Sei sulla pagina 1di 12

JOURNAL OF BONE AND MINERAL RESEARCH

Volume 20, Number 9, 2005


Published online on May 23, 2005; doi: 10.1359/JBMR.050514
© 2005 American Society for Bone and Mineral Research

Advanced Glycation End-Products Attenuate Human Mesenchymal


Stem Cells and Prevent Cognate Differentiation Into Adipose Tissue,
Cartilage, and Bone*
Shinichiro Kume,1,2 Seiya Kato,2 Sho-ichi Yamagishi,3 Yosuke Inagaki,3 Seiji Ueda,4 Nobuyuki Arima,2
Takahiro Okawa,1 Masamichi Kojiro,2 and Kensei Nagata1

ABSTRACT: The impact of AGEs on human MSCs was studied. AGEs inhibited the proliferation of MSCs,
induced apoptosis, and prevented cognate differentiation into adipose tissue, cartilage, and bone, suggesting
a deleterious effect of AGEs in the pathogenesis of musculoskeletal disorders in aged and diabetic patients.
Introduction: Advanced glycation end-products (AGEs) are accumulated on long-lived proteins of various
tissues in advanced age and diabetes mellitus and have been implicated in chronic complication, including
musculoskeletal disorders. Human mesenchymal stem cells (MSCs) potentially differentiate into mature mus-
culoskeletal tissues during tissue repair, but the pathogenetic role of AGEs on MSCs is unclear.
Materials and Methods: AGEs were prepared by incubating BSA with glucose, glyceraldehydes, or glycol-
aldehyde (designated as AGE-1, AGE-2, or AGE-3, respectively). Proliferation, apoptosis, and reactive
oxygen species (ROS) generation were assayed in AGE-treated cells. The expression of the receptor for AGE
(RAGE) was examined by immunohistochemistry and Western blotting. Involvement of RAGE-mediated
signaling was examined using a neutralizing antiserum against RAGE. Differentiation into adipose tissue,
cartilage, and bone were morphologically and biochemically monitored with specific markers for each.
Results: AGE-2 and AGE-3, but not control nonglycated BSA and AGE-1, reduced the viable cell number
and 5-bromo-2’deoxyuridine (BrdU) incorporation with increased intracellular ROS generation and the per-
centage of apoptotic cells. MSCs expressed RAGE and its induction was stimulated by AGE-2 and AGE-3.
These AGEs inhibited adipogenic differentiation (assayed by oil red O staining, lipoprotein lipase production,
and intracellular triglyceride content) and chondrogenic differentiation (assayed by safranin O staining and
type II collagen production). On osteogenic differentiation, AGE-2 and AGE-3 increased alkaline phospha-
tase activity and intracellular calcium content; however, von Kossa staining revealed the loss of mineralization
and mature bone nodule formation. The antiserum against RAGE partially prevented AGE-induced cellular
events.
Conclusion: AGE-2 and AGE-3 may lead to the in vivo loss of MSC mass and the delay of tissue repair by
inhibiting the maturation of MSC-derived cells. The AGE–RAGE interaction may be involved in the del-
eterious effect of AGEs on MSCs.
J Bone Miner Res 2005;20:1647–1658. Published online on May 23, 2005; doi: 10.1359/JBMR.050514

Key words: mesenchymal stem cells, advanced glycation end-products, receptor for advanced glycation end-
products, proliferation, differentiation

INTRODUCTION reversibly cross-linked and fluorescent moieties.(1–3) The


formation and accumulation of AGEs are a characteristic
A DVANCED GLYCATION END - PRODUCTS (AGEs) are
formed by reducing sugars nonenzymatically with the
amino groups of proteins to initiate a complex series of
feature of tissues in aged people, especially in patients with
diabetes mellitus, and these products have also been
rearrangements and dehydrations to produce a class of ir- strongly implicated in the pathogenesis of age-related and
diabetic complications.(4,5) It has been reported that AGEs
are involved in musculoskeletal diseases such as osteoar-
*Part of this work was presented as abstract form at the 25th
thritis (OA), which is the most common chronic disabling
Congress of the International Academy of Pathology, Brisbane,
Australia, October 10–15, 2004. disorder for aged people.(6) Accumulation of AGEs in the
The authors have no conflict of interest. articular cartilage in OA patients biochemically affects cel-

1
Department of Orthopaedic Surgery, Kurume University School of Medicine, Kurume, Japan; 2Department of Pathology, Kurume
University School of Medicine, Kurume, Japan; 3Department of Internal Medicine III and the Cardiovascular Research Institute, Kurume
University School of Medicine, Kurume, Japan; 4Department of Nephrology, Kurume University School of Medicine, Kurume, Japan.

1647
1648 KUME ET AL.

lular characteristics and increases stiffness and brittleness of Preparation of AGEs


the tissue.(7) Accumulation of AGEs increases stiffness of
the collagen network in the bone as well, which may explain AGEs were prepared as described previously.(25) Briefly,
some of the age-related increase in skeletal fragility and 50 mg/ml of BSA (Sigma) was incubated under sterile con-
fracture risk.(8) Indeed, decreased bone strength and the ditions with D-glucose for 8 weeks or 0.1 M D-glyceralde-
increased risk of fracture have been shown in human and hyde or D-glycolaldehyde in 0.2 M NaPO4 buffer (pH 7.4)
rodent models of diabetes, as well as of diabetic nephrop- for 7 days. The unincorporated sugars were removed by
athy.(9–11) In the craniotomy model in type 1 diabetic mice, dialysis against 0.2 M PBS (pH 7.4). These products of
the degree of bone healing is reported to be ∼40% of that AGE proteins were designated as AGE-1, AGE-2, or
in nondiabetic animals, and the receptor for AGEs AGE-3, respectively. Nonglycated BSA was incubated un-
(RAGE) is expressed at higher levels in the healing bone der the same conditions except for the absence of D-
tissue in diabetic than in control animals.(12) Taken to- glucose, D-glyceraldehyde, or D-glycolaldehyde as a nega-
gether, these findings suggest that AGEs may play a role in tive control. Preparations were tested for endotoxin using
the pathogenesis of diabetic osteopenia as well as that of an Endospecy ES-20S system (Seikagaku Co., Tokyo, Ja-
various bone and joint disorders in nondiabetic patients; pan), and no endotoxin was detectable.
however, the details of such deleterious mechanisms remain
Proliferation assay
to be clarified.
Mesenchymal stem cells (MSCs) are multipotent cells Total accumulation of viable cells was estimated by hand
present in various tissues during human development and counting with a hemocytometer after Trypan blue exclu-
are capable of being isolated from adult bone marrow and sion. For the measurement of proliferating cells, 5-bromo-
to be differentiating into bone, cartilage, muscle, and adi- 2’deoxyuridine (BrdU) incorporation(26) was performed us-
pose tissues.(13,14) In injured adults, MSCs are involved in ing a cell proliferation ELISA kit (Roche) following the
the formation, remodeling, and repair of musculoskeletal manufacturer’s protocol. Briefly, MSCs were seeded on a
tissues, suggesting the potential for therapeutic approaches 96-well multititer plate at 5 × 103 cells/well. After incuba-
using MSCs in orthopedic and regenerative medicine.(15,16) tion under a serum-starved condition for 24 h, the growing
In bone formation, pre-osteoblasts, osteoblasts, and osteo- medium was applied, and MSCs were further cultured for
cytes all originate from MSCs.(17) Defects in the number 24 h. At the end of the culture course, 10 ␮l/well of BrdU
and proliferative potential of MSCs have been suggested to diluted with PBS was added for 4 h. Absorbance at 450–690
underlie age-related defects in osteoblast number and func- nm was measured using a scanning multiwell spectropho-
tion.(18,19) In streptozotocin-induced diabetic rats, AGE- tometer (Eimax A-4; Fujirebio, Tokyo, Japan).
modified type I collagen dose-dependently inhibits the phe-
notypic expression of osteoblasts.(20) It has also been Semiquantitative analysis of apoptosis
reported that AGEs influence osteoblast development
The level of apoptosis was tested by the terminal deoxy-
through specific receptors, including RAGE.(21,22) In hu-
nucleotidyl transferase (TdT)-mediated deoxyuridine tri-
man articular cartilage, an increase in AGE levels nega-
phosphate (dUTP) nick end labeling (TUNEL) method.
tively affects the proteoglycan synthesis, thereby reducing
The multitude of newly generated 3⬘-OH ends by DNA
cellular turnover and repair capacity and contributing to the
fragmentation was identified visually using an in situ cell
degradation of tissue.(23) It has been reported that CD36, a
death detection kit (Roche). A positive control was estab-
cell surface glycoprotein, functions as a receptor for AGEs
lished by incubating the specimens in DNase I, RNase-free
in the adipogenic differentiation of mouse 3T3-L1 cells.(24)
(Roche) for 10 minutes before the reaction with TdT en-
These reports may suggest that AGEs have the ability to
zyme. For the assay, the cells inoculated on a coverslip were
regulate proliferation and differentiation of musculoskele-
washed with PBS, fixed in 4% paraformaldehyde in PBS,
tal lineage cells, but there have been no studies investigat-
and permeabilized with 0.2% Triton X-100 in citrate buffer.
ing whether AGEs directly affect the function of MCSs. In
Samples were incubated with TdT and fluorescein-labeled
this context, we studied the effects of AGEs on the prolif-
dUTP, counterstained with propidium iodide, and observed
eration and differentiation of cultured human MSCs in
with a fluorescence microscope (Leica). Percentages of
vitro. Here we first report that AGEs inhibited prolifera-
apoptotic cells were estimated by counting a total of 300
tion of MSCs with apoptosis and prevented cognate differ-
cells from random fields.
entiation into adipose tissue, cartilage, and bone with the
partial involvement of AGE–RAGE interaction. Measurement of intracellular reactive oxygen species
generation
MATERIALS AND METHODS
The intracellular formation of reactive oxygen species
Cell culture condition of human MSCs (ROS) was detected using a fluorescent probe CM-
Human MSCs (Poietics) and mesenchymal stem cell H2DCFDA (Molecular Probes) as described previously.(27)
growth medium (MSCGM) were purchased from BioWhit- Briefly, cells were plated at 5 × 103 cells/well in a 96-well
taker (Walkersville, MD, USA). The cells were cultured at plate. The cells were loaded with 10 ␮⌴ CM-H2DCFDA,
37°C in a humidified 5% CO2–95% air mixture and were incubated for 60 minutes at 37°C, and analyzed using an
kept at a subconfluent density. Cells passaged less than five EZS-FL fluorescent plate reader (Asahi Techno Glass, To-
times were used in this study. kyo, Japan) using the program EZScan-FL for Windows.
EFFECTS OF AGES ON MESENCHYMAL STEM CELLS 1649

Western blotting Adipogenic differentiation of MSCs


The cells were lysed in TNE buffer (20 mM Tris-HCl, pH Adipogenic differentiation was induced with commer-
7.6, 150 mM NaCl, 2 mM EDTA, 50 mM NaF, 1 mM cially available adipogenic differentiation medium (Bio-
Na3VO4, 1 mM phenylmethanesulfanyl fluoride (PMSF), 5 Whittaker). Briefly, confluent MSCs were cultured with
mM 2-mercaptoethanol, 0.8 ␮M of aprotinin, 20 ␮M of leu- medium containing 1 ␮M dexamethasone, 10 ␮g/ml bovine
peptin, 50 ␮M of pepstatin, 10 ␮M of pepstatin A, and 1% insulin, 0.2 mM indomethacin, 0.5 mM isobutylmethylxan-
NP-40) at 4°C. The cell lysate, which was determined to thine, and 10% FCS for a period of 3 days followed by the
contain 30 ␮g of protein using a Bradford protein assay kit removal of dexamethasone, indomethacin, and isobutyl-
(Bio-Rad), was further dissolved in SDS-PAGE sample methylxanthine, and additional culturing for 2 days. Adipo-
buffer (25 mM Tris-HCl, pH 6.8, 5% glycerol, 1% SDS, genic differentiation was terminated after 2–3 weeks in cul-
0.01% bromophenol blue, 50 mM DTT). The sample was ture. For the oil red O staining, cells were fixed in 10%
subsequently resolved by 12% SDS-PAGE. The gels were neutral buffered formalin, washed in water, and stained
electronically transferred to polyvinylidene fluoride with a 0.6% (w/v) oil red O solution (60% isopropanol,
(PVDF) membranes (Millipore) followed by incubation 40% water) for 1 h at room temperature, followed by wash-
with a goat polyclonal anti-RAGE antibody (clone C-20; ing with water to remove unbound dye. The concentration
Santa Cruz), at a dilution of 1:500 for 2 h at room tempera- of lipoprotein lipase (LPL) in the conditioned medium was
ture. Blots were visualized by enhanced chemiluminescence measured using an enzyme-linked immunosorbent assay
(ECL; Amersham), using horseradish peroxidase– (ELISA) kit (Daiichi Pure Chemicals, Tokyo, Japan). For
conjugated secondary antibodies (Zymed). To confirm that the measurement of intracellular triglyceride (TG) content,
equal amounts of the proteins were subjected to Western cells were scraped off into 25 mM Tris-HCl (pH 7.5) con-
blotting analysis, the membranes were reprobed with a taining 1 mM EDTA. After homogenization, TG was ex-
mouse monoclonal antibody against ␤-actin (Sigma). Den- tracted using chloroform-methanol (2:1, vol/vol) and quan-
sitometric analysis was performed with NIH image soft- tified enzymatically using a commercially available TG test
ware, and the relative ratio to ␤-actin expression was cal- kit (WAKO, Tokyo, Japan).
culated in each sample.
Chondrogenic differentiation of MSCs
Immunofluorescence microscopy
Chondrogenic differentiation was induced with a com-
MSCs were grown on the Laboratory-Tek chamber mercially available chondrogenic differentiation medium
(NUNC). At the end of the culture period, the cells were (BioWhittaker). Briefly, incomplete chondrogenic differen-
fixed with 4% paraformaldehyde in PBS (pH7.4) for 10 tiation medium (ICDM) containing 0.1 ␮M dexametha-
minutes and permeabilized with 0.2% Triton X-100. The sone, 0.17 mM ascorbic acid 2-phosphate, 6.25 ␮g/ml bovine
cells were incubated with a 1:200 dilution of polyclonal an- insulin, 6.25 ␮g/ml transferrin, and an additional 10 ng/ml of
tibody for RAGE (Santa Cruz) for 1 h, followed by incu- TGF-␤3 (PeproTech) was added for the complete chondro-
bation with a 1:1000 dilution of FITC-conjugated secondary genic differentiation medium (CCDM). MSCs suspended in
antibodies (Bio-Rad). After counterstaining with 0.1 ␮g/ml ICDM (∼7.5 × 105 cells/ml) were centrifuged at 150g for 5
of propidium iodide (Sigma), the specimens were observed
minutes, and ICDM was aspirated and replaced by CCDM.
with a fluorescence microscope (Leica). Negative control
MSCs resuspended in CCDM (5 × 105 cells/ml) were cul-
staining in which a primary antibody was omitted was also
tured in polypropylene tubes (Corning). The cell pellets
tested.
were fed every 3 days by completely replacing the CCDM
Immunoelectron microscopy and harvested after 2–4 weeks in culture. The clusters of the
cells were fixed with 10% neutral buffered formalin, and
The distribution of RAGE on the cell surface of MSCs
paraffin-embedded sections were stained with the safranin
was also examined by electron microscopy using the immu-
O method for glycosaminoglycan synthesis. Type II colla-
nogold technique. Briefly, MSCs were fixed with 4% para-
gen production into the media was measured by ELISA
formaldehyde in PBS for 1 h. After washing with PBS, cells
using a native type II collagen detection kit (Chodrex, Red-
were rinsed with 0.5% BSA and 0.02% sodium azide
mond, WA, USA).
(Sigma) for 10 minutes followed by the incubation with a
1:200 dilution of anti-RAGE antibody (Santa Cruz) for 1 h Osteogenic differentiation of MSCs
at room temperature. Positive signals were visualized using
an immunogold staining kit (EY Laboratories, San Mateo, Osteogenic differentiation was induced with a commer-
CA, USA) with 15-nm Protein A-gold colloidal particles. cially available osteogenic differentiation medium (Bio-
Labeled cells were fixed with 1% glutaraldehyde, mounted Whittaker) containing 0.1 ␮M dexamethasone, 0.05 mM
on polylysin-coated coverslips, and postfixed with 1% os- ascorbic acid 2-phosphate, and 10 nM ␤-glycerophosphate.
mium tetroxide in PBS. Samples were examined with a Hi- MSCs were seeded at 5 × 103 cells/cm2 and were cultured up
tachi S-800 field emission scanning electron microscope to 3 weeks with replacement of the fresh osteogenesis in-
equipped with a detector for backscattered electrons. duction medium every 4 days. For the detection of osteo-
genic differentiation, bone nodule formation was examined
Neutralizing antibody for RAGE under a phase-contrast microscope (Nikon) after von Kossa
Antiserum against human RAGE was prepared for neu- staining as previously described.(29) For alkaline phospha-
tralizing assays as described previously.(28) tase (ALP) activity, MSCs were washed three times with
1650 KUME ET AL.

PBS and harvested with 0.2% NP-40 (Sigma) plus 2 mM


MgCl2 in PBS. After centrifugation at 1500g for 15 minutes,
the supernatant was subjected to for ALP activity assay
with p-nitrophenol phosphate as a substrate using an
Osteolinks-BAP kit (a diagnostic enzyme immunoassay kit
for bone-specific ALP; Smitomo Pharmaceuticals Co., To-
kyo, Japan). For the measurement of intracellular calcium
content, the cells dissolved in NP-40 lysis buffer were ap-
plied to a Clinical analyzer (Hitachi), and the assay was
performed by the method of Connerty.(30) Type I collagen
production into the medium was measured using a human
type I capture ELISA kit (Chodrex).

Statistical analysis
Experimental groups were compared by ANOVA, and
when appropriate, with Scheffe’s test for multiple compari-
sons. All data are expressed as mean ± SD. A level of p <
0.05 was accepted as statistically significant.

RESULTS
Effects of AGEs on proliferation and apoptosis
of MSCs
Cultured MSCs were incubated with 100 ␮g/ml of non-
glycated BSA (as a control), AGE-1, AGE-2, or AGE-3,
which dosage has been shown to be sufficient for inducing
biological events in vascular endothelial cells(31) and retinal
pericytes.(32) The conditioned medium was changed every 3
days with fresh AGEs, and the viable cell number was
counted up to 5 days after the initial addition. As shown in
Fig. 1A, the growth curve of MSCs was shifted downwards
by the treatments with AGE-2 and AGE-3. At day 5, the FIG. 1. Growth suppression of human MSCs by AGE-2 and
AGE-3. (A) Cell counting assay. MSCs were seeded at a density
viable cell numbers were decreased by 45 ± 3.5% with of 1 × 103 cells/cm2 on 6-well culture dishes at day 0. The cells
AGE-2 (p < 0.01) and by 41 ± 7.0% with AGE-3 (p < 0.05). were cultured with MSCGM containing 100 ␮g/ml of nonglycated
Both AGE-2 and AGE-3, but not AGE-1, significantly re- BSA, AGE-1, AGE-2, or AGE-3. The cell counting was per-
duced DNA synthesis, which was measured by the BrdU formed at days 1–5. Data are shown as the mean ± SD (n ⳱ 3).
incorporation assay, in a dose-dependent fashion (Fig. 1B). (B) BrdU incorporation assay. MSCs were seeded on a 96-well
culture plate at a density of 1 × 103 cells/well. After 24 h of serum
Next, MSCs were incubated with AGEs for 3 days, and the starvation, cells were cultured with MSCGM containing 10–100
level of apoptosis was measured by semiquantitative ␮g/ml of nonglycated BSA (open bars), AGE-1 (hatched bars),
TUNEL staining. The cells treated with AGE-2 and AGE-2 (gray bars), or AGE-3 (filled bars), respectively. The val-
AGE-3 showed a dose-dependent increase in apoptotic cell ues of A450-A690 are shown as the mean ± SD (n ⳱ 6). *p < 0.05.
These data are representative of one of two independent experi-
death (Fig. 2). In contrast, AGE-1 did not show the inhibi- ments performed under the same conditions.
tory effect on the growth of MSC in terms of proliferation
and apoptosis.

Effects of AGEs on ROS generation in MSCs RAGE expression in MSCs

We have previously shown that cellular events mediated The immunohistochemical study showed RAGE expres-
by AGEs were associated with an enhancement of intracel- sion in MSCs (Fig. 4A). We further used an immunoelec-
lular ROS generation.(31,32) Thus, ROS generation was tron microscopic imaging with the immunogold technique
tested in AGE-treated MSCs. As shown in Fig. 3, both to show the cell surface expression of RAGE (Fig. 4B).
AGE-2 and AGE-3, but not AGE1, stimulated ROS gen- Negative control experiments performed without anti-
eration in a dose-dependent fashion. The ROS generation RAGE antibody but with Protein-A gold conjugate did not
stimulated by 100 ␮g/ml of these AGEs was about 3-fold display any specific gold label (data not shown). Recently,
higher than the control levels (p < 0.01). The following we have shown that AGEs upregulate RAGE expression in
experiments were performed with AGE-2 and AGE-3 at a endothelial cells,(31) retinal pericytes,(32) and renal mesan-
concentration of 100 ␮g/ml, because this dosage has been gial cells.(33) Also in MSCs, Western blotting showed that
shown to be sufficient to induce biological events against AGE-2 as well as AGE-3 upregulated RAGE expression
MSCs. about 1.5-fold (Fig. 4C).
EFFECTS OF AGES ON MESENCHYMAL STEM CELLS 1651

FIG. 2. Apoptosis of human MSCs induced by AGE-2 and


AGE-3. MSCs were seeded on a coverslip and cultured with
MSCGM containing 10–100 ␮g/ml of nonglycated BSA (open
bars), AGE-1 (hatched bars), AGE-2 (gray bars), or AGE-3
(filled bars). After 24 h of cultivation, an in situ TUNEL assay for
semiquantitation of apoptosis was performed, and the percent-
ages of apoptotic cells were estimated by counting a total of 300
cells from random fields. Data are shown as the mean ± SD (n ⳱
3). *p < 0.05. These data are representative of one of two inde-
pendent experiments performed under the same conditions.

FIG. 4. Expression of RAGE in human MSCs. (A) Immunoflu-


orescence microscopy. MSCs were seeded on the Laboratory-Tek
chamber (NUNC), and immunohistochemical staining was carried
out for the expression of RAGE. (B) Immunoelectron micro-
graphs showing immunoreactive gold particles (15 nm in diam-
eter, arrowheads) against a goat anti-RAGE antibody at the cell
surface of MSCs. (a) Low magnification of a secondary electron
image, (b) high magnification of a backscattered electron image,
and (c) the same area of b with a secondary electron image are
shown. (C) Upregulation of RAGE expression by AGE-2 and
AGE-3. MSCs were cultured with MSCGM containing 100 ␮g/ml
of nonglycated BSA, AGE-2, or AGE-3 for 48 h. The results are
also expressed in relative densitometric units (RAGE/␤-actin,
n ⳱ 2).
FIG. 3. Intracellular ROS generation of human MCSs induced
by AGE-2 and AGE-3. MSCs were seeded on a 96-well culture
plate at a density of 2 × 104 cells/well and were cultured for 24 h
with MSCGM containing 10–100 ␮g/ml of nonglycated BSA AGE-2 or AGE-3 was partly recovered by the treatment
(open bars), AGE-1 (hatched bars), AGE-2 (gray bars), or with anti-RAGE neutralizing antibody (Fig. 5A). The ROS
AGE-3 (filled bars). Data are shown as the mean ± SD (n ⳱ 6).
*p < 0.05; **p < 0.01. These data are representative of one of two generation induced by these AGEs was also reduced by the
independent experiments performed under the same conditions. treatment with RAGE-neutralizing antibody (Fig. 5B).

Effects of AGEs on adipogenic differentiation


Effect of neutralizing anti-RAGE antibody in of MSCs
AGEs-treated MSCs
We further studied whether AGE-2 and AGE-3 could
Next, we tested the potential involvement of AGE- influence the differentiation of MSCs. First, the differentia-
RAGE interaction in the regulation of MSC growth. A cell tion of MSCs into adipocyte was induced with dexametha-
counting assay revealed that the growth inhibition by sone-containing medium, and lipid droplets in the cells
1652 KUME ET AL.

FIG. 6. Effects of AGEs on adipogenic differentiation of MSCs.


(A) Oil red O staining. MSCs were induced adipogenic differen-
tiation for 3 weeks by treatment with 100 ␮g/ml of nonglycated
BSA, AGE-2, or AGE-3. (B) ELISA for lipoprotein lipase (LPL)
concentration and (C) the measurement of intracellular triglycer-
ide (TG). MSCs were induced to adipogenic differentiation by
treatment with 100 ␮g/ml of nonglycated BSA (open bar), AGE-2
(gray bars), or AGE-3 (filled bars) in the presence or absence of
a neutralizing anti-RAGE antiserum (0.1% in concentration).
Data are shown as the mean ± SD (n ⳱ 6). *p < 0.05; **p < 0.01;
FIG. 5. AGE-RAGE interaction in MSCs. (A) Cell counting ns, not statistically significant. These data are representative of
assay. MSCs were seeded as shown in Fig. 1 and cultured with one of two independent experiments performed under the same
MSCGM containing 100 ␮g/ml of nonglycated BSA (open bars), conditions.
AGE-1 (hatched bars), AGE-2 (gray bars), or AGE-3 (filled bars)
in the presence or absence of a neutralizing anti-RAGE anti-
serum (0.1% in concentration) for 48 h. The results of cell count-
ing were expressed as percentages in relation to the control non- Effects of AGEs on chondrogenic differentiation
glycated BSA-treated cells. The values shown are the mean ± SD of MSCs
(n ⳱ 3). *p < 0.05. (B) Intracellular ROS generation. MCSs were
seeded as shown in Fig. 3 and cultured with MSCGM containing As a result of chondrogenic differentiation, MSCs
100 ␮g/ml of nonglycated BSA (open bars), AGE-2 (gray bars), or formed a cluster of hypertrophic polygonal cells, within
AGE-3 (filled bars) in the presence or absence of a neutralizing which proteoglycan deposition was apparent by staining
anti-RAGE anti-serum (0.1% in concentration) for 24 h. The with the safranin O method. However, only a faint staining
measured ROS values were expressed as percentages in relation
to the control nonglycated BSA-treated cells. Data are shown as was found in AGE-2– or AGE-3–treated cells (Fig. 7A).
the mean ± SD (n ⳱ 6). *p < 0.05. These data are representative ELISA showed that the production of type II collagen, a
of one of two independent experiments performed under the chondrocyte-specific matrix component, was reduced al-
same conditions. most by one-half by treatment with theses AGEs, and this
reduction was partially recovered by addition of anti-
RAGE antibody by 20% in AGE-2–treated cells or by 30%
were visualized by oil red O staining. Treatment with in AGE-3–treated cells (Fig. 7B).
AGE-2 or AGE-3 reduced the accumulation of lipid drop-
Effects of AGEs on osteogenic differentiation
lets in MSCs (Fig. 6A). Consistently, reduced LPL expres-
sion and intracellular TG content were revealed in cells
of MSCs
treated with AGEs, and the addition of anti-RAGE anti- Under osteogenic stimulation, bone nodule formation
body did not influence AGE-mediated inhibition of adipo- was seen in the control culture, but very little was observed
genesis (Figs. 6B and 6C). in AGE-treated cells (Fig. 8, left panels). Von Kossa stain-
EFFECTS OF AGES ON MESENCHYMAL STEM CELLS 1653

DISCUSSION
AGEs on MSCs growth regulation
It has been shown that AGEs inhibit cell growth in sev-
eral cell lines. In a previous study, we showed that pericyte
loss is an important aspect of the early stage of diabetic
retinopathy, and AGE-2 inhibits proliferation of cultured
bovine retinal pericytes in addition to promoting apoptosis
through the downregulation of expression ratio of bcl-2/
bax.(34) Apoptosis of retinal pericytes has been shown to be
associated with intracellular diacylglycerol/ceramide pro-
duction and activation of the caspase-10 pathway.(35) In rat
Schwann cells, reduced cell replication and induction of
apoptosis by AGE-2 and AGE-3, but not by AGE-1, have
been shown, suggesting that AGEs may be implicated in
diabetic neuropathy.(36) On the other hand, AGEs have the
ability to promote proliferation of cells under specific con-
ditions. In bovine retinal endothelial cells, AGEs enhance
the ROS generation, protein kinase C activation, and vas-
cular endothelial cell growth factor expression, implicating
FIG. 7. Effects of AGEs on chondrogenic differentiation of the excess vascular formation in diabetic retinopathy.(37,38)
MSCs. (A) Safranin O staining. MCSs were induced chondrogenic The AGE-RAGE interaction mediates proliferation of vas-
differentiation for 3 weeks by addition of 100 ␮g/ml of nongly- cular smooth muscle cells, which plays a role in neointimal
cated BSA, AGE-2, or AGE-3. (B) ELISA for type II collagen hyperplasia.(39–41) Furthermore, it has been reported that
synthesis. MCSs were induced to chondrogenic differentiation by
low concentrations (<1 ␮M) of AGEs stimulate mesangial
addition of 100 ␮g/ml of nonglycated BSA (open bar), AGE-2
(gray bars), or AGE-3 (filled bars) in the presence or absence of cell proliferation, whereas higher concentrations (>10 ␮M)
a neutralizing anti-RAGE antiserum (0.1% in concentration). reduce it.(42) In osteoblastic cell lines, AGE-collagen has
The values of A490 are shown as the mean ± SD (n ⳱ 6). *p < been reported to have the opposite effect on the prolifera-
0.05; **p < 0.01; ns, not statistically significant. These data are tion and the number of surviving cells, dependent on the
representative of one of two independent experiments performed
under the same conditions. stage of osteoblastic differentiation.(43) In this study, we
observed that relatively higher doses (1–100 ␮g/ml) of ex-
tracellular AGE-2 and AGE-3 attenuated MSCs with inhi-
bition of proliferation and increased apoptosis (Figs. 1 and
ing revealed a large mass of calcium deposition in accor- 2). Because the bimodal effects of AGEs presumably vary
dance with the bone nodule formation in controls; however, according to the source of AGEs, the type of cells, and
in AGE-treated cells, the loss of mineralization was ob- culture conditions, further study will be needed to clarify
served (Fig. 8, right panels). The intracellular calcium con- the details of the mechanism of AGE-mediated growth
tents of AGE-2– or AGE-3–treated cells were increased control of MSCs, particularly studies modeling the in vivo
1.3-fold in each condition (p < 0.05), and these increases microenvironment for the developmental or disease pro-
were completely inhibited by the neutralizing anti-RAGE cesses.
antibody (Fig. 9A). The activity of bone-specific ALP was
upregulated 2.7-fold in AGE-2–treated cells (p < 0.01) and AGE–RAGE interaction and its relationship with
3.3-fold in AGE-3–treated cells (p < 0.01), and these up- ROS in MSCs
regulations were partially recovered by the neutralizing
Among various subtypes of AGEs, it has been shown
anti-RAGE antibody (Fig. 9B). On the other hand, type I
that AGE-2 (glyceraldehyde-derived AGEs) and AGE-3
collagen synthesis was downregulated by about 20–30% in
(glycolaldehyde-derived AGEs) are the main structures of
both AGE-2– and AGE-3–treated cells (p < 0.01), and AGEs that are detectable in the serum of diabetic patients,
these downregulations were slightly but significantly recov- and both display highly toxic bioactivities on vascular cells,
ered by the neutralizing anti-RAGE antibody (p < 0.05; Fig. mesangial cells, Schwann cells, melanoma cells, and cortical
9C). In addition, we performed time-course analysis for neurons.(44) In this study, we consistently found that
osteogenic differentiation. In control cells (untreated cells AGE-2 and AGE-3, but not control BSA or AGE-1 (glu-
and control-BSA–treated cells), intracellular calcium con- cose-derived AGE), significantly suppressed the growth of
tent increased linearly with time in culture, and upregula- MSCs. Our results may further support the idea that
tion of ALP activity was temporarily induced in association AGE-2 and AGE-3 function as toxic AGEs and play a
with elevated type I collagen synthesis. In AGE-2– and central role in the pathophysiological processes associated
AGE-3–treated cells, intracellular calcium content and with AGEs formation.(44) AGEs have been considered as a
ALP activity were higher than those in control cells at the possible contributor mainly in the diabetic vascular compli-
end of the culture period; however, type I collagen synthesis cations, and now, AGEs play a central role in the patho-
remained at a low level (Fig. 10). physiological processes not only in diabetic but also in non-
1654 KUME ET AL.

FIG. 8. Effects of AGEs on osteogenic dif-


ferentiation of MSCs (Part I). Phase-contrast
microscopy (left panels) and von Kossa stain-
ing (right panels). MSCs were induced osteo-
genic differentiation for 3 weeks by treat-
ment with 100 ␮g/ml of nonglycated BSA,
AGE-2, or AGE-3. The arrows indicate bone
nodule formation, and the high magnifica-
tion inset is also shown.

diabetic patients such as Alzheimer’s disease and chronic the ability to stimulate RAGE expression in osteoblastic
alcoholism.(44) Collagenous proteins are major structural cell lines.(22) Augmentation of RAGE expression by such a
component of connective tissues and play important roles positive feedback mechanism may enhance the biological
in the regulation of cellular function, suggesting that AGE- significance of AGEs, which may exacerbate the disease
modified collagen affects MSCs proliferation and differen- process. Further studies are needed to determine the ex-
tiation.(20) In this study, we focused on the effects of AGE-2 pression pattern of AGE receptors and their downstream
and AGE-3 that have been recognized as toxic(44); how- signaling in many cell types, including MSCs.
ever, further study is needed to understand the effects of
AGE-modified collagen on the cellular function of MSCs. Differentiation of MSCs and AGEs
AGE-2 and AGE-3 may be recognized by RAGE, and
the neutralizing anti-RAGE antibody successfully abolishes There has been little information about the influence of
AGE-mediated cellular events.(28,33) It has been shown that AGEs on the differentiation process of MSCs, and this
the cellular event induced by AGE–RAGE interaction is study was the first to report that AGE-2 and AGE-3 inter-
mediated by intracellular ROS generation.(33,45) In this fered with differentiation of MSCs into adipocytes, chon-
study, we first showed that RAGE is expressed by MSCs drocytes, and osteocytes. The treatment with anti-RAGE
(Fig. 4). The addition of anti-RAGE neutralizing antibody neutralizing antibody revealed that AGE-RAGE interac-
completely inhibited the ROS generation induced by AGEs tion was involved in chondrogenic and osteogenic differen-
(Fig. 5). Thus, the ROS generation observed in MSCs may tiation but not in adipogenic differentiation of MSCs (Figs.
be largely dependent on RAGE signaling. On the other 6–8). It has been reported that a series of gene expressions
hand, the treatment with neutralizing anti-RAGE antibody are involved in adipogenic differentiation of human
partially recovered the growth inhibition of MSCs. There MSCs,(52) and that CD36, a cell surface glycoprotein, func-
are several molecules that have been reported to act as tions as a receptor for AGEs in the adipogenic differentia-
AGE receptors in addition to RAGE, including OST-48 tion of mouse 3T3-L1 cells.(53) Although we do not provide
(ARE-R1)/80K-H/galectin-3,(46) scavenger receptor class direct evidence, AGE receptors other than RAGE may
AI/AII(SR-A),(47) scavenger receptor class B type I (SR- contribute to the modification of adipogenesis. Although
BI),(48) CD36,(49) lectin-like oxidized low density lipopro- the clinical implication of the impact of AGEs on adipo-
tein receptor-1,(50) and FEEL-1,-2.(51) It may be possible genesis is still unclear, we conjecture that AGEs may play a
that AGEs regulate the growth of MSCs in both RAGE- role in soft tissue remodeling, obesity, or insulin-resistance,
dependent and RAGE-independent manners. We found through the functional alteration of MSCs.
that AGE-2 and AGE-3 upregulated RAGE expression MSCs have been shown to be present in adult human
(Fig. 4C). It has been shown that AGEs themselves have articular cartilage other than bone marrow tissue.(54) Our in
EFFECTS OF AGES ON MESENCHYMAL STEM CELLS 1655

FIG. 10. Effects of AGEs on osteogenic differentiation of MSCs


(Part III). (A) Intracellular calcium content, (B) Bone ALP ac-
tivity, and (C) type I collagen synthesis. MSCs were induced os-
teogenic differentiation for 3 weeks in the presence or absence
(untreated) of 100 ␮g/ml of nonglycated BSA (control-BSA),
AGE-2, or AGE-3. Samples were collected every week and were
analyzed. Data are shown as the mean ± SD (n ⳱ 3). *p < 0.05;
**p < 0.01 against the value obtained in untreated cells. These
data are representative of one of two independent experiments
performed under the same conditions.

needed to determine whether there is a causative link be-


FIG. 9. Effects of AGEs on osteogenic differentiation of MSCs
(Part II). (A) Intracellular calcium content, (B) Bone ALP activ- tween accumulation of AGEs and the disease process and
ity, and (C) type I collagen synthesis. MSCs were induced osteo- to develop the appropriate therapies.
genic differentiation for 3 weeks by addition of 100 ␮g/ml of non- Several steps have been identified in osteogenesis, such
glycated BSA (open bar), AGE-2 (gray bars), or AGE-3 (filled as cell proliferation, extracellular matrix production, and
bars) in the presence or absence of a neutralizing anti-RAGE
anti-serum (0.1% in concentration) and analyzed. Data are shown mineralization.(17,56) Proliferation of osteoblasts with the
as the mean ± SD (n ⳱ 6). *p < 0.05; **p < 0.01. These data are production of type I collagen and fibronectin occurs ini-
representative of one of two independent experiments performed tially, followed by enhanced expression of ALP and calcium
under the same conditions. deposition.(56) It has been reported that the enhanced ex-
pression of ALP metabolizes calcium phosphate into in-
soluble phosphate salts, thereby mediating the calcifica-
vitro data may support the idea that excess accumulation of tion.(57) The pattern of the changes in osteogenic markers
AGEs inhibits the intrinsic repair capacity based on the in control MSCs was almost compared with those described
chondrogenesis of MSCs. Consistent with this hypothesis, it in the previous report using more mature osteoblastic cells
has been reported that an increase in AGE levels nega- (Figs. 8–10).(56–58) In this study, AGE-2 and AGE-3 inhib-
tively affects the proteoglycan synthesis and degradation of ited bone nodule formation (Fig. 8). It has been indicated
articular cartilage in OA patients.(55) Further studies will be that von Kossa staining is not a sufficient marker for the in
1656 KUME ET AL.

vitro mineralization of osteoblastic cells.(58) However, we 8. Vashishth D, Gibson GJ, Khoury JI, Schaffler MB, Kimura J,
also observed changes in the other mineralization markers Fyhrie DP 2001 Influence of nonenzymatic glycation on bio-
mechanical properties of cortical bone. Bone 28:195–201.
such as ALP between controls and AGE-treated cells (Figs.
9. Schwartz AV 2003 Diabetes mellitus: Does it affect bone? Cal-
9 and 10). Thus, dystrophic mineralization may occur in cif Tissue Int 73:515–519.
AGE-2– and AGE-3–treated cells. Interestingly, intracellu- 10. Verhaeghe J, van Herck E, Visser WJ, Suiker AM, Thomasset
lar calcium contents and ALP activity were rather increased M, Einhorn TA, Faierman E, Bouillon R 1990 Bone and min-
in AGE-2– and AGE-3–treated cells at the later culture eral metabolism in BB rats with long-term diabetes. Decreased
bone turnover and osteoporosis. Diabetes 39:477–482.
period, without successful induction of type I collagen (Figs.
11. Yamamoto T, Ozono K, Miyauchi A, Kasayama S, Kojima Y,
9 and 10). The treatment with anti-RAGE antibody par- Shima M, Okada S 2001 Role of advanced glycation end prod-
tially recovered the AGE-mediated inhibition of osteogen- ucts in adynamic bone disease in patients with diabetic ne-
esis, and thus AGE-RAGE interaction may be involved in phropathy. Am J Kidney Dis 38:S161–S164.
the intracellular mechanism (Fig. 9). These data may sup- 12. Santana RB, Xu L, Chase HB, Amar S, Graves DT, Trackman
PC 2003 A role for advanced glycation end products in dimin-
port the hypothesis that AGE-2 and AGE-3 exert harmful
ished bone healing in type 1 diabetes. Diabetes 52:1502–1510.
influence on the differentiation of MSCs through the 13. Prockop DJ 1997 Marrow stromal cells as stem cells for non-
AGE–RAGE interaction. hematopoietic tissues. Science 276:71–74.
Several specific signaling have been identified in osteo- 14. Tocci A, Forte L 2003 Mesenchymal stem cell: Use and per-
genesis, such as MAP kinase(59) and Notch(60) signaling. spectives. Hematol J 4:92–96.
15. Bruder SP, Fink DJ, Caplan AI 1994 Mesenchymal stem cells
Bone-specific gene expression such as osteocalcin has been
in bone development, bone repair, and skeletal regeneration
influenced by specific transcriptional factors such as Runx2/ therapy. J Cell Biochem 56:283–294.
Cbfa1.(61) Further studies would be needed to determine 16. Peng H, Huard J 2003 Stem cells in the treatment of muscle
how AGEs influence the differentiation of MSCs in terms and connective tissue diseases. Curr Opin Pharmacol 3:329–
of intracellular signaling pathways. It has been shown that 333.
17. Long MW 2001 Osteogenesis and bone-marrow-derived cells.
aldehyde dehydrogenase (ALDH) activity is elevated in
Blood Cells Mol Dis 27:677–690.
hematopoietic stem cells.(62) Betaine aldehyde dehydroge- 18. Bergman RJ, Gazit D, Kahn AJ, Gruber H, McDougall S,
nase (ALDH-9) is able to catalyze the oxidation of 2-oxo- Hahn TJ 1996 Age-related changes in osteogenic stem cells in
aldehyde methylglyoxal, a precursor of AGEs.(63) Al- mice. J Bone Miner Res 11:568–577.
though the role of ALDH-like activity in MSCs has not 19. D’Ippolito G, Schiller PC, Ricordi C, Roos BA, Howard GA
1999 Age-related osteogenic potential of mesenchymal stromal
been clearly described, further study should be needed to
stem cells from human vertebral bone marrow. J Bone Miner
identify the endogenous regulatory factors for AGE- Res 14:1115–1122.
mediated cellular response in these cells. 20. Katayama Y, Akatsu T, Yamamoto M, Kugai N, Nagata N
In conclusion, in this study, AGEs, especially AGE-2 and 1996 Role of nonenzymatic glycosylation of type I collagen in
AGE-3, attenuated MSC mass in association with an inhi- diabetic osteopenia. J Bone Miner Res 11:931–937.
bition of their proliferation and an increase in the number 21. McCarthy AD, Etcheverry SB, Cortizo AM 1999 Advanced
glycation endproduct-specific receptors in rat and mouse os-
of apoptotic cells. These AGEs also prevented MSCs from teoblast-like cells: Regulation with stages of differentiation.
differentiating into adipocytes, chondrocytes, and osteo- Acta Diabetol 36:45–52.
cytes. Thus, we showed an impact of AGEs in the cellular 22. Cortizo AM, Lettieri MG, Barrio DA, Mercer N, Etcheverry
behavior of MSCs, which may further indicate the delete- SB, McCarthy AD 2003 Advanced glycation end-products
(AGEs) induce concerted changes in the osteoblastic expres-
rious effects of AGEs in the pathogenesis of musculoskel-
sion of their receptor RAGE and in the activation of extracel-
etal diseases in aged and diabetic patients. lular signal-regulated kinases (ERK). Mol Cell Biochem 250:1–
10.
23. DeGroot J, Verzijl N, Jacobs KM, Budde M, Bank RA, Bi-
REFERENCES jlsma JW, TeKoppele JM, Lafeber FP 2001 Accumulation of
advanced glycation endproducts reduces chondrocyte-
1. Brownlee M, Cerami A, Vlassara H 1988 Advanced glycosyla- mediated extracellular matrix turnover in human articular car-
tion end products in tissue and the biochemical basis of dia- tilage. Osteoarthritis Cartilage 9:720–726.
betic complications. N Engl J Med 318:1315–1321. 24. Kuniyasu A, Ohgami N, Hayashi S, Miyazaki A, Horiuchi S,
2. Grandhee SK, Monnier VM 1991 Mechanism of formation of Nakayama H 2003 CD36-mediated endocytic uptake of ad-
the Maillard protein cross-link pentosidine. Glucose, fructose, vanced glycation end products (AGE) in mouse 3T3-L1 and
and ascorbate as pentosidine precursors. J Biol Chem human subcutaneous adipocytes. FEBS Lett 537:85–90.
266:11649–11653. 25. Takeuchi M, Makita Z, Bucala R, Suzuki T, Koike T, Kameda
3. Dyer DG, Blackledge JA, Thorpe SR, Baynes JW 1991 For- Y 2000 Immunological evidence that non-carboxymethyllysine
mation of pentosidine during nonenzymatic browning of pro- advance glycation endproducts are produced from short chain
teins by glucose. Identification of glucose and other carbohy- sugars and decarbonyl compounds in vivo. Mol Med 6:114–125.
drates as possible precursors of pentosidine in vivo. J Biol 26. Gratzner HG 1982 Monoclonal antibody to 5-bromo-and 5-io-
Chem 266:11654–11660. dodeoxyuridine: A new reagent for detection of DNA replica-
4. Thorpe SR, Baynes JW 1996 Role of the Maillard reaction in tion. Science 218:474–475.
diabetes mellitus and diseases of aging. Drugs Aging 9:69–77. 27. Yamagishi S, Edelstein D, Du XL, Kaneda Y, Guzman M,
5. Vlassara H 1997 Recent progress in advanced glycation end Brownlee M 2001 Leptin induces mitochondrial superoxide
products and diabetic complications. Diabetes 46:S19–S25. production and monocyte chemoattractant protein-1 expres-
6. Saudek DM, Kay J 2003 Advanced glycation endproducts and sion in aortic endothelial cells by increasing fatty acid oxidation
osteoarthritis. Curr Rheumatol Rep 5:33–40. via protein kinase A. J Biol Chem 276:25096–25100.
7. Verzijl N, Bank RA, TeKoppele JM, DeGroot J 2003 AGEing 28. Abe R, Shimizu T, Sugawara H, Watanabe H, Nakamura H,
and osteoarthritis: A different perspective. Curr Opin Rheu- Choei H, Sasaki N, Yamagishi S, Takeuchi M, Shimizu H 2004
matol 15:616–622. Regulation of human melanoma growth and metastasis by
EFFECTS OF AGES ON MESENCHYMAL STEM CELLS 1657

AGE-AGE receptor interactions. J Invest Dermatol 122:461– 44. Takeuchi M, Yamagishi S 2004 TAGE (toxic AGEs) hypoth-
467. esis in various chronic diseases. Med Hypotheses 63:449–452.
29. Hanada K, Dennis JE, Caplan AI 1997 Stimulatory effects of 45. Wautier MP, Chappey O, Corda S, Stern DM, Schmidt AM,
basic fibroblast growth factor and bone morphogenetic pro- Wautier JL 2001 Activation of NADPH oxidase by AGE links
tein-2 on osteogenic differentiation of rat bone marrow- oxidant stress to altered gene expression via RAGE. Am J
derived mesenchymal stem cells. J Bone Miner Res 12:1606– Physiol Endocrinol Metab 280:E685–E694.
1614. 46. Vlassara H, Li YM, Imani F, Wojciechowicz D, Yang Z, Liu
30. Connerty HV, Briggs AR 1966 Determination of serum cal- FT, Cerami A 1995 Identification of galectin-3 as a high-
cium by means of orthocresolphthalein complexone. Am J Clin affinity binding protein for advanced glycation end products
Pathol 45:290–296. (AGE): A new member of the AGE-receptor complex. Mol
31. Yamagishi S, Fujimori H, Yonekura H, Yamamoto Y, Yama- Med 1:634–646.
moto H 1998 Advanced glycation endproducts inhibit prosta- 47. Araki N, Higashi T, Mori T, Shibayama R, Kawabe Y,
cyclin production and induce plasminogen activator inhibitor-1 Kodama T, Takahashi K, Shichiri M, Horiuchi S 1995 Macro-
in human microvascular endothelial cells. Diabetologia phage scavenger receptor mediates the endocytic uptake and
41:1435–1441. degradation of advanced glycation end products of the Mail-
32. Yamagishi S, Fujimori H, Yonekura H, Tanaka N, Yamamoto lard reaction. Eur J Biochem 230:408–415.
H 1999 Advanced glycation endproducts accelerate calcifica- 48. Ohgami N, Nagai R, Miyazaki A, Ikemoto M, Arai H, Hori-
tion in microvascular pericytes. Biochem Biophys Res Com- uchi S, Nakayama H 2001 Scavenger receptor class B type
mun 258:353–357. I-mediated reverse cholesterol transport is inhibited by ad-
33. Fukami K, Ueda S, Yamagishi S, Kato S, Inagaki Y, Takeuchi vanced glycation end products. J Biol Chem 276:13348–13355.
M, Motomiya Y, Bucala R, Iida S, Tamaki K, Imaizumi T, 49. Ohgami N, Nagai R, Ikemoto M, Arai H, Kuniyasu A, Hori-
Cooper ME, Okuda S 2004 AGEs activate mesangial TGF-␤- uchi S, Nakayama H 2001 Cd36, a member of the class b scav-
Smad signaling via an angiotensin II-type I receptor interac- enger receptor family, as a receptor for advanced glycation end
tion. Kidney Int 66:2137–2147. products. J Biol Chem 276:3195–3202.
34. Yamagishi S, Hsu CC, Taniguchi M, Harada S, Yamamoto Y, 50. Jono T, Miyazaki A, Nagai R, Sawamura T, Kitamura T,
Ohsawa K, Kobayashi K, Yamamoto H 1995 Receptor- Horiuchi S 2002 Lectin-like oxidized low density lipoprotein
mediated toxicity to pericytes of advanced glycosylation end receptor-1 (LOX-1) serves as an endothelial receptor for ad-
products: A possible mechanism of pericyte loss in diabetic vanced glycation end products (AGE). FEBS Lett 511:170–
microangiopathy. Biochem Biophys Res Commun 213:681– 174.
687. 51. Tamura Y, Adachi H, Osuga J, Ohashi K, Yahagi N, Sekiya M,
35. Lecomte M, Denis U, Ruggiero D, Lagarde M, Wiernsperger Okazaki H, Tomita S, Iizuka Y, Shimano H, Nagai R, Kimura
N 2004 Involvement of caspase-10 in advanced glycation end- S, Tsujimoto M, Ishibashi S 2003 FEEL-1 and FEEL-2 are
product-induced apoptosis of bovine retinal pericytes in cul- endocytic receptors for advanced glycation end products. J
ture. Biochim Biophys Acta 1689:202–211. Biol Chem 278:12613–12617.
36. Sekido H, Suzuki T, Jomori T, Takeuchi M, Yabe-Nishimura 52. Sekiya I, Larson BL, Vuoristo JT, Cui JG, Prockop DJ 2004
C, Yagihashi S 2004 Reduced cell replication and induction of Adipogenic differentiation of human adult stem cells from
apoptosis by advanced glycation end products in rat Schwann bone marrow stroma. J Bone Miner Res 19:256–264.
cells. Biochem Biophys Res Commun 320:241–248. 53. Kuniyasu A, Ohgami N, Hayashi S, Miyazaki A, Horiuchi S,
37. Adamis AP 1998 Advanced glycation end products increase Nakayama H 2003 CD36-mediated endocytic uptake of ad-
retinal vascular endothelial growth factor expression. J Clin vanced glycation end products (AGE) in mouse 3T3-L1 and
Invest 101:1219–1224. human subcutaneous adipocytes. FEBS Lett 537:85–90.
38. Mamputu JC, Renier G 2002 Advanced glycation end products 54. Alsalameh S, Amin R, Gemba T, Lotz M 2004 Identification of
increase, through a protein kinase C-dependent pathway, vas- mesenchymal progenitor cells in normal and osteoarthritic hu-
cular endothelial growth factor expression in retinal endothe- man articular cartilage. Arthritis Rheum 50:1522–1532.
lial cells. Inhibitory effect of gliclazide. J Diabetes Complica- 55. DeGroot J, Verzijl N, Jacobs KM, Budde M, Bank RA, Bi-
tions 16:284–293. jlsma JW, TeKoppele JM, Lafeber FP 2001 Accumulation of
39. Seki N, Hashimoto N, Sano H, Horiuchi S, Yagui K, Makino advanced glycation endproducts reduces chondrocyte-
H, Saito Y 2003 Mechanisms involved in the stimulatory effect mediated extracellular matrix turnover in human articular car-
of advanced glycation end products on growth of rat aortic tilage. Osteoarthritis Cartilage 9:720–726.
smooth muscle cells. Metabolism 52:1558–1563. 56. Siggelkow H, Rebenstorff K, Kurre W, Niedhart C, Engel I,
40. Zhou Z, Wang K, Penn MS, Marso SP, Lauer MA, Forudi F, Schulz H, Atkinson MJ, Hufner M 1999 Development of the
Zhou X, Qu W, Lu Y, Stern DM, Schmidt AM, Lincoff AM, osteoblast phenotype in primary human osteoblasts in culture:
Topol EJ 2003 Receptor for AGE (RAGE) mediates neointi- Comparison with rat calvarial cells in osteoblast differentia-
mal formation in response to arterial injury. Circulation tion. J Cell Biochem 75:22–35.
107:2238–2243. 57. Beck GR Jr, Sullivan EC, Moran E, Zerler B 1998 Relation-
41. Sakaguchi T, Yan SF, Yan SD, Belov D, Rong LL, Sousa M, ship between alkaline phosphatase levels, osteopontin expres-
Andrassy M, Marso SP, Duda S, Arnold B, Liliensiek B, sion, and mineralization in differentiating MC3T3-E1 osteo-
Nawroth PP, Stern DM, Schmidt AM, Naka Y 2003 Central blasts. J Cell Biochem 68:269–280.
role of RAGE-dependent neointimal expansion in arterial re- 58. Bonewald LF, Harris SE, Rosser J, Dallas MR, Dallas SL,
stenosis. J Clin Invest 111:959–972. Camacho NP, Boyan B, Boskey A 2003 Von Kossa staining
42. Geoffroy K, Wiernsperger N, Lagarde M, El Bawab S 2004 alone is not sufficient to confirm that mineralization in vitro
Bimodal effect of advanced glycation end products on mesan- represents bone formation. Calcif Tissue Int 72:537–547.
gial cell proliferation is mediated by neutral ceramidase regu- 59. Higuchi C, Myoui A, Hashimoto N, Kuriyama K, Yoshioka K,
lation and endogenous sphingolipids. J Biol Chem 279:34343– Yoshikawa H, Itoh K 2002 Continuous inhibition of MAPK
34352. signaling promotes the early osteoblastic differentiation and
43. McCarthy AD, Etcheverry SB, Cortizo AM 2001 Effect of mineralization of the extracellular matrix. J Bone Miner Res
advanced glycation endproducts on the secretion of insulin-like 17:1785–1794.
growth factor-I and its binding proteins: Role in osteoblast 60. Tezuka K, Yasuda M, Watanabe N, Morimura N, Kuroda K,
development. Acta Diabetol 38:113–122. Miyatani S, Hozumi N 2002 Stimulation of osteoblastic cell
43. McCarthy AD, Etcheverry SB, Bruzzone L, Cortizo AM 1997 differentiation by Notch. J Bone Miner Res 17:231–239.
Effects of advanced glycation end-products on the prolifera- 61. Lian JB, Stein JL, Stein GS, van Wijnen AJ, Montecino M,
tion and differentiation of osteoblast-like cells. Mol Cell Bio- Javed A, Gutierrez S, Shen J, Zaidi SK, Drissi H 2003 Runx2/
chem 170:43–51. Cbfa1 functions: Diverse regulation of gene transcription by
1658 KUME ET AL.

chromatin remodeling and co-regulatory protein interactions. Address reprint requests to:
Connect Tissue Res 44(Suppl 1):141–148. Seiya Kato, MD, PhD
62. Hess DA, Meyerrose TE, Wirthlin L, Craft TP, Herrbrich PE, Department of Pathology
Creer MH, Nolta JA 2004 Functional characterization of Kurume University School of Medicine
highly purified human hematopoietic repopulating cells iso-
67 Asahi-machi
lated according to aldehyde dehydrogenase activity. Blood
104:1648–1655. Kurume 830-0011, Japan
63. Vander Jagt DL, Hunsaker LA 2003 Methylglyoxal metabo-
E-mail: seikato@med.kurume-u.ac.jp
lism and diabetic complications: Roles of aldose reductase, gly-
oxalase-I, betaine aldehyde dehydrogenase and 2-oxoaldehyde Received in original form November 30, 2004; revised form May
dehydrogenase. Chem Biol Interact 143-144:341–351. 11, 2005; accepted May 20, 2005.

Potrebbero piacerti anche