Вы находитесь на странице: 1из 9

Biol Blood Marrow Transplant 19 (2013) 538e546

Mesenchymal Stem Cells Ameliorate Podocyte Injury and


Proteinuria in a Type 1 Diabetic Nephropathy Rat Model ASBMT
American Society for Blood
and Marrow Transplantation

Shuai Wang 1, 2, Yi Li 1, Jinghong Zhao 1, Jingbo Zhang 1, Yunjian Huang 1, *


1
Institute of Nephrology of Chongqing and Department of Nephrology, Xinqiao Hospital, Third Military Medical University,
Chongqing, China
2
Department of Nephrology, Chengdu Military General Hospital, Chengdu, Sichuan, China

Article history: a b s t r a c t
Received 26 May 2012 Mesenchymal stem cells (MSC) attenuate albuminuria and preserve normal renal histology in diabetic mice.
Accepted 2 January 2013 However, the effects of MSC on glomerular podocyte injury remain uncertain. The aim of this study was to
evaluate the effects of MSC on podocyte injury in streptozotocin (STZ)-induced diabetic rats. Thirty days after
Key Words: diabetes induction by STZ injection (65 mg/kg, intraperitoneally) in Sprague-Dawley rats, the diabetic rats
Mesenchymal stem cells received medium or 2  106 enhanced green uorescent protein-labeled MSC via the renal artery. In vivo
Cell therapy tracking of MSC was followed by immunouorescence analysis. Diabetes-related physical and biochemical
Diabetic nephropathy
parameters were measured on day 60 after the MSC infusion. The expression of podocyte markers (nephrin
Podocyte injury
and podocin), podocyte survival factors (VEGF and BMP-7), and the ultrastructural pathology of podocytes
Proteinuria
Bone morphogenetic protein-7 were also assessed. MSC were only detected in the glomeruli from the left kidney receiving MSC infusion.
Compared with medium-treated diabetic rats, rats treated with MSC showed a suppressed increase in kidney
weight, kidney to body weight index, creatinine clearance rate, and urinary albumin to creatinine ratio;
however, the treatment had no effect on blood glucose or body weight levels. Furthermore, the MSC treat-
ment reduced the loss of podocytes, effacement of foot processes, widening of foot processes, thickening of
glomerular basal membrane (GBM), and loss of glomerular nephrin and podocin. Most important, MSC-
injected kidneys expressed higher levels of BMP-7 but not of VEGF. Our results clearly demonstrated that
intra-arterial administration of MSC prevented the development of albuminuria as well as any damage to or
loss of podocytes, though there was no improvement in blood sugar levels. The protective effects of MSC may
be mediated in part by increasing BMP-7 secretion.
2013 American Society for Blood and Marrow Transplantation.

INTRODUCTION mesenchymal stem cell (MSC) therapy improves micro-


Diabetic nephropathy (DN) is a major complication of albuminuria and preserves the normal renal histology of
diabetes and represents the leading cause of end-stage renal diabetic mice [7,8]. However, the exact mechanisms under-
disease worldwide [1]. To date, there is no cure for DN. Drugs lying their therapeutic effects on microalbuminuria have not
that decrease blood glucose, lower blood pressure, or inhibit been clearly dened. It remains unknown whether the
the actions of the hormone angiotensin can delay, but not contribution of MSC to reducing microalbuminuria is asso-
eliminate, the onset of DN. Thus, the development of novel ciated with the improvement of podocyte injury. Based on
therapeutic strategies that could specically target DN is these ndings, we hypothesized that the transplantation of
necessary. MSC may be a potential therapeutic approach for diabetic
Podocytes are highly specialized cells with important podocyte injury.
roles in maintaining the glomerular ltration barrier and In the present study, we aim to evaluate whether MSC
producing growth factors for both the mesangial and endo- could exert their protective effects against diabetic podocyte
thelial cells [2]. Recent studies have shown that progressive injury. Here, our study provides the rst evidence that MSC
podocyte injury, called podocytopathy, is one of the key therapy may constitute a new therapeutic intervention to
events in the pathogenesis of DN [3]. Podocyte loss and target podocyte damage.
injury are associated with the development of albuminuria
and the acceleration of glomerular structural abnormalities MATERIALS AND METHODS
[2,4,5]. Thus, there is a need to clarify the underlying path- Experimental Animal
Adult male Sprague-Dawley (SD) rats (weight, 180 g to 200 g) from the
ogenesis of podocyte injury and the associated alterations in
laboratory animal center of Xinqiao hospital were maintained under specic
the function of the glomerular ltration barrier to develop pathogen-free conditions at the animal care facilities. After 1 week of
a novel therapeutic strategy for the prevention and amelio- adaptation, diabetes was induced by a single intraperitoneal injection of
ration of podocyte injury. streptozotocin (STZ) (65 mg/kg; Sigma, St Louis, MO) in the rats following
Stem cell therapy is a novel strategy for various diseases overnight fasting. Rats with a blood glucose level over 16.7 mmol/L were
considered diabetes-induced rats and were used in the study. Diabetic rats
and has the potential to be more effective than single-agent received daily injections of long-acting insulin in doses adjusted individually
drug therapies [6]. Several studies have shown that (ranging from 1 U to 4 U) to maintain blood glucose levels between 16
mmol/L and 28 mmol/L and to avoid ketonuria. All animal procedures were
performed according to the guidelines of the Animal Ethics Committee of
the Third Military Medical University, which are consistent with the NIH
Guide for the Care and Use of Laboratory Animals.
Financial disclosure: See Acknowledgments on page 545.
* Correspondence and reprint requests: Yunjian Huang, MD, Department
of Nephrology, Xinqiao Hospital, The Third Military Medical University, Isolation, Culture, and Characterization of MSC
Chongqing, 400037, P.R. China. MSC were obtained from the femurs and tibias of adult male SD rats and
E-mail address: h55769@yahoo.com.cn (Y. Huang). cultured in DMEM/F12 medium (Gibco, Gaithersburg, MD) containing 10%
1083-8791/$ e see front matter 2013 American Society for Blood and Marrow Transplantation.
http://dx.doi.org/10.1016/j.bbmt.2013.01.001
S. Wang et al. / Biol Blood Marrow Transplant 19 (2013) 538e546 539

fetal bovine serum (Gibco), 100 U/mL penicillin, and 100 mg/mL strepto- presenting mesangial expansion [12] or glomerulosclerosis, as described
mycin. The phenotypic properties of MSC were performed by ow cytom- previously [13]. For ultrastructural evaluation, 1 mm3 pieces of renal cortex
etry analysis using the following markers: CD29, CD44, CD34, and CD45 were xed in 2.5% glutaraldehyde in .1 M phosphate buffer (pH, 7.4) for
[9,10]. MSC were expanded in osteogenic or adipogenic differentiation transmission electron microscopy. Electron micrographs of 5 blocks per
media (Cyagen Biosciences, Guangzhou, China) in 6-well plates after the kidney (10 photographs per block) were obtained at a nal magnication of
third passage. Osteogenic and adipogenic differentiations were tested using 8900- or 12,500-fold for each rat using a systematic uniform random
standard protocol from Cyagen Biosciences Inc. sampling protocol. To ensure randomized sampling and avoid repetitious
counting for podocytes in cross-sectional proles, .5-mm sections taken at
Cell Labeling for In Vivo Tracking least 200 mm (average glomerular diameter) apart were cut and collected.
To assess the intrarenal localization of MSC, we used enhanced green The average number of podocytes was measured by manually tracing along
uorescent protein (EGFP) (Cyagen Biosciences) as a cell tracker in the GBM on a video screen. Podocytes were dened as those cells residing
tracking experiments. Cells were labeled using transduction with lentiviral- within the glomerular tuft but outside the GBM. The podocyte quantity,
transduced EGFP, according to the manufacturers protocol. Briey, MSC expressed as the number of podocytes per 2.5 mm GBM, was calculated by
after the third passage were seeded at 2  106 cells in 6-well plates 24 hours computerized measurement using the Image Measurement System (Leica
before transduction. The next day, 1 mL MSC medium per well containing 8 Qwin Lite, Wetzlar, Germany) [14]. At the higher magnication, GBM
mg/mL polybrene (Sigma) and viral were added to the cells at 20 multi- thickness was dened as the distance perpendicular to the GBM between
plicities of infection. After viral addition, cells were incubated at 37 C in 5% the endothelial and podocyte plasma membranes, measured with the aid of
CO2 for 6 hours. The medium was replaced by a fresh complete culture Image-Pro Plus (Media Cybernetics, Silver Spring, MD). The analysis of
solution, and cells were cultured until further experimentation. Selection podocyte foot process effacement was expressed as the arithmetic mean of
was completed 72 hours after incubation with 10 mg/mL puromycin. the podocyte foot processes width (FPW), which was calculated by dividing
the total length of the GBM length by the total number of foot processes
according to published methods [15].
MSC Administration and Experimental Design
Thirty days after the injection of STZ, DN rats were randomly divided X .X 
into 2 groups: DN rats treated with medium (DNmedium, n 10) and DN FPW p=4  GBM length foot process
rats treated with MSC (DNMSC, n 14). Nondiabetic rats were used as the
A correction factor of p/4 was used to correct for presumed random
normal control group (NC, n 6). MSC-treated DN rats were injected with
variation in the angle of the section relative to the long axis of the
2  106 MSC (suspended in 1 mL serum-free medium) via the left renal
podocyte. All measurements were performed by the same technician
artery, as described previously [11], and DNmedium rats received an equal
who was unaware of their origin [16]. The podocyte and GBM data were
volume of medium. In this procedure, 2 rats that died of anesthesia or during
shown as box-and-whisker plots using the SigmaPlot 12.0 software (Systat,
surgery were excluded; 1 died in each group. Then, the rats were housed
San Jose, CA).
under specic pathogen-free conditions with a standard diet and water for
60 days before they were sacriced (Figure 1). At the end of the experiment,
the surviving rats were sacriced, and the left and right kidneys of MSC-
Immunouorescence
treated rats (n 9) and medium-treated rats (n 8) were harvested,
Immunouorescent staining was performed with the specic podocyte
weighed, and processed for histological and biochemical evaluation.
markers nephrin and podocin for localization. Frozen sections (5 mm) were
xed in cold acetone for 10 minutes at 20 C and incubated with wash
EGFP-MSC Detection buffer containing .1% Triton X-100 at room temperature for 20 minutes to
At 24 hours and on day 60 after MSC injection, 2 MSC-treated rats were prevent nonspecic binding. Subsequently, the sections were incubated
sacriced, and their kidney tissues were embedded in Tissue-Tek O.C.T. overnight at 4 C with polyclonal rabbit anti-nephrin (1:200 dilution; Santa
compound (Sakura Finetek, Torrance, CA) and frozen. The tissues were Cruz Biotechnology, Santa Cruz, CA) and polyclonal rabbit anti-podocin
sectioned into 8-mm samples using a cryostat microtome at 22 C. After (1:100 dilution; Santa Cruz Biotechnology) antibodies diluted in bovine
xation in acetone for 5 minutes, the sections were then covered with serum albumin at 1%. After being rinsed in phosphate buffered saline (PBS),
uorescent antifade mounting medium. The 488-nm line of the laser was the TRITC-labeled goat antirabbit secondary antibody was incubated away
used for EGFP uorescence excitation, and renal tissue background auto- from light at 37 C for 30 minutes. Negative controls were run by replacing
uorescence was detected at 555 nm. We used laser scanning confocal the primary antibody with PBS. Staining was evaluated under laser scanning
microscopy (Leica Microsystems, Wetzlar, Germany) for the 2 uorescent confocal microscopy (Leica Microsystems, Wetzlar, Germany).
analyses.

Physical and Biochemical Analysis Western Blot Analysis


The body weight, kidney weight, and blood glucose were measured at The renal cortex was homogenized in lysis buffer (Beyotime Institute of
the conclusion of the experiment. The serum creatinine levels were Biotechnology, Shanghai, China) on ice for 30 minutes. The proteins were
measured in the blood obtained from the retro-orbital sinus at the time of separated by sodium dodecyl sulfate polyacrylamide gel electrophoresis and
the sacrice using an automatic biochemistry analyzer (Hitachi, Tokyo, electrotransferred to polyvinylidene diuoride nitrocellulose membranes
Japan). The creatinine clearance rate was calculated as urinary creatinine  (Roche Molecular Biochemicals, Mannheim, Germany). After blocking in 5%
urine volume/serum creatinine and expressed as milliliters per minute. nonfat dried milk for 1 hour at room temperature, the membranes were
Urinary albumin excretion was expressed as the urinary albumin to creati- incubated with polyclonal rabbit antinephrin (1:700 dilution; Santa Cruz)
nine ratio. Urinary albumin levels were measured in morning spot urine and polyclonal antipodocin (1:500 dilution; Santa Cruz) antibodies over-
samples by a protein assay kit (Bio-Rad, Hercules, CA). After diabetes night at 4 C. After washing, the membrane was incubated for 1 hour at room
induction, albumin to creatinine ratios were determined every 30 days. temperature with goat antirabbit IgG horseradish peroxidase-linked anti-
body (1:2000 dilution; Zhongshan Goldenbridge Biotechnology, Beijing,
Renal Morphology China). The membrane-bound antibody detected was incubated with
The parafn sections were stained with Periodic Acid Schiff (PAS). The a chemiluminescent reagent plus (Thermo Fisher Scientic Inc, Rockford, IL)
extent of glomerular damage was expressed as the percentage of glomeruli and captured on Gel Documentation systems (Alpha Innotech, Santa Clara,

Figure 1. Mesenchymal stem cells (MSC) administration and experimental design. To induce diabetes, the rats were injected with 65 mg/kg streptozotocin. Thirty
days later, diabetic nephropathy (DN) was conrmed, and the DN rats received intra-arterial 2  106 MSC (n 14) or an equal volume of control medium (n 10). On
day 90 post-DN induction (60 days after MSC injection), the left and right kidneys of MSC-treated rats and medium-treated rats were harvested, weighed, and
processed for histological and correlative protein evaluation.
540 S. Wang et al. / Biol Blood Marrow Transplant 19 (2013) 538e546

Figure 2. In vitro differentiation and in vivo tracking of mesenchymal stem cells (MSC). (A) In vitro differentiation of MSC to osteoblasts and adipocytes. MSC in the
primary culture phase shows a broblast-like spindle shape appearance. Magnication 100. The osteogenic differentiation of MSC is indicated by the presence of red
calcium-rich hydroxyapatite stained with Alizarin red. Magnication 100. Adipogenic differentiation is indicated by intracellular mass lipid droplet sediment and by
Oil Red O staining. Magnication 100. Transduction enhanced green uorescent protein (EGFP) into MSC by lentiviral vector resulted in a high transduction
efciency, conrmed by nearly 90% of MSC exhibiting green uorescence 3 days after transduction. Magnication 200. (B) In vivo tracking of EGFP-labeled MSC. The
positive glomeruli, which contained at least 1 to 2 specic GFP uorescence areas, was observed in glomeruli 24 hours and 60 days after MSC injection (arrows).
Autouorescence detection showed signicant autouorescence in tubular areas. Merging both channels showed intrarenal migration of MSC. Magnication 400.
(C) Morphology of adipocytes in MSC-treated rats on day 60 after MSC injection. PAS staining shows the MSC-treated glomeruli contained fat cells (Ca). There was
a focal accumulation of Oil Red O staining in the glomeruli of MSC-treated rats (Cb). Electron microscopy showed the individual large vacuoles as well as several small
droplets within the glomerular podocyte cells (Cc)

CA). Band intensities were quantied by semiquantitative analysis software RESULTS


(Media Cybernetics) and normalized to b-actin. Characterization of Rat MSC
Bone marrow-derived MSC from SD rats exhibited typical
ELISA Measurement of Cytokines broblast-like morphology. MSC identity was proved by
For the analysis of the growth factor production in MSC cultures, the
supernatant was collected upon conuence in the third passage. The
differentiation into osteogenic and adipogenic cells
amounts of VEGF and BMP-7 in cell culture supernatants were quantied by (Figure 2A). Flow cytometric analysis showed that the MSC
ELISA (R&D Systems, Minneapolis, MN). The renal cortical homogenate VEGF were positive for CD29 (99.97%) and CD44 (99.97%) and
and BMP-7 protein levels were also determined using ELISA. All experiments negative for CD34 (1.53%) and CD45 (.43%).
were performed in triplicate.

Statistical Analysis MSC Localization


All values are presented as the means  SD. Statistical signicance was Transduction EGFP into MSC by lentiviral vector resulted
evaluated using 1-way ANOVA with modied t test performed with the
Bonferroni correction. The paired t test was used to compare directly the left
in a high-transduction efciency, conrmed by nearly 90% of
and right kidneys of an animal. A P value <.05 was considered statistically MSC exhibiting green uorescence 3 days after transduction
signicant. (Figure 2A). At 24 hours after MSC injection, EGFP-positive
S. Wang et al. / Biol Blood Marrow Transplant 19 (2013) 538e546 541

Table 1 extracellular matrix deposition and frequent brin cap


Physical and Metabolic Parameters in Animals formation inside the glomeruli. In contrast, 60 days after MSC
Variable NC (n 6) DNmedium DNMSC injection, there was a signicant decrease in mesangial
(n 8) (n 9) matrix deposition (Figure 4B) and in the glomerulosclerotic
Blood glucose 5.87  .65 26.91  4.71* 24.43  4.03* index (Figure 4C) of the left kidneys of MSC-treated rats,
(mmol/L) compared to the left and right kidneys of medium-treated
y
Kidney weight (g) 1.20  .05 1.56  .06* 1.41  .04*,
rats and the right kidneys of MSC-treated rats.
Body weight (g) 387.0  10.02 203.38  5.90* 210.33  7.45*
Kidney/body 3.09  .09 7.67  .42* 6.68  .36*,
y
Electron microscopy studies were performed to deter-
weight (g/kg) mine the early structural changes in podocyte morphology
y
Creatinine clearance 1.52  .06 1.92  .07* 1.63  .05*, (Figure 5A). Sixty days after MSC injection, compared with
rate (mL/min) the left kidneys of medium-treated rats and the right kidneys
NC indicates normal control; DNmedium, medium-treated rats; DNMSC, of MSC-treated rats, the MSC-injected kidneys exhibited
MSC-treated rats. a signicant improvement in ultrastructural abnormalities,
Data are presented as the means  SD. Kidney weight, kidney/body weight,
such as loss of podocytes (Figure 5B), GBM thickening
and creatinine clearance rate levels were reduced signicantly by mesen-
chymal stem cells (MSC) treatment. Glucose and body weight did not (Figure 5C), and podocyte foot process effacement
change signicantly between the 2 DN rat groups. (Figure 5D).
* P < .05 versus NC group. Oil Red O staining was performed to determine the renal
y
P < .05 versus DNmedium group. accumulation of neutral lipids. As shown in Figure 2C, 60
days after MSC injection, approximately 10% of the MSC-
treated glomeruli contained fat cells (Figure 2Ca). There
cells were only observed in the left kidney receiving EGFP-
was a focal accumulation of Oil Red O staining in the
labeled MSC but not in the contralateral right kidneys (data
glomeruli of MSC-treated rats (Figure 2Cb) and almost no
not shown). EGFP-positive cells were observed in glomeruli
staining in the glomeruli of medium-treated rats or in the
but not in tubulointerstitial areas (data not shown). About
right glomeruli of MSC-treated rats (data not shown). Elec-
20% and 3% glomeruli showed positive uorescence at 24
tron microscopy showed the individual large vacuoles as well
hours and 60 days after MSC infusion, respectively
as several small droplets within the glomerular podocyte
(Figure 2B).
cells (Figure 2Cc).
Effect of MSC on Physical and Biochemical Parameters
Effect of MSC on the Renal Expression of Nephrin and
The blood glucose, body weight, kidney weight, kidney to
Podocin
body weight index, and creatinine clearance rate levels of the
To further reveal the mechanism responsible for the
experimental animals are shown in Table 1. Compared with
prevention of albuminuria in MSC-treated diabetic rats, we
medium-treated DN rats, the MSC treatment suppressed the
studied the expression of nephrin and podocin proteins,
increase in kidney weight, kidney to body weight index, and
which are the 2 major proteins involved in the formation of
creatinine clearance rate levels but had no effect on blood
a slit diaphragm. Comparing the left kidneys of medium-
glucose and body weight levels on day 60 after MSC infusion.
treated rats with the right kidneys of MSC-treated rats,
As shown in Figure 3, the ratio of urinary albumin to
MSC treatment led to a signicant improvement in the loss of
creatinine 30 and 60 days after MSC injection was signi-
glomerular nephrin and podocin staining (Figure 6A).
cantly lower in MSC-treated DN rats than in medium-treated
In line with the results from immunouorescence stain-
rats, suggesting that MSC have an antiproteinuric effect in
ing, western blot analysis showed that MSC injection
DN rats.
signicantly attenuated the diabetic-induced decrease of
glomerular nephrin and podocin expression, compared with
Renal Histopathological and Ultrastructural Changes
the left kidneys of medium-treated rats and the right kidneys
Light microscopy studies (PAS-staining) were performed
of MSC-treated rats (Figure 6B).
to determine the extent of the glomerular damage
(Figure 4A). As shown in Figure 4A, 90 days after STZ injec-
MSC Express High Levels of VEGF and BMP-7
tion, the left and right kidneys of medium-treated rats and
Next, we aimed to evaluate whether MSC could exert
the right kidneys of MSC-treated rats exhibited profound
their protective effects against podocyte damage in part by
increasing VEGF and BMP-7 secretion, which have been
shown to be podocyte survival factors to rescue podocytes
from injury [17,18]. Both factors could be secreted by MSC, as
previously reported [19]. After 24 hours of incubation, MSC
released higher amounts of VEGF (1231  123 pg/mL versus
10  2 pg/mL) and BMP-7 (1868  67 pg/mL versus 49 
12 pg/mL) in vitro in cell culture supernatants than in fresh
medium (Figure 7A).
As shown in Figure 7B, the renal cortical VEGF levels in
the left kidneys from MSC-treated rats (1928  63 pg/mg
protein) did not signicantly increase on day 60 post-MSC
injection compared with the left kidneys of medium-
treated rats (1804  97 pg/mg protein) and the right
kidneys of MSC-treated rats (1838  87 pg/mg protein). In
Figure 3. Urinary albumin excretion. At 30 days and 60 days after mesen-
chymal stem cells injection, the ratio of urinary albumin to creatinine was
contrast, the BMP-7 levels were higher in the left kidneys of
signicantly lower in MSC-treated DN rats than in medium-treated rats. The MSC-treated rats (1431  39 pg/mg protein) than those in the
results are presented as the mean  SD. *P < .05 versus medium-treated rats. left kidneys of medium-treated rats (837  61 pg/mg protein)
542 S. Wang et al. / Biol Blood Marrow Transplant 19 (2013) 538e546

Figure 4. Mesangial expansion and glomerulosclerosis. (A) Ninety days after streptozotocin injection, the left and right kidneys of medium-treated rats and the right
kidneys of mesenchymal stem cells (MSC)-treated rats exhibited profound extracellular matrix deposition and frequent brin cap formation (arrows) inside the
glomeruli. Sixty days after the MSC injection, there was a signicant decrease in mesangial matrix deposition (B) and glomerulosclerotic index (C) in the left kidneys
of MSC-treated rats, compared to the left kidneys of medium-treated rats and the right kidneys of MSC-treated rats. Data are presented as the mean  SD. *P < .05
versus the kidneys of NC rats; #P < .05 versus the left kidneys of medium-treated rats; :P < .05 versus the right kidneys of MSC-treated rats.

and the right kidneys of MSC-treated rats (889  100 pg/mg expanded in culture [25]. However, in vitro manipulations
protein), suggesting that the protective effects of MSC on may also alter or inuence their natural phenotypes, leading
podocyte injury in DN may be mediated in part by an to different, as-yet-undened activities and responses. In this
increased BMP-7 secretion. study, we established that adherent, bone marrow-derived,
spindle-shaped cells expressed mesenchymal cell pheno-
DISCUSSION types (CD29, CD44) rather than CD34 (hematopoietic cell
MSC are multipotent cells present in bone marrow and in marker) and CD45 (leukocyte marker). In addition, the MSC
mesenchymal tissues that can differentiate in vitro into possessed osteogenic and adipogenic potential; we
adipocytic, chondrocytic, and osteocytic lineages. These cells conrmed with this approach that the subsequently
do not exhibit hemopoietic markers but rather express CD29, administered cells retained the characteristics of MSC.
CD44, CD105, and alpha smooth muscle actin [10]. In vitro, We found that adequate homing of MSC to the injured
MSC may give rise to insulin-producing cells [20,21]. In vivo, tissue is important for effective therapy. In most studies, MSC
MSC may differentiate into renal cells [22,23] and reconsti- is administered through a standard intravenous route. A
tute necrotic segments of damaged kidneys. Thus, bone disadvantage of the systemic intravenous delivery of MSC
marrow-derived MSC have been used successfully in cell can be low uptake at the site of injury. Indeed, signicant
therapy to treat animal models with acute and chronic renal engraftment of injured tissue was observed in some studies
failure, such as ischemia-reperfusion injury, 5/6 nephrec- [26-28], but not all [29,30]. Schrepfer et al. [31] demon-
tomy, and unilateral ureteral obstruction, as well as in kidney strated that the systemic intravenous route of administration
transplantation models [24]. The present study provides was not appropriate for MSC to reach their site of activation.
clear evidence that although there was no improvement in Zonta [32] showed that the intra-arterial administration of
blood sugar levels, the injected MSC did prevent the devel- MSC were the most effective route to achieve immunomo-
opment of albuminuria and the loss of podocytes. dulating effects in experimental kidney transplantation,
The primary advantage of MSC for utilization in cell which primarily occurs because large MSC (15 mm to 19 mm)
therapy is the ease with which they can be harvested from remain trapped in the capillaries of the small lung lter,
the bone marrow, isolated by plastic adherence, and which in turn causes the inadequate homing of MSC to the
S. Wang et al. / Biol Blood Marrow Transplant 19 (2013) 538e546 543

Figure 5. Ultrastructural alterations of the podocyte and glomerular basal membrane (A). Sixty days after mesenchymal stem cells (MSC) injection, compared with
the left kidneys of medium-treated rats and the right kidneys of MSC-treated rats, a signicant improvement in ultrastructural abnormalities, such as loss of
podocytes (B), GBM thickening (C), and podocyte foot process effacement (D) was found in MSC-injected kidneys. The degree of podocyte detachment was calculated
as the mean FPW. The podocyte number per 2.5 mm of GBM and GBM thickness are presented as median values using box-and-whisker plots. The results are
presented as the means  SD. M indicates glomerular basal membrane; P, podocyte; FP, foot process; NC, normal control rats. *P < .05 versus the kidneys of NC rats; #P
< .05 versus the left kidneys of medium-treated rats; :P < .05 versus the right kidneys of MSC-treated rats.

injured tissue. However, using the renal artery as the injec- day 60 after the injection into a renal artery, approximately
tion route to administer MSC to treat DN may be associated 10% of the MSC-treated glomeruli contained cells that
with the following 2 major complications: (1) renal infarcts exhibited features typical of adipocytes. Electron microscopy
and loss of function, and (2) ectopic differentiation into showed that large or multiple small fatty vacuoles were
adipocytes within glomeruli [33]. Recently, Ho et al. [34] found within the glomerular podocyte cells. This phenom-
demonstrated that multiple intravenous transplantations of enon had been previously reported by Kunter et al. [33].
MSC effectively restored the long-term blood glucose Although it seems highly likely that these cells originated
homeostasis during 15 weeks in STZ-induced diabetic mice; from the transplanted MSC, the exact mechanisms of MSC
thus, multiple intravenous transplantations of MSC may transformation are not completely understood. There is
serve as a new therapeutic strategy for DM patients. The evidence that BMP-7 promotes the differentiation of MSC
following 2 factors may contribute to MSC for the treatment into adipocyte cells [35]. In our study, we found that the
of DN: (1) blood sugar reduction and (2) renal protective expression of BMP-7 was signicantly increased in MSC-
actions of the MSC within the glomeruli. To examine the treated kidneys compared to medium-treated kidneys,
second factor, we could only choose the renal artery as the which might help explain these results. These phenomena
injection route instead of systemic administration. In our seem to suggest that the long-term effects of intrarenal,
study, intra-arterial injection led to 20% of glomeruli con- syngeneic MSC transplantation may result in the maldiffer-
taining EGFP cells at 24 hours, as observed in the targeted entiation of glomerular MSC into adipocytes, thus calling into
kidney but not in the lung, liver, or spleen (data not shown). question the potential benet of MSC treatment for DN.
However, 60 days after MSC injection, only 3% of glomeruli Taken together, our observation is consistent with the
containing EGFP cells were found in MSC-treated kidneys. homing capacity of MSC to injured areas via intra-arterial
This low level of engraftment suggests a paracrine, rather injection in several animal models [11,36].
than a cell differentiation, contribution to the benecial Type 1 Diabetes (T1DM), a multifactorial autoimmune
effects of the MSC. However, the positive status of the 3% of disease involving genetic and environmental factors, is
the glomeruli in the kidney on 60 days after injection is still hallmarked by the T cell- and macrophages-mediated
higher than the 0% in the lung, liver, and spleen and suggests destruction of pancreatic b-cells, resulting in an irreversible
that there was some targeting of the cell therapy to the insulin deciency. STZ or other diabetogenic agents (eg,
kidney, likely due to the renal artery injection. In addition, on alloxan) with b-cell-toxic abilities have also been used for
544 S. Wang et al. / Biol Blood Marrow Transplant 19 (2013) 538e546

Figure 6. Glomerular expression of nephrin and podocin. (A) The staining scores of nephrin and podocin were signicantly reduced in the left and right kidneys of
medium-treated rats and in the right kidneys of mesenchymal stem cells (MSC)-treated rats. The sources of nephrin and podocin in MSC-injected kidneys were
restored. Magnication 400. (B) Western blot analysis showed that nephrin and podocin expressions were restored in MSC-injected kidneys compared to the left
kidneys of medium-treated rats and the right kidneys of MSC-treated rats. Both proteins expression was quantied by semi-quantitative analysis of the western blots.
The results are presented as the means  SD. *P < .05 versus the kidneys of normal control (NC) rats; #P < .05 versus the left kidneys of medium-treated rats; :P < .05
versus the right kidneys of MSC-treated rats.

producing chemically induced T1DM models through their to obtain valuable information about understanding the
administration in a large dose or repeated low doses for molecular bases that contribute to the induction of T1DM in
several days. However, although the STZ model results in humans.
hyperglycemia and insulinopenia, it does not bear strong Recent studies have shown that the systemic adminis-
autoimmune features [37-39], suggesting that the STZ- tration of MSC into STZ-induced type 1 diabetic C57BL/6
induced diabetic model is not the equivalent of T1DM. mice [7] and type 2 diabetic NOD/scid mice [8] reduced
Although an ideal experimental model of T1DM does not microalbuminuria and preserved normal renal histology. In
exist, we believe that the use of this STZ model may allow us contrast, untreated diabetic mice presented glomerular

Figure 7. The expression of VEGF and BMP-7 in cultured mesenchymal stem cells (MSC) and kidneys of diabetic nephropathy rats. (A) After 24 hours of incubation,
MSC released high amounts of VEGF and BMP-7 in vitro in cell culture supernatants (black bars) as opposed to in fresh medium (white bars). xP < .05 versus fresh
medium. (B) Sixty days after MSC injection, there were no signicant differences in the renal cortical VEGF levels between the 2 groups. In contrast, BMP-7 levels
were signicantly higher in the MSC-treated kidneys than in the left kidneys of medium-treated rats or the right kidneys of the MSC-treated rats. The results were
presented as the means  SD. *P < .05 versus the kidneys of normal control rats; #P < .05 versus the left kidneys of medium-treated rats; :P < .05 versus the right
kidneys of MSC-treated rats.
S. Wang et al. / Biol Blood Marrow Transplant 19 (2013) 538e546 545

hyalinosis and mesangial expansion. The observed renopro- did not reach statistical signicance. These data suggested
tection was associated with a decrease in glycemic levels that that MSC exert their protective effects against podocyte
could be explained by beta-pancreatic islet regeneration damage possibly by increasing BMP-7 secretion. Both in vitro
resulting from MSC administration. However, in these and in vivo studies have indicated that BMP-7 is important
studies, neither group demonstrated a signicant histologic for the maintenance of podocyte viability and differentiation
amelioration of podocyte damage in response to MSC injec- [18,51,52]. In adult rodent glomeruli in vivo, BMP-7 and its
tion; furthermore, their data could not completely distin- receptors are expressed in podocytes [18]. In cultured mouse
guish these benecial effects from a reduction in the serum podocytes, high glucose decreases BMP-7 expression, and
glucose levels or direct protection of the podocytes. In this the treatment of podocytes with rhBMP-7 restores podocin
study, we successfully established an experimental DN, and synaptopodin expression [51]. In STZ-induced diabetic
similar to human type 1 DN and characterized by hypergly- mice, the maintenance of renal and glomerular podocyte
cemia and albuminuria, that was also associated with the BMP-7 levels by means of a transgene reduces the onset and
loss of podocytes, effacement of foot process, widening of progression of nephropathy, especially podocyte dropout
foot process, and thickening of the GBM [1,40-42]. These [52]. Moreover, the ability of MSC to contribute to podocyte
changes contribute to progressive glomerulosclerosis, which regeneration has been recently shown in a mouse model of
is a feature of experimental DN. More important, our results Alport syndrome [53]. In addition to BMP-7 and VEGF, MSC
revealed that MSC effectively ameliorated the phenotypic can secrete a number of factors, such as HGF, bFGF, and IGF-I,
changes of podocytes in this animal model. Another inter- all known to improve renal function in acute kidney injury,
esting nding of the present study was that the renopro- mediated by their antiapoptotic, mitogenic, and other cyto-
tective effect of MSC in podocyte injury was only found in the kine actions [49,50]. Based on these observations, we deduce
left kidneys, not in the right kidneys, of MSC-treated DN rats. that the mechanisms that mediate the protective effects of
In addition, the renoprotection of MSC were not attributable MSC may be primarily paracrine actions.
to glycemic control because MSC via intra-arterial injection In summary, our current study provided further evidence
had no impact on the elevated serum glucose levels in the that MSC could not only exert antialbuminuric effects but
diabetic rats. As shown in Table 1, MSC administration by also, more important, prevent early phenotypic changes in
intra-arterial injection reversed neither hyperglycemia nor podocytes and, subsequently, glomerulosclerosis. We believe
body weight loss, the latter of which was associated with that the successful treatment of DN with MSC demonstrated
poor blood glucose control. Thus, our study clearly suggested herein holds substantial promise for the development of
that MSC themselves may have had a direct benecial effect novel, MSC-based interventions that can prevent the devel-
on the ultrastructural alterations in podocytes. opment of DN. However, because our study is an animal
Podocytes play a major role in maintaining the integrity study, these ndings must be conrmed after further study,
and permeability of the glomerular ltration barrier [43,44]. such as clinical trial, on human subjects.
In DN, the effacement of podocyte foot processes and the loss
of slit diaphragm proteins result in a leakage of albumin and
proteinuria [45,46]. The results of the present study showed ACKNOWLEDGMENTS
that the levels of urinary protein were signicantly increased We thank the laboratory animal center of Xinqiao hospital
in diabetic rats and could be signicantly lowered by MSC. To for the excellent rat care. We also thank Dr. Xiaohua Guo
identify the molecular basis of the observed phenomena, we (University of Utah) for his invaluable advice.
performed immunostaining for nephrin and podocin, which Financial disclosure: This study was supported by
are constituents of the slit diaphragm and act as a barrier for the National Natural Science Foundation of China (No.
glomerular capillary walls [47]. Our study revealed that MSC 30570871) and the Natural Science Foundation of Chongqing
administration could restore the expression of nephrin and (No. 2007BB1017).
podocin in diabetic rats. Hence, our data suggested that the Conict of Interest Statement: There are no conicts of
antiproteinuric effects of MSC in diabetes-induced protein- interest to report.
uria may be associated with the restoration of the expres-
sional patterns of podocyte-associated proteins and the
maintenance of podocyte foot process in the glomerular REFERENCES
ltration barrier. 1. Molitch ME, DeFronzo RA, Franz MJ, et al. Nephropathy in diabetes.
What are the mechanisms underlying the benet of MSC Diabetes Care. 2004;27(Suppl 1):S79-S83.
in our model? Several recent studies suggest that MSC may 2. Steffes MW, Schmidt D, McCrery R, Basgen JM. Glomerular cell number
in normal subjects and in type 1 diabetic patients. Kidney Int. 2001;59:
mediate their benecial effects, in part, through paracrine 2104-2113.
and/or endocrine activity, not through a differentiation 3. Ziyadeh FN, Wolf G. Pathogenesis of the podocytopathy and protein-
mechanism as had been described in some acute renal uria in diabetic glomerulopathy. Curr Diabetes Rev. 2008;4:39-45.
4. Dalla Vestra M, Masiero A, Roiter AM, et al. Is podocyte injury relevant
damage models [48]. To elucidate the potential mechanism in diabetic nephropathy? Studies in patients with type 2 diabetes.
of MSC, we focused on evaluating the expression of VEGF and Diabetes. 2003;52:1031-1035.
BMP-7 in vitro and in vivo, which are podocyte survival 5. White KE, Bilous RW. Structural alterations to the podocyte are related
to proteinuria in type 2 diabetic patients. Nephrol Dial Transplant. 2004;
factors secreted by MSC. In this respect, it was noteworthy 19:1437-1440.
that our MSC released high amounts of VEGF and BMP-7 6. Togel F, Westenfelder C. Adult bone marrow-derived stem cells for
in vitro. Our ndings were in line with the data of others organ regeneration and repair. Dev Dyn. 2007;236:3321-3331.
7. Ezquer FE, Ezquer ME, Parrau DB, et al. Systemic administration of
showing that MSC release VEGF and BMP-7, exerting bene-
multipotent mesenchymal stromal cells reverts hyperglycemia and
cial effects in chronic kidney disease and acute kidney prevents nephropathy in type 1 diabetic mice. Biol Blood Marrow
injury models [19,49,40]. We found that the expression of Transplant. 2008;14:631-640.
BMP-7 was signicantly increased in MSC-treated kidneys 8. Lee RH, Seo MJ, Reger RL, et al. Multipotent stromal cells from human
marrow home to and promote repair of pancreatic islets and renal
compared to medium-treated kidneys. There were small glomeruli in diabetic NOD/scid mice. Proc Natl Acad Sci U S A. 2006;103:
increases in VEGF in MSC-treated kidneys. However, these 17438-17443.
546 S. Wang et al. / Biol Blood Marrow Transplant 19 (2013) 538e546

9. Jiang TS, Cai L, Ji WY, et al. Reconstruction of the corneal epithelium liver integrity of irradiation damage. Methods Mol Biol. 2012;826:
with induced marrow mesenchymal stem cells in rats. Mol Vis. 2010; 179-188.
16:1304-1316. 31. Schrepfer S, Deuse T, Reichenspurner H, et al. Stem cell trans-
10. Ma P, Hindocha S, Ma R, et al. Adult mesenchymal stem cells and cell plantation: the lung barrier. Transplant Proc. 2007;39:573-576.
surface characterization - a systematic review of the literature. Open 32. Zonta S, De Martino M, Bedino G, et al. Which is the most suitable and
Orthop J. 2011;5:253-260. effective route of administration for mesenchymal stem cell-based
11. Kunter U, Rong S, Djuric Z, et al. Transplanted mesenchymal stem cells immunomodulation therapy in experimental kidney transplantation:
accelerate glomerular healing in experimental glomerulonephritis. endovenous or arterial? Transplant Proc. 2010;42:1336-1340.
J Am Soc Nephrol. 2006;17:2202-2212. 33. Kunter U, Rong S, Boor P, et al. Mesenchymal stem cells prevent
12. Lee EA, Seo JY, Jiang Z, et al. Reactive oxygen species mediate high progressive experimental renal failure but maldifferentiate into
glucose-induced plasminogen activator inhibitor-1 up-regulation in glomerular adipocytes. J Am Soc Nephrol. 2007;18:1754-1764.
mesangial cells and in diabetic kidney. Kidney Int. 2005;67:1762-1771. 34. Ho JH, Tseng TC, Ma WH, et al. Multiple intravenous transplantations
13. Thallas-Bonke V, Thorpe SR, Coughlan MT, et al. Inhibition of NADPH of mesenchymal stem cells effectively restore long-term blood
oxidase prevents advanced glycation end product-mediated damage in glucose homeostasis by hepatic engraftment and beta cell differen-
diabetic nephropathy through a protein kinase C-alpha-dependent tiation in streptozosin-induced diabetic mice. Cell Transplant. 2012;
pathway. Diabetes. 2008;57:460-469. 21:997-1009.
14. Macedo CS, Lerco MM, Capelletti SM, et al. Reduction of podocytes 35. Neumann K, Endres M, Ringe J, et al. BMP7 promotes adipogenic but
number in late diabetic alloxan nephropathy: prevention by glycemic not osteo-/chondrogenic differentiation of adult human bone marrow-
control. Acta Cir Bras. 2007;22:337-341. derived stem cells in high-density micro-mass culture. J Cell Biochem.
15. van den Berg JG, van den Bergh Weerman MA, Assmann KJ, et al. 2007;102:626-637.
Podocyte foot process effacement is not correlated with the level of 36. Yoo JH, Park C, Jung DI, et al. In vivo cell tracking of canine allogenic
proteinuria in human glomerulopathies. Kidney Int. 2004;66: mesenchymal stem cells administration via renal arterial catheteriza-
1901-1906. tion and physiopathological effects on the kidney in two healthy dogs.
16. Gundersen HJ, Seefeldt T, Osterby R. Glomerular epithelial foot J Vet Med Sci. 2011;73:269-274.
processes in normal man and rats. Distribution of true width and its 37. Rees DA, Alcolado JC. Animal models of diabetes mellitus. Diabet Med.
intra- and inter-individual variation. Cell Tissue Res. 1980;205:147-155. 2005;22:359-370.
17. Ma J, Matsusaka T, Yang HC, et al. Induction of podocyte-derived VEGF 38. Lenzen S. The mechanisms of alloxan- and streptozotocin-induced
ameliorates podocyte injury and subsequent abnormal glomerular diabetes. Diabetologia. 2008;51:216-226.
development caused by puromycin aminonucleoside. Pediatr Res. 39. Szkudelski T. The mechanism of alloxan and streptozotocin action in B
2011;70:83-89. cells of the rat pancreas. Physiol Res. 2001;50:537-546.
18. Mitu GM, Wang S, Hirschberg R. BMP7 is a podocyte survival factor and 40. Menini S, Iacobini C, Oddi G, et al. Increased glomerular cell (podocyte)
rescues podocytes from diabetic injury. Am J Physiol Renal Physiol. apoptosis in rats with streptozotocin-induced diabetes mellitus: role in
2007;293:F1641-F1648. the development of diabetic glomerular disease. Diabetologia. 2007;50:
19. Ninichuk V, Gross O, Segerer S, et al. Multipotent mesenchymal stem 2591-2599.
cells reduce interstitial brosis but do not delay progression of chronic 41. Barutta F, Corbelli A, Mastrocola R, et al. Cannabinoid receptor 1
kidney disease in collagen4A3-decient mice. Kidney Int. 2006;70: blockade ameliorates albuminuria in experimental diabetic nephrop-
121-129. athy. Diabetes. 2010;59:1046-1054.
20. Neshati Z, Matin MM, Bahrami AR, Moghimi A. Differentiation of 42. Sohn E, Kim J, Kim CS, et al. Extract of the aerial parts of Aster kor-
mesenchymal stem cells to insulin-producing cells and their impact on aiensis reduced development of diabetic nephropathy via anti-
type 1 diabetic rats. J Physiol Biochem. 2010;66:181-187. apoptosis of podocytes in streptozotocin-induced diabetic rats. Bio-
21. Sun Y, Chen L, Hou XG, et al. Differentiation of bone marrow-derived chem Biophys Res Commun. 2010;391:733-738.
mesenchymal stem cells from diabetic patients into insulin- 43. Saleem MA. Biology of the human podocyte. Nephron Exp Nephrol.
producing cells in vitro. Chin Med J (Engl). 2007;120:771-776. 2003;95:e87-e92.
22. Huls M, Russel FG, Masereeuw R. Insights into the role of bone 44. Kawachi H, Miyauchi N, Suzuki K, et al. Role of podocyte slit diaphragm
marrow-derived stem cells in renal repair. Kidney Blood Press Res. as a ltration barrier. Nephrology (Carlton). 2006;11:274-281.
2008;31:104-110. 45. Ihalmo P, Wessman M, Kaunisto MA, et al. Association analysis of
23. Morigi M, Imberti B, Zoja C, et al. Mesenchymal stem cells are reno- podocyte slit diaphragm genes as candidates for diabetic nephropathy.
tropic, helping to repair the kidney and improve function in acute renal Diabetologia. 2008;51:86-90.
failure. J Am Soc Nephrol. 2004;15:1794-1804. 46. White KE. Research into the glomerular podocyteeis it relevant to
24. Asanuma H, Meldrum DR, Meldrum KK. Therapeutic applications of diabetic nephropathy? Diabet Med. 2006;23:715-719.
mesenchymal stem cells to repair kidney injury. J Urol. 2010;184: 47. Kerjaschki D. Caught at-footed: podocyte damage and the molecular
26-33. bases of focal glomerulosclerosis. J Clin Invest. 2001;108:1583-1587.
25. Meirelles Lda S, Fontes AM, Covas DT, Caplan AI. Mechanisms involved 48. Masereeuw R. Contribution of bone marrow-derived cells in renal
in the therapeutic properties of mesenchymal stem cells. Cytokine repair after acute kidney injury. Minerva Urol Nefrol. 2009;61:373-384.
Growth Factor Rev. 2009;20:419-427. 49. Togel F, Hu Z, Weiss K, et al. Administered mesenchymal stem cells
26. Ezquer F, Ezquer M, Simon V, et al. Endovenous administration of protect against ischemic acute renal failure through differentiation-
bone-marrow-derived multipotent mesenchymal stromal cells independent mechanisms. Am J Physiol Renal Physiol. 2005;289:
prevents renal failure in diabetic mice. Biol Blood Marrow Transplant. F31-F42.
2009;15:1354-1365. 50. Togel F, Weiss K, Yang Y, et al. Vasculotropic, paracrine actions of
27. Morigi M, Introna M, Imberti B, et al. Human bone marrow mesen- infused mesenchymal stem cells are important to the recovery from
chymal stem cells accelerate recovery of acute renal injury and prolong acute kidney injury. Am J Physiol Renal Physiol. 2007;292:F1626-F1635.
survival in mice. Stem Cells. 2008;26:2075-2082. 51. De Petris L, Hruska KA, Chiechio S, Liapis H. Bone morphogenetic
28. Semedo P, Correa-Costa M, Antonio Cenedeze M, et al. Mesenchymal protein-7 delays podocyte injury due to high glucose. Nephrol Dial
stem cells attenuate renal brosis through immune modulation and Transplant. 2007;22:3442-3450.
remodeling properties in a rat remnant kidney model. Stem Cells. 2009; 52. Wang S, de Caestecker M, Kopp J, et al. Renal bone morphogenetic
27:3063-3073. protein-7 protects against diabetic nephropathy. J Am Soc Nephrol.
29. Walczak P, Zhang J, Gilad AA, et al. Dual-modality monitoring of tar- 2006;17:2504-2512.
geted intraarterial delivery of mesenchymal stem cells after transient 53. Prodromidi EI, Poulsom R, Jeffery R, et al. Bone marrow-derived
ischemia. Stroke. 2008;39:1569-1574. cells contribute to podocyte regeneration and amelioration of renal
30. Mouiseddine M, Francois S, Souidi M, Chapel A. Intravenous human disease in a mouse model of Alport syndrome. Stem Cells. 2006;24:
mesenchymal stem cells transplantation in NOD/SCID mice preserve 2448-2455.

Вам также может понравиться