Você está na página 1de 13

Cell Transplantation, Vol. 24, pp. 865–877, 2015 0963-6897/15 $90.00 + .

00
Printed in the USA. All rights reserved. DOI: http://dx.doi.org/10.3727/096368913X676231
Copyright © 2015 Cognizant Comm. Corp. E-ISSN 1555-3892
www.cognizantcommunication.com

The Human Umbilical Cord Tissue-Derived MSC Population UCX® Promotes


Early Motogenic Effects on Keratinocytes and Fibroblasts and G-CSF-Mediated
Mobilization of BM-MSCs When Transplanted In Vivo
Joana P. Miranda,*1 Elysse Filipe,*1 Ana Sofia Fernandes,*† Joana M. Almeida,‡ José P. Martins,‡
Alexandre De la Fuente,§ Miguel Abal,§ Rita N. Barcia,‡ Pedro Cruz,‡ Helder Cruz,‡
Matilde Castro,* and Jorge M. Santos‡

*Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
†Centro de Biociências da Universidade Lusófona (CBios), Lisbon, Portugal
‡ECBio S.A., Amadora, Portugal
§Translational Medical Oncology, Health Research Institute of Santiago (IDIS),
Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Spain

Mesenchymal stromal cells (MSCs) play an important role in tissue regeneration mainly through the secretion of
trophic factors that enhance the repair of damaged tissues. The main goal of this work was to study the paracrine
mechanisms by which an umbilical cord tissue-derived MSC population (UCX®) promotes the migration capacity
of human dermal fibroblasts and keratinocytes, which is highly relevant for skin regeneration. Furthermore, the
differences between paracrine activities of MSCs from the umbilical cord tissue and the bone marrow (BM-MSCs)
were also evaluated. In vitro scratch assays revealed that conditioned media (CM) obtained from both growing
and stationary-phase UCX® cultures induced human dermal fibroblast (HDF) and keratinocyte (HaCaT) migra-
tion. These assays showed that the motogenic activity of UCX® CM to HaCaTs was significantly higher than to
HDFs, in opposition to the effect seen with CM produced by BM-MSCs that preferentially induced HDF migra-
tion. Accordingly, a comparative quantification of key factors with vital importance in the consecutive stages of
wound healing revealed very different secretome profiles between UCX® and BM-MSCs. The relatively higher
UCX® expression of EGF, FGF-2, and KGF strongly supports early induction of keratinocyte migration and
function, whereas the UCX®-specific expression of G-CSF suggested additional roles in mobilization of healing-
related cells including CD34−/CD45− precursors (MSCs) known to be involved in tissue regeneration. Accord-
ingly, in vitro chemotaxis assays and an in vivo transplantation model for chemoattraction confirmed that UCX®
are chemotactic to CD34−/CD45− BM-MSCs via a cell-specific mobilization mechanism mediated by G-CSF.
Overall, the results strongly suggest different paracrine activities between MSCs derived from different tissue
sources, revealing the potential of UCX® to extend the regenerative capacity of the organism by complementing
the role of endogenous BM-MSCs.

Key words: Mesenchymal stromal cells (MSCs); UCX®; Stem cell transplantation; Fibroblast and keratinocyte
migration; BM-MSC mobilization; G-CSF; Skin regeneration

INTRODUCTION complications and increased morbidity. Hence, a number of


The healing of cutaneous wounds, like any other type of novel strategies have been attempted, such as the local
injury, represents a complex process, integrating a number application of growth factors and stem cells, to improve the
of sequential molecular events involving clotting, inflam- regeneration process (6,35).
mation, cellular migration, proliferation, extracellular matrix Mesenchymal stromal cells (MSCs) are a subset of stem
(ECM) deposition, angiogenesis, vasculogenesis, and cells that can be isolated from a number of adult and neona-
remodeling of the mature scar tissue (29,36,50,58). In a tal tissues (13,16,56). MSCs have been shown to possess
number of medical conditions, such as diabetes, chronic modulatory properties of both immune and inflammatory
renal failure, or arterial or venous insufficiency, the success reactions, although the specific mechanism by which MSCs
of this process is jeopardized, resulting in numerous display such behaviors is still under investigation (18,26,57).

Received July 19, 2013; final acceptance December 17, 2013. Online prepub date: December 30, 2013.
1
These authors provided equal contribution to this work.
Address correspondence to Jorge Miguel Santos, ECBio S.A., Rua Henrique Paiva Couceiro, Nº 27, 2700-451 Amadora, Portugal. Tel: +351 214997579;
Fax: +351 214997594; E-mail: miguel.santos@ecbio.com
865
866 MIRANDA ET AL.

In situ engraftment of MSCs to sites of tissue damage the UCX® capacity for modulating the inflammatory
after systemic infusion or localized transplantation has reaction and for suppressing the immune system was
been demonstrated, yet increasing evidence also shows that shown to be higher than that of BM-MSCs (46). These
MSCs have the ability to induce tissue regeneration with- observations have led us to propose that MSCs from
out local engraftment and differentiation. Indeed, several different tissue sources should not be treated as equal in
authors have hypothesized that the MSC-secreted factors terms of their regenerative properties and should there-
enhance and assist in the repair and regeneration of dam- fore be discussed and evaluated as separate therapeutic
aged tissues via paracrine activation of surrounding cells entities (46).
(7,8,12,24,38). Secreted bioactive factors may induce cell In this work, the motogenic effect of UCX® to human
migration, proliferation, and even differentiation, resulting dermal fibroblasts (HDFs) and human keratinocytes
in, for example, angiogenesis and vasculogenesis (6,38,44). (HaCaTs) was demonstrated and shown to be very differ-
The paracrine secretion of trophic factors has the potential ent from that caused by BM-MSCs, further supporting
to induce profound effects on the MSC local environment, a distinct, but nevertheless complementary, regenerative
as suggested by the cytokine-mediated control of hemato- role for UCX® (55). Accordingly, a comparative quantifi-
poiesis, which is thought to be regulated by MSCs in the cation of a selection of paracrine factors with vital impor-
vascular niche of the bone marrow (8,44,59). The paracrine tance during the consecutive phases of wound healing
action of MSCs has been implicated in cutaneous wound revealed very different secretome profiles between the
healing, where it is suggested that these cells secrete a two MSC types. While the UCX® trophic factor profile
number of promitogenic and motogenic growth factors suggested a direct impact on the earlier homeostasis and
and cytokines, thus leading to enhanced tissue regenera- inflammation stages of wound healing, the BM-MSC tro-
tion (10,59,60). phic profile was better suited to promote later prolifera-
Most of the data so far have been generated with the tive and final remodeling activities. Additionally, the
prospective autologous application, using bone marrow- UCX®-specific expression of granulocyte-colony stimulat-
derived MSCs (BM-MSCs) (9,60). BM-MSCs are known ing factor (G-CSF) was shown to attribute UCX® with a
to be endogenously mobilized and to coexist within sites chemotactic recruitment activity to other circulating or
of injury, actively participating in the healing process resident BM-MSCs. Such capacity was confirmed in vivo
(9,10,60). However, increasing evidence has been sup- through transplantation of UCX® and BM-MSC-seeded
porting neonatal tissues, such as the umbilical cord tissue scaffolds in the inner wall of the peritoneal cavity of
and placenta, as better sources of MSCs due to several severe combined immunodeficient (SCID) mice and by
reasons. Neonatal tissues confer ease of isolation from monitoring interscaffold-specific MSC migration.
noninvasive and noncontroversial human sources. Further- Overall, our results are consistent with different regen-
more, a relatively higher cell yield is obtained, and the erative activities between MSCs from different tissue
resulting MSCs have higher expansion capacity and often sources and confirm the potential of UCX® as an active
higher regenerative potency (2,28,60,61). UCX® in partic- substance for developing ATMPs.
ular are MSCs obtained from the umbilical cord stromal
tissue using a proprietary method that has recently been MATERIALS AND METHODS
adapted according to advanced therapy medicinal product Ethical and Regulatory Aspects
(ATMP) certification requirements [EMEA/CAT/486831/ This study was approved by the Ethics Committee of
2008/corr (17)] (36a). UCX® comply with the MSC defi- the Cascais Hospital Dr. José de Almeida (Portugal), in the
nition provided by the International Society for Cellular scope of a research protocol with ECBio, S.A. (Amadora,
Therapy (ISCT) (15). Namely, UCX® present fibroblast- Portugal). Umbilical cord procurement involving donations,
like morphology while adherent to plastic; more than with written informed consent, was made according to
98% of UCX® within the population express the markers Directive 2004/23/EC. All the experimental procedures
cluster of differentiation 105 (CD105), CD73, CD90, and involving animal models were carried out with the per-
CD44, whereas less than 2% express CD45, CD34, mission of the local laboratory animal committees and in
CD31, CD14, CD19, and human leukocyte antigen accordance with internationally accepted guidelines.
(HLA)-DR. Finally, UCX® can be differentiated into oste-
oblast, chondrocyte, and adipocyte-like cells (46). UCX® UCX® Isolation and Culture
have proven to be nonimmunogenic in vitro and in vivo and UCX® used in this work were derived from a total of
to promote angiogenesis in the scope of an acute myo- seven umbilical cords (three females and four males) from
cardial infarction model (38a). Furthermore, UCX® have healthy donors, isolated according to Santos et al. (47) and,
recently been demonstrated to have the capacity of when necessary, thawed from a low passage (P3–P5)
repressing T-cell activation and promoting the expansion working cell bank (WCB). In brief, UCX® were isolated
of regulatory T-cells (Tregs) in vitro. Most importantly, from human umbilical cord tissue sections that were
UCX® REGENERATIVE ACTIVITY 867

digested using a precise ratio between tissue mass, tissue phosphate. The medium was replaced every 3 days dur-
digestion enzyme activity units, digestion solution vol- ing 21 days. To induce osteogenic differentiation (n = 4),
ume, and void volume using collagenase. Cells dissoci- cells were incubated in a-MEM supplemented with 10%
ated from the tissue were recovered by adherence to the FBS, 2 mM glutamine, 10 mM b-glycerol phosphate, 50 µg/
surface of a culture flask (Nunc, Schnelldorf, Germany) ml ascorbate-2-phosphate, and 100 nM dexamethasone.
during a static horizontal incubation period and cultured The medium was replaced every 3 days during 21 days.
with a-modified minimum essential medium (a-MEM; For cytochemical staining, cells were fixed with 4% (w/v)
Sigma-Aldrich, Madrid, Spain) supplemented with 20% paraformaldehyde (Sigma-Aldrich) for 20 min. In adipo-
fetal bovine serum (FBS; Gibco, Life Technologies, Madrid, genic and osteogenic differentiation protocols, cells were
Spain), herein designated as MSC complete medium, at stained with oil red O (Sigma-Aldrich) and alkaline phos-
37°C humidified atmosphere with 7% CO2 in air and cryo- phatase (Sigma-Aldrich), respectively. For chondrogenic
preserved (47). The WCB was generated by expanding differentiation (n = 3), spheroids were cut into sections
UCX® in standard MSC culture conditions (46). Cells were and stained with alcian blue (Sigma-Aldrich). The pre-
characterized by their capacity to adhere to plastic in stan- sence of stained cells was confirmed by use of an inverted
dard MSC culture conditions, specific surface marker microscope with phase contrast (DMIL HC; Leica,
expression, and trilineage differentiation capacity (46). All Wetzlar, Germany).
cell culture media and supplements were purchased from
Sigma-Aldrich, unless otherwise indicated. Production of UCX® and BM-MSC Conditioned
Media (CM)
Cell Characterization by Flow Cytometry For conditioned media (CM) production, UCX® and
For surface marker expression analyses (n = 7), cells BM-MSCs (Cat. P10576, batch 2095; Innoprot, Bizkaia,
were incubated for 1 h at 4°C with the antibodies in 2% Spain) were first sequentially adapted to low serum con-
(w/v) bovine serum albumin (Sigma-Aldrich) solution and centrations by a reduction of the amount of FBS in the
analyzed by flow cytometry using a Gallios imaging flow cell culture medium from 20% to 2% (Fig. 1a). When at
cytometer (Beckman Coulter, Inc., Pasadena, CA, USA). 2% FBS, cultures were maintained until 80% confluence
The antibodies used (anti-human) and their conjugates after which (i) either UCX® or BM-MSCs were trypsinized
were CD105-phycoerythrin (PE), CD73-allophycocyanine (Gibco, Life Technologies, Madrid, Spain) and seeded at
(APC), CD90-PE, CD44-APC, CD14-Peridinin-chlorophyll a 1 × 104 cells/cm2 density in MSC medium supplemented
protein/cyanine 5.5 (PerCp/Cy5.5), CD45-PerCp/Cy5.5, with 2% FBS and cultured for a conditioning period of 7
CD34-fluorescein isothiocyanate (FITC), CD31-FITC, and 4 days, respectively (growing UCX®-CM and grow-
CD19-Pacific blue, and HLA-DR-Pacific blue. The isotype ing BM-MSC-CM), or (ii) UCX® medium was simply
controls used were Pacific blue IgG1, Pacific blue IgG2a, replaced with MSC medium supplemented with 2% FBS
IgG1k PerCp/Cy5.5, IgG2a PerCp/Cy5.5, IgG1k PE, and maintained in culture for a conditioning period of
IgG1k APC, and IgG1k FITC. The antibodies’ final dilu- 3 days (confluent UCX®-CM). The control sample con-
tion was 1:100, with the exception of the antibodies con- sisted of MSC complete medium containing 2% FBS that
jugated with PerCp/Cy5.5, where a dilution of 1:400 was never in contact with cells. CM and control were con-
was used. All antibodies were purchased from Biolegend centrated with 5 kDa cutoff spin concentrators (Agilent
(San Diego, CA, USA), except for CD105-PE, which was Technologies, Santa Clara, CA, USA) as per manufactur-
acquired from eBioscience (San Diego, CA, USA). er’s recommendations. CM used for comparative studies
based on scratch assays were normalized for total protein
Cell Characterization by Trilineage Differentiation concentration. Protein content was quantified using the
To induce adipogenic differentiation (n = 7), cultured bicinchoninic acid protein assay kit (Thermo Scientific,
cells were incubated in MSC complete medium, supple- Waltham, MA, USA) according to the manufactur-
mented with 10 µg/ml insulin, 200 µM indomethacin, er’s instructions.
0.5 mM isobutylmetylxantine, and 1 µM dexamethasone,
for 3 days, and subsequently 1 day in adipogenic main- In Vitro Scratch Assays
tenance medium, consisting of MSC complete medium sup- The in vitro scratch assays were performed on primary
plemented with 10 µg/ml insulin. Medium replacement HDFs (from a 38-year-old Caucasian female, Cat. DF-F;
cycles were repeated during 21 days. For chondrogenic Zen-Bio, Research Triangle Park, NC, USA) and on
differentiation, cells were grown in suspension as pellets, HaCaTs (from a 62-year-old Caucasian male, Cat. 300-493;
incubated in Dulbecco’s modified Eagle’s medium (DMEM), CLS Cell Lines Service GmbH, Eppelheim, Germany)
1% FBS, 2 mM L-glutamine, 6.25 µg/ml insulin, 10 ng/ essentially as described by Liang et al. (33). Primary HDF
ml transforming growth factor-b (TGF-b1) (Tebu-bio, and HaCaT cells were seeded into 24-well plates (Nunc,
Le-Perray-en-Yvelines, France), and 50 µM ascorbate-2- Schnelldorf, Germany) at a density of 2.5 × 104 cells/cm2
868

Figure 1. Motogenic effect of UCX®-CM and BM-MSC-CM to HDF and HaCaT cells. (a) Scheme of CM production as described in Materials and Methods. (b) Images of the
scratch area on HDF and HaCaT cell cultures at time 0 h and 20 h or 40 h, respectively, after CM treatment. (c) Graphical representation of the integrated area migrated by
HDF and HaCaT cells after contact with different CMs. *p < 0.05; **p < 0.01. Scale bar: 500 µm. UCX®, umbilical cord tissue-derived mesenchymal stromal cell (MSC) popu-
lation; BM, bone marrow; FBS, fetal bovine serum.
MIRANDA ET AL.
UCX® REGENERATIVE ACTIVITY 869

and 4.0 × 104 cells/cm2, respectively, with DMEM-High and were allowed to migrate through the porous mem-
Glucose (DMEM basal medium) supplemented with 10% brane (8 mm) for 48 h in response to the following stimuli
FBS, herein designated as DMEM complete medium. from the lower compartment: (i) serum-free media (nega-
Once cultures reached 80% confluence, culture medium tive control), (ii) serum-free media supplemented with the
was replaced with DMEM basal medium without FBS for universal chemoattractant EGF at 10 ng/ml (positive con-
24 h to guarantee the inhibition of cell proliferation, while trol), (iii) 200,000 nonlabeled BM-MSCs in serum-free
enabling cell migration. Scratches of approximately 0.5 mm media seeded on the bottom of the well (control for self-
in width were then performed on the monolayer with a mediated chemotaxis), (iv) 200,000 nonlabeled UCX® in
sterile P200 pipette tip (Orange, Braine-L’Alleud, Belgium). serum-free media seeded on the bottom of the well, and
After performing the scratch, the culture medium was (v) 200,000 UCX® in serum-free media supplemented with
replaced with 200 µl of DMEM basal medium supple- anti-G-CSF as an inhibitor (hG-CSF, Anti-G-CSF Human,
mented with the different groups of CM and control 500-P43-B; Tebu-Bio). Upon incubation, migratory BM-
diluted to a final concentration of 500 µg/ml total protein. MSCs were removed from the bottom of the membrane
Additional controls consisting of CM generated from using trypsin and quantified using a FLUOstar OPTIMA
HDF or HaCaT cultures were also performed in order to fluorimeter (BMG Labtech, Ortengerg, Germany) oper-
monitor for self-chemotactic effects (data not shown). The ated at 549 nm excitation/565 nm emission. Results from
area of the scratch was measured at 0 h for both cell three independent experiments are represented as ratios
types and at 20 h and 40 h postscratch for HDF and compared to negative controls.
HaCaT cells, respectively. UCX®-CMs were derived from
a total of n = 3 umbilical cord samples, and a total of In Vivo Scaffold-Based Transplantation Model
n = 6 independent experiments was performed. Photographs for Chemoattraction
were taken at an amplification of 40× on a Nikon Eclipse Either BM-MSCs or UCX® labeled with 1 µM DiD
Ti-U inverted microscope with a conjugated Nikon DS- was suspended in culture medium at an appropriate con-
Qi1Mc camera (Nikon, Tokyo, Japan). Cellular migra- centration (50,000 cells/20 ml) and seeded into 3D Insert-
tion was analyzed in the NIS-Elements BR 3.10 program polystyrene/polycaprolactone (PS/PCL) with nanomesh
by integrating the final cell-free area within the scratch, scaffolds (3D-Biotek, Hillsborough, NJ, USA) 24 h before
calculated in relation to the total initial area of the scratch implantation in the inner wall of the peritoneal cavity of
at 0 h. 8- to 9-week SCID mice (female SCID beige mice; Charles
River, Barcelona, Spain). For this, mice were anesthetized
Trophic Factor Quantification with Ketolar® (75 mg/kg; Parke-Davis, Pfizer, Madrid,
A commercially available kit (FlowCytomix™; Spain) and metomidine (1 mg/kg; Orion Pharma, Barcelona,
eBioscience) was used to determine the concentrations of Spain) and an incision performed in the peritoneum to intro-
epidermal growth factor (EGF), fibroblast growth factor-2 duce and fix the scaffolds with BM-MSCs and UCX® in
(FGF-2), G-CSF, hepatocyte growth factor (HGF), inter- the left upper part and lower part of the peritoneal cavity
leukin-6 (IL-6), TGF-b1, and vascular endothelial growth at 1 cm of distance approximately, with the help of surgi-
factor-A (VEGF-A) in CM from growing UCX®, grow- cal glue (Vetbond®; 3M, St. Paul, MN, USA). Atipamezol
ing BM-MSCs, and control samples. Samples were (1 mg/kg; Orion Pharma), a metomidine antagonist, was
acquired on a Gallios imaging flow cytometer, and the supplied after operation. One week after implantation, mice
results were obtained using FlowCytomix Pro 3.0 Soft- were sacrificed, and MSC migration was evaluated using an
ware. Quantification of keratinocyte growth factor (KGF) in vivo imaging system (IVIS; Perkin-Elmer, Santa Clara,
in these samples was performed using an enzyme-linked CA, USA) operated at 644 nm excitation/665 nm emis-
immunosorbent assay kit (Quantikine; R&D Systems, sion. Figures are representative images of two independent
Minneapolis, MN, USA). The quantification of all growth experiments using a minimum of four animals per group.
factors was obtained from a total of n = 2 umbilical cord
samples (300 events acquired/growth factor) and was Statistical Analysis
expressed as pg/106 cells/day and normalized against the All statistical analyses were done with SPSS 21.0
control background. (Chicago, IL, USA). Results are presented as mean and with
standard errors of the mean (SEM) where possible. To esti-
In Vitro Chemotaxis Assay mate the significance of the differences between groups, the
BM-MSCs were labeled with octadecyl indocarbo- parametric Student’s t-test was used. As a result of multi-
cyanine (DiD, 1 µM; Life Technologies, Madrid, Spain) ple subgroup assessments, a Bonferroni correction was
for 30 min at 37°C. Fifty thousand labeled BM-MSCs used to adjust the p value of the in vitro scratch assays
were seeded in serum-free media in the upper compartment (a/4) and in vitro chemotaxis assays (a/3). Differences
of a Boyden chamber (Corning, Amsterdam, Netherlands) were considered to be significant for values of p < 0.05.
870 MIRANDA ET AL.

RESULTS UCX® and BM-MSCs to HDFs and HaCaTs, respec-


In this work, UCX® were freshly isolated from a total tively, a number of essential trophic factors known to be
of seven umbilical cord samples, and when necessary for representative of the several phases of wound healing
experimentation, cells were thawed from a low passage were quantified. Thus, the concentrations of HGF, G-
WCB (P3–P5). UCX® and BM-MSCs were expanded in CSF, TGF-b1, KGF, EGF, FGF-2, VEGF-A, and IL-6
all cases and consistently presented fibroblast-like mor- were determined in growing UCX®-CM, growing BM-
phology while adherent to plastic; more than 98% of the MSC-CM, and CM control samples (Fig. 2a). The results
cells within the corresponding populations consistently clearly show different secretome profiles between UCX®
expressed CD105, CD73, CD90, and CD44, whereas less and BM-MSCs under our growth conditions (Fig. 2a). In
than 2% expressed CD45, CD34, CD31, CD14, CD19, turn, a pie chart representation of trophic factor expression
and HLA-DR. Finally, both types of MSCs could consis- distribution between growing UCX®-CM and growing
tently be differentiated into osteoblast, chondrocyte, and BM-MSC-CM revealed that i) factors secreted preferen-
adipocyte-like cells following the MSC criteria as defined tially by UCX® (such as G-CSF, EGF, FGF-2, and KGF)
by the ISCT (15) (results not shown). have been commonly implicated in earlier stages of
wound healing, namely, homeostasis and inflammation
In Vitro Motogenic Effects of UCX® and BM-MSC to (1,5,27,30,40–42,48,53,62), while ii) factors secreted pref-
HDFs and HaCaTs erentially by BM-MSCs (such as TGF-b1, IL-6, and
VEGF-A) have been more closely associated with the later
In order to evaluate the motogenic potential of UCX® to
proliferation and remodeling phases of wound resolution
HDFs and HaCaTs, CM was derived from UCX® and its
(4,19,20,22,32,34,39) (Fig. 2b). Moreover, given what is
activity compared to that derived from BM-MSCs (Fig. 1a).
known about G-CSF-mediated mobilization of circulating
Both CMs were supplied to HDF and HaCaT cell cultures
BM-MSCs to sites of injury (14,21,43,45,54), a signifi-
and specific cell migration evaluated using in vitro scratch
cant UCX®-specific expression of G-CSF led us to pro-
assays. Prior to CM production, both types of MSCs were
pose a mechanism where UCX® could further promote
progressively adapted to growth conditions containing
chemotactic recruitment of BM-MSCs (Fig. 2b).
sequentially lower FBS concentrations in order to reduce the
amount of serum components in the CM that could poten- In Vitro Evaluation of G-CSF-Mediated UCX®
tially mask their trophic activity (Fig. 1a). CM was finally Chemotactic Mobilization of BM-MSCs
produced by cultures growing in 2% FBS, the lowest con-
The hypothesis that UCX® could promote chemotactic
centration that still allowed for MSC growth and expansion.
mobilization of BM-MSCs was first evaluated in vitro.
Also, in an attempt to elucidate for the importance of meta-
For this, DiD-labeled BM-MSCs were placed in the upper
bolic rate in the quality of the secreted trophic factors, CM
compartment of Boyden chambers and allowed to migrate
were produced by UCX® cultures in exponential phase (grow-
toward the lower compartment in response to different
ing UCX®-CM) and stationary phase (confluent UCX®-
chemotactic stimuli. As shown in Figure 3, BM-MSCs
CM). Both HDF and HaCaT cells in contact with either
migrate in the presence of EGF (universal chemoattract-
growing UCX®-CM or confluent UCX®-CM exhibited a
ant positive control) or BM-MSCs (self-mediated chemo-
significantly increased migration capacity, with a slightly
taxis positive control). In turn, UCX® seeded on the
more prominent effect being observed for CM produced by
bottom of the well were clearly chemotactic to BM-MSCs,
more metabolically active, exponentially growing UCX®
resulting in a 1.7-fold migration increase when compared
(Fig. 1b, c). Interestingly, the motogenic effect of growing
to self-mediated chemotaxis. Moreover, when the chemo-
UCX®-CM was considerably higher with keratinocytes
tactic assay was performed in the presence of hG-CSF
(more than threefold higher than control) than fibroblasts
neutralizing antibodies (a–G-CSF), the UCX®-mediated
(1.8-fold higher than control), contrarily to the cell migra-
recruitment was significantly hindered (Fig. 3). These results
tion promoted by CM produced by exponential phase BM-
indicate that (i) UCX® are chemotactic to BM-MSCs, and
MSC cultures (growing BM-MSC-CM) that showed a
(ii) cell-specific expression of G-CSF by UCX® is an
clear selectivity for fibroblasts (twofold higher than control)
important factor determining the paracrine chemotactic
(Fig. 1c). The results demonstrated that the two different
activity to BM-MSCs in vitro.
types of MSCs have diverse paracrine activities, resulting in
differential motogenic effects to HDFs and HaCaTs. In Vivo Evaluation of UCX® Potential for Chemotactic
Mobilization of BM-MSCs
Trophic Factor Quantification in UCX® and BM-MSC In order to confirm whether the UCX® chemotactic
Conditioned Media effect to BM-MSCs was kept intact in vivo, two 3D-
In order to establish a correlation between the differen- Biotek Insert-PS/PCL scaffolds were transplanted into the
tial motogenic effects caused by the paracrine activity of peritoneum of SCID mice, one seeded with BM-MSCs and
UCX® REGENERATIVE ACTIVITY

Figure 2. Trophic factor quantification in growing UCX®-CM and growing BM-MSC-CM. (a) Quantification of HGF, G-CSF, TGF-b1, KGF, EGF, FGF-2, VEGF-A, and IL-6
in growing UCX®-CM and growing BM-MSC-CM normalized against control background and expressed in pg of growth factor/cell number in culture/number of conditioning
days (pg/106 cells/day). *p < 0.05; **p < 0.01. (b) Pie chart representation of the relative trophic factor concentrations in growing UCX ®-CM and growing BM-MSC-CM revealed
that factors secreted preferentially by UCX® have been commonly implicated in earlier stages of wound healing, while factors secreted preferentially by BM-MSCs have been
more closely associated with the later phases of wound resolution. A proposed mechanism where UCX ® can further promote a G-CSF-mediated chemotactic recruitment of BM-
MSCs is represented by the arrow line on top.
871
872 MIRANDA ET AL.

Figure 3. G-CSF-mediated chemotactic activity of UCX® cells to BM-MSCs in vitro. Graphical representation of the relative BM-
MSC migration through the Boyden chamber pores (8 mm) and schematic representation of experimental setup. UCX® seeded on
the bottom of the well were chemotactic to BM-MSCs. Moreover, when the chemotactic assay was performed in the presence of
a-G-CSF neutralizing antibodies, the UCX®-mediated recruitment was significantly hindered. Relative migration was determined with
respect to the negative control. *p < 0.05.

the other with UCX®. MSC-specific chemotactic mobiliza- from the bottom scaffold at day 7, discarding any migra-
tion was assessed by coupling either fluorescently labeled tion due to chemotactic inflammatory signals caused by
UCX® or BM-MSCs with their nonlabeled counterparts the scaffold implantation procedure alone (Fig. 4E, F).
(Fig. 4). The left panel on Figure 4 shows fluorescence The signal detected around the incision scar in experi-
emission by MSCs at time 0, right after scaffold implan- ments 1 and 2 was most likely due to nonspecific MSC
tation. In turn, the center panels show the location of both homing to the inflammation site caused by the incision
scaffolds in the internal wall of the peritoneum 1 week past (Fig. 4B, D, white arrows). Overall, our observations clearly
cell transplantation, at sacrifice day 7. Panels on the right demonstrate the existence of a UCX®-specific chemo-
show the corresponding fluorescent image at day 7, and the tactic activity to BM-MSCs in vivo.
smaller insets, on the bottom right-hand corner of these
panels, are fluorescent images of the corresponding scaffolds DISCUSSION
ex vivo. The experiment with DiD-labeled BM-MSCs can In a still widely accepted classical model of wound
be followed in the upper panels of Figure 4 (A, B). DiD- healing, four consecutive and overlapping phases can be
labeled BM-MSCs seeded in the upper scaffold migrated considered: homeostasis, inflammation, proliferation, and
toward the lower scaffold containing unlabeled UCX® as remodeling, although many authors congregate homeosta-
demonstrated by a strong fluorescent signal emanating from sis and inflammation in a single inflammatory stage (37).
the bottom scaffold, as well from a migration path con- Figure 2b depicts the wound healing process attempting to
necting both scaffolds (Fig. 4A, B). In turn, when labeled divide the four healing phases within a temporal sequence
UCX® were located in the upper compartment, a mobili- of events. Nevertheless, in vivo, there are overlapping
zation of much lower significance was detected toward the sequences occurring simultaneously within the wound site.
lower scaffold seeded with unlabeled BM-MSCs (Fig. 4C, Accumulated evidence in this field has drawn up an
D). The column histogram in Figure 4 shows the significant extensive list of growth factors and cytokines secreted by
difference between the quantified fluorescence intensity, MSCs that are involved in the various processes leading
in arbitrary units, in the UCX® scaffold, resulting from to tissue regeneration (4,20,32,58). Within the cutaneous
the mobilization of DiD-labeled BM-MSCs (Fig. 4B), wound milieu, inflammation, cellular migration, prolifera-
compared to fluorescence intensity in the BM-MSC scaf- tion, differentiation, ECM deposition, angiogenesis, vascu-
fold, resulting from a much lower mobilization of DiD- logenesis, maturation, and remodeling are all necessary
labeled UCX® (p < 0.001) (Fig. 4D). Lastly, in a control responses where MSC-secreted trophic factors may play a
experiment, DiD-labeled BM-MSCs were seeded in the role (9,10,24,25,38,49–51,55,59).
upper scaffold and coupled with an empty scaffold on In line with the previously demonstrated differential capa-
the bottom. The result was that no signal was detected city between UCX® and BM-MSCs for immune suppression
UCX® REGENERATIVE ACTIVITY

Figure 4. Chemotactic activity of UCX® cells to BM-MSCs in vivo using a SCID mouse intraperitoneal, two-scaffold, implant model. (Left panels): Photos of mice right after
small-incision scaffold implantation, at day 0. (Right panels) Postmortem images of mice at day 7. (A, C, E) Photos showing scaffold location. (B, D, F) Images of mice with
superimposed in vivo imaging system (IVIS) detection, representing the density (color gradient) of octadecyl indocarbocyanine (DiD)-labeled cells. The bottom right-hand panels
are ex vivo IVIS images of scaffolds, after excision from animals. The column histogram consists of quantified fluorescence intensity, in arbitrary units, in the UCX ® scaffold,
resulting from the mobilization of DiD-labeled BM-MSCs (B), compared to fluorescence intensity in the BM-MSC scaffold, resulting from a much lower mobilization of DiD-
labeled UCX® (D) (p < 0.001). Data were obtained from two independent experiments using a minimum of four animals per group; ***p < 0.001. SCID, severe com-
bined immunodeficient.
873
874 MIRANDA ET AL.

(46), we herein report a clear, distinct, but nonetheless com- Similarly, VEGF-A-inducible matrix metalloproteinase 1
plementary effect of their paracrine motogenic activities to (MMP-1), MMP-2, and tissue inhibitor of metalloproteinase
HDFs and HaCaTs. In agreement with what had been 1, all peaking at 2 to 5 days after excisional wounding (22),
previously reported for BM-MSCs (55), we demonstrate support a role of VEGF-A during fibroblast proliferation and
a preferential trophism of BM-MSCs to HDFs when migration, ECM synthesis leading to granulation tissue for-
compared to HaCaTs. Conversely, our results showed mation, angiogenesis, and vasculogenesis (20). In accordance
that the effect of UCX® on inducing HaCaT migration with our results, VEGF-A has been targeted primarily at
was significantly higher when compared to the corre- fibroblasts (and macrophages) in deeper wounds (39), thus
sponding effect on HDFs. Given the relatively higher correlating the BM-MSC-CM fibroblast-specific motogenic
importance of keratinocyte migration in the early stages of activity seen by us with its VEGF-A contents. Like VEGF-
wound healing, we hypothesized that the paracrine healing A, IL-6 was also mainly expressed by BM-MSCs in our
potential of UCX® could therefore be more relevant dur- assays. Pursuant to what is known about VEGF-A, IL-6 has
ing the initial stages leading to skin regeneration, where also been shown to be crucial to epithelialization and to
early keratinocyte migration plays a major role (9,55). influence granulation tissue formation during the later stages
Accordingly, UCX® and BM-MSCs presented clearly of tissue regeneration (34).
distinct secretome profiles. Furthermore, within our spe- In summary, the functional analysis of the factors
cific pool, those factors either exclusively or majorly included in our test pool does substantiate our hypothesis
expressed by UCX® could in fact be directly correlated to that, contrasting to BM-MSCs, the direct impact of
events occurring during the earlier phases of the healing UCX® paracrine activity could be centered on the earlier
process. The relatively secluded expression of EGF, FGF-2, phases of the regenerative process. Nevertheless, equiva-
and KGF by UCX®, contrasting with the relatively clois- lent expression of other important factors, such as HGF
tered expression of VEGF-A (and to some extent IL-6) and TGF-b1, further suggest overlapping roles between
by BM-MSCs, could alone explain the differential effects UCX® and BM-MSCs. HGF is a cytokine known to play
observed on HDF and HaCaT migration. EGF, for exam- multiple roles during the various stages of wound healing,
ple, has been shown not only to stimulate HaCaT migration, from neutrophil, monocyte, and mast cell recruitment to
but also to induce the autocrine secretion of all members the secretion of matrix metalloproteinases, ECM proteins,
of the EGF family. Some of these EGF family members integrins, and collagenases necessary for keratinocyte
possess even higher promitogenic and motogenic-inducing migration across the fibronectin-rich provisional wound
capacities upon HaCaTs so important for hemostasis. matrix (4,19,20,23,32,58). In turn, TGF-b1 is highly
In addition, increasing evidence suggests that the EGF expressed by keratinocytes and has been shown, along
receptor pathway has a major impact on the early inflam- with elements from the VEGF family, to be a key factor
matory/immune reactions necessary for proper skin regen- for the formation of granulation tissue due to its pro-
eration early after physical injury (31,40,48,53). In turn, mitogenic effect upon dermal fibroblasts, while also
FGF-2 is essential for the organization and morphogenesis inducing the expression of ECM proteins and matrix
of suprabasal HaCaTs acting through a chemotactic mech- deposition by these same cells (32). Furthermore, TGF-b1
anism (5,27,42). Additionally, FGF-2 has been shown to aids in the angiogenic process and has been reported as
induce leukocyte recruitment on endothelium, implicating necessary for fibroblast differentiation into myofibroblasts
this growth factor on early inflammatory stages of the that are responsible for wound contraction (4,20,32).
healing process (62). Finally, although KGF has preferen- Finally, our secretome analysis has revealed a quite
tially been correlated with epithelialization events (1), its remarkable UCX®-specific expression of G-CSF in our
early action on wound healing has been recently shown growth conditions. In the context of tissue regeneration, it
in vivo in a mouse model (41). Experiments demonstrated has come to our understanding that circulating factors and
that KGF is essential to the proliferation of keratinocytes systemic-derived cells play an important role in numerous
and that the early activity of KGF on keratinocyte function aspects of homeostasis and repair. The capacity of G-CSF
may also play a role in the regulation of VEGF gene to mobilize platelets, infiltrating inflammatory cells and cir-
expression and consequently on angiogenesis during fol- culating hematopoietic stem cells has been well documented
lowing stages of the healing process (41). On the other and currently widely used for clinical transplantation (11).
hand, in our culture conditions, factors such as the domi- However, a putative circulating population of CD45−/
nant VEGF isoform VEGF-A (VEGF165) and IL-6 were CD34− nucleated cells has also been found in the periph-
secreted preferentially by BM-MSCs. VEGF-A, in partic- eral blood of human adults that could be mobilized to the
ular, has been shown to be expressed in a specific tempo- injured tissue through a mechanism involving G-CSF (14,21,
ral pattern in wounds, namely, in the later proliferation 43,45,54). Studies using parabiotic models have demon-
phase occurring 3 to 7 days postwound, when capillary strated the participation of circulating MSCs in wound
growth and differentiation are at a maximum (Fig. 2b) (3). repair (52). Autologous BM-MSCs multiply in the bone
UCX® REGENERATIVE ACTIVITY 875

Figure 5. UCX®-mediated therapeutic pathways. Representation of the different pathways by which UCX® can actively impact the
wound healing process. Gray arrows highlight G-CSF-mediated mechanisms, namely, early impact recruitment of platelets, immune infil-
trate cells, and CD34+ hematopoietic precursors, and later-impact recruitment of CD34−/CD45− BM-MSCs. ECM, extracellular matrix.

marrow and migrate to the injured sites immediately after important during the later phases of healing, would be
burn injuries, contributing to the inflammatory response, compensated by inducing the chemotactic mobilization of
autolysis, and neovascularization along with hematopoi- either resident or circulating BM-MSCs with potential to
etic and endothelial progenitor cells (6). More recently, promote the formation of granulation tissue, contraction
the relative contribution of grafted human MSCs and host by myofibroblasts, angiogenesis, vasculogenesis, and epi-
stem/progenitor cells in promoting wound healing has been thelialization (Fig. 5).
evaluated by using a novel asymmetric wound model in Overall, the results presented in this work validate the
normal and impaired healing diabetic (db/db) mice (49). motogenic potential of UCX® to dermal fibroblasts and
MSCs significantly improved healing in both normal and keratinocytes and consolidate distinct paracrine activities
impaired animals by inducing wingless-related mouse of MSCs from different tissue sources. The G-CSF-
mammary tumor virus integration site 3a (Wnt3a), VEGF, mediated BM-MSC recruiting capacity of UCX® extends
and platelet-derived growth factor receptor-a and increas- its potential to a full range of events leading to tissue
ing the number of preexisting host MSCs recruited to the regeneration and homeostasis.
wound bed. Improvement was seen in both the grafted ACKNOWLEDGMENTS: The authors thank Prof. Filomena
and nongrafted sides, suggesting a systemic response to Nunes, Director of the Hospital of Cascais Dr. José Almeida,
MSC engraftment. Most importantly, healing was enhanced for the umbilical cord sample collection. This work was cofunded
despite the rapid loss of grafted human MSCs, suggesting by FCT (EXPL/DTP-FTO/0308/2013, Pest-OE/SAU/UI4013/2011,
and Ciência2008 to J.P.M.), QREN 2008/1467 - TERACEL to
that mobilizing the host response was the major outcome ECBio, and ECBio’s own research funds. H. Cruz and P. Cruz
of grafting MSCs to tissue repair (49). are shareholders of ECBio; J. M. Santos, M. Filipe, M. Teixeira,
As demonstrated in vitro, a G-CSF-dependent pathway and R. N. Barcia are employees of ECBio; J. P. Miranda is the
is possibly implicated in the motogenic potential of spouse of J. M. Santos. No other competing financial interests exist.
UCX® in vivo. Such a discovery, together with distinct
motogenic activities to skin cells, could constitute impor- REFERENCES
tant distinctive factors between UCX® and BM-MSCs in 1. Andreadis, S. T.; Hamoen, K. E.; Yarmush, M. L.; Morgan,
J. R. Keratinocyte growth factor induces hyperproliferation
terms of their future applications. During the early phases and delays differentiation in a skin equivalent model sys-
of healing, the paracrine activity of UCX® can directly tem. FASEB J. 15(6):898–906; 2001.
benefit the process in two ways: (i) by mitogenic and 2. Baksh, D.; Yao, R.; Tuan, R. S. Comparison of proliferative
motogenic activity to keratinocytes and (ii) by promoting and multilineage differentiation potential of human mesen-
G-CSF-mediated direct recruitment of platelets, infiltrate chymal stem cells derived from umbilical cord and bone
marrow. Stem Cells 25(6):1384–1392; 2007.
cells, such as neutrophils, macrophages, mast cells, and 3. Bao, P.; Kodra, A.; Tomic-Canic, M.; Golinko, M. S.; Ehrlich,
CD34+ hematopoietic precursor cells (Fig. 5). In turn, the H. P.; Brem, H. The role of vascular endothelial growth factor
relatively lower trophism of UCX® to fibroblasts, so in wound healing. J. Surg. Res. 153(2):347–358; 2009.
876 MIRANDA ET AL.

4. Baum, C. L.; Arpey, C. J. Normal cutaneous wound participate in ischemic neovascularization. Plast. Reconstr.
healing: Clinical correlation with cellular and molecular Surg. 123(2 Suppl):45S–55S; 2009.
events. Dermatol. Surg. 31(6):674–686; 2005. 22. Howard, E. W.; Soo, C.; Hunter, S.; Zhang, X.; Shaw, W. W.;
5. Bikfalvi, A.; Klein, S.; Pintucci, G.; Rifkin, D. B. Biolo- Longaker, M. T. Differences in MMP and TIMP gene expres-
gical roles of fibroblast growth factor-2. Endocr. Rev. 18(1): sion during wound repair. Surg. Forum 48:688–691; 1997.
26–45; 1997. 23. Kawada, A.; Hiruma, M.; Noguchi, H.; Ishibashi, A.;
6. Butler, K. L.; Goverman, J.; Ma, H.; Fischman, A.; Yu, Y. M.; Motoyoshi, K.; Kawada, I. Granulocyte and macrophage
Bilodeau, M.; Rad, A. M.; Bonab, A. A.; Tompkins, R. G.; colony-stimulating factors stimulate proliferation of human
Fagan, S. P. Stem cells and burns: Review and therapeutic keratinocytes. Arch. Dermatol. Res. 289(10):600–602; 1997.
implications. J. Burn. Care Res. 31(6):874–881; 2010. 24. Kim, C. H.; Lee, J. H.; Won, J. H.; Cho, M. K. Mesenchymal
7. Caplan, A. I.; Correa, D. The MSC: An injury drugstore. stem cells improve wound healing in vivo via early activa-
Cell Stem Cell 9(1):11–15; 2011. tion of matrix metalloproteinase-9 and vascular endothe-
8. Caplan, A. I.; Dennis, J. E. Mesenchymal stem cells as tro- lial growth factor. J. Korean Med. Sci. 26(6):726–733; 2011.
phic mediators. J. Cell. Biochem. 98(5):1076–1084; 2006. 25. Kim, W. S.; Park, B. S.; Sung, J. H.; Yang, J. M.; Park,
9. Chen, J. S.; Wong, V. W.; Gurtner, G. C. Therapeutic S. B.; Kwak, S. J.; Park, J. S. Wound healing effect of
potential of bone marrow-derived mesenchymal stem cells adipose-derived stem cells: A critical role of secretory factors on
for cutaneous wound healing. Front. Immunol. 3:192; 2012. human dermal fibroblasts. J. Dermatol. Sci. 48(1):15–24; 2007.
10. Chen, L.; Tredget, E. E.; Wu, P. Y.; Wu, Y. Paracrine 26. Krampera, M.; Cosmi, L.; Angeli, R.; Pasini, A.; Liotta, F.;
factors of mesenchymal stem cells recruit macrophages and Andreini, A.; Santarlasci, V.; Mazzinghi, B.; Pizzolo, G.;
endothelial lineage cells and enhance wound healing. PLoS Vinante, F.; Romagnani, P.; Maggi, E.; Romagnani, S.;
One 3(4):e1886; 2008. Annunziato, F. Role for interferon-gamma in the immuno-
11. Clark, S. C.; Kamen, R. The human hematopoietic colony- modulatory activity of human bone marrow mesenchymal
stimulating factors. Science 236(4806):1229–1237; 1987. stem cells. Stem Cells 24(2):386–398; 2006.
12. da Silva Meirelles, L.; Caplan, A. I.; Nardi, N. B. In search 27. Kurita, Y.; Tsuboi, R.; Ueki, R.; Rifkin, D. B.; Ogawa, H.
of the in vivo identity of mesenchymal stem cells. Stem Immunohistochemical localization of basic fibroblast growth
Cells 26(9):2287–2299; 2008. factor in wound healing sites of mouse skin. Arch.
13. da Silva Meirelles, L.; Chagastelles, P. C.; Nardi, N. B. Dermatol. Res. 284(4):193–197; 1992.
Mesenchymal stem cells reside in virtually all post-natal 28. La Rocca, G.; Anzalone, R.; Corrao, S.; Magno, F.; Loria,
organs and tissues. J. Cell Sci. 119(Pt 11):2204–2213; 2006. T.; Lo Iacono, M.; Di Stefano, A.; Giannuzzi, P.; Marasa,
14. Deng, J.; Zou, Z. M.; Zhou, T. L.; Su, Y. P.; Ai, G. P.; L.; Cappello, F.; Zummo, G.; Farina, F. Isolation and char-
Wang, J. P.; Xu, H.; Dong, S. W. Bone marrow mesenchy- acterization of Oct-4+/HLA-G+ mesenchymal stem cells
mal stem cells can be mobilized into peripheral blood by from human umbilical cord matrix: Differentiation potential
G-CSF in vivo and integrate into traumatically injured and detection of new markers. Histochem. Cell. Biol. 131(2):
cerebral tissue. Neurol. Sci. 32(4):641–651; 2011. 267–282; 2009.
15. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, 29. Lau, K.; Paus, R.; Tiede, S.; Day, P.; Bayat, A. Exploring
I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, the role of stem cells in cutaneous wound healing. Exp.
D.; Horwitz, E. Minimal criteria for defining multipotent Dermatol. 18(11):921–933; 2009.
mesenchymal stromal cells. The International Society for 30. Lewkowicz, N.; Lewkowicz, P.; Banasik, M.; Kurnatowska,
Cellular Therapy position statement. Cytotherapy 8(4):315– A.; Tchorzewski, H. Predominance of Type 1 cytokines and
317; 2006. decreased number of CD4(+)CD25(+high) T regulatory
16. Erices, A.; Conget, P.; Minguell, J. J. Mesenchymal progen- cells in peripheral blood of patients with recurrent aphthous
itor cells in human umbilical cord blood. Br. J. Haematol. ulcerations. Immunol. Lett. 99(1):57–62; 2005.
109(1):235–242; 2000. 31. Lewkowicz, P.; Tchorzewski, H.; Dytnerska, K.; Banasik,
17. European Medicines Agency. Guideline on the minimum M.; Lewkowicz, N. Epidermal growth factor enhances
quality and non-clinical data for certification of advanced TNF-alpha-induced priming of human neutrophils. Immunol.
therapy medicinal products. London; 2010:22. http://www. Lett. 96(2):203–210; 2005.
ema.europa.eu/docs/en_GB/document_library/Scientific_ 32. Li, W.; Fan, J.; Chen, M.; Woodley, D. T. Mechanisms of
guideline/2010/01/WC500070031.pdf; accessed on April human skin cell motility. Histol. Histopathol. 19(4):1311–
1st 2013. 1324; 2004.
18. Ghannam, S.; Bouffi, C.; Djouad, F.; Jorgensen, C.; Noel, D. 33. Liang, C. C.; Park, A. Y.; Guan, J. L. In vitro scratch
Immunosuppression by mesenchymal stem cells: Mechanisms assay: A convenient and inexpensive method for analysis
and clinical applications. Stem Cell Res. Ther. 1(1):2; 2010. of cell migration in vitro. Nat. Protoc. 2(2):329–333; 2007.
19. Grossman, R. M.; Krueger, J.; Yourish, D.; Granelli- 34. Lin, Z. Q.; Kondo, T.; Ishida, Y.; Takayasu, T.; Mukaida,
Piperno, A.; Murphy, D. P.; May, L. T.; Kupper, T. S.; N. Essential involvement of IL-6 in the skin wound-healing
Sehgal, P. B.; Gottlieb, A. B. Interleukin 6 is expressed in process as evidenced by delayed wound healing in
high levels in psoriatic skin and stimulates proliferation of IL-6-deficient mice. J. Leukoc. Biol. 73(6):713–721; 2003.
cultured human keratinocytes. Proc. Natl. Acad. Sci. USA 35. Liu, Y.; Dulchavsky, D. S.; Gao, X.; Kwon, D.; Chopp, M.;
86(16):6367–6371; 1989. Dulchavsky, S.; Gautam, S. C. Wound repair by bone mar-
20. Gurtner, G. C.; Werner, S.; Barrandon, Y.; Longaker, M. T. row stromal cells through growth factor production. J. Surg.
Wound repair and regeneration. Nature 453(7193):314– Res. 136(2):336–341; 2006.
321; 2008. 36. Martin, P. Wound healing-aiming for perfect skin regenera-
21. Hamou, C.; Callaghan, M. J.; Thangarajah, H.; Chang, E.; tion. Science 276(5309):75–81; 1997.
Chang, E. I.; Grogan, R. H.; Paterno, J.; Vial, I. N.; 36a.Martins, J. P.; Santos, J. M.; de Almeida, J. M.; Felipe,
Jazayeri, L.; Gurtner, G. C. Mesenchymal stem cells can M. A.; Teixeria de Almeida, M. V.; Almeida, S. C. P.;
UCX® REGENERATIVE ACTIVITY 877

Água-Doce, A.; Varela, A.; Gilljam, M.; Stellan, B.; 47. Santos, J. M.; Coelho, M. C.; Martins, J. P.; Basto, V.;
Pohl, S.; Dittmar, K.; Lindenmaier, W.; Alici, E.; Graça, L.; Cruz, P.; Soares, R.; Cruz, H. Optimized and defined
Cruz, P. E.; Cruz, H. J.; Bárcia, N. Towards an advanced method for isolation and preservation of precursor cells
therapy medicinal product based on mesenchymal stromal from human umbilical cord. European Patent EP2203555
cells isolated from the umbilical cord tissue: Quality A2. Portugal; 2008.
and safety data. Stem Cell Research & Therapy 5(1). 48. Schultz, G.; Rotatori, D. S.; Clark, W. EGF and TGF-alpha
doi:10.1186/scrt398 in wound healing and repair. J. Cell. Biochem. 45(4):
37. Maxson, S.; Lopez, E. A.; Yoo, D.; Danilkovitch-Miagkova, 346–352; 1991.
A.; Leroux, M. A. Concise review: Role of mesenchymal 49. Shin, L.; Peterson, D. A. Human mesenchymal stem cell
stem cells in wound repair. Stem Cells Transl. Med. 1(2): grafts enhance normal and impaired wound healing by
142–149; 2012. recruiting existing endogenous tissue stem/progenitor cells.
38. Meirelles Lda, S.; Fontes, A. M.; Covas, D. T.; Caplan, Stem Cells Transl. Med. 2(1):33–42; 2013.
A. I. Mechanisms involved in the therapeutic properties of 50. Singer, A. J.; Clark, R. A. Cutaneous wound healing. N.
mesenchymal stem cells. Cytokine Growth Factor Rev. Engl. J. Med. 341(10):738–746; 1999.
20(5–6):419–427; 2009. 51. Smith, A. N.; Willis, E.; Chan, V. T.; Muffley, L. A.; Isik,
38a.Nascimento, D. S.; Mosqueira, D.; Sousa, L. M.; Teixeria, F. F.; Gibran, N. S.; Hocking, A. M. Mesenchymal stem
M.; Felipe, M.; Resende, T. P.; Araújo, A. F.; Valente, M.; cells induce dermal fibroblast responses to injury. Exp. Cell
Almeida, J.; Martins, J. P.; Santos, J. M.; Bárcia, R. N.; Res. 316(1):48–54; 2010.
Cruz, P.; Cruz, H.; Pinto-do-Ó, P. Human umbilical cord 52. Song, G.; Nguyen, D. T.; Pietramaggiori, G.; Scherer, S.;
tissue- derived stromal celss attenuate remodeling after Chen, B.; Zhan, Q.; Ogawa, R.; Yannas, I. V.; Wagers,
myocardial infarction by proangiogenic, antiapoptotic, and A. J.; Orgill, D. P.; Murphy, G. F. Use of the parabiotic
endogenous cell-activation mechanisms. Stem Cell model in studies of cutaneous wound healing to define the
Research & Therapy 5(1). doi:10.1186/scrt394 participation of circulating cells. Wound Repair Regen.
39. Nissen, N. N.; Polverini, P. J.; Koch, A. E.; Volin, M. V.; 18(4):426–432; 2010.
Gamelli, R. L.; DiPietro, L. A. Vascular endothelial growth 53. Uddin, M.; Seumois, G.; Lau, L. C.; Rytila, P.; Davies,
factor mediates angiogenic activity during the proliferative D. E.; Djukanovic, R. Enhancement of neutrophil function
phase of wound healing. Am. J. Pathol. 152(6):1445–1452; by the bronchial epithelium stimulated by epidermal growth
1998. factor. Eur. Respir. J. 31(4):714–724; 2008.
40. Pastore, S.; Mascia, F.; Mariani, V.; Girolomoni, G. The 54. Vallabhaneni, K. C.; Tkachuk, S.; Kiyan, Y.; Shushakova,
epidermal growth factor receptor system in skin repair and N.; Haller, H.; Dumler, I.; Eden, G. Urokinase receptor
inflammation. J. Invest. Dermatol. 128(6):1365–1374; 2008. mediates mobilization, migration, and differentiation of mes-
41. Peng, C.; He, Q.; Luo, C. Lack of keratinocyte growth fac- enchymal stem cells. Cardiovasc. Res. 90(1):113–121; 2011.
tor retards angiogenesis in cutaneous wounds. J. Int. Med. 55. Walter, M. N.; Wright, K. T.; Fuller, H. R.; MacNeil, S.;
Res. 39(2):416–423; 2011. Johnson, W. E. Mesenchymal stem cell-conditioned medium
42. Peplow, P. V.; Chatterjee, M. P. A review of the influence accelerates skin wound healing: An in vitro study of fibro-
of growth factors and cytokines in in vitro human blast and keratinocyte scratch assays. Exp. Cell Res. 316(7):
keratinocyte migration. Cytokine 62(1):1–21; 2013. 1271–1281; 2010.
43. Petit, I.; Szyper-Kravitz, M.; Nagler, A.; Lahav, M.; Peled, A.; 56. Wang, H. S.; Hung, S. C.; Peng, S. T.; Huang, C. C.; Wei,
Habler, L.; Ponomaryov, T.; Taichman, R. S.; Arenzana- H. M.; Guo, Y. J.; Fu, Y. S.; Lai, M. C.; Chen, C. C. Mes-
Seisdedos, F.; Fujii, N.; Sandbank, J.; Zipori, D.; Lapidot, enchymal stem cells in the Wharton’s jelly of the human
T. G-CSF induces stem cell mobilization by decreasing bone umbilical cord. Stem Cells 22(7):1330–1337; 2004.
marrow SDF-1 and up-regulating CXCR4. Nat. Immunol. 57. Weiss, M. L.; Troyer, D. L. Stem cells in the umbilical
3(7):687–694; 2002. cord. Stem Cell Rev. 2(2):155–162; 2006.
44. Pontikoglou, C.; Deschaseaux, F.; Sensebe, L.; Papadaki, 58. Werner, S.; Grose, R. Regulation of wound healing by growth
H. A. Bone marrow mesenchymal stem cells: Biological factors and cytokines. Physiol. Rev. 83(3):835–870; 2003.
properties and their role in hematopoiesis and hemato- 59. Wu, Y.; Zhao, R. C.; Tredget, E. E. Concise review: Bone
poietic stem cell transplantation. Stem Cell Rev. 7(3): marrow-derived stem/progenitor cells in cutaneous repair
569–589; 2011. and regeneration. Stem Cells 28(5):905–915; 2010.
45. Ripa, R. S.; Haack-Sorensen, M.; Wang, Y.; Jorgensen, E.; 60. Yang, S.; Huang, S.; Feng, C.; Fu, X. Umbilical cord-
Mortensen, S.; Bindslev, L.; Friis, T.; Kastrup, J. Bone marrow derived mesenchymal stem cells: Strategies, challenges, and
derived mesenchymal cell mobilization by granulocyte- potential for cutaneous regeneration. Front. Med. 6(1):41–
colony stimulating factor after acute myocardial infarction: 47; 2012.
Results from the Stem Cells in Myocardial Infarction 61. Zeddou, M.; Briquet, A.; Relic, B.; Josse, C.; Malaise,
(STEMMI) trial. Circulation 116(11 Suppl):I24–I30; 2007. M. G.; Gothot, A.; Lechanteur, C.; Beguin, Y. The umbili-
46. Santos, J. M.; Barcia, R. N.; Simoes, S. I.; Gaspar, M. M.; cal cord matrix is a better source of mesenchymal stem cells
Calado, S.; Agua-Doce, A.; Almeida, S. C.; Almeida, J.; (MSC) than the umbilical cord blood. Cell. Biol. Int. 34(7):
Filipe, M.; Teixeira, M.; Martins, J. P.; Graca, L.; Cruz, M. E.; 693–701; 2010.
Cruz, P.; Cruz, H. The role of human umbilical cord 62. Zittermann, S. I.; Issekutz, A. C. Basic fibroblast growth
tissue-derived mesenchymal stromal cells (UCX(R)) in the factor (bFGF, FGF-2) potentiates leukocyte recruitment to
treatment of inflammatory arthritis. J. Transl. Med. 11: inflammation by enhancing endothelial adhesion molecule
18; 2013. expression. Am. J. Pathol. 168(3):835–846; 2006.

Você também pode gostar