Vous êtes sur la page 1sur 11

Cell Tissue Res (2007) 329:301–311

DOI 10.1007/s00441-007-0417-3

REGULAR ARTICLE

Treatment of diabetic wounds with fetal murine


mesenchymal stromal cells enhances wound closure
Andrea T. Badillo & Robert A. Redden & Liping Zhang &
Edward J. Doolin & Kenneth W. Liechty

Received: 10 December 2006 / Accepted: 8 March 2007 / Published online: 24 April 2007
# Springer-Verlag 2007

Abstract Diabetes impairs multiple aspects of the wound- Keywords Stromal progenitor cells .
healing response. Delayed wound healing continues to be a Mesenchymal stromal cells . Mesenchymal stem cells .
significant healthcare problem for which effective therapies are Wound healing . Diabetes . Mouse (C57BKS strains)
lacking. We have hypothesized that local delivery of mesen-
chymal stromal cells (MSC) at a wound might correct many of
the wound-healing impairments seen in diabetic lesions. We Introduction
treated excisional wounds of genetically diabetic (Db-/Db-)
mice and heterozygous controls with either MSC, CD45+ Diabetic wound-related morbidity is a major clinical problem
cells, or vehicle. At 7 days, treatment with MSC resulted in a resulting in prolonged hospitalization, lost time from work,
decrease in the epithelial gap from 3.2±0.5 mm in vehicle- and significant healthcare expenditure. Normal wound healing
treated wounds to 1.3±0.4 mm in MSC-treated wounds and is an intricate process involving various cell types, coordinat-
an increase in granulation tissue from 0.8±0.3 mm2 to 2.4± ed processes, and complex signaling interactions. In diabetic
0.6 mm2, respectively (mean±SD, P<0.04). MSC-treated wound healing, many of these processes are impaired, and this
wounds also displayed a higher density of CD31+ vessels and intricate orchestration is disrupted. Alterations in growth-
exhibited increases in the production of mRNA for epidermal factor production, cellular responses to inflammatory media-
growth factor, transforming growth factor beta 1, vascular tors, matrix production, angiogenesis, and wound contraction
endothelial growth factor, and stromal-derived growth factor have all been shown to contribute to the delayed healing of
1-alpha. MSC also demonstrated greater contractile ability diabetic wounds (Blakytny and Jude 2006). Consequently, the
than fibroblast controls in a collagen gel contraction assay. effectiveness of treatments focusing on a single deficit tends
The effects of locally applied MSC are thus sufficient to to be limited. For example, the local delivery of platelet-
improve healing in diabetic mice. Possible mechanisms of derived growth factor (PDGF) requires repeated application
this effect include augmented local growth-factor production, to achieve modest improvements in wound closure (Wieman
improved neovascularization, enhanced cellular recruitment et al. 1998). The relative insufficiency of isolated growth-
to wounds, and improved wound contraction. factor delivery may also be related to a simultaneous lack of
responsive cellular targets within the wound (Falanga 2005).
A. T. Badillo : R. A. Redden : L. Zhang : E. J. Doolin : Because of the multifactorial etiologies of wound-healing
K. W. Liechty (*) impairment in these wounds, potential therapies should have
The Center for Fetal Research at The Children’s a broad impact on the wound-healing response and any
Hospital of Philadelphia,
associated problems with this response.
The University of Pennsylvania School of Medicine,
Philadelphia, PA 19104, USA Cell-based therapy is an attractive approach for the
e-mail: liechty@email.chop.edu treatment of wounds with multiple impairments. Cells are
biologically active and possess the machinery to provide a
K. W. Liechty
wide range of support to healing wounds and the capacity to
Abramson Research Center,
Rm 1116E, 3615 Civic Center Blvd, regulate responses according to the local wound environment.
Philadelphia 19104, USA Mesenchymal stromal cells (MSC) are multipotent cells
302 Cell Tissue Res (2007) 329:301–311

derived from the stroma of bone marrow and other tissues (da anesthetized with inhaled isofluorane after which the dorsal
Silva Meirelles et al. 2006). MSC have demonstrated a skin was shaved, depilatated, and cleaned with ethanol. Full
number of properties in vitro that can promote tissue repair, thickness wounds (through the panniculus carnosus) of
including the production of multiple growth factors, cyto- 8 mm were created on the midback of animals by using a
kines, collagens, and matrix metalloproteinases (Fathke et al. dermal punch (Miltex) and then covered with a Tegaderm
2004; Kim et al. 2005; Kinnaird et al. 2004; Parikka et al. dressing (3 M). Db-/Db- wounds were treated with either
2005; Pittenger et al. 1999) and the ability to promote 1×106 MSC, 1×106 CD45+ fetal liver cells, or vehicle
migration of other skin cells such as keratinocytes (Akino et (phosphate-buffered saline; PBS) alone. Non-diabetic
al. 2005). MSC have also been implicated as participators in wounds were treated with either 1×106 MSC or vehicle
normal tissue-repair processes based on their location in alone. Wound treatments (cells or vehicle alone) were
areas of tissue injury, such as injured spinal cord (S. Wu et al. administered immediately after wounding and took the form
2003), heart allografts in chronic rejection (G.D. Wu et al. of a group of four intradermal injections spaced in a radial
2003), and areas of dermal injury (Liechty et al. 2000). pattern from the wound edge in a total volume of 40 μl
Based on their in vitro properties, MSC have the potential to (10 μl per injection) and applied via a Hamilton syringe.
improve impaired wound healing through a variety of Experimental protocols were approved by the Institutional
mechanisms; however, the effects of MSC on cutaneous Animal Care and Use Committee and followed guidelines
wound healing have not been extensively tested in vivo. set forth in the National Institutes of Health Guide for the
Until recently, the inability to isolate and culture-expand Care and Use of Laboratory Animals.
enriched murine MSC populations has precluded the study of
the specific effects of MSC and sub-fractions of bone marrow Tissue processing and wound measurement
in established murine models of impaired wound healing.
Whole bone marrow has been used to treat wounds of patients At 3, 7, and 28 days after wounding, animals were euthanized,
with diabetes and peripheral vascular disease and have and the wounds were harvested. Full thickness skin was
demonstrated improved wound closure (Badiavas and Falanga excised and viewed under a Leica MZ16 FA fluorescent
2003). The precise cell type, within bone marrow, that is stereomicroscope for the presence of GFP. Wounds were then
responsible for this improvement has yet to be identified. We bisected. One half was fixed overnight in 10% neutral
have hypothesized that the local delivery of MSC to diabetic buffered formalin at 4°C, paraffin embedded, and sectioned
wounds might correct wound-healing impairment both indi- at 4 μm thickness. The other half was frozen in liquid nitrogen
rectly, by reversing local growth-factor deficiencies, and for RNA isolation.
directly, by improving wound contraction through interaction Sections were stained with hematoxylin and eosin for the
with the extracellular matrix. To test this hypothesis, we have measurement of the epithelial gap and the granulation tissue
isolated MSC from the livers of allogeneic fetal transgenic area by using Spot Advanced Version 4.5 software (Diagnos-
mice expressing green fluorescent protein (GFP). MSC have tic Instruments) by an investigator blinded to the group and
been used here to treat wounds created in a murine model of treatment. The epithelial gap was defined as the distance
obesity-related diabetes and non-diabetic control mice. Wound between encroaching epidermal elements (at least three cell
healing was assessed by measuring the epithelial gap, the layers thick). The granulation tissue area was defined as the
volume of granulation tissue, and growth-factor production. cellular area from the lateral transition zone from the normal
We have also examined the location and differentiation profile epidermis to the hypertrophic epidermis, inferior to the level
of MSC in wounds and the ability of MSC to promote of the panniculus carnosus and superior to the epithelial
contraction of the extracellular matrix in vitro. basement membrane or open wound surface.

Histology
Materials and methods
For immunoperoxidase staining, endogenous peroxidase
Animal wounding model activity was blocked with 0.3% hydrogen peroxide in
methanol. GFP was detected with rabbit anti-GFP primary
Twelve 14-week-old female C57BKS.Cg-m+/+Leprdb/J antiserum (1:100; Invitrogen) followed by biotinylated anti-
mice (Db-/Db-) and age-matched non-diabetic heterozy- rabbit secondary antibody (1:200) and development with
gous C57BKS mice (Db+/Db-) were used in wounding avidin-biotin complex (Vector Laboratories). Slides were
experiments (Jackson Laboratories). Db-/Db- mice weighed counterstained with hematoxylin. For double-immunofluores-
greater than 50 g, with blood glucose levels in excess of cent staining, sections were blocked with 1% sodium
400 g/dl, whereas Db+/Db- mice weighed an average of 22 g, borohydrate (Sigma) in PBS. CD31 was detected by using
with blood glucose levels less than 200 g/dl. Mice were rat anti-mouse CD31 antibody (1:20 dilution; Invitrogen)
Cell Tissue Res (2007) 329:301–311 303

followed by biotinylated rabbit anti-rat secondary antibody were thawed and expanded for a maximum of four passages
and detection with the alkaline phosphatase detection for use in experiments.
system (Vector Laboratories). GFP was detected by using the MSC characterization included the performance of fibro-
same rabbit anti-GFP primary antiserum followed by Alexa- blast-colony forming unit (CFU-f) assays, differentiation
Fluor-488-conjugated F(ab’)2 secondary antibodies (1:100; assays, and flow cytometric analysis of cell-surface antigen
Invitrogen). Slides were counterstained with 4,6-diamidino- profiles. CFU-f assays were performed in triplicate according
2-phenylindole (Vector Laboratories) and examined. Five to the manufacturer’s instructions (StemCell Technologies).
high-power fields per sample were counted. For alpha Osteogenic and adipogenic differentiation assays were carried
smooth muscle actin (α-SMA)/GFP double-staining, anti- out at passage 5 as previously described (Digirolamo et al.
α-SMA antibody directly conjugated to fluorescein isothiocya- 1999; Javazon et al. 2001). Flow cytometry was performed
nate was applied (1:200 dilution; Jackson ImmunoResearch) by using a FACS Calibur and CellQuest Pro Version 5.0.1 fl
followed by GFP staining as previously described. software. Primary antibodies tested included phycoerythrin
(PE)-conjugated antibodies with specificity for the surface
RNA isolation and polymerase chain reaction antigens CD3, CD11b, CD13, CD19, CD31, CD34, CD40,
CD44, CD45, CD73, CD90, c-Kit (CD117), MHC I, MHC
Total cellular RNA from wounds was isolated by using II, Sca–1, and purified anti CD49a (BD Biosciences). CD49a
TRIzol followed by preparation of cDNA with the was detected by PE-conjugated goat anti-Armenian hamster
SuperScript First-Strand Synthesis System (Invitrogen) IgG secondary antibody (Jackson ImmunoResearch).
according to the manufacturer’s instructions. The polymer- For CD45+ cell isolation, a single cell suspension of fetal
ase chain reaction (PCR) was performed with REDExtract liver cells was created and then incubated with CD45
N-Amp PCR ReadyMix (Sigma) and primers specific for microbeads. MACS-positive selection for CD45 cells was
murine epidermal growth factor (EGF), PDGF-BB, kerati- performed b using an autoMACS separator according to the
nocyte growth factor, transforming growth factor beta-1 manufacturer’s instructions (Miltenyi Biotech).
(TGF-β1), vascular endothelial growth factor (VEGF), Fibroblasts were isolated from age-matched fetal mice
interleukin-6 (IL–6), major intrinsic protein-2 (MIP2), IL– by explant culture. Fetal back skin was harvested, placed
10, placental-derived growth factor (PlGF), stromal-derived dermal side down onto tissue culture plastic, and grown in
growth factor 1–alpha (SDF–1α), and D-glyceraldehyde-3- Dulbecco’s Modified Eagle’s media (MEM) with 10% fetal
phosphate dehydrogenase (GAPDH; Table 1). Aliquots of bovine serum (Gibco). Cells migrating onto tissue culture
each reaction were subjected to gel electrophoresis in a 2% plates from the dermal surface were expanded and
agarose gel, and densitometry for individual sample cryopreserved at passage 1 for later use in experiments.
message level was performed by using Image J analysis
software (version 1.33 u) and standardized to GAPDH. Cell-populated collagen gel assay

Cell isolation and characterization On ice, a collagen solution (Type I from rat tail; BD
Biosciences, Bedford, Mass.) in 0.1% acetic acid was
MSC were isolated from fetal livers of embryonic day-15 adjusted to physiologic pH (7.6) and ionic strength with
gestational C57BL/6TbN (act-GFP) OsbY01 transgenic mice 1 N NaOH and 10× MEM respectively. A concentrated cell
expressing GFP. Fetal livers were mechanically disrupted into suspension of MSC was incorporated to achieve a final cell
a single cell suspension followed by the lysis of red cells with density of 2×105 cells/ml. The cell-populated solution was
ammonium chloride. For MSC isolation, cells were suspended added to 12-well dishes (750 μl/gel) and polymerized for
in “complete medium” containing MesenCult Basal Medium 1.5 h at 37°C. The gels were released by gentle tapping
for Murine Mesenchymal Stem Cells and Mesenchymal Stem and the use of a spatula into 6-well dishes containing 3 ml
Cell Stimulatory Supplements (StemCell Technologies) and complete growth media as previously described (Redden
plated at a density of 7.5×105 cells/cm2 at passage 0. Medium and Doolin 2006). The cell-populated collagen gels (CPCG)
was changed every 3 days until cells reached 90% confluency were incubated in a humidified incubator (5% CO2, 37°C).
(approximately 2 weeks), at which point, adherent fetal cells Digital images of the gels were taken daily with a Bio-Rad
were detached with 0.05% trypsin (Life Technologies/ Chemi Doc System by using Quantity One Software
Invitrogen). Cells were then enriched for MSC by CD11b (version 4.1; Bio-Rad Laboratories, Hercules, Calif.) and
immunodepletion by using streptavidin magnetic beads an in-picture centimeter ruler for scale. The images were
(Dynal Biotech) coated with biotinylated antibody against imported into image-analysis software (SigmaScan Pro,
CD11b (BD Biosciences) as previously described (Tropel et SPSS Science), and pixel-per-centimeter calibrations were
al. 2004). Depleted cells were expanded in complete growth made. The gel outlines were manually traced, and gel areas
media for 1 week and then cryopreserved at passage 1. MSC were calculated by the software in square centimeters.
304 Cell Tissue Res (2007) 329:301–311

Statistical analysis network within the subcutaneous fat (Fig. 1c), whereas
vehicle-treated controls had no such visible vessels (Fig. 1d).
Wound measurements are presented as the mean±SD. PCR Histologic examination of MSC-treated diabetic wounds
data were normalized to the percent of mRNA expression in confirmed the presence of a robust granulation tissue bed with
untreated control wounds and are presented as the mean± re-epithelialization and increased cellularity in the wound base
SD. Differences between experimental and control groups (Fig. 2a). In contrast, vehicle-treated control diabetic wounds
were determined by using the two-tailed Student’s t-test for were largely devoid of granulation tissue with little evidence
samples with unequal variance (Excel 2003, Microsoft). of re-epithelialization and less overall cellularity (Fig. 2b). In
Probability values of P<0.05 were used to denote statistical order to compare the effect of MSC with a cellular control, we
significance. isolated the CD45+ fraction of fetal liver and treated wounds
with an equal number of these cells (the CD45+ fraction
represents the MSC-negative portion of fetal liver and also
Results serves as a hematopoietic cellular control, being the predom-
inant cell population in whole bone marrow). Quantitative
Effect of MSC in diabetic wound healing analysis revealed that the epithelial gap of MSC-treated
diabetic wounds (1.3±0.9 mm) was significantly smaller than
To assess the early effects of MSC on diabetic wound healing, that of CD45+-treated wounds (4.1±0.7 mm, P<0.004) and
we chose the 7-day time-point for morphologic and quantitative vehicle-treated controls (3.2±0.5 mm, P<0.001, Fig. 2g).
analysis. At 7 days, gross examination of MSC-treated diabetic Indeed, the epithelial gap of CD45+-treated wounds was
wounds (n=10) revealed near closure with a fibrous tissue greater than that of vehicle-treated wounds, but this was not
filling of the wound base and evidence of wrinkling around statistically significant (P=0.06). The granulation tissue area
the wound edge implying wound contraction (Fig. 1a). By of MSC-treated diabetic wounds (2.4±0.6 mm2) was also
comparison, vehicle-treated wounds (n=10) appeared largely significantly higher than that of wounds administered vehicle
unhealed with a bare wound base and little evidence of new alone (0.8±0.3 mm2, P<0.05, Fig. 2h). There was no
tissue in-growth or wound contraction (Fig. 1b). The underside significant difference, however, in the volume of granulation
of MSC-treated diabetic wounds revealed a visible vascular tissue between MSC- and CD45+-treated wounds.

Fig. 1 Gross appearance


of MSC versus vehicle-treated
wounds at 7 days. Representa-
tive gross photographs of
wounds were taken at the time
of wound-tissue harvest.
a MSC-treated wound
exhibiting wrinkled skin around
the wound and fibrous tissue
growth filling the wound base.
b Largely unhealed wound
treated with vehicle alone.
c Underside of MSC-treated
wound demonstrating growing
vessels. d Vehicle-treated
wound; no vessels are visible in
the subcutaneous fat underlying
the wound base. Bars 5 mm
Cell Tissue Res (2007) 329:301–311 305

Fig. 2 Histologic analysis of MSC-, vehicle-, or CD45+-treated diabetic epithelial margin (boxed areas) demonstrating the encroaching epithelial
wounds. An 8-mm excisional diabetic wound was immediately treated tongue over top of cellular granulation tissue. g Epithelial gap of diabetic
with an intradermal injection of 1×106 MSC, 1×106 CD45+ fetal liver wounds treated with MSC (n=10; solid bar, MSC), CD45+ (n=10;
cells, or PBS vehicle alone, placed radially around the wound. Wounds striped bar, CD45), or vehicle alone (n=10; open bar, PBS). *P<0.001
were harvested at 7 days and stained with hematoxylin and eosin. vs vehicle, †P<0.004 vs CD45+. h Granulation tissue area (GTA) of
a MSC-treated wound. b Vehicle-treated wound. c CD45+-treated wound. diabetic wounds treated with MSC (n=10; solid bar, MSC), CD45+ (n=
The edge of the encroaching epithelium is indicated by black arrow- 10; striped bar, CD45), or vehicle alone (n=10; open bar, PBS). ‡P<0.05
heads. Bars1 mm. d-f Corresponding higher power views of the right vs vehicle

Effect of MSC on wound healing of non-diabetic mice used to confirm the presence GFP in the wound histologically
and to allow for a more precise determination of MSC location
In order to assess the effect of MSC on normal wound within the wounds. MSC were located within the dermis and
healing, non-diabetic (Db+/Db-) heterozygous mice were wound base. No GFP+ cells were seen in the epidermis
wounded in the same fashion and treated with MSC or PBS (Fig. 3c), and no GFP signal was detected in vehicle-treated
vehicle alone. At 7 days post-wounding, MSC-treated (n= control wounds either grossly or histologically (Fig. 3e,f).
10) and vehicle-treated (n=10) wounds had a similar To determine any potential differentiation of MSC in
histologic appearance, and there was no significant differ- wounds, we performed double-immunofluorescent staining
ence in the epithelial gap (MSC 3.7±0.5 mm vs PBS 4.0± for GFP and either CD31, an endothelial cell marker, or α-
0.8 mm, P=0.6) or granulation tissue area (MSC 3.9± SMA, a marker of myofibroblast differentiation. MSC-treated
1.6 mm2 vs PBS 2.6±1.4 mm2, P=0.3). diabetic wounds demonstrated higher levels of CD31 staining
(Fig. 4a) compared with vehicle-treated wounds (Fig. 4b).
Localization and differentiation of MSC in wounds Quantitative comparison revealed a significant increase in the
number of vessels per high-powered field in MSC versus
To determine the persistence of MSC, we analyzed wounds vehicle-treated wounds at 7 days (MSC 30.8±7.8 versus PBS
for MSC content, both grossly and histologically, by using 9.2±4.6, P<0.0005). In addition, GFP+ MSC were seen in
GFP as a cell marker. At 7 days post-wounding, fluorescent close association with CD31+ vessels, but no double-positive
stereomicroscopy of samples immediately after harvesting cells were identified (Fig. 4a). MSC in diabetic wounds,
demonstrated GFP expression both in the wound base and at however, did demonstrate double-positive staining for GFP
the wound margin (Fig. 3b). Immunoperoxidase staining was and α-SMA, consistent with myofibroblast differentiation
306 Cell Tissue Res (2007) 329:301–311

Fig. 3 Presence of GFP+ cells in treated diabetic wounds at 7 days. idase staining for GFP confirms the presence of GFP-expressing MSC
MSC and vehicle treated wounds were harvested at 7 days and in the wound margin located exclusively within the dermis. Bar100 μm.
visualized by using fluorescent stereomicroscopy and immunohisto- d-f Corresponding images of a vehicle-treated wound (8 mm long)
chemistry. a Representative gray-scale image of MSC-treated wound demonstrating minimal healing at 7 days and an absence of GFP signal
(8 mm long) demonstrating near closure. b Corresponding fluorescent by fluorescent stereomicroscopy and immunoperoxidase staining
image demonstrating GFP signal at the wound edge. c Immunoperox-

(Fig. 4c), whereas CD45+ cells stained positively for GFP wound healing, wounds were harvested 3 days after
only (Fig. 4d). wounding, and mRNA production for various growth
factors was analyzed. MSC-treated diabetic wounds
Analysis of wound closure exhibited increased mRNA production for EGF, TGFβ–
1, and VEGF compared with vehicle-treated controls and
MSC have the potential for induced differentiation into with CD45+-treated wounds (n=6 per group, P<0.05),
other cell types of the mesenchymal lineage in vitro. To whereas levels of PDGF-BB were not significantly
exclude the possibility of late-onset differentiation of MSC different (Fig. 6a). Because of the enhanced cellularity of
into other cell types of the mesenchymal linage, we ana- MSC-treated wounds, we also examined mRNA produc-
lyzed the histology of wounds at 28 days post-wounding. tion for factors important in cellular recruitment. Of the
Based on the morphologic appearance of healed wounds factors tested, there was a two-fold increase in SDF–1α
stained with hematoxylin and eosin, both MSC-treated mRNA. Levels of PlGF and the inflammatory cytokines
diabetic wounds and untreated diabetic control wounds MIP2 and IL–6 were not significantly elevated. In
were completely re-epithelialized, with an absence of contrast, CD45+-treated wounds demonstrated levels of
epidermal appendages, and typical scar formation with the chemotactic factors that were below those of controls
resolution of granulation tissue and increased connective (Fig. 6b).
tissue content compared with 7-day wounds (Fig. 5). In all To determine the level of SDF–1α mRNA expression
healed MSC-treated wounds examined, no histologic during normal wound healing, we analyzed SDF-1α mes-
evidence of bone or cartilage formation was noted (n=5, sage levels at 3 days in non-diabetic heterozygous animals
5 sections examined per animal). (Db+/Db-). Relative to GAPDH, SDF–1α mRNA xpression
was significantly higher in wounded Db+/Db- (1.5±0.2-fold)
Production growth factors and cytokines in MSC-treated compared with Db-/Db- animals (1.1±0.2-fold, P<0.05). In
wounds diabetic wounds treated with MSC, SDF-1α production was
similar (at 1.7±0.1-fold higher than GAPDH) to SDF–1α
To determine the contribution of MSC treatment to growth- production in non-wound-healing-impaired non-diabetic het-
factor production during the early proliferative phase of erozygote animals.
Cell Tissue Res (2007) 329:301–311 307

Fig. 4 MSC differentiation in wounds. To assess for endothelial exhibiting fewer CD31+ vessels. Bars50 μm. To assess for myofibro-
differentiation, wounds were harvested at 7 days and stained for both blast differentiation, sections were stained for α-SMA (green) and
GFP (green, white arrowheads) and CD31 (red, white arrows). GFP (red). c Double-staining for α-SMA and GFP (yellow, open
Representative images were taken of the wound base of MSC and arrowheads) in an MSC-treated wound, demonstrating myofibroblast
vehicle-treated control wounds. a MSC-treated wound demonstrating differentiation. d In CD45+-treated wounds, cells stained positive for
increased vascularity, with GFP cells located in close proximity to GFP only (red, white arrowheads). Other wound cells stained for α-
vessels. No double-positive cells were identified to indicate endothelial SMA only (green, white arrow). No double-positive cells were
cell differentiation by MSC. b Vehicle-treated wound in comparison identified. Bars 100 μm

Collagen gel contraction thought to mediate wound contraction, we chose age-


matched fetal fibroblasts for the control cell population in
To determine potential direct mechanisms by which MSC the gel contraction assay. At all time-points, MSC were
might improve diabetic wound healing, we utilized the free- more efficient than dermal fibroblasts with regard to
floating CPCG contraction assay. Since fibroblasts are contraction of the collagen matrix (P<0.05, Fig. 7).

Fig. 5 Histology of healed wounds at 28 days post-wounding. epithelialization and typical scar formation with no epidermal
Wounds were harvested from MSC- and vehicle-treated animals at appendages and an increased connective tissue content similar to that
28 days post-wounding and stained with hematoxylin and eosin. The of vehicle-treated wounds (b). No evidence of bone or cartilage
area of the healed wound was identified by the absence of epidermal formation was seen in MSC-treated wounds. Bars 50 μm
appendages. MSC-treated wounds (a) demonstrated complete re-
308 Cell Tissue Res (2007) 329:301–311

Fig. 6 Cytokine production in MSC versus CD45+-treated wounds at


Fig. 7 MSC-mediated extracellular matrix interactions. MSC were
3 days. Total cellular mRNA was isolated from wounds at 3 days post-
observed in a free-floating cell-populated collagen gel contraction
wounding, standardized to GAPDH for each sample, and analyzed by
assay and compared with fibroblast-seeded control gels. a Gel area
semi-quantitative PCR for the expression of various cytokines. mRNA
was calculated daily from digital images. At the same cell density,
levels from MSC-treated (black bars) and CD45+-treated (open bars)
MSC-seeded gels contracted faster then fibroblast-populated gels.
wounds are presented as the percent expression of vehicle-treated
b At individual time-points, the percent gel area of MSC-seeded gels
control wounds (dotted line control level of expression). a MSC-
was less than that with an equal density of fibroblasts, indicating
treated wounds demonstrated an elevation in message levels for
greater overall contraction of MSC-seeded gels. *P<0.05
epidermal growth factor (EGF), transforming growth factor beta 1
(TGF-β1), and vascular endothelial growth factor (VEGF) compared
with CD45+-treated wounds at P<0.05 (PDGF-BB platelet-derived the expansion of MSC yielding a dramatic increase in
growth factor). b MSC-treated wounds also exhibited a significant
elevation in stromal-derived growth factor 1 alpha (SDF–1α) mRNA
CD49a levels with >90% of cells expressing CD49a by
(IL-6 interleukin-6, MIP2 major intrinsic protein-2, PLGF placental- passage 5. MSC also demonstrated multipotent capacity for
derived growth factor) induced osteogenic and adipogenic differentiation and
spontaneous myofibroblast differentiation as defined by
the expression of α-SMA.
MSC isolation and characterization

MSC are identified by a combination of functional criteria Discussion


and by their cell-surface antigen profile. To date, no
specific MSC marker has been identified. We have demonstrated the improved healing and closure of
In order to test the adequacy of our MSC enrichment and diabetic wounds following the local application of MSC.
the reproducibility of our isolation protocol, we performed This improvement is characterized by enhanced epithelial-
CFU-f assays (a quantitative measure of mesenchymal ization, granulation tissue production, and neovasculariza-
progenitor content) and differentiation assays and charac- tion. These dramatic wound-healing effects might be
terized the cell-surface antigen profile of cells at each mediated through a combination of direct and indirect
passage (n=10). With our isolation protocol, the value of mechanisms, including myofibroblast differentiation and
CFU-f per 100 cells plated increased from 1.7×10−3% pre- promotion of wound contraction, augmented growth-factor
depletion to 2% immediately following CD11b depletion, a production, and enhanced cellular recruitment.
1,000-fold enrichment. In the wound environment, MSC differentiation is
Myeloid cells, identified as CD11b+, are the largest restricted to the mesenchymal lineage. MSC differentiation
contaminating cell population in MSC cultures. Following into myofibroblasts may contribute to the contracted smaller
CD11b depletion, the percentage of CD11b+ cells was less appearance of MSC-treated wounds compared with untreated
than 2%. The cell-surface antigen profile of the remaining diabetic wounds as early as 7 days post-wounding. This is a
cell population was CD31−, CD34−, and CD45−, but significant change from the typical pattern of Db-/Db- wound
strongly positive for CD44, CD90.2, Sca–1, and CD49a closure since, unlike normal rodent wound healing, Db-/Db-
(data not shown). Short-term culturing further selected for wound healing relies more on new tissue production than does
Cell Tissue Res (2007) 329:301–311 309

wound contraction to achieve wound closure (Sullivan et al. in MSC-treated wounds might not be MSC, but rather
2004). In addition to these in vivo observations, the endogenous cells that are induced by MSC to increase the
contractile abilities of MSC have been specifically tested production of growth-factor mRNA. Regardless of the
and compared with that of fibroblasts in a contraction assay. cellular source of growth factor, inappropriate growth-factor
In collagen gels, MSC demonstrate not only their ability to delivery can contribute to the establishment of a chronic
interact directly with extracellular matrix components, but inflammatory state. Therefore, it is important to note that all
also their greater contractile capacity than fibroblasts. MSC-treated wounds healed completely in our experiments,
Fibroblasts are generally regarded as the principal cellular with the full resolution of inflammation. MSC-mediated
mediators of wound contraction in response to growth increases in local growth-factor levels may provide a
factors and alterations in mechanical forces within the powerful therapeutic tool for the improvement of diabetes-
wound (Broughton et al. 2006). Therefore, the ability of related healing impairments.
MSC to generate contractile forces on the local wound Interestingly, MSC-treated wounds demonstrate signifi-
matrix may be superior to that of fibroblasts. In human cant increases in SDF–1α, one of the most important
diabetic ulcers, the failure of timely wound contraction is factors in the cellular recruitment of stem and progenitor
known to increase the risk of infection, to prolong inflamma- cells to tissues (Dar et al. 2006). MSC and inflammatory
tion, and to exacerbate wound-healing difficulties (Singer and cells have been shown to migrate in response to SDF–1α
Clark 1999). MSC-mediated decreases in wound size (Bhakta et al. 2006). More recently, SDF–1α has also been
through contraction alone might have the potential to offset shown to have a role in dermal wound healing and to be
diabetes-related wound-healing impairment significantly. down-regulated by inflammatory cytokines (Fedyk et al.
In contrast to other reports, we have not observed 2001). We have found a deficiency of SDF–1α in diabetic
epidermal or endothelial cell differentiation following wounds. These differences in SDF–1α production suggest
MSC treatment. To our knowledge, epidermal transdiffer- that the impairment in wound healing may be attributable in
entiation has only been described for human embryonic part to a defect in progenitor cell recruitment and/or
mesenchymal stem cell populations (M. Wu et al. 2006). In function. The deficiency of SDF–1α seen in diabetic
our study, MSC are distinct from the epidermal layer. To wounds may be explained by the down-regulation of SDF
assess for endothelial differentiation, we have chosen a in response to protracted inflammation in the wound. Of
time-period with robust granulation tissue and high vessel interest, MSC does not significantly improve the rate of
density but have failed to observe transdifferentiation wound healing in non-diabetic mice. Perhaps, wound
events. Although transduced progenitor cells might down- healing in these animals proceeds at a maximal rate, and
regulate GFP expression with terminal differentiation, our MSC do not have a detectable effect on healing in the
cell population is derived from transgenic animals, and setting of intact host-progenitor-cell recruitment and mobi-
therefore, GFP expression should be maintained in differ- lization to wounds. MSC-mediated increases in SDF–1α
entiated cell types. Previous reports of endothelial differ- production may facilitate the mobilization and recruitment
entiation come from in vitro observations, which are of of cells to healing wound for their contribution to tissue
unclear relevance to their actual differentiation potential in repair; however, since no specific MSC marker exists, we
vivo (Gang et al. 2006; Oswald et al. 2004). Claims of such have been unable to determine the proportions of these cells
transdifferentiation in vivo are extremely rare events in the that represent endogenous progenitor cells or other inflam-
absence of severe tissue injury and selection pressures matory cells.
(Nagaya et al. 2004) and are unlikely to make a significant The observed improvements in wound closure seen with
contribution to the therapeutic benefit of MSC observed in placement of MSC in the periphery of the wound is
this study. supportive of the importance of growth-factor production
MSC may also influence healing indirectly through local and paracrine-based mechanisms of MSC wound-healing
increases in the production of VEGF, EGF, TGFβ–1, and effects. In our study, we have used intradermal application
SDF–1α. Growth factors are important in modulating the of MSC, since this mode of delivery results in better cell
wound-healing response; however, the effect of a growth persistence than topical cell application in the wound base
factor is highly dependent on the concentration and timing of (data not shown). Since MSC have been shown to migrate
its delivery. Theoretically, a possible explanation for the in response to an SDF–1α gradient, and since the
success of MSC in our study is their persistence in the wound concentration of SDF–1α is likely to be the highest at the
during early wound healing and their potential for regulated site of tissue injury (Ceradini et al. 2004), we would expect
delivery of multiple growth factors in response to local that any migration of MSC would be toward the area of
environmental cues without the need for re-administration. maximal tissue injury within the wound. Delivery of cells
Whereas MSC have been shown to produce a variety of within a prefabricated biologic matrix is an attractive
growth factors in vitro, the cellular origin of growth factors approach to improving the retention of cells in the wound
310 Cell Tissue Res (2007) 329:301–311

base. However, the use of such scaffolds also has the creation of such constructs can be complicated and time-
potential to alter MSC properties and to elicit host consuming. Our isolation protocol produces a defined
inflammatory reactions (Pandit et al. 1999; Sung et al. reproducible MSC population in just 2 weeks. For our
2004). Further studies are required to determine the study, we have chosen a fetal liver source because of the
influence of different scaffolds on MSC properties and species-related difficulty in the isolation of MSC. Human
any overall wound-healing benefit. MSC, by comparison, are easily isolated in sufficient
Bone marrow cells have been hypothesized to be numbers from donor and from autologous bone marrow.
recruited to wounds and to participate in tissue repair, but Autologous derivation has the advantage of avoiding the
the specific type of bone marrow cell responsible for such transmission of infectious diseases and the need for
repair remains unidentified. Indirect evidence suggests the immunosuppression. The autologous isolation of fibro-
presence of a non-hematopoietic population of cells with blasts, however, requires the creation of a new wound to
tissue-repair functions (Badiavas et al. 2003; Fathke et al. obtain tissue in a patient with impaired healing. Such
2004; Opalenik and Davidson 2005). Identification of this practices have the potential to result in additional wound
tissue-repair population has been limited by an inability to morbidity.
isolate and characterize purified subpopulations from Locally applied MSC can overcome many of the
hematopoietic organs for subsequent transplantation stud- pathophysiologic abnormalities in diabetic wounds. This
ies. To address this question, we have compared the effect improvement appears to be mediated through direct and
of MSC with that of the hematopoietic fraction of fetal indirect mechanisms, including promotion of contraction
liver. We have chosen fetal liver as our tissue source through myofibroblast differentiation, direct interaction
because of the higher frequency of MSC and the ability to with the extracellular matrix, and augmentation of growth-
eliminate hematopoietic cell contamination by early immuno- factor production. Together, these mechanisms contribute to
depletion. Of note, our fetal liver MSC population is enhanced neovascularization and cellular recruitment.
uniformly stromal and entirely devoid of hematopoietic These cells may also be responsible for the tissue-repair
contamination, unlike early-passage adult murine bone- properties observed with regard to cell preparations from
marrow-derived MSC. We have observed that MSC-treated hematopoietic organs. Recognition of the impact of MSC
wounds heal better than wounds treated with the CD45+ on impaired wound healing has significant implications for
hematopoietic cell fraction of fetal liver. Indeed, CD45+- the development of novel therapeutic strategies to facilitate
treated wounds demonstrate a trend toward lower growth- wound closure, and further studies are needed to optimize
factor production and epithelialization compared with controls. this therapy and to compare directly its efficacy with that of
MSC also tend to persist in wounded skin, whereas CD45+ other cell-based therapies.
cells are cleared. This comparison directly supports the role
of stromal non-hematopoietic progenitor cells as being the
principal effectors of tissue repair within hematopoietic
organs. References
Of note, MSC properties may potentially vary depending
on the tissue of derivation. However, the only differences in Akino K, Mineda T, Akita S (2005) Early cellular changes of human
differentiation properties have been seen in MSC isolated mesenchymal stem cells and their interaction with other cells.
from adult spleen, umbilical cord blood, and adult liver (in ’t Wound Repair Regen 13:434–440
Anker PS in ’t, Noort WA, Scherjon SA, Kleijburg-van der Keur C,
Anker et al. 2003; Kern et al. 2006). In addition, a direct
Kruisselbrink AB, Bezooijen RL van, Beekhuizen W, Willemze
comparison of growth-factor profiles between human adult R, Kanhai HH, Fibbe WE (2003) Mesenchymal stem cells in
bone-marrow-derived and human adult adipose-derived human second-trimester bone marrow, liver, lung, and spleen
MSC has failed to show any differences (Wang et al. exhibit a similar immunophenotype but a heterogeneous multi-
lineage differentiation potential. Haematologica 88:845–852
2006). To date, no formal comparisons have been made
Badiavas EV, Falanga V (2003) Treatment of chronic wounds with
between bone-marrow-derived and other tissue-derived bone marrow-derived cells. Arch Dermatol 139:510–516
MSC preparations with regard to their tissue-repair proper- Badiavas EV, Abedi M, Butmarc J, Falanga V, Quesenberry P (2003)
ties. Such a comparison might be helpful in identifying the Participation of bone marrow derived cells in cutaneous wound
healing. J Cell Physiol 196:245–250
optimal tissue source for MSC-based wound-healing
Bhakta S, Hong P, Koc O (2006) The surface adhesion molecule cxcr4
applications. stimulates mesenchymal stem cell migration to stromal cell-
MSC offer several additional advantages over other cell- derived factor–1 in vitro but does not decrease apoptosis under
based therapies. Cultured keratinocytes and fibroblasts have serum deprivation. Cardiovasc Revasc Med 7:19–24
Blakytny R, Jude E (2006) The molecular biology of chronic wounds
been incorporated into scaffolds and bioengineered for use
and delayed healing in diabetes. Diabet Med 23:594–608
as wound coverage and vehicles for growth-factor delivery Broughton G 2nd, Janis JE, Attinger CE (2006) The basic science of
(Dinh and Veves 2006; Veves et al. 2001), although the wound healing. Plast Reconstr Surg 117:12S–34S
Cell Tissue Res (2007) 329:301–311 311

Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Oswald J, Boxberger S, Jorgensen B, Feldmann S, Ehninger G,
Kleinman ME, Capla JM, Galiano RD, Levine JP, Gurtner GC Bornhauser M, Werner C (2004) Mesenchymal stem cells can be
(2004) Progenitor cell trafficking is regulated by hypoxic gradients differentiated into endothelial cells in vitro. Stem Cells 22:377–384
through hif–1 induction of SDF–1. Nat Med 10:858–864 Pandit A, Ashar R, Feldman D (1999) The effect of TGF-beta
Dar A, Kollet O, Lapidot T (2006) Mutual, reciprocal sdf–1/cxcr4 delivered through a collagen scaffold on wound healing. J Invest
interactions between hematopoietic and bone marrow stromal Surg 12:89–100
cells regulate human stem cell migration and development in Parikka V, Vaananen A, Risteli J, Salo T, Sorsa T, Vaananen HK,
nod/scid chimeric mice. Exp Hematol 34:967–975 Lehenkari P (2005) Human mesenchymal stem cell derived
Digirolamo CM, Stokes D, Colter D, Phinney DG, Class R, Prockop osteoblasts degrade organic bone matrix in vitro by matrix
DJ (1999) Propagation and senescence of human marrow stromal metalloproteinases. Matrix Biol 24:438–447
cells in culture: a simple colony-forming assay identifies samples Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca
with the greatest potential to propagate and differentiate. Br J JD, Moorman MA, Simonetti DW, Craig S, Marshak DR (1999)
Haematol 107:275–281 Multilineage potential of adult human mesenchymal stem cells.
Dinh TL, Veves A (2006) The efficacy of apligraf in the treatment of Science 284:143–147
diabetic foot ulcers. Plast Reconstr Surg 117:152S–157S Redden RA, Doolin EJ (2006) Complementary roles of microtubules
Falanga V (2005) Wound healing and its impairment in the diabetic and microfilaments in the lung fibroblast-mediated contraction of
foot. Lancet 366:1736–1743 collagen gels: dynamics and the influence of cell density. In Vitro
Fathke C, Wilson L, Hutter J, Kapoor V, Smith A, Hocking A, Cell Dev Biol Anim 42:70–74
Isik F (2004) Contribution of bone marrow-derived cells to Silva Meirelles L da, Chagastelles PC, Nardi NB (2006) Mesenchymal
skin: collagen deposition and wound repair. Stem Cells 22: stem cells reside in virtually all post-natal organs and tissues. J
812–822 Cell Sci 119:2204–2213
Fedyk ER, Jones D, Critchley HO, Phipps RP, Blieden TM, Springer Singer AJ, Clark RA (1999) Cutaneous wound healing. N Engl J Med
TA (2001) Expression of stromal-derived factor–1 is decreased 341:738–746
by IL–1 and TNF and in dermal wound healing. J Immunol Sullivan SR, Underwood RA, Gibran NS, Sigle RO, Usui ML, Carter
166:5749–5754 WG, Olerud JE (2004) Validation of a model for the study of
Gang EJ, Jeong JA, Han S, Yan Q, Jeon CJ, Kim H (2006) In vitro multiple wounds in the diabetic mouse (Db/Db). Plast Reconstr
endothelial potential of human uc blood-derived mesenchymal Surg 113:953–960
stem cells. Cytotherapy 8:215–227 Sung HJ, Meredith C, Johnson C, Galis ZS (2004) The effect of
Javazon EH, Colter DC, Schwarz EJ, Prockop DJ (2001) Rat marrow scaffold degradation rate on three-dimensional cell growth and
stromal cells are more sensitive to plating density and expand angiogenesis. Biomaterials 25:5735–5742
more rapidly from single-cell-derived colonies than human Tropel P, Noel D, Platet N, Legrand P, Benabid AL, Berger F (2004)
marrow stromal cells. Stem Cells 19:219–225 Isolation and characterisation of mesenchymal stem cells from
Kern S, Eichler H, Stoeve J, Kluter H, Bieback K (2006) Com- adult mouse bone marrow. Exp Cell Res 295:395–406
parative analysis of mesenchymal stem cells from bone Veves A, Falanga V, Armstrong DG, Sabolinski ML (2001) Graftskin,
marrow, umbilical cord blood, or adipose tissue. Stem Cells a human skin equivalent, is effective in the management of
24:1294–1301 noninfected neuropathic diabetic foot ulcers: a prospective
Kim DH, Yoo KH, Choi KS, Choi J, Choi SY, Yang SE, Yang YS, Im randomized multicenter clinical trial. Diabetes Care 24:290–295
HJ, Kim KH, Jung HL, Sung KW, Koo HH (2005) Gene Wang M, Crisostomo PR, Herring C, Meldrum KK, Meldrum DR
expression profile of cytokine and growth factor during differ- (2006) Human progenitor cells from bone marrow or adipose
entiation of bone marrow-derived mesenchymal stem cell. tissue produce VEGF, HGF, and IGF-I in response to TNF by a
Cytokine 31:119–126 p38 MAPK-dependent mechanism. Am J Physiol Regul Integr
Kinnaird T, Stabile E, Burnett MS, Shou M, Lee CW, Barr S, Fuchs S, Comp Physiol 291:R880–R884
Epstein SE (2004) Local delivery of marrow-derived stromal Wieman TJ, Smiell JM, Su Y (1998) Efficacy and safety of a topical
cells augments collateral perfusion through paracrine mecha- gel formulation of recombinant human platelet-derived growth
nisms. Circulation 109:1543–1549 factor-BB (becaplermin) in patients with chronic neuropathic
Liechty KW, MacKenzie TC, Shaaban AF, Radu A, Moseley AM, diabetic ulcers. A phase III randomized placebo-controlled
Deans R, Marshak DR, Flake AW (2000) Human mesenchymal double-blind study. Diabetes Care 21:822–827
stem cells engraft and demonstrate site-specific differentiation Wu GD, Nolta JA, Jin YS, Barr ML, Yu H, Starnes VA, Cramer DV
after in utero transplantation in sheep. Nat Med 6:1282–1286 (2003) Migration of mesenchymal stem cells to heart allografts
Nagaya N, Fujii T, Iwase T, Ohgushi H, Itoh T, Uematsu M, during chronic rejection. Transplantation 75:679–685
Yamagishi M, Mori H, Kangawa K, Kitamura S (2004) Wu S, Suzuki Y, Ejiri Y, Noda T, Bai H, Kitada M, Kataoka K, Ohta
Intravenous administration of mesenchymal stem cells improves M, Chou H, Ide C (2003) Bone marrow stromal cells enhance
cardiac function in rats with acute myocardial infarction through differentiation of cocultured neurosphere cells and promote
angiogenesis and myogenesis. Am J Physiol Heart Circ Physiol regeneration of injured spinal cord. J Neurosci Res 72:343–351
287:H2670–H2676 Wu M, Yang L, Liu S, Li H, Hui N, Wang F, Liu H (2006)
Opalenik SR, Davidson JM (2005) Fibroblast differentiation of bone Differentiation potential of human embryonic mesenchymal stem
marrow-derived cells during wound repair. FASEB J 19:1561–1563 cells for skin-related tissue. Br J Dermatol 155:282–291

Vous aimerez peut-être aussi