Reddleman K, Keating ME, Klay A, Ritter MA, Faris PM, et al. Total knee
replacement in patients with diabetes mellitus. Clin Orthop Relat Res. 2003; 416:208–16.
26. Jämsen E, Nevalainen P, Kalliovalkama J, Moilanen T. Preoperative hyperglycemia predicts
infected total knee replacement. Eur J Intern Med. 2010; 21:196–201. [PubMed: 20493422]
27. Jämsen E, Nevalainen P, Eskelinen A, Huotari K, Kalliovalkama J, Moilanen T. Obesity, diabetes,
and preoperative hyperglycemia as predictors of periprosthetic joint infection: a single-center
analysis of 7181 primary hip and knee replacements for osteoarthritis. J Bone Joint Surg Am.
2012; 94:e101. [PubMed: 22810408]
28. Namba RS, Inacio MC, Paxton EW. Risk factors associated with deep surgical site infections after
primary total knee arthroplasty: an analysis of 56,216 knees. J Bone Joint Surg Am. 2013; 95:775–
82. [PubMed: 23636183]

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 11

29. Mraovic B, Hipszer BR, Epstein RH, Pequignot EC, Parvizi J, Joseph JI. Preadmission
hyperglycemia is an independent risk factor for in-hospital symptomatic pulmonary embolism
after major orthopedic surgery. J Arthroplasty. 2010; 25:64–70. [PubMed: 19056217]
30. Dy CJ, Bozic KJ, Pan TJ, Wright TM, Padgett DE, Lyman S. Risk factors for early revision after
total hip arthroplasty. Arthritis Care Res (Hoboken). 2014; 66:907–15. [PubMed: 24285406]
31. Pedersen AB, Mehnert F, Johnsen SP, Sorensen HT. Risk of revision of a total hip replacement in
patients with diabetes mellitus: a population-based follow up study. J Bone Joint Surg Br. 2010;
VA Author Manuscript

92:929–34. [PubMed: 20595109]


32. Sams JD, Francis ML, Scaife SL, Robinson BS, Novicoff WM, Saleh KJ. Redefining revision total
hip arthroplasty based on hospital admission status. J Arthroplasty. 2012; 27:758–63. [PubMed:
22019324]
33. Athanasiou KA, Fleischli JG, Bosma J, Laughlin TJ, Zhu CF, Agrawal CM, et al. Effects of
diabetes mellitus on the biomechanical properties of human ankle cartilage. Clin Orthop Relat Res.
1999:182–9.
34. Saito M, Marumo K. Collagen cross-links as a determinant of bone quality: a possible explanation
for bone fragility in aging, osteoporosis, and diabetes mellitus. Osteoporos Int. 2010; 21:195–214.
[PubMed: 19760059]
35. Reddy GK. Glucose-mediated in vitro glycation modulates biomechanical integrity of the soft
tissues but not hard tissues. J Orthop Res. 2003; 21:738–43. [PubMed: 12798076]
36. Reddy GK. Cross-linking in collagen by nonenzymatic glycation increases the matrix stiffness in
rabbit Achilles tendon. Exp Diabesity Res. 2004; 5:143–53. [PubMed: 15203885]
37. Li Y, Fessel G, Georgiadis M, Snedeker JG. Advanced glycation end-products diminish tendon
VA Author Manuscript

collagen fiber sliding. Matrix Biol. 2013; 32:169–77. [PubMed: 23348249]


38. Oren TW, Botolin S, Williams A, Bucknell A, King KB. Arthroplasty in veterans: analysis of
cartilage, bone, serum, and synovial fluid reveals differences and similarities in osteoarthritis with
and without comorbid diabetes. J Rehabil Res Dev. 2011; 48:1195–210. [PubMed: 22234664]
39. de Oliveira RR, Lemos A, de Castro Silveira PV, da Silva RJ, de Moraes SR. Alterations of
tendons in patients with diabetes mellitus: a systematic review. Diabet Med. 2011; 28:886–95.
[PubMed: 21749441]
40. Seino Y, Ishida H. Diabetic osteopenia: Pathophysiology and clinical aspects. Diabetes Metab Rev.
1995; 11:21–35. [PubMed: 7600905]
41. Schwartz AV, Sellmeyer DE, Ensrud KE, Cauley JA, Tabor HK, Schreiner PJ, et al. Older women
with diabetes have an increased risk of fracture: a prospective study. J Clin Endocrinol Metab.
2001; 86:32–8. [PubMed: 11231974]
42. Janghorbani M, Van Dam RM, Willett WC, Hu FB. Systematic review of type 1 and type 2
diabetes mellitus and risk of fracture. Am J Epidemiol. 2007; 166:495–505. [PubMed: 17575306]
43. Patsch JM, Burghardt AJ, Yap SP, Baum T, Schwartz AV, Joseph GB, et al. Increased cortical
porosity in type 2 diabetic postmenopausal women with fragility fractures. J Bone Min Res. 2013;
VA Author Manuscript

28:313–24.
44. Pritchard JM, Papaioannou A, Tomowich C, Giangregorio LM, Atkinson SA, Beattie KA, et al.
Bone mineralization is elevated and less heterogeneous in adults with type 2 diabetes and
osteoarthritis compared to controls with osteoarthritis alone. Bone. 2013; 54:76–82. [PubMed:
23356988]
45. Garcia-Martin A, Rozas-Moreno P, Reyes-Garcia R, Morales-Santana S, Garcia-Fontana B,
Garcia-Salcedo JA, et al. Circulating levels of sclerostin are increased in patients with type 2
diabetes mellitus. J Clin Endocrinol Metab. 2012; 97:234–41. [PubMed: 22031520]
46. Reyes-Garcia R, Rozas-Moreno P, Lopez-Gallardo G, Garcia-Martin A, Varsavsky M, Aviles-Perez
MD, et al. Serum levels of bone resorption markers are decreased in patients with type 2 diabetes.
Acta Diabetol. 2013; 50:47–52. [PubMed: 22042129]
47. Hansen BC. Investigation and treatment of type 2 diabetes in nonhuman primates. Methods Mol
Biol. 2012; 933:177–85. [PubMed: 22893407]
48. Nadeau KJ, Ehlers LB, Aguirre LE, Moore RL, Jew KN, Ortmeyer HK, et al. Exercise training and
calorie restriction increase SREBP-1 expression and intramuscular triglyceride in skeletal muscle.
Am J Physiol Endocrinol Metab. 2006; 291:E90–8. [PubMed: 16449296]

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 12

49. Wang J, Wan R, Mo Y, Zhang Q, Sherwood LC, Chien S. Creating a long-term diabetic rabbit
model. Exp Diabetes Res. 2010; 2010:289614. [PubMed: 21234414]
50. King AJ. The use of animal models in diabetes research. Br J Pharmacol. 2012; 166:877–94.
[PubMed: 22352879]
51. Fajardo RJ, Karim L, Calley VI, Bouxsein ML. A review of rodent models of type 2 diabetic
skeletal fragility. J Bone Min Res. 2014; 29:1025–40.
VA Author Manuscript

52. Cantarelli E, Citro A, Marzorati S, Melzi R, Scavini M, Piemonti L. Murine animal models for
preclinical islet transplantation: no model fits all (research purposes). Islets. 2013; 5:79–86.
[PubMed: 23751893]
53. Wang J, Takeuchi T, Tanaka S, Kubo SK, Kayo T, Lu D, et al. A mutation in the insulin 2 gene
induces diabetes with severe pancreatic beta-cell dysfunction in the Mody mouse. J Clin Invest.
1999; 103:27–37. [PubMed: 9884331]
54. Mathews CE, Langley SH, Leiter EH. New mouse model to study islet transplantation in insulin-
dependent diabetes mellitus. Transplantation. 2002; 73:1333–6. [PubMed: 11981430]
55. Deeds MC, Anderson JM, Armstrong AS, Gastineau DA, Hiddinga HJ, Jahangir A, et al. Single
dose streptozotocin-induced diabetes: considerations for study design in islet transplantation
models. Lab Anim. 2011; 45:131–40. [PubMed: 21478271]
56. Weiss RE, Gorn AH, Nimni ME. Abnormalities in the biosynthesis of cartilage and bone
proteoglycans in experimental diabetes. Diabetes. 1981; 30:670–7. [PubMed: 7250535]
57. Spanheimer RG, Umpierrez GE, Stumpf V. Decreased collagen production in diabetic rats.
Diabetes. 1988; 37:371–6. [PubMed: 3378683]
58. Sarmiento J, Wallis RH, Ning T, Marandi L, Chao GY, Paterson AD, et al. Genetic dissection of
VA Author Manuscript

Iddm26 in the spontaneously diabetic BBDP rat. Genes Immun. 2014; 15:378–91. [PubMed:
24920533]
59. Leiter EH, Strobel M, O’Neill A, Schultz D, Schile A, Reifsnyder PC. Comparison of two new
mouse models of polygenic type 2 diabetes at the Jackson Laboratory, NON-cNZO10Lt/J and
TALLYHO/JngJ. J Diabetes Res. 2013; 2013:165327. [PubMed: 23671854]
60. Leiter EH. Selecting the “right” mouse model for metabolic syndrome and type 2 diabetes
research. Methods Mol Biol. 2009; 560:1–17. [PubMed: 19504239]
61. McGuinness OP, Ayala JE, Laughlin MR, Wasserman DH. NIH experiment in centralized mouse
phenotyping: the Vanderbilt experience and recommendations for evaluating glucose homeostasis
in the mouse. Am J Physiol Endocrinol Metab. 2009; 297:E849–55. [PubMed: 19638507]
62. Andrikopoulos S, Blair AR, Deluca N, Fam BC, Proietto J. Evaluating the glucose tolerance test in
mice. Am J Physiol Endocrinol Metab. 2008; 295:E1323–32. [PubMed: 18812462]
63. Stenholm S, Koster A, Alley DE, Visser M, Maggio M, Harris TB, et al. Adipocytokines and the
metabolic syndrome among older persons with and without obesity: the InCHIANTI study. Clin
Endocrinol (Oxf). 2010; 73:55–65. [PubMed: 19878507]
64. Muhlhausler BS. Nutritional models of type 2 diabetes mellitus. Methods Mol Biol. 2009; 560:19–
VA Author Manuscript

36. [PubMed: 19504240]


65. Ravussin E, Smith SR. Increased fat intake, impaired fat oxidation, and failure of fat cell
proliferation result in ectopic fat storage, insulin resistance, and type 2 diabetes mellitus. Ann N Y
Acad Sci. 2002; 967:363–78. [PubMed: 12079864]
66. Rossmeisl M, Rim JS, Koza RA, Kozak LP. Variation in type 2 diabetes–related traits in mouse
strains susceptible to diet-induced obesity. Diabetes. 2003; 52:1958–66. [PubMed: 12882911]
67. Wang B, Chandrasekera PC, Pippin JJ. Leptin- and leptin receptor-deficient rodent models:
relevance for human type 2 diabetes. Curr Diabetes Rev. 2014; 10:131–45. [PubMed: 24809394]
68. Iwatsuka H, Shino A, Suzuoki Z. General survey of diabetic features of yellow KK mice.
Endocrinol Jpn. 1970; 17:23–35. [PubMed: 5468422]
69. Duran M, Shum L, Clark R, McNulty M, King KB. A new animal model for determining the
effects of diabetes on osteoarthritis. Osteoarthritis Cartilage. 2014; 22:S347–8.
70. Kim JH, Sen S, Avery CS, Simpson E, Chandler P, Nishina PM, et al. Genetic analysis of a new
mouse model for non-insulin-dependent diabetes. Genomics. 2001; 74:273–86. [PubMed:
11414755]

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 13

71. Wallis RH, Wang K, Marandi L, Hsieh E, Ning T, Chao GY, et al. Type 1 diabetes in the BB rat: a
polygenic disease. Diabetes. 2009; 58:1007–17. [PubMed: 19168599]
72. Kim JH, Stewart TP, Soltani-Bejnood M, Wang L, Fortuna JM, Mostafa OA, et al. Phenotypic
characterization of polygenic type 2 diabetes in TALLYHO/JngJ mice. J Endocrinol. 2006;
191:437–46. [PubMed: 17088413]
73. Chen YJ, Chan DC, Lan KC, Wang CC, Chen CM, Chao SC, et al. PPARgamma is involved in the
hyperglycemia-induced inflammatory responses and collagen degradation in human chondrocytes
VA Author Manuscript

and diabetic mouse cartilages. J Orthop Res. 2014


74. Mooney RA, Sampson ER, Lerea J, Rosier RN, Zuscik MJ. High-fat diet accelerates progression of
osteoarthritis after meniscal/ligamentous injury. Arthritis Res Ther. 2011; 13:R198. [PubMed:
22152451]
75. Griffin TM, Huebner JL, Kraus VB, Yan Z, Guilak F. Induction of osteoarthritis and metabolic
inflammation by a very high-fat diet in mice: effects of short-term exercise. Arthritis Rheum.
2012; 64:443–53. [PubMed: 21953366]
76. Umpierrez GE, Zlatev T, Spanheimer RG. Correction of altered collagen metabolism in diabetic
animals with insulin therapy. Matrix (Stuttgart, Germany). 1989; 9:336–42.
77. Silberberg R, Hirshberg GE, Lesker P. Enzyme studies in the articular cartilage of diabetic rats and
of rats bearing transplanted pancreatic islets. Diabetes. 1977; 26:732–5. [PubMed: 142034]
78. Cohen MP, Surma ML. Effect of diabetes on in vivo metabolism of [35S]-labeled glomerular
basement membrane. Diabetes. 1984; 33:8–12. [PubMed: 6690347]
79. Kayal RA, Tsatsas D, Bauer MA, Allen B, Al-Sebaei MO, Kakar S, et al. Diminished bone
formation during diabetic fracture healing is related to the premature resorption of cartilage
VA Author Manuscript

associated with increased osteoclast activity. J Bone Min Res. 2007; 22:560–8.
80. Alblowi J, Kayal RA, Siqueria M, McKenzie E, Krothapalli N, McLean J, et al. High levels of
tumor necrosis factor-[alpha] contribute to accelerated loss of cartilage in diabetic fracture healing.
Am J Pathol. 2009; 175:1574–85. [PubMed: 19745063]
81. de Oliveira RR, Martins CS, Rocha YR, Braga AB, Mattos RM, Hecht F, et al. Experimental
diabetes induces structural, inflammatory and vascular changes of Achilles tendons. PLoS One.
2013; 8:e74942. [PubMed: 24130676]
82. Fox AJ, Bedi A, Deng XH, Ying L, Harris PE, Warren RF, et al. Diabetes mellitus alters the
mechanical properties of the native tendon in an experimental rat model. J Orthop Res. 2011;
29:880–5. [PubMed: 21246619]
83. Odetti P, Aragno I, Rolandi R, Garibaldi S, Valentini S, Cosso L, et al. Scanning force microscopy
reveals structural alterations in diabetic rat collagen fibrils: role of protein glycation. Diabetes
Metab Res Rev. 2000; 16:74–81. [PubMed: 10751746]
84. David MA, Jones KH, Inzana JA, Zuscik MJ, Awad HA, Mooney RA. Tendon repair is
compromised in a high fat diet-induced mouse model of obesity and type 2 diabetes. PLoS One.
2014; 9:e91234. [PubMed: 24658034]
VA Author Manuscript

85. Egemen O, Ozkaya O, Ozturk MB, Sen E, Akan M, Sakiz D, et al. The biomechanical and
histological effects of diabetes on tendon healing: experimental study in rats. J Hand Microsurg.
2012; 4:60–4. [PubMed: 24293952]
86. Ahmed AS, Schizas N, Li J, Ahmed M, Ostenson CG, Salo P, et al. Type 2 diabetes impairs tendon
repair after injury in a rat model. J Appl Physiol. 1985; 2012(113):1784–91.
87. Connizzo BK, Bhatt PR, Liechty KW, Soslowsky LJ. Diabetes alters mechanical properties and
collagen fiber re-alignment in multiple mouse tendons. Ann Biomed Eng. 2014; 42:1880–8.
[PubMed: 24833253]
88. Friel NA, Chu CR. The role of ACL injury in the development of posttraumatic knee osteoarthritis.
Clin Sports Med. 2013; 32:1–12. [PubMed: 23177457]
89. Atayde SA, Yoshinari NH, Nascimento DP, Catanozi S, Andrade PC, Velosa AP, et al.
Experimental diabetes modulates collagen remodelling of joints in rats. Histol Histopathol. 2012;
27:1471–9. [PubMed: 23018246]
90. Pessler F, Dai L, Diaz-Torne C, Gomez-Vaquero C, Paessler ME, Zheng DH, et al. The synovitis of
“non-inflammatory” orthopaedic arthropathies: a quantitative histological and
immunohistochemical analysis. Ann Rheum Dis. 2008; 67:1184–7. [PubMed: 18203762]

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 14

91. Cecil DL, Johnson K, Rediske J, Lotz M, Schmidt AM, Terkeltaub R. Inflammation-induced
chondrocyte hypertrophy is driven by receptor for advanced glycation end products. J Immunol.
2005; 175:8296–302. [PubMed: 16339570]
92. Rajamani U, Jialal I. Hyperglycemia induces toll-like receptor-2 and -4 expression and activity in
human microvascular retinal endothelial cells: implications for diabetic retinopathy. J Diabetes
Res. 2014; 2014:15.
93. McNulty AL, Stabler TV, Vail TP, McDaniel GE, Kraus VB. Dehydroascorbate transport in human
VA Author Manuscript

chondrocytes is regulated by hypoxia and is a physiologically relevant source of ascorbic acid in


the joint. Arthritis Rheum. 2005; 52:2676–85. [PubMed: 16142743]
94. Rosa SC, Goncalves J, Judas F, Mobasheri A, Lopes C, Mendes AF. Impaired glucose transporter-1
degradation and increased glucose transport and oxidative stress in response to high glucose in
chondrocytes from osteoarthritic versus normal human cartilage. Arthritis Res Ther. 2009; 11:R80.
[PubMed: 19490621]
95. Lin Y, Berg AH, Iyengar P, Lam TKT, Giacca A, Combs TP, et al. The hyperglycemia-induced
inflammatory response in adipocytes: the role of reactive oxygen species. J Biol Chem. 2005;
280:4617–26. [PubMed: 15536073]
96. Brocker C, Thompson DC, Vasiliou V. The role of hyperosmotic stress in inflammation and
disease. Biomol Concepts. 2012; 3:345–64. [PubMed: 22977648]
97. Chung SSM, Ho ECM, Lam KSL, Chung SK. Contribution of polyol pathway to diabetes-induced
oxidative stress. J Am Soc Nephrol. 2003; 14:S233–6. [PubMed: 12874437]
98. Scanzello CR, Umoh E, Pessler F, Diaz-Torne C, Miles T, Dicarlo E, et al. Local cytokine profiles
in knee osteoarthritis: elevated synovial fluid interleukin-15 differentiates early from end-stage
disease. Osteoarthritis Cartilage. 2009; 17:1040–8. [PubMed: 19289234]
VA Author Manuscript

99. Scanzello CR, Plaas A, Crow MK. Innate immune system activation in osteoarthritis: is
osteoarthritis a chronic wound? Curr Opin Rheumatol. 2008; 20:565–72. [PubMed: 18698179]
100. Rosa SC, Rufino AT, Judas F, Tenreiro C, Lopes MC, Mendes AF. Expression and function of the
insulin receptor in normal and osteoarthritic human chondrocytes: modulation of anabolic gene
expression, glucose transport and GLUT-1 content by insulin. Osteoarthritis Cartilage. 2011;
19:719–27. [PubMed: 21324373]
101. Rosa SC, Rufino AT, Judas FM, Tenreiro CM, Lopes MC, Mendes AF. Role of glucose as a
modulator of anabolic and catabolic gene expression in normal and osteoarthritic human
chondrocytes. J Cell Biochem. 2011; 112:2813–24. [PubMed: 21608018]
102. Rufino AT, Rosa SC, Judas F, Mobasheri A, Lopes MC, Mendes AF. Expression and function of
K(ATP) channels in normal and osteoarthritic human chondrocytes: possible role in glucose
sensing. J Cell Biochem. 2013; 114:1879–89. [PubMed: 23494827]
103. Mobasheri A. Glucose: an energy currency and structural precursor in articular cartilage and bone
with emerging roles as an extracellular signaling molecule and metabolic regulator. Front
Endocrinol (Lausanne). 2012; 3:153. [PubMed: 23251132]
104. Loeser RF, Yammani RR, Carlson CS, Chen H, Cole A, Im HJ, et al. Articular chondrocytes
VA Author Manuscript

express the receptor for advanced glycation end products: potential role in osteoarthritis. Arthritis
Rheum. 2005; 52:2376–85. [PubMed: 16052547]
105. Yammani RR, Carlson CS, Bresnick AR, Loeser RF. Increase in production of matrix
metalloproteinase 13 by human articular chondrocytes due to stimulation with S100A4: role of
the receptor for advanced glycation end products. Arthritis Rheum. 2006; 54:2901–11. [PubMed:
16948116]
106. DeGroot J, Verzijl N, Jacobs KM, Budde M, Bank RA, Bijlsma JW, et al. Accumulation of
advanced glycation end-products reduces chondrocyte-mediated extracellular matrix turnover in
human articular cartilage. Osteoarthritis Cartilage. 2001; 9:720–6. [PubMed: 11795991]
107. Vos PA, DeGroot J, Barten-van Rijbroek AD, Zuurmond AM, Bijlsma JW, Mastbergen SC, et al.
Elevation of cartilage AGEs does not accelerate initiation of canine experimental osteoarthritis
upon mild surgical damage. J Orthop Res. 2012; 30:1398–404. [PubMed: 22388985]
108. Zhang P, Xing K, Randazzo J, Blessing K, Lou MF, Kador PF. Osmotic stress, not aldose
reductase activity, directly induces growth factors and MAPK signaling changes during sugar
cataract formation. Exp Eye Res. 2012; 101:36–43. [PubMed: 22710095]

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 15

109. Cheng X, Ni B, Zhang Z, Liu Q, Wang L, Ding Y, et al. Polyol pathway mediates enhanced
degradation of extracellular matrix via p38 MAPK activation in intervertebral disc of diabetic
rats. Connect Tissue Res. 2013; 54:118–22. [PubMed: 23215968]
110. McNulty AL, Miller MR, O’Connor SK, Guilak F. The effects of adipokines on cartilage and
meniscus catabolism. Connect Tissue Res. 2011; 52:523–33. [PubMed: 21787135]
111. Wu CL, Jain D, McNeill JN, Little D, Anderson JA, Huebner JL, et al. Dietary fatty acid content
regulates wound repair and the pathogenesis of osteoarthritis following joint injury. Ann Rheum
VA Author Manuscript

Dis. 2014
112. Al-Hamodi Z, Al-Habori M, Al-Meeri A, Saif-Ali R. Association of adipokines, leptin/
adiponectin ratio and c-reactive protein with obesity and type 2 diabetes mellitus. Diabetol Metab
Syndr. 2014; 6:99. [PubMed: 25276234]
113. Tsai WC, Liang FC, Cheng JW, Lin LP, Chang SC, Chen HH, et al. High glucose concentration
up-regulates the expression of matrix metalloproteinase-9 and -13 in tendon cells. BMC
Musculoskelet Disord. 2013; 14:255. [PubMed: 23981230]
114. Jiang L, Zhang X, Zheng X, Ru A, Ni X, Wu Y, et al. Apoptosis, senescence, and autophagy in rat
nucleus pulposus cells: implications for diabetic intervertebral disc degeneration. J Orthop Res.
2013; 31:692–702. [PubMed: 23238821]
115. Won HY, Park JB, Park EY, Riew KD. Effect of hyperglycemia on apoptosis of notochordal cells
and intervertebral disc degeneration in diabetic rats. J Neurosurg Spine. 2009; 11:741–8.
[PubMed: 19951028]
116. Gazzarrini C, Stagni N, Pollesello P, D’Andrea P, De Bernard B. Possible mechanism of
inhibition of cartilage alkaline phosphatase by insulin. Acta Diabetol Lat. 1989; 26:321–7.
[PubMed: 2698041]
VA Author Manuscript
VA Author Manuscript

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


King and Rosenthal Page 16

As there is no perfect animal model that entirely recapitulates either T1DM or T2DM,
compromises are likely to be required with any one model. Ideally the selected model(s)
should be well characterized in terms of the following:

1. Level and consistency of hyperglycemia. Chemically induced models of


T1DM have been known to lose their hyperglycemic state over time due to β-
VA Author Manuscript

cell regeneration. Hyperglycemia drops among some monogenic T2DM


models.

2. Age at onset of diabetes. T2DM and OA are more common amongst older
humans, therefore the use of older animals in these research studies should be
considered and the lifespan of the model should be sufficient to allow
development of OA.

3. Sex differences. There is a gender bias in diabetes severity and age of onset In
many rodent models of diabetes.

4. Appropriateness of control group. Different background strains may have


greatly different susceptibility to obesity and change in blood glucose.

5. Comorbid conditions. It may be desirable to have present comorbid


VA Author Manuscript

conditions to answer the specific research questions. Scientists should be


aware that not all DM models have been fully characterized for comorbid
conditions.
VA Author Manuscript

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


VA Author Manuscript VA Author Manuscript VA Author Manuscript

Table I

Commonly used rodent models for type 1 and type 2 DM

Model name* (short name) DM type Preferred control Advantages Disadvantages References

C57BL/6-Ins2Akita (Akita) T1DM Same strain background Available on other backgrounds Genetic background modulates DM 50,52–54
phenotype
King and Rosenthal

Streptozotocin (STZ) T1DM Same strain with vehicle treatment Economical Drug toxicity 50,52,55
Can be used in most rodent strains
and other species
Alloxan monohydrate (Alloxan) T1DM Same strain with vehicle treatment Economical Less efficacious than STZ method 49,50,52
Can be used in most rodent strains
and other species
Biobreeding diabetes-prone rat T1DM Biobreeding diabetes-resistant rat (BBDR) Larger size (rat) Will not develop DM in a pathogen-free 58,71
(BBDP) Polygenic environment
80–95% incidence
No sex bias
Diet-induced obesity (DIO) T2DM Same strain with vehicle treatment (low fat Economical Mild or no hyperglycemia 51,64,66
chow) Can model pre-diabetes state
Works well with C56BL/6 and
NON strains
C57BLKS-Leprdb (db/db) T2DM Heterozygous (+/db) littermates One of the most studied models Monogenic 51,59,67
No sex bias Juvenile-onset
Early death
C57BLKS-Lepob (ob/ob) T2DM Heterozygous (+/ob) littermates A well-studied model Monogenic 511,67
No sex bias Mild, short-term hyperglycemia
Zucker diabetic fatty rats (ZDF) T2DM ZDF lean Larger size (rat) Monogenic 51,67
No or mild hyperglycemia
DM in males only
KK.Cg-Ay/J (KKAy) T2DM Homozygous, non-diabetic siblings Polygenic T2DM Females have lower blood glucose levels 68
Maturity onset
NONcNZO10/LtJ (NcZ10) T2DM NON/LtJ, SWR or FVB (Swiss-derived strains) Polygenic T2DM DM in males only 59,60
on low fat diet Maturity onset Requires high fat diet for high penetrance

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.


TALLYHO/JngJ (TH) T2DM NON/LtJ, SWR, or FVB (Swiss-derived Polygenic T2DM DM in males only 59,70,72
strains). Maturity onset
C57Bl/6 strain has also been used. High penetrance

*
Where relevant, the most commonly used background strain is indicated although other backgrounds may be available. DM.
Page 17
King and Rosenthal Page 18

Table II

Mechanisms present in diabetes that can damage joint tissues


VA Author Manuscript

Potential DM mechanisms Contributing pathways Examples of major Examples of References


mediators molecular
participants
Inflammation AGE/RAGE, innate immune Cytokines, Adipokines, MMPs, TLR 91,92
pathways Reactive oxygen species
Oxidative stress Glucose transporters AGE/RAGE Reactive oxygen species GLUT, ATP, ADP 93–95

Osmotic stress Polyol pathway Sorbitol p38 MAPK 96,97

AGE = advanced glycation end products, RAGE = receptor for AGE, MMP = matrix metalloproteinase, TLR = toll-like receptors, GLUT = glucose
transporter family, ATP = adenosine triphosphate, ADP = adenosine diphosphate, MAPK = mitogen-activated protein kinase.
VA Author Manuscript
VA Author Manuscript

Osteoarthritis Cartilage. Author manuscript; available in PMC 2017 July 27.

You might also like