Você está na página 1de 31

TISSUE ENGINEERING

Volume 13, Number 6, 2007


# Mary Ann Liebert, Inc.
DOI: 10.1089/ten.2006.0278

Autologous Bone MarrowDerived Cultured Mesenchymal


Stem Cells Delivered in a Fibrin Spray Accelerate
Healing in Murine and Human Cutaneous Wounds*

VINCENT FALANGA, M.D.,1,35 SATORI IWAMOTO, M.D., Ph.D.,1,3,5 MOLLY CHARTIER, M.D.,1
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

TATYANA YUFIT, M.D.,1,3 JANET BUTMARC, B.A.,1,3 NICHOLAS KOUTTAB, Ph.D.,2,3


DAVID SHRAYER, M.D., Ph.D.,1 and POLLY CARSON, C.W.S.1,3

ABSTRACT

The nonhematopoietic component of bone marrow includes multipotent mesenchymal stem cells (MSC)
capable of differentiating into fat, bone, muscle, cartilage, and endothelium. In this report, we describe the
cell culture and characterization, delivery system, and successful use of topically applied autologous MSC
to accelerate the healing of human and experimental murine wounds. A single bone marrow aspirate of
3550 mL was obtained from patients with acute wounds (n = 5) from skin cancer surgery and from
patients with chronic, long-standing, nonhealing lower extremity wounds (n = 8). Cells were grown in vitro
under conditions favoring the propagation of MSC, and flow cytometry and immunostaining showed a
profile (CD29+, CD44+, CD105+, CD166+, CD34, CD45) highly consistent with published reports of
human MSC. Functional induction studies confirmed that the MSC could differentiate into bone, carti-
lage, and adipose tissue. The cultured autologous MSC were applied up to four times to the wounds using a
fibrin polymer spray system with a double-barreled syringe. Both fibrinogen (containing the MSC) and
thrombin were diluted to optimally deliver a polymerized gel that immediately adhered to the wound,
without run-off, and yet allowing the MSC to remain viable and migrate from the gel. Sequential adjacent
sections from biopsy specimens of the wound bed after MSC application showed elongated spindle cells,
similar to their in vitro counterparts, which immunostained for MSC markers. Generation of new elastic
fibers was evident by both special stains and antibodies to human elastin. The application of cultured cells
was safe, without treatment-related adverse events. A strong direct correlation was found between the
number of cells applied (greater than 1106 cells per cm2 of wound area) and the subsequent decrease in
chronic wound size ( p = 0.0058). Topical application of autologous MSC also stimulated closure of full-
thickness wounds in diabetic mice (db/db). Tracking of green fluorescent protein (GFP)+ MSC in mouse
wounds showed GFP+ blood vessels, suggesting that the applied cells may persist as well as act to stimu-
late the wound repair process. These findings indicate that autologous bone marrowderived MSC can be
safely and effectively delivered to wounds using a fibrin spray system.

Departments of 1Dermatology, 2Pathology, and 3Center of Biomedical Research Excellence (COBRE), Roger Williams Medical
Center, Providence, Rhode Island.
Departments of 4Dermatology and 5Biochemistry, Boston University School of Medicine, Boston, Massachusetts.
*This work was supported by NIH grants DK067836 (VF) and P20RR018757 (VF).

1299
1300 FALANGA ET AL.

INTRODUCTION Roger Williams Medical Center. To determine the validity


of the topical delivery method and the safety of the ap-

T HERE IS AN IMPORTANT NEED to stimulate the healing of


acute and chronic wounds to a level that is not presently
possible with standard care measures or recently developed
proach, two types of human wounds were studied: acute
wounds resulting from the removal of a nonmelanoma skin
cancer from the trunk or limbs and considered likely to heal
innovative approaches. An area of research that holds prom- properly with secondary intention but not ideally suitable
ise for the treatment of difficult-to-heal wounds is stem cell for primary closure; and chronic wounds of the lower ex-
application. The bone marrow is an important source of he- tremities and feet, which had not been healing for a period
matopoietic stem cells that regularly regenerate components of at least 12 months in spite of appropriate standard care
of the blood, and nonhematopoietic stem cells including and more innovative approaches including topical appli-
mesenchymal stem cells (MSC). There are several poten- cation of PDGF and bioengineered skin products.
tial mechanisms by which autologous stem cells could sig- A single bone marrow aspirate (3550 mL) was taken from
nificantly contribute to wound healing. Under appropriate
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

each patients iliac crest. To prevent clotting, the aspiration


conditions stem cells can rejuvenate or rebuild tissue com- needle was primed with sterile heparin sulfate (1000 U/mL).
partments.1 Considerable evidence from our laboratory and The bone marrow aspirate was aliquoted immediately after
from others suggests that the resident dermal fibroblasts in collection. Each 4 mL of the aspirate was layered onto 3 mL
nonhealing wounds have acquired an abnormal phenotype of sterile Ficoll-Paque TM Plus (GE Healthcare Biosciences,
that is not conducive to appropriate tissue repair.25 Spe- Piscataway, NJ) in 15 mL conical tissue culture polypropyl-
cifically, fibroblasts cultured from nonhealing wounds are ene tubes (Becton Dickinson Labware, Franklin Lakes, NJ).
unresponsive to the action of certain growth factors, such as The resulting suspension was centrifuged at 400 g for 30
platelet-derived growth factor-BB (PDGF-BB)2 and trans- minutes at room temperature. The mononuclear layer at the
forming growth factor-b1 (TGF-b1).4,5 The unresponsive- interface between the Ficoll-Paque and plasma was removed
ness to TGF-b1 may be due to down regulation of type II and plated onto T-25 tissue culture flasks containing 7 mL
TGF-b receptors and decreased phosphorylation of key of media, consisting of basic MSC media supplemented
signaling molecules, including Smad3 and MAPK.5 Ac- with 10% mesenchymal stimulatory supplements (StemCell
cordingly, the addition and incorporation of stem cells to the Technologies, Vancouver, BC, Canada). The flasks were in-
wound, particularly if they are potentially capable of dif- cubated and kept at 378C, 5% carbon dioxide (CO2) for the
ferentiating into a number of cells types, is promising. initial 48 hours. The media were then removed and 7 mL
Recently, increasing focus is being placed on the use of of fresh media were added every 34 days. When the cell
bone marrowderived MSC. In a first but preliminary report, monolayers achieved confluence (generally 1 week later),
we showed that such autologous cultured cells could bring they were passaged to T-75 flasks and supplemented with
about closure of long-standing and hard-to-heal wounds.6 15 mL of new media every 34 days. Periodically, cell cul-
However, there are several difficulties encountered in the tures were tested for the presence of mycoplasma using the
use of this type of emerging technology. First, MSC or other Mycoalert mycoplasma detection assay (Cambrex Bio Sci-
stem cell types must be grown in sufficient numbers for ence, Baltimore, MD), and with appropriate positive and neg-
meaningful topical application. Proper characterization of ative controls. The measurements were made by performing
the cultured cells is also important. This step requires ap- a bioluminescent assay.
propriate collection and separation of the bone marrow and
the use of tissue culture techniques that can yield cells with a
stable phenotype. An even greater challenge is the actual Flow cytometry to characterize cultured cells
application of stem cells to a wound. The cells need to be Flow cytometry analysis was used to determine specific
delivered in a preparation that remains in contact with the markers on the cultured cells, from both mouse and human
wound bed and keeps them viable in the often-hostile wound bone marrow aspirates and preparations. Cells in suspen-
microenvironment. Ultimately, the cells have to enter the sion were mixed with fluorochrome-conjugated antibodies
wound to effect a therapeutic response. Using human acute against CD29, CD31, CD34, CD44, CD45, CD105, CD166,
and chronic nonhealing wounds, we provide in this report SCA-1 (BD Biosciences, St. Jose, CA). Appropriate isotype
promising solutions to these difficulties. The results also antibodies were used to control for nonspecific staining.
indicate that the cultured MSC can accelerate healing of Staining was performed according to manufacturers re-
both murine and human cutaneous wounds. commendations. Immunostained cells were acquired and an-
alyzed on a FACS Calibur flow cytometer (BD Biosciences),
using cellQUEST software (BD Biosciences). Cells were
MATERIALS AND METHODS first visualized on forward scatter versus side scatter, and a
gate was constructed around viable cells, thus eliminating
Culture of bone marrowderived cells nonviable cells and debris. To insure the stability of the
All clinical materials and components used in this study phenotype of cultured cells, flow cytometry was performed
were approved by the Institutional Review Board (IRB) of at passages 35 and again at passages 1011. On the day of
DELIVERY OF STEM CELLS TO HUMAN AND MURINE WOUNDS 1301

cultured cell application, cell monolayers in T-75 flasks adipogenic maintenance media alone. The cells were ob-
were trypsinized with 0.05% trypsin-EDTA (Invitrogen, served for oil droplet formation and were also stained with Oil
Carlsbad, CA), collected in regular mesenchymal media, Red O. This technique allowed bright orange-red staining of
and centrifuged for 5 minutes, 400 g at room temperature. fat and blue staining of nuclei. To better emphasize cell out-
The cells were then resuspended and washed twice with lines and nuclei, the cells were incubated with 1 mL of he-
sterile saline, repelleting at the same centrifuge conditions. matoxylin counterstain for 2.5 minutes. Pictures were taken
A sample of cell suspension was taken prior to the final spin using a Nikon Coolpix camera on an Olympus phase-contrast
for determining cell count using a hemacytometer and for inverted scope. Osteogenic induction requires cells aliquoted
calculating the number of cells delivered per cm2 of the into 6-well plates to be grown to confluence in osteogenic in-
wound surface. duction media. To measure calcium deposition as a marker
of bone formation, cells were washed, scraped off the plates,
and digested overnight with hydrochloric acid. A calcium re-
Immunohistochemistry for further characterization porter assay (Stanbio Total Calcium LiquiColor) was then
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

of cultured cells used to determine the amount of calcium in the cell lysate.
Per this procedure, 550 nm absorbance ratings were made us-
Immunohistochemistry was performed as previously de-
ing a spectrophotometer. Cells being induced into chondro-
scribed.7 Cells from passages 5 to 7 were trypsinized with
cytes were counted and spun into pellets containing 3104 to
0.05% trypsin-EDTA (Gibco, Grand Island, NY), diluted in
4105 cells. The pellets were supplemented with chondro-
MSC media, and allowed to adhere to sterile glass slides for
genic induction media containing TGF-b3 every 23 days for
15 minutes. These slides were then placed in 10015 mm
4 weeks and kept under standard conditions. The pellets were
dishes, covered with 10 mL of MSC media, and allowed to
then processed, externally stained with eosin, embedded, and
grow at 378C, 5% CO2 for 24 hours. The slides were washed
cut into 4-mm sections for staining with Safranin O. Known
in phosphate buffered saline (PBS), air-dried, fixed in cold
cartilage tissue was used as a positive staining control and cell
acetone, placed in 1 PBS, and immunostained. Cluster des-
pellets that had been kept in regular mesenchymal media
ignation marker primary antibodies included the following:
without induction served as negative controls.
monoclonal CD29 (Serotec, Raleigh, NC), monoclonal CD34
(Immunotech, Marseilles, FR), monoclonal CD44, mono-
clonal CD45, monoclonal CD105, polyclonal CD117 (Dako
Cytomation, Carpinteria, CA), monoclonal CD90 (Oncogene, Fibrin spray system to deliver cells topically
San Diego, CA). In addition, adjacent 4-mm tissue sections to wounds
from formalin-fixed biopsy specimens of the wound bed were
The fibrin delivery method made use of the Tisseel VH
also analyzed with identical antibodies for MSC markers,
fibrin sealant system and the Tissomat application device
as well as using a monoclonal antibody to human prolyl-4-
and spray set (Baxter Healthcare, Glendale, CA). This prep-
hydroxylase beta (Acris antibodies, Germany, distributed by
aration contains human fibrinogen and thrombin. The fol-
Novus Biologicals, Littleton, CO). To analyze whether tissue
lowing protocol was used to make a fibrin gel with final
regeneration might be occurring, we focused on the reestab-
concentrations of 5 mg/mL fibrinogen and 25 U/mL throm-
lishment of elastic fibers. This was done using special stains
bin, utilizing a 1 mL Tisseel VH kit (Baxter). This kit uti-
(Verhoeff-van Gieson) as well as a specific elastin monoclonal
lizes two liquid phases that can be either extruded through a
antibody obtained from Vision Biosystems/Novocastra (Nor-
dual chamber applicator or sprayed through the applicator
well, MA).
with an inert gas carrier. To make the thrombin component,
1 mL of calcium chloride (CaCl2) mixture from the kit was
added using a sterile syringe to the 1 mL bottle of thrombin,
Functional assays to determine the differentiation
and the mixture was allowed to dissolve. One part of this
capacity of cultured MSC solution was then added to 9 parts of sterile 30 mM CaCl2
Bone marrowderived cultured cells between passages in normal saline (0.9% sodium chloride [NaCl]). The
3 and 9 were tested for their ability to differentiate into os- fibrinogen/sealer protein component was made by adding
teocytes, chondrocytes, and adipocytes using specific dif- 1 mL of sterile normal saline to the sealer protein bottle. One
ferentiation assays (Cambrex). For positive controls an MSC part of this solution was then added to 9 parts of sterile
strain (Cambrex PoeticsTM human mesenchymal stem cells) normal saline. The protease inhibitor aprotinin included with
was utilized. Negative controls consisted of the patients cells the kit was not used. All solutions were used within 4 hours.
grown in standard MSC media, and were used to demonstrate The total volume of fibrin gel was predetermined by the size
that the phenotypic changes were nonspontaneous. Briefly, of the wound to be covered. At the time of application to
for adipocyte differentiation, cells were plated onto 6-well each wound, a small amount of the fibrin gel was placed on a
plates and grown to confluence. They were then exposed to tissue culture plate, covered in media, and incubated under
3 cycles of adipogenic induction media alternating with adi- standard conditions to verify and confirm cell viability and
pogenic maintenance media. Negative control cells received migration of the cells from the fibrin.
1302 FALANGA ET AL.

Mouse experiments to determine cell delivery


RESULTS
and efficacy with the fibrin spray system
For initial in vivo screening of cell delivery using the Cell culture and characterization, and delivery
fibrin spray, red fluorescencelabeled fluorescent protein in a fibrin spray
(FP) mouse MSC were delivered to wounds created in the
No adverse events occurred during the harvesting of bone
back of C57BL/6 mice. For red-fluorescence labeling, cells
marrow from patients, and separation of the bone marrow
were stained and washed using the PKH26 red fluorescent
aspirate using Ficoll and subsequent cell culture also pro-
linker kit (Sigma, St. Louis, MO) 2 hours prior to applica-
ceeded without problems. All cell cultures tested negative
tion. Just prior to wounding, the backs of mice were shaved
for mycoplasma. We first focused on the morphology of the
and anesthesia was administered. A 11.5 cm elliptical full-
cultured cells. This is shown in Figures 1AD. After pri-
thickness wound was made in the upper back of the mice. A
mary plating of the Ficoll-separated bone marrow aspirate
polymer film (Cavilon, 3M, St. Paul, MN) was used to cover
and change of media at 48 hours, fusiform or polygonal
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

the wound, followed by silicone dressings (Mepitel, Mol-


cells with multiple small, knob-like cytoplasmic projec-
nilcke, Sweden). The dressing was secured to the unwounded
tions could be seen adhering to the tissue culture plastic
dorsal skin with clips. On day 5, the mice were sacrificed
(Figure 1A). By approximately 5 days, many of the flasks
by CO2 inhalation. The dressing was removed and the
contained spindle-shaped cells. In many cases, the cells were
wounded area was excised. Frozen sections were prepared
extremely elongated, and in fact appeared to be aligning
by snap freezing the skin in isopentane on OTC mounting
themselves end to end along their long axis (Figure 1B).
compound. Thereafter, 5 mm sections were cut and observed
Cells for application to patients were used within the first 10
for red fluorescence. In other experiments, mouse MSC
passages. After multiple passages (>1012) the cell mor-
cultures were established from the bone marrow of green
phology became more bizarre and polygonal (Figure 1C).
fluorescent protein (GFP) mice (UBI-GFP/BL/6; Jackson
Occasionally and in very early passages (<5), one in ap-
Laboratory, Bar Harbor, ME) and db/db mice (BKS.Cg-m/
proximately 10 culture dishes, more complex structures
Leprdb/J, former name C57BLKS-m Leprdb, Jackson
formed, consisting of many cells adhering to one another
Laboratory).8 For actual wounding experiments, we used
along their sides and their long axis, appearing like a three-
female mice (2530 grams) under previously described
dimensional structure (Figure 1D). In general, within a week
conditions.3 The fibrin spray system was then used to spray
the cells became confluent. Cell proliferation slowed with
the GFP cells onto the wound. In experiments designed to
increasing in vitro passage, so that doubling times, initially
measure efficacy and tracking, we delivered GFP cultured
34 days, increased to 14 days or more with passages greater
MSC to full-thickness tail wounds of db/db mice and control
than 1012. When placed in tissue culture flasks within the
(db/) littermates using a model we have recently de-
fibrin polymer matrix described in the Materials and Meth-
scribed.3 Imaging of histological and immunofluorescent
ods section, the cells were able to migrate out of the fibrin
sections for these experiments was performed using the Zeiss
onto tissue culture plastic as early as 4 hours (Figure 1E).
Axioplan 2 system with the Axio CAM, and sections were
This migration from the fibrin polymer could also be seen
analyzed by filters for fluorescein isothiocyanate (FITC)
in vivo (see below). Indeed, we used the mouse wounds to
(510560 nm), light microscopy, Texas Red (645/75 nm),
identify an ideal and suitable make up of the fibrin gel. The
and DAPI (435475 nm). Using these filters, a composite
exact concentration of gel components was selected by ti-
photomicrograph was obtained, which helped to determine
tration to provide the most dilute system that would still form
the specificity of the green fluorescence.
an adherent semi-solid. Eventually, using the mouse wounds
for initial screening, the fibrin system we selected provided
a minimum amount of fibrin so that cell migration and gel
Statistical analysis breakdown would be maximized. We found that a final di-
The work on human subjects was analyzed by deter- lution of less than 5 mg/mL of fibrinogen and 25 U/mL of
mining the effect of each application of MSC and any fibrin was associated with a visible run-off of the sprayed
correlation with the cell numbers on subsequent change in solution, before a gel formed on contact with the wound.
wound size within the immediate 2 4 weeks following the Therefore, this was used as the most suitable concentration of
topical application. This analysis was done using the non- both fibrinogen and thrombin and for cell delivery. Increas-
parametric Spearman Rank Correlation with no Gaussian as- ing the concentration of thrombin, while keeping the fibrin-
sumption. For further analysis of the number of cells needed ogen concentration constant at 5 mg/mL also led to a suitable
for a clinically meaningful biological effect, the two-sided gel. However, it was our goal to minimize all reagents for
Fishers Exact Test was used, which helped to determine the application to wounds. As explained in the Materials and
effect of greater or less than 1106 cells/cm2 of the wound on Methods section, the protease inhibitor aprotinin was not
any decrease (at least 10%) in ulcer size. Comparisons with included in the spray system, since gel stability was not our
mice experiments were obtained using the Bonferroni Multi- goal and the purpose was to create a gel that would break
ple Comparisons Test. down relatively quickly and release cells.
DELIVERY OF STEM CELLS TO HUMAN AND MURINE WOUNDS 1303
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

FIG. 1. Morphologic appearance of bone marrowderived cul-


tured cells and their exit from fibrin. (A) Appearance of cultured
cells on tissue culture plastic by day 2 after seeding from Ficoll-
separated bone marrow aspirate. A variety of morphologies is
seen, ranging from round to spindle cells. (B) As early as day 5 in
culture, cells began to assume a spindle shape, often with cells
FIG. 3. Functional assays for differentiation of cultured MSC.
joined together along their long axis. (C) At later times in culture
(A) Representative results of bone formation, using calcium as-
(1012 passages), cells became larger and generally lost the
says, for four consecutively tested cultured cells from four dif-
spindle-shape morphology, and assumed a more bizarre polygonal
ferent patients. Positive (established strain of MSC) and negative
shape. (D) During early passage of cells grown on tissue culture
(human dermal fibroblasts) controls are also included in the graph.
plastic (up to 10 passages), one can often see extremely spindle cell
(B) Adipose tissue formation, using Oil Red O, of lipid droplets in
morphology, with the development of islands of three-dimensional
a representative example of cultured cells; the red-staining cells
structures. (E) Migration by 4 hours of spindle-shaped bone
indicate fat-laden cells. (C) Representative example of a pellet of
marrowderived cells seeded in culture as a spray of fibrin con-
cultured cells staining positive for cartilage, using Safranin O.
taining bone marrowderived cells. All figures are at a magnification
(D) Negative control for cartilage, using human dermal fibro-
of 10.
blasts. (E) Positive control for cartilage, using archival cartilage
tissue. Magnification 10 in all cases.

There is no single specific marker for MSC. Therefore,


for flow cytometry and immunohistochemistry a panel of
markers was chosen on the basis of published reports on
human MSC.9,10 In agreement with these reports, the bone
marrowderived cells in the present study demonstrated the
following characteristics and percentage positivity of sur-
face markers: CD29 (99%), CD44 (99%), CD90 (81%),
CD105 (99%), CD166 (99%), CD34 (1.5%), and CD45
(<1%). Therefore, the cultured cells were virtually negative
for CD34 and CD45. This overall flow cytometry profile
was consistently present in cells through passages 48 and
corresponded well with immunostaining of cells plated on
glass slides, which was performed on cells in passages 47.
The representative results for glass slides in Figure 2 were
quite consistent with the flow cytometry studies, indicating
positivity for MSC markers (CD29, CD44, CD90) and neg-
ativity for the hematopoietic marker CD34. It has been
reported that the most primitive bone marrow stromal cells
FIG. 2. Immunostaining of cultured cells for selected CD mark-
ers. Bone marrowderived cultured cells were grown on glass are CD34.11 The cells we cultured were consistently CD34
slides and immunostained with CD markers. These representative negative. While this may indicate that our cultured bone
examples show that the cultured cells were positive for MSC marrowderived cells may have had less stemness, it also
markers ((A) CD29, (B) CD44, (C) CD90) and negative for hemato- demonstrates that they were unlikely to be hematopoietic
poietic markers ((D) CD34). Magnification 10. precursors. To further demonstrate the stem cell characteristics
1304 FALANGA ET AL.

of the cultured cells, we performed functional assays to de- wound bed. Indeed, as Figure 4B shows, the spray could be
termine whether the cells could be induced to differentiate delivered to the wound in an upright position, and still
into osteocytes, adipocytes, and chondrocytes, which is an without run-off of the spray or of the formed gel. Generally,
established characteristic of MSC.12 Results are shown in the syringe was held at 458 to 608 from the wound bed and
Figure 3, which for a total of four representative cell strains in pointing from the edge toward the center of the wound. We
passages 39 confirmed the conversion of the MSC to a found that the use of CO2 flows greater than 5 psi would
phenotype of calcium production (Figure 3A), adipocyte and cause too forceful a spray and would result in splashing of
fat staining (Figure 3B), and cartilage formation (Figures 3C considerable amounts of the gel outside the wound area.
E). It should be noted that in Figure 3A, negative control refers Healing of the wound following a total of three applications
to human dermal fibroblasts, while positive control represents per patient at least 1 week apart occurred uneventfully and
established and commercially available MSC. by no later than week 8 (Figure 5). First, the wound bed
filled with granulation tissue by 2 weeks. Pain relief was
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

considerable, practically disappearing with the cell-based


application. By 6 weeks, the large full-thickness wound al-
Application of cultured MSC to human
most completely resurfaced (Figure 4C) and healed com-
acute wounds pletely a week later. Figure 4D shows that complete wound
Having determined that bone marrowderived cultured closure was durable, and that the wound remained healed at
cells consistently display characteristics of MSC and could week 12. Follow-up of patients has continued to show per-
migrate from the fibrin matrix in vitro and in murine wounds, sistent wound closure by 412 months after the procedure.
we next applied autologous cultured MSC to human wounds. Although the study did not have effectiveness as a primary
We tested this approach with a fibrin spray delivery of the outcome, some interesting observations emerged. Two of
cells to both acute (n 5) and chronic nonhealing wounds the four subjects enrolled had more than one wound, thus
(n 8). For the acute wounds, one subject had her bone allowing the use of cell-containing fibrin in one wound and
marrow aspirated but later declined to participate in the fibrin alone in the other. Figure 5A shows the healing tra-
study. Therefore, a total of four patients with postsurgi- jectory in these four patients. One patient (#3) had two
cal acute wounds were treated. As per protocol, the acute wounds, while another (#4) had three wounds. In each of
wounds consisted of defects left by removal of skin cancers these two subjects, one of the wounds was treated with MSC
(basal cell and squamous cell carcinomas) and that would in fibrin, while the other(s) was treated with the fibrin spray
not be ideally suited for primary closure or flaps.13 Patients alone. No delay in healing was observed with the use of cells
had their bone marrow aspirated approximately 2 weeks in these two subjects. All wounds healed completely between
before the surgery to allow for proper in vitro establishment weeks 7 and 8 after the surgery. Interestingly, however, the
of MSC cultures by the day of surgery. The cells were ap- one wound that was dramatically larger at baseline, right
plied immediately after the removal of the skin cancer by after surgery (Figure 5A, subject #1, also shown in Figure 4),
Mohs surgery, an established procedure that helps insure healed more rapidly than the smaller wounds and by week 7.
complete cancer removal and tumor-free margins. There- This finding suggests that in acute wounds, which generally
fore, the applied cells were in their first 24 in vitro pas- heal uneventfully, MSC application could lead to more ac-
sages at the first application. Up to three applications were celerated resurfacing.
performed during the clinical course of the wound and, as In the subjects with the acute wounds described above,
stated in the Materials and Methods section, cells were not biopsies of the wound bed were obtained at day 8 after the
used beyond the 10th in vitro passage. The total fibrin application of cultured cells. Using sequential and adjacent
volume (fibrinogen and an equal volume of thrombin) was histological sections, we then determined whether immuno-
no greater than 2 mL, and the same applies to the treatment staining for specific markers could help support the hypoth-
of chronic wounds (see below). These postsurgical acute esis that the cultured cells had indeed migrated from the
wounds received approximately 2106 cells/cm2. Figure 4 fibrin and were present in the superficial layers of the wound.
illustrates this general approach of MSC application in a Figure 6 shows representative immunostaining results using
representative acute wound (subject #1 of Figure 5). Figure antibodies to CD29, CD45, and prolyl hydroxylase, a spe-
4A shows the double-barreled syringe used to load the cells cific human fibroblast marker, in a subject who received
in the fibrinogen fraction of one barrel and the thrombin either fibrin plus cells (left two panels) or fibrin alone (right
solution in the other. A gentle push on the common plunger panel).14 Labels a, b, and c in Figure 6 refer to
while activating the CO2 gas (2.55.0 psi) flowing through magnifications of 4, 10, and 2, respectively. CD29 is
the tip of the syringe allowed an even mist of mixed fi- one of the markers for mesenchymal type of cells.9 We
brinogen and thrombin to reach the wound as a fibrin spray found CD29 immunostaining in the upper levels of the
(Figure 4B). The syringe was held approximately 13 cm wound bed, and unassociated with immunostaining for
away from the wound bed, and the delivered spray poly- CD45, the leukocyte common antigen and a marker that
merized very quickly to a gel consistency adhering to the was not present in our cultured cells (see previous para-
DELIVERY OF STEM CELLS TO HUMAN AND MURINE WOUNDS 1305
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

FIG. 4. Application of bone marrowderived cultured MSC to


acute human wounds. (A) The cells were applied directly to the
wound using a fibrin polymer spray delivered from a double-
barreled syringe. The arrows point to the individual barrels filled
with either thrombin or the cell-containing fibrinogen solution.
The tubing, attached to the common spray jet area below the
syringe, was connected to CO2. (B) Application of the cultured
cells to the wound, in this panel at baseline and immediately after FIG. 5. Rate of healing of the human wounds. The autologous
surgery, was done by pressing on the common plunger of the bone marrowderived cultured MSC were applied using a fibrin
double-barreled syringe shown in (A), and approximately 2 cm spray to both acute and chronic human wounds. (A) Healing
away from the wound bed. The inset shows the large wound on the trajectory of four subjects with acute wounds after removal of skin
back of the subject, who was sitting up. No run-off of the sprayed cancer. The numbers refer to the four individual patients, and
material is observed. (C) Appearance of the wound at week 6, or next to the numbers indicates wounds treated with either
showing complete filling of the wound bed and almost complete MSC in the fibrin spray or the fibrin spray alone, respectively. The
epithelial resurfacing. (D) Complete healing of the wound oc- dashed lines also represent the healing of wounds treated with
curred by week 7, and the wound remained healing by week 12, as fibrin alone. Patient #4 had one wound treated with cells, and two
shown. The pink area to the right of the healed wound indicates a with fibrin alone (-a and -b). (B) Healing trajectory of subjects
healed biopsy site. with chronic wounds treated with MSC in a fibrin spray.

with a specific antibody directed against human elastin.


graph). Similar spindle cells in the upper layer of the Comparable immunostaining for elastin was not observed
wound bed also stained with the prolyl hydroxylase fibro- in control biopsies (not shown).
blast marker (FibM). Taken together, these results suggest
that at least some of the cultured cells may have migrated
Application of cultured MSC to human
into the upper layers of the wound bed and possibly may
have differentiated into a fibroblast phenotype. In contrast,
chronic wounds
as shown in the right panel (c) of Figure 6 for the wound Chronic wounds are very difficult to heal. In the context
in the same subject treated with fibrin alone, the upper of studying the feasibility of our experimental approach in
layers of the wound bed do not contain a substantial density humans, we applied cultured MSC from autologous bone
of CD29 cells. Indeed, there is a deeper infiltrate that marrow to wounds of more than 1 year duration in the leg
appears to be CD29 and FibM positive and probably rep- and foot of eight subjects that had not healed with a number
resents endogenous cells that are participating in the heal- of therapeutic approaches, including standard care, topically
ing process. We also determined the possibility of tissue applied PDGF-BB, and living bioengineered skin constructs.
regeneration by focusing on the deposition of new elastic These chronic wounds were due to venous insufficiency or
fibers.1517 Since these wounds were full-thickness, one diabetic neuropathy, but there was no evidence of significant
would not expect the presence of elastic fibers in the upper arterial insufficiency. As with acute wounds, the patients
wound bed. However, as shown in Figure 7, definite elastic were treated with MSC delivered topically in a fibrin spray,
fibers were present when looked for both by the traditional using the same methodology with respect to fibrinogen and
Verhoeff-van Gieson special stain and by immunostaining thrombin concentrations and total volume of fibrin (no more
1306 FALANGA ET AL.

than 2 mL). Because chronic wounds show considerable var- wound were able to migrate from the fibrin matrix and had
iation in baseline size, we tracked very carefully the number a mesenchymal phenotypic morphology.
of cells delivered per cm2 of wound surface, which was
measured by computerized planimetry. A total of six sub-
Use of autologous MSC in full-thickness
jects with chronic wounds were evaluated from the eight
enrolled; two had to be excluded. One excluded subject was murine wounds
a woman with mild mental retardation who was found to Tracking of MSC in humans is obviously difficult because
chronically manipulate her foot wound. The other excluded of our inability to safely and reliably label cells prior to
subject was a man who was concomitantly found to have a application. Moreover, we could only biopsy the human
systemic malignancy and thus could no longer continue to wounds early on after MSC application, and not after com-
be in the protocol. No safety problems occurred during bone plete closure had occurred. Therefore, we next used mouse
marrow aspiration and the application of cultured MSC in a models of wounding to determine whether MSC would persist
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

fibrin spray. As with acute wounds, the protocol called for in the healed wound and lead to the formation of new
up to three applications of the stem cells. Figure 5B shows structures, and whether a biological effect could be demon-
the healing trajectory of the six patients, and indicates a trend strated in animals, such as the db/db diabetic mouse, which
toward a decrease in ulcer size or complete wound closure are known to heal with more difficulty. We approached this
by 1620 weeks. Figure 8 shows the example of one woman problem in the mouse in two different ways. We labeled
with a nonhealing venous ulcer of the ankle, complicated autologous bone marrow derived cultured MSC with a red
by rheumatoid arthritis, which achieved complete wound fluorescence dye (see Materials and Methods) and also
closure using this approach. Her wound had not healed in used GFP MSC. Figures 11A and B show the histological
over 10 years. Of the remaining five subjects, a mean wound hematoxylin and eosin appearance and the red fluorescence
area reduction of 40% was found in four, while one subject of adjacent tissue sections 5 days after the application of red
showed no change in the size of his wound. No adverse fluorescently labeled MSC to wounds made on the back of
events were noted. Of great importance is the healing re- C57BL/6 mice. As the figures indicate, the labeled cells are
sponse within the first 24 weeks after each application of able to penetrate into the wound bed. It should be noted that
MSC in the subjects with chronic wounds. We analyzed this some of these studies were done just prior to the application
by determining the number of cells applied per cm2 of total of MSC to human wounds, and actually were instrumental in
wound surface, as measured by computerized planimetry. determining the proper application technique and concen-
The graph in Figure 9 represents the correlation between the trations of fibrinogen and thrombin for the delivery of cells
number of MSC applied to chronic wounds and the percent in a spray system (see first part of Results section). However,
change in ulcer area at 2 4 weeks after each application wounds made in the back of mice have the drawback that
(n 17 data points). There was a very strong correlation they heal mostly by contraction and a large dead space makes
indicating that, the greater the number of applied cells, the it difficult to know for certain whether the applied cells will
larger the reduction in ulcer area. Thus, using Spearmen remain in place. Therefore, we turned to a full-thickness
Rank Correlation, we found r 0.6389 (corrected for ties) model we recently established and that is now being used by
with a 95% confidence interval of 0.8606 to 0.2135; other investigators also.18 This model consists of creating
p 0.0058. As also suggested by the data points in Figure 9, full-thickness wounds, down to fascia, on the dorsal aspect of
additional statistical evaluation showed that only applica- the tail, approximately 1 cm distal to the body.3 These exci-
tions of greater than 1106 cells/cm2 of the wound were sions take up to 3 weeks to heal in normal mice, and reflect
associated with a subsequent (24 weeks) decrease in ulcer resurfacing by epithelial migration rather than by contraction.
size of at least 10% (Fishers Exact Test two-sided; p Using this model in db/db mice and their control (db/) lit-
0.0345). termates, we delivered immediately after wounding a single
One of the treated subjects had bilateral plantar ulcers application of either fibrin alone or fibrin containing autolo-
from diabetes, and had his wounds treated with either the gous bone marrowderived cultured MSC. As with human
fibrin spray alone or cultured cells in a fibrin spray. Biopsies MSC, we determined mouse cluster designation markers
were taken from each of his plantar wounds, and sequential by flow cytometry and immunohistochemistry. The applied
adjacent sections were analyzed by immunostaining. Figure cells had the following profile: CD29, CD44, SCA-1,
10 shows that there was minimal or no overlap between CD34, CD45/LCA, CD31. Figure 11C shows the re-
CD45 and CD29 immunostaining in the wound bed of the sults in tail wounds in db/db mice and their control littermates
MSC-treated wound. Interestingly, CD29 immunostaining treated with either fibrin alone or MSC-containing fibrin. We
was dramatically present in the MSC-treated wound but not used the Bonferroni Multiple Comparisons Test to determine
in the one treated with fibrin alone. The dermal cells were the significance of the results. By day 10 after wounding, the
spindle shaped, a pattern that was also observed when se- control mice showed accelerated healing with MSC appli-
quential sections were immunostained with antibodies to a cation ( p < 0.01) compared to fibrin alone, while a strong
specific fibroblast marker (Figure 10). These results, as with trend but no statistically significant difference was detected in
the acute wounds, suggest that the cells delivered to the the db/db mice. By day 20 after wounding, MSC application
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

FIG. 6. Immunostaining of acute wounds at day 8 after MSC application. Sequential sections of formalin-fixed biopsy specimens were
immunostained for CD29, CD45, and prolyl hydroxylase as a specific marker for human fibroblasts (fibroblast marker: FibM). The a,
b, and c refer to magnifications of 4, 10, and 2, respectively. The blue staining is due to the marker blue ink applied to the top
of the biopsy specimens. The left two columns of photomicrographs represent sequential sections of a representative wound treated with
MSC in fibrin, while the right single column of photomicrographs represents a control wound treated with fibrin alone. For the MSC-
treated wounds, spindle-shaped cells positive for both CD29 and the fibroblast marker (FibM) are present in the superficial layers of the
wound bed, which is where the cultured cells were applied. The immunostained cells appear to be distinct from those positive for CD45,
the leukocyte common antigen (LCA). Conversely, as seen in the fibrin-only group, the superficial bed of the wound treated with fibrin
alone is cellular poor for CD29 and the fibroblast marker (FibM). Only the deep aspect of the wounds shows immunostaining for these
markers, most likely representing an endogenous and deeper cellular component.

FIG. 7. Elastic fibers in acute human wounds at day 8 after MSC


application. The left side of the pictures indicates the two different
methods of elastic fibrin staining. The Verhoeff-van Gieson stains FIG. 8. Application of bone marrowderived cultured cells to
the elastic fibers black, while the elastic fibers immunostained with human chronic wounds. (A) Nonhealing wound over the ankle of
a specific elastic antibody stain red. Arrows point to individual and a subject at baseline; (B) Third application of MSC in a fibrin
representative elastic fibers. The left upper and lower panels spray to the now healing wound; (C) At 3 months, the wound is
(A) and (C) represent control normal skin, while the right upper almost healed. The wound bed has filled in completely, and only a
and lower panels (B) and (D) are from the superficial wound bed to small eroded surface is present; (D) The wound then went on to
which the MSC had been applied. Using both stains, there appears heal. The photographs show final documentation of complete
to be definite new formation of elastic fibers. Magnification 10. wound closure at 6 months.

1307
1308 FALANGA ET AL.

ters and overlay of images to eliminate false positivity,


indicate a blood vessel that shows definite green fluores-
cence. These results suggest that the applied cells, at least
those that are GFP, may not persist in great numbers in the
healed wound. The acceleration of healing clearly present
with the application of MSC to db/db mice wounds indicates
that the MSC may play a stimulatory role in spite of the lack
of long-term persistence.

DISCUSSION
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

We report the successful culture and propagation of MSC


from human and mouse bone marrow for topical delivery to
autologous animal and human wounds. The establishment
of these cell cultures was rapid, and their characterized
MSC phenotype was evident and stable in culture, as shown
FIG. 9. Correlation of wound healing with the number of MSC by morphology, flow cytometry, immunostaining, and func-
applied. The graph represents the correlation between the number tional assays. We show that these cells can be successfully
of MSC applied to wounds in a fibrin spray and the percent change applied to the wound bed using a fibrin spray system. For
in ulcer area at 24 weeks after each application in patients with this, we modified the concentrations of both fibrinogen and
chronic wounds (n 17 data points). Analysis was done using thrombin so as to deliver a fine spray that polymerized to
Spearmen Rank Correlation; r 0.6389 (corrected for ties) with fibrin immediately and on contact with the wound bed.
the 95% confidence interval of 0.8606 to 0.2135; p 0.0058.
Using a very low CO2 flow for fibrin delivery, there was no
Application of greater than 1106 cells/cm2 of the wound was
highly associated with a subsequent (24 weeks) decrease of at
run-off of the preparation from the wound bed. MSC in the
least 10% in ulcer size (Fishers Exact Test two-sided; p 0.0345). fibrin spray remained viable and were able to migrate from
the fibrin matrix, as determined by both in vitro and in vivo
studies. Thus, the application of autologous cultured MSC
led to a statistically significant difference from fibrin alone to human acute and chronic wounds is safely accomplished.
in the db/db mice ( p < 0.001), and this difference continued The applied cells appear to establish themselves in the
to be present by day 25 ( p < 0.05). The application of fibrin wound bed. A decrease in size of the chronic wounds cor-
alone did not stimulate healing when compared to air-exposed related very strongly and with a great degree of statistical
wounds ( p > 0.05, not shown). significance with the number of MSC applied per cm2 of
In other mouse tail-wound experiments designed to track the wound surface. Indeed, a concentration of 1106 cells
the fate of topically delivered cells, we applied mouse au- per cm2 was clearly required to stimulate a decrease in
tologous GFP MSC immediately after tail wounding and wound size. Animal studies showed that mouse autologous
analyzed frozen sections from the wound site for the pres- bone marrowderived MSC can accelerate the healing of
ence of green fluorescence. Because of the very friable full-thickness wounds in db/db mice as well as their con-
nature of the wounded site at early time points, which made trol littermates. Tracking of GFP MSC in full-thickness
it difficult to obtain intact sections, we took shave biop- mouse wounds suggests that most of these cultured cells may
sies for frozen sections at later times and immediately af- not persist, in spite of the stimulation of wound healing. Taken
ter wound closure. For imaging, we used different filters together, these results indicate the feasibility of using a
capable of detecting false green fluorescence positivity modified fibrin spray system to deliver cells to wounds, and
from GFP (see legend to Figure 12). Thus, we used filters for also offer considerable promise that bone marrowderived
FITC (510560 nm), light microscopy, Texas Red (645/ cultured MSC can accelerate healing. Of crucial importance is
75 nm), and DAPI (435475 nm). Using these filters, we that our studies were performed not only in experimental
were also able to construct composite photomicrographs, animal wounds but also in difficult-to-heal human wounds.
again to confirm the specificity of any green fluorescence. There is substantial and increasing interest in the use of
We found that, by day 18 after wounding, definite clusters of adult stem cells to accelerate healing.1,19,20 While there have
GFP cells could no longer be demonstrated at the wound been advances in the treatment of difficult-to-heal wounds
site, and only rare individual GFP cells, not due to auto- in the last few years, there is still a considerable percentage
fluorescence, could be detected. Similarly, keratinocytes (up to 50%) of chronic wounds, particularly those that are of
were not GFP. Interestingly, however, we uncommonly more than 1 year in duration, that remain unresponsive to
found stable endothelial structures that were clearly GFP advanced treatment approaches.21,22 With the inherent dif-
(approximately one per 20 high-power fields). The repre- ficulties involved in using embryonic stem cells, both from
sentative photomicrographs in Figure 12, using different fil- the technical and regulatory standpoints, adult autologous
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

FIG. 12. Identification of GFP MSC in mouse wounds. Photo-


FIG. 10. Immunostaining of chronic wounds before and at micrographs of C57BL/6 mouse tail wounds 18 days after wounding.
week 3 after MSC application. Sequential sections of formalin- The tail wounds were treated with a fibrin spray containing GFP
fixed biopsy specimens from a representative chronic wound were syngeneic bone marrowderived cultured MSC. Frozen sections
immunostained for CD29, CD45, and prolyl hydroxylase as a were analyzed using the Zeiss Axioplan 2 imaging system with the
specific marker for human fibroblasts (fibroblast marker: FibM). Axio CAM. GFP blood vessels (arrows) were identified by FITC
All magnifications are 10. The left and right panels refer to the (510560 nm; (A)). Adjacent sections were analyzed by filters for
wound treated with fibrin alone or MSC-containing fibrin spray, light microscopy (C), Texas Red (645/75 nm; (D)), and DAPI (435
respectively. The blue staining is due to the marker blue ink ap- 475 nm; (E)). Using these filters, a composite photomicrograph was
plied to the top of the biopsy specimens. The panels show spindle- obtained, showing the specificity of the green fluorescence (B). Mag-
shaped cells positive for both CD29 and the fibroblast marker in nification 200.
the superficial layers of the wound bed, where the cultured cells
were applied. The immunostained cells appear to be distinct from
those positive for CD45, the leukocyte common antigen (LCA).

FIG. 11. Histology and effect on healing in mouse wounds treated with MSC. (A) Histology of a representative mouse wound 5 days
after application of syngeneic MSC in a fibrin spray. The wound bed contains a large number of cells; 4. (B) Red fluorescently labeled
MSC were applied in a fibrin spray to the mouse wounds. By day 5, the cells had migrated into the dermal component of the wound bed.
The photomicrograph represents a histological section adjacent to that shown in (A); 10. (C) The graphs represent the results of healing
when treating mouse tail wounds with syngeneic MSC. Both db/db mice and their control littermates were treated either with fibrin
alone or with MSC-containing fibrin spray. Each point represents the mean SEM from four mice.
1309
1310 FALANGA ET AL.

bone marrowderived stem cells become an attractive alter- not hematopoietic cells or leukocyte precursors. Some in-
native. Indeed, in a previous report, we showed promising vestigators have proposed that early or primitive stromal
results with the use of autologous bone marrowderived elements should be CD34 and thus exhibit more stem-
cultured cells in three subjects with previously nonhealing ness.11 However, our experiments do show that the cultured
leg ulcers.6 In that study, however, characterization of the cells could differentiate into bone, adipose, and cartilage
cultured cells was not performed. More importantly, at that components. Moreover, our goal was to develop an easily
time we did not have a proper method for cell delivery, and reproducible culture system that would insure that we were
the cultured cells were simply placed as a suspension under dealing with MSC and that MSC could be properly delivered
an occlusive film applied over the wound. That system topically and accelerate healing.
proved unsatisfactory, in that the cell suspension could easily An important feature of this report is that we studied hu-
run off the wound and definite and reliable delivery of the man wounds and determined the feasibility of fibrin as a de-
cultured cells could not be insured. In this report, we fully livery system in both acute and chronic wounds. Ultimately,
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

characterize the autologous bone marrow derived cultured the feasibility of the fibrin spray had to be tested in human
stem cells and show their mesenchymal phenotype. More- wounds, and both in the acute and more hostile chronic
over, we found that a delivery system using a fibrin spray, wound microenvironments. The results presented here appear
which we have modified for this purpose, seems ideal in this very promising and, equally important, no adverse effects
situation. The choice of fibrin as a delivery method was were noted. We found that, for a possible therapeutic effect of
based on a number of previous observations and the fact that this approach in human acute wounds, larger wounds would
fibrin is a well-tested system already in use to stop bleeding need to be tested in future studies. This is actually not sur-
during surgery.2325 We have previously shown that cross- prising, for acute wounds generally heal without much dif-
linked fibrin stimulates cell attachment and spreading.26 ficulty. For chronic wounds, we chose subjects with very
This and work from others suggested that fibrin may be safe difficult-to-heal wounds, which were present for a long time
for cells, including MSC,27 and for the healing process.2830 and were unresponsive to even advanced therapies. Thus, the
The next important question was whether and how to modify results in these subjects are extremely promising. We found a
the fibrinogen and/or thrombin concentrations for the de- strong and statistically significant correlation between the
livery of cells to a wound. A recent report has shown ideal number of applied MSC and acceleration of healing. The
concentrations of these components for developing fibrin optimal number of applied cells needs to be at least 1106
constructs that, at least in vitro, allow fibroblasts to migrate cells per cm2 of the wound surface. This is very important
onto tissue culture plastic.27 In agreement with that report, information that could only be discovered empirically and by
we also found that the readily available commercial fibrin actually studying these difficult wounds. Identifying the num-
preparation, which is used to control bleeding, could not be ber of cells is also critical for larger studies in the future. The
used in the same way to deliver cells. In fact, our in vivo results obtained with the acceleration of healing of full-
studies found that full concentrations of fibrinogen and thickness wounds in db/db mice provide additional evidence
thrombin encased the stem cells to the point that they ap- for the effectiveness of MSC in stimulating wound repair. As
peared to be nonviable (not shown). The concentrations of expected, control littermates healed faster than db/db mice,
fibrinogen and thrombin used in this report, however, appear although they too showed a statistically significant healing
to be effective in cell delivery, as indicated by our histo- response to autologous mouse MSC.
logical and immunofluorescence analysis and by the accel- An understandable and obvious disadvantage of using
eration of wound healing. human wounds is that one cannot properly and safely mark
In culturing the Ficoll-separated bone marrow aspirate, cells and track them from the fibrin gel into the wound. We
we chose to test for a subset of high-yield cluster designa- therefore immunostained sequential adjacent sections for
tion markers to provide information on the lineage of the different markers. The results in both acute and chronic
cultured cells. While the cells were plastic adherent and had wounds showed that the introduced cells, positive for CD29
similar morphology to stromal cells, we were interested in and for prolyl hydroxylase as a specific human fibroblast
confirming that the cells were indeed mesenchymal in origin. marker,14 can be found in the dermis, immediately under the
There is no specific MSC marker, and we therefore selected site of fibrin gel delivery. The highly spindled morphologi-
a group of markers. We chose CD29, CD44, CD90, CD105, cal appearance we found in histological and immunostained
and CD166 as they are commonly cited as MSC markers sections, remarkably similar to what we documented in tis-
in the literature.9,10 For the sake of practicality and for de- sue culture, is also supportive of the fact that the stem cells
veloping a method that could be repeated on cultures from did indeed migrate from the applied fibrin gel and mobilize
multiple patients, we chose this previously reported subset. into the dermis. Moreover, using two separate methods to
Other markers described in reports of human MSC include detect elastic fibers, we found strong evidence that new
STRO-1, CD73, and CD49e.11 We also tested for CD34 and elastic fibers may have been deposited in the dermis of full-
CD45 to insure that we were culturing the stromal compo- thickness wounds treated with MSC. This possible regen-
nent of the bone marrow, and not hematopoietic cells. Our eration of elastic fibers, which normally does not occur in
cells were CD34 and CD45 negative, indicating that they are healing wounds or in scars,1517 is quite interesting. Similar
DELIVERY OF STEM CELLS TO HUMAN AND MURINE WOUNDS 1311

accumulation of spindle cells having these markers in the 5. Kim, B.C., Kim, H.T., Park, S.H., Cha, J.S., Yufit, T., Kim,
upper dermis or new elastic fiber formation were not found S.J., and Falanga, V. Fibroblasts from chronic wounds show
in control sections where the wound received fibrin alone. altered TGF-beta-signaling and decreased TGF-beta type II
In order to further track the MSC applied to wounds, we receptor expression. J Cell Physiol 195(3): 33136, 2003.
performed experiments in mice by using GFP autologous 6. Badiavas, E.V., and Falanga, V. Treatment of chronic wounds
with bone marrow-derived cells. Arch Dermatol 139(4): 510
MSC isolated from syngeneic strains. For imaging the im-
16, 2003.
munofluorescence, we used a stringent system of optical fil-
7. Butmarc, J., Yufit, T., Carson, P., and Falanga, V. Human
ters to exclude the possibility of autofluorescence and other beta-defensin-2 expression is increased in chronic wounds.
situations of false positive green fluorescence. Our results Wound Repair Regen 12(4): 43943, 2004.
indicate that, at least at later time points, labeled MSC could 8. Sudres, M., Norol, F., Trenado, A., Gregoire, S., Charlotte, F.,
not be found in clusters, except for very occasional indi- Levacher, B., Lataillade, J.J., Bourin, P., Holy, X., Vernant,
vidual cells. We cannot fully explain these findings, but we J.P., Klatzmann, D., and Cohen, J.L. Bone marrow mesen-
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

propose some possibilities. One is that cultured and topically chymal stem cells suppress lymphocyte proliferation in vitro
applied MSC may participate in the stimulation of wound but fail to prevent graft-versus-host disease in mice. J Immunol
healing by the production of cytokines and/or stimulation of 176(12): 776167, 2006.
endogenous resident cells. Another explanation is that, be- 9. Kassis, I., Zangi, L., Rivkin, R., Levdansky, L., Samuel, S.,
Marx, G., and Gorodetsky, R. Isolation of mesenchymal stem
cause GFP is quite immunogenic,31 cells engineered with this
cells from G-CSF-mobilized human peripheral blood using
protein are removed by the host immune system. Studies are
fibrin microbeads. Bone Marrow Transplant 37(10): 96776,
going on to answer some of these questions. We did observe 2006.
rare but definitely GFP dermal blood vessels in the healed 10. Tuli, R., Tuli, S., Nandi, S., Wang, M.L., Alexander, P.G.,
wound. At this point, we do not know whether this could Haleem-Smith, H., Hozack, W.J., Manner, P.A., Danielson,
represent cell fusion or a rare event of MSC conversion to K.G., and Tuan, R.S. Characterization of multipotential mes-
endothelium. enchymal progenitor cells derived from human trabecular bone.
In summary, we describe methods for reliably culturing Stem Cells 21(6): 68193, 2003.
human and mouse MSC from bone marrow and for the de- 11. Simmons, P.J., and Torok-Storb, B. CD34 expression by stro-
livery of these cells to human wounds and experimental mu- mal precursors in normal human adult bone marrow. Blood
rine wounds. The approach appears reliable and safe and is 78(11): 284853, 1991.
12. Short, B., Brouard, N., Occhiodoro-Scott, T., Ramakrishnan,
very promising in terms of effectively stimulating the repair
A., and Simmons, P.J. Mesenchymal stem cells. Arch Med Res
process in injured tissue.
34(6): 56571, 2003.
13. Bowen, G.M., White, G.L., Jr., and Gerwels, J.W. Mohs mi-
crographic surgery. Am Fam Physician 72(5): 84548, 2005.
ACKNOWLEDGMENTS 14. Olerud, J.E., Chiu, D.S., Usui, M.L., Gibran, N.S., and Ansel,
J.C. Protein gene product 9.5 is expressed by fibroblasts in
We thank Dr. Gerald Colvin of the Division of Hema- human cutaneous wounds. J Invest Dermatol 111(4): 56572,
tology for performing the bone marrow aspiration on human 1998.
subjects. Dr. Peter Libbey, of the Department of Pathology, 15. Kumagai, N., Nishina, H., Tanabe, H., Hosaka, T., Ishida, H.,
helped us in the studies of MSC conversion to cartilage and and Ogino, Y. Clinical application of autologous cultured
adipose tissue. epithelia for the treatment of burn wounds and burn scars.
Plast Reconstr Surg 82(1): 99110, 1988.
16. Moiemen, N.S., Vlachou, E., Staiano, J.J., Thawy, Y., and
REFERENCES Frame, J.D. Reconstructive surgery with Integra dermal regen-
eration template: histologic study, clinical evaluation, and cur-
1. Falanga, V. Wound healing and its impairment in the diabetic rent practice. Plast Reconstr Surg 117(7 Suppl): 160S174S,
foot. Lancet 366(9498): 173643, 2005. 2006.
2. Agren, M.S., Steenfos, H.H., Dabelsteen, S., Hansen, J.B., and 17. Stuzin, J.M., Baker, T.J., Baker, T.M., and Kligman, A.M.
Dabelsteen, E. Proliferation and mitogenic response to PDGF- Histologic effects of the high-energy pulsed CO2 laser on
BB of fibroblasts isolated from chronic venous leg ulcers is photoaged facial skin. Plast Reconstr Surg 99(7): 203650,
ulcer-age dependent. J Invest Dermatol 112(4): 46369, 1999. 1997.
3. Falanga, V., Schrayer, D., Cha, J., Butmarc, J., Carson, P., 18. Cho, C.H., Sung, H.K., Kim, K.T., Cheon, H.G., Oh, G.T.,
Roberts, A.B., and Kim, S.J. Full-thickness wounding of the Hong, H.J., Yoo, O.J., and Koh, G.Y. COMP-angiopoietin-1
mouse tail as a model for delayed wound healing: accelerated promotes wound healing through enhanced angiogenesis,
wound closure in Smad3 knock-out mice. Wound Repair Regen lymphangiogenesis, and blood flow in a diabetic mouse model.
12(3): 32026, 2004. Proc Natl Acad Sci USA 103(13): 494651, 2006.
4. Hasan, A., Murata, H., Falabella, A., Ochoa, S., Zhou, L., 19. Fu, X., Fang, L., Li, X., Cheng, B., and Sheng, Z. Enhanced
Badiavas, E., and Falanga, V. Dermal fibroblasts from venous wound-healing quality with bone marrow mesenchymal stem
ulcers are unresponsive to the action of transforming growth cells autografting after skin injury. Wound Repair Regen
factor-beta 1. J Dermatol Sci 16(1): 5966, 1997. 14(3): 32535, 2006.
1312 FALANGA ET AL.

20. Roh, C., and Lyle, S. Cutaneous stem cells and wound heal- binding of growth factors. Scand J Gastroenterol 39(10): 927
ing. Pediatr Res 59(4 Pt 2): 100R103R, 2006. 32, 2004.
21. Falanga, V., and Sabolinski, M. A bilayered living skin con- 29. Catelas, I., Sese, N., Wu, B.M., Dunn, J.C., Helgerson, S., and
struct (APLIGRAF) accelerates complete closure of hard-to- Tawil, B. Human mesenchymal stem cell proliferation and os-
heal venous ulcers. Wound Repair Regen 7(4): 2017, 1999. teogenic differentiation in fibrin gels in vitro. Tissue Eng 12:
22. Phillips, T.J., Machado, F., Trout, R., Porter, J., Olin, J., and 23852396, 2006.
Falanga, V. Prognostic indicators in venous ulcers. J Am 30. Cox, S., Cole, M., and Tawil, B. Behavior of human dermal
Acad Dermatol 43(4): 62730, 2000. fibroblasts in three-dimensional fibrin clots: dependence on
23. Dunn, C.J., and Goa, K.L. Fibrin sealant: a review of its use in fibrinogen and thrombin concentration. Tissue Eng 10(5-6):
surgery and endoscopy. Drugs 58(5): 86386, 1999. 94254, 2004.
24. Katkhouda, N. New hemostatic agents in general open and 31. Re, F., Srinivasan, R., Igarashi, T., Marincola, F., and Childs,
laparoscopic surgery. Surg Technol Int 13: 6570, 2004. R. Green fluorescent protein expression in dendritic cells en-
25. Schips, L., Dalpiaz, O., Cestari, A., Lipsky, K., Gidaro, S., hances their immunogenicity and elicits specific cytotoxic T-
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

Zigeuner, R., and Petritsch, P. Autologous fibrin glue using the cell responses in humans. Exp Hematol 32(2): 21017, 2004.
Vivostat system for hemostasis in laparoscopic partial ne-
phrectomy. Eur Urol 50: 801805, 2006. Address reprint requests to:
26. Weiss, E., Yamaguchi, Y., Falabella, A., Crane, S., Tokuda, Y., Vincent Falanga, M.D.
and Falanga, V. Un-cross-linked fibrin substrates inhibit kera-
Professor and Chairman
tinocyte spreading and replication: correction with fibronectin
and factor XIII cross-linking. J Cell Physiol 174(1): 5865, 1998.
Department of Dermatology and Skin Surgery
27. Ho, W., Tawil, B., Dunn, J.C., and Wu, B.M. The behavior of Roger Williams Medical Center
human mesenchymal stem cells in 3D fibrin clots: dependence Elmhurst Building
on fibrinogen concentration and clot structure. Tissue Eng 50 Maude Street
12(6): 158795, 2006. Providence, RI 02908
28. Becker, J.C., Domschke, W., and Pohle, T. Biological in vitro
effects of fibrin glue: fibroblast proliferation, expression and E-mail: vfalanga@bu.edu
This article has been cited by:

1. Chi-Hong Ho, Chen-Wei Lan, Chen-Yi Liao, Shih-Chieh Hung, Hsin-Yang Li, Yen-Jen Sung. 2017. Mesenchymal stem cells
and their conditioned medium can enhance the repair of uterine defects in a rat model. Journal of the Chinese Medical Association
. [Crossref]
2. Schwartz Dana M., Pehlivaner Kara Meryem O., Goldstein Allan M., Ott Harald C., Ekenseair Adam K.. 2017. Spray Delivery
of Intestinal Organoids to Reconstitute Epithelium on Decellularized Native Extracellular Matrix. Tissue Engineering Part C:
Methods 23:9, 565-573. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
3. Dijkstra Koen, Hendriks Jan, Karperien Marcel, Vonk Lucienne A., Saris Danil B.F.. 2017. Arthroscopic Airbrush-Assisted
Cell Spraying for Cartilage Repair: Design, Development, and Characterization of Custom-Made Arthroscopic Spray Nozzles.
Tissue Engineering Part C: Methods 23:9, 505-515. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
[Supplemental Material]
4. Elizabeth Thompson. 2017. Debridement Techniques and NonNegative Pressure Wound Therapy Wound Management.
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

Veterinary Clinics of North America: Small Animal Practice . [Crossref]


5. Anjali Somal, Irfan A. Bhat, Indu B., Anuj P. Singh, Bibhudatta S.K. Panda, Perumal A. Desingu, Sriti Pandey, Mukesh K.
Bharti, Amar Pal, Guttula Saikumar, Vikash Chandra, Guttula Taru Sharma. 2017. Impact of Cryopreservation on Caprine Fetal
Adnexa Derived Stem Cells and Its Evaluation for Growth Kinetics, Phenotypic Characterization, and Wound Healing Potential
in Xenogenic Rat Model. Journal of Cellular Physiology 232:8, 2186-2200. [Crossref]
6. Mrs. Dana M. Schwartz, Mrs. Meryem O. Pehlivaner Kara, Dr. Allan Goldstein, Dr. Harold C. Ott, Dr. Adam Ekenseair. Spray
Delivery of Intestinal Organoids to Reconstitute Epithelium on Decellularized Native Extracellular Matrix. Tissue Engineering
Part C: Methods 0:ja. . [Abstract] [Full Text PDF] [Full Text PDF with Links]
7. van de Vyver Mari. 2017. Intrinsic Mesenchymal Stem Cell Dysfunction in Diabetes Mellitus: Implications for Autologous Cell
Therapy. Stem Cells and Development 26:14, 1042-1053. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with
Links]
8. Bower Andrew J., Mahmassani Ziad, Zhao Youbo, Chaney Eric J., Marjanovic Marina, Lee Min Kyung, Graf Benedikt W., De
Lisio Michael, Kong Hyunjoon, Boppart Marni D., Boppart Stephen A.. 2017. In Vivo Assessment of Engineered Skin Cell
Delivery with Multimodal Optical Microscopy. Tissue Engineering Part C: Methods 23:7, 434-442. [Abstract] [Full Text HTML]
[Full Text PDF] [Full Text PDF with Links]
9. Amy L. Strong, Michael W. Neumeister, Benjamin Levi. 2017. Stem Cells and Tissue Engineering. Clinics in Plastic Surgery
44:3, 635-650. [Crossref]
10. E. I. Domaratskaya, O. V. Payushina. 2017. Mesenchymal stromal cells as a resource for regeneration of damaged skin. Biology
Bulletin Reviews 7:4, 333-343. [Crossref]
11. Tomas de Mayo, Paulette Conget, Silvia Becerra-Bayona, Claudia L. Sossa, Virgilio Galvis, Martha L. Arango-Rodrguez. 2017.
The role of bone marrow mesenchymal stromal cell derivatives in skin wound healing in diabetic mice. PLOS ONE 12:6, e0177533.
[Crossref]
12. Ho Jasmine, Walsh Claire, Yue Dominic, Dardik Alan, Cheema Umber. 2017. Current Advancements and Strategies in Tissue
Engineering for Wound Healing: A Comprehensive Review. Advances in Wound Care 6:6, 191-209. [Abstract] [Full Text HTML]
[Full Text PDF] [Full Text PDF with Links]
13. Yixiao Dong, Sigen A, Melanie Rodrigues, Xiaolin Li, Sun H. Kwon, Nina Kosaric, Sacha Khong, Yongsheng Gao, Wenxin Wang,
Geoffrey C. Gurtner. 2017. Injectable and Tunable Gelatin Hydrogels Enhance Stem Cell Retention and Improve Cutaneous
Wound Healing. Advanced Functional Materials 27:24, 1606619. [Crossref]
14. Calver Pang, Amel Ibrahim, Neil W Bulstrode, Patrizia Ferretti. 2017. An overview of the therapeutic potential of regenerative
medicine in cutaneous wound healing. International Wound Journal 14:3, 450-459. [Crossref]
15. Holly M. Wobma, David Liu, Gordana Vunjak-Novakovic. 2017. Paracrine Effects of Mesenchymal Stromal Cells Cultured in
Three-Dimensional Settings on Tissue Repair. ACS Biomaterials Science & Engineering . [Crossref]
16. Sei-ichiro Motegi, Osamu Ishikawa. 2017. Mesenchymal stem cells: The roles and functions in cutaneous wound healing and
tumor growth. Journal of Dermatological Science 86:2, 83-89. [Crossref]
17. Volker Enzmann, Stphanie Lecaud, Anna Kruschinski, Axel Vater. 2017. CXCL12/SDF-1-Dependent Retinal Migration of
Endogenous Bone Marrow-Derived Stem Cells Improves Visual Function after Pharmacologically Induced Retinal Degeneration.
Stem Cell Reviews and Reports 13:2, 278-286. [Crossref]
18. Farshad Zarei, Babak Negahdari, Ali Eatemadi. 2017. Diabetic ulcer regeneration: stem cells, biomaterials, growth factors. Artificial
Cells, Nanomedicine, and Biotechnology 28, 1-7. [Crossref]
19. K.H. Kim, G. Blasco-Morente, N. Cuende, S. Arias-Santiago. 2017. Mesenchymal stromal cells: properties and role in
management of cutaneous diseases. Journal of the European Academy of Dermatology and Venereology 31:3, 414-423. [Crossref]
20. Alessandra Moore, Clement Marshall, Michael Longaker. 2017. Minimizing Skin Scarring through Biomaterial Design. Journal
of Functional Biomaterials 8:1, 3. [Crossref]
21. Lisa N. Kasiewicz, Kathryn A. Whitehead. 2017. Recent advances in biomaterials for the treatment of diabetic foot ulcers. Biomater.
Sci. 23. . [Crossref]
22. Yusef Yousuf, Saeid Amini-Nik, Marc G. Jeschke. Use of Stem Cells in Acute and Complex Wounds 195-226. [Crossref]
23. Kangquan Shou, Yahui Niu, Xun Zheng, Zhanjun Ma, Chao Jian, Baiwen Qi, Xiang Hu, Aixi Yu. 2017. Enhancement of Bone-
Marrow-Derived Mesenchymal Stem Cell Angiogenic Capacity by NPWT for a Combinatorial Therapy to Promote Wound
Healing with Large Defect. BioMed Research International 2017, 1-13. [Crossref]
24. H.-G. Machens, Christina Irene Gnter. Innovative Therapien der Wundheilung 177-197. [Crossref]
25. Samuel J. Barke, Richard N. Villar. The Use of Fibrin Adhesive for Cartilage Repair in Hip Arthroscopy 681-686. [Crossref]
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

26. Q. Zeng, L.K. Macri, A. Prasad, R.A.F. Clark, D.I. Zeugolis, C. Hanley, Y. Garcia, A. Pandit, D.I. Leavesley, D. Stupar, M.L.
Fernandez, C. Fan, Z. Upton. 6.20 Skin Tissue Engineering 334-382. [Crossref]
27. Toshio Hasegawa, Shigaku Ikeda. 2017. Mesenchymal Stem Cells for the Treatment of Skin Diseases. AIMS Cell and Tissue
Engineering 1:2, 104-117. [Crossref]
28. M. Vitacolonna, D. Belharazem, P. Hohenberger, E. D. Roessner. 2016. In-vivo quantification of the revascularization of a human
acellular dermis seeded with EPCs and MSCs in co-culture with fibroblasts and pericytes in the dorsal chamber model in pre-
irradiated tissue. Cell and Tissue Banking 105. . [Crossref]
29. Kim Sally Yunsun, Burgess Janette K., Wang Yiwei, Kable Eleanor P.W., Weiss Daniel J., Chan Hak-Kim, Chrzanowski Wojciech.
2016. Atomized Human Amniotic Mesenchymal Stromal Cells for Direct Delivery to the Airway for Treatment of Lung Injury.
Journal of Aerosol Medicine and Pulmonary Drug Delivery 29:6, 514-524. [Abstract] [Full Text HTML] [Full Text PDF] [Full
Text PDF with Links] [Supplemental Material]
30. Pei Li, Yuming Zhao, Lihong Ge. 2016. Therapeutic effects of human gingiva-derived mesenchymal stromal cells on murine
contact hypersensitivity via prostaglandin E2EP3 signaling. Stem Cell Research & Therapy 7:1. . [Crossref]
31. Bruno Amato, Rita Compagna, Maurizio Amato, Lucia Butrico, Francesco Fugetto, Mariia D Chibireva, Andrea Barbetta, Marco
Cannistr, Stefano de Franciscis, Raffaele Serra. 2016. The role of adult tissue-derived stem cells in chronic leg ulcers: a systematic
review focused on tissue regeneration medicine. International Wound Journal 13:6, 1289-1298. [Crossref]
32. Martin Boh, Mria Csbnyeiov, Ida Kupcov, Radoslav Zamborsk, Jozef Fedele, Jn Koller. 2016. Stem cell regenerative
potential for plastic and reconstructive surgery. Cell and Tissue Banking 17:4, 735-744. [Crossref]
33. Annelies Bronckaers, Ivo Lambrichts. The role of mesenchymal stem/stromal cells in angiogenesis 347-365. [Crossref]
34. Stefan Zwingenberger, Ishaq Ojodu, Maik Stiehler, Stuart B. Goodman. The role of mesenchymal stromal cells in the
management of musculoskeletal disorders 677-689. [Crossref]
35. Unnikrishnan Sivan, K. Jayakumar, Lissy K. Krishnan. 2016. Matrix-directed differentiation of human adipose-derived
mesenchymal stem cells to dermal-like fibroblasts that produce extracellular matrix. Journal of Tissue Engineering and Regenerative
Medicine 10:10, E546-E558. [Crossref]
36. Valerio Ian L., Sabino Jennifer M., Dearth Christopher L.. 2016. Plastic Surgery Challenges in War Wounded II: Regenerative
Medicine. Advances in Wound Care 5:9, 412-419. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
37. Mayer Tenenhaus, Hans-Oliver Rennekampff. 2016. Current Concepts in Tissue Engineering. Plastic and Reconstructive Surgery
138, 42S-50S. [Crossref]
38. Sarah Sorice, Kristine C. Rustad, Alexander Y Li, Geoffrey C. Gurtner. 2016. The Role of Stem Cell Therapeutics in Wound
Healing. Plastic and Reconstructive Surgery 138, 31S-41S. [Crossref]
39. Foubert Philippe, Gonzalez Andreina D., Teodosescu Stephan, Berard Felipe, Doyle-Eisele Melanie, Yekkala Krishna, Tenenhaus
Mayer, Fraser John K.. 2016. Adipose-Derived Regenerative Cell Therapy for Burn Wound Healing: A Comparison of Two
Delivery Methods. Advances in Wound Care 5:7, 288-298. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with
Links]
40. Sweta Rani, Thomas Ritter. 2016. The Exosome - A Naturally Secreted Nanoparticle and its Application to Wound Healing.
Advanced Materials 28:27, 5542-5552. [Crossref]
41. Napoleo M. Argolo Neto, Ricardo J. Del Carlo, Betnia S. Monteiro, Nance B. Nardi, Pedro C. Chagastelles, Ana Flora S. Brito,
Amanda M.S. Reis, Las M.C. Silva. 2016. Role of the autologous mesenchymal stem cells compared with platelet rich plasma
on cicatrization of cutaneous wounds in diabetic mice. Pesquisa Veterinria Brasileira 36:7, 617-624. [Crossref]
42. Wojtek Tutak, Gili Kaufman, Grant Gelven, Chris Markle, Caitlyn Maczka. 2016. Uniform, fast, high concentration delivery
of bone marrow stromal cells and gingival fibroblasts by gas-brushing. Biomedical Physics & Engineering Express 2:3, 035007.
[Crossref]
43. Amanda M. Meppelink, Xing-Hua Wang, Gino Bradica, Kathryn Barron, Kathleen Hiltz, Xiang-Hong Liu, Scott M. Goldman,
Joseph P. Vacanti, Armand Keating, David M. Hoganson. 2016. Rapid isolation of bone marrow mesenchymal stromal cells using
integrated centrifuge-based technology. Cytotherapy 18:6, 729-739. [Crossref]
44. M. van de Vyver, C. Niesler, K.H. Myburgh, W.F. Ferris. 2016. Delayed wound healing and dysregulation of IL6/STAT3 signalling
in MSCs derived from pre-diabetic obese mice. Molecular and Cellular Endocrinology 426, 1-10. [Crossref]
45. Cerqueira Mariana Teixeira, Pirraco Rogrio Pedro, Marques Alexandra Pinto. 2016. Stem Cells in Skin Wound Healing: Are We
There Yet?. Advances in Wound Care 5:4, 164-175. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
46. Otero-Vias Marta, Falanga Vincent. 2016. Mesenchymal Stem Cells in Chronic Wounds: The Spectrum from Basic to Advanced
Therapy. Advances in Wound Care 5:4, 149-163. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

47. Thiebes Anja Lena, Reddemann Manuel Armin, Palmer Johannes, Kneer Reinhold, Jockenhoevel Stefan, Cornelissen Christian
Gabriel. 2016. Flexible Endoscopic Spray Application of Respiratory Epithelial Cells as Platform Technology to Apply Cells in
Tubular Organs. Tissue Engineering Part C: Methods 22:4, 322-331. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text
PDF with Links] [Supplemental Material]
48. Juli R. Bag, Guillaume J. Pegna, Onyi Okolie, Shawn D. Hingtgen. 2016. Fibrin matrices enhance the transplant and efficacy
of cytotoxic stem cell therapy for post-surgical cancer. Biomaterials 84, 42-53. [Crossref]
49. Michael K. Lichtman, Marta Otero-Vinas, Vincent Falanga. 2016. Transforming growth factor beta (TGF-) isoforms in wound
healing and fibrosis. Wound Repair and Regeneration 24:2, 215-222. [Crossref]
50. Carlos Zgheib, Maggie Hodges, Junyi Hu, David P. Beason, Louis J. Soslowsky, Kenneth W. Liechty, Junwang Xu. 2016.
Mechanisms of mesenchymal stem cell correction of the impaired biomechanical properties of diabetic skin: The role of miR-29a.
Wound Repair and Regeneration 24:2, 237-246. [Crossref]
51. Rui Tostoes, Paula M. Alves. Culturing Entrapped Stem Cells in Continuous Bioreactors 201-226. [Crossref]
52. Summer E. Hanson, Kyle R. Kleinbeck, David Cantu, Jaeyhup Kim, Michael L. Bentz, Lee D. Faucher, W. John Kao, Peiman
Hematti. 2016. Local delivery of allogeneic bone marrow and adipose tissue-derived mesenchymal stromal cells for cutaneous
wound healing in a porcine model. Journal of Tissue Engineering and Regenerative Medicine 10:2, E90-E100. [Crossref]
53. Carolina Caliari-Oliveira, Juliana Navarro Ueda Yaochite, Leandra N?ira Zambelli Ramalho, Patr?cia Vianna Bonini Palma, Daniela
Carlos, Fernando De Queir?z Cunha, Daurea Abadia De Souza, Marco Andrey Cipriani Frade, Dimas Tadeu Covas, Kelen Cristina
Ribeiro Malmegrim, Maria Carolina Oliveira, Julio C?sar Voltarelli. 2016. Xenogeneic Mesenchymal Stromal Cells Improve Wound
Healing and Modulate the Immune Response in an Extensive Burn Model. Cell Transplantation 25:2, 201-215. [Crossref]
54. Daniele Tartarini, Elisa Mele. 2016. Adult Stem Cell Therapies for Wound Healing: Biomaterials and Computational Models.
Frontiers in Bioengineering and Biotechnology 3. . [Crossref]
55. Phuc Van Pham. Mesenchymal Stem Cells in Clinical Applications 37-69. [Crossref]
56. Qinan Wu, Bing Chen, Ziwen Liang. 2016. Mesenchymal Stem Cells as a Prospective Therapy for the Diabetic Foot. Stem Cells
International 2016, 1-18. [Crossref]
57. Claudia Cappuzzello, Andrea Doni, Erica Dander, Fabio Pasqualini, Manuela Nebuloni, Barbara Bottazzi, Alberto Mantovani,
Andrea Biondi, Cecilia Garlanda, Giovanna DAmico. 2016. Mesenchymal Stromal Cell-Derived PTX3 Promotes Wound Healing
via Fibrin Remodeling. Journal of Investigative Dermatology 136:1, 293-300. [Crossref]
58. Bommie Florence Seo, Sung-No Jung. 2016. The Immunomodulatory Effects of Mesenchymal Stem Cells in Prevention or
Treatment of Excessive Scars. Stem Cells International 2016, 1-8. [Crossref]
59. Rano Matta, John E. Davies. Bioengineering and Regenerative Medicine in Surgery 189-203. [Crossref]
60. Deyun Chen, Haojie Hao, Xiaobing Fu, Weidong Han. 2016. Insight into Reepithelialization: How Do Mesenchymal Stem Cells
Perform?. Stem Cells International 2016, 1-9. [Crossref]
61. M. Tenenhaus, H.O. Rennekampff, G. Mulder. Living cell products as wound healing biomaterials 201-225. [Crossref]
62. N. Urao, T.J. Koh. Manipulating inflammation to improve healing 117-150. [Crossref]
63. Samir Malhotra, Michael S. Hu, Clement D. Marshall, Tripp Leavitt, Alexander T. M. Cheung, Jennifer G. Gonzalez, Harleen
Kaur, H. Peter Lorenz, Michael T. Longaker. 2016. Mesenchymal Stromal Cells as Cell-Based Therapeutics for Wound Healing.
Stem Cells International 2016, 1-6. [Crossref]
64. Anthony R. Sheets, Catalina K. Hwang, Ira M. Herman. Developing Smart Point-of-Care Diagnostic Tools for Next-
Generation Wound Care 251-264. [Crossref]
65. L. Estronca, L. Ferreira. Stem Cells for the Regeneration of Chronic Wounds 291-312. [Crossref]
66. R. Snchez-Snchez, E. Martnez-Arredondo, V. Martnez-Lpez, Y. Melgarejo-Ramrez, A. Brena-Molina, H. Lugo-
Martnez, R. Gmez-Garca, D. Garciadiego-Czares, P. Silva-Bermdez, E. Mrquez-Gutirrez, C. Ibarra, C. Velasquillo. 2016.
Development of Hydrogel with Anti-Inflammatory Properties Permissive for the Growth of Human Adipose Mesenchymal Stem
Cells. Journal of Nanomaterials 2016, 1-8. [Crossref]
67. F. Akter, J. Ibanez, N. Bulstrode. Skin Engineering 17-27. [Crossref]
68. Sean V. Murphy, Mohammad Z. Albanna. Cutaneous Applications of Stem Cells for Skin Tissue Engineering 317-336. [Crossref]
69. A. Abdullahi, S. Amini-Nik, M.G. Jeschke. Stem cell therapies for wounds 177-200. [Crossref]
70. G.A. Limb, M.F. Jones, K. Eastlake, S. Becker. Biomaterials for repair and regeneration of the neural retina 243-261. [Crossref]
71. Flavia Bruna, David Contador, Paulette Conget, Benjamn Erranz, Claudia L. Sossa, Martha L. Arango-Rodrguez. 2016.
Regenerative Potential of Mesenchymal Stromal Cells: Age-Related Changes. Stem Cells International 2016, 1-15. [Crossref]
72. Thiebes Anja Lena, Albers Stefanie, Klopsch Christian, Jockenhoevel Stefan, Cornelissen Christian G.. 2015. Spraying Respiratory
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

Epithelial Cells to Coat Tissue-Engineered Constructs. BioResearch Open Access 4:1, 278-287. [Abstract] [Full Text HTML]
[Full Text PDF] [Full Text PDF with Links]
73. Jun How Low, Premdass Ramdas, Ammu Kutty Radhakrishnan. 2015. Modulatory effects of mesenchymal stem cells on
leucocytes and leukemic cells: A double-edged sword?. Blood Cells, Molecules, and Diseases 55:4, 351-357. [Crossref]
74. Hatixhe Latifi-Pupovci, Zyrafete Kui, Sibylle Wehner, Halvard Bnig, Ralf Lieberz, Thomas Klingebiel, Peter Bader, Selim
Kui. 2015. In vitro migration and proliferation (wound healing) potential of mesenchymal stromal cells generated from human
CD271+ bone marrow mononuclear cells. Journal of Translational Medicine 13:1. . [Crossref]
75. Shuo Huang, Liangliang Xu, Yuxin Sun, Yifeng Zhang, Gang Li. 2015. The fate of systemically administrated allogeneic
mesenchymal stem cells in mouse femoral fracture healing. Stem Cell Research & Therapy 6:1. . [Crossref]
76. Hocking Anne M.. 2015. The Role of Chemokines in Mesenchymal Stem Cell Homing to Wounds. Advances in Wound Care
4:11, 623-630. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
77. Ejaj Ahmad, Munazza Tamkeen Fatima, Mehboob Hoque, Mohammad Owais, Mohammed Saleemuddin. 2015. Fibrin matrices:
The versatile therapeutic delivery systems. International Journal of Biological Macromolecules 81, 121-136. [Crossref]
78. Kuan-Chih Kuo, Ruei-Zeng Lin, Han-Wen Tien, Pei-Yun Wu, Yen-Cheng Li, Juan M. Melero-Martin, Ying-Chieh Chen.
2015. Bioengineering vascularized tissue constructs using an injectable cell-laden enzymatically crosslinked collagen hydrogel
derived from dermal extracellular matrix. Acta Biomaterialia 27, 151-166. [Crossref]
79. Fernando A. Fierro, Adam J. O'Neal, Julie R. Beegle, Myra N. Chvez, Thomas R. Peavy, Roslyn R. Isseroff, Jos T. Egaa.
2015. Hypoxic pre-conditioning increases the infiltration of endothelial cells into scaffolds for dermal regeneration pre-seeded
with mesenchymal stem cells. Frontiers in Cell and Developmental Biology 3. . [Crossref]
80. Nkemcho Ojeh, Irena Pastar, Marjana Tomic-Canic, Olivera Stojadinovic. 2015. Stem Cells in Skin Regeneration, Wound Healing,
and Their Clinical Applications. International Journal of Molecular Sciences 16:10, 25476-25501. [Crossref]
81. Yang Zeng, Lin Zhu, Qin Han, Wei Liu, Xiaojing Mao, Yaqian Li, Nanze Yu, Siyu Feng, Qinyouen Fu, Xiaojun Wang, Yanan
Du, Robert Chunhua Zhao. 2015. Preformed gelatin microcryogels as injectable cell carriers for enhanced skin wound healing.
Acta Biomaterialia 25, 291-303. [Crossref]
82. Sinan Ozturk, Huseyin Karagoz. 2015. Experimental stem cell therapies on burn wound: Do source, dose, timing and method
matter?. Burns 41:6, 1133-1139. [Crossref]
83. Xiaoling Li, Kexin Gan, Guangyao Song, Chao Wang. 2015. VEGF gene transfected umbilical cord mesenchymal stem cells
transplantation improve the lower limb vascular lesions of diabetic rats. Journal of Diabetes and its Complications 29:7, 872-881.
[Crossref]
84. Tom Hodgkinson, Ardeshir Bayat. 2015. Ex vivo evaluation of acellular and cellular collagen-glycosaminoglycan flowable matrices.
Biomedical Materials 10:4, 041001. [Crossref]
85. Jong Kyu Hong, Jisoo Yun, Hyunyun Kim, Sang-Mo Kwon. 2015. Three-dimensional culture of mesenchymal stem cells. Tissue
Engineering and Regenerative Medicine 12:4, 211-221. [Crossref]
86. Ji-Yong Jung, Joong Shim, Hyun Choi, Tae Lee, Dong Shin. 2015. Human Dermal Stem/Progenitor Cell-Derived Conditioned
Medium Improves Senescent Human Dermal Fibroblasts. International Journal of Molecular Sciences 16:8, 19027-19039. [Crossref]
87. Meirong Li, Yali Zhao, Haojie Hao, Weidong Han, Xiaobing Fu. 2015. Mesenchymal stem cell-based therapy for nonhealing
wounds: today and tomorrow. Wound Repair and Regeneration 23:4, 465-482. [Crossref]
88. Sona Zare, Maryam Anjomshoa, Soleiman Kurd, Jia-Ke Chai, Mostafa Dahmardei, Mohammad Ali Nilforoushzadeh, Ayoob
Rostamzadeh. 2015. Regenerative Medicine: Novel Approach in Burn Wound Healing. Journal of Skin and Stem Cell 2:2. .
[Crossref]
89. Hannes Heublein, Augustinus Bader, Shibashish Giri. 2015. Preclinical and clinical evidence for stem cell therapies as treatment
for diabetic wounds. Drug Discovery Today 20:6, 703-717. [Crossref]
90. Anthony J.P. Clover, Arun H.S. Kumar, Matthew Isakson, Derek Whelan, Alecia Stocca, Birgitta M. Gleeson, Noel M. Caplice.
2015. Allogeneic mesenchymal stem cells, but not culture modified monocytes, improve burn wound healing. Burns 41:3, 548-557.
[Crossref]
91. Stephen M. Cronk, Molly R. Kelly-Goss, H. Clifton Ray, Thomas A. Mendel, Kyle L. Hoehn, Anthony C. Bruce, Bijan K.
Dey, Alexander M. Guendel, Daniel N. Tavakol, Ira M. Herman, Shayn M. Peirce, Paul A. Yates. 2015. Adipose-Derived Stem
Cells From Diabetic Mice Show Impaired Vascular Stabilization in a Murine Model of Diabetic Retinopathy. STEM CELLS
Translational Medicine 4:5, 459-467. [Crossref]
92. Julie A. Rytlewski, M. Alejandra Aldon, Evan W. Lewis, Laura J. Suggs. 2015. Mechanisms of tubulogenesis and endothelial
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

phenotype expression by MSCs. Microvascular Research 99, 26-35. [Crossref]


93. Noh Seong-Seo, Bhang Suk Ho, La Wan-Geun, Lee Seahyoung, Shin Jung-Youn, Ma Yoon-Ji, Jang Hyeon-Ki, Kang Seokyung,
Jin Min, Park Jooyeon, Kim Byung-Soo. 2015. A Dual Delivery of Substance P and Bone Morphogenetic Protein-2 for
Mesenchymal Stem Cell Recruitment and Bone Regeneration. Tissue Engineering Part A 21:7-8, 1275-1287. [Abstract] [Full Text
HTML] [Full Text PDF] [Full Text PDF with Links]
94. Jennifer J. Bara, Sarah Turner, Sally Roberts, Gareth Griffiths, Rod Benson, Jayesh M. Trivedi, Karina T. Wright. 2015. High
content and high throughput screening to assess the angiogenic and neurogenic actions of mesenchymal stem cells in vitro.
Experimental Cell Research 333:1, 93-104. [Crossref]
95. Damian Garcia-Olmo, Hector Guadalajara, Ines Rubio-Perez, Maria Dolores Herreros, Paloma de-la-Quintana, Mariano Garcia-
Arranz. 2015. Recurrent anal fistulae: Limited surgery supported by stem cells. World Journal of Gastroenterology 21:11, 3330-3336.
[Crossref]
96. Ashley B. Allen, Lauren B. Priddy, Mon-Tzu A. Li, Robert E. Guldberg. 2015. Functional Augmentation of Naturally-Derived
Materials for Tissue Regeneration. Annals of Biomedical Engineering 43:3, 555-567. [Crossref]
97. J.W. Kuhbier, K. Reimers, C. Radtke, P.M. Vogt. 2015. Regenerative Therapieanstze in der plastischen Chirurgie. Der Chirurg
86:3, 214-222. [Crossref]
98. M?rcia Z Bellini, Carolina Caliari-Oliveira, Amanda Mizukami, Kamilla Swiech, Dimas T Covas, Eduardo A Donadi, Pedro
Oliva-Neto, ?ngela M Moraes. 2015. Combining xanthan and chitosan membranes to multipotent mesenchymal stromal cells as
bioactive dressings for dermo-epidermal wounds. Journal of Biomaterials Applications 29:8, 1155-1166. [Crossref]
99. Riopel Matthew, Trinder Mark, Wang Rennian. 2015. Fibrin, a Scaffold Material for Islet Transplantation and Pancreatic
Endocrine Tissue Engineering. Tissue Engineering Part B: Reviews 21:1, 34-44. [Abstract] [Full Text HTML] [Full Text PDF]
[Full Text PDF with Links]
100. Roux Stephan, Bodivit Gwellaouen, Bartis Widy, Lebouvier Anglique, Chevallier Nathalie, Fialaire-Legendre Anne, Bierling
Philippe, Rouard Helene. 2015. In Vitro Characterization of Patches of Human Mesenchymal Stromal Cells. Tissue Engineering
Part A 21:3-4, 417-425. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
101. Karen L Andrews, Matthew T Houdek, Lester J Kiemele. 2015. Wound management of chronic diabetic foot ulcers: From the
basics to regenerative medicine. Prosthetics and Orthotics International 39:1, 29-39. [Crossref]
102. Julio J. Mendez, Mahboobe Ghaedi, Amogh Sivarapatna, Sashka Dimitrievska, Zhen Shao, Chinedum O. Osuji, Derek M.
Steinbacher, David J. Leffell, Laura E. Niklason. 2015. Mesenchymal stromal cells form vascular tubes when placed in fibrin
sealant and accelerate wound healing invivo. Biomaterials 40, 61-71. [Crossref]
103. Renea A. Faulknor, Melissa A. Olekson, Nir I. Nativ, Mehdi Ghodbane, Andrea J. Gray, Franois Berthiaume. 2015. Mesenchymal
stromal cells reverse hypoxia-mediated suppression of-smooth muscle actin expression in human dermal fibroblasts. Biochemical
and Biophysical Research Communications 458:1, 8-13. [Crossref]
104. Park Dong-Soo, Park Jung-Chul, Lee Jung-Seok, Kim Tae-Wan, Kim Ki-Joon, Jung Byung-Joo, Shim Eun-Kyung, Choi Eun-
Young, Park So-Yon, Cho Kyoo-Sung, Kim Chang-Sung. 2015. Effect of FGF-2 on Collagen Tissue Regeneration by Human
Vertebral Bone Marrow Stem Cells. Stem Cells and Development 24:2, 228-243. [Abstract] [Full Text HTML] [Full Text PDF]
[Full Text PDF with Links]
105. Michael S. Hu, Tripp Leavitt, Samir Malhotra, Dominik Duscher, Michael S. Pollhammer, Graham G. Walmsley, Zeshaan N.
Maan, Alexander T. M. Cheung, Manfred Schmidt, Georg M. Huemer, Michael T. Longaker, H. Peter Lorenz. 2015. Stem
Cell-Based Therapeutics to Improve Wound Healing. Plastic Surgery International 2015, 1-7. [Crossref]
106. Giles T. S. Kirby, Stuart J. Mills, Allison J. Cowin, Louise E. Smith. 2015. Stem Cells for Cutaneous Wound Healing. BioMed
Research International 2015, 1-11. [Crossref]
107. Junwang Xu, Carlos Zgheib, Kenneth W. Liechty. miRNAs in Bone MarrowDerived Mesenchymal Stem Cells 111-136.
[Crossref]
108. Jazmin Ozsvar, Suzanne M. Mithieux, Richard Wang, Anthony S. Weiss. 2015. Elastin-based biomaterials and mesenchymal stem
cells. Biomater. Sci. 3:6, 800-809. [Crossref]
109. Erszebet Szilagyi, Premenand Sundivakkam, Tamara Nunez, Kavitha Premenand, Norma Kenyon, Amelia Bartholomew. Clinical
Aspects of Regenerative Medicine 507-526. [Crossref]
110. Elizabeth A. Wahl, Fernando A. Fierro, Thomas R. Peavy, Ursula Hopfner, Julian F. Dye, Hans-Gnther Machens, Jos T.
Egaa, Thilo L. Schenck. 2015. In Vitro Evaluation of Scaffolds for the Delivery of Mesenchymal Stem Cells to Wounds. BioMed
Research International 2015, 1-14. [Crossref]
111. M. Isakson, C. de Blacam, D. Whelan, A. McArdle, A. J. P. Clover. 2015. Mesenchymal Stem Cells and Cutaneous Wound
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

Healing: Current Evidence and Future Potential. Stem Cells International 2015, 1-12. [Crossref]
112. T.S. de Windt, L.A. Vonk, J.K. Buskermolen, J. Visser, M. Karperien, R.L.A.W. Bleys, W.J.A. Dhert, D.B.F. Saris. 2015.
Arthroscopic airbrush assisted cell implantation for cartilage repair in the knee: a controlled laboratory and human cadaveric study.
Osteoarthritis and Cartilage 23:1, 143-150. [Crossref]
113. Roberto Snchez-Snchez, Ana Brena-Molina, Valentn Martnez-Lpez, Yaaziel Melgarejo-Ramrez, Lenin Tamay de Dios,
Ricardo Gmez-Garca, Ma. de Lourdes Reyes-Fras, Lourdes Rodrguez-Rodrguez, David Garciadiego-Czares, Hayde Lugo-
Martnez, Clemente Ibarra, Mara Esther Martnez-Pardo, Cristina Velasquillo-Martnez. 2015. Generation of Two Biological
Wound Dressings as a Potential Delivery System of Human Adipose-Derived Mesenchymal Stem Cells. ASAIO Journal 61:6,
718-725. [Crossref]
114. Johanna Kober, Alfred Gugerell, Melanie Schmid, Lars-Peter Kamolz, Maike Keck. 2015. Generation of a Fibrin Based Three-
Layered Skin Substitute. BioMed Research International 2015, 1-8. [Crossref]
115. S. A. Eming, P. Martin, M. Tomic-Canic. 2014. Wound repair and regeneration: Mechanisms, signaling, and translation. Science
Translational Medicine 6:265, 265sr6-265sr6. [Crossref]
116. Sivan Unnikrishnan, Jayakumar K., Krishnan Lissy K.. 2014. Constitution of Fibrin-Based Niche for In Vitro Differentiation of
Adipose-Derived Mesenchymal Stem Cells to Keratinocytes. BioResearch Open Access 3:6, 339-347. [Abstract] [Full Text HTML]
[Full Text PDF] [Full Text PDF with Links]
117. McAndrews Kathleen M., Kim Min Jeong, Lam Tuyet Y., McGrail Daniel J., Dawson Michelle R.. 2014. Architectural and
Mechanical Cues Direct Mesenchymal Stem Cell Interactions with Crosslinked Gelatin Scaffolds. Tissue Engineering Part A
20:23-24, 3252-3260. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links] [Supplemental Material]
118. Thekkeparambil Chandrabose Srijaya, Thamil Selvee Ramasamy, Noor Hayaty Abu Kasim. 2014. Advancing stem cell therapy
from bench to bedside: lessons from drug therapies. Journal of Translational Medicine 12:1. . [Crossref]
119. D. Whelan, N.M. Caplice, A.J.P. Clover. 2014. Fibrin as a delivery system in wound healing tissue engineering applications.
Journal of Controlled Release 196, 1-8. [Crossref]
120. Alfred Gugerell, Waltraud Pasteiner, Sylvia Nrnberger, Johanna Kober, Alexandra Meinl, Sabine Pfeifer, Joachim Hartinger,
Susanne Wolbank, Andreas Goppelt, Heinz Redl, Rainer Mittermayr. 2014. Thrombin as important factor for cutaneous wound
healing: Comparison of fibrin biomatrices in vitro and in a rat excisional wound healing model. Wound Repair and Regeneration
22:6, 740-748. [Crossref]
121. King Alice, Balaji Swathi, Keswani Sundeep G., Crombleholme Timothy M.. 2014. The Role of Stem Cells in Wound
Angiogenesis. Advances in Wound Care 3:10, 614-625. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with
Links]
122. Junwang Xu, Carlos Zgheib, Junyi Hu, Wenjie Wu, Liping Zhang, Kenneth W. Liechty. 2014. The role of microRNA-15b in
the impaired angiogenesis in diabetic wounds. Wound Repair and Regeneration 22:5, 671-677. [Crossref]
123. Yan Xiang-Zhen, van den Beucken Jeroen J.J.P., Both Sanne K., Yang Pi-Shan, Jansen John A., Yang Fang. 2014. Biomaterial
Strategies for Stem Cell Maintenance During In Vitro Expansion. Tissue Engineering Part B: Reviews 20:4, 340-354. [Abstract]
[Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
124. Dimitrios Baltzis, Ioanna Eleftheriadou, Aristidis Veves. 2014. Pathogenesis and Treatment of Impaired Wound Healing in
Diabetes Mellitus: New Insights. Advances in Therapy 31:8, 817-836. [Crossref]
125. Nkemcho O. Ojeh, Harshad A. Navsaria. 2014. An in vitro skin model to study the effect of mesenchymal stem cells in wound
healing and epidermal regeneration. Journal of Biomedical Materials Research Part A 102:8, 2785-2792. [Crossref]
126. Dong Won Lee, Yeo Reum Jeon, Eul Je Cho, Jong Hwa Kang, Dae Hyun Lew. 2014. Optimal administration routes for adipose-
derived stem cells therapy in ischaemic flaps. Journal of Tissue Engineering and Regenerative Medicine 8:8, 596-603. [Crossref]
127. Annelies Bronckaers, Petra Hilkens, Wendy Martens, Pascal Gervois, Jessica Ratajczak, Tom Struys, Ivo Lambrichts. 2014.
Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate angiogenesis. Pharmacology &
Therapeutics 143:2, 181-196. [Crossref]
128. Michael S. Hu, Zeshaan N. Maan, Jen-Chieh Wu, Robert C. Rennert, Wan Xing Hong, Tiffany S. Lai, Alexander T. M. Cheung,
Graham G. Walmsley, Michael T. Chung, Adrian McArdle, Michael T. Longaker, H. Peter Lorenz. 2014. Tissue Engineering
and Regenerative Repair in Wound Healing. Annals of Biomedical Engineering 42:7, 1494-1507. [Crossref]
129. Grace C. Davey, Swapnil B. Patil, Aonghus OLoughlin, Timothy OBrien. 2014. Mesenchymal Stem Cell-Based
Treatment for Microvascular and Secondary Complications of Diabetes Mellitus. Frontiers in Endocrinology 5. . [Crossref]
130. Mohan R. Dasu, Sandra R. Ramirez, Thi Dinh La, Farzam Gorouhi, Chuong Nguyen, Benjamin R. Lin, Chelcy Mashburn,
Heather Stewart, Thomas R. Peavy, Jan A. Nolta, Roslyn R. Isseroff. 2014. Crosstalk Between Adrenergic and Toll-Like Receptors
in Human Mesenchymal Stem Cells and Keratinocytes: A Recipe for Impaired Wound Healing. STEM CELLS Translational
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

Medicine 3:6, 745-759. [Crossref]


131. Lijun Ding, Xinan Li, Haixiang Sun, Jing Su, Nacheng Lin, Bruno Pault, Tianran Song, Jun Yang, Jianwu Dai, Yali Hu.
2014. Transplantation of bone marrow mesenchymal stem cells on collagen scaffolds for the functional regeneration of injured
rat uterus. Biomaterials 35:18, 4888-4900. [Crossref]
132. Somaya A.A. Mohammed, Manal H. Moussa, Safaa M. Shaker, Shahinaz A. Ahmed. 2014. The effect of bone marrow-derived
mesenchymal stem cells on the healing of experimental skin wound in diabetic adult male albino rats. The Egyptian Journal of
Histology 37:2, 360-372. [Crossref]
133. Ratti Ram Sharma, Kathryn Pollock, Allison Hubel, David McKenna. 2014. Mesenchymal stem or stromal cells: a review of
clinical applications and manufacturing practices. Transfusion 54:5, 1418-1437. [Crossref]
134. B. Laverdet, L. Micallef, C. Lebreton, J. Mollard, J.-J. Lataillade, B. Coulomb, A. Desmoulire. 2014. Use of mesenchymal stem
cells for cutaneous repair and skin substitute elaboration. Pathologie Biologie 62:2, 108-117. [Crossref]
135. Jamie A. Textor, Kaitlin C. Murphy, J. Kent Leach, Fern Tablin. 2014. Ultrastructure and growth factor content of equine
platelet-rich fibrin gels. American Journal of Veterinary Research 75:4, 392-401. [Crossref]
136. Ann Katharin Reckhenrich, Bianca Manuela Kirsch, Elizabeth Ann Wahl, Thilo Ludwig Schenck, Farid Rezaeian, Yves Harder,
Peter Foehr, Hans-Gnther Machens, Jos Toms Egaa. 2014. Surgical Sutures Filled with Adipose-Derived Stem Cells Promote
Wound Healing. PLoS ONE 9:3, e91169. [Crossref]
137. Miao Teng, Yuesheng Huang, Hengshu Zhang. 2014. Application of stems cells in wound healing-An update. Wound Repair
and Regeneration 22:2, 151-160. [Crossref]
138. Melissa Hu, David Ludlow, J. Steven Alexander, Jerry McLarty, Timothy Lian. 2014. Improved wound healing of postischemic
cutaneous flaps with the use of bone marrow-derived stem cells. The Laryngoscope 124:3, 642-648. [Crossref]
139. A. Bakhtina, M. Tohfafarosh, A. Lichtler, T. Livingston Arinzeh. 2014. Characterization and differentiation potential of rabbit
mesenchymal stem cells for translational regenerative medicine. In Vitro Cellular & Developmental Biology - Animal 50:3, 251-260.
[Crossref]
140. Dash Surjya Narayan, Dash Nihar Ranjan, Guru Bhikaricharan, Mohapatra Prakash Chandra. 2014. Towards Reaching the Target:
Clinical Application of Mesenchymal Stem Cells for Diabetic Foot Ulcers. Rejuvenation Research 17:1, 40-53. [Abstract] [Full
Text HTML] [Full Text PDF] [Full Text PDF with Links]
141. Laurie A. McDuffee, Blanca P. Esparza Gonzalez, Rodolfo Nino-Fong, Enrique Aburto. 2014. Evaluation of an in vivo heterotopic
model of osteogenic differentiation of equine bone marrow and muscle mesenchymal stem cells in fibrin glue scaffold. Cell and
Tissue Research 355:2, 327-335. [Crossref]
142. Yu Qi, Dongsheng Jiang, Anca Sindrilaru, Agatha Stegemann, Susanne Schatz, Nicolai Treiber, Markus Rojewski, Hubert
Schrezenmeier, Seppe Vander Beken, Meinhard Wlaschek, Markus Bhm, Andreas Seitz, Natalie Scholz, Lutz Drselen, Jrgen
Brinckmann, Anita Ignatius, Karin Scharffetter-Kochanek. 2014. TSG-6 Released from Intradermally Injected Mesenchymal
Stem Cells Accelerates Wound Healing and Reduces Tissue Fibrosis in Murine Full-Thickness Skin Wounds. Journal of
Investigative Dermatology 134:2, 526-537. [Crossref]
143. Simon Myers, Harshad Navsaria, Nkemcho Ojeh. Skin Engineering and Keratinocyte Stem Cell Therapy 497-528. [Crossref]
144. Morgan T. Sutton, Tracey L. Bonfield. 2014. Stem Cells: Innovations in Clinical Applications. Stem Cells International 2014,
1-9. [Crossref]
145. Witold N Nowak, Sebastian Borys, Katarzyna Kusiska, Karolina Bukowska-Strakova, Przemysaw Witek, Teresa Koblik, Alicja
Jzkowicz, Maciej Tadeusz Maecki, Jzef Dulak. 2014. Number of circulating pro-angiogenic cells, growth factor and anti-
oxidative gene profiles might be altered in type 2 diabetes with and without diabetic foot syndrome. Journal of Diabetes Investigation
5:1, 99-107. [Crossref]
146. Austin Nuschke, Melanie Rodrigues, Donna B Stolz, Charleen T Chu, Linda Griffith, Alan Wells. 2014. Human mesenchymal
stem cells/multipotent stromal cells consume accumulated autophagosomes early in differentiation. Stem Cell Research & Therapy
5:6, 140. [Crossref]
147. Reto Wettstein, Miodrag Savic, Gerhard Pierer, Oliver Scheufler, Martin Haug, Jrg Halter, Alois Gratwohl, Michael Baumberger,
Dirk Schaefer, Daniel Kalbermatten. 2014. Progenitor cell therapy for sacral pressure sore: a pilot study with a novel human
chronic wound model. Stem Cell Research & Therapy 5:1, 18. [Crossref]
148. Yasumasa Kuroda, Mari Dezawa. 2014. Mesenchymal Stem Cells and Their Subpopulation, Pluripotent Muse Cells, in Basic
Research and Regenerative Medicine. The Anatomical Record 297:1, 98-110. [Crossref]
149. Austin Nuschke. 2014. Activity of mesenchymal stem cells in therapies for chronic skin wound healing. Organogenesis 10:1, 29-37.
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

[Crossref]
150. Ganary Dabiri, Douglas Heiner, Vincent Falanga. 2013. The emerging use of bone marrow-derived mesenchymal stem cells in
the treatment of human chronic wounds. Expert Opinion on Emerging Drugs 18:4, 405-419. [Crossref]
151. Ludovic Zimmerlin, Tea Soon Park, Elias T. Zambidis, Vera S. Donnenberg, Albert D. Donnenberg. 2013. Mesenchymal stem
cell secretome and regenerative therapy aftercancer. Biochimie 95:12, 2235-2245. [Crossref]
152. Satori Iwamoto, Xiaofeng Lin, Ron Ramirez, Polly Carson, David Fiore, Jane Goodrich, Tatyana Yufit, Vincent Falanga. 2013.
Bone Marrow Cell Mobilization by the Systemic Use of Granulocyte Colony-Stimulating Factor (GCSF) Improves Wound Bed
Preparation. The International Journal of Lower Extremity Wounds 12:4, 256-264. [Crossref]
153. David Antonio Cantu, W. John Kao. 2013. Combinatorial Biomatrix/Cell-Based Therapies for Restoration of Host Tissue
Architecture and Function. Advanced Healthcare Materials 2:12, 1544-1563. [Crossref]
154. Matthew B Murphy, Kathryn Moncivais, Arnold I Caplan. 2013. Mesenchymal stem cells: environmentally responsive therapeutics
for regenerative medicine. Experimental & Molecular Medicine 45:11, e54. [Crossref]
155. Daniel C. Roy, Nancie A. Mooney, Carol H. Raeman, Diane Dalecki, Denise C. Hocking. 2013. Fibronectin Matrix Mimetics
Promote Full-Thickness Wound Repair in Diabetic Mice. Tissue Engineering Part A 19:21-22, 2517-2526. [Abstract] [Full Text
HTML] [Full Text PDF] [Full Text PDF with Links] [Supplemental Material]
156. Ganary Dabiri, Vincent Falanga. 2013. Connective tissue ulcers. Journal of Tissue Viability 22:4, 92-102. [Crossref]
157. Damien P. Kuffler. 2013. Platelet-Rich Plasma and the Elimination of Neuropathic Pain. Molecular Neurobiology 48:2, 315-332.
[Crossref]
158. Ling-Ying Liu, Yu-Sen Hou, Jia-Ke Chai, Quan Hu, Hong-Jie Duan, Yong-Hui Yu, Hui-Nan Yin, Dai-Feng Hao, Guang Feng,
Tao Li, Jun-Dong Du. 2013. Basic fibroblast growth factor/vascular endothelial growth factor in the serum from severe burn
patients stimulates the proliferation of cultured human umbilical cord mesenchymal stem cells via activation of Notch signaling
pathways. Journal of Trauma and Acute Care Surgery 75:5, 789-797. [Crossref]
159. Seok-Ho Hong, Leila Maghen, Shlomit Kenigsberg, Anouk-Martine Teichert, Ashley W. Rammeloo, Ekaterina Shlush, Peter
Szaraz, Schreiber Pereira, Ayub Lulat, Rong Xiao, Shang-Mian Yie, Andre Gauthier-Fisher, Clifford L. Librach. 2013. Ontogeny
of Human Umbilical Cord Perivascular Cells: Molecular and Fate Potential Changes During Gestation. Stem Cells and Development
22:17, 2425-2439. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links] [Supplemental Material]
160. Naoki Morimoto, Kenichi Yoshimura, Miyuki Niimi, Tatsuya Ito, Rino Aya, Junpei Fujitaka, Harue Tada, Satoshi Teramukai,
Toshinori Murayama, Chikako Toyooka, Kazumi Miura, Satoru Takemoto, Norikazu Kanda, Katsuya Kawai, Masayuki Yokode,
Akira Shimizu, Shigehiko Suzuki. 2013. Novel Collagen/Gelatin Scaffold with Sustained Release of Basic Fibroblast Growth
Factor: Clinical Trial for Chronic Skin Ulcers. Tissue Engineering Part A 19:17-18, 1931-1940. [Abstract] [Full Text HTML]
[Full Text PDF] [Full Text PDF with Links]
161. William J. Ennis, Audrey Sui, Amelia Bartholomew. 2013. Stem Cells and Healing: Impact on Inflammation. Advances in Wound
Care 2:7, 369-378. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
162. Stuart Mills, Allison Cowin, Pritinder Kaur. 2013. Pericytes, Mesenchymal Stem Cells and the Wound Healing Process. Cells
2:3, 621-634. [Crossref]
163. D. H. You, M. J. Nam. 2013. Effects of human epidermal growth factor gene-transfected mesenchymal stem cells on fibroblast
migration and proliferation. Cell Proliferation 46:4, 408-415. [Crossref]
164. Yanan Jiang, Bing Chen, Yongbo Liu, Zhongyin Zhufu, Xin Yan, Xianglin Hou, Jianwu Dai, Qian Tan. 2013. Effect of collagen
scaffold with adipose-derived stromal vascular fraction cells on diabetic wound healing: A study in a diabetic porcine model. Tissue
Engineering and Regenerative Medicine 10:4, 192-199. [Crossref]
165. Joong Hyun Shim, Ju-Yearl Park, Mi-Gi Lee, Hak Hee Kang, Tae Ryong Lee, Dong Wook Shin. 2013. Human Dermal Stem/
Progenitor Cell-Derived Conditioned Medium Ameliorates Ultraviolet A-Induced Damage of Normal Human Dermal Fibroblasts.
PLoS ONE 8:7, e67604. [Crossref]
166. A. O'Loughlin, M. Kulkarni, M. Creane, E. E. Vaughan, E. Mooney, G. Shaw, M. Murphy, P. Dockery, A. Pandit, T. O'Brien.
2013. Topical Administration of Allogeneic Mesenchymal Stromal Cells Seeded in a Collagen Scaffold Augments Wound Healing
and Increases Angiogenesis in the Diabetic Rabbit Ulcer. Diabetes 62:7, 2588-2594. [Crossref]
167. Jasper Killat, Kerstin Reimers, Claudia Choi, Sabrina Jahn, Peter Vogt, Christine Radtke. 2013. Cultivation of Keratinocytes and
Fibroblasts in a Three-Dimensional Bovine Collagen-Elastin Matrix (Matriderm) and Application for Full Thickness Wound
Coverage in Vivo. International Journal of Molecular Sciences 14:7, 14460-14474. [Crossref]
168. Chris A. Bashur, Raj R. Rao, Anand Ramamurthi. 2013. Perspectives on Stem Cell-Based Elastic Matrix Regenerative Therapies
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

for Abdominal Aortic Aneurysms. STEM CELLS Translational Medicine 2:6, 401-408. [Crossref]
169. Simon Lee, Erzsebet Szilagyi, Lin Chen, Kavitha Premanand, Luisa A. DiPietro, William Ennis, Amelia M. Bartholomew. 2013.
Activated mesenchymal stem cells increase wound tensile strength in aged mouse model via macrophages. Journal of Surgical
Research 181:1, 20-24. [Crossref]
170. Kiarash Khosrotehrani. 2013. Mesenchymal stem cell therapy in skin: why and what for?. Experimental Dermatology 22:5, 307-310.
[Crossref]
171. Robert J. Harman. Stem Cell Therapy in Veterinary Dermatology 99-107. [Crossref]
172. Ju-Won Kim, Jong-Hwan Lee, Young S. Lyoo, Dong-In Jung, Hee-Myung Park. 2013. The effects of topical mesenchymal stem
cell transplantation in canine experimental cutaneous wounds. Veterinary Dermatology 24:2, 242-e53. [Crossref]
173. Yukun Cao, Ting Yang, Chunhu Gu, Dinghua Yi. 2013. Pigment epithelium-derived factor delays cellular senescence of human
mesenchymal stem cells in vitro by reducing oxidative stress. Cell Biology International 37:4, 305-313. [Crossref]
174. Joyce Doorn, Hugo A.M. Fernandes, Bach Q. Le, Jeroen van de Peppel, Johannes P.T.M. van Leeuwen, Margreet R. De Vries,
Zeen Aref, Paul H.A. Quax, Ola Myklebost, Daniel B.F. Saris, Clemens A. van Blitterswijk, Jan de Boer. 2013. A small molecule
approach to engineering vascularized tissue. Biomaterials 34:12, 3053-3063. [Crossref]
175. Hannu Larjava, Yi Yang, Edward Putnins, Jyrki Heino, Lari Hkkinen. Biological Agents and Cell Therapies in Periodontal
Regeneration 261-285. [Crossref]
176. Fabio Castiglione, Petter Hedlund, Frank Van der Aa, Trinity J. Bivalacqua, Patrizio Rigatti, Hein Van Poppel, Francesco Montorsi,
Dirk De Ridder, Maarten Albersen. 2013. Intratunical Injection of Human Adipose Tissuederived Stem Cells Prevents Fibrosis
and Is Associated with Improved Erectile Function in a Rat Model of Peyronie's Disease. European Urology 63:3, 551-560.
[Crossref]
177. Olle Ringdn, Tom Erkers, Silvia Nava, Mehmet Uzunel, Erik Iwarsson, Rka Conrad, Magnus Westgren, Jonas Mattsson, Helen
Kaipe. 2013. Fetal Membrane Cells for Treatment of Steroid-Refractory Acute Graft-Versus-Host Disease. STEM CELLS 31:3,
592-601. [Crossref]
178. Dongsheng Jiang, Yu Qi, Nathan G. Walker, Anca Sindrilaru, Adelheid Hainzl, Meinhard Wlaschek, Sheila MacNeil, Karin
Scharffetter-Kochanek. 2013. The effect of adipose tissue derived MSCs delivered by a chemically defined carrier on full-thickness
cutaneous wound healing. Biomaterials 34:10, 2501-2515. [Crossref]
179. Radbeh Torabi, Vincenzo Villani, Christopher A. Mallard, Curtis L. Cetrulo. The Translational Potential of Perinatal Stem Cells
in Clinical Medicine: Mesenchymal Stem Cells 105-117. [Crossref]
180. Federica Casiraghi, Giuseppe Remuzzi, Mauro Abbate, Norberto Perico. 2013. Multipotent Mesenchymal Stromal Cell Therapy
and Risk of Malignancies. Stem Cell Reviews and Reports 9:1, 65-79. [Crossref]
181. Vitali Alexeev, Adele Donahue, Jouni Uitto, Olga Igoucheva. 2013. Analysis of chemotactic molecules in bone marrow-derived
mesenchymal stem cells and the skin: Ccl27-Ccr10 axis as a basis for targeting to cutaneous tissues. Cytotherapy 15:2, 171-184.e1.
[Crossref]
182. Robert J. Harman. 2013. Stem cell therapy in veterinary dermatology. Veterinary Dermatology 24:1, 90-e24. [Crossref]
183. Christina Gnter, Hans-Gnther Machens. Innovative Solutions for Wound Therapy 475-494. [Crossref]
184. Supakanda Sukpat, Nipan Isarasena, Jutamas Wongphoom, Suthiluk Patumraj. 2013. Vasculoprotective Effects of Combined
Endothelial Progenitor Cells and Mesenchymal Stem Cells in Diabetic Wound Care: Their Potential Role in Decreasing Wound-
Oxidative Stress. BioMed Research International 2013, 1-8. [Crossref]
185. Jiangbo Wan, Liulu Xia, Wenjia Liang, Yi Liu, Qian Cai. 2013. Transplantation of Bone Marrow-Derived Mesenchymal Stem
Cells Promotes Delayed Wound Healing in Diabetic Rats. Journal of Diabetes Research 2013, 1-11. [Crossref]
186. Chunli Hou, Lei Shen, Qiang Huang, Jianhong Mi, Yangxiao Wu, Mingcan Yang, Wen Zeng, Li Li, Wen Chen, Chuhong Zhu.
2013. The effect of heme oxygenase-1 complexed with collagen on MSC performance in the treatment of diabetic ischemic ulcer.
Biomaterials 34:1, 112-120. [Crossref]
187. Ludovic Zimmerlin, J. Peter Rubin, Melanie E. Pfeifer, Linda R. Moore, Vera S. Donnenberg, Albert D. Donnenberg. 2013.
Human adipose stromal vascular cell delivery in a fibrin spray. Cytotherapy 15:1, 102-108. [Crossref]
188. Julia van de Kamp, Willi Jahnen-Dechent, Bjoern Rath, Ruth Knuechel, Sabine Neuss. 2013. Hepatocyte Growth Factor-Loaded
Biomaterials for Mesenchymal Stem Cell Recruitment. Stem Cells International 2013, 1-9. [Crossref]
189. Yunling Li, Lei Zheng, Xia Xu, Lili Song, Yin Li, Wei Li, Suhan Zhang, Feng Zhang, Haiyan Jin. 2013. Mesenchymal stem cells
modified with angiopoietin-1 gene promote wound healing. Stem Cell Research & Therapy 4:5, 113. [Crossref]
190. Georgette Oni, Charlotte Lequeux, Min-Jeong Cho, Daniel Zhang, Eric Lazcano, Spencer A. Brown, Jeffrey M. Kenkel. 2013.
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

Transdermal Delivery of Adipocyte-Derived Stem Cells Using a Fractional Ablative Laser. Aesthetic Surgery Journal 33:1, 109-116.
[Crossref]
191. Chandrama Shrestha, Liling Zhao, Ke Chen, Honghui He, Zhaohui Mo. 2013. Enhanced Healing of Diabetic Wounds by
Subcutaneous Administration of Human Umbilical Cord Derived Stem Cells and Their Conditioned Media. International Journal
of Endocrinology 2013, 1-10. [Crossref]
192. Mei Yang, Lingling Sheng, Tian R. Zhang, Qingfeng Li. 2013. Stem Cell Therapy for Lower Extremity Diabetic Ulcers: Where
Do We Stand?. BioMed Research International 2013, 1-8. [Crossref]
193. Soon-Jung Park, Sung-Hwan Moon, Hye-Jin Lee, Joa-Jin Lim, Jung-Mo Kim, Joseph Seo, Ji-Woon Yoo, Ok-Jung Kim, Sun-
Woong Kang, Hyung-Min Chung. 2013. A comparison of human cord blood- and embryonic stem cell-derived endothelial
progenitor cells in the treatment of chronic wounds. Biomaterials 34:4, 995-1003. [Crossref]
194. Daniela Dreymueller, Bernd Denecke, Andreas Ludwig, Willi Jahnen-Dechent. 2013. Embryonic stem cell-derived M2-like
macrophages delay cutaneous wound healing. Wound Repair and Regeneration 21:1, 44-54. [Crossref]
195. Gerard Marx. Fibrinogen: Science and Biotechnology 117-135. [Crossref]
196. Kai Wang, Yong Guan, Yi Liu, Meifeng Zhu, Ting Li, Di An, Lailiang Ou, Yongzhe Che, Guorong Zhang, Jun Zhang, Xi-
Long Zheng, Deling Kong. 2012. Fibrin Glue with Autogenic Bone Marrow Mesenchymal Stem Cells for Urethral Injury Repair
in Rabbit Model. Tissue Engineering Part A 18:23-24, 2507-2517. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text
PDF with Links] [Supplemental Material]
197. Gerit D Mulder, Daniel K Lee, Nathan S Jeppesen. 2012. Comprehensive review of the clinical application of autologous
mesenchymal stem cells in the treatment of chronic wounds and diabetic bone healing. International Wound Journal 9:6, 595-600.
[Crossref]
198. Sheng-Lian Yang, Erica Harnish, Thomas Leeuw, Uwe Dietz, Erika Batchelder, Paul S. Wright, Jane Peppard, Paul August, Cecile
Volle-Challier, Francoise Bono, Jean-Marc Herbert, Juan Carlos Izpisua Belmonte. 2012. Compound screening platform using
human induced pluripotent stem cells to identify small molecules that promote chondrogenesis. Protein & Cell 3:12, 934-942.
[Crossref]
199. J. J. Heffner, J. W. Holmes, J. P. Ferrari, J. Krontiris-Litowitz, H. Marie, D. L. Fagan, J. C. Perko, H. A. Dorion. 2012. Bone
marrow-derived mesenchymal stromal cells and platelet-rich plasma on a collagen matrix to improve fascial healing. Hernia 16:6,
677-687. [Crossref]
200. Ji-Ping Zou, Sha Huang, Yan Peng, Hong-Wei Liu, Biao Cheng, Xiao-Bing Fu, Xiao-Fei Xiang. 2012. Mesenchymal Stem Cells/
Multipotent Mesenchymal Stromal Cells (MSCs). The International Journal of Lower Extremity Wounds 11:4, 244-253. [Crossref]
201. Xining Pang. 2012. Direct comparison of the potency of human mesenchymal stem cells derived from amnion tissue, bone marrow
and adipose tissue at inducing dermal fibroblast responses to cutaneous wounds. International Journal of Molecular Medicine .
[Crossref]
202. J. Xu, W. Wu, L. Zhang, W. Dorset-Martin, M. W. Morris, M. E. Mitchell, K. W. Liechty. 2012. The Role of MicroRNA-146a
in the Pathogenesis of the Diabetic Wound-Healing Impairment: Correction With Mesenchymal Stem Cell Treatment. Diabetes
61:11, 2906-2912. [Crossref]
203. Yi Liu, Songlin Wang, Songtao Shi. 2012. The role of recipient T cells in mesenchymal stem cell-based tissue regeneration. The
International Journal of Biochemistry & Cell Biology 44:11, 2044-2050. [Crossref]
204. Diego M. Castilla, Zhao-Jun Liu, Runxia Tian, Yan Li, Alan S. Livingstone, Omaida C. Velazquez. 2012. A Novel Autologous
Cell-Based Therapy to Promote Diabetic Wound Healing. Annals of Surgery 256:4, 560-572. [Crossref]
205. Marius Strioga, Sowmya Viswanathan, Adas Darinskas, Ondrej Slaby, Jaroslav Michalek. 2012. Same or Not the Same?
Comparison of Adipose Tissue-Derived Versus Bone Marrow-Derived Mesenchymal Stem and Stromal Cells. Stem Cells and
Development 21:14, 2724-2752. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
206. Weerapong Prasongchean, Patrizia Ferretti. 2012. Autologous stem cells for personalised medicine. New Biotechnology 29:6,
641-650. [Crossref]
207. Ryutaro Shohara, Akihito Yamamoto, Sachiko Takikawa, Akira Iwase, Hideharu Hibi, Fumitaka Kikkawa, Minoru Ueda.
2012. Mesenchymal stromal cells of human umbilical cord Wharton's jelly accelerate wound healing by paracrine mechanisms.
Cytotherapy 14:10, 1171-1181. [Crossref]
208. Swathi Balaji, Sundeep G. Keswani, Timothy M. Crombleholme. 2012. The Role of Mesenchymal Stem Cells in the Regenerative
Wound Healing Phenotype. Advances in Wound Care 1:4, 159-165. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text
PDF with Links]
209. O. Dereure. 2012. Cellules souches en dermatologie: concepts, identification et intrt. Annales de Dermatologie et de Vnrologie
139:8-9, 568-578. [Crossref]
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

210. Andrea D. Maderal, Alejandra C. Vivas, Thomas G. Zwick, Robert S. Kirsner. 2012. Diabetic Foot Ulcers: Evaluation and
Management. Hospital Practice 40:3, 102-115. [Crossref]
211. Gerit Mulder, Kelly Wallin, Mayer Tenenhaus. 2012. Regenerative Materials That Facilitate Wound Healing. Clinics in Plastic
Surgery 39:3, 249-267. [Crossref]
212. Xiao-Yan Jiang, De-Bin Lu, Bing Chen. 2012. Progress in stem cell therapy for the diabetic foot. Diabetes Research and Clinical
Practice 97:1, 43-50. [Crossref]
213. Sadanori Akita, Hiroshi Yoshimoto, Kozo Akino, Akira Ohtsuru, Kenji Hayashida, Akiyoshi Hirano, Keiji Suzuki, Shunichi
Yamashita. 2012. Early Experiences with Stem Cells in Treating Chronic Wounds. Clinics in Plastic Surgery 39:3, 281-292.
[Crossref]
214. Summer Hanson, Peiman Hematti. Clinical Applications of Mesenchymal Stem CellBiomaterial Constructs for Tissue
Reconstruction 479-492. [Crossref]
215. Zin Z. Khaing, Christine E. Schmidt. 2012. Advances in natural biomaterials for nerve tissue repair. Neuroscience Letters 519:2,
103-114. [Crossref]
216. Vincent Falanga. 2012. Stem Cells in Tissue Repair and Regeneration. Journal of Investigative Dermatology 132:6, 1538-1541.
[Crossref]
217. Jinling Wang, Wugeng Cui, Jianhong Ye, Shufeng Ji, Xiufeng Zhao, Linghui Zhan, Jiachun Feng, Zhigang Zhang, Yilin Zhao.
2012. A cellular delivery system fabricated with autologous BMSCs and collagen scaffold enhances angiogenesis and perfusion in
ischemic hind limb. Journal of Biomedical Materials Research Part A 100A:6, 1438-1447. [Crossref]
218. Joung-Hwan Oh, Hye-Jin Kim, Tae-Il Kim, Jeong-Hwa Baek, Hyun-Mo Ryoo, Kyung Mi Woo. 2012. The effects of the
modulation of the fibronectin-binding capacity of fibrin by thrombin on osteoblast differentiation. Biomaterials 33:16, 4089-4099.
[Crossref]
219. Xiuwen Wu, Jianan Ren, Jieshou Li. 2012. Fibrin glue as the cell-delivery vehicle for mesenchymal stromal cells in regenerative
medicine. Cytotherapy 14:5, 555-562. [Crossref]
220. Anthony J. P. Clover, Billy Lane O'Neill, Arun H. S. Kumar. 2012. Analysis of attitudes toward the source of progenitor cells in
tissue-engineered products for use in burns compared with other disease states. Wound Repair and Regeneration 20:3, 311-316.
[Crossref]
221. Vincent C. Van Der Veen, Marcel Vlig, Florine J. Van Milligen, Sharon I. De Vries, Esther Middelkoop, Magda M. W. Ulrich.
2012. Stem Cells in Burn Eschar. Cell Transplantation 21:5, 933-942. [Crossref]
222. Zhaoxia Zou, Erin Denny, Christine E. Brown, Michael C. Jensen, Gang Li, Tatsuhiro Fujii, Josh Neman, Rahul Jandial, Mike
Chen. 2012. Cytotoxic T Lymphocyte Trafficking and Survival in an Augmented Fibrin Matrix Carrier. PLoS ONE 7:4, e34652.
[Crossref]
223. Nynke M.S. van den Akker, Felix F. Kolk, Fabinne Jeukens, Sanne Verbruggen, Mick Gagliardi, Stefan Dullens, Ingo Heschel,
Mark J. Post, Danil G.M. Molin, Johannes Waltenberger. 2012. Vascular Potency of Sus Scrofa Bone MarrowDerived
Mesenchymal Stem Cells: A Progenitor Source of Medial but Not Endothelial Cells. Tissue Engineering Part A 18:7-8, 828-839.
[Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links] [Supplemental Material]
224. Joyce Doorn, Guido Moll, Katarina Le Blanc, Clemens van Blitterswijk, Jan de Boer. 2012. Therapeutic Applications of
Mesenchymal Stromal Cells: Paracrine Effects and Potential Improvements. Tissue Engineering Part B: Reviews 18:2, 101-115.
[Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
225. Sheila N. Blumberg, Alexandra Berger, Lisa Hwang, Irena Pastar, Stephen M. Warren, Weiliam Chen. 2012. The role of stem
cells in the treatment of diabetic foot ulcers. Diabetes Research and Clinical Practice 96:1, 1-9. [Crossref]
226. Kacey G. Marra, J. Peter Rubin. 2012. The potential of adipose-derived stem cells in craniofacial repair and regeneration. Birth
Defects Research Part C: Embryo Today: Reviews 96:1, 95-97. [Crossref]
227. . 207-218. [Crossref]
228. Hannu Larjava, Yi Yang, Edward Putnins, Jyrki Heino, Lari Hkkinen. 2012. Biological agents and cell therapies in periodontal
regeneration. Endodontic Topics 26:1, 18-40. [Crossref]
229. Zhi-Yong Zhang, Swee-Hin Teoh, James H.P. Hui, Nicholas M. Fisk, Mahesh Choolani, Jerry K.Y. Chan. 2012. The potential of
human fetal mesenchymal stem cells for off-the-shelf bone tissue engineering application. Biomaterials 33:9, 2656-2672. [Crossref]
230. Nathan G. Walker, Anita R. Mistry, Louise E. Smith, Paula C. Eves, Grigorios Tsaknakis, Simon Forster, Suzanne M. Watt, Sheila
MacNeil. 2012. A Chemically Defined Carrier for the Delivery of Human Mesenchymal Stem/Stromal Cells to Skin Wounds.
Tissue Engineering Part C: Methods 18:2, 143-155. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

231. Zoe Marie MacIsaac, Hulan Shang, Hitesh Agrawal, Ning Yang, Anna Parker, Adam J. Katz. 2012. Long-term in-vivo tumorigenic
assessment of human culture-expanded adipose stromal/stem cells. Experimental Cell Research 318:4, 416-423. [Crossref]
232. Hitomi Sano, Shigeru Ichioka, Sachio Kouraba, Ai Minamimura, Tomoya Sato, Naomi Sekiya, Masato Yasuta. 2012. Treatment of
venous ulcers with bone marrow-impregnated collagen matrix. Journal of Plastic Surgery and Hand Surgery 46:1, 37-44. [Crossref]
233. Benjamin W. Hale, Laurie R. Goodrich, David D. Frisbie, C. Wayne McIlwraith, John D. Kisiday. 2012. Effect of scaffold dilution
on migration of mesenchymal stem cells from fibrin hydrogels. American Journal of Veterinary Research 73:2, 313-318. [Crossref]
234. Scott Maxson, Erasmo A. Lopez, Dana Yoo, Alla Danilkovitch-Miagkova, Michelle A. LeRoux. 2012. Concise Review: Role of
Mesenchymal Stem Cells in Wound Repair. STEM CELLS Translational Medicine 1:2, 142-149. [Crossref]
235. Wesley M Jackson, Leon J Nesti, Rocky S Tuan. 2012. Mesenchymal stem cell therapy for attenuation of scar formation during
wound healing. Stem Cell Research & Therapy 3:3, 20. [Crossref]
236. Kristine C. Rustad, Victor W. Wong, Michael Sorkin, Jason P. Glotzbach, Melanie R. Major, Jayakumar Rajadas, Michael T.
Longaker, Geoffrey C. Gurtner. 2012. Enhancement of mesenchymal stem cell angiogenic capacity and stemness by a biomimetic
hydrogel scaffold. Biomaterials 33:1, 80-90. [Crossref]
237. Kenichi Tamama, Dominique J. Barbeau. 2012. Early Growth Response Genes Signaling Supports Strong Paracrine Capability
of Mesenchymal Stem Cells. Stem Cells International 2012, 1-7. [Crossref]
238. Victor W. Wong, Benjamin Levi, Jayakumar Rajadas, Michael T. Longaker, Geoffrey C. Gurtner. 2012. Stem Cell Niches for
Skin Regeneration. International Journal of Biomaterials 2012, 1-8. [Crossref]
239. Emily C Keats, Zia A Khan. 2012. Vascular stem cells in diabetic complications: evidence for a role in the pathogenesis and the
therapeutic promise. Cardiovascular Diabetology 11:1, 37. [Crossref]
240. Wesley M. Jackson, Leon J. Nesti, Rocky S. Tuan. 2012. Concise Review: Clinical Translation of Wound Healing Therapies
Based on Mesenchymal Stem Cells. STEM CELLS Translational Medicine 1:1, 44-50. [Crossref]
241. Jason P. Glotzbach, Sae Hee Ko, Geoffrey C. Gurtner, Michael T. Longaker. Regenerative Medicine 178-187. [Crossref]
242. Mairead Hayes, Gerard Curley, Bilal Ansari, John G Laffey. 2012. Clinical review: Stem cell therapies for acute lung injury/acute
respiratory distress syndrome - hope or hype?. Critical Care 16:2, 205. [Crossref]
243. Peter J. Amos, Esra Cagavi Bozkulak, Yibing Qyang. 2012. Methods of Cell Purification: A Critical Juncture for Laboratory
Research and Translational Science. Cells Tissues Organs 195:1-2, 26-40. [Crossref]
244. Sonal R. Tuljapurkar, John D. Jackson, Susan K. Brusnahan, Barbara J. OKane, John G. Sharp. 2012. Characterization of a
mesenchymal stem cell line that differentiates to bone and provides niches supporting mouse and human hematopoietic stem cells.
Stem Cell Discovery 02:01, 5-14. [Crossref]
245. Alan Yan, Tomer Avraham, Jamie C Zampell, Yosef S Haviv, Evan Weitman, Babak J Mehrara. 2011. Adipose-derived stem
cells promote lymphangiogenesis in response to VEGF-C stimulation or TGF-1 inhibition. Future Oncology 7:12, 1457-1473.
[Crossref]
246. Anna Arno, Alexandra H. Smith, Patrick H. Blit, Mohammed Al Shehab, Gerd G. Gauglitz, Marc G. Jeschke. 2011. Stem Cell
Therapy: A New Treatment for Burns?. Pharmaceuticals 4:12, 1355-1380. [Crossref]
247. Jonathan Brower, Sheila Blumberg, Emily Carroll, Irena Pastar, Harold Brem, Weiliam Chen. 2011. Mesenchymal Stem Cell
Therapy and Delivery Systems in Nonhealing Wounds. Advances in Skin & Wound Care 24:11, 524-532. [Crossref]
248. Alexander R Badiavas, Evangelos V Badiavas. 2011. Potential benefits of allogeneic bone marrow mesenchymal stem cells for
wound healing. Expert Opinion on Biological Therapy 11:11, 1447-1454. [Crossref]
249. Gerd G. Gauglitz, Marc G. Jeschke. 2011. Combined Gene and Stem Cell Therapy for Cutaneous Wound Healing. Molecular
Pharmaceutics 8:5, 1471-1479. [Crossref]
250. Guorui Jin, Molamma P. Prabhakaran, Seeram Ramakrishna. 2011. Stem cell differentiation to epidermal lineages on electrospun
nanofibrous substrates for skin tissue engineering. Acta Biomaterialia 7:8, 3113-3122. [Crossref]
251. Mohsen Khosravi Maharlooei, Mansooreh Bagheri, Zhabiz Solhjou, Behnam Moein Jahromi, Majid Akrami, Lili Rohani, Ahmad
Monabati, Ali Noorafshan, Gholamhossein Ranjbar Omrani. 2011. Adipose tissue derived mesenchymal stem cell (AD-MSC)
promotes skin wound healing in diabetic rats. Diabetes Research and Clinical Practice 93:2, 228-234. [Crossref]
252. T. Hodgkinson, A. Bayat. 2011. Dermal substitute-assisted healing: enhancing stem cell therapy with novel biomaterial design.
Archives of Dermatological Research 303:5, 301-315. [Crossref]
253. Jin Wang, Lianming Liao, Jianming Tan. 2011. Mesenchymal-stem-cell-based experimental and clinical trials: current status and
open questions. Expert Opinion on Biological Therapy 11:7, 893-909. [Crossref]
254. Ziyang Zhang, Wulf D. Ito, Ursula Hopfner, Bjrn Bhmert, Mathias Kremer, Ann K. Reckhenrich, Yves Harder, Natalie Lund,
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

Charli Kruse, Hans-Gnther Machens, Jos T. Egaa. 2011. The role of single cell derived vascular resident endothelial progenitor
cells in the enhancement of vascularization in scaffold-based skin regeneration. Biomaterials 32:17, 4109-4117. [Crossref]
255. Tu-Lai Yew, Yeh-Ting Hung, Hsin-Yang Li, Hsin-Wei Chen, Ling-Lan Chen, Kuo-Shu Tsai, Shih-Hwa Chiou, Kuan-Chong
Chao, Tung-Fu Huang, Hen-Li Chen, Shih-Chieh Hung. 2011. Enhancement of Wound Healing by Human Multipotent
Stromal Cell Conditioned Medium: The Paracrine Factors and p38 MAPK Activation. Cell Transplantation 20:5, 693-706.
[Crossref]
256. Richard Eglen, Terry Reisine. 2011. Primary Cells and Stem Cells in Drug Discovery: Emerging Tools for High-Throughput
Screening. ASSAY and Drug Development Technologies 9:2, 108-124. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text
PDF with Links]
257. T. Leclerc, C. Thepenier, P. Jault, E. Bey, J. Peltzer, M. Trouillas, P. Duhamel, L. Bargues, M. Prat, M. Bonderriter, J.-J.
Lataillade. 2011. Cell therapy of burns. Cell Proliferation 44, 48-54. [Crossref]
258. Tamer A.E. Ahmed, Emma V. Dare, Max Hincke. 2011. Fibrin: A Versatile Scaffold for Tissue Engineering Applications. Tissue
Engineering Part B: Reviews 77, 110306231744007. [Crossref]
259. Jason P. Glotzbach, Victor W. Wong, Geoffrey C. Gurtner, Michael T. Longaker. 2011. Regenerative Medicine. Current Problems
in Surgery 48:3, 148-212. [Crossref]
260. Benoit Hendrickx, Jan J. Vranckx, Aernout Luttun. 2011. Cell-Based Vascularization Strategies for Skin Tissue Engineering.
Tissue Engineering Part B: Reviews 17:1, 13-24. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
261. Gerwen Lammers, Pauline D.H.M. Verhaegen, Magda M.W. Ulrich, Joost Schalkwijk, Esther Middelkoop, Daniela Weiland,
Suzan T.M. Nillesen, Toin H. Van Kuppevelt, Willeke F. Daamen. 2011. An Overview of Methods for the In Vivo Evaluation of
Tissue-Engineered Skin Constructs. Tissue Engineering Part B: Reviews 17:1, 33-55. [Abstract] [Full Text HTML] [Full Text
PDF] [Full Text PDF with Links]
262. Mario Cherubino, J. Peter Rubin, Natasa Miljkovic, Arta Kelmendi-Doko, Kacey G. Marra. 2011. Adipose-Derived Stem Cells
for Wound Healing Applications. Annals of Plastic Surgery 66:2, 210-215. [Crossref]
263. Marcus Jger, Monika Herten, Ulrike Fochtmann, Johannes Fischer, Philippe Hernigou, Christoph Zilkens, Christian Hendrich,
Rdiger Krauspe. 2011. Bridging the gap: Bone marrow aspiration concentrate reduces autologous bone grafting in osseous defects.
Journal of Orthopaedic Research 29:2, 173-180. [Crossref]
264. William R Otto, Nicholas A Wright. 2011. Mesenchymal stem cells: from experiment to clinic. Fibrogenesis & Tissue Repair
4:1, 20. [Crossref]
265. Vladislav Volarevic, Nebojsa Arsenijevic, Miodrag L. Lukic, Miodrag Stojkovic. 2011. Concise Review: Mesenchymal Stem Cell
Treatment of the Complications of Diabetes Mellitus. STEM CELLS 29:1, 5-10. [Crossref]
266. Allison Nauta, Barrett Larson, Michael T. Longaker, H. Peter Lorenz. Scarless Wound Healing 103-127. [Crossref]
267. B.J. Larson, A. Nauta, K. Kawai, M.T. Longaker, H.P. Lorenz. Scarring and scarless wound healing 77-111. [Crossref]
268. Chul Han Kim, Jang Hyun Lee, Jong Ho Won, Moon Kyun Cho. 2011. Mesenchymal Stem Cells Improve Wound Healing
In Vivo via Early Activation of Matrix Metalloproteinase-9 and Vascular Endothelial Growth Factor. Journal of Korean Medical
Science 26:6, 726. [Crossref]
269. Vitali Alexeev, Jouni Uitto, Olga Igoucheva. 2011. Gene expression signatures of mouse bone marrow-derived mesenchymal stem
cells in the cutaneous environment and therapeutic implications for blistering skin disorder. Cytotherapy 13:1, 30-45. [Crossref]
270. G.G. Gauglitz, M.G. Jeschke. Stem cell therapies for wound repair 552-567. [Crossref]
271. Sae Hee Ko, Allison Nauta, Victor Wong, Jason Glotzbach, Geoffrey C. Gurtner, Michael T. Longaker. 2011. The Role of Stem
Cells in Cutaneous Wound Healing: What Do We Really Know?. Plastic and Reconstructive Surgery 127, 10S-20S. [Crossref]
272. Q. Zeng, L.K. Macri, A. Prasad, R.A.F. Clark, D.I. Zeugolis, C. Hanley, Y. Garcia, A. Pandit. Skin Tissue Engineering 467-499.
[Crossref]
273. James D. Kretlow, Patrick P. Spicer, John A. Jansen, Charles A. Vacanti, F. Kurtis Kasper, Antonios G. Mikos. 2010. Uncultured
Marrow Mononuclear Cells Delivered Within Fibrin Glue Hydrogels to Porous Scaffolds Enhance Bone Regeneration Within
Critical-Sized Rat Cranial Defects. Tissue Engineering Part A 16:12, 3555-3568. [Abstract] [Full Text HTML] [Full Text PDF]
[Full Text PDF with Links]
274. Ivan Hadad, Brian H. Johnstone, Jeffrey G. Brabham, Matthew W. Blanton, Pamela I. Rogers, Cory Fellers, James L. Solomon,
Stephanie Merfeld-Clauss, Colleen M. DesRosiers, Joseph R. Dynlacht, John J. Coleman, Keith L. March. 2010. Development of
a Porcine Delayed Wound-Healing Model and Its Use in Testing a Novel Cell-Based Therapy. International Journal of Radiation
Oncology*Biology*Physics 78:3, 888-896. [Crossref]
275. Kathryn L. Butler, Jeremy Goverman, Harry Ma, Alan Fischman, Yong-Ming Yu, Maryelizabeth Bilodeau, Ali M. Rad, Ali A.
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

Bonab, Ronald G. Tompkins, Shawn P. Fagan. 2010. Stem Cells and Burns: Review and Therapeutic Implications. Journal of
Burn Care & Research 31:6, 874-881. [Crossref]
276. Bo-Young Yoo, Youn-Ho Shin, Hee-Hoon Yoon, Young-Kwon Seo, Kye-Yong Song, Jung-Keug Park. 2010. Application of
mesenchymal stem cells derived from bone marrow and umbilical cord in human hair multiplication. Journal of Dermatological
Science 60:2, 74-83. [Crossref]
277. Nazlee Zebardast, David Lickorish, John E. Davies. 2010. Human umbilical cord perivascular cells (HUCPVC). Organogenesis
6:4, 197-203. [Crossref]
278. Ignacio Garca-Gmez, Gema Elvira, Agustn G Zapata, Mara L Lamana, Manuel Ramrez, Javier Garca Castro, Mariano Garca
Arranz, Angeles Vicente, Juan Bueren, Damin Garca-Olmo. 2010. Mesenchymal stem cells: biological properties and clinical
applications. Expert Opinion on Biological Therapy 10:10, 1453-1468. [Crossref]
279. Edward K. Merritt, Megan V. Cannon, David W. Hammers, Long N. Le, Rohit Gokhale, Apurva Sarathy, Tae J. Song, Matthew
T. Tierney, Laura J. Suggs, Thomas J. Walters, Roger P. Farrar. 2010. Repair of Traumatic Skeletal Muscle Injury with Bone-
Marrow-Derived Mesenchymal Stem Cells Seeded on Extracellular Matrix. Tissue Engineering Part A 16:9, 2871-2881. [Abstract]
[Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
280. Stephanie C. Wu, William Marston, David G. Armstrong. 2010. Wound Care. Journal of the American Podiatric Medical Association
100:5, 385-394. [Crossref]
281. Stephanie C. Wu, William Marston, David G. Armstrong. 2010. Wound care: The role of advanced wound healing technologies.
Journal of Vascular Surgery 52:3, 59S-66S. [Crossref]
282. Erika Deak, Erhard Seifried, Reinhard Henschler. 2010. Homing Pathways of Mesenchymal Stromal Cells (MSCs) and Their
Role in Clinical Applications. International Reviews of Immunology 29:5, 514-529. [Crossref]
283. Christina Dieckmann, Regina Renner, Linda Milkova, Jan C. Simon. 2010. Regenerative medicine in dermatology: biomaterials,
tissue engineering, stem cells, gene transfer and beyond. Experimental Dermatology 19:8, 697-706. [Crossref]
284. Anne M. Hocking, Nicole S. Gibran. 2010. Mesenchymal stem cells: Paracrine signaling and differentiation during cutaneous
wound repair. Experimental Cell Research 316:14, 2213-2219. [Crossref]
285. Mercedes Zurita, Laura Otero, Concepcin Aguayo, Celia Bonilla, Edgar Ferreira, Avelino Parajn, Jess Vaquero. 2010. Cell
therapy for spinal cord repair: optimization of biologic scaffolds for survival and neural differentiation of human bone marrow
stromal cells. Cytotherapy 12:4, 522-537. [Crossref]
286. Haruhisa Kawasaki, Jianjun Guan, Kenichi Tamama. 2010. Hydrogen gas treatment prolongs replicative lifespan of bone marrow
multipotential stromal cells in vitro while preserving differentiation and paracrine potentials. Biochemical and Biophysical Research
Communications 397:3, 608-613. [Crossref]
287. Peter J. Amos, Sahil K. Kapur, Peter C. Stapor, Hulan Shang, Stefan Bekiranov, Moshe Khurgel, George T. Rodeheaver, Shayn
M. Peirce, Adam J. Katz. 2010. Human Adipose-Derived Stromal Cells Accelerate Diabetic Wound Healing: Impact of Cell
Formulation and Delivery. Tissue Engineering Part A 16:5, 1595-1606. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text
PDF with Links] [Supplemental Material]
288. Von R. King, Alla Alovskaya, Diana Y.T. Wei, Robert A. Brown, John V. Priestley. 2010. The use of injectable forms of fibrin
and fibronectin to support axonal ingrowth after spinal cord injury. Biomaterials 31:15, 4447-4456. [Crossref]
289. Susan W. Volk. Mesenchymal Stem Cells in Ischemic Wound Healing 471-476. [Abstract] [Full Text PDF] [Full Text PDF
with Links]
290. Dustin M. Bermudez, Kenneth W. Liechty. Mesenchymal Stem Cells in Diabetic Wound Healing 477-482. [Abstract] [Full Text
PDF] [Full Text PDF with Links]
291. John Kowalczyk, Evangelos Badiavas. Bone Marrow Adult Stem and Progenitor Cells in the Treatment of Chronic Wounds
490-494. [Abstract] [Full Text PDF] [Full Text PDF with Links]
292. . Advances in Wound Care: Volume 1 . [Citation] [Full Text PDF] [Full Text PDF with Links]
293. Vincent Ronfard, Thomas Williams. Developments in Cell-Based Therapy for Wounds 412-418. [Abstract] [Full Text PDF]
[Full Text PDF with Links]
294. M.N.M. Walter, K.T. Wright, H.R. Fuller, S. MacNeil, W.E.B. Johnson. 2010. Mesenchymal stem cell-conditioned medium
accelerates skin wound healing: An in vitro study of fibroblast and keratinocyte scratch assays. Experimental Cell Research 316:7,
1271-1281. [Crossref]
295. Reut Shainer, Elena Gaberman, Lilia Levdansky, Raphael Gorodetsky. 2010. Efficient isolation and chondrogenic differentiation
of adult mesenchymal stem cells with fibrin microbeads and micronized collagen sponges. Regenerative Medicine 5:2, 255-265.
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

[Crossref]
296. L. Senseb, M. Krampera, H. Schrezenmeier, P. Bourin, R. Giordano. 2010. Mesenchymal stem cells for clinical application.
Vox Sanguinis 98:2, 93-107. [Crossref]
297. Summer E. Hanson, Michael L. Bentz, Peiman Hematti. 2010. Mesenchymal Stem Cell Therapy for Nonhealing Cutaneous
Wounds. Plastic and Reconstructive Surgery 125:2, 510-516. [Crossref]
298. J. Michael Sorrell, Marilyn A. Baber, Arnold I. Caplan. Influence of Adult Mesenchymal Stem Cells on In Vitro Vascular
Formation 230-240. [Abstract] [Full Text PDF] [Full Text PDF with Links]
299. . Advances in Tissue Engineering . [Citation] [Full Text PDF] [Full Text PDF with Links]
300. Lee C. Rogers, David G. Armstrong. Podiatry Care 1747-1760. [Crossref]
301. H. Drago, G.H. Marn, F. Sturla, G. Roque, K. Mrtire, V. Daz Aquino, R. Lamonega, C. Gardiner, T. Ichim, N. Riordan, J.C.
Raimondi, S. Bossi, A. Samadikuchaksaraei, M. van Leeuwen, J.M. Tau, L. Nez, G. Larsen, R. Spretz, E. Mansilla. 2010.
The Next Generation of Burns Treatment: Intelligent Films and Matrix, Controlled Enzymatic Debridement, and Adult Stem
Cells. Transplantation Proceedings 42:1, 345-349. [Crossref]
302. Andria N. Smith, Elise Willis, Vincent T. Chan, Lara A. Muffley, F. Frank Isik, Nicole S. Gibran, Anne M. Hocking. 2010.
Mesenchymal stem cells induce dermal fibroblast responses to injury. Experimental Cell Research 316:1, 48-54. [Crossref]
303. S Kumar, T R Nagy, S Ponnazhagan. 2010. Therapeutic potential of genetically modified adult stem cells for osteopenia. Gene
Therapy 17:1, 105-116. [Crossref]
304. Kenichi Tamama, Haruhisa Kawasaki, Alan Wells. 2010. Epidermal Growth Factor (EGF) Treatment on Multipotential Stromal
Cells (MSCs). Possible Enhancement of Therapeutic Potential of MSC. Journal of Biomedicine and Biotechnology 2010, 1-10.
[Crossref]
305. Katherine Lau, Ralf Paus, Stephan Tiede, Philip Day, Ardeshir Bayat. 2009. Exploring the role of stem cells in cutaneous wound
healing. Experimental Dermatology 18:11, 921-933. [Crossref]
306. Thomas Hodgkinson, Xue-Feng Yuan, Ardeshir Bayat. 2009. Adult stem cells in tissue engineering. Expert Review of Medical
Devices 6:6, 621-640. [Crossref]
307. Nihar Ranjan Dash, Surjya Narayan Dash, Padmanav Routray, Sribatsha Mohapatra, Prakash C. Mohapatra. 2009. Targeting
Nonhealing Ulcers of Lower Extremity in Human Through Autologous Bone Marrow-Derived Mesenchymal Stem Cells.
Rejuvenation Research 12:5, 359-366. [Abstract] [Full Text PDF] [Full Text PDF with Links]
308. Joseph Wagner, Thomas Kean, Randell Young, James E Dennis, Arnold I Caplan. 2009. Optimizing mesenchymal stem cell-
based therapeutics. Current Opinion in Biotechnology 20:5, 531-536. [Crossref]
309. Alon Ben-Ari, Rachel Rivkin, Miryam Frishman, Elena Gaberman, Lilia Levdansky, Raphael Gorodetsky. 2009. Isolation and
Implantation of Bone MarrowDerived Mesenchymal Stem Cells with Fibrin Micro Beads to Repair a Critical-Size Bone Defect
in Mice. Tissue Engineering Part A 15:9, 2537-2546. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
310. J. Michael Sorrell, Marilyn A. Baber, Arnold I. Caplan. 2009. Influence of Adult Mesenchymal Stem Cells on In Vitro Vascular
Formation. Tissue Engineering Part A 15:7, 1751-1761. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with
Links]
311. Liliana Guerra, Elena Dellambra, Laura Panacchia, Emanuel Paionni. 2009. Tissue Engineering for Damaged Surface and Lining
Epithelia: Stem Cells, Current Clinical Applications, and Available Engineered Tissues. Tissue Engineering Part B: Reviews 15:2,
91-112. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
312. Ailish Breen, Timothy O'Brien, Abhay Pandit. 2009. Fibrin as a Delivery System for Therapeutic Drugs and Biomolecules. Tissue
Engineering Part B: Reviews 15:2, 201-214. [Abstract] [Full Text HTML] [Full Text PDF] [Full Text PDF with Links]
313. Kim L. Kroeze, Wouter J. Jurgens, Behrouz Z. Doulabi, Florine J. van Milligen, Rik J. Scheper, Susan Gibbs. 2009. Chemokine-
Mediated Migration of Skin-Derived Stem Cells: Predominant Role for CCL5/RANTES. Journal of Investigative Dermatology
129:6, 1569-1581. [Crossref]
314. Jaymie Panuncialman, Vincent Falanga. 2009. The Science of Wound Bed Preparation. Surgical Clinics of North America 89:3,
611-626. [Crossref]
315. Alexander T. Hillel, Shyni Varghese, Jennifer Petsche, Michael J. Shamblott, Jennifer H. Elisseeff. 2009. Embryonic Germ Cells
Are Capable of Adipogenic Differentiation In Vitro and In Vivo. Tissue Engineering Part A 15:3, 479-486. [Abstract] [Full Text
HTML] [Full Text PDF] [Full Text PDF with Links]
316. Ludwik K. Branski, Gerd G. Gauglitz, David N. Herndon, Marc G. Jeschke. 2009. A review of gene and stem cell therapy in
cutaneous wound healing. Burns 35:2, 171-180. [Crossref]
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

317. Kim L Kroeze, Wouter J Jurgens, Behrouz Z Doulabi, Florine J van Milligen, Rik J Scheper, Susan Gibbs. 2009. Chemokine-
Mediated Migration of Skin-Derived Stem Cells: Predominant Role for CCL5/RANTES. Journal of Investigative Dermatology
. [Crossref]
318. John Girdlestone, Vasanti A. Limbani, Antony J. Cutler, Cristina V. Navarrete. 2009. Efficient expansion of mesenchymal stromal
cells from umbilical cord under low serum conditions. Cytotherapy 11:6, 738-748. [Crossref]
319. D. M. Choumerianou, H. Dimitriou, C. Perdikogianni, G. Martimianaki, M. Riminucci, M. Kalmanti. 2008. Study of oncogenic
transformation in ex vivo expanded mesenchymal cells, from paediatric bone marrow. Cell Proliferation 41:6, 909-922. [Crossref]
320. M. P. Alfaro, M. Pagni, A. Vincent, J. Atkinson, M. F. Hill, J. Cates, J. M. Davidson, J. Rottman, E. Lee, P. P. Young. 2008.
The Wnt modulator sFRP2 enhances mesenchymal stem cell engraftment, granulation tissue formation and myocardial repair.
Proceedings of the National Academy of Sciences 105:47, 18366-18371. [Crossref]
321. Agnieszka Banas, Takumi Teratani, Yusuke Yamamoto, Makoto Tokuhara, Fumitaka Takeshita, Mitsuhiko Osaki, Masaki
Kawamata, Takashi Kato, Hitoshi Okochi, Takahiro Ochiya. 2008. IFATS Collection: In Vivo Therapeutic Potential of Human
Adipose Tissue Mesenchymal Stem Cells After Transplantation into Mice with Liver Injury. STEM CELLS 26:10, 2705-2712.
[Crossref]
322. V. Falanga. 2008. Bone marrow cells can manipulate healing. Blood 113:5, 982-983. [Crossref]
323. Cui-ping ZHANG, Xiao-bing FU. 2008. Therapeutic potential of stem cells in skin repair and regeneration. Chinese Journal of
Traumatology (English Edition) 11:4, 209-221. [Crossref]
324. Isabel Zwart, Andrew J. Hill, John Girdlestone, Maria F. Manca, Roberto Navarrete, Cristina Navarrete, LingSun Jen. 2008.
Analysis of neural potential of human umbilical cord bloodderived multipotent mesenchymal stem cells in response to a range
of neurogenic stimuli. Journal of Neuroscience Research 86:9, 1902-1915. [Crossref]
325. Isabel Zwart, Andrew J. Hill, John Girdlestone, Maria F. Manca, Roberto Navarrete, Cristina Navarrete, LingSun Jen. 2008.
Analysis of neural potential of human umbilical cord bloodderived multipotent mesenchymal stem cells in response to a range
of neurogenic stimuli. Journal of Neuroscience Research 86:9, 1902-1915. [Crossref]
326. Tamer A.E. Ahmed, Emma V. Dare, Max Hincke. 2008. Fibrin: A Versatile Scaffold for Tissue Engineering Applications. Tissue
Engineering Part B: Reviews 14:2, 199-215. [Abstract] [Full Text PDF] [Full Text PDF with Links]
327. Marek Dudas, Annette Wysocki, Brian Gelpi, Tai-Lan Tuan. 2008. Memory Encoded Throughout Our Bodies: Molecular and
Cellular Basis of Tissue Regeneration. Pediatric Research 63:5, 502-512. [Crossref]
328. Pankaj Godara, Robert E Nordon, Clive D McFarland. 2008. Mesenchymal stem cells in tissue engineering. Journal of Chemical
Technology & Biotechnology 83:4, 397-407. [Crossref]
329. Despoina M. Choumerianou, Helen Dimitriou, Maria Kalmanti. 2008. Stem Cells: Promises Versus Limitations. Tissue
Engineering Part B: Reviews 14:1, 53-60. [Abstract] [Full Text PDF] [Full Text PDF with Links]
330. Z. T. Bloomgarden. 2008. The Diabetic Foot. Diabetes Care 31:2, 372-376. [Crossref]
331. Despoina M. Choumerianou, Helen Dimitriou, Maria Kalmanti. 2008. Stem Cells: Promises Versus Limitations. Tissue
Engineering 105, 110306233438005. [Crossref]
332. Courtney Quinn, Alan W. Flake. 2008. In vivo Differentiation Potential of Mesenchymal Stem Cells: Prenatal and Postnatal
Model Systems. Transfusion Medicine and Hemotherapy 35:3, 239-247. [Crossref]
333. Pankaj Godara, Robert E Nordon, Clive D McFarland. 2008. Mesenchymal stem cells in tissue engineering. Journal of Chemical
Technology & Biotechnology 83:4, 397. [Crossref]
334. Markus Thomas Rojewski, Barbara Maria Weber, Hubert Schrezenmeier. 2008. Phenotypic Characterization of Mesenchymal
Stem Cells from Various Tissues. Transfusion Medicine and Hemotherapy 35:3, 168-184. [Crossref]
335. B. Tawil, H. Duong, B. Wu. Fibrin matrices in tissue engineering 533-548. [Crossref]
336. B.J. Herdrich, R.C. Lind, K.W. Liechty. 2008. Multipotent adult progenitor cells: their role in wound healing and the treatment
of dermal wounds. Cytotherapy 10:6, 543-550. [Crossref]
337. Li Yan, Ying Han, Yuanlong He, Huahong Xie, Jingmei Liu, Lina Zhao, Jingbo Wang, Liuchun Gao, Daiming Fan. 2007. Cell
Tracing Techniques in Stem Cell Transplantation. Stem Cell Reviews 3:4, 265-269. [Crossref]
338. Jaymie Panuncialman, Vincent Falanga. 2007. The Science of Wound Bed Preparation. Clinics in Plastic Surgery 34:4, 621-632.
[Crossref]
339. Li Yan, Ying Han, Yuanlong He, Huahong Xie, Jingmei Liu, Lina Zhao, Jingbo Wang, Liuchun Gao, Daiming Fan. 2007. Cell
Tracing Techniques in Stem Cell Transplantation. Stem Cell Reviews 3:4, 265. [Crossref]
Downloaded by 36.79.6.204 from online.liebertpub.com at 09/21/17. For personal use only.

Você também pode gostar