Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

ARTICLE IN PRESS

+ MODEL
Journal of Plastic, Reconstructive & Aesthetic Surgery (2009) xx, 1e9

Adipose-derived stem cells enhance peripheral


nerve regeneration
P.G. di Summa a,b, P.J. Kingham b,c, W. Raffoul a, M. Wiberg c,
G. Terenghi b, D.F. Kalbermatten a,c,*

a
Chirurgie Plastique et Reconstructive CHUV, Université de Lausanne, Rue de Bugnon 46, 1005 Lausanne, CH, Switzerland
b
Blond McIndoe Research Laboratories. The University of Manchester, Manchester, UK
c
Departments of Surgical and Perioperative Science (Hand surgery) and Integrative Medical Biology (Anatomy), Umea
University, Umea, Sweden

Received 30 March 2009; accepted 10 September 2009

KEYWORDS Summary Traumatic injuries resulting in peripheral nerve lesions often require a graft to
Adipose-derived bridge the gap. Although autologous nerve auto-graft is still the first-choice strategy in recon-
stem cells; structions, it has the severe disadvantage of the sacrifice of a functional nerve. Cell transplan-
Peripheral nerve tation in a bioartificial conduit is an alternative strategy to create a favourable environment
regeneration; for nerve regeneration. We decided to test new fibrin nerve conduits seeded with various cell
Differentiated cells; types (primary Schwann cells and adult stem cells differentiated to a Schwann cell-like pheno-
Sciatic nerve type) for repair of sciatic nerve injury.
Two weeks after implantation, the conduits were removed and examined by immunohisto-
chemistry for axonal regeneration (evaluated by PGP 9.5 expression) and Schwann cell pres-
ence (detected by S100 expression). The results show a significant increase in axonal
regeneration in the group of fibrin seeded with Schwann cells compared with the empty fibrin
conduit. Differentiated adipose-derived stem cells also enhanced regeneration distance in
a similar manner to differentiated bone marrow mesenchymal stem cells. These observations
suggest that adipose-derived stem cells may provide an effective cell population, without the
limitations of the donor-site morbidity associated with isolation of Schwann cells, and could be
a clinically translatable route towards new methods to enhance peripheral nerve repair.
ª 2009 British Association of Plastic, Reconstructive and Aesthetic Surgeons. Published by
Elsevier Ltd. All rights reserved.

Numerous surgical procedures are performed each year for working days with corresponding economic consequences.1
peripheral nerve repair, with a significant amount of lost Nerve lesions without defect or with a short gap are usually
treated by end-to-end coaptation. Traumatic injuries
* Corresponding author. Tel.: þ41 21 314 25 06; Fax: þ41 21 314 resulting in longer peripheral nerve lesions often require
25 31. a graft to bridge the gap. Unfortunately, full recovery may
E-mail address: daniel.kalbermatten@bluewin.ch (D.F. never be achieved, particularly with extended lesions.2
Kalbermatten). Although autologous nerve autograft is still the first-choice

1748-6815/$ - see front matter ª 2009 British Association of Plastic, Reconstructive and Aesthetic Surgeons. Published by Elsevier Ltd. All rights reserved.
doi:10.1016/j.bjps.2009.09.012

Please cite this article in press as: di Summa PG et al., Adipose-derived stem cells enhance peripheral nerve regeneration, J Plast
Reconstr Aesthet Surg (2009), doi:10.1016/j.bjps.2009.09.012
ARTICLE IN PRESS
+ MODEL
2 P.G. di Summa et al.

strategy in reconstructions, they have several disadvan- adipose tissue stromal cells.24e26 Although literature
tages caused by the limited availability of donor tissue, terminology can be confusing,27,28 we will use the generic
sacrifice of functional nerve and potential formation of nomenclature of adipose-derived stem cells (ADSCs) to
neuroma.2e4 describe these cells.
Nerve autografts have been mimicked by venous and As previously cited, stem and progenitor cells from adult
arterial conduit grafts, which show no functional benefits stromal tissue retain the capacity to differentiate towards
compared with standard nerve grafts.5 Allografts have also mesenchymal and non-mesenchymal lineages, similar to bone
been tested, but they require an undesirable long-term marrow stromal cells,24,29 showing pluripotency above mul-
immunosuppressive therapy.6 Many attempts have been tipotency.30 Regarding neural differentiation, several studies
made to develop conduits made of biodegradable mate- proved trans-differentiation of ADSC.23,24,31e33 This suggests
rials, which could guide the regenerating axons without that adult stem cells may not be as limited by lineage as
inhibiting the process of growth and maturation.7 Recently, previously thought and, in fact, manifest profound plasticity.
we described a bioresorbable nerve constructed from fibrin Recently, we showed how Schwann cell-like differenti-
glue,8 which shows good tissue acceptance and a porous ated ADSC could enhance neurite outgrowth in vitro.4 In
structure that allows neurotrophic growth factors to this current study, we have tested the ability of the
penetrate into the lumen of the conduit. Moreover, the differentiated ADSC to enhance nerve regeneration in vivo.
resorption of the conduit within 4 weeks avoids compres- We compared these cells with SC-differentiated MSCs and
sion syndromes and enhances biocompatibility.9 primary adult SCs seeded in fibrin conduits.
Our aim was to optimise the fibrin conduit, previously
described, by seeding it with regenerative cells. Cell
Materials and Methods
transplantation in a bioartificial conduit is an alternative
strategy to create a favourable environment for nerve
regeneration10: in this study, we compared different cell Fibrin conduit
populations such as primary Schwann cells (SCs) and stem
cells differentiated towards to a Schwann cell phenotype. The fibrin conduit was prepared from two-compound fibrin
SCs offer a highly preferred substrate for axon migration glue (Tisseel Kit VH 1.0, Baxter SA, USA). Tisseel
and release of bioactive factors that further enhance contains fibrinogen, 70e110 mg ml1; plasma fibronectin,
migration. They synthesise adhesion molecules (CAMs), 2e9 mg ml1; factor XIII, 10e50 U ml1; plasminogen,
release growth factors (GFs) and build up basement 40e120 mg ml1; aprotinin solution 3000 KIU ml1; thrombin
membranes for the support and guidance of the sprouting 4 IU ml1; and calcium chloride 40 mmol l1. Fibrin glue was
axons.11,12 Despite showing regeneration enhancement,13 dispensed in a mould prepared from silicone around
SCs have limited clinical applications. The culture of a stainless steel core and pressed into shape for 5 min
a sufficient amount of SC to achieve optimal conditions for (Figure 1). This allows the generation of uniform conduits
transplantation in nerve conduits is time consuming and measuring 14 mm in length, with a 2-mm lumen and 1-mm
requires particular care for in vitro expansion and wall thickness designed to bridge a 10-mm gap in the left
a constant input of GFs.14 Moreover, SCs are not easily sciatic nerve of 250 g adult Sprague-Dawley rats (Janvier,
accessible without nerve biopsy and bear the need to France).
sacrifice an autologous nerve, with the related complica-
tions of anaesthesia and pain at the harvest site.4,15 Cell harvest
Instead, the ideal transplantable cell should be easily
accessible, should proliferate rapidly in culture and should Schwann cells
successfully integrate into the host tissue with immunological All cells were obtained from Sprague-Dawley rats (Janvier,
tolerance.16,17 The growing applicability of stem cells has France). These were euthanised by CO2 according to the
opened new frontiers to nerve regeneration: the advantage local veterinary commission in Lausanne (Switzerland) and
of using these cell lines is that cell differentiation may be study was carried out in accordance with the European
stimulated by advancing axons and pluripotency, allows Community Council directive 86/609/ECC for the care and
multiple differentiation paths, which can create a positive use of laboratory animals.
environment for axonal regeneration.1 To harvest Schwann cells, sciatic nerves were exposed,
Bone-marrow-derived mesenchymal stem cells (MSCs) removed and kept in Dulbecco’s modified Eagle’s medium
can be induced to differentiate into non-mesenchymal fates plus glutamax (DMEM, Invitrogen, UK) containing 1% pen-
in vitro, such as Schwann cells, improving myelin formation icillinestreptomycin. Nerves were then dissected in trunks,
and nerve regeneration in vivo after their transplantation de-sheathed and finally chopped in 1-mm segments. The
into different models of peripheral nerve injury.18e20 segments were then plated in a Petri dish with SC growth
Adipose tissue is a mesodermally derived complex tissue medium supplemented by 14 mM forskolin and 100 ng ml1
that, besides adipocytes and pre-adipocytes, contains neuregulin-1 b1 (R&D Systems, UK). The cells were incu-
a stromal population, which includes microvascular endo- bated for 2 weeks at 37  C with 5% CO2 with fresh medium
thelial cells, smooth muscle cells, resident monocytes, added approximately every 72 h. After these 2 weeks,
lymphocytes and stem cells.21,22 These non-adipocyte cells medium was aspirated and 0.125% (w/v) collagenase type
are known as stromal vascular fraction (SVF) and can be IV and 117 U mg1 dispase were added to the Petri dish.
isolated by centrifugation of collagenase-digested adipose After 24-h incubation, the cell suspension was filtered
tissue.23 In the past few years, it has been shown how through a 70-mm cell strainer (Falcon; BD Biosciences
cultured SVFs give rise to multipotent precursor cell from Discovery Labware, Bedford, MA), centrifuged at 900 rpm

Please cite this article in press as: di Summa PG et al., Adipose-derived stem cells enhance peripheral nerve regeneration, J Plast
Reconstr Aesthet Surg (2009), doi:10.1016/j.bjps.2009.09.012
ARTICLE IN PRESS
+ MODEL
Adipose-derived stem cells enhance peripheral nerve regeneration 3

Figure 1 Preparation of fibrin conduits.A. The fibrin conduit (Tisseel) with stainless steel wire inserted in the lumen as a spacer
while the conduit is stored before implantation.B. The fibrin conduit shows an intact lumen after removal of the stainless steel
wire.C. The fibrin conduits are stored in DMEM medium before being seeded with regenerative cells.D. A fibrin conduit implanted
in vivo to repair a rat sciatic nerve injury.

for 5 min to obtain the cell pellet. Finally, the cell pellet a 21-guage needle. The resulting cell suspension was tritu-
was re-suspended in SC growth medium, seeded into rated, filtered through a 70-mm Falcon filter and centrifuged
25-cm2 flask precoated with poly-D-lysine (Sigma, St Louis, for 5 min at 600 g. The supernatant was aspirated, the cell
MO, USA) and incubated in the same conditions. The bolus re-suspended in mesenchymal cell growth medium and
following day, the medium was freshly changed and the the cells plated in 75-cm2 tissue culture flasks and incubated
cells were left to proliferate. At confluence, the SCs were in 5% CO2 at 37  C. Haematopoietic cells were eliminated by
purified by an antibody complement method to eradicate washing daily with DMEM until all the non-adherent cells were
the remaining fibroblasts.13,17,34 removed. The cells were then allowed to grow to confluence.

Adipose-derived stem cells Stem cell differentiation


Adipose-derived stem cells were harvested as described
previously.4 After euthanasia, the inguinal fat pad was care- The multi-potent nature of the cell cultures was tested by
fully dissected and minced using a sterile razor blade. The ensuring their ability to differentiate along various lineages,
tissue was enzymatically digested for 2 h at 37  C using 0.15% as previously described.4,34 Further, undifferentiated ADSCs
(w/v) collagenase type I (Invitrogen, UK). The solution was and MSCs were differentiated into an SC-like phenotype at
passed through a 70-mm filter to remove undissociated tissue, early passages.4,18 Growth medium was removed from sub-
neutralised by adding modified Eagle’s medium (a-MEM; confluent cultures and replaced with medium supplemented
Invitrogen, UK) containing 10% (v/v) foetal bovine serum with 1 mM b-mercaptoethanol (Sigma-Aldrich, UK) for 24 h.
(FBS) and centrifuged at 800  g for 5 min. The stromal cell The cells were then washed and fresh medium supplemented
pellet was re-suspended in MEM containing 10% (v/v) FBS and with 35 ng ml1 all-trans-retinoic acid was added. A further
1% (v/v) penicillin/streptomycin solution. The cultures were 72 h later, the cells were washed and the medium replaced
maintained at sub-confluent levels in a 37  C incubator with with differentiation medium; cell growth medium supple-
5% CO2 and passaged with trypsin/ethylenediaminetetra- mented with 5 ng mle1 platelet-derived growth factor (PDGF;
acetic acid (EDTA) (Invitrogen, UK) when required. PeproTech Ltd., UK), 10 ng ml1 basic fibroblast growth factor
(bFGF; PeproTech Ltd., UK), 14 mM forskolin and 200 ng ml1
Bone-marrow-derived mesenchymal stem cells neuregulin-1 b1. The cells were incubated for 2 weeks under
MSCs were harvested from adult Sprague-Dawley rat femoral these conditions with fresh medium added approximately
bones34: mesenchymal cell growth medium (MEM- a; Invi- every 72 h. To confirm the effectiveness of the differentiation
trogen; supplemented with 10% FBS (v/v) and 1% penicilline of ADSC and their similarity to the Schwann cell population,
streptomycin) was injected through each marrow cavity using we performed cell immunostaining for typical Schwann cells

Please cite this article in press as: di Summa PG et al., Adipose-derived stem cells enhance peripheral nerve regeneration, J Plast
Reconstr Aesthet Surg (2009), doi:10.1016/j.bjps.2009.09.012
ARTICLE IN PRESS
+ MODEL
4 P.G. di Summa et al.

markers S100 and GFAP.4 Cultures of differentiated ADSC conduit were evaluated using an optical microgrid: the
(dADSC) and SC were compared by immunostaining: the cells length was measured from the beginning of the conduit to
were incubated with mouse anti-glial fibrillary acidic protein the last visible sprout of the regenerating conus.
(GFAP; 1:500; LabVision, USA) and rabbit anti-S100 (1:500;
Dako, Denmark) overnight at 4  C. Phosphate-buffered solu- Statistical analysis
tion (PBS)-washed slides were then treated with goat anti-
rabbit FITC (1:100; Vector Labs, UK) and goat anti-mouse CY3 One-way analysis of variance (ANOVA) with Bonferroni
(1:200; Amersham, UK) conjugated secondary antibodies. multiple comparison test was used to statistically analyse
Slides were examined using an Olympus IX51 inverted fluo- data (GraphPad Prism). Significance was determined as
rescence microscope. The dADSCs showed similar immuno- *P < 0.05, **P < 0.01 and ***P < 0.001.
cytochemical properties when compared with SCs (Figure 2).
Bone-marrow-derived MSCs were differentiated (dMSC) in
Results
a similar manner as previously shown.34

Cell seeding and microsurgery Cell seeding and conduit properties

Prior to in vivo experiments, we tested the fibrin conduit and


A couple of hours before implantations in rats, the cells
its capacity to retain cells within. We filled two different
were trypsinised and, after centrifugation, 2  106 cells
fibrin conduits with ADSC (80  106 ml1 cell concentration),
were suspended in 50 ml of differentiation medium and
then we fixed them at 48 h. By DAPI staining, we found a good
injected into the fibrin tubes. To avoid cell dispersion during
capacity of fibrin of retaining cells inside the lumen
the procedure, one side of the fibrin tube was previously
(Figure 3). Fibrin conduits were easy to handle and micro-
capped by a fibrin glue clot. Before implantation, the fibrin
surgical suture was uneventful. All animals survived; none of
conduit was easily decapped from the fibrin clots at its ends.
the animals developed infections or surgical complications
The control group in the absence of cells (empty) was
except autotomy, which was present in one case out of 24.
similarly filled with 50 ml of differentiation medium alone.
After 2 weeks in vivo, fibrin conduits showed an intact
The sciatic nerve was transected and nerve ends were fixed
structure, with proximal and distal ends partially resorbed.
to the conduit by a single epineural suture (9/0 Prolene,
No signs of haematoma or infection were detected.
Ethicon): proximal and distal nerve stumps were inserted
2 mm into the tube, thus leaving a 10-mm gap. Muscles and
fascia layers were closed with single resorbable stitches (4/ Axon regeneration
0 Softcat, Braun) and the skin by a continuous running
suture (4/0 Prolene, Ethicon). The various groups of fibrin Sciatic nerve axonal regeneration was different depending on
conduits (each one formed by six animals for a total n Z 24) the type of cells which had been seeded in the fibrin conduits.
were left in place for 2 weeks and subsequently harvested Axonal regeneration distance was assessed proximally by PGP
together with the proximal and distal nerve stumps. 9.5 immunohistochemistry (Figure 4). Conduits filled with SCs
showed the best regeneration distance (5.758  0.12 mm)
followed by fibrin containing dMSC (4.986  0.19 mm) or
Nerve conduit harvesting and dADSC (4.968  0.22 mm) (Figure 6A). All three groups with
immunohistochemistry cells resulted in better regeneration than the empty fibrin
conduit group (4.155  0.07 mm). PGP 9.5 positive axons were
Animals were sacrificed after 2 weeks by CO2 chamber associated with S100 positive cells at the proximal regener-
euthanasia. Harvested conduits were fixed by para- ating stumps (Figure 5). The distances of S100 positive cell
formaldehyde (PFA) for 16 h, then rinsed 24 h at 4  C in PBS migration at the stumps showed a similar pattern to PGP 9.5:
containing 30% sucrose and 0.1% sodium azide, with PBS the SC group (5.858  0.13 mm) followed by conduits con-
changes after about 12 h to eliminate any PFA remnants. taining dMSC (5.035  0.21 mm) or dADSC (5.032  0.20 mm)
The specimens were then embedded in OCT freezing media and empty fibrin conduits (4.108  0.08 mm) (Figure. 6B).
(Tissue-tek, Sakura, Japan), and longitudinal cryo-sections Statistical analysis showed that fibrin conduits seeded
(14 mm) were prepared onto slides (Superfrost plus, with SC significantly improved axonal regeneration
Menzel-Gläser, Germany). distance, and proximal SC invasion compared with the
After 3  5 min PBS washes, a casein blocking solution empty fibrin conduit (** p < 0.01). Differentiated adult stem
was applied for 1 h before incubating slides overnight with cells also showed a positive impact on nerve regeneration
primary antibodies rabbit anti-S100 (1:500, Dako, UK) and with a significantly enhanced nerve regeneration distance
rabbit anti-PGP 9.5 (1:500, Dako, UK). The following day (dADSC vs. empty: p < 0.05 PGP 9.5 and p < 0.01 S100;
after 3  5 min PBS washes, slides were incubated with dMSC vs. empty: p < 0.01 for both PGP 9.5 and S100). No
secondary antibody (1:500, Alexa Fluor goat anti-rabbit, significant difference was noticed between the dADSC
Invitrogen, USA) at room temperature in the dark for group and the dMSC group with either staining.
40 min. After 3  5 min PBS washes, the slides were finally
mounted with Vectashield with DAPI (Vector Labs, UK) and
examined under the fluorescence microscope Discussion
(10  magnification) with the regenerating conus directed
towards the distal end.35 Axonal regeneration distance In this study, we analysed the behaviour of fibrin conduits
(PGP 9.5) and S100 positive cell distribution inside the seeded with different cell populations as a nerve guide to

Please cite this article in press as: di Summa PG et al., Adipose-derived stem cells enhance peripheral nerve regeneration, J Plast
Reconstr Aesthet Surg (2009), doi:10.1016/j.bjps.2009.09.012
ARTICLE IN PRESS
+ MODEL
Adipose-derived stem cells enhance peripheral nerve regeneration 5

Figure 2 Differentiation of stem cells, dADSC and SC immunostained for S100 (green), GFAP (red) and DAPI (blue). Differenti-
ation of ADSC resulted in 40% of cells co-expressing the SC markers and with a bipolar elongated morphology. SC cultures were
routinely >97% positive for both S100 and GFAP indicating the absence of significant quantities of contaminating fibroblast cells.

Please cite this article in press as: di Summa PG et al., Adipose-derived stem cells enhance peripheral nerve regeneration, J Plast
Reconstr Aesthet Surg (2009), doi:10.1016/j.bjps.2009.09.012
ARTICLE IN PRESS
+ MODEL
6 P.G. di Summa et al.

Figure 3 Analysis of cell seeding. A. Haematoxylin staining of a fibrin conduit seeded with cells in vitro and (B) shown at high
magnification.C DAPI staining of a fibrin conduit seeded with cells in vitro and (D) shown at high magnification.

repair sciatic nerve injury. We compared the regeneration edges were noted after 2 weeks in vivo, displaying biode-
achievable with SC, dADSC, dMSC and with an empty gradability of the fibrin conduit. The porous structure of
conduit. First, we found that all conduits seeded with cells the conduit, already noted with in vitro experiments,8
supported axonal regeneration without collapsing and seems to allow passage of neurotrophic factors into the
retained a lumen, which provides a permissive environment lumen, the surface of fibrin forms a barrier preventing
for regeneration. fibrous tissue to invade the lumen.36
No signs of haematoma or infection were detected at Fibrin has been previously used as a matrix for MSC cell
explantations, confirming the good tissue acceptance of suspension37 and as a new nerve guide8: our results show
fibrin without altering the tissue environment at the that, at least in the short term, the fibrin conduit can be
implantation side. Moreover, first signs of resorption at the

Figure 5 S100 staining shows the proximal Schwann cell


Figure 4 PGP 9.5 staining shows axonal regeneration in the migration in the fibrin conduits either empty or seeded
fibrin conduits either empty or seeded with different regenera- with different regenerative cells. A. Empty fibrin conduit.
tive cells. A. Empty fibrin conduit. B. Fibrin conduit þ dADSC. B. Fibrin conduit þ dADSC. C. Fibrin conduit þ dMSC. D. Fibrin
C. Fibrin conduit þ dMSC. D. Fibrin conduit þ SC. conduit þ SC.

Please cite this article in press as: di Summa PG et al., Adipose-derived stem cells enhance peripheral nerve regeneration, J Plast
Reconstr Aesthet Surg (2009), doi:10.1016/j.bjps.2009.09.012
ARTICLE IN PRESS
+ MODEL
Adipose-derived stem cells enhance peripheral nerve regeneration 7

Figure 6 Quantification of regeneration distances measured with (A) PGP 9.5 and (B) S100 cell staining. Graphs show
a significant improvement in axon regeneration distance in all groups (dADSC, dMSC, SC), when compared with the empty fibrin
group. Values are mean distances (mm)  SEM. (* p < 0.05; ** p< 0.01; *** p < 0.001).

optimised by cell delivery, resulting in higher peripheral promoting peripheral nerve regeneration can be promising,
nerve regeneration. considering the already successful application of MSC
The best regenerative values were found combining in vivo in the longer-term group.14,34,35 Stem cells could aid
fibrin conduits and SC. This should be expected, given the regeneration by the release of soluble nerve growth factors
crucial role covered by SCs in nerve regeneration and in such as brain-derived neurotrophic factor41 or angiogenic
creating the adequate environment through which regen- molecules, including vascular endothelial growth factor.42
erating axons grow to reach their peripheral targets.12 Other studies have shown that dMSC can physically engraft
Indeed, SCs were injected for the first time into a fibrin and myelineate regenerating axons in vivo20, suggesting
conduit, giving encouraging results, with a better regen- that multiple mechanisms may exist in stem cell therapy.
erating distance when compared statistically with the In terms of future clinical use, humans have abundant
empty fibrin conduit. These data seem to strengthen the subcutaneous fat deposits, and ADSCs can be isolated in
argument that SC can help in enhancing nerve regeneration large quantities by conventional liposuction procedure
when seeded in a conduit.10,14 under local anaesthesia, thus overcoming the discomfort
Concerning dMSC and dADSC, we also detected a statis- and the tissue morbidity associated with bone marrow
tically significant improvement in nerve regeneration aspiration and showing potential advantages for tissue
distance (PGP 9.5 staining, respectively, 4.986 and 4.968 engineering application, compared with MSC.40,43,44
for dMSC and dADSC vs. 4.155 in the empty fibrin conduit). Furthermore, the frequency of MSCs in bone marrow ranges
From these data, we could argue that, even if these cells between 1 in 25 000 to 1 in 100 000,45 whereas ADSCs
still not reach the full regenerative potential of SC, they constitute a more abundant proportion, approximately 2%
can play an effective role in nerve regeneration. Interest- of lipoaspirate cells.25,28
ingly, we could find that dADSC promote regeneration in the The promising results achieved from the combination of
same manner as dMSC (difference between the two groups regenerative cells and fibrin conduit are hopeful steps for
p > 0.05) as we saw reporting overlapping results when we future clinical applications in cases of peripheral nerve
compared the axonal regeneration distance (PGP 9.5) and injury. Similarly, the interesting outcomes obtained from
SC invasion (S100) for the two groups. the dADSCs, which appear comparable to the dMSC,
This seems to confirm the previously reported similari- underline the effectiveness of this cell population and their
ties between these two cell populations, concerning promising applications in nerve regeneration. A longer-term
morphology,38 immune phenotype39 and trans-differentia- group will be necessary to confirm the positive effects on
tion capacity along mesodermal27 and non-mesodermal functional recovery and to see if dADSC will maintain this
lineages.26,40 This equivalence of dADSC and dMSC in similarity compared with dMSC.

Please cite this article in press as: di Summa PG et al., Adipose-derived stem cells enhance peripheral nerve regeneration, J Plast
Reconstr Aesthet Surg (2009), doi:10.1016/j.bjps.2009.09.012
ARTICLE IN PRESS
+ MODEL
8 P.G. di Summa et al.

Acknowledgements regeneration and functional recovery after complete transec-


tion of adult rat spinal cord. J Neuropathol Exp Neurol 2005;
64:37e45.
We thank Professor Daniel V. Egloff, director of the 16. Azizi SA, Stokes D, Augelli BJ, et al. Engraftment and migration
department for plastic surgery CHUV for his support and of human bone marrow stromal cells implanted in the brains of
useful comments to this project and Catherine Pythoud albino ratsesimilarities to astrocyte grafts. Proc Natl Acad Sci
BSc, Jean Francois Brunet PhD for technical support. The U S A 1998;95:3908e13.
authors are grateful to the SwissLife Foundation, the SUVA 17. Tohill M, Terenghi G. Stem-cell plasticity and therapy for
and the University of Lausanne FBM for their financial injuries of the peripheral nervous system. Biotechnol Appl
support. Biochem 2004;40:17e24.
18. Dezawa M, Takahashi I, Esaki M, et al. Sciatic nerve regeneration
in rats induced by transplantation of in vitro differentiated
Conflict of Interest bone-marrow stromal cells. Eur J Neurosci 2001;14:1771e6.
19. Tohill MP, Mann DJ, Mantovani CM, et al. Green fluorescent
None. protein is a stable morphological marker for schwann cell
transplants in bioengineered nerve conduits. Tissue Eng 2004;
10:1359e67.
References 20. Keilhoff G, Goihl A, Langnase K, et al. Transdifferentiation of
mesenchymal stem cells into Schwann cell-like myelinating
1. Chalfoun CT, Wirth GA, Evans GR. Tissue engineered nerve cells. Eur J Cell Biol 2006;85:11e24.
constructs: where do we stand? J Cell Mol Med 2006;10: 21. Kokai LE, Rubin JP, Marra KG. The potential of adipose-derived
309e17. adult stem cells as a source of neuronal progenitor cells. Plast
2. Hu J, Zhu QT, Liu XL, et al. Repair of extended peripheral Reconstr Surg 2005;116:1453e60.
nerve lesions in rhesus monkeys using acellular allogenic nerve 22. Prunet-Marcassus B, Cousin B, Caton D, et al. From heteroge-
grafts implanted with autologous mesenchymal stem cells. Exp neity to plasticity in adipose tissues: site-specific differences.
Neurol 2007;204:658e66. Exp Cell Res 2006;312:727e36.
3. Marchesi C, Pluderi M, Colleoni F, et al. Skin-derived stem cells 23. Safford KM, Rice HE. Stem cell therapy for neurologic disor-
transplanted into resorbable guides provide functional nerve ders: therapeutic potential of adipose-derived stem cells. Curr
regeneration after sciatic nerve resection. Glia 2007;55: Drug Targets 2005;6:57e62.
425e38. 24. Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human
4. Kingham PJ, Kalbermatten DF, Mahay D, et al. Adipose-derived adipose tissue: implications for cell-based therapies. Tissue
stem cells differentiate into a Schwann cell phenotype and Eng 2001;7:211e28.
promote neurite outgrowth in vitro. Exp Neurol 2007;207: 25. Strem BM, Hicok KC, Zhu M, et al. Multipotential differentiation
267e74. of adipose tissue-derived stem cells. Keio J Med 2005;54:132e41.
5. Battiston B, Tos P, Conforti LG, et al. Alternative techniques 26. Guilak F, Lott KE, Awad HA, et al. Clonal analysis of the
for peripheral nerve repair: conduits and end-to-side neuro- differentiation potential of human adipose-derived adult stem
rrhaphy. Acta Neurochir Suppl 2007;100:43e50. cells. J Cell Physiol 2006;206:229e37.
6. Bain JR, Mackinnon SE, Hudson AR, et al. The peripheral nerve 27. Schaffler A, Buchler C. Concise review: adipose tissue-derived
allograft in the primate immunosuppressed with Cyclosporin A: stromal cellsebasic and clinical implications for novel cell-
I. Histologic and electrophysiologic assessment. Plast Reconstr based therapies. Stem Cells 2007;25:818e27.
Surg 1992;90:1036e46. 28. Fraser JK, Wulur I, Alfonso Z, et al. Fat tissue: an underap-
7. Mohanna PN, Terenghi G, Wiberg M. Composite PHB-GGF preciated source of stem cells for biotechnology. Trends
conduit for long nerve gap repair: a long-term evaluation. Biotechnol 2006;24:150e4.
Scand J Plast Reconstr Surg Hand Surg 2005;39:129e37. 29. Sanchez-Ramos J, Song S, Cardozo-Pelaez F, et al. Adult bone
8. Kalbermatten DF, Pettersson J, Kingham PJ, et al. New fibrin marrow stromal cells differentiate into neural cells in vitro.
conduit for peripheral nerve repair. J Reconstr Microsurg 2009; Exp Neurol 2000;164:247e56.
25:27e33. 30. Safford KM, Hicok KC, Safford SD, et al. Neurogenic differen-
9. Kalbermatten DF, Kingham PJ, Mahay D, et al. Fibrin matrix for tiation of murine and human adipose-derived stromal cells.
suspension of regenerative cells in an artificial nerve conduit. Biochem Biophys Res Commun 2002;294:371e9.
J Plast Reconstr Aesthet Surg 2008;61:669e75. 31. Ashjian PH, Elbarbary AS, Edmonds B, et al. In vitro differen-
10. Stang F, Fansa H, Wolf G, et al. Structural parameters of tiation of human processed lipoaspirate cells into early neural
collagen nerve grafts influence peripheral nerve regeneration. progenitors. Plast Reconstr Surg 2003;111:1922e31.
Biomaterials 2005;26:3083e91. 32. Safford KM, Safford SD, Gimble JM, et al. Characterization of
11. Terenghi G. Peripheral nerve regeneration and neurotrophic neuronal/glial differentiation of murine adipose-derived adult
factors. J Anat 1999;194:1e14. stromal cells. Exp Neurol 2004;187:319e28.
12. Rodriguez FJ, Verdu E, Ceballos D, et al. Nerve guides seeded 33. Wang B, Han J, Gao Y, et al. The differentiation of rat
with autologous schwann cells improve nerve regeneration. adipose-derived stem cells into OEC-like cells on collagen
Exp Neurol 2000;161:571e84. scaffolds by co-culturing with OECs. Neurosci Lett 2007;421:
13. Mosahebi A, Woodward B, Wiberg M, et al. Retroviral labeling 191e6.
of Schwann cells: in vitro characterization and in vivo trans- 34. Caddick J, Kingham PJ, Gardiner NJ, et al. Phenotypic and
plantation to improve peripheral nerve regeneration. Glia functional characteristics of mesenchymal stem cells differ-
2001;34:8e17. entiated along a Schwann cell lineage. Glia 2006;54:840e9.
14. Pereira Lopes FR, Camargo de Moura Campos L, Dias 35. Tohill M, Mantovani C, Wiberg M, et al. Rat bone marrow
Correa Jr J, et al. Bone marrow stromal cells and resorbable mesenchymal stem cells express glial markers and stimulate
collagen guidance tubes enhance sciatic nerve regeneration in nerve regeneration. Neurosci Lett 2004;362:200e3.
mice. Exp Neurol 2006;198:457e68. 36. Takazawa R, Yamato M, Kageyama Y, et al. Mesothelial cell
15. Kamada T, Koda M, Dezawa M, et al. Transplantation of bone sheets cultured on fibrin gel prevent adhesion formation in an
marrow stromal cell-derived Schwann cells promotes axonal intestinal hernia model. Tissue Eng 2005;11:618e25.

Please cite this article in press as: di Summa PG et al., Adipose-derived stem cells enhance peripheral nerve regeneration, J Plast
Reconstr Aesthet Surg (2009), doi:10.1016/j.bjps.2009.09.012
ARTICLE IN PRESS
+ MODEL
Adipose-derived stem cells enhance peripheral nerve regeneration 9

37. Bensaid W, Triffitt JT, Blanchat C, et al. A biodegradable fibrin 42. Rehman J, Traktuev D, Li J, et al. Secretion of angiogenic and
scaffold for mesenchymal stem cell transplantation. Bioma- antiapoptotic factors by human adipose stromal cells. Circu-
terials 2003;24:2497e502. lation 2004;109:1292e8.
38. Kern S, Eichler H, Stoeve J, et al. Comparative analysis of 43. Tholpady SS, Katz AJ, Ogle RC. Mesenchymal stem cells from
mesenchymal stem cells from bone marrow, umbilical cord rat visceral fat exhibit multipotential differentiation in vitro.
blood, or adipose tissue. Stem Cells 2006;24:1294e301. Anat Rec A Discov Mol Cell Evol Biol 2003;272:398e402.
39. Gimble JM, Katz AJ, Bunnell BA. Adipose-derived stem cells for 44. Matsumoto T, Kano K, Kondo D, et al. Mature adipocyte-
regenerative medicine. Circ Res 2007;100:1249e60. derived dedifferentiated fat cells exhibit multilineage poten-
40. Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source tial. J Cell Physiol 2008;215:210e22.
of multipotent stem cells. Mol Biol Cell 2002;13:4279e95. 45. Banfi A, Bianchi G, Galotto M, et al. Bone marrow stromal
41. Zhao L, Wei X, Ma Z, et al. Adipose stromal cells-conditional damage after chemo/radiotherapy: occurrence, consequences
medium protected glutamate-induced CGNs neuronal death by and possibilities of treatment. Leuk Lymphoma 2001;42:
BDNF. Neurosci Lett 2009;452:238e40. 863e70.

Please cite this article in press as: di Summa PG et al., Adipose-derived stem cells enhance peripheral nerve regeneration, J Plast
Reconstr Aesthet Surg (2009), doi:10.1016/j.bjps.2009.09.012

You might also like