You are on page 1of 13

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/266576860

Combining xanthan and chitosan membranes to multipotent mesenchymal


stromal cells as bioactive dressings for dermo-epidermal wounds

Article  in  Journal of Biomaterials Applications · October 2014


DOI: 10.1177/0885328214553959 · Source: PubMed

CITATIONS READS

20 88

8 authors, including:

Carolina Caliari-Oliveira Amanda Mizukami


University of Campinas University of São Paulo
12 PUBLICATIONS   118 CITATIONS    12 PUBLICATIONS   86 CITATIONS   

SEE PROFILE SEE PROFILE

Kamilla Swiech Dimas Tadeu Covas


University of São Paulo University of São Paulo
38 PUBLICATIONS   332 CITATIONS    516 PUBLICATIONS   4,491 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

3D Biofabrication of biological tissue and organs: production of spheroids of mesenchymal stem cells aiming at obtaining chondral structures View project

Modification of T cells with anti-CD19 chimeric antigen receptor View project

All content following this page was uploaded by Carolina Caliari-Oliveira on 24 April 2015.

The user has requested enhancement of the downloaded file.


Soft Tissues and Materials
Journal of Biomaterials Applications
2015, Vol. 29(8) 1155–1166
! The Author(s) 2014
Combining xanthan and chitosan Reprints and permissions:
sagepub.co.uk/journalsPermissions.nav
membranes to multipotent DOI: 10.1177/0885328214553959
jba.sagepub.com
mesenchymal stromal cells as bioactive
dressings for dermo-epidermal wounds

Márcia Z Bellini1,2, Carolina Caliari-Oliveira3, Amanda Mizukami3, Kamilla Swiech4,


Dimas T Covas3, Eduardo A Donadi3, Pedro Oliva-Neto5 and Ângela M Moraes1

Abstract
The association between tridimensional scaffolds to cells of interest has provided excellent perspectives for obtaining
viable complex tissues in vitro, such as skin, resulting in impressive advances in the field of tissue engineering applied to
regenerative therapies. The use of multipotent mesenchymal stromal cells in the treatment of dermo-epidermal wounds
is particularly promising due to several relevant properties of these cells, such as high capacity of proliferation in culture,
potential of differentiation in multiple skin cell types, important paracrine and immunomodulatory effects, among others.
Membranes of chitosan complexed with xanthan may be potentially useful as scaffolds for multipotent mesenchymal
stromal cells, given that they present suitable physico-chemical characteristics and have adequate tridimensional struc-
ture for the adhesion, growth, and maintenance of cell function. Therefore, the purpose of this work was to assess the
applicability of bioactive dressings associating dense and porous chitosan-xanthan membranes to multipotent mesen-
chymal stromal cells for the treatment of skin wounds. The membranes showed to be non-mutagenic and allowed
efficient adhesion and proliferation of the mesenchymal stromal cells in vitro. In vivo assays performed with mesenchymal
stromal cells grown on the surface of the dense membranes showed acceleration of wound healing in Wistar rats, thus
indicating that the use of this cell-scaffold association for tissue engineering purposes is feasible and attractive.

Keywords
Scaffolds, chitosan, xanthan gum, stem cells, dressings, regenerative medicine, skin lesions

Introduction
to provide faster and more effective healing of the
The skin, the largest organ of the human body, consti- damaged skin are of great relevance.
tutes the first barrier of organism defense, playing
important roles in homeostasis, such as body tempera-
ture regulation and protection against dehydration, as
well as providing support to blood vessels and nerves.1,2 1
School of Chemical Engineering, University of Campinas (UNICAMP),
In severe burns and chronic wounds of various etiolo- Campinas, SP, Brazil
2
gies, the spontaneous regeneration process is severely Integrated Adamantinenses Colleges (FAI), Adamantina, SP, Brazil
3
School of Medicine of Ribeirão Preto, University of São Paulo (USP),
impaired, raising the need for therapeutic intervention. Ribeirão Preto, SP, Brazil
The rapid healing of the skin area damaged or lost 4
School of Pharmaceutical Sciences of Ribeirão Preto, University of
may represent an improvement of prognosis of the São Paulo (USP), Ribeirão Preto, SP, Brazil
5
patient, and for this purpose, conventional dressings School of Sciences and Languages of Assis, São Paulo State University
are extensively used. However, ideally, a dressing (UNESP), Assis, SP, Brazil
should not only protect the wound but also promote These first two authors contributed equally to this work.
the healing process by providing a suitable microenvir- Corresponding author:
Ângela M Moraes, Department of Engineering of Materials and
onment, hydrated and with thermal insulation, allow- Bioprocesses, University of Campinas (UNICAMP), Campinas, SP,
ing the elimination of the excess of exudate and CEP 13083-852, Brazil.
promoting gas exchange.3 Thus, approaches intended Email: ammoraes@feq.unicamp.br

Downloaded from jba.sagepub.com at CIDADE UNIVERSITARIA on April 24, 2015


1156 Journal of Biomaterials Applications 29(8)

Human skin may be successfully reconstituted predisposition to infections. In this sense, artificial epi-
in vitro through the culture of skin cells on biological dermis produced from multipotent cells, even those of
scaffolds such as de-epidermized human dermis4 and allogenous origin, could function as a temporary sub-
bovine collagen,5 being morphologically and function- stitute while the patient waits for an autologous graft
ally compatible with in vivo human skin. Examples obtained in vitro.
of products for skin tissue engineering already Several factors are favorable to the use of multipo-
approved by the Food and Drug Administration tent mesenchymal stromal cells (MSCs) in the treat-
(FDA, USA) are ApligrafÕ (produced by ment of skin wounds, such as their regenerating and
Organogenesis, Massachusetts, USA) and OrCelÕ immunoregulation properties, their easy isolation and
(Forticell Bioscience, New York, USA), both consti- in vitro expansion, added to satisfactory results in
tuted of human keratinocytes and fibroblasts cultivated recent preclinical and clinical studies.15 Moreover,
in scaffolds of bovine collagen type I. MSCs are poorly immunogenic,16 allowing therapies
Approaches involving the use of scaffolds obtained based on previously established cultures of healthy
from animal origin, human or not, as supports for cell donors, what represents a considerable benefit for
growth may raise concerns due to shortage of donors patients with severe wounds requiring immediate
and to the possibility of transmission of pathogens. treatment.
However, the use of certain bioactive polymers as Thus, in the search for alternatives for more effective
scaffolds for tissue engineering is attractive due to treatments for dermo-epidermal lesions, this study
the fact that it is possible, through their use, to repro- aimed to demonstrate the feasibility of the association
duce relevant characteristics of the native extracellular of Xn and Ch membranes with MSCs and the develop-
matrix and also to obtain materials with rates of deg- ment of a biodressing for the therapy of this type of
radation in vivo compatible to those of new tissue wounds.
development.6
Several studies have demonstrated the potential of
natural-origin bioabsorbable polymers such as chitosan Materials and methods
(Ch) combined or not with alginate,7,8 gelatin,9 cellu-
lose and its derivatives10 in tissue engineering. Lamellar
Animals and cells
dense (compacted) and porous scaffolds of Ch com- Wistar rats weighting 200 to 300 g with 4 to 6 weeks in
plexed with xanthan (Xn) seems to be particularly pro- age were kept in boxes lined with sawdust, in a room
mising for this purpose, given that Xn is a with controlled temperature and light (22 C, 12 h light
polysaccharide obtained by fermentation, with more and 12 h dark). Throughout the procedure, the animals
controlled quality in contrast to the natural counter- received food and filtered water ad libtum.
parts processed from animal or vegetable sources. Multipotent mesenchymal stromal cells were isolated
Dense and porous Ch-Xn scaffolds were analyzed in from the bone marrow of at least three animals per
detail and extensively characterized by Veiga and batch, through flushing with PBS/EDTA of their
Moraes11 and Bellini et al.12 The dense membranes femurs and tibias after the removal of bone epiphysis.17
are thin, transparent in both wet and dry states, and The animals were sacrificed prior to bone extraction
present no apparent pores on their surfaces, having a and the collected cells were cultivated in Alpha
highly compacted structure when dry. The porous Minimum Essential Medium with 0.58 g/L L-gluta-
Ch-Xn membranes, on the other hand, are opaque mine, 4.50 g/L D-glucose (Gibco, Grand Island, NY),
and have a large number of uniformly distributed supplemented with 3.70 g/L NaHCO3 (Merck,
pores. Both membrane types show high capacity of Germany), antibiotic solution containing 10,000 UI/L
absorption of physiological solutions and body fluids, penicillin and 10,000 mg/L streptomycin (Gibco,
excellent stability in cell culture medium, mechanical Grand Island, NY), 5.96 g/L Hepes (Gibco, Grand
properties compatible with those of the skin and negli- Island, NY), and 15% (v/v) fetal bovine serum (SFB,
gible cytotoxicity to L929 cells, comprising a set of rele- Thermo Scientific, Rockford, IL, EUA) and main-
vant characteristics for application as wound dressings tained at 37 C and 5% of CO2 in an incubator
and scaffolds for cell growth. (Tecnal, Brazil, model TE-399). Cells at the third or
Artificial autologous epidermis can be successfully fourth passage were then tested regarding adhesion
obtained by inoculation of epidermal and dermal cells and proliferation on the Ch:Xn membranes as well as
derived from the patient himself on polymeric scaffolds for substrate consumption and metabolite production
based on Ch.13,14 However, this approach is frequently during cultivation and for cell therapy assays in an
hampered by the long time required for obtaining it, in vivo model.
about 4 to 5 weeks.5 This waiting period may result in The homogeneity of the MSCs population after suc-
dehydration of the patient, increasing his or her cessive subculture procedures was assessed by flow

Downloaded from jba.sagepub.com at CIDADE UNIVERSITARIA on April 24, 2015


Bellini et al. 1157

cytometry at the Flow Cytometry Laboratory of the Analysis of MSCs behavior after inoculation on the
Regional Blood Center (Ribeirão Preto/FMRP-USP,
São Paulo, Brazil). Monoclonal antibodies against the
Ch:Xn membranes
markers CD45, CD11b, CD31, CD29, CD90, CD105, Membrane samples (1.5 cm in diameter) were individu-
and CD44 (eBioscience, USA) were used. ally arranged in 24-well flat-bottom plates (TPP, Brazil)
The procedures in vitro and in vivo are consistent and washed three times with PBS/EDTA buffer. After
with the standards of the Brazilian College of Animal washing, 700 mL of a-MEM supplemented medium
Experimentation (COBEA) and were reviewed and were added to each well and the plates were incubated
approved by the Ethics Committee on Animal at 37 C and 5% CO2 overnight to swell. Afterwards,
Experimentation (CETEA) of the School of Medicine the excess of culture medium was removed and 100 mL
of Ribeirão Preto, University of São Paulo, where the of a suspension of 3.0  105 cells/mL were inoculated
experiments were conducted under the registration into each well. After 2 to 3 h in the incubator, 900 mL
number 158/2010. of a-MEM supplemented medium were added and the
samples were maintained in the incubator for up to
168 h, with exchange of the culture medium every
Membrane preparation and characterization 48 h. As a control, the adhesion of cells to the 24-well
The dense and porous membranes of Ch complexed plate without the membranes was also monitored.
with Xn were prepared according to the procedures During this period, samples were collected to determine
described by Bellini et al.,12 at a mass ratio of Xn to cell adhesion kinetics and the concentrations of glucose,
Ch equal to 1:1, using 96% deacetylated Ch, xanthan lactate, glutamine, and ammonium in the culture
gum, and Pluronic F68Õ from Sigma-Aldrich medium and of cells grown in the biomaterials, as
(St. Louis, MO, USA) and glacial acetic acid from well as MSCs morphology on the scaffolds as described
Merck (São Paulo, Brazil). The water used was distilled below. All experiments were performed in at least
and deionized in a MilliQ system (Millipore, São Paulo, triplicate.
Brazil).
After sterilization with ethylene oxide, the mem- Kinetics of cell adhesion. Cell adhesion was determined by
branes showed the characteristics summarized in monitoring the decrease in the number of cells sus-
Table 1. Prior to in vitro and in vivo tests with MSCs, pended in the culture medium after inoculation.
the mutagenic potential of the membranes was evalu- Samples from the supernatant were collected at every
ated by the AMES test, which is based on the induction hour during 6 h and analyzed for cell concentration and
of reverse mutation in histidine auxotrophic Salmonella viability using the Vi-Cell XR analyzer Beckman
typhimurium bacteria18 at the Tecam Laboratórios (São Coulter (Brea, CA, USA).
Paulo, Brazil), following the procedures described in
the OECD Guidelines for Testing of Chemicals Concentration of glucose, lactate, glutamine, and
(1997),19 ISO10993-3 (1992),20 ISO10993-1 (2003),21 ammonium. Samples of culture medium supernatant
and ISO10993-12 (2007).22 were collected every 24 h to determine the basic

Table 1. Properties of the dense and porous Ch-Xn membranes used (summarized from Bellini et al.12).

Values

Dense Ch-Xn Porous Ch-Xn


Property membrane membrane

Thickness of dry membranes (mm) 0.10  0.04 1.84  0.23


Uptake of water after 24 h at 37 C (g/g) 85.6  3.7 29.8  1.8
Uptake of simulated body fluid after 24 h at 37 C (g/g) 15.8  0.1 18.0  0.3
Membrane mass loss after exposure to water for 24 h at 37 C (%) 14.0  0.9 16.9  0.4
Membrane mass loss after exposure to simulated body fluid for 24 h at 37 C (%) 10.3  1.4 34.7  7.5
Tensile strength of dry membranes (MPa) 25.1  7.8 1.25  0.2
Elongation at break of dry membranes (%) 2.0  0.7 2.1  0.6
Cytotoxicity of membrane extracts to L929 cells (%) 1.6  0.7 2.6  0.4
Ch: chitosan; Xn: xanthan.

Downloaded from jba.sagepub.com at CIDADE UNIVERSITARIA on April 24, 2015


1158 Journal of Biomaterials Applications 29(8)

metabolic activities of MSCs in the biomaterial. the wounds were treated with dense membranes of
Glucose, lactate, and glutamine were determined 1.5 cm in diameter previously hydrated with a-MEM
using an automatic analyzer (YSI 2700, Yellow culture medium supplemented with 15% fetal bovine
Springs Instruments, OH) and ammonium was deter- serum, but with no cells inoculated on the surface. In
mined using an ion-selective electrode (Thermo the third group (MSCs), 1  105 MSCs were directly
Scientific, ORION 720A). applied at the edges of the wound, and no coverage
with Ch:Xn membrane was provided. Depositing the
Concentration of cells adhered to the scaffolds. The concen- cells in the incision edges facilitated their attachment
tration of viable cells was determined indirectly by the to the lesion and prevented their displacement to
MTT method (3-(4,5-dimethylthiazol-2-yil)-2,5-diphe- regions away from the lesion during the early stages
nyltetrazolium bromide) (Sigma-Aldrich, St. Louis, of cell adhesion. In the last group (Memb þ MSCs),
USA). The membrane samples were removed from the wounds were dressed with the dense scaffold
the plate, washed once with PBS/EDTA buffer, and inoculated with the MSCs 96 h before the in vivo test
incubated with 200 mL of a-MEM without phenol red as previously described. The blood exuded from the
and 20 mL of MTT reagent (5 mg/mL in PBS) at 37 C lesions aided in the adhesion of the membrane to the
for 4 h. The formazan crystals formed were dissolved wound bed, avoiding its displacement thereafter. No
with 200 mL of 0.1 N HCl in anhydrous isopropanol secondary dressings were used as additional protection
and the absorbance of the supernatant was measured of the tested treatments and as a preventive measure
at 570 nm in a plate reader (Multiskan FC Thermo against the occurrence of trauma injuries, individual
Scientific) and compared to a calibration curve previ- cages relatively large in size (49  34  16 cm3) were
ously established. Three independent samples were ana- used to accommodate the animals during the healing
lyzed daily and subsequently discarded. period.
Photos were taken (Fuji S2000HD FilmFinepix digi-
Morphology of cells adhered to the scaffolds. The morph- tal camera, resolution of 10.0 megapixels) and wound
ology of cells attached to the biomaterial was analyzed healing percentage was analyzed immediately after
by scanning electron microscopy (SEM, model JSM the start of the experiment (day 0) and 20 days after-
5800 LV, JEOL) by immersing the samples into different wards. The wound healing percentage was calculated
fixative solutions, one consisting of 4% paraformalde- by the ratio between the final and the initial area of
hyde (v/v) and glutaraldehyde 2% (m/v) in 0.2 M caco- the wound through the Image JÕ software. Statistical
dylate buffer and the other of osmium tetroxide (1% m/ analyses were based on one-way ANOVA and the
v) in cacodylate buffer at 0.2 M, all from Sigma Tukey’s test to determine the differences among the
Chemical Co, Brazil. Afterwards, the samples were experimental groups. Differences were considered
sequentially dehydrated in alcoholic solutions of differ- statistically significant when the p value was lower
ent concentrations and dried through the critical point than 5% (p < 0.05). These tests were performed using
drying procedure with CO2. The samples were then the software PRISM 5.0.
metal-coated through depositing a thin gold layer (92 Å).

In vivo wound healing assay Results


Wistar rats were anaesthetized intraperitoneally with 1 mg
of anesthetic solution per gram of animal. The anesthetic
Mutagenicity of the Ch-Xn membranes
solution consisted of a 1:1 (v/v) mixture of ketamine solu- The dense and porous Ch-Xn membranes tested
tion at 10% (Agener Union, Brazil) and xylazine at herein were extensively characterized referring to sev-
20 mg/mL (Dopaser-Calier, Brazil). After the anesthesia, eral relevant physico-chemical properties in a previous
the back of the animals was shaved and two circular work,12 showing a set of features to a large extent
wounds of 1.5 cm in diameter were performed with sur- adequate for their potential application in the tissue
gical scissors in previously marked areas. All layers of the engineering area (Table 1), however, their mutageni-
skin, including the panniculus carnosus, were removed city potential was not assessed at that time. Hence,
with the aid of the scissors, ensuring the production of their potential to cause mutations was analyzed in
lesions of about the same thickness in all animals. After the present work. It was observed (Table 2) that
15 min of completion, the animals still anesthetized were after 72 h of incubation of the membranes with
divided into four groups of at least three rats each and TA98, TA100, TA102, TA1535, and TA1537 strains
subjected to the different treatments tested. of Salmonella typhimurium in the absence and presence
In the control group (Ctrl), the wounds were not of metabolic activation, none of the samples showed
covered by any dressing. In the second group (Memb), mutagenic characteristics.

Downloaded from jba.sagepub.com at CIDADE UNIVERSITARIA on April 24, 2015


Bellini et al. 1159

Table 2. Results of the AMES mutagenicity test performed on the dense and porous Ch-Xn membranes.

Salmonella typhimurium

Sample description TA98 TA100 TA102 TA1535 TA1537

In the absence of metabolic activation


Negative controla () () () () ()
Positive controlb (þ) (þ) (þ) (þ) (þ)
Dense Ch-Xn membrane () () () () ()
Porous Ch-Xn membrane () () () () ()
In the presence of metabolic activation
Negative controla () () () () ()
Positive controlc (þ) (þ) (þ) (þ) (þ)
Dense Ch-Xn membrane () () () () ()
Porous Ch-Xn membrane () () () () ()
Ch: chitosan; Xn: xanthan; (þ): mutagenic; (): not mutagenic.
a
Deionized water.
b
5.0 mg per dish of sodium azide.
c
2.5 mg/dish of 2-aminoantracene.

Behavior of MSCs inoculated on the Ch:Xn Table 3. Expression of surface antigen markers by mesenchy-
mal stromal cells (MSCs) on 3rd passage.
membranes
Surface antigen Mean expression value (%)
All experiments involving MSCs were performed with
cultures established from the third or fourth passage, CD45 5.5  4.7
with CD45, CD11b, and CD31 expression levels lower CD11b 4.8  1.8
than 5.5% and high expression of CD29 and CD44 CD31 2.5  1.8
surface markers, as characterized by flow cytometry CD29 92.4  1.2
(Table 3). CD44 85.8  8.6
The adhesion of MSCs to the materials tested, in
terms of the gradual reduction in the number of cells
in suspension, is shown in Figure 1. More than 50% of dense and porous Ch:Xn membrane samples, respect-
the total initial population had already adhered to the ively (Figure 2). Afterwards, cell death occurred.
materials 2 h after MSCs inoculation in all tested sys- The concentrations of glucose, lactate, glutamine,
tems, however, the cells adhered more rapidly to the and ammonium in the culture media were monitored
surface of the control culture plate than to the mem- over 168 h (Figure 3). Due to the frequent exchange of
branes (Figure 1a). Three hours after the inoculation, culture medium, depletion of glucose and glutamine
the number of cells in the supernatant of the control was not observed. Lactic acid was produced as a bypro-
culture plate and of the vessels containing dense mem- duct of the glucose metabolism, reaching maximum
brane samples was similar and more than 98% of the values of 3.86 and 7.78 mM in porous and dense mem-
cells inoculated was capable of adhering to the branes, respectively. The maximum accumulation of
membranes. ammonium attained was 0.64 mM in the supernatant
As shown in Figure 1(b), the viability of the MSCs of the porous membranes and equal to 0.51 mM for
in the supernatant of all systems decreased with the dense Ch-Xn membranes.
time, indicating that cells that were unable to adhere
to the materials tested were not able to survive in Morphology of MSCs. The morphological aspect of the
suspension. cells cultured on the surface of the tested materials
was examined by SEM and it is shown in Figure 4.
Growth and metabolism of MSCs in the membranes. MSCs MSCs were distributed over the surface of both formu-
were inoculated at an initial concentration of 3.0  104 lations studied (Figure 4e and f). Cells cultured in the
cells per well on the surface of each membrane sample. porous sample proliferated intensely within the pores
Intense cell growth was noticed for up to 96 h of culti- (Figure 4f), in which it is possible to observe the pres-
vation and cell concentration reached maximum values ence of cytoplasmic extensions as well as the production
of 1.1  105 and 1.27  105 cells/per membrane for of extracellular matrix. The dense membranes Ch-Xn

Downloaded from jba.sagepub.com at CIDADE UNIVERSITARIA on April 24, 2015


1160 Journal of Biomaterials Applications 29(8)

(a) (b)
1.2x10 5 100
Cell population in the supernatant
Porous Ch-Xn Membrane Porous Ch-Xn Membrane
Dense Ch-Xn Membrane Dense Ch-Xn Membrane
5
1.0x10 Control - plate Control - plate
80
4
8.0x10

Viability (%)
60
6.0x10 4

40
4.0x10 4

2.0x10 4 20

0.0
0
0 1 2 3 4 5 0 1 2 3 4 5
Time (hours) Time (hours)

Figure 1. Adhesion of MSCs to control polystyrene culture plates and to dense and porous Ch:Xn membranes: (a) cell population
remaining in the supernatant; (b) percentage of viable cells during culture. Data are reported as means  SD for experiments in
triplicate.

marrow cells were extracted because those animals


2.0x105 were sacrificed prior to extraction.
Dense Ch-Xn Membrane
Viable cell density (cells/ membrane)

Porous Ch-Xn Membrane The dense Ch-Xn membrane was selected over the
porous biomaterial due to the lower thickness and the
1.5x105
higher transparency of the former. These characteristics
might contribute positively to aspects related to
1.0x105 improved comfort for the patient with the thinner dres-
sing and also of better visual monitoring of lesion
healing.
5.0x104 The usual aspect of the lesions before treatment, the
percentage of wound healing 20 days after the begin-
0.0
ning of the treatment, and the typical aspect of the
0 24 48 72 96 120 144 168 192 wounds of animals representative from each group
Time (hours)
are respectively shown in Figure 5(a–c).
Figure 2. MSCs proliferation on dense and porous Ch-Xn It was observed that healing progressed well in all
membranes during cultivation for 7 days in supplemented a-MEM animals and that on day 20 after the beginning of the
medium, assessed through the use of the MTT assay. Data are treatment all animals were already in the final process
reported as means  SD for experiments in triplicate. of reepithelialization, even those which did not receive
membranes or MSCs.
The group treated with the membrane containing
adhered MSCs (Memb þ MSCs) showed healing of
remained transparent even after inoculation and culti- wounds significantly higher (96.49  1.62%) when com-
vation of cells and this is a characteristic of great pared to the Ctrl group (90.01  2.38%, p ¼ 0.03). The
importance, since it would allow the effective observa- difference was also significant when compared to the
tion of the wound bed during the lesion recovery group treated with the membrane without cells
process. (Memb), which reached 87.24  3.10% (p ¼ 0.02).
Statistically significant difference was not noticed
when comparing the percentage of healing in groups
In vivo healing assays Memb þ MSCs and MSCs (animals with lesions that
The in vivo wound healing assays were performed using received only MSCs and reached 99.27  0.73% of
Wistar rats subjected to surgical wounds aiming to healing). Consequently, the percentage of wound heal-
assess the applicability of the tested approach upon ing in group MSCs was significantly higher than that of
the principles of translational medicine using allogeneic the Ctrl group and of the Memb group that received the
MSCs, given that it was not possible to use in this part acellular biomembrane (p ¼ 0.003 and p ¼ 0.001,
of the study the same animals from which the bone respectively).

Downloaded from jba.sagepub.com at CIDADE UNIVERSITARIA on April 24, 2015


Bellini et al. 1161

(a) 10 (b) 5
Glucose Glutamine
Lactate Ammonium
8 4
Concentration (mM)

Concentration (mM)
6 3

4 2

2 1

0 0
0 24 48 72 96 120 144 168 192 0 24 48 72 96 120 144 168 192
Time (hours) Time (hours)

(c) 10 (d) 5
Glucose Glutamine
Lactate Ammonium

8 4

Concentration (mM)
Concentration (mM)

6 3

4 2

2 1

0 0
0 24 48 72 96 120 144 168 192 0 24 48 72 96 120 144 168 192
Time (hours) Time (hours)

Figure 3. Variation in the concentrations of glucose, glutamine, lactate, and ammonium during MSCs cell cultivation on: (a and b)
dense Ch-Xn membranes; (a, c and d) porous Ch-Xn membranes. Dashed lines refer to the time points of culture medium changes.

agents in vivo could lead to consequences as serious as


Discussion
cancer induction, even when negative results are
Over the last years tissue engineering has advanced sig- obtained in in vitro cytocompatibility analysis.23 The
nificantly, especially in the dermatology field. One of negative mutagenic activity results attained evidenced
the trends in this field is the use of biomaterials pro- then that compounds with potential to alter cell growth
duced by the combination of inert and low-complexity were not released.
materials with cells that satisfactorily meet require- Once the safety of use of the biomaterials has been
ments of biosafety and are active on the healing of established, assays involving the association of the
the type of wound focused. In this sense, the develop- membranes with MSCs were performed. Despite sup-
ment of a dermal biodressing through the association of posedly higher area for cell accommodation would be
Ch-Xn membranes which were already characterized in available in the porous biomaterial than in the dense
a previous study12 with multipotent MSCs, a widely membranes, the cells cultivated in the biomaterials pre-
studied cell type especially interesting for regenerative sented similar behavior in both membrane formulations
medicine and tissue engineering, was investigated studied. High population increase was noticed in the
in vitro and in vivo on Wistar rats. The results obtained first 96 h of culture, followed by marked decline in
indicate that the formulations tested present suitable viable cells in the subsequent hours. The decrease in
properties aiming application as scaffolds for cell cell viability observed after 96 h cannot be attributed
growth. to the exhaustion of the substrates glucose and glutam-
None of the formulations tested were mutagenic. ine, since they were replenished during culture medium
This property is of particular importance in the devel- exchange, or to the accumulation of toxic byproducts
opment of biomaterials, since the release of genotoxic such as lactic acid and ammonium, which reached

Downloaded from jba.sagepub.com at CIDADE UNIVERSITARIA on April 24, 2015


1162 Journal of Biomaterials Applications 29(8)

Figure 4. Scanning electron micrographs of scaffolds. Dense Ch-Xn membranes: (a) before MSCs inoculation; (b) 24 h after cell
inoculation; (c) 96 h after cell inoculation. Porous Ch-Xn membranes: (d) before MSCs inoculation; (e) 24 h after cell inoculation;
(f) 96 h after cell inoculation.

concentrations of only 3.86 and 0.64 mM, respectively, the dense biomaterials, more severe limitation to cell
for the Ch-Xn porous membranes, and 7.78 and growth in the inner parts of the scaffolds would be
0.51 mM, respectively, for the Ch-Xn dense mem- expected for the porous scaffolds. However, the
branes. Since lactic acid concentrations over 28.0 mM growth curves for both types of membranes are very
and ammonium concentrations over 3.0 mM are con- similar regarding cell population in both systems in
sidered inhibitory for MSCs,24 and those high concen- all periods, then the hypotheses pointing to more
trations were not observed in the supernatant culture severe problems in the porous systems alone cannot
medium, the decrease in the number of viable cells in fully satisfactorily explain the results obtained.
the biomaterials evaluated might be attributed to mass Another possible explanation for the decrease in cell
transfer issues related both to limited diffusion of nutri- population in the dense and porous scaffolds could be
ents (even of oxygen) to the membranes and of toxic the degradation of the polymer matrix during cell
metabolites to the medium outside the scaffolds growth. According to Bellini et al.,12 Ch-Xn dense
(including carbon dioxide produced during cell respir- and porous membranes exhibit weight loss between 17
ation), causing then fast depletion and accumulation and 34% after 144 h of exposure to a sterile culture
respectively of nutrients and metabolites in the inner medium traditionally used for mammalian cell in vitro
parts of the scaffolds. Given that the thickness of the (RPMI supplemented with 10% fetal bovine serum).
porous membranes is significantly higher than that of The presence of cells in the scaffold could have further

Downloaded from jba.sagepub.com at CIDADE UNIVERSITARIA on April 24, 2015


Bellini et al. 1163

(a) #
#
*
(b) 100 *

Wound healing (%)


75

Ctrl Memb MSCs Memb+MSCs

(c)

0 days 7 days 15 days 20 days


Ctrl
Memb
MSCs
Memb+
MSCs

Figure 5. Wound healing progression in animals of the different groups: control group (Ctrl); animals with lesions treated with only
the dense membrane (Memb); animals with wounds treated only with the mesenchymal stromal cells (MSCs); and rats treated with the
dense membrane inoculated with cells (Memb þ MSCs). (a) Usual aspect of the wound in day 0; (b) Percentage of wound healing in
animals of each study group 20 days after the beginning of the treatment (values determined by photographic analysis with the
software Image JÕ and statistical analysis performed using One-Way ANOVA, PRISMA 5.0: *groups with statistical difference from the
Ctrl group; #groups with statistical difference from the Memb group); and (c) Typical aspect of the lesions of animals representative of
each study group 20 days after the beginning of the treatment.

accelerated the degradation process. Nevertheless, bio- satisfactorily to both membrane formulations.
degradation may be seen as a positive factor for a bio- Evidence of extracellular matrix formation was also
material intended to the application as a bioactive noticed. Similar successful results in the culture of
dermal healing device, since in vivo it would facilitate MSCs in scaffolds based on Ch have also been reported
the delivery of bioactive agents incorporated therein, in by Ragetly et al.25,26 The production of extracellular
the present case, the cells cultivated on it, to the deepest matrix is important to provide additional physical sup-
layers of the damaged tissue. port for growing cells and is also relevant in processes
The morphological analysis of the cells grown in the of tissue morphogenesis, differentiation, and homeosta-
membranes indicated that the MSCs adhered sis.27 According to Yang,28 scaffolds containing Ch

Downloaded from jba.sagepub.com at CIDADE UNIVERSITARIA on April 24, 2015


1164 Journal of Biomaterials Applications 29(8)

may stimulate the production of extracellular matrix. solely of Ch or of Ch complexed with alginate have low
Due to its charge distribution at physiological condi- permeability to oxygen (unpublished results), that
tions, Ch can increase, by adsorption, the local concen- might not be the case for the Ch:Xn membrane, since
tration of important negatively charged molecules such no significant difference was observed when comparing
as glycosaminoglycans (GAG) and proteoglycans, Ctrl and Memb groups. These results confirm that the
which are essential components of the extracellular membrane is a biocompatible biomaterial and appar-
matrix. ently inert in this microenvironment, representing a
Morphological differences were observed when com- good alternative for the administration of these cells
paring MSCs cultured on the Ch-Xn scaffolds to the and consisting in a non-invasive and faster alternative
cells cultivated on the polysterene plate (data not for the treatment of skin wounds. Furthermore, the fact
shown). This result was expected, given that MSCs that the membranes need to be applied to the surface of
behave differently when grown on scaffolds with differ- the lesions only in the beginning of the treatment is
ent stiffness.29 As observed by Engler et al.,29 soft quite attractive in the clinical point of view, and given
matrices mimicking the brain are neurogenic, while stif- that the produced dressings are very transparent, their
fer matrices mimicking muscles are myogenic, and periodic removal to inspect the progress of the lesion is
matrices with higher rigidity may be osteogenic not required.
and there is evidence that Ch scaffolds have physical Several works demonstrate the therapeutic potential
and mechanical characteristics effectively useful for the of local application of MSCs in the healing of skin
cultivation of cells such as human dermal fibroblasts wounds. Most of them use topical or intravenous
for dermal regeneration.30 application of MSCs,31–34 whereas others use fibrin
The results obtained when assessing the adhesion sealant for the fixation of the cells to the
and proliferation of MSCs on both dense and porous wound bed.35,36 Effective results were also reported
formulations showed that the two membrane types are by Liu et al.37 when using allogeneic MSCs in a
suitable for application in tissue engineering as sup- collagen scaffold for the reepithelialization of superfi-
ports for cell culture. Lower thickness and higher trans- cial dermal burns in mini pigs. Despite successful out-
parency before and after cell inoculation though come is attained with fibrin or collagen-based
pointed that the dense formulation might be more biomaterials, less limitations might be involved with
appropriate for application in vivo as a bioactive the use of Ch and xanthan membranes as MSCs sup-
dermal dressing. In addition, the processing time to ports for the treatment of skin wounds, since these
obtain porous membranes is significantly higher when polysaccharides have lower probability of transference
compared to that of dense films. of adventitious agents than proteins from mammalian
Highly satisfactory results were observed in the sources. Also, less potential to cause immunogenic
Memb þ MSCs group treated with the biodressing problems might be associated to Ch:Xn scaffolds
combining the dense membrane with the MSCs. The than with supports obtained from proteins of heterol-
cells were able to remain viable and active in the mem- ogous sources.
brane, showing promising therapeutic perspectives. Keratinocytes may be also used for skin lesion regen-
Despite the cells in suspension also showing very effect- eration, but MSCs cells have a significant advantage
ive healing performance (MSCs group), their applica- over these cells. MSCs may be used in allogeneic sys-
tion to the wound site is not as straightforward as that tems, given that they are not too immunogenic, present-
of the biodressing. The transference of the cells already ing low MHC-I expression and absence of expression of
adhered to the membrane surface is much simpler and, MHC-II and of costimulatory molecules.38,39 Since
in addition, the membrane provides greater mechanical relatively long periods of culture are required for both
protection to the wound and to the MSCs themselves. keratinocytes and MSCs, the former type of cells could
Another advantage is that the membrane can function be successfully used for the treatment of chronic ulcers,
as a physical barrier, preventing microbial contamin- being isolated from the patient himself, while MSCs
ation from the outside environment. previously cultured and stored in banks might be
Local application of Ch:Xn acellular biomembrane more useful in the therapy of acute injuries, such as
on the bed ulcers apparently did not interfere in the severe burns, that require immediate treatment and
healing process in comparison to the untreated animals for which limited availability of autologous cells
(Ctrl group), suggesting that the improved therapeutic might be a concern.
potential in groups MSCs and Memb þ MSCs may be
attributed to the MSCs themselves. Despite the fact
Conclusions
that the use of the membrane to cover the wound
implies in an additional barrier to gas permeation In this work, the association of dense and porous mem-
from and to the wound and that membranes composed branes of Ch-Xn with mesenchymal stromal cells was

Downloaded from jba.sagepub.com at CIDADE UNIVERSITARIA on April 24, 2015


Bellini et al. 1165

tested to verify their applicability as bioactive dermal 7. Bueno C and Moraes AM. Development of porous lamel-
healing dressings. The results showed that the mem- lar chitosan-alginate membranes: effect of different sur-
branes are not mutagenic and present favorable archi- factants on biomaterial properties. J Appl Polym Sci
tecture for adhesion, maintenance, and proliferation of 2011; 122: 624–631.
8. Meng X, Tian F, Yang J, et al. Chitosan and alginate
MSCs. In vivo assays performed with the non-porous
polyelectrolyte complex membranes and their properties
membranes containing MSCs on the surface showed sig- for wound dressing application. J Mater Sci Mater Med
nificant acceleration of dermo-epidermal wound heal- 2010; 21: 1751–1759.
ing, leading to the conclusion that this membrane-cells 9. Yang B, Yin Z, Cao J, et al. In vitro cartilage tissue engin-
association might be considered viable in tissue engin- eering using cancellous bone matrix gelatin as a bio-
eering and should be considered for further studies. degradable scaffold. Biomed Mater 2010; 5: 1–8.
10. Ito T, Yeo Y, Highley CB, et al. The prevention of peri-
Acknowledgements toneal adhesions by in situ cross-linking hydrogels of hya-
The authors are grateful to the São Paulo Research luronic acid and cellulose derivatives. Biomaterials 2007;
Foundation (FAPESP, Brazil) for the financial support to 28: 975–983.
this research. MZ Bellini would also like to acknowledge 11. Veiga IV and Moraes AM. Study of the swelling and
the Coordination for the Improvement of Higher Education stability properties of chitosan–xanthan membranes.
Personnel (CAPES, Brazil) for a PhD fellowship and ÂM J Appl Polym Sci 2012; 24: E154–E160.
Moraes acknowledges the National Council for Scientific 12. Bellini MZ, Pires ALR, Vasconcelos MO, et al.
and Technological Development (CNPq, Brazil) for a fellow- Comparison of the properties of compacted and porous
ship. Thanks are also due to the assistance of Alessandra lamellar chitosan–xanthan membranes as dressings and
Almeida for the English review and of Sandra Navarro scaffolds for the treatment of skin lesions. J Appl Polym
Bresciani for the graphic design. Sci 2012; 125: E421–E431.
13. Chen KY, Liao WJ, Kuo SM, et al. Asymmetric chitosan
membrane containing collagen I nanospheres for skin
Declaration of conflicting interests
tissue engineering. Biomacromolecules 2009; 10:
None declared. 1642–1649.
14. Pajoum-Shariati SR, Shokrgozar MA, Vossoughi M,
Funding et al. In vitro co-culture of human skin keratinocytes
This research received grants from São Paulo Research and fibroblasts on a biocompatible and biodegradable
Foundation (FAPESP, Brazil), the Coordination for the scaffold. Iran Biomed J 2009; 13: 169–177.
Improvement of Higher Education Personnel (CAPES, 15. Meirelles SL, Fontes AM, Covas DT, et al. Mechanisms
Brazil) and from the National Council for Scientificand involved in the therapeutic properties of
Technological Development (CNPq, Brazil). mesenchymal stem cells. Cytokine Growth Factor Rev
2009; 20: 419–427.
16. Uccelli A, Moretta L and Pistoia V. Immunoregulatory
function of mesenchymal stem cells. Eur J Immunol 2006;
References
36: 2566–2573.
1. Rodrigues AP, Sanchez EMS, Costa AC, et al. The influ- 17. Soleimani M and Nadri S. A protocol for isolation and
ence of preparation conditions on the characteristics of culture of mesenchymal stem cells from mouse bone
chitosan-alginate dressings for skin lesions. J Appl Polym marrow. Nat Protoc 2009; 4: 102–106.
Sci 2008; 109: 2703–2710. 18. Mortelmans K and Zeiger E. The AMES Salmonella/
2. Ratner BD, Hoffman AS, Schoen FJ, et al. Biomaterials microsome mutagenicity assay. Mutat Res 2000; 455:
science, an introduction to materials in medicine, 2nd ed. 29–60.
San Diego, USA: Elsevier Academic Press, 2004. 19. OECD Guideline for Testing of Chemicals. Bacterial
3. Jayakumar R, Prabaharan M, Sudheesh KPT, et al. Reverse Mutation Test 1997; 471.
Biomaterials based on chitin and chitosan in wound dres- 20. ISO10993-3: Biological evaluation of medical devices –
sing applications. Biotechnol Adv 2011; 29: 322–337. Tests for genotoxicity, carcinogenicity and reproductive
4. Rehder J, Souto LRM, Issa CMBM, et al. Model of toxicity, 1992.
human epiderm reconstructed in vitro with keratinocytes 21. ISO10993-1: Biological evaluation of medical devices –
and melanocytes on dead d-epidermized human dermis. Evaluation and testing, 2003.
São Paulo Med J 2004; 122: 22–25. 22. ISO10993-12: Sample Preparation and Reference
5. Souto LRM, Rehder J, Vassalo J, et al. Model Materials, 2007.
for human skin reconstructed in vitro composed of asso- 23. Keong LC and Halim AS. In vitro models in biocompati-
ciated dermis and epidermis. Sao Paulo Med J 2006; 124: bility assessment for biomedical-grade chitosan deriva-
71–76. tives in wound management. Int J Mol Sci 2009; 10:
6. Holzapfel BM, Reichert JC, Schantz JT, et al. How smart 1300–1313.
do biomaterials need to be? A translational science and 24. Schop D, Janssen FW, Van Rijn LD, et al. Growth,
clinical point of view. Adv Drug Deliv Rev 2013; 65: metabolism, and growth inhibitors of mesenchymal
581–603. stem cells. Tissue Eng Part A 2009; 15: 1877–1886.

Downloaded from jba.sagepub.com at CIDADE UNIVERSITARIA on April 24, 2015


1166 Journal of Biomaterials Applications 29(8)

25. Ragetly GR, Griffon DJ, Lee HB, et al. Effect of chitosan 34. Sasaki M, Abe R, Fujita Y, et al. Mesenchymal stem cells
scaffold microstructure on mesenchymal stem cell chon- are recruited into wounded skin and contribute to wound
drogenesis. Acta Biomater 2010; 6: 1430–1436. repair by transdiferentiation into multiple skin cell type.
26. Ragetly GR, Griffon DJ, Chung YS, et al. The effect of J Immunol 2008; 180: 2581–2587.
type II collagen coating of chitosan fibrous scaffolds on 35. Falanga V, Iwamoto S, Chartier M, et al. Autologous
mesenchymal stem cell adhesion and chondrogenesis. bone marrow-derived cultured mesenchymal stem cells
Acta Biomater 2010; 6: 3988–3997. delivered in a fibrin spray accelerate healing in murine
27. Frantz C, Stewart KM and Weaver VM. The extracellu- and human cutaneous wounds. Tissue Eng 2007; 13:
lar matrix at a glance. J Cell Sci 2010; 123: 4195–200. 1299–1311.
28. Yang T. Chitin-based materials in tissue engineering: 36. Fu X, Fang L, Li X, et al. Enhanced wound-healing qual-
applications in soft tissue and epithelial organ. Int J ity with bone marrow mesenchymal stem cells autograft-
Mol Sci 2011; 12: 1936–1963. ing after skin injury. Wound Repair Regen 2006; 14:
29. Engler AJ, Sen S, Sweeney HL, et al. Matrix elasticity 325–335.
directs stem cell lineage specification. Cell 2006; 126: 37. Liu P, Deng Z, Han S, et al. Tissue-engineered skin con-
677–689. taining mesenchymal stem cells improves burn wounds.
30. Hilmi ABM, Halim AS, Hassan A, et al. In vitro charac- ArtiOcial Organs 2008; 32: 925–931.
terization of a chitosan skin regenerating template as a 38. De Miguel MP, Fuentes-Julián S, Blázquez-Martı́nez A,
scaffold for cells cultivation. SpringerPlus 2013; 2: 79. et al. Immunosuppressive properties of mesenchymal
31. Wu Y, Chen L, Scott PG, et al. Mesenchymal stem cells stem cells: Advances and applications. Curr Mol Med
enhance wound healing through differentiation and 2012; 12: 574–591.
angiogenesis. Stem cells 2007; 25: 2648–2659. 39. Tyndall A, Walker U, Cope A, et al. Immunomodulatory
32. Volk SW, Radu A, Zhang LE, et al. Stromal progenitor properties of mesenchymal stem cells: a review based on
cell therapy corrects the wound-healing defect in the an interdisciplinary meeting held at the Kennedy Institute
ischemic rabbit ear model of chronic wound repair. of Rheumatology Division, London, UK. Arthritis Res
Wound Repair Regen 2007; 15: 736–747. Ther 2007; 9: 301–311.
33. Kwon DS, Gao X, Liu YB, et al. Treatment with bone
marrow-derived stromal cells accelerates wound healing
in diabetic rats. Int Wound J 2008; 5: 453–463.

Downloaded from jba.sagepub.com at CIDADE UNIVERSITARIA on April 24, 2015


View publication stats

You might also like