One of the most common complications of diabetes is diabetic foot ulcer. Diabetic ulcers do not heal easily due to diabetic neuropathy and reduced blood flow, and nonhealing ulcers may progress to gangrene, which necessitates amputation of the patient’s foot. This study attempted to develop a new cell-based therapy for nonhealing diabetic ulcers using a full-thickness skin defect in a rat model of type 2 diabetes and obesity. Allogeneic adipose-derived stem cells (ASCs) were harvested from the inguinal fat of normal rats, and ASC sheets were created using cell sheet technology and transplanted into full-thickness skin defects in Zucker diabetic fatty rats. The results indicate that the transplantation of ASC sheets combined with artificial skin accelerated wound healing and vascularization, with significant differences observed 2 weeks after treatment. The ASC sheets secreted large amounts of several angiogenic growth factors in vitro, and transplanted ASCs were observed in perivascular regions and incorporated into the newly constructed vessel structures in vivo. These results suggest that ASC sheets accelerate wound healing both directly and indirectly in this diabetic wound-healing model. In conclusion, allogeneic ASC sheets exhibit potential as a new therapeutic strategy for the treatment of diabetic ulcers.

The population of patients with diabetes is growing worldwide and reached ∼400 million in 2013 (1). An estimated 15–25% of patients with diabetes are at risk for developing a lower-extremity diabetic ulcer in their lifetime (2). Among patients with diabetic foot ulcers, 7–20% will subsequently require an amputation, and 85% of lower-extremity amputations in diabetic patients are caused by foot ulcers (3). An urgent need exists to develop new therapies for the treatment of diabetic wounds to prevent foot ulcers from leading to amputations.

Artificial skin is one of the commercially available treatments for full-thickness skin defects after debridement. Artificial skin typically comprises two layers: an outer silicone sheet layer and an inner collagen sponge layer that act as the epidermis and dermis, respectively (4). However, difficulties often are experienced with the use of artificial skin treatments for diabetic wounds in patients with neuropathy, impaired blood flow, or relatively large wounds. In these cases, the formation of neodermal tissue is delayed, thus prolonging the treatment period (5).

Recombinant basic fibroblast growth factor has been widely used in wound healing to promote angiogenesis and granulation. Although recombinant basic fibroblast growth factor successfully accelerates wound healing, it requires frequent administration because of its short half-life (6,7).

Cell-based therapy has emerged as a new application for the treatment of ulcers (8,9). In particular, mesenchymal stem cells (MSCs) exhibit an excellent potential for increasing the rate of wound healing given their self-renewal capacity, immunomodulatory effects, and ability to differentiate into various cell lineages (10). Adipose-derived stem cells (ASCs), a type of MSC, exhibit various advantageous properties, such as paracrine activity and angiogenic potential (11,12). ASCs have been used experimentally in wound-healing applications (13,14).

Single-cell suspensions of MSCs have been directly injected around wounds in numerous studies, and accelerated wound healing has been observed (15,16). Despite reports of improved wound healing in diabetic ulcer models following the injection of single-cell suspensions, the residence time of transplanted cells at the wound site is unclear. Yang et al. (17) reported that the injection of single-cell suspensions result in the formation of island-like aggregates and visible necrosis of the injected cells.

Cell sheet engineering improves the efficacy of cell transplantation. Okano et al. (18) reported that temperature-responsive culture dishes can be prepared by covalently grafting the temperature-responsive polymer N-isopropylacrylamide onto a culture dish surface. The grafted polymer layer permits temperature-controlled cell adhesion/detachment on the culture dish surface. Specifically, the surface is hydrophobic at 37°C, allowing cells to adhere and proliferate. In contrast, the surface becomes hydrophilic at <32°C, causing cells to spontaneously detach from the surface. These cells can be subsequently harvested as a contiguous cell sheet with intact cell-cell junctions and extracellular matrix (ECM). This approach avoids the use of proteolytic enzymes, such as trypsin, that damage the ECM, and the cell sheets can be immediately transplanted to the wound site (19).

Artificial skin has been used to achieve great therapeutic results, even in chronic wounds such as diabetic wounds (20). In addition, cellular artificial skin has been developed, and a significant body of evidence supports its use in the treatment of diabetic wounds (21). As previously stated, ASCs exhibit therapeutic potential; therefore, ASC sheets with artificial skin have the potential to accelerate chronic wound healing and offer a feasible therapeutic strategy for diabetic wound treatment. The efficacy of ASC sheets with artificial skin requires further evaluation in diabetic wounds with exposed bone. In this study, the artificial skin used provided a three-dimensional framework for ASCs, maintaining transplanted ASC sheets and wounds in a moist environment, preventing wounds from spontaneous contraction, and protecting wounds from infection and external forces (22). The aim of the current study was to evaluate the efficacy of allogeneic ASC sheet transplantation with artificial skin to promote the healing and vascularization of full-thickness skin defects in a rat diabetic wound model.

Animals

All experimental protocols were approved by the Animal Welfare Committee of Tokyo Women’s Medical University School of Medicine. Zucker diabetic fatty (ZDF) rats (ZDF-Leprfa/CrlCrlj) were used as a type 2 diabetic obesity model. Adipose tissue was isolated from Lewis rats (LEW/CrlCrlj) and EGFP rats [SD-Tg (CAG-EGFP)] and used to prepare cell sheets.

Isolation of Rat ASCs

Rat ASCs (rASCs) were isolated from the inguinal adipose tissue of Lewis rats (20–33 weeks old, male), which was processed according to a previously reported method (23). Briefly, the isolated adipose tissue was enzymatically digested with 0.1% type A collagenase (Roche Diagnostics, Mannheim, Germany) at 37°C for 1 h. The stromal vascular fraction was collected after centrifugation at 700g for 5 min. Cells in the stromal vascular fraction were plated on a 60-cm2 Primaria tissue culture dish (BD Biosciences, Franklin Lakes, NJ) and cultured in complete culture medium (MEM α with GlutaMAX [Invitrogen, Carlsbad, CA] with 20% FBS [Moregate Biotech, Bulimba, QLD, Australia] and 1% penicillin/streptomycin [Sigma-Aldrich, St. Louis, MO]) at 37°C in a 5% CO2 incubator. After 24 h, debris was removed by washing with PBS (Life Technologies, Grand Island, NY), and fresh medium was added. The cells were passaged with 0.25% trypsin-EDTA (Life Technologies) on day 3 and transferred to a new dish. Subcultures were plated at a density of 1.7 × 103 cells/cm2 every 3 days until passage 3.

rASC Culture and Characterization

rASCs were characterized by measuring their colony-forming, adipogenic, and osteogenic abilities using previously reported methods (24). For each assay, 100 rASCs at passage 3 were plated in a 60-cm2 dish and cultured in complete medium for 7 days. For the colony-forming assay, the cells were fixed with 4% paraformaldehyde (PFA) (Muto Pure Chemicals, Tokyo, Japan) and strained with 0.5% crystal violet in methanol for 5 min.

For adipogenesis, the medium was switched to an adipogenic medium consisting of complete medium supplemented with 0.5 μmol/L dexamethasone (Fuji Pharma, Tokyo, Japan), 0.5 mmol/L isobutyl-1-methyl xanthine (Sigma-Aldrich), and 50 μmol/L indomethacin (Wako Pure Chemical Industries, Osaka, Japan). After 14 days, the cells were fixed with 4% PFA for at least 1 h and stained for 2 h with fresh Oil Red O solution (Wako). For osteogenesis, the medium was switched to calcification medium consisting of complete medium supplemented with 100 nmol/L dexamethasone, 10 mmol/L β-glycerophosphate (Sigma-Aldrich), and 50 μmol/L ascorbic acid (Wako). The cells were incubated for 21 days and then stained with 1% alizarin red S solution. The number of stained colonies was counted for each assay (n = 3).

Flow Cytometry Assay

One million rASCs at passage 4 were suspended in 100 μL PBS containing 10 μg/mL fluorescein isothiocyanate–conjugated primary antibodies to characterize their surface marker expression (Table 1). After incubation for 30 min at 4°C, the cells were washed with PBS and then suspended in 1 mL PBS for analysis. Cell fluorescence was evaluated using a Gallios flow cytometer (Beckman Coulter, Tokyo, Japan), and data were analyzed using Kaluza for Gallios software (Beckman Coulter).

Table 1

Fluorescein isothiocyanate–conjugated antibodies used for flow cytometry

TargetCatalog numberIsotype controlSource
CD11b 554982 Mouse IgA BD Pharmingen* 
CD29 561796 Hamster IgM BD Pharmingen 
CD31 MCA1334FA Mouse IgG AbD Serotec 
CD45 MCA43FA Mouse IgG AbD Serotec 
CD90 MCA47FA Mouse IgG AbD Serotec 
TargetCatalog numberIsotype controlSource
CD11b 554982 Mouse IgA BD Pharmingen* 
CD29 561796 Hamster IgM BD Pharmingen 
CD31 MCA1334FA Mouse IgG AbD Serotec 
CD45 MCA43FA Mouse IgG AbD Serotec 
CD90 MCA47FA Mouse IgG AbD Serotec 

*BD Pharmingen, Franklin Lakes, NJ;

†AbD Serotec, Raleigh, NC.

Creation of rASC Sheets

rASCs derived from Lewis rats or EGFP rats at passage 3 were seeded on 35-mm diameter temperature-responsive culture dishes (UpCell; CellSeed, Tokyo, Japan) at a density of 1.5 × 105 cells/dish and cultured in complete medium for 3 days. The cells were then cultured in complete medium with 16.4 μg/mL ascorbic acid for an additional 4–5 days. After reducing the temperature to room temperature, the cells spontaneously detached as contiguous cell sheets and were harvested from the dishes with forceps.

Preparation of the Full-Thickness Skin Defect Wound Model and Transplantation of rASC Sheets

ZDF rats (n = 48; 16 weeks old; male; 520–600 g) were used as a rat wound-healing model for type 2 diabetes and obesity. rASC sheets from Lewis rats were used for transplantation (Fig. 1A–D). The blood glucose levels of the ZDF rats were measured using a blood glucose monitor (Glutest Neo Sensor; Sanwa Kagaku Kenkyusho, Nagoya, Japan), and their body weights were monitored both before the operation and at kill.

Figure 1

Schematic of the experimental procedure for transplanting an allogeneic rASC sheet with artificial skin into a rat wound-healing model of type 2 diabetes and obesity. A: Rat subcutaneous adipose tissue was surgically excised from Lewis rats or EGFP rats. rASCs were isolated and seeded onto a 60-cm2 Primaria tissue culture dish and cultured at 37°C for 7–8 days. B: Passage 3 rASCs were seeded onto a temperature-responsive culture dish and cultured at 37°C for 7–8 days. rASCs were harvested as an rASC sheet with intact cell-cell junctions and ECM by reducing the temperature to 20°C. C: rASC sheets from Lewis rats or EGFR rats were transplanted into a 15 × 10-mm full-thickness skin defect with exposed bone created on the heads of ZDF rats (n = 48). D: In the transplantation group, an rASC sheet was placed on the defect, and a 15 × 10-mm sheet of artificial skin (PELNAC) was overlaid on the transplanted rASC sheet (n = 24). In the control group, only the artificial skin was placed on the defect (n = 24). The defect with the artificial skin was closed with 10 stitches using 5-0 silk sutures. GFP, green fluorescent protein.

Figure 1

Schematic of the experimental procedure for transplanting an allogeneic rASC sheet with artificial skin into a rat wound-healing model of type 2 diabetes and obesity. A: Rat subcutaneous adipose tissue was surgically excised from Lewis rats or EGFP rats. rASCs were isolated and seeded onto a 60-cm2 Primaria tissue culture dish and cultured at 37°C for 7–8 days. B: Passage 3 rASCs were seeded onto a temperature-responsive culture dish and cultured at 37°C for 7–8 days. rASCs were harvested as an rASC sheet with intact cell-cell junctions and ECM by reducing the temperature to 20°C. C: rASC sheets from Lewis rats or EGFR rats were transplanted into a 15 × 10-mm full-thickness skin defect with exposed bone created on the heads of ZDF rats (n = 48). D: In the transplantation group, an rASC sheet was placed on the defect, and a 15 × 10-mm sheet of artificial skin (PELNAC) was overlaid on the transplanted rASC sheet (n = 24). In the control group, only the artificial skin was placed on the defect (n = 24). The defect with the artificial skin was closed with 10 stitches using 5-0 silk sutures. GFP, green fluorescent protein.

Close modal

ZDF rats were anesthetized by inhalation of 4% isoflurane (Pfizer Japan, Tokyo, Japan) and ventilated with a rodent mechanical ventilator (Stoelting, Wood Dale, IL). Then, 15 × 10-mm full-thickness skin defects were created on the heads of the ZDF rats by removing the cutaneous tissue from the epidermis to the periosteum (25), and the wound healing of the defects was observed at predetermined time points. ZDF rats were randomly divided into two groups—a control group and an rASC sheet transplantation group—to investigate the efficacy of cell sheets for wound healing. In the transplantation group, an rASC sheet was placed on the defect. The defects with or without an rASC sheet were covered with 15 × 10 mm of artificial skin (PELNAC; Smith & Nephew, Tokyo, Japan). Defects were closed with 10 stitches using 5-0 silk sutures (Alfresa, Osaka, Japan) (Fig. 1D). To protect the wound, 20 × 15 mm of nonadhesive dressing (Hydrosite Plus [known as ALLEVYN Non-Adhesive in the U.S.]; Smith & Nephew) was placed on top of the artificial skin with 5-0 silk sutures to maintain a moist wound environment and absorb exudate.

Transplantation of the rASC Sheet From EGFP Rats

EGFP-expressing rASCs were obtained from EGFP rats to track the fate of the transplanted cells. EGFP-expressing rASC sheets were created and transplanted into full-thickness defects in ZDF rats (16–18 weeks old; male; n = 2) using the aforementioned procedure.

Gross Wound Measurement and Complete Wound Closure Time

Gross wounds were observed and photographs taken at 0, 3, 7, 10, and 14 days (n = 12) after the operation and every 3–7 days thereafter (n = 6) until complete wound closure was observed (42 days). The wound area was measured by tracing the wound margin on the photograph and calculating the pixel data using ImageJ software (National Institutes of Health, Bethesda, MD). The time to complete wound closure was assessed (n = 6).

Tissue Preparation for Immunohistochemistry

Three rats from each group were killed 3, 7, 10, and 14 days after the operation. The wound areas were excised for histological analysis, and the blood vessel density was analyzed immunohistochemically. After fixation with 4% PFA for 24 h at 4°C, the trimmed specimens were decalcified with Morse’s solution (10% sodium citrate [Wako] and 22.5% formic acid [Wako]) (26) for 3 days at 4°C with gentle agitation. The Morse’s solution was exchanged daily. After decalcification, the specimens were rinsed with PBS, embedded in paraffin, cut into 6-μm–thick sagittal sections, and stained with hematoxylin-eosin (H-E). The sections of the wound area were then observed for histological analysis.

To quantify vascularization within the wound, blood vessel endothelial cells were immunohistochemically stained with an anti-CD31 antibody (rabbit polyclonal antibody; Thermo Fisher Scientific Anatomical Pathology, Fremont, CA). The specimens were pretreated through heating followed by blocking with 1% BSA (Sigma-Aldrich) and then incubated with the anti-CD31 antibody at 4°C overnight. After primary antibody staining, the specimens were washed with PBS, incubated with a secondary antibody (Alexa Fluor 488 Goat Anti-Rabbit IgG [H+L] Antibody; Life Technologies), mounted on coverslips with Prolong Gold, and stained with DAPI (Invitrogen). Serial sections of the specimens were observed with a fluorescence microscope (U-RFL-T; Olympus, Tokyo, Japan), and the images were analyzed with application software (DP2-BSW; Olympus).

The blood vessel densities of six animals in each group were determined 14 days after the operation by measuring the area of CD31-positive vessels in the wound area of the specimen. The center of the wound in each section was selected. The vessel area in the selected field of each specimen was then observed with a microscope (Eclipse E800; Nikon, Tokyo, Japan), and the area of CD31-positive vessels was quantified using ImageJ software. The relative area of CD31-positive vessels (VA) was calculated using Eq. 1:

formula
(Eq. 1)

where VAact and Af are the actual area of CD31-positive vessels and the total area of the field, respectively.

EGFP-expressing rASC sheets were transplanted to trace the fate of the transplanted rASC sheets. At 3, 5, 7, and 14 days after the transplantation, the rats’ chests were opened, and their vasculature was perfused from the left ventricle with 100 mL of 2% PFA in 0.01 mol/L PBS (pH 7.4) at a pressure of 120 mmHg. The wound tissue, including the skull bone and surrounding cutaneous tissue, was then removed and soaked in 4% PFA for 24 h at 4°C. The specimens were subsequently washed with PBS and decalcified with 10% EDTA weight for volume (Wako) in PBS for 5 days at 4°C with gentle agitation. The EDTA solution was changed daily. After decalcification, the specimens were rinsed >10 times with PBS, cryoprotected in an ascending series of sucrose-PBS (15 and 30 weight for volume percent), embedded in optimal cutting temperature compound (Sakura Finetek Japan, Tokyo, Japan), and snap-frozen in liquid nitrogen. Then, 14-μm–thick frozen sections were prepared with a cryostat (Leica CM1850; Finetek) and processed for immunohistochemistry. The frozen sections were first incubated with 4% Block-Ace (Dainippon Sumitomo Seiyaku, Osaka, Japan) and then incubated with rat antiendothelial cell antibody-1 (RECA-1) (Abcam, Cambridge, U.K.) as the primary antibody at a dilution of 1:200 in PBS containing 1% BSA (Sigma-Aldrich) at 4°C overnight. After several PBS washes, the specimens were incubated with a Cy3-conjugated secondary antibody at a 1:200 dilution (Jackson ImmunoResearch Laboratories, West Grove, PA) for 3 h at room temperature. Finally, the specimens were observed and photographed with a Keyence BZ-9000 fluorescence microscope (Keyence Corp., Osaka, Japan).

ELISA of the Supernatant From rASC Sheets

rASCs at passage 3 were seeded onto temperature-responsive culture dishes at a density of 1.5 × 105 cells/dish and cultured at 37°C for 72 h. The cells achieved subconfluence (80–90%) at 72 h after seeding, and this time point was defined as day 0. The medium was then collected and replaced with complete medium containing 16.4 μg/mL ascorbic acid on days 0, 2, and 4. The rASC sheets were transplanted on day 4 in this study. After the collected medium was centrifuged at 300g for 3 min at 4°C, the supernatant was collected, immediately frozen, and maintained at −80°C for further experiments.

The number of cultured cells per dish was assessed on days 0, 2, and 4 (n = 4). The levels of vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), transforming growth factor-β1 (TGF-β1), IGF-I, epidermal growth factor (EGF), and keratinocyte growth factor (KGF) were determined using RRV00, MHG00, MB100B, MG100 (R&D Systems, Minneapolis, MN), SE560Ra (Cloud-Clone Corp., Houston, TX), and CSB-E12905r (CUSABIO, Wuhan, China) quantitative ELISA kits, respectively. The protein concentrations were measured by duplicate ELISA assays, which were performed according to the manufacturer’s instructions. The amounts of the various growth factors in the supernatant were calculated to examine the amount secreted from the rASC sheets at each time point.

Statistical Analysis

Data are expressed as the mean ± SD. Comparisons between the two groups (i.e., transplant, control) were performed using Student t test. P < 0.05 was considered statistically significant.

Characterization of rASCs

To evaluate the characteristics of the rASCs, crystal violet, Oil Red O, and alizarin red S staining were performed to confirm rASC self-renewal, adipogenesis, and osteogenesis, respectively. rASCs exhibited colony-forming potential (Fig. 2A) as well as adipogenic and osteogenic differentiation potential in vitro (Fig. 2B and C), suggesting that the rASCs used in this study possessed MSC-like properties.

Figure 2

Confirmation of the characteristics and bipotency of rASCs obtained from normal Lewis rats. A: One hundred cells were cultured in a 60-cm2 dish for 7 days and stained with crystal violet to assess the number of cell colonies and confirm the self-renewal ability of rASCs. rASCs were able to differentiate into adipogenic and osteogenic lineages. B: After culture in adipogenic induction medium for 2 weeks, rASCs stained positively with Oil Red O. C: After culture in osteogenic induction medium for 4 weeks, the rASCs stained positively with alizarin red S, confirming their osteogenic differentiation potential. DH: The expression of surface antigens on the rASCs was analyzed by flow cytometry. rASCs at passage 3 exhibited a typical MSC phenotype because they were positive for CD29 and CD90 and negative for CD11b, CD31, and CD45.

Figure 2

Confirmation of the characteristics and bipotency of rASCs obtained from normal Lewis rats. A: One hundred cells were cultured in a 60-cm2 dish for 7 days and stained with crystal violet to assess the number of cell colonies and confirm the self-renewal ability of rASCs. rASCs were able to differentiate into adipogenic and osteogenic lineages. B: After culture in adipogenic induction medium for 2 weeks, rASCs stained positively with Oil Red O. C: After culture in osteogenic induction medium for 4 weeks, the rASCs stained positively with alizarin red S, confirming their osteogenic differentiation potential. DH: The expression of surface antigens on the rASCs was analyzed by flow cytometry. rASCs at passage 3 exhibited a typical MSC phenotype because they were positive for CD29 and CD90 and negative for CD11b, CD31, and CD45.

Close modal

Flow cytometry was used to characterize cell surface markers (Fig. 2D–H). rASCs strongly expressed CD29 and CD90. In contrast, low expression levels of CD11b, CD31, and CD45 were observed.

Blood Glucose Level and Body Weight of 16-Week-Old ZDF Rats

ZDF rats in each group exhibited an average blood glucose level >250 mg/dL, and these levels were ∼2.5-fold higher than those observed in 16-week-old nondiabetic rats. ZDF rats exhibited an average body weight of 578.2 g in the transplantation group and 582.6 g in the control group. These body weights were ∼1.5-fold higher than those observed in 16-week-old nondiabetic rats.

Gross Wound Area

Digital photographs were obtained 0, 3, 7, 10, and 14 days after the operation and every 3–7 days thereafter until complete wound closure (Fig. 3A–L). The average wound area was significantly smaller in the transplantation group than in the control group beginning on the 7th day (Fig. 4A). The wounds in both groups were digitally photographed and observed until complete wound closure (n = 6). The average observed wound closure time was significantly reduced in the transplantation group compared with the control group (Fig. 4B).

Figure 3

Macroscopic images of full-thickness skin defects. A macroscopic photograph of a typical full-thickness skin defect immediately after the creation of a 15 × 10-mm wound (A); the wound was covered with artificial skin using 10 stitches (B). Macroscopic photographs of full-thickness skin defects in the control (C, E, G, I, and K) and transplant (D, F, H, J, and L) groups at 3 (C and D), 7 (E and F), and 14 days (G and H) after creation of the wound (n = 12) and at 21 days (I and J) and 28 days (K and L) after creation of the wound (n = 6). White arrows indicate the area of scar-like appearance with complete epithelialization.

Figure 3

Macroscopic images of full-thickness skin defects. A macroscopic photograph of a typical full-thickness skin defect immediately after the creation of a 15 × 10-mm wound (A); the wound was covered with artificial skin using 10 stitches (B). Macroscopic photographs of full-thickness skin defects in the control (C, E, G, I, and K) and transplant (D, F, H, J, and L) groups at 3 (C and D), 7 (E and F), and 14 days (G and H) after creation of the wound (n = 12) and at 21 days (I and J) and 28 days (K and L) after creation of the wound (n = 6). White arrows indicate the area of scar-like appearance with complete epithelialization.

Close modal
Figure 4

A: Measurement and analysis of the wound area with or without rASC sheet transplantation. Macroscopic photographs of the wounds were digitized, and the wound area at 0, 3, 7, 10, and 14 days (n = 12) and 18 and 21 days (n = 6) after surgery was quantified using ImageJ software. The average wound areas at 0, 3, 7, 10, 14, 18, and 21 days after the operation were 1.56, 1.42, 1.23, 1.11, 0.87, 0.62, and 0.38 cm2 in the control group and 1.58, 1.30, 1.16, 0.93, 0.65, 0.40, and 0.25 cm2 in the transplantation group, respectively. The average wound area in the transplantation group 7 days after creation of the wound was significantly reduced compared with the control group, indicating accelerated wound healing in the transplantation group. B: The times required to complete wound closure of full-thickness skin defects are presented on a scatter diagram of the data for both the control and the transplantation groups (n = 6). The red bars on the diagram indicate the mean time to complete wound closure for both groups. The mean (range) times to complete wound closure were 34.2 (28–42) and 25.6 (18–35) days in the control and transplantation groups, respectively. The time to complete wound closure was significantly reduced in the transplantation group compared with the control group (P = 0.02). *P < 0.05.

Figure 4

A: Measurement and analysis of the wound area with or without rASC sheet transplantation. Macroscopic photographs of the wounds were digitized, and the wound area at 0, 3, 7, 10, and 14 days (n = 12) and 18 and 21 days (n = 6) after surgery was quantified using ImageJ software. The average wound areas at 0, 3, 7, 10, 14, 18, and 21 days after the operation were 1.56, 1.42, 1.23, 1.11, 0.87, 0.62, and 0.38 cm2 in the control group and 1.58, 1.30, 1.16, 0.93, 0.65, 0.40, and 0.25 cm2 in the transplantation group, respectively. The average wound area in the transplantation group 7 days after creation of the wound was significantly reduced compared with the control group, indicating accelerated wound healing in the transplantation group. B: The times required to complete wound closure of full-thickness skin defects are presented on a scatter diagram of the data for both the control and the transplantation groups (n = 6). The red bars on the diagram indicate the mean time to complete wound closure for both groups. The mean (range) times to complete wound closure were 34.2 (28–42) and 25.6 (18–35) days in the control and transplantation groups, respectively. The time to complete wound closure was significantly reduced in the transplantation group compared with the control group (P = 0.02). *P < 0.05.

Close modal

Histology

In low-magnification microphotographs of sagittal sections of H-E–stained specimens collected 14 days after the operation (Fig. 5A–D), a thin dermal layer was observed for the control group (Fig. 5A). In contrast, dense connective tissue was observed in the transplantation group (Fig. 5B). Higher-magnification microphotographs indicated the same results (Fig. 5C and D).

Figure 5

Histological analysis of wounds treated with rASC sheets. H-E–stained sagittal cross-sections of the control (A) and rASC sheet transplant (B) groups evaluated 14 days after surgery. The arrowheads indicate the epidermal margins. The region between the two arrowheads corresponds to the epidermal defect. Scale bars = 500 μm. C and D: Microphotographs display higher-magnification images of the boxed regions of the dermal layers in A and B, respectively. Scale bars = 100 μm. E and F: Quantitative analysis of neovascularization of the wounds. Control and transplant specimens were stained with CD31, a blood vessel endothelium cell marker (green). G: Blood vessel density in the wound area was measured at 14 days after wound creation and was calculated by dividing the area of CD31-positive vessels by the total area. The average CD31-positive area was 1.39% (SD 0.94) in the control group (E) and 3.77% (SD 1.74) in the transplantation group (F) (n = 6). The average blood vessel density in the transplantation group was increased by ∼2.5-fold compared with the control group, and this difference was statistically significant (P < 0.001). Scale bar = 50 μm. *P < 0.05.

Figure 5

Histological analysis of wounds treated with rASC sheets. H-E–stained sagittal cross-sections of the control (A) and rASC sheet transplant (B) groups evaluated 14 days after surgery. The arrowheads indicate the epidermal margins. The region between the two arrowheads corresponds to the epidermal defect. Scale bars = 500 μm. C and D: Microphotographs display higher-magnification images of the boxed regions of the dermal layers in A and B, respectively. Scale bars = 100 μm. E and F: Quantitative analysis of neovascularization of the wounds. Control and transplant specimens were stained with CD31, a blood vessel endothelium cell marker (green). G: Blood vessel density in the wound area was measured at 14 days after wound creation and was calculated by dividing the area of CD31-positive vessels by the total area. The average CD31-positive area was 1.39% (SD 0.94) in the control group (E) and 3.77% (SD 1.74) in the transplantation group (F) (n = 6). The average blood vessel density in the transplantation group was increased by ∼2.5-fold compared with the control group, and this difference was statistically significant (P < 0.001). Scale bar = 50 μm. *P < 0.05.

Close modal

Blood Vessel Density

Blood vessel density was quantified 14 days after the operation (Fig. 5E–G). The blood vessel density in the wound was increased by ∼2.5-fold in the transplantation group compared with the control group after 14 days, and the difference was statistically significant (n = 6) (Fig. 5G).

Number of rASCs Per rASC Sheet

The average number of cultured cells per dish was 8.76 × 105 cells on day 4 at the time of transplantation (Fig. 6A).

Figure 6

The number of rASCs per rASC sheet and the levels of growth factors secreted by an rASC sheet as measured by ELISA. rASCs achieved subconfluency (80–90%) in a 35-mm diameter dish 72 h after seeding, and this time point was considered as day 0 (A). The measured time points in the remaining panels (BG) were the same. A: The number of rASCs per cell sheet was assessed on days 0, 2, and 4 (n = 8). The average number of rASCs per rASC sheet was 3.88 × 105, 6.59 × 105, and 8.76 × 105 cells at days 0, 2, and 4, respectively (n = 8). The levels of VEGF, HGF, TGF-β1, IGF-I, EGF, and KGF were determined in vitro with quantitative ELISA kits for these growth factors. B: VEGF levels were 32.58, 13.90, and 18.60 ng/sheet/day at days 0, 2, and 4, respectively (n = 4). C: HGF levels were 1.32, 3.73, and 3.27 ng/sheet/day at days 0, 2, and 4, respectively (n = 4). D: TGF-β1 levels were 0.01, 0.49, and 0.76 ng/sheet/day at days 0, 2, and 4, respectively (n = 4). E: IGF-I levels were 0.04, 0.77, and 2.78 ng/sheet/day at days 0, 2, and 4, respectively (n = 4). F: EGF levels were 0.32, 0.47, and 0.44 ng/sheet/day at days 0, 2, and 4, respectively (n = 4). G: KGF levels were 0, 0.06, and 0.42 ng/sheet/day at days 0, 2, and 4, respectively (n = 4).

Figure 6

The number of rASCs per rASC sheet and the levels of growth factors secreted by an rASC sheet as measured by ELISA. rASCs achieved subconfluency (80–90%) in a 35-mm diameter dish 72 h after seeding, and this time point was considered as day 0 (A). The measured time points in the remaining panels (BG) were the same. A: The number of rASCs per cell sheet was assessed on days 0, 2, and 4 (n = 8). The average number of rASCs per rASC sheet was 3.88 × 105, 6.59 × 105, and 8.76 × 105 cells at days 0, 2, and 4, respectively (n = 8). The levels of VEGF, HGF, TGF-β1, IGF-I, EGF, and KGF were determined in vitro with quantitative ELISA kits for these growth factors. B: VEGF levels were 32.58, 13.90, and 18.60 ng/sheet/day at days 0, 2, and 4, respectively (n = 4). C: HGF levels were 1.32, 3.73, and 3.27 ng/sheet/day at days 0, 2, and 4, respectively (n = 4). D: TGF-β1 levels were 0.01, 0.49, and 0.76 ng/sheet/day at days 0, 2, and 4, respectively (n = 4). E: IGF-I levels were 0.04, 0.77, and 2.78 ng/sheet/day at days 0, 2, and 4, respectively (n = 4). F: EGF levels were 0.32, 0.47, and 0.44 ng/sheet/day at days 0, 2, and 4, respectively (n = 4). G: KGF levels were 0, 0.06, and 0.42 ng/sheet/day at days 0, 2, and 4, respectively (n = 4).

Close modal

Growth Factor Levels in the rASC Culture Supernatant

VEGF, HGF, TGF-β1, IGF-I, EGF, and KGF were present in the conditioned medium from cultured rASCs at passage 3 (Fig. 6B–G). Specifically, the levels of VEGF, HGF, TGF-β1, IGF-I, EGF, and KGF per rASC sheet were 18.60, 3.27, 0.76, 2.78, 0.44, and 0.42 ng/sheet/day at day 4 (n = 4), respectively, as determined using ELISA kits for each protein. These results confirmed that the rASC sheets were secreting growth factors at the time of transplantation.

Fate of the Transplanted rASCs

EGFP-expressing rASC sheets were transplanted into the wounds (Fig. 7A and B). Fourteen days after the transplantation, the transplanted EGFP-expressing rASCs (stained green [Fig. 7C]) and endothelial cells of regenerated blood vessels (stained red with RECA-1 [Fig. 7D]) were visualized. Overlaid microphotographs revealed that EGFP-expressing rASCs were located on the periphery of the RECA-1–positive blood vessels (Fig. 7E).

Figure 7

Transplantation of rASC sheets prepared from rASCs isolated from EGFP rats. Bright-field (A) and dark-field (B) photographs depict the wound immediately after transplanting an EGFP-positive rASC sheet and before overlaying the artificial skin. Fluorescent microphotography (B) confirmed that the transplanted EGFP-positive rASC sheet expressed green fluorescence (green). Scale bars = 10 mm. C: The differentiation fate of the transplanted rASC sheet, which expressed green fluorescence, was examined. D: Frozen sections of the wounds were immunostained with RECA-1 (red) to visualize blood vessel endothelial cells. E: An overlaid photograph of C depicting EGFP-positive rASCs (green) and D depicting RECA-1–positive endothelial cells (red) revealed that the EGFP-expressing rASCs (green) were located on the periphery of the RECA-1–stained areas (red).

Figure 7

Transplantation of rASC sheets prepared from rASCs isolated from EGFP rats. Bright-field (A) and dark-field (B) photographs depict the wound immediately after transplanting an EGFP-positive rASC sheet and before overlaying the artificial skin. Fluorescent microphotography (B) confirmed that the transplanted EGFP-positive rASC sheet expressed green fluorescence (green). Scale bars = 10 mm. C: The differentiation fate of the transplanted rASC sheet, which expressed green fluorescence, was examined. D: Frozen sections of the wounds were immunostained with RECA-1 (red) to visualize blood vessel endothelial cells. E: An overlaid photograph of C depicting EGFP-positive rASCs (green) and D depicting RECA-1–positive endothelial cells (red) revealed that the EGFP-expressing rASCs (green) were located on the periphery of the RECA-1–stained areas (red).

Close modal

Although streptozotocin-induced type 1 diabetic rats without obesity are most frequently used for diabetic wound-healing studies, this study used a wound-healing model with type 2 diabetes, which is known to affect >90% of patients with diabetes. Specifically, ZDF rats were used as a wound-healing model of type 2 diabetes and obesity in this study. These rats spontaneously develop obesity at ∼4 weeks of age and then develop type 2 diabetes at 8–12 weeks, exhibiting hyperglycemia combined with insulin resistance, dyslipidemia, and hypertriglyceridemia (27). Delayed wound healing, impeded blood flow in peripheral blood vessels, and diabetic nephropathy are also observed (2830).

ASC sheets have been used to accelerate wound healing in previous studies. Lin et al. (31) and McLaughlin and Marra (32) reported that transplantation of human ASC sheets with fibrin-coated membranes accelerates wound healing in a 12-mm–diameter full-thickness skin defect on the backs of 6-week-old athymic nude mice. Cerqueira et al. (33) reported that human ASC sheet transplantation promotes wound regeneration in 10-mm–diameter full-thickness skin defects on the backs of 5-week-old normal mice. In contrast to previous studies, the current study created a wider full-thickness skin defect with exposed bone (wound area 15 mm2) on the parietal region of 16-week-old ZDF rats. In addition, the periosteum, which may support the regeneration of the skin, was removed to increase the severity of the wound (25). Diabetic wounds with exposed bone are often observed clinically after debridement. To assess the clinical applicability of the method for severe diabetic foot ulcers, this study attempted to produce a wider full-thickness skin defect model with exposed bone and removed periosteum in rats with type 2 diabetes and obesity.

The differences in wound-healing mechanisms noted between humans and rodents are attributed to anatomical differences of the skin (e.g., panniculus carnosus layer). Normal rats exhibit contraction-based wound healing, whereas humans display re-epithelialization and granulation tissue formation–based wound healing. Typically, wound splinting in rodent models helps to minimize wound contracture, which allows for the gradual formation of granulation tissue (34). Slavkovsky et al. (28) reported that the contraction of diabetic wounds in ZDF rats is impaired, whereas nondiabetic wounds are almost entirely closed by contraction. Epithelialization also contributes to repair in this diabetic wound model in ZDF rats. Furthermore, the bilayer artificial skin we used in the current study prevented wound contraction and promoted new connective tissue matrix synthesis, resembling the true dermis (35). In the current study, artificial skin was placed on a recipient wound and fixed with nylon threads to reduce wound contraction and prevent wound enlargement as a result of the rat’s loose skin. Even if considerable contraction of the wound was observed, it appeared similar across all conditions between the control and the transplantation groups except in the presence or absence of ASC sheet transplantation. Under these conditions, significant differences in wound area and vascular density were observed between the groups. This experiment may be useful for evaluating ASC sheets.

Estrogen deficiency is associated with impaired cutaneous wound healing. Female rats exhibit a faster rate of wound healing with a higher rate of wound contraction because of their thinner skin (36). Accordingly, to reduce potential hormonal influences on wound healing, only male rats were used in this study.

ASCs exhibit several advantages compared with MSCs derived from other tissues (12). Adipose tissue is relatively abundant in the human body. These tissues are accessible and can be collected using a minimally invasive liposuction procedure. ASCs are also abundant in adipose tissue (11). Moreover, many reports have suggested that ASCs play an important role in accelerating wound healing (37,38). Thus, ASCs are considered a good cell source for stem cell therapies.

This study investigated whether allogeneic ASC sheets together with artificial skin are effective at accelerating wound healing. Xing et al. (39) reported that postoperative wound healing after laparotomy is impaired in obese rats, suggesting that the risk of laparotomy dehiscence may be increased by obesity. In clinical practice, patients with diabetic ulcers often exhibit severe diabetes complications, including uncontrolled high blood glucose and a high BMI. The collection of adipose tissue from patients with acute diabetic ulcers is difficult. Furthermore, after obtaining autologous ASCs, ASC sheet preparation requires several weeks. rASCs from diabetic animals exhibit altered properties and impaired functions (40). In fact, our group attempted to create rASC sheets using rASCs from ZDF rats, but cell proliferation was slow (data not shown). Given that MSCs, including ASCs, suppress immune responses (41), rASC sheets prepared from normal rats were transplanted into defects in ZDF rats to study allogeneic transplantation. In the current study, accelerated wound healing was observed in the transplantation group, and no obvious clinical signs of immune rejection, such as erythema, other local inflammatory signs, or visible signs of necrosis as described by Falanga et al. (42) and Briscoe et al. (43), were observed in the transplantation group during the experimental period.

rASC sheets were successfully created from Lewis rats exhibiting a wide range of ages (8, 10, 16, 20, and 33 weeks) (data not shown), and rASC sheets from 20–33-week-old rats were used for the transplantations. This study showed that rASC sheets from 33-week-old rats secreted growth factors and accelerated wound healing compared with the control group, suggesting that adipose tissue from middle-aged rats created functional rASC sheets. ASC sheets from patients over a wide range of ages could potentially be useful for treatment in clinical settings. In clinical practice, the age of patients undergoing liposuction operations ranges from young to elderly, and most patients are adults. This study intentionally used cells from middle-aged rats to simulate actual clinical practice.

In this study, rASC sheets contained ∼9 × 105 rASCs at the time of transplantation. A previous study based on the topical administration of MSCs in a collagen scaffold reported that 1 × 106 MSCs were transplanted into a 6-mm–diameter wound, which was the highest dose in that experiment. The highest percentage of wound closure and angiogenesis was achieved in 1 week with this cell dose compared with lower doses, suggesting that the wound-healing effects of MSCs may be dose dependent (44). In the current study, approximately the same number of rASCs was transplanted into larger defects (15 × 10 mm) as a cell sheet, and wound closure in the transplantation group was significantly accelerated 2 weeks after the operation. These results suggest that cell sheet technology might enhance the cellular activity and efficacy of cell transplantation because cell sheets are harvested with intact cell-cell junctions and undamaged ECM and can be immediately transplanted into the defect site (19). Moreover, a cell sheet is thin and exhibits sufficient flexibility to allow it to adhere well to the rough surface of a wound. In fact, cell sheet technologies have been used for the reconstruction of various tissues, with curative effects (45,46).

In this study, the time to complete wound closure was significantly faster in the transplantation group, and increased blood vessel density was confirmed in the transplantation group 14 days after transplantation compared with the control group. The regeneration and introduction of capillaries inside the wound are important because tissue regeneration commonly requires blood flow to supply oxygen and nutrition and remove waste. Increased blood vessel density at the wound site may help to promote wound healing.

Wound healing requires complex processes, including a coordinated interplay between cells and growth factors; it also requires multiple growth factors acting synergistically (47). A previous study reported that the rate of VEGF secretion from surrounding cells, such as macrophages and fibroblasts, is reduced in diabetic wounds (48). Numerous physiological factors contribute to wound-healing deficiency in individuals with diabetes (48).

In this study, various secreted growth factors (VEGF, HGF, TGF-β1, IGF-I, EGF, and KGF) played important roles in wound repair and were detected in large quantities in the conditioned medium from rASC sheets. ASCs secrete angiogenic growth factors (49), such as VEGF and HGF (50), and these growth factors could contribute to neovascularization (51,52) and accelerate wound healing in both normal and diabetic rats (15). These findings, which are consistent with the current results, suggest that the paracrine effects of transplanted rASCs might enhance neovascularization and wound epithelialization.

In this study, EGFP-expressing rASCs were surrounded with blood vessel endothelial cells 14 days after transplantation, suggesting that the rASCs remained at the wound site for at least 14 days and were able to differentiate into mural cells at the wound site and support the reconstitution of new blood vessels (53,54). MSCs originate from and are natively associated with blood vessel walls, suggesting that MSCs belong to a subset of perivascular cells (55,56). Some studies reported that ASCs have the potential to differentiate into endothelial cells (57,58). Also previously reported was that the majority of transplanted ASCs are adjacent to microvessels and differentiate into pericytes (59,60).

The results from this animal study demonstrate a promising approach for the application of ASC sheets together with artificial skin to accelerate diabetic wound healing. However, additional studies in larger animals are necessary to confirm and validate the efficacy of ASC sheets with artificial skin for clinical trials.

In conclusion, this study demonstrated that allogeneic transplantation of ASC sheets in combination with artificial skin accelerates wound healing in a rat model of type 2 diabetes and obesity. The rASC sheets displayed effective paracrine activity because they secreted angiogenic growth factors and potentially differentiated into perivascular cells to support the construction of new blood vessels. Thus, ASC sheets exhibited therapeutic value in this rat model and may be useful for accelerating the healing of diabetic ulcers in patients with type 2 diabetes and obesity.

See accompanying article, p. 2717.

Acknowledgments. The authors thank Toshiyuki Yoshida and Kaoru Washio of the Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, for valuable advice and suggestions; Hozue Kuroda of the Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, for excellent technical support; Taichi Ezaki of the Department of Anatomy and Developmental Biology, Tokyo Women’s Medical University School of Medicine, for expert technical advice and support in immunohistochemistry; Yukiko Koga of the Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, for practical advice; and Norio Ueno, of the Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, for English editing and valuable advice. They also thank Yuka Tsumanuma and Supreda Suphanantachat of Tokyo Medical and Dental University; Issei Komatsu and Daisuke Fujisawa of the Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University; Nana Mori of the Department of Odontology, Periodontology Section, Fukuoka Dental College; and Aya Matsui of the Department of Anatomy and Developmental Biology, Tokyo Women’s Medical University School of Medicine, for technical support. Finally, the authors thank Kazuki Ikura of the Diabetic Center, Tokyo Women’s Medical University School of Medicine, for expert advice on diabetic feet.

Funding. This study was supported by the Creation of Innovation Centers for Advanced Interdisciplinary Research Areas Program of the Project for Developing Innovation Systems “Cell Sheet Tissue Engineering Center (CSTEC)” from the Ministry of Education, Culture, Sports, Science and Technology (MEXT), Japan.

Duality of Interest. T.O. is a founder and director of the board of CellSeed, Inc., and holds technology licensing and patents from Tokyo Women’s Medical University. T.O. also is a stakeholder of CellSeed, Inc. Tokyo Women’s Medical University receives research funds from CellSeed, Inc. No other potential conflicts of interest relevant to this article were reported.

Author Contributions. Y.K. wrote the manuscript and researched data. T.I. and S.M. researched data, contributed to the discussions, and reviewed and edited the manuscript. M.Y., T.O., and Y.U. participated in discussions and reviewed and edited the manuscript. T.I. and T.O. are the guarantors of this work and, as such, had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Parts of this study were presented in abstract form at the 74th Scientific Sessions of the American Diabetes Association, San Francisco, CA, 13–17 June 2014.

1.
Aguiree
F
,
Brown
A
,
Cho
NH
, et al
.
IDF Diabetes Atlas
.
Brussels
,
International Diabetes Federation
,
2013
2.
Boulton
AJ
,
Vileikyte
L
,
Ragnarson-Tennvall
G
,
Apelqvist
J
.
The global burden of diabetic foot disease
.
Lancet
2005
;
366
:
1719
1724
[PubMed]
3.
Frykberg
RG
,
Zgonis
T
,
Armstrong
DG
, et al.;
American College of Foot and Ankle Surgeons
.
Diabetic foot disorders. A clinical practice guideline (2006 revision)
.
J Foot Ankle Surg
2006
;
45
(
Suppl.
):
S1
S66
[PubMed]
4.
Suzuki
S
,
Matsuda
K
,
Isshiki
N
,
Tamada
Y
,
Yoshioka
K
,
Ikada
Y
.
Clinical evaluation of a new bilayer “artificial skin” composed of collagen sponge and silicone layer
.
Br J Plast Surg
1990
;
43
:
47
54
[PubMed]
5.
Greenhalgh
DG
.
Wound healing and diabetes mellitus
.
Clin Plast Surg
2003
;
30
:
37
45
[PubMed]
6.
Tsuboi
R
,
Rifkin
DB
.
Recombinant basic fibroblast growth factor stimulates wound healing in healing-impaired db/db mice
.
J Exp Med
1990
;
172
:
245
251
[PubMed]
7.
Richard
JL
,
Parer-Richard
C
,
Daures
JP
, et al
.
Effect of topical basic fibroblast growth factor on the healing of chronic diabetic neuropathic ulcer of the foot. A pilot, randomized, double-blind, placebo-controlled study
.
Diabetes Care
1995
;
18
:
64
69
[PubMed]
8.
Kim
BM
,
Suzuki
S
,
Nishimura
Y
, et al
.
Cellular artificial skin substitute produced by short period simultaneous culture of fibroblasts and keratinocytes
.
Br J Plast Surg
1999
;
52
:
573
578
[PubMed]
9.
Blumberg
SN
,
Berger
A
,
Hwang
L
,
Pastar
I
,
Warren
SM
,
Chen
W
.
The role of stem cells in the treatment of diabetic foot ulcers
.
Diabetes Res Clin Pract
2012
;
96
:
1
9
[PubMed]
10.
Zannettino
AC
,
Paton
S
,
Arthur
A
, et al
.
Multipotential human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in vivo
.
J Cell Physiol
2008
;
214
:
413
421
[PubMed]
11.
Kern
S
,
Eichler
H
,
Stoeve
J
,
Klüter
H
,
Bieback
K
.
Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue
.
Stem Cells
2006
;
24
:
1294
1301
[PubMed]
12.
Casteilla
L
,
Planat-Benard
V
,
Laharrague
P
,
Cousin
B
.
Adipose-derived stromal cells: their identity and uses in clinical trials, an update
.
World J Stem Cells
2011
;
3
:
25
33
[PubMed]
13.
Zuk
PA
,
Zhu
M
,
Mizuno
H
, et al
.
Multilineage cells from human adipose tissue: implications for cell-based therapies
.
Tissue Eng
2001
;
7
:
211
228
[PubMed]
14.
Zuk
P
.
The ASC: critical participants in paracrine-mediated tissue health and function
. In
Regenerative Medicine and Tissue Engineering
.
Andrades
JA
, Ed.
Rijeka
,
InTech Open
,
2013
15.
Nie
C
,
Yang
D
,
Xu
J
,
Si
Z
,
Jin
X
,
Zhang
J
.
Locally administered adipose-derived stem cells accelerate wound healing through differentiation and vasculogenesis
.
Cell Transplant
2011
;
20
:
205
216
[PubMed]
16.
Shin
L
,
Peterson
DA
.
Human mesenchymal stem cell grafts enhance normal and impaired wound healing by recruiting existing endogenous tissue stem/progenitor cells
.
Stem Cells Transl Med
2013
;
2
:
33
42
[PubMed]
17.
Yang
J
,
Yamato
M
,
Kohno
C
, et al
.
Cell sheet engineering: recreating tissues without biodegradable scaffolds
.
Biomaterials
2005
;
26
:
6415
6422
[PubMed]
18.
Okano
T
,
Yamada
N
,
Sakai
H
,
Sakurai
Y
.
A novel recovery system for cultured cells using plasma-treated polystyrene dishes grafted with poly(N-isopropylacrylamide)
.
J Biomed Mater Res
1993
;
27
:
1243
1251
[PubMed]
19.
Yamato
M
,
Utsumi
M
,
Kushida
A
,
Konno
C
,
Kikuchi
A
,
Okano
T
.
Thermo-responsive culture dishes allow the intact harvest of multilayered keratinocyte sheets without dispase by reducing temperature
.
Tissue Eng
2001
;
7
:
473
480
[PubMed]
20.
Greaves
NS
,
Iqbal
SA
,
Baguneid
M
,
Bayat
A
.
The role of skin substitutes in the management of chronic cutaneous wounds
.
Wound Repair Regen
2013
;
21
:
194
210
[PubMed]
21.
Felder
JM
 3rd
,
Goyal
SS
,
Attinger
CE
.
A systematic review of skin substitutes for foot ulcers
.
Plast Reconstr Surg
2012
;
130
:
145
164
[PubMed]
22.
Priya
SG
,
Jungvid
H
,
Kumar
A
.
Skin tissue engineering for tissue repair and regeneration
.
Tissue Eng Part B Rev
2008
;
14
:
105
118
[PubMed]
23.
Watanabe
N
,
Ohashi
K
,
Tatsumi
K
, et al
.
Genetically modified adipose tissue-derived stem/stromal cells, using simian immunodeficiency virus-based lentiviral vectors, in the treatment of hemophilia B
.
Hum Gene Ther
2013
;
24
:
283
294
[PubMed]
24.
Yoshimura
H
,
Muneta
T
,
Nimura
A
,
Yokoyama
A
,
Koga
H
,
Sekiya
I
.
Comparison of rat mesenchymal stem cells derived from bone marrow, synovium, periosteum, adipose tissue, and muscle
.
Cell Tissue Res
2007
;
327
:
449
462
[PubMed]
25.
Koga
Y
,
Komuro
Y
,
Yamato
M
, et al
.
Recovery course of full-thickness skin defects with exposed bone: an evaluation by a quantitative examination of new blood vessels
.
J Surg Res
2007
;
137
:
30
37
[PubMed]
26.
Morse
A
.
Formic acid-sodium citrate decalcification and butyl alcohol dehydration of teeth and bones for sectioning in paraffin
.
J Dent Res
1945
;
24
:
143
153
27.
Kuhlmann
J
,
Neumann-Haefelin
C
,
Belz
U
, et al
.
Intramyocellular lipid and insulin resistance: a longitudinal in vivo 1H-spectroscopic study in Zucker diabetic fatty rats
.
Diabetes
2003
;
52
:
138
144
[PubMed]
28.
Slavkovsky
R
,
Kohlerova
R
,
Tkacova
V
, et al
.
Zucker diabetic fatty rat: a new model of impaired cutaneous wound repair with type II diabetes mellitus and obesity
.
Wound Repair Regen
2011
;
19
:
515
525
[PubMed]
29.
Oltman
CL
,
Coppey
LJ
,
Gellett
JS
,
Davidson
EP
,
Lund
DD
,
Yorek
MA
.
Progression of vascular and neural dysfunction in sciatic nerves of Zucker diabetic fatty and Zucker rats
.
Am J Physiol Endocrinol Metab
2005
;
289
:
E113
E122
[PubMed]
30.
Coppey
LJ
,
Gellett
JS
,
Davidson
EP
,
Dunlap
JA
,
Yorek
MA
.
Changes in endoneurial blood flow, motor nerve conduction velocity and vascular relaxation of epineurial arterioles of the sciatic nerve in ZDF-obese diabetic rats
.
Diabetes Metab Res Rev
2002
;
18
:
49
56
[PubMed]
31.
Lin
YC
,
Grahovac
T
,
Oh
SJ
,
Ieraci
M
,
Rubin
JP
,
Marra
KG
.
Evaluation of a multi-layer adipose-derived stem cell sheet in a full-thickness wound healing model
.
Acta Biomater
2013
;
9
:
5243
5250
[PubMed]
32.
McLaughlin
MM
,
Marra
KG
.
The use of adipose-derived stem cells as sheets for wound healing
.
Organogenesis
2013
;
9
:
79
81
[PubMed]
33.
Cerqueira
MT
,
Pirraco
RP
,
Santos
TC
, et al
.
Human adipose stem cells cell sheet constructs impact epidermal morphogenesis in full-thickness excisional wounds
.
Biomacromolecules
2013
;
14
:
3997
4008
[PubMed]
34.
Galiano
RD
,
Michaels
J
 5th
,
Dobryansky
M
,
Levine
JP
,
Gurtner
GC
.
Quantitative and reproducible murine model of excisional wound healing
.
Wound Repair Regen
2004
;
12
:
485
492
[PubMed]
35.
Matsuda
K
,
Suzuki
S
,
Isshiki
N
,
Ikada
Y
.
Re-freeze dried bilayer artificial skin
.
Biomaterials
1993
;
14
:
1030
1035
[PubMed]
36.
Dorsett-Martin
WA
.
Rat models of skin wound healing: a review
.
Wound Repair Regen
2004
;
12
:
591
599
[PubMed]
37.
Trottier
V
,
Marceau-Fortier
G
,
Germain
L
,
Vincent
C
,
Fradette
J
.
IFATS collection: using human adipose-derived stem/stromal cells for the production of new skin substitutes
.
Stem Cells
2008
;
26
:
2713
2723
[PubMed]
38.
Maxson
S
,
Lopez
EA
,
Yoo
D
,
Danilkovitch-Miagkova
A
,
Leroux
MA
.
Concise review: role of mesenchymal stem cells in wound repair
.
Stem Cells Transl Med
2012
;
1
:
142
149
[PubMed]
39.
Xing
L
,
Culbertson
EJ
,
Wen
Y
,
Robson
MC
,
Franz
MG
.
Impaired laparotomy wound healing in obese rats
.
Obes Surg
2011
;
21
:
1937
1946
[PubMed]
40.
Cianfarani
F
,
Toietta
G
,
Di Rocco
G
,
Cesareo
E
,
Zambruno
G
,
Odorisio
T
.
Diabetes impairs adipose tissue-derived stem cell function and efficiency in promoting wound healing
.
Wound Repair Regen
2013
;
21
:
545
553
[PubMed]
41.
Ali
MM
,
Li
F
,
Zhang
Z
, et al
.
Rolling circle amplification: a versatile tool for chemical biology, materials science and medicine
.
Chem Soc Rev
2014
;
43
:
3324
3341
[PubMed]
42.
Falanga
V
,
Margolis
D
,
Alvarez
O
, et al.;
Human Skin Equivalent Investigators Group
.
Rapid healing of venous ulcers and lack of clinical rejection with an allogeneic cultured human skin equivalent
.
Arch Dermatol
1998
;
134
:
293
300
[PubMed]
43.
Briscoe
DM
,
Dharnidharka
VR
,
Isaacs
C
, et al
.
The allogeneic response to cultured human skin equivalent in the hu-PBL-SCID mouse model of skin rejection
.
Transplantation
1999
;
67
:
1590
1599
[PubMed]
44.
O’Loughlin
A
,
Kulkarni
M
,
Creane
M
, et al
.
Topical administration of allogeneic mesenchymal stromal cells seeded in a collagen scaffold augments wound healing and increases angiogenesis in the diabetic rabbit ulcer
.
Diabetes
2013
;
62
:
2588
2594
[PubMed]
45.
Iwata
T
,
Washio
K
,
Yoshida
T
, et al
.
Cell sheet engineering and its application for periodontal regeneration
.
J Tissue Eng Regen Med
. 23 July
2013
. [Epub ahead of print]. DOI:
[PubMed]
46.
Elloumi-Hannachi
I
,
Yamato
M
,
Okano
T
.
Cell sheet engineering: a unique nanotechnology for scaffold-free tissue reconstruction with clinical applications in regenerative medicine
.
J Intern Med
2010
;
267
:
54
70
[PubMed]
47.
Barrientos
S
,
Stojadinovic
O
,
Golinko
MS
,
Brem
H
,
Tomic-Canic
M
.
Growth factors and cytokines in wound healing
.
Wound Repair Regen
2008
;
16
:
585
601
[PubMed]
48.
Brem
H
,
Tomic-Canic
M
.
Cellular and molecular basis of wound healing in diabetes
.
J Clin Invest
2007
;
117
:
1219
1222
[PubMed]
49.
Kim
WS
,
Park
BS
,
Sung
JH
, et al
.
Wound healing effect of adipose-derived stem cells: a critical role of secretory factors on human dermal fibroblasts
.
J Dermatol Sci
2007
;
48
:
15
24
[PubMed]
50.
Rehman
J
,
Traktuev
D
,
Li
J
, et al
.
Secretion of angiogenic and antiapoptotic factors by human adipose stromal cells
.
Circulation
2004
;
109
:
1292
1298
[PubMed]
51.
Nakagami
H
,
Maeda
K
,
Morishita
R
, et al
.
Novel autologous cell therapy in ischemic limb disease through growth factor secretion by cultured adipose tissue-derived stromal cells
.
Arterioscler Thromb Vasc Biol
2005
;
25
:
2542
2547
[PubMed]
52.
Asahara
T
,
Takahashi
T
,
Masuda
H
, et al
.
VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells
.
EMBO J
1999
;
18
:
3964
3972
[PubMed]
53.
Morikawa
S
,
Baluk
P
,
Kaidoh
T
,
Haskell
A
,
Jain
RK
,
McDonald
DM
.
Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors
.
Am J Pathol
2002
;
160
:
985
1000
[PubMed]
54.
Morikawa
S
,
Ezaki
T
.
Phenotypic changes and possible angiogenic roles of pericytes during wound healing in the mouse skin
.
Histol Histopathol
2011
;
26
:
979
995
[PubMed]
55.
da Silva Meirelles
L
,
Caplan
AI
,
Nardi
NB
.
In search of the in vivo identity of mesenchymal stem cells
.
Stem Cells
2008
;
26
:
2287
2299
[PubMed]
56.
Crisan
M
,
Yap
S
,
Casteilla
L
, et al
.
A perivascular origin for mesenchymal stem cells in multiple human organs
.
Cell Stem Cell
2008
;
3
:
301
313
[PubMed]
57.
Cao
Y
,
Sun
Z
,
Liao
L
,
Meng
Y
,
Han
Q
,
Zhao
RC
.
Human adipose tissue-derived stem cells differentiate into endothelial cells in vitro and improve postnatal neovascularization in vivo
.
Biochem Biophys Res Commun
2005
;
332
:
370
379
[PubMed]
58.
Planat-Benard
V
,
Silvestre
JS
,
Cousin
B
, et al
.
Plasticity of human adipose lineage cells toward endothelial cells: physiological and therapeutic perspectives
.
Circulation
2004
;
109
:
656
663
[PubMed]
59.
Traktuev
DO
,
Merfeld-Clauss
S
,
Li
J
, et al
.
A population of multipotent CD34-positive adipose stromal cells share pericyte and mesenchymal surface markers, reside in a periendothelial location, and stabilize endothelial networks
.
Circ Res
2008
;
102
:
77
85
[PubMed]
60.
Szöke
K
,
Brinchmann
JE
.
Concise review: therapeutic potential of adipose tissue-derived angiogenic cells
.
Stem Cells Transl Med
2012
;
1
:
658
667
[PubMed]