Sie sind auf Seite 1von 10

DOI:10.1111/j.1600-0625.2010.01087.

x
www.blackwellpublishing.com/EXD
Review Article

Regenerative medicine in dermatology: biomaterials,


tissue engineering, stem cells, gene transfer and beyond
Christina Dieckmann1*, Regina Renner2*, Linda Milkova1,2 and Jan C. Simon1,2
1
Translational Centre for Regenerative Medicine, Leipzig University, Leipzig, Germany;
2
Department of Dermatology, Venerology and Allergology, Leipzig University Medical Center, Leipzig, Germany
Correspondence: Regina Renner, MD, Department of Dermatology, Venerology and Allergology, Leipzig University Medical Center,
Philipp-Rosenthal-Straße 23, D-04103 Leipzig, Germany, Tel.: +49-341-9718600, Fax: +49-341-9718609,
e-mail: regina.renner@medizin.uni-leipzig.de
*These authors contributed equally to this work.

Accepted for publication 9 March 2010

Abstract: The term ‘regenerative medicine’ refers to a new and review recent developments in regenerative medicine and their
expanding field in biomedical research that focuses on the potential relevance for dermatology with a particular emphasis on
development of innovative therapies allowing the body to replace, biomaterials, tissue engineering, skin substitutes and stem cell-
restore and regenerate damaged or diseased cells, tissues and based therapies for skin reconstitution in patients suffering from
organs. It combines several technological approaches including the chronic wounds and extensive burns.
use of soluble molecules, biomaterials, tissue engineering, gene
Key words: biomaterials – gene transfer – regenerative medicine –
therapy, stem cell transplantation and the reprogramming of cell
skin substitutes – stem cells – tissue engineering
and tissue types. Because of its easy accessibility, skin is becoming
an attractive model organ for regenerative medicine. Here, we

Please cite this paper as: Regenerative medicine in dermatology: biomaterials, tissue engineering, stem cells, gene transfer and beyond. Experimental Derma-
tology 2010; 19: 697–706.

character [also referred to induced pluripotent stem (iPS)


Introduction
cells], featuring a versatile growth and differentiation
Regenerative medicine is an emerging interdisciplinary field potential become increasingly important for fighting severe
of research and clinical application focussing on the repair, and yet incurable diseases. Stem cell therapies have already
replacement or regeneration of cells, tissue or organs to been demonstrated (in clinical trials or the laboratory) to
restore impaired function because of congenital defects, heal ischaemic heart diseases (3–5), auto immune diseases,
disease, trauma and ageing. It combines several technologi- like multiple sclerosis (6) and neurological diseases, like
cal approaches including, the use of soluble molecules, gene Parkinson’s disease and stroke (7,8). Table S1 shows a rep-
therapy, stem cell transplantation, tissue engineering and resentation of completed or pending human clinical trials
the reprogramming of cell and tissue types (Fig. 1) (1). of stem cell therapies, indexed by clinical application and
The earliest successful example of regenerative medicine source of stem cells used.
can be traced back to the late 1950s, when the idea of Skin is an attractive model organ to test novel concepts
reversing heart failure by transplanting a heart form one of regenerative medicine, with a particular emphasis on
individual to another had been realized, in the beginning skin regeneration for acute or chronic wounds. Chronic
in animal models. In 1967, the first heart transplantation in wounds present a worldwide growing health and economic
humans was carried out (2). Because of ethical concerns problem because of a steadily increasing number of
and the lack of donor organs as well as the risk of graft patients, high morbidity and risk of amputations, unsatis-
failure, scientists and clinicians now are pursuing a differ- factory results of existing therapies and heavy socio-
ent strategy in regenerative medicine. Instead of replacing economic burden (9). Tissue-engineered skin substitutes
whole organs, they intend to transplant biologically compe- represent an innovative therapeutical option for the treat-
tent cells and engineered tissues or to stimulate tissue-resi- ment of acute and chronic skin wounds. Bioengineered
dent stem cells to restore tissue or organ function. Both skin replacements are not only supposed to substitute the
adult stem cells and embryonic stem (ES) cells as well as major physiological functions by providing a rapid and
reprogrammed somatic cells with a multipotent, ES cell-like reliable cover of the defect but also should be easily appli-

ª 2010 John Wiley & Sons A/S, Experimental Dermatology, 19, 697–706 697
Dieckmann et al.

Soluble favour compared to other wound dressings [reviewed by


molecules
VEGF EGF
O¢Donnell et al. and by Dini et al. (12,13)], in particular
IGF
FGF VEGF products like Oasis (Healthpoint Ltd., Fortworth, Texas,
IGF
G FGF
EGF USA), Dermagraft (Advanced BioHealing, Westport, Con-
Stem cell
necticut, USA) and Apligraf (Organogenesis Inc., Canton,
therapy Biomaterials Massachusetts, USA). Tissue engineering usually requires an
Regenerative artificial extracellular matrix (ECM) allowing for infiltration
medicine of surrounding cells. For the generation of an artificial
ECM, both naturally occurring and synthetically manufac-
Gene
tured substances are processed. Examples of natural materi-
therapy als include polypeptides, hydroxyapatites, hyaluronan,
glycosaminoglycans (GAG), fibronectin, collagen, chitosan
and alginates. Because of their abundance in skin and their
recognition by cell surface receptors, these natural materials
display low toxicity and low chronic inflammatory response.
Figure 1. Research areas in regenerative medicine. Regenerative
medicine is an emerging multidisciplinary field of research and clinical
However, concerning their predominant xenogeneic origin,
applications focused on the repair, replacement or regeneration of they harbour an intrinsic risk for transmission of animal
tissue or organs. The approaches may include the use of soluble viruses (9,14).
molecules, gene therapy, stem cell therapy and biomaterials. Examples of synthetic, fully degradable materials include
polyglycolide, polylactide, polylactide-coglycolide, polytetra-
cable under routine use conditions and reduce pain and fluoroethylene, polycaprolactone and polyethylene tere-
discomfort for the patient. Furthermore, they should elicit phthalate, while polyurethane (PUR) represents a frequently
the regeneration process from the wound bed without used non-degradable substance. New technologies like three-
causing inflammation or rejection. Skin substitutes should dimensional printing and electrospinning emerged for accu-
be available immediately, and be non-toxic nor immuno- rate manufacture, creating scaffolds of both a defined shape
genic. From an aesthetic point of view, skin substitutes and pore sizes, facilitating infiltration of fibroblasts and
should be durable, elastic, with minimized scar formation, vascularization (10,14–19). A major drawback of synthetic
and pigmentation should resemble natural skin. Finally, yet materials is the lack of cell-recognition signals. An approach
importantly, the cost–benefit ratio has to be taken into enabling the repopulation and regeneration of a new natural
consideration (10,11). matrix consisted in the incorporation of adhesion peptides
In this review, we are surveying the existing bioengi- e.g. using RGD sequences (Arg-Gly-Asp) into biomatrices
neered skin substitutes that are already in clinical applica- (20,21). A so-called smart matrix is provided by biomaterials
tions, without claiming to be complete. Additionally, we or scaffolds capable of directing cell differentiation and
give an outlook on recent progress in the development metabolism and therefore accelerating tissue regeneration
of innovative approaches in skin reconstitution, by stem (11,22). For example, this can be accomplished via integra-
cell-based therapies. tion of polyethylene glycol (PEG) hydrogels into the scaffold.
PEG acts as an inert structural platform because of its hydro-
philicity and resistance to protein adsorption (23). The gel
Biomaterials as skin substitutes
can be modified by addition of cell anchoring points like
Here, we will discuss the key requirements of biomaterials RGD-containing peptides or networking functional domains
for the generation of synthetically engineered skin substi- linked to a hyaluronan backbone. Furthermore, the degree of
tutes. Biomaterials are acellular natural or synthetic sub- degradability by proteases can be influenced by linkage of
stances used for creation of the backbone of skin protease-sensitive oligopeptides (24–27).
substitutes in clinical applications. As the crucial role of growth factors for cell migration,
Skin substitutes composed of biomaterials can be used as proliferation, differentiation and differentiation in the
temporary wound cover for all thickness wounds, like wounded area has been recognized, efforts in tissue engi-
chronic ulcers or superficial and second-degree wounds neering have focused on the incorporation of growth fac-
prior to autologous skin grafting. They can be used as stim- tors like fibroblast growth factor, vascular endothelial
ulating agents for cell proliferation and angiogenesis or as growth factor (VEGF), insulin-like growth factor (IGF) and
permanent dermal replacement, according to the intended platelet-derived growth factor (PDGF) into matrix scaffolds
use and the individual constitution of the wound and the (28–30). Thus, the bioactivity of a polymer backbone can
patients general condition In a systematic review of ran- be augmented and the healing process markedly accelerated
domized controlled trials, most skin equivalents are in especially in chronic wounds (11,28). The challenge consists

698 ª 2010 John Wiley & Sons A/S, Experimental Dermatology, 19, 697–706
Skin substitution by tissue engineering

in the identification of the required factors and cytokines USA). Despite an overall rather negligible exposure, porcine
according to the current condition of the wound. In the matrices implicate the risk of transmission of prion disease
following, acellular epidermal and dermal skin substitutes and porcine retroviruses, a concern that needs to be
comprising biomaterials are discussed. addressed wherever xenogeneic material is used for skin
A currently used double-layered biosynthetic epidermal substitutes (9). An approach to resolve this problem con-
substitute consists of an outer silicone membrane and a knit- sists in the use of alloplastic material. The common scaffold
ted nylon mesh (Biobrane, Smith & Nephew Healthcare, of the commercially available alloplastic skin substitutes is a
London, UK). Both layers enclose a chemically cross-linked foil made of PUR that is coated with various substances.
porcine type 1 collagen forming a 3D structure that allows Epigard (Orthomed Medizintechnik GmbH, Wien,
rapid adherence to the wound surface and initiation of the Austria) possesses a teflon layer upside that renders the
wound healing. Indications for Biobrane include superfi- membrane permeable to air, but not to bacteria or wound
cial and second-degree burns and large epithelial defects secretion. If changes of dressing are performed regularly,
(14). A new product synthetically polymerized of the three removal of necrotic tissue and wound exudate as well as in-
components DL-lactidetrimethytencarbonate, trimethylen- growth of fibroblasts and vascular endothelium is facilitated
carbonate and e-caprolactone with a porous membrane is (39). Syspurderm (Paul Hartmann AG, Heidenheim,
called Suprathel. Hydrolytic degradation of this epidermal Germany) is a double-layered pad of flexible foam of PUR.
substitute occurs about 4 weeks after application. Principal The inner layer provides an open porous matrix for tissue
domains of Suprathel (PolyMedics Innovations GmbH, granulation, whereas the outer condensated surface serves
Denkendorf, Germany) are second-degree burns and donor as a barrier preventing secondary infection. Cell debris,
sites of skin transplantation. Promising advantages are a necrotic material and bacteria can also be easily removed
rapid painless re-epithelialization and the possibility of during wound dressing. In contrast, Lyomousse ⁄ Lyofoam
application next to utilized regions like joints (31). (Cosanum AG, Swiss) is composed of a hydrophilic porous
Being engineered already since 1981, Integra (Intergra- membrane that does not adhere to the wound ground
life Sciences, Plainsboro, New Jersey, USA) represents the and an outer hydrophobic surface ensuring bacterial imper-
oldest available dermal equivalent. A double-layered syn- meability. The mentioned alloplastic products represent a
thetic skin substitute is composed of a three-dimensional temporary solution for debrided wound beds until bio-
porous matrix of bovine collagen with about 10–15% logical skin substitutes can provide a permanent wound
chondroitin-6-sulphate derived from shark cartilage and an cover (40,41).
outer silicone sheet. After the infiltration of fibroblasts and A temporary cover can also be achieved by cadaver skin.
initiation of the vascularization process within 3 weeks, the Allografts function often as scaffolds and allow repopula-
application procedure requires removement of the silicone tion of the recipient’s endothelial and dermal cells. But
layer and wound covering with a sheet autograft (14,17,32– they are subjected to host rejection (17,42). A modification
34). Indications for Integra are treatment of full-thickness of human cadaver skin is Alloderm (LifeCell Corporation,
burns and correction of scars in joint proximity. However, Branchburg, New Jersey, USA). The immunologically inert
healing of keratinocyte autografts on Integra is limited acellular dermal matrix facilitates the regeneration of the
(about 60%) possibly because of an increased frequency of underlying dermis (17,36,43).
infections, and this material cannot be applied in persons
bearing a sensitization against bovine products (14,35).
Cellular skin substitutes
Matriderm (Skin & Health Care AG, Suwelack, Biller-
beck, Germany) is a porous and thin matrix composed of There are several cellular tissue-engineered skin substitutes
bovine type I, II and V collagen covered with bovine elastin currently on the market that can be characterized and
hydrolysate. Within 2 weeks after application, the matrix is distinguished by their origin: xenogeneic (from other spe-
degraded and replaced by the recipients own collagen. cies), allogeneic (from a non-genetically identical individual
Depending on the thickness of the sheet used for applica- of the same species), autologous (from the patient itself)
tion, a one-step procedure is possible, however, sheets of and syngeneic grafts (from a genetic identical individual
2 mm thickness and above should be used only in suffi- like monozygotic twins). Xenogeneic skin grafts bear the
ciently vascularized wound beds during an interval of 7 days intrinsic risk of transferring prion diseases or porcine retro-
before transplantation of split thickness autografts (36–38). viruses. In particular, the use of porcine tissue in humans
Convincing results in the treatment of patients with can evoke an immune response against the porcine mem-
chronic venous leg ulcers or superficial and second-degree brane glycoprotein (GAL epitope), which can cause acute
burns, respectively, have been received using porcine small rejection of the donated skin. It is absolutely necessary
intestinal submucosa acellular collagen matrix (Oasis) or that this membrane protein is removed in the final skin
porcine skin (Permacol, Covidien, Norwalk, Connecticut, product (44). Allogeneic transplants offer the possibility of

ª 2010 John Wiley & Sons A/S, Experimental Dermatology, 19, 697–706 699
Dieckmann et al.

large prefabrication and cryo-preservation mostly with mal sheets to more complex bilayered cutaneous constructs.
comparable results in regard to vitality or effectivity of Mainly, dermal constructs are created with or without a tem-
fresh allografts (45,46). Furthermore, they allow for porary synthetic epidermis. The dermal cellular component
repeated applications. However, the keratinocytes within is composed either of autologous dermal fibroblasts or of
these allografts are being replaced within a few weeks by allogeneic neonatal foreskin fibroblasts, as they are more
the infiltrating recipient cells and sometimes fail to produce responsive to mitogens than adult cells (60). The scaffold for
a satisfactory result in full-thickness wounds (47,48). the cells consists mostly of biocompatible and biodegradable
The manufactured skin substitutes can further be distin- materials, like benzyl-esterified derivatives of hyaluronic acid
guished by the different dermal components they replace: (Hyaff-11) (Hyalograft 3D, Fidia farmaceutici S.p.A.,
There are (i) epidermal equivalents, (ii) dermal equivalents Abano Terme (PD), Italy), polyglycolic acid or polyglactin
and (iii) composite substitutes. (Dermagraft, Advanced BioHealing, Westport, Connecti-
cut, USA). TransCyte (formerly Dermagraft-TC, Advanced
Epidermal equivalents BioHealing, Westport, Connecticut, USA) is a comparable
Epidermal grafts consist of keratinocytes that are differenti- product containing a nylon mesh coated with porcine der-
ated in vitro building a stratified epidermal layer. They can mal collagen that is seeded with newborn human fibroblasts
be combined with other biocompatible substrates (bovine and fixed to an outer silicone membrane (61,62). The major
collagen, hyaluronic acid), acellular natural human or indication of application consists in temporary wound cover
porcine materials, nylon or polyglactin meshes. The first in surgically excised full-thickness and partial-thickness
commercialized cultured epidermal autograft (Epicel (Gen- burns. One of the most successfully bioengineered products
zyme Corporate Offices, Cambridge, Massachusettts, USA)) is Dermagraft, an allogenic human neonatal-derived der-
is composed of autologous keratinocytes grown in vitro mal fibroblast culture, grown on a biodegradable scaffold,
in the presence of proliferation-arrested, murine (Swiss is able to produce several growth factors, to stimulate angio-
3T3 ⁄ J2) fibroblasts (49). It is currently used as cover for full- genesis, tissue expansion and re-epithelialization from the
thickness burns in the USA and Europe. Another epidermal wound edge, even after cryopreservation and thawing
product is represented by EpiDex (Euroderm GmbH, Leip- (63,64). This seems to be of particular advantage in diabetic
zig, Germany), which is manufactured from autologous foot ulcers as shown in various randomized controlled trials
outer root sheath (ORS) cells of plucked hair follicles for the (63). However, Dermagraft turned out to be also as safe
treatment of chronic venous leg ulcers (Fig. S1) (50–53). and as efficacious as allograft in burns (14). Furthermore, it
The proliferation potential of the ORS derived keratinocytes can be used to provide a dermal matrix that helps facilitating
is not restricted by the age of the recipient, enabling a suc- re-epithelization by the patient’s own keratinocytes (17).
cessful treatment also of elderly patients (54). To improve In clinical trials, treatment of venous leg ulcers with Der-
mechanical stability some epidermal skin substitutes are magraft in combination with compression therapy was
combined with a polyvinyl chloride polymer coated with a superior to compression therapy alone. Total ulcer area rate
plasma-polymerized surface (Altrika Ltd., Sheffield, UK) or of healing and linear rate of healing was significantly
with a perforated hyaluronic acid membrane (Laserskin, improved in patients treated with Dermagraft, they also
Fidia farmaceutici S.p.A., Abano Terme (PD), Italy) (55). had a greater increase in periulcer skin perfusion but this
Another administration strategy is pursued by BioSeed-S was not statistically significant (65).
(not available, formerly by BioTissue Technologies GmbH,
Freiburg, Germany). Autologous keratinocytes are propa- Composite substitutes
gated in vitro and then suspended within a gel-like fibrin Composite skin equivalents are defined by epidermal cells
adhesive (56,57). The gel-like skin graft is applied to the growing on fibroblast-containing dermal substitutes. They
patient’s wound using a syringe. The fibrin adhesive fixes the have been demonstrated to provide benefit in chronic ulcers
cells to the wound bed and allows a better in-growth (56,57). (16). The additional dermal component reduces wound
Although the product is approved to treat chronic leg ulcers, contraction, provides better mechanical stability and
it is not commercially available at present. The clinical out- reduces the time necessary for self-assembly of host-own
come of the epidermal transplants is not always satisfying granulation tissue. Furthermore, production and long-
which can be partially explained by the absence of a dermal distance transport are facilitated using composites. On the
component and of an adequate support of the undifferenti- other hand, collagen-based dermal substitutes promote ter-
ated keratinocytes (58,59). minal differentiation and apoptosis of fibroblasts as well as
of keratinocytes (9,66,67), and high-density fibrin carriers
Dermal substitutes have anti-migratory and survival-compromising effects (57).
The development of cutaneous substitutes by tissue engi- PermaDerm (previously known as Cincinnati skin
neering has evolved from simple cultured autologous epider- substitute, Calibrex Bioscience, Walkersville, Maryland,

700 ª 2010 John Wiley & Sons A/S, Experimental Dermatology, 19, 697–706
Skin substitution by tissue engineering

USA) is an example of collagen-based autologous dermo- potent stem cells to replace damaged tissue in patients with
epidermal skin substitutes. The dermal component is compromised wound healing (50,51,79,80). Two distinct
composed of type I bovine collagen seeded with human subpopulations of epidermal stem cells occur in the skin: a
fibroblasts and an overlying epidermal layer of human basal keratinocyte population found in the interfollicular
keratinocytes. Similar to OrCel (Forticell Bioscience, epithelium and stem cells residing in the bulge region of the
formerly Ortec International Inc., New York, USA), it is hair follicle (81,82).
applied for the permanent skin replacement in severe Epithelial keratinocytes are applied today with success in
burn patients (33,68). skin-grafting technologies, e.g. burn patients routinely have
Apligraf (formerly Graftskin) is an allogeneic dermo- cultured skin keratinocytes engrafted (68,83–85). They are
epidermal product consisting of cultivated keratinocytes isolated from skin biopsy and plated onto a mitotically
and a dermal layer of fibroblasts on a collagen-type-I- inactivated and lethally irradiated layer of murine fibro-
matrix. As it does not contain any antigen-presenting cells blasts. The feeder layer supports clonal expansion of prolif-
such as Langerhans cells, dermal dendritic cells, endothelial erative epithelial cells. Changing the culture conditions to a
cells, melanocytes or inflammatory cells, such as leucocytes, higher calcium level subsequently supports the partial dif-
it is thought to be immunologically inert. It has been ferentiation and stratification of the cells (49,86,87). How-
approved for the treatment of venous and diabetic ulcers ever, several studies have demonstrated the superiority of
(69–71). Clinical trials showed that the healing rate of the progenitor cells over more differentiated keratinocytes in
ulcers treated with Apligraf (63% vs. 49% control) as well the generation of tissue-engineered skin (88–90). High a6
as the healing time (61 vs. 181 days in the control group) integrin expression (a6bri) and low expression of the trans-
was improved (72). The mechanism of action in the pro- ferrin receptor (CD71dim) is the most accepted combina-
motion of wound healing is not exactly clear. First of all, it tion of epidermal stem cell markers (91–93). Studies
behaves as physical wound cover but secondary, it is sup- analysing the regenerative capacity of these epidermal stem
posed to induce the production of a number of cytokines cell populations revealed that skin equivalents created from
and growth factors (73–75) which seems to be responsible a6bri ⁄ CD71dim keratinocytes give rise to a stratified and
for stimulation of differentiation and proliferation of the thick epidermis, while skin equivalents from a6bri ⁄ CD71bri
surrounding fibroblasts and keratinocytes. Healing occurs keratinocytes produced a thin and less well-differentiated
with less fibrosis which might be because of the construc- epidermis (93). However, several other cell surface proteins
tion of the graft itself containing neonatal cells, stimulating like high expression of b1 integrin (94), the ability to
a more foetal-like scarless wound healing (76). Clinical tri- exclude Hoechst 33342 dye (95–97), expression of phos-
als demonstrated that ulcers treated with OrCel were heal- pho-glycoproteins (98), CD200 (99,100), frizzled homo-
ing better in comparison with standard care (77,78). Yet, logue 1 (FZD1) (99), p63, a homologue of p53 tumor
the major drawbacks of dermo-epidermal substitutes are suppressor gene) (101), Keratin 19 (102), Keratin 15 (103–
the technically challenging and time-consuming two-step 105) or b-catenin (106) have been described recently for
production and early clinical failures because of a delayed epidermal stem cells. However, the identification of one or
vascularization of the wound bed or because of their com- rather a combination of specific epidermal stem cells mark-
ponents from xenogenic origin. ers is mandatory, to achieve a pure cell population.
Table S2 summarizes skin substitutes that are or have The second reservoir for skin stem cells is the bulge
been available in the past for the treatment of skin injuries region of the ORS of hair follicles (107–111). Under physi-
in humans. ological conditions, these stem cells regenerate the hair
bulb, while after injury they can also regenerate the seba-
ceous gland and the epidermis (112,113). Stem cells from
The potential of stem cells for skin
hair follicles could be differentiated experimentally into
replacement
neurons, glia cells, keratinocytes, smooth muscle cells and
The use of stem cells as the basic material for skin engineer- melanocytes, indicating that the hair follicle contains stem
ing offers the potential to improve significantly the clinical cell populations of both neuroectodermal and mesodermal
outcome, both in wound healing and in gene therapeutic origin (89,100,114–118).
approaches. Cell-based therapies with adult stem cells, ES Because of their multiple differentiation capacity and the
cells or reprogrammed somatic cells, respectively, are attrac- facile accessibility hair follicle stem cells offer an attractive
tive, particularly if used in autologous transplantation regi- source of stem cells for skin substitution. However, the
mens, as inherent problems with rejections of the transplant issue of a reliable and specific identification and discrimi-
do not exist, and ethical and moral objections are avoided. nation of the various stem cell populations has not been
Recent studies on tissue-engineered skin indicated that epi- solved yet properly. Additionally, because of the limited
dermal stem cells might provide a superior source of multi- number of cells within the interfollicular epithelium and

ª 2010 John Wiley & Sons A/S, Experimental Dermatology, 19, 697–706 701
Dieckmann et al.

the ORS of the hair follicle, further investigations are also can be potentially applied in skin replacement strategies.
needed for determination of appropriate culture conditions ASCs are a population of pluripotent cells, which have
to prevent stem cell loss. characteristics similar to bone marrow-derived MSCs (146–
Another fascinating application for epidermal stem cells 148). Generally, ASCs are isolated from lipoaspirates,
might be their use for gene therapy approaches. Consider- obtained by suction-assisted lipectomy (147,149,150). After
ing that the epidermis is a self-renewing tissue, any perma- extensive washing, removal of the red cells and enzymatic
nent genetic correction must be aimed at the stem cell break-down of the ECM, the resulting cells are known as
population, to achieve long-lasting therapeutical effects. stromal vascular fraction (SVF). SVF contains mesenchymal
Particularly, a gene therapy approach is perfectly eligible stromal cells, but also other cells, like endothelial cells,
for the treatment of diseases involving recessive loss-of- smooth muscle cells, pericytes, fibroblasts, leucocytes and
function mutations, as it occurs in junctional or recessive hematopoietic stem cells (151). Finally, from this mixed
dystrophic epidermolysis bullosa (EB) and other recessive cell population, the selection for ASCs occurs simply by
genetic skin disorders like X-linked ichthyosis or xeroderma their ability to adhere to plastic ware. (152,153). Some
pigmentosus. The re-introduction of the wild-type gene via researchers have performed additionally purification by
viral or non-viral insertion is sufficient for correction of the magnetic bead coupling to deplete cells of the hematopoi-
phenotype. In a striking and pioneering clinical trial, a team etic and of the endothelial lineages. ASCs are capable of
of researchers and clinicians demonstrated for the first time differentiating into other mesenchymal tissue types, like
that cutaneous gene therapy has the potential to cure a adipocytes, chondrocytes, myocytes and osteoblasts and
patient with inherited EB. The treatment relied on ex vivo show angiogenic properties (147,148,154–156). They seem
transduction of autologous epidermal stem cells with a nor- also to be applicable for skin regeneration, because in
mal copy of the defective gene, here laminin beta-3 chain, experimental wound models ASCs accelerated wound clo-
followed by generation of epithelial sheets from these genet- sure by improved re-epithelization and angiogenesis (157–
ically modified cells. The transplantation of the epidermal 162). In some of these studies, the stem cells had been
grafts led to functional correction of the disease (119). delivered via a human acellular dermal matrix or a matrix
Mesenchymal stem cells (MSCs; also referred to as mes- composed of atelocollagen or silk fibroin-chitosan. It could
enchymal stromal cells) either from bone marrow or adi- be shown, that these ASCs persisted locally and did not
pose tissue present further suitable candidates for cell distribute systemically, and by this providing anatomically
therapy. Bone marrow MSCs (BM-MSCs) are a heteroge- directed support to tissue regeneration at the desired site of
neous group of multipotent progenitor cells with the surgical engraftment (158–161).
capacity of self-renewal and differentiation into cells with These observations are particularly promising, because
mesodermal, ectodermal and endodermal characteristics BM-MSCs can only be obtained in a limited amount and
(120–128). They have the intrinsic capacity to leave the their differentiation abilities decrease with age. Addition-
bone marrow, circulate in the blood and home to injured ally, the bone marrow procurement is extremely painful for
tissues (129–133). Not surprisingly, they have also the patients. In contrast, adipose tissue is ubiquitous and cells
capability of accelerating cutaneous wound regeneration are easily obtainable in adequate quantities with little
both in animal models and in humans (134–137). Several patient discomfort. Therefore, ASCs may provide a superior
reports on the clinical application of BM-MSCs in wound source of stem cells for skin regeneration purposes
therapies have clearly revealed that grafted MSCs facilitate (155,157,161).
skin regeneration, both in acute and in chronic wounds Despite of the eminent potential of MSCs in tissue
(138–142). For example, Badiavas et al. (138) could dem- engineering, unintentional side effects in connection with
onstrate that autologous BM-MSCs delivered in a fibrin MSCs engrafting, like their immunosuppressive properties,
spray accelerate healing in human cutaneous wounds. have to be taken into consideration before these cells will
However, the underlying mechanisms of BM-MSCs on skin be utilized in clinical applications (163–165). And very
healing are multiple and have not been clarified in detail importantly is a distinct molecular characterization of the
yet, but include cell differentiation, wound contraction, cells, e.g. appropriate marker proteins to allow for (i) the
release of proangiogenic factors and production and main- selection of cells or cell types, respectively, that are most
tenance of the ECM (137,143–145). beneficial to wound healing and (ii) the standardization of
BM-MSCs are usually isolated from the mononuclear technique improving the clinical usefulness.
layer of bone marrow after separation by density gradient Since their discovery in 1998 (166), human ES cells have
centrifugation and are achieved through expansion of plas- been recognized for their regenerative properties and viewed
tic-adherent cells. as a potential application in tissue regeneration. However,
In recent years, in analogy to BM-MSCs, researchers are their use in clinical application is limited by ethical and
investigating whether also adipose-derived stem cells (ASC) moral objections and the risk of graft rejection reactions or

702 ª 2010 John Wiley & Sons A/S, Experimental Dermatology, 19, 697–706
Skin substitution by tissue engineering

teratoma formation (167–169). At least the first two prob- and stem cell-targeted cutaneous gene transfer will be
lems can be prevented by the use of reprogrammed somatic essential for the successful gene therapy.
cells that can be autologously derived. Several scientists have In conclusion, already today regenerative medicine has
succeeded in generating iPS cells from adult human dermal proven to have a tremendous potential in dermatological
fibroblasts and from keratinocytes by transduction with a applications that will increase in the future, as the interdis-
combination of various transcription factors, involved in ciplinary research on stem cell biology, biomaterial science,
reprogramming. These cells were phenotypically and func- gene therapy and tissue engineering will progress.
tionally indistinguishable from ES cells (170–174). These
cells could potentially be used in the construction of tissue- Acknowledgement
engineered skin, after considerations of potential tumorge- This work was supported by funding from the German Federal Ministry of
nicity caused by cell regulations and genetic manipulations Education and Research (BMBF, PtJ-Bio 0313909 to C.D. and L.M.) and
have been sorted out. by the German Research Council (DFG, TRR-SFB 67, TP B3, http://
Table S3 summarizes the differentiation potential of fol- www.trr67.de to JCS).
licular epidermal stem cells, stem cells from the bulge
region, bone marrow-derived MSCs, adipose stem cells, ES References
cells and iPS-cells. 1 Greenwood H L, Singer P A, Downey G P, Martin D K, Thorsteinsdottir H,
Daar A S. Regenerative medicine and the developing world. PLoS Med 2006:
3: e381.
2 Chien K R. Regenerative medicine and human models of human disease. Nat-
Conclusion ure 2008: 453: 302–305.
3 Abdel-Latif A, Bolli R, Tleyjeh I M et al. Adult bone marrow-derived cells for
New progress in stem cell technologies has extended greatly cardiac repair: a systematic review and meta-analysis. Arch Intern Med 2007:
167: 989–997.
our knowledge about the function and relevance of adult 4 Schachinger V, Tonn T, Dimmeler S, Zeiher A M. Bone-marrow-derived pro-
genitor cell therapy in need of proof of concept: design of the REPAIR-AMI
stem cells for tissue development and tissue regeneration. trial. Nat Clin Pract Cardiovasc Med 2006: 3 (Suppl 1): S23–S28.
Additionally, new technologies in material sciences are 5 Lipinski M J, Biondi-Zoccai G G, Abbate A et al. Impact of intracoronary cell
therapy on left ventricular function in the setting of acute myocardial infarc-
emerging for the accurate manufacture of degradable poly- tion: a collaborative systematic review and meta-analysis of controlled clinical
mers, creating materials with defined and customized trials. J Am Coll Cardiol 2007: 50: 1761–1767.
6 Schippling S, Heesen C, Zander A, Martin R. Stem cell transplantation in mul-
mechanical properties for the repair or replacement of tiple sclerosis. J Neurol 2008: 255 (Suppl 6): 43–47.
7 Lindvall O, Kokaia Z, Martinez-Serrano A. Stem cell therapy for human neuro-
impaired tissue or organs. Particularly in dermatology, degenerative disorders-how to make it work. Nat Med 2004: 10 (Suppl):
numerous developments in the field of tissue engineering S42–S50.
8 Lindvall O, Kokaia Z. Stem cells for the treatment of neurological disorders.
have been translated already into therapeutical applications, Nature 2006: 441: 1094–1096.
like for the treatment of extensive chronic wounds, e.g. skin 9 Clark R A, Ghosh K, Tonnesen M G. Tissue engineering for cutaneous
wounds. J Invest Dermatol 2007: 127: 1018–1029.
burns, venous or diabetic leg ulcers. Initial success in treat- 10 Ikada Y. Challenges in tissue engineering. J R Soc Interface 2006: 3: 589–
601.
ing recessive genetic skin disorders by a combined approach 11 Metcalfe A D, Ferguson M W. Tissue engineering of replacement skin: the
using gene therapy and tissue engineering indicates clearly crossroads of biomaterials, wound healing, embryonic development, stem
cells and regeneration. J R Soc Interface 2007: 4: 413–437.
that skin is an appropriate target for innovative therapeutic 12 O’Donnell T F Jr, Lau J. A systematic review of randomized controlled trials of
strategies in regenerative medicine. Despite of all this scien- wound dressings for chronic venous ulcer. J Vasc Surg 2006: 44: 1118–1125.
13 Dini V, Romanelli M, Piaggesi A, Stefani A, Mosca F. Cutaneous tissue engi-
tific progress, the skin substitutes used at present are yet neering and lower extremity wounds (part 2). Int J Low Extrem Wounds
2006: 5: 27–34.
not fully functional as they lack differentiated structures 14 Pham C, Greenwood J, Cleland H, Woodruff P, Maddern G. Bioengineered
as nerves, sweat glands, pilosebaceous structure and blood skin substitutes for the management of burns: a systematic review. Burns
2007: 33: 946–957.
supply. Still, intensive research has to proceed to attain a 15 Metcalfe A D, Ferguson M W. Skin stem and progenitor cells: using regenera-
durable and cosmetically acceptable tissue-engineered skin. tion as a tissue-engineering strategy. Cell Mol Life Sci 2008: 65: 24–32.
16 Li M, Guo Y, Wei Y, MacDiarmid A G, Lelkes P I. Electrospinning polyaniline-
The choice of suitable cells in sufficient quantity and purity, contained gelatin nanofibers for tissue engineering applications. Biomaterials
2006: 27: 2705–2715.
adequate culture conditions to maintain stem cell proper- 17 Supp D M, Boyce S T. Engineered skin substitutes: practices and potentials.
ties, as well as appropriate biomaterials for transient or per- Clin Dermatol 2005: 23: 403–412.
18 Griffith L G, Swartz M A. Capturing complex 3D tissue physiology in vitro.
manent replacement are factors important for long-lasting Nat Rev Mol Cell Biol 2006: 7: 211–224.
engraftment of a regenerated skin. 19 Luu Y K, Kim K, Hsiao B S, Chu B, Hadjiargyrou M. Development of a nano-
structured DNA delivery scaffold via electrospinning of PLGA and PLA-PEG
The tissue-engineered skin of the future will resemble block copolymers. J Control Release 2003: 89: 341–353.
20 Li B, Chen J, Wang J H. RGD peptide-conjugated poly(dimethylsiloxane)
morphologically and functionally natural skin. Epidermal promotes adhesion, proliferation, and collagen secretion of human fibro-
stem cells will be utilized for generation of the epidermal blasts. J Biomed Mater Res A 2006: 79: 989–998.
21 Fittkau M H, Zilla P, Bezuidenhout D et al. The selective modulation of endo-
component, probably along with the addition of melanocy- thelial cell mobility on RGD peptide containing surfaces by YIGSR peptides.
tic stem cells. For the dermal construct, endothelial, mesen- Biomaterials 2005: 26: 167–174.
22 Stoop R. Smart biomaterials for tissue engineering of cartilage. Injury 2008:
chymal and neuronal stem cells may support the 39 (Suppl 1): S77–S87.
23 Desai N P, Hubbell J A. Solution technique to incorporate polyethylene oxide
generation of the dermal component with a functional vas- and other water-soluble polymers into surfaces of polymeric biomaterials.
culature. Furthermore, improving tissue-engineered skin Biomaterials 1991: 12: 144–153.

ª 2010 John Wiley & Sons A/S, Experimental Dermatology, 19, 697–706 703
Dieckmann et al.

24 van de Wetering P, Metters A T, Schoenmakers R G, Hubbell J A. Poly(ethyl- 53 Renner R, Harth W, Simon J C. Transplantation of chronic wounds with epi-
ene glycol) hydrogels formed by conjugate addition with controllable swell- dermal sheets derived from autologous hair follicles–the Leipzig experience.
ing, degradation, and release of pharmaceutically active proteins. J Control Int Wound J 2009: 6: 226–232.
Release 2005: 102: 619–627. 54 Limat A, Mauri D, Hunziker T. Successful treatment of chronic leg ulcers with
25 Raeber G P, Lutolf M P, Hubbell J A. Molecularly engineered PEG hydrogels: epidermal equivalents generated from cultured autologous outer root sheath
a novel model system for proteolytically mediated cell migration. Biophys J cells. J Invest Dermatol 1996: 107: 128–135.
2005: 89: 1374–1388. 55 Lam P K, Chan E S, To E W, Lau C H, Yen S C, King W W. Development and
26 Rizzi S C, Ehrbar M, Halstenberg S et al. Recombinant protein-co-PEG net- evaluation of a new composite Laserskin graft. J Trauma 1999: 47: 918–922.
works as cell-adhesive and proteolytically degradable hydrogel matrixes. Part 56 Vanscheidt W, Ukat A, Horak V et al. Treatment of recalcitrant venous leg
II: biofunctional characteristics. Biomacromolecules 2006: 7: 3019–3029. ulcers with autologous keratinocytes in fibrin sealant: a multinational ran-
27 Seliktar D, Zisch A H, Lutolf M P, Wrana J L, Hubbell J A. MMP-2 sensitive, domized controlled clinical trial. Wound Repair Regen 2007: 15: 308–315.
VEGF-bearing bioactive hydrogels for promotion of vascular healing. J Biomed 57 Hartmann A, Quist J, Hamm H, Brocker E B, Friedl P. Transplantation of
Mater Res A 2004: 68: 704–716. autologous keratinocyte suspension in fibrin matrix to chronic venous leg
28 Macri L, Silverstein D, Clark R A. Growth factor binding to the pericellular ulcers: improved long-term healing after removal of the fibrin carrier. Derma-
matrix and its importance in tissue engineering. Adv Drug Deliv Rev 2007: tol Surg 2008: 34: 922–929.
59: 1366–1381. 58 Kangesu T, Navsaria H A, Manek S, Fryer P R, Leigh I M, Green C J. Kerato-
29 Davidson J M. First-class delivery: getting growth factors to their destination. dermal grafts: the importance of dermis for the in vivo growth of cultured
J Invest Dermatol 2008: 128: 1360–1362. keratinocytes. Br J Plast Surg 1993: 46: 401–409.
30 Branski L K, Pereira C T, Herndon D N, Jeschke M G. Gene therapy in 59 Myers S R, Partha V N, Soranzo C, Price R D, Navsaria H A. Hyalomatrix: a
wound healing: present status and future directions. Gene Ther 2007: 14: temporary epidermal barrier, hyaluronan delivery, and neodermis induction
1–10. system for keratinocyte stem cell therapy. Tissue Eng 2007: 13: 2733–2741.
31 Schwarze H, Kuntscher M, Uhlig C et al. Suprathel, a new skin substitute, in 60 Phillips T J. New skin for old: developments in biological skin substitutes. Arch
the management of donor sites of split-thickness skin grafts: results of a clini- Dermatol 1998: 134: 344–349.
cal study. Burns 2007: 33: 850–854. 61 Kumar R J, Kimble R M, Boots R, Pegg S P. Treatment of partial-thickness
32 Helgeson M D, Potter B K, Evans K N, Shawen S B. Bioartificial dermal substi- burns: a prospective, randomized trial using Transcyte. ANZ J Surg 2004: 74:
tute: a preliminary report on its use for the management of complex combat- 622–626.
related soft tissue wounds. J Orthop Trauma 2007: 21: 394–399. 62 Noordenbos J, Dore C, Hansbrough J F. Safety and efficacy of TransCyte for
33 MacNeil S. Progress and opportunities for tissue-engineered skin. Nature the treatment of partial-thickness burns. J Burn Care Rehabil 1999: 20: 275–
2007: 445: 874–880. 281.
34 Burke J F, Yannas I V, Quinby W C Jr, Bondoc C C, Jung W K. Successful use 63 Marston W A. Dermagraft, a bioengineered human dermal equivalent for the
of a physiologically acceptable artificial skin in the treatment of extensive treatment of chronic nonhealing diabetic foot ulcer. Expert Rev Med Devices
burn injury. Ann Surg 1981: 194: 413–428. 2004: 1: 21–31.
35 Muangman P, Deubner H, Honari S et al. Correlation of clinical outcome of 64 Mansbridge J. Commercial considerations in tissue engineering. J Anat 2006:
integra application with microbiologic and pathological biopsies. J Trauma 209: 527–532.
2006: 61: 1212–1217. 65 Omar A A, Mavor A I, Jones A M, Homer-Vanniasinkam S. Treatment of
36 Kamolz L P, Lumenta D B, Kitzinger H B, Frey M. Aktueller Stand und venous leg ulcers with Dermagraft. Eur J Vasc Endovasc Surg 2004: 27: 666–
Möglichkeiten der Wundbehandlung durch Tissue engineering. Eur Surg 672.
2008: 40: 19–26. 66 Fluck J, Querfeld C, Cremer A, Niland S, Krieg T, Sollberg S. Normal human
37 Ryssel H, Gazyakan E, Germann G, Ohlbauer M. The use of MatriDerm in primary fibroblasts undergo apoptosis in three-dimensional contractile colla-
early excision and simultaneous autologous skin grafting in burns–a pilot gen gels. J Invest Dermatol 1998: 110: 153–157.
study. Burns 2008: 34: 93–97. 67 Tuan T L, Song A, Chang S, Younai S, Nimni M E. In vitro fibroplasia: matrix
38 Wetzig T, Gebhardt C, Simon J C. New indications for artificial collagen-elas- contraction, cell growth, and collagen production of fibroblasts cultured in
tin matrices? Covering exposed tendons. Dermatology 2009: 219: 272–273. fibrin gels. Exp Cell Res 1996: 223: 127–134.
39 Alexander J W, Wheeler L M, Rooney R C, McDonald J J, MacMillan B G. 68 Boyce S T, Kagan R J, Greenhalgh D G et al. Cultured skin substitutes reduce
Clinical evaluation of Epigard, a new synthetic substitute for homograft and requirements for harvesting of skin autograft for closure of excised, full-thick-
heterograft skin. J Trauma 1973: 13: 374–383. ness burns. J Trauma 2006: 60: 821–829.
40 Turner T D. Dressings in wound management. In: Swarbrick J, Boylan J C, 69 Wong T, McGrath J A, Navsaria H. The role of fibroblasts in tissue engineer-
(Eds.) Encyclopedia of Pharmaceutical Technology (New York, Basel, Hong ing and regeneration. Br J Dermatol 2007: 156: 1149–1155.
Kong: Marcel Dekker, Inc.) 1991: 283–301. 70 Curran M P, Plosker G L. Bilayered bioengineered skin substitute (Apligraf): a
41 Rosdy M, Clauss L C. Cytotoxicity testing of wound dressings using normal review of its use in the treatment of venous leg ulcers and diabetic foot
human keratinocytes in culture. J Biomed Mater Res 1990: 24: 363–377. ulcers. BioDrugs 2002: 16: 439–455.
42 Snyder R J. Treatment of nonhealing ulcers with allografts. Clin Dermatol 71 Falanga V, Sabolinski M. A bilayered living skin construct (APLIGRAF) acceler-
2005: 23: 388–395. ates complete closure of hard-to-heal venous ulcers. Wound Repair Regen
43 Wainwright D, Madden M, Luterman A et al. Clinical evaluation of an acellu- 1999: 7: 201–207.
lar allograft dermal matrix in full-thickness burns. J Burn Care Rehabil 1996: 72 Falanga V, Margolis D, Alvarez O et al. Rapid healing of venous ulcers and
17: 124–136. lack of clinical rejection with an allogeneic cultured human skin equivalent.
44 McPherson T B, Liang H, Record R D, Badylak S F. Galalpha(1,3)Gal epitope Human Skin Equivalent Investigators Group. Arch Dermatol 1998: 134: 293–
in porcine small intestinal submucosa. Tissue Eng 2000: 6: 233–239. 300.
45 Teepe R G, Koebrugge E J, Ponec M, Vermeer B J. Fresh versus cryopreserved 73 Streit M, Braathen L R. Apligraf–a living human skin equivalent for the treat-
cultured allografts for the treatment of chronic skin ulcers. Br J Dermatol ment of chronic wounds. Int J Artif Organs 2000: 23: 831–833.
1990: 122: 81–89. 74 Falanga V, Isaacs C, Paquette D et al. Wounding of bioengineered skin: cellu-
46 Gibbs S, van den Hoogenband H M, Kirtschig G et al. Autologous full-thick- lar and molecular aspects after injury. J Invest Dermatol 2002: 119: 653–660.
ness skin substitute for healing chronic wounds. Br J Dermatol 2006: 155: 75 Brem H, Young J, Tomic-Canic M, Isaacs C, Ehrlich H P. Clinical efficacy and
267–274. mechanism of bilayered living human skin equivalent (HSE) in treatment of
47 Oliver A M, Kaawach W, Mithoff E W, Watt A, Abramovich D R, Rayner diabetic foot ulcers. Surg Technol Int 2003: 11: 23–31.
C R. The differentiation and proliferation of newly formed epidermis on 76 Zaulyanov L, Kirsner R S. A review of a bi-layered living cell treatment (Aplig-
wounds treated with cultured epithelial allografts. Br J Dermatol 1991: 125: raf) in the treatment of venous leg ulcers and diabetic foot ulcers. Clin Interv
147–154. Aging 2007: 2: 93–98.
48 Williamson J S, Snelling C F, Clugston P, Macdonald I B, Germann E. Cultured 77 Lipkin S, Chaikof E, Isseroff Z, Silverstein P. Effectiveness of Bilayered Cellular
epithelial autograft: five years of clinical experience with twenty-eight Matrix in Healing of Neuropathic Diabetic Foot Ulcers: results of a Multicenter
patients. J Trauma 1995: 39: 309–319. Pilot Trial. Wounds 2003: 15: 230–236.
49 Wright K A, Nadire K B, Busto P, Tubo R, McPherson J M, Wentworth B M. 78 Still J, Glat P, Silverstein P, Griswold J, Mozingo D. The use of a collagen
Alternative delivery of keratinocytes using a polyurethane membrane and the sponge ⁄ living cell composite material to treat donor sites in burn patients.
implications for its use in the treatment of full-thickness burn injury. Burns Burns 2003: 29: 837–841.
1998: 24: 7–17. 79 Pellegrini G, Ranno R, Stracuzzi G et al. The control of epidermal stem cells
50 Hunziker T, Limat A. Cultured keratinocyte grafts. Curr Probl Dermatol 1999: (holoclones) in the treatment of massive full-thickness burns with autologous
27: 57–64. keratinocytes cultured on fibrin. Transplantation 1999: 68: 868–879.
51 Limat A, Hunziker T. Use of epidermal equivalents generated from follicular 80 Lau K, Paus R, Tiede S, Day P, Bayat A. Exploring the role of stem cells in
outer root sheath cells in vitro and for autologous grafting of chronic cutaneous wound healing. Exp Dermatol 2009: 18: 921–933.
wounds. Cells Tissues Organs 2002: 172: 79–85. 81 Blanpain C, Lowry W E, Geoghegan A, Polak L, Fuchs E. Self-renewal, multi-
52 Tausche A K, Skaria M, Bohlen L et al. An autologous epidermal equivalent potency, and the existence of two cell populations within an epithelial stem
tissue-engineered from follicular outer root sheath keratinocytes is as effec- cell niche. Cell 2004: 118: 635–648.
tive as split-thickness skin autograft in recalcitrant vascular leg ulcers. Wound 82 Blanpain C, Fuchs E. Epidermal stem cells of the skin. Annu Rev Cell Dev Biol
Repair Regen 2003: 11: 248–252. 2006: 22: 339–373.

704 ª 2010 John Wiley & Sons A/S, Experimental Dermatology, 19, 697–706
Skin substitution by tissue engineering

83 Rheinwald J G, Green H. Serial cultivation of strains of human epidermal 113 Ito M, Liu Y, Yang Z et al. Stem cells in the hair follicle bulge contribute to
keratinocytes: the formation of keratinizing colonies from single cells. Cell wound repair but not to homeostasis of the epidermis. Nat Med 2005: 11:
1975: 6: 331–343. 1351–1354.
84 Boyce S T, Goretsky M J, Greenhalgh D G, Kagan R J, Rieman M T, Warden G 114 Sieber-Blum M, Hu Y. Epidermal neural crest stem cells (EPI-NCSC) and pluri-
D. Comparative assessment of cultured skin substitutes and native skin auto- potency. Stem Cell Rev 2008: 4: 256–260.
graft for treatment of full-thickness burns. Ann Surg 1995: 222: 743–752. 115 Heng B C, Cao T, Liu H, Phan T T. Directing stem cells into the keratinocyte
85 Hybbinette S, Bostrom M, Lindberg K. Enzymatic dissociation of keratinocytes lineage in vitro. Exp Dermatol 2005: 14: 1–16.
from human skin biopsies for in vitro cell propagation. Exp Dermatol 1999: 8: 116 Amoh Y, Yang M, Li L et al. Nestin-linked green fluorescent protein trans-
30–38. genic nude mouse for imaging human tumor angiogenesis. Cancer Res 2005:
86 Meana A, Iglesias J, Del R M et al. Large surface of cultured human epithe- 65: 5352–5357.
lium obtained on a dermal matrix based on live fibroblast-containing fibrin 117 Yu H, Fang D, Kumar S M et al. Isolation of a novel population of multipo-
gels. Burns 1998: 24: 621–630. tent adult stem cells from human hair follicles. Am J Pathol 2006: 168:
87 Barrandon Y, Green H. Cell size as a determinant of the clone-forming ability 1879–1888.
of human keratinocytes. Proc Natl Acad Sci U S A 1985: 82: 5390–5394. 118 Limat A, Noser F K. Serial cultivation of single keratinocytes from the outer
88 Bianco P, Robey P G. Stem cells in tissue engineering. Nature 2001: 414: root sheath of human scalp hair follicles. J Invest Dermatol 1986: 87: 485–
118–121. 488.
89 Dunnwald M, Tomanek-Chalkley A, Alexandrunas D, Fishbaugh J, Bickenbach 119 Mavilio F, Pellegrini G, Ferrari S et al. Correction of junctional epidermolysis
J R. Isolating a pure population of epidermal stem cells for use in tissue engi- bullosa by transplantation of genetically modified epidermal stem cells. Nat
neering. Exp Dermatol 2001: 10: 45–54. Med 2006: 12: 1397–1402.
90 Pouliot N, Redvers R P, Ellis S, Saunders N A, Kaur P. Optimization of a trans- 120 Battula V L, Bareiss P M, Treml S et al. Human placenta and bone marrow
plant model to assess skin reconstitution from stem cell-enriched primary derived MSC cultured in serum-free, b-FGF-containing medium express cell
human keratinocyte populations. Exp Dermatol 2005: 14: 60–69. surface frizzled-9 and SSEA-4 and give rise to multilineage differentiation.
91 Li A, Simmons P J, Kaur P. Identification and isolation of candidate human Differentiation 2007: 75: 279–291.
keratinocyte stem cells based on cell surface phenotype. Proc Natl Acad Sci 121 Schultz S S, Lucas P A. Human stem cells isolated from adult skeletal muscle
U S A 1998: 95: 3902–3907. differentiate into neural phenotypes. J Neurosci Methods 2006: 152: 144–
92 Tani H, Morris R J, Kaur P. Enrichment for murine keratinocyte stem cells 155.
based on cell surface phenotype. Proc Natl Acad Sci U S A 2000: 97: 10960– 122 Jiang Y, Henderson D, Blackstad M, Chen A, Miller R F, Verfaillie C M. Neu-
10965. roectodermal differentiation from mouse multipotent adult progenitor cells.
93 Kim D S, Cho H J, Choi H R, Kwon S B, Park K C. Isolation of human epider- Proc Natl Acad Sci U S A 2003: 100 (Suppl 1): 11854–11860.
mal stem cells by adherence and the reconstruction of skin equivalents. Cell 123 Woodbury D, Reynolds K, Black I B. Adult bone marrow stromal stem cells
Mol Life Sci 2004: 61: 2774–2781. express germline, ectodermal, endodermal, and mesodermal genes prior to
94 Jones P H, Harper S, Watt F M. Stem cell patterning and fate in human epi- neurogenesis. J Neurosci Res 2002: 69: 908–917.
dermis. Cell 1995: 80: 83–93. 124 Wang G, Bunnell B A, Painter R G et al. Adult stem cells from bone marrow
95 Terunuma A, Jackson K L, Kapoor V, Telford W G, Vogel J C. Side population stroma differentiate into airway epithelial cells: potential therapy for cystic
keratinocytes resembling bone marrow side population stem cells are distinct fibrosis. Proc Natl Acad Sci U S A 2005: 102: 186–191.
from label-retaining keratinocyte stem cells. J Invest Dermatol 2003: 121: 125 Deng W, Han Q, Liao L et al. Engrafted bone marrow-derived flk-(1 + ) mes-
1095–1103. enchymal stem cells regenerate skin tissue. Tissue Eng 2005: 11: 110–119.
96 Triel C, Vestergaard M E, Bolund L, Jensen T G, Jensen U B. Side population 126 Silva G V, Litovsky S, Assad J A et al. Mesenchymal stem cells differentiate
cells in human and mouse epidermis lack stem cell characteristics. Exp Cell into an endothelial phenotype, enhance vascular density, and improve heart
Res 2004: 295: 79–90. function in a canine chronic ischemia model. Circulation 2005: 111: 150–
97 Zhou J X, Jia L W, Yang Y J, Peng S, Cao Y J, Duan E K. Enrichment and 156.
characterization of mouse putative epidermal stem cells. Cell Biol Int 2004: 127 Bosnakovski D, Mizuno M, Kim G, Takagi S, Okumura M, Fujinaga T. Isola-
28: 523–529. tion and multilineage differentiation of bovine bone marrow mesenchymal
98 Sleeman M A, Watson J D, Murison J G. Neonatal murine epidermal cells stem cells. Cell Tissue Res 2005: 319: 243–253.
express a functional multidrug-resistant pump. J Invest Dermatol 2000: 115: 128 Oshima Y, Watanabe N, Matsuda K, Takai S, Kawata M, Kubo T. Behavior of
19–23. transplanted bone marrow-derived GFP mesenchymal cells in osteochondral
99 Ohyama M, Terunuma A, Tock C L et al. Characterization and isolation of stem defect as a simulation of autologous transplantation. J Histochem Cytochem
cell-enriched human hair follicle bulge cells. J Clin Invest 2006: 116: 249–260. 2005: 53: 207–216.
100 Cotsarelis G. Epithelial stem cells: a folliculocentric view. J Invest Dermatol 129 Urbich C, Dimmeler S. Endothelial progenitor cells: characterization and role
2006: 126: 1459–1468. in vascular biology. Circ Res 2004: 95: 343–353.
101 Mills A A, Zheng B, Wang X J, Vogel H, Roop D R, Bradley A. p63 is a p53 130 Asahara T, Masuda H, Takahashi T et al. Bone marrow origin of endothelial
homologue required for limb and epidermal morphogenesis. Nature 1999: progenitor cells responsible for postnatal vasculogenesis in physiological and
398: 708–713. pathological neovascularization. Circ Res 1999: 85: 221–228.
102 Stasiak P C, Purkis P E, Leigh I M, Lane E B. Keratin 19: predicted amino acid 131 Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for
sequence and broad tissue distribution suggest it evolved from keratinocyte organ vascularization and regeneration. Nat Med 2003: 9: 702–712.
keratins. J Invest Dermatol 1989: 92: 707–716. 132 Kawada H, Fujita J, Kinjo K et al. Nonhematopoietic mesenchymal stem cells
103 Webb A, Li A, Kaur P. Location and phenotype of human adult keratinocyte can be mobilized and differentiate into cardiomyocytes after myocardial
stem cells of the skin. Differentiation 2004: 72: 387–395. infarction. Blood 2004: 104: 3581–3587.
104 Lyle S, Christofidou-Solomidou M, Liu Y, Elder D E, Albelda S, Cotsarelis G. 133 Orlic D, Kajstura J, Chimenti S et al. Mobilized bone marrow cells repair the
The C8 ⁄ 144B monoclonal antibody recognizes cytokeratin 15 and defines infarcted heart, improving function and survival. Proc Natl Acad Sci U S A
the location of human hair follicle stem cells. J Cell Sci 1998: 111 (Pt 21): 2001: 98: 10344–10349.
3179–3188. 134 Fathke C, Wilson L, Hutter J et al. Contribution of bone marrow-derived cells
105 Whitbread L A, Powell B C. Expression of the intermediate filament keratin to skin: collagen deposition and wound repair. Stem Cells 2004: 22: 812–822.
gene, K15, in the basal cell layers of epithelia and the hair follicle. Exp Cell 135 Ishii G, Sangai T, Sugiyama K et al. In vivo characterization of bone marrow-
Res 1998: 244: 448–459. derived fibroblasts recruited into fibrotic lesions. Stem Cells 2005: 23: 699–
106 Zhu A J, Watt F M. beta-catenin signalling modulates proliferative potential 706.
of human epidermal keratinocytes independently of intercellular adhesion. 136 Liu P, Deng Z, Han S et al. Tissue-engineered skin containing mesenchymal
Development 1999: 126: 2285–2298. stem cells improves burn wounds. Artif Organs 2008: 32: 925–931.
107 Morris R J, Liu Y, Marles L et al. Capturing and profiling adult hair follicle 137 McFarlin K, Gao X, Liu Y B et al. Bone marrow-derived mesenchymal stromal
stem cells. Nat Biotechnol 2004: 22: 411–417. cells accelerate wound healing in the rat. Wound Repair Regen 2006: 14:
108 Kloepper J E, Tiede S, Brinckmann J et al. Immunophenotyping of the 471–478.
human bulge region: the quest to define useful in situ markers for human 138 Badiavas E V, Falanga V. Treatment of chronic wounds with bone marrow-
epithelial hair follicle stem cells and their niche. Exp Dermatol 2008: 17: derived cells. Arch Dermatol 2003: 139: 510–516.
592–609. 139 Falanga V, Iwamoto S, Chartier M et al. Autologous bone marrow-derived
109 Taylor G, Lehrer M S, Jensen P J, Sun T T, Lavker R M. Involvement of follicu- cultured mesenchymal stem cells delivered in a fibrin spray accelerate healing
lar stem cells in forming not only the follicle but also the epidermis. Cell in murine and human cutaneous wounds. Tissue Eng 2007: 13: 1299–1312.
2000: 102: 451–461. 140 Yoshikawa T, Mitsuno H, Nonaka I et al. Wound therapy by marrow mesen-
110 Cotsarelis G, Sun T T, Lavker R M. Label-retaining cells reside in the bulge chymal cell transplantation. Plast Reconstr Surg 2008: 121: 860–877.
area of pilosebaceous unit: implications for follicular stem cells, hair cycle, 141 Vojtassak J, Danisovic L, Kubes M et al. Autologous biograft and mesenchy-
and skin carcinogenesis. Cell 1990: 61: 1329–1337. mal stem cells in treatment of the diabetic foot. Neuro Endocrinol Lett 2006:
111 Fuchs E, Tumbar T, Guasch G. Socializing with the neighbors: stem cells and 27 (Suppl 2): 134–137.
their niche. Cell 2004: 116: 769–778. 142 Rasulov M F, Vasilchenkov A V, Onishchenko N A et al. First experience of
112 Tiede S, Kloepper J E, Bodo E, Tiwari S, Kruse C, Paus R. Hair follicle stem the use bone marrow mesenchymal stem cells for the treatment of a patient
cells: walking the maze. Eur J Cell Biol 2007: 86: 355–376. with deep skin burns. Bull Exp Biol Med 2005: 139: 141–144.

ª 2010 John Wiley & Sons A/S, Experimental Dermatology, 19, 697–706 705
Dieckmann et al.

143 Wu Y, Chen L, Scott P G, Tredget E E. Mesenchymal stem cells enhance 174 Takahashi K, Tanabe K, Ohnuki M et al. Induction of pluripotent stem cells
wound healing through differentiation and angiogenesis. Stem Cells 2007: from adult human fibroblasts by defined factors. Cell 2007: 131: 861–872.
25: 2648–2659. 175 Kang H J, Lee H Y, Na S H et al. Differential effect of intracoronary
144 Li H, Fu X, Ouyang Y, Cai C, Wang J, Sun T. Adult bone-marrow-derived infusion of mobilized peripheral blood stem cells by granulocyte colony-
mesenchymal stem cells contribute to wound healing of skin appendages. stimulating factor on left ventricular function and remodeling in patients
Cell Tissue Res 2006: 326: 725–736. with acute myocardial infarction versus old myocardial infarction: the
145 Badillo A T, Redden R A, Zhang L, Doolin E J, Liechty K W. Treatment of dia- MAGIC Cell-3-DES randomized, controlled trial. Circulation 2006: 114 (1
betic wounds with fetal murine mesenchymal stromal cells enhances wound Suppl): I145–I151.
closure. Cell Tissue Res 2007: 329: 301–311. 176 Veltman C E, Soliman O I, Geleijnse M L et al. Four-year follow-up of treat-
146 Taura D, Noguchi M, Sone M et al. Adipogenic differentiation of human ment with intramyocardial skeletal myoblasts injection in patients with ischae-
induced pluripotent stem cells: comparison with that of human embryonic mic cardiomyopathy. Eur Heart J 2008: 29: 1386–1396.
stem cells. FEBS Lett 2009: 583: 1029–1033. 177 Bang O Y, Lee J S, Lee P H, Lee G. Autologous mesenchymal stem cell trans-
147 Zuk P A, Zhu M, Mizuno H et al. Multilineage cells from human adipose tis- plantation in stroke patients. Ann Neurol 2005: 57: 874–882.
sue: implications for cell-based therapies. Tissue Eng 2001: 7: 211–228. 178 Planat-Benard V, Menard C, Andre M et al. Spontaneous cardiomyocyte dif-
148 Zuk P A, Zhu M, Ashjian P et al. Human adipose tissue is a source of multipo- ferentiation from adipose tissue stroma cells. Circ Res 2004: 94: 223–229.
tent stem cells. Mol Biol Cell 2002: 13: 4279–4295. 179 Planat-Benard V, Silvestre J S, Cousin B et al. Plasticity of human adipose line-
149 Boquest A C, Shahdadfar A, Brinchmann J E, Collas P. Isolation of stromal age cells toward endothelial cells: physiological and therapeutic perspectives.
stem cells from human adipose tissue. Methods Mol Biol 2006: 325: 35–46. Circulation 2004: 109: 656–663.
150 Aust L, Devlin B, Foster S J et al. Yield of human adipose-derived adult stem 180 Erickson G R, Gimble J M, Franklin D M, Rice H E, Awad H, Guilak F. Chon-
cells from liposuction aspirates. Cytotherapy 2004: 6: 7–14. drogenic potential of adipose tissue-derived stromal cells in vitro and in vivo.
151 Tholpady S S, Llull R, Ogle R C, Rubin J P, Futrell J W, Katz A J. Adipose Biochem Biophys Res Commun 2002: 290: 763–769.
tissue: stem cells and beyond. Clin Plast Surg 2006: 33: 55–62, vi. 181 Safford K M, Hicok K C, Safford S D et al. Neurogenic differentiation of mur-
152 Mitchell J B, McIntosh K, Zvonic S et al. Immunophenotype of human ine and human adipose-derived stromal cells. Biochem Biophys Res Commun
adipose-derived cells: temporal changes in stromal-associated and stem cell- 2002: 294: 371–379.
associated markers. Stem Cells 2006: 24: 376–385. 182 Petersen B E, Bowen W C, Patrene K D et al. Bone marrow as a potential
153 Ogawa R. The importance of adipose-derived stem cells and vascularized source of hepatic oval cells. Science 1999: 284: 1168–1170.
tissue regeneration in the field of tissue transplantation. Curr Stem Cell Res 183 Kopen G C, Prockop D J, Phinney D G. Marrow stromal cells migrate
Ther 2006: 1: 13–20. throughout forebrain and cerebellum, and they differentiate into astrocytes
154 Gronthos S, Franklin D M, Leddy H A, Robey P G, Storms R W, Gimble J M. after injection into neonatal mouse brains. Proc Natl Acad Sci U S A 1999:
Surface protein characterization of human adipose tissue-derived stromal 96: 10711–10716.
cells. J Cell Physiol 2001: 189: 54–63. 184 Haynesworth S E, Goshima J, Goldberg V M, Caplan A I. Characterization
155 Gimble J, Guilak F. Adipose-derived adult stem cells: isolation, characteriza- of cells with osteogenic potential from human marrow. Bone 1992: 13:
tion, and differentiation potential. Cytotherapy 2003: 5: 362–369. 81–88.
156 Gimble J M, Guilak F. Differentiation potential of adipose derived adult stem 185 Bruder S P, Jaiswal N, Haynesworth S E. Growth kinetics, self-renewal, and
(ADAS) cells. Curr Top Dev Biol 2003: 58: 137–160. the osteogenic potential of purified human mesenchymal stem cells during
157 Ebrahimian T G, Pouzoulet F, Squiban C et al. Cell therapy based on adipose extensive subcultivation and following cryopreservation. J Cell Biochem 1997:
tissue-derived stromal cells promotes physiological and pathological wound 64: 278–294.
healing. Arterioscler Thromb Vasc Biol 2009: 29: 503–510. 186 Pittenger M F, Mackay A M, Beck S C et al. Multilineage potential of adult
158 Altman A M, Matthias N, Yan Y et al. Dermal matrix as a carrier for in vivo human mesenchymal stem cells. Science 1999: 284: 143–147.
delivery of human adipose-derived stem cells. Biomaterials 2008: 29: 1431– 187 Tateishi K, He J, Taranova O, Liang G, D’Alessio A C, Zhang Y. Generation of
1442. insulin-secreting islet-like clusters from human skin fibroblasts. J Biol Chem
159 Altman A M, Yan Y, Matthias N et al. IFATS collection: human adipose- 2008: 283: 31601–31607.
derived stem cells seeded on a silk fibroin-chitosan scaffold enhance 188 Karumbayaram S, Novitch B G, Patterson M et al. Directed differentiation of
wound repair in a murine soft tissue injury model. Stem Cells 2009: 27: human-induced pluripotent stem cells generates active motor neurons. Stem
250–258. Cells 2009: 27: 806–811.
160 Nambu M, Ishihara M, Nakamura S et al. Enhanced healing of mitomycin C- 189 Taura D, Sone M, Homma K et al. Induction and isolation of vascular cells
treated wounds in rats using inbred adipose tissue-derived stromal cells from human induced pluripotent stem cells–brief report. Arterioscler Thromb
within an atelocollagen matrix. Wound Repair Regen 2007: 15: 505–510. Vasc Biol 2009: 29: 1100–1103.
161 Nambu M, Kishimoto S, Nakamura S et al. Accelerated wound healing in 190 Zhang J, Wilson G F, Soerens A G et al. Functional cardiomyocytes derived
healing-impaired db ⁄ db mice by autologous adipose tissue-derived stromal from human induced pluripotent stem cells. Circ Res 2009: 104: e30–e41.
cells combined with atelocollagen matrix. Ann Plast Surg 2009: 62: 317– 191 Doetschman T C, Eistetter H, Katz M, Schmidt W, Kemler R. The in vitro
321. development of blastocyst-derived embryonic stem cell lines: formation of vis-
162 Trottier V, Marceau-Fortier G, Germain L, Vincent C, Fradette J. IFATS collec- ceral yolk sac, blood islands and myocardium. J Embryol Exp Morphol 1985:
tion: using human adipose-derived stem ⁄ stromal cells for the production of 87: 27–45.
new skin substitutes. Stem Cells 2008: 26: 2713–2723. 192 Czyz J, Wiese C, Rolletschek A, Blyszczuk P, Cross M, Wobus A M. Potential
163 Spees J L, Olson S D, Ylostalo J et al. Differentiation, cell fusion, and of embryonic and adult stem cells in vitro. Biol Chem 2003: 384: 1391–1409.
nuclear fusion during ex vivo repair of epithelium by human adult stem
cells from bone marrow stroma. Proc Natl Acad Sci U S A 2003: 100:
2397–2402. Supporting Information
164 Caplan A I, Dennis J E. Mesenchymal stem cells as trophic mediators. J Cell
Biochem 2006: 98: 1076–1084. Additional Supporting Information may be found in the online version of
165 Docheva D, Popov C, Mutschler W, Schieker M. Human mesenchymal stem
cells in contact with their environment: surface characteristics and the inte- this article:
grin system. J Cell Mol Med 2007: 11: 21–38. Figure S1. Scheme of a mature anagen hair follicle, plucked hair follicles
166 Thomson J A, Itskovitz-Eldor J, Shapiro S S et al. Embryonic stem cell lines in their anagen phase, the manufacturing process of outer root sheath
derived from human blastocysts. Science 1998: 282: 1145–1147.
167 Grinnemo K H, Sylven C, Hovatta O, Dellgren G, Corbascio M. Immunogenic- (ORS) cells to an epidermal equivalent (EpiDex) and finally the clinical
ity of human embryonic stem cells. Cell Tissue Res 2008: 331: 67–78. application in the treatment of chronic venous leg ulcers.
168 Grinnemo K H, Kumagai-Braesch M, Mansson-Broberg A et al. Human Table S1. Representative human clinical trials of stem cell therapies,
embryonic stem cells are immunogenic in allogeneic and xenogeneic settings.
Reprod Biomed Online 2006: 13: 712–724. both completed or pending, indexed by clinical application and source of
169 Knoepfler P S. Deconstructing stem cell tumorigenicity: a roadmap to safe stem cell.
regenerative medicine. Stem Cells 2009: 27: 1050–1056. Table S2. Examples of various types of acellular and cellular skin substi-
170 Aasen T, Raya A, Barrero M J et al. Efficient and rapid generation of induced
pluripotent stem cells from human keratinocytes. Nat Biotechnol 2008: 26: tutes currently commercially available without intending to be complete.
1276–1284. Table S3. Differentiation potential of human epidermal stem cells, bone
171 Yu J, Vodyanik M A, Smuga-Otto K et al. Induced pluripotent stem cell lines marrow-derived mesenchymal stromal cells, adipose stem cells, embryonic
derived from human somatic cells. Science 2007: 318: 1917–1920.
172 Lowry W E, Richter L, Yachechko R et al. Generation of human induced plu- stem cells and induced pluripotent stem cells.
ripotent stem cells from dermal fibroblasts. Proc Natl Acad Sci U S A 2008: Please note: Wiley-Blackwell are not responsible for the content or func-
105: 2883–2888. tionality of any supporting materials supplied by the authors. Any queries
173 Nakagawa M, Koyanagi M, Tanabe K et al. Generation of induced pluripo-
tent stem cells without Myc from mouse and human fibroblasts. Nat Biotech- (other than missing material) should be directed to the corresponding
nol 2008: 26: 101–106. author for the article.

706 ª 2010 John Wiley & Sons A/S, Experimental Dermatology, 19, 697–706

Das könnte Ihnen auch gefallen