Sie sind auf Seite 1von 6

Available online at www.sciencedirect.

com

Methods 45 (2008) 115–120


www.elsevier.com/locate/ymeth

Adipose-derived stem cells: Isolation, expansion and differentiation q


Bruce A. Bunnell a,b,c,*, Mette Flaat a,d, Christine Gagliardi a,c, Bindiya Patel a,c,
Cynthia Ripoll a,c
a
Division of Gene Therapy, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, USA
b
Center for Gene Therapy, School of Medicine, Tulane University, New Orleans, LA, USA
c
Department of Pharmacology, School of Medicine, Tulane University, New Orleans, LA, USA
d
Department of Biochemistry, School of Medicine, Tulane University, New Orleans, LA, USA

Accepted 17 March 2008


Available online 29 May 2008

Abstract

The emerging field of regenerative medicine will require a reliable source of stem cells in addition to biomaterial scaffolds and cytokine
growth factors. Adipose tissue has proven to serve as an abundant, accessible and rich source of adult stem cells with multipotent prop-
erties suitable for tissue engineering and regenerative medical applications. There has been increased interest in adipose-derived stem cells
(ASCs) for tissue engineering applications. Here, methods for the isolation, expansion and differentiation of ASCs are presented and
described in detail. While this article has focused on the isolation of ASCs from human adipose tissue, the procedure can be applied
to adipose tissues from other species with minimal modifications.
Ó 2008 Elsevier Inc. All rights reserved.

Keywords: Adipose-derived stem cells (ASCs); Biopsy; Differentiation; Expansion; Isolation; Lipoaspirate; Mesenchymal stem cells (MSCs)

1. Introduction safely and effectively transplanted to either an autologous


or allogeneic host [1].
By definition, a stem cell is characterized by its ability to Tissue specific stem cells comprise the second group and
undergo self-renewal and its ability to undergo multilineage are derived from specific organs, such as brain, gut, lung,
differentiation and form terminally differentiated cells. Ide- liver, adipose tissue and bone marrow [2]. It has become
ally, a stem cell for regenerative medicinal applications evident that these stem cells persist in adult tissues,
should meet the following set of criteria: (i) should be although they represent a rare population localized in small
found in abundant quantities (millions to billions of cells); niches [3]. Postnatal (adult) stem cells are not totipotent;
(ii) can be collected and harvested by a minimally invasive however, they are pluripotent. These cells retain a broad
procedure; (iii) can be differentiated along multiple cell differentiation potential, but their developmental potential
lineage pathways in a reproducible manner; (iv) can be is more restricted than embryonic stem cells. Adult stem
cells were initially thought to have the differentiation
q
capacity limited to their tissue of origin, however recent
The research was supported by the National Center for Research
Resources, National Institutes of Health, Grant No. RR00164, and a
studies have demonstrated that stem cells have the capacity
grant from the State of Louisiana Millennium Health Excellence Fund to differentiate into cells of mesodermal, endodermal and
and the Louisiana Gene Therapy Research Consortium. ectodermal origins [4–10]. The plasticity of MSCs most
*
Corresponding author. Address: Center for Gene Therapy, Depart- often refers to the inherent ability retained within stem cells
ment of Pharmacology, Division of Gene Therapy, Tulane National to cross lineage barriers and to adopt the phenotypic, bio-
Primate Research Center, Tulane University Health Sciences Center,
18703 Three Rivers Road, Covington, LA 70433, USA. Fax: +1 985 871
chemical and functional properties of cells unique to other
6564. tissues. Adult MSCs can be isolated from bone marrow
E-mail address: bbunnell@tulane.edu (B.A. Bunnell). and adipose tissue. With the increased incidence of obesity

1046-2023/$ - see front matter Ó 2008 Elsevier Inc. All rights reserved.
doi:10.1016/j.ymeth.2008.03.006
116 B.A. Bunnell et al. / Methods 45 (2008) 115–120

in the U.S. and abroad, subcutaneous adipose tissue is stem cells” (ASCs) to identify the isolated, plastic-adherent,
abundant and readily accessible [11,12]. Approximately multipotent cell population. The ASCs have been distin-
400,000 liposuction surgeries are performed in the U.S. guished from the plastic adherent adult stem/progenitor
each year and these procedures yield anywhere from cells from bone marrow originally referred to as fibroblas-
100 ml to >3 L of lipoaspirate tissue [12]. This material is toid colony forming units, then in the hematological litera-
routinely discarded. As discussed below, adipose-derived ture as marrow stromal, subsequently as mesenchymal
stem cells are multipotent and hold promise for a range stem cells, and most recently as multipotent mesenchymal
of therapeutic applications. stromal cells (MSCs).
Adipocytes develop from mesenchymal cells via a com-
plex cascade of transcriptional and non-transcriptional
events that occurs throughout human life. Stromal cells 2. Isolation of mesenchymal stem cells from adipose tissue
that have preadipocyte characteristics can be isolated from
adipose tissue of adult subjects, propagated in vitro and The initial methods to isolate cells from adipose tissue
induced to differentiate into adipocytes [13–16]. Adipocyte were pioneered by Rodbell and colleagues in the 1960s
differentiation is a complex process accompanied by coor- [20–22]. They minced rat fat pads, washed extensively to
dinated changes in cell morphology, hormone sensitivity remove contaminating hematopoietic cells, incubated the
and gene expression that have been studied primarily in tissue fragments with collagenase and centrifuged the
murine preadipocyte cell lines rather than in human preadi- digest, thereby separating the floating population of
pocytes. This protocol describes primary in vitro culture of mature adipocytes from the pelleted stromal vascular frac-
stromal cell isolated from either large or small quantities of tion (SVF) (Fig. 1). The SVF consisted of a heterogeneous
human adipose tissue. While historically in the literature cell population, including circulating blood cells, fibro-
adipose-derived stromal cells have been termed ‘‘pre-adipo- blasts, pericytes and endothelial cells as well as ‘‘pre-adipo-
cytes” [14,15], there is a growing appreciation that they are cytes” or adipocyte progenitors [20–22]. The final isolation
multipotent, with chondrogenic, neurogenic and osteogenic step selected for the plastic adherent population within the
capability [14,15,17–19]. SVF cells, which enriched for the ‘‘pre-adipocytes”. Subse-
A variety of names have been used to describe the plastic quently, this procedure has been modified for the isolation
adherent cell population isolated from collagenase digests of cells from human adipose tissue specimens
of adipose tissue. The following terms have been used to [23,24,14,25,26]. Initially, fragments of human tissue were
identify the same adipose tissue cell population: adipose- minced by hand; however, with the development of lipo-
derived stem/stromal cells (ASCs), adipose derived adult suction surgery, this procedure has been simplified. During
stem (ADAS) cells, adipose derived adult stromal cells, adi- tumescent liposuction, plastic surgeons infuse the subcuta-
pose derived stromal cells (ADSC), adipose stromal cells neous tissues with a saline solution containing anesthetic
(ASC), adipose mesenchymal stem cells (AdMSC), lipo- and/or epinephrine via a cannula and then remove both
blast, pericyte, pre-adipocyte, processed lipoaspirate the liquid and tissue under suction [27]. The procedure gen-
(PLA) cells. The use of this diverse nomenclature has lead erates finely minced tissue fragments whose size depends on
to significant confusion in the literature. To address this the cannula’s dimensions. Independent studies have deter-
issue, the International Fat Applied Technology Society mined that liposuction aspiration alone does not signifi-
reached a consensus to adopt the term ‘‘adipose-derived cantly alter the viability of isolated SVF cells [28–30].

Fig. 1. Scheme for processing of adipose tissue and isolation of adipose-derived stem cells.
B.A. Bunnell et al. / Methods 45 (2008) 115–120 117

Indeed, adherent stromal cells with characteristics of adi- 3. Culture and expansion of ASCs
pocyte progenitors can be found directly within the lipo-
suction aspiration fluid, as well as in SVF derived from Seventy-two hours after plating, aspirate the entire med-
the tissue fragment digests [31]. However, when ultra- ium from the wells. Again, it is essential to note that the
sound-assisted liposuction is performed, the number of percent of preadipocytes obtained from the SVF after
cells recovered from tissue digests is reduced, as is their digestion is patient-dependent. If the SVF does not expand
proliferative capacity [30]. The recovery of ASCs can be well, increase the FBS contained in the stromal medium to
improved further by manipulating the centrifugation speed 25%; however this may promote premature adipogenesis.
[31]. Investigators have achieved optimal cell recovery Signs of deterioration such as granularity around the
using a centrifugation speed of 1200g based on the subse- nucleus, cytoplasmic vacuolations and/or detachment of
quent formation of a human-derived adipose tissue depot the cells from the plastic surface may indicate inadequate
following implantation in an immunodeficient murine or toxic medium, microbial contamination or senescence
model [31]. of the primary cells.
Adipose tissue is collected by needle biopsy or liposuc- Wash the cells with prewarmed PBS (1% antibiotic can
tion aspiration. The adipose sample can be kept at room be added to the solution). Pipette the solution over the cell
temperature for no more than 24 h prior to use (Fig. 1). layer several times in order to clean the cells thoroughly
ASCs can be isolated from adipose tissue by first washing from any tissue fragments and/or blood cells. Add a volume
the tissue sample extensively with phosphate-buffered sal- of fresh stromal medium according to the well capacity of
ine (PBS) containing 5% penicillin/streptomycin (P/S). the culture plate. Maintain the cells in a humidified tissue
Upon removal of debris, place the sample in a sterile tissue culture incubator at 37 °C with 5%CO2. Change the
culture plate with 0.075% collagenase Type I prepared in medium every second day until the cells reach 80–90% con-
PBS containing 2% P/S for tissue digestion. Mince the adi- fluence, and then there are two options: either harvest the
pose tissue sample using two scalpels and pipette the sam- cells or directly induce the adipocyte differentiation. For
ple up and down with a 25 or 50 ml pipette several times to harvesting viable ASCs, add a small volume (250–500 ll)
further facilitate the digestion. Incubate the sample for of sterile, warm PBS to the wells and allow PBS to remain
30 min at 37 °C, 5% CO2, and then neutralize the collage- on cells briefly. Replace the PBS with 500 ll of Trypsin/
nase Type I activity by adding 5 ml of a-MEM containing EDTA solution (0.5%). Place in an incubator for 5 min
20% heat inactivated fetal bovine serum (FBS, Atlanta Bio- and verify that the cells have detached by assessment using
logical, Atlanta, GA), to the tissue sample. Pipette the sam- a microscope. Verify under microscope that more than 90%
ple up and down several times to further disintegrate of the cells have detached and then add 500 ll of stromal
aggregates of the adipose tissue. Upon disintegration, medium to allow the serum contained in the solution to neu-
transfer the sample to a 50 ml tube, avoiding the solid tralize the trypsin reaction. Transfer the medium containing
aggregates. The SVF, containing the ASCs, is obtained the suspended cells from the well to a sterile 2 ml tube. Cen-
by centrifuging the sample at 2000 rpm for 5 min. Take trifuge at 1200 rpm for 5 min. Aspirate the supernatant and
the samples out of the centrifuge and shake them vigor- suspend the cells in a small volume of stromal medium
ously to thoroughly disrupt the pellet and to mix the cells. (250 ll). Proceed to cell counting by taking an aliquot of
This step completes the separation of the stromal cells from cells diluted in trypan blue (for a 1:2 dilution: add 12.5 ll
the primary adipocytes. Repeat the centrifugation step. of suspended cells to 12.5 ll of trypan blue). Count cells
After spinning, aspirate all the collagenase solution above using the hemocytometer. The cells can then be replated
the pellet without disturbing the cells. Resuspend the pellet according to the well capacity in cell culture plates.
in 1 ml of lysis buffer, incubate for 10 min on ice and wash When expanded from a frozen vial, the cells are rapidly
with 20 ml of PBS/2% P/S and centrifuge at 2000 rpm for thawed at 37 °C and immediately seeded in a 15 cm2 plate
5 min. Aspirate the supernatant and resuspend the cell with complete stromal medium. Medium is changed the
pellet in a maximum of 3 ml of stromal medium (alpha- day after seeding and then every second day.
MEM, Mediatech, Herndon, VA) supplemented with
20% FBS, 1% L-glutamine (Mediatech), and 1% P/S and 4. Freezing and long-term storage of ASCs
the cell suspension is filtered through 70 mm cell strainer.
It is important to note that all fetal bovine serum should ASCs should be harvested at 80% confluence for freez-
be pre-screened prior to purchase for its ability to support ing. To collect cells, remove the culture medium and
both cell proliferation and adipocyte differentiation. Wash replace with a small volume of sterile, warm PBS. Remove
the cell strainer with an additional 2 ml of stromal medium the PBS and replace with trypsin-EDTA solution. Incubate
to obtain any additional cells. Plate the sample containing the culture dish at 37 °C for at least 5 min, or until approx-
the cells in a lysine coated culture plate and incubate at imately 90% of the cells have detached from the bottom of
37 °C, 5% CO2. Inoculate the cells in a single well of a the dish. Progress can be monitored under a microscope;
12-well plate for an amount of about 500 mg of adipose the treated cells will be rounded and floating. Add an equal
tissue or in a single well of a 24-well plate for an amount volume of stromal medium to inactivate the trypsin, and
of 250–150 mg of adipose tissue. transfer the suspension to a conical centrifuge tube. Centri-
118 B.A. Bunnell et al. / Methods 45 (2008) 115–120

fuge the tube at 1200 rpm for 5 min to pellet the cells. Care-
fully remove the supernatant, and resuspend the cell pellet
in 1–2 ml of room temperature cryopreservation medium.
Cryopreservation medium consists of 80% fetal bovine
serum, 10% dimethylsulfoxide (DMSO) and 10%
DMEM/Ham’s F-12, and should be used within two weeks
of its preparation. Determine the cell concentration, and
then dilute to a final concentration of 1–2 million viable
cells per milliliter of cryopreservation medium. Aliquot
the suspension into labeled cryovials, 1 ml/vial. Place the Fig. 3. Mesenchymal lineage differentiation assays for adipose-derived
vials in an alcohol freezing container, and store at stem cells. Adipogenic differentiation: As the stem cells proliferate, some
of these cells differentiate into preadipocytes. The preadipocytes undergo a
80 °C overnight. The freezing container will cool the vials
second differentiation step and begin to fill with lipid. Lipid accumulates
slowly, at approximately 1 °C every minute, until they within the cell in small vacuoles, which appear as droplets. Osteogenic
reach 80 °C. The next day, the frozen vials can be trans- differentiation: As cells undergo osteogenic differentiation they proliferate
ferred to a liquid nitrogen container for long-term storage. rapidly and form tightly packed colonies. In some cases, these colonies
Vials of cells should always be stored in the vapor phase of give rise to dense nodules from which radiate highly elongated spindle-
shaped cells with large nuclei. There are several methods to determine
the LN2 tank.
osteogenesis, such as demonstration of mineralization by staining with
Alizarin red, measurement of alkaline phosphatase (AP) activity, level of
calcium, and detection of lineage specific gene and protein regulations.
5. Mesenchymal differentiation assays for ASCs Chondrogenic differentiation: chondrogenesis occurs in three stages.
Cartilage formation initiates when the dividing MSCs begin expressing
ASCs display multipotency, meaning they retain the extra cellular matrix proteins that tell them to condense into nodules. Cells
ability to differentiate into cell types of multiple different in these nodules become chondrocytes and begin secreting the proteogly-
cans and collagen necessary for cartilage formation.
lineages (Figs. 2 and 3). The multipotency of these cells
has generated interest in their potential therapeutic value
for regenerative medicine. Differentiation of these cells
can be directed by the addition of specific cocktails of a factor, since some studies suggest that the differentiation
chemical inducers or cytokines. The differentiation effi- capacity is higher in culture from younger subjects com-
ciency is patient dependent. The age of the donor can be pared to older people.

Fig. 2. Multilineage differentiation potential for adipose-derived stem cells.


B.A. Bunnell et al. / Methods 45 (2008) 115–120 119

5.1. Adipogenic differentiation after a three day rest period; i.e., 3 days on in the presence
of the differentiation medium and three days off in the pres-
When the cells reach between 80–90% confluence, the ence of the adipocyte medium [32]. Repeat this cycle until
preadipocytes can be induced to differentiate. Aspirate mature adipocytes are obtained.
the medium, add a small volume (about 1.5 ml for a 6-well
plate) of prewarmed PBS + 1% antibiotic to wash the cells, 5.2. Osteogenic differentiation
and then remove the PBS by aspiration. Do not dry the
well when changing the medium since adipocytes tend to Osteogenesis can be induced using culture medium sup-
float when new medium is added. Next, add the differenti- plemented with 1 nM dexamethasone, 2 mM b-glycerol-
ation medium. phosphate and 50 lM ascorbate-2-phosphate. The cells
Adipogenic differentiation can be induced using culture are induced in this medium for approximately 14 days
medium supplemented with 0.5 mM isobutylmethylxan- and the osteogenic medium is replaced every 2–3 days.
thine, 50 lM indomethacin and 0.5 lM dexamethasone. Mineralization is assessed by staining the cells with
Change the medium every 3 days until mature adipocytes 40 mM Alizarin Red (pH 4.1) after fixation in 10%
are obtained After 12–14 days of differentiation, the cells formalin.
can be fixed either using a 10% formalin solution or 70% eth-
anol. After removing the medium and washing the cells with 5.3. Chondrogenic differentiation
PBS, immerse the cells in the fixative solution, either 10%
formalin or 70% ethanol for 30 min or 1 h, respectively. For chondrogenic differentiation, the pellet culture sys-
Remove the fixative and air dry before staining or add water tem described by Sekiya et al. can be used [33]. The cell pel-
until performing the dye staining. The fixed cells can be lets are cultured in chondrogenic differentiation medium,
stored at 4 or 20 °C for as long as several months. It is which consists of high glucose DMEM supplemented with
important to note that when using ethanol as the fixative, 500 ng/ml bone morphogenic protein-6, 10 ng/ml TGF-b3,
there is a risk that the lipids will be eluted from the cells. 10 7 M dexamethasone, 50 lg/ml ascorbate 2-phosphate,
The accumulation of neutral lipids can be detected by 40 lg/ml proline, 100 lg/ml pyruvate and 50 mg/ml
staining the cells in a solution of 0.5% Oil Red-O at room ITS + premix (Becton Dickinson: 6.25 lg/ml insulin,
temperature for at least 60 min, and can be performed out- 6.25 lg/ml transferrin, 6.25 ng/ml selenous acid, 1.25 mg/
side of a biosafety hood. As an alternative, a two-step dye ml bovine serum albumin, 5.35 mg/ml linoleic acid). The
staining is performed by using a neutral lipid fluorescent medium is replaced every 2–3 days for 21 days. The pellets
dye (BODIPY, maximum fluorescence emission range is are then fixed in formalin, embedded in paraffin and sec-
from 510 to 665 nm) and a nuclear fluorescent dye (DAPI, tioned. The sections can then be stained with Toluidine Blue.
maximum excitation is about 360 nm and the emission
maximum is 460 nm). All the procedures are performed 6. Neural differentiation of ASCs
at room temperature.
Immediately after drying the cells from the fixative solu- Neurospheres are generated as described in Kang et al.
tion or after removing the water from the wells, add 10 mg/ [34]. Undifferentiated adipose stem cells cultured at high
ml BODIPY working solution and stain for 20 min. It is densities will spontaneously form spherical clumps of cells
recommended to perform the staining in a semi-dark envi- that can be isolated in 0.25% trypsin/2.21 mM EDTA.
ronment. After 20 min, remove all BODIPY solution and Free-floating neurospheres released from the cell culture
immediately wash with H2O several times. For the DAPI surface into the culture medium can also be collected.
staining, remove all H2O from the wells and air dry the The spheres of cells are transferred to an ultra low cluster
cells for 1–2 min. Add 300 nM DAPI working solution to culture dish and cultured in neurobasal medium supple-
the cells and counterstain for 20 min at room temperature. mented with B27 (1:50), 20 ng/ml bFGF and 20 ng/ml
The percentage of cells undergoing adipogenesis can be EGF for 4–7 days. The culture density of the spheroid
calculated by microscopic inspection. The number of cells bodies should be maintained at 10–20 spheroids/cm2 to
in a field staining positive with BODIPY for lipid droplets prevent self aggregation.
can be determined as a percentage relative to the total num- Neurospheres derived from adipose stem cells are lay-
ber of cells in the field, i.e., the number of cells determined ered on a Nunc tissue culture dish and maintained in Neu-
by positive staining with the DAPI nuclear stain. robasal medium containing only the B27 supplement.
If adipogenic differentiation does not occur efficiently, During differentiation, 70% of the medium is replaced
adipogenesis can be enhanced using the following alterna- every 3–4 days.
tives: (i) different PPARc agonists (troglitazone, pioglitaz- The induction of neural differentiation can be confirmed
one among others) can be added to the medium; (ii) 5% using RT-PCR, immunohistochemistry or Western blot-
rabbit serum (RS) can be added to the differentiation med- ting for the expression of neuronal or glial antigens. The
ium to enhance differentiation (the ethyl acetate contained expression the neuronal associated markers such as nestin,
in the RS has been found 35-fold more abundant than in NeuN, intermediate filament, MAP2, b-III tubulin and glu-
FBS); or (iii) add the differentiation medium multiple times tamate receptor subunits NR1 and NR2or the oligoden-
120 B.A. Bunnell et al. / Methods 45 (2008) 115–120

drocyte marker, S-100 or glial fibrillary acidic protein [11] G.A. Bray, J. Clin. Endocrinol. Metab. 89 (2004) 2583–2589.
(GFAP) are routinely detected after differentiation. [12] A.J. Katz, R. Llull, M.H. Hedrick, J.W. Futrell, Clin. Plast. Surg. 26
(1999) 587–603.
[13] S.M. Rangwala, M.A. Lazar, Annu. Rev. Nutr. 20 (2000) 535–
7. Conclusion 559.
[14] S. Deslex, R. Negrel, C. Vannier, J. Etienne, G. Ailhaud, Int. J. Obes.
In summary, ASCs provide unique opportunities for 11 (1987) 19–27.
[15] H. Hauner, G. Entenmann, M. Wabitsch, D. Gaillard, G. Ailhaud, R.
investigating novel treatments for a vast array of inherited Negrel, E.F. Pfeiffer, J. Clin. Invest. 84 (1989) 663–1670.
and acquired diseases. In addition, ASCs may also provide [16] Y.D. Halvorsen, A. Bond, A. Sen, D.M. Franklin, Y.R. Lea-Currie,
an opportunity to identify new molecular targets for drug D. Sujkowski, P.N. Ellis, W.O. Wilkison, J.M. Gimble, Metabolism
discovery. In this review article, a series of detailed proto- 50 (2001) 407–413.
cols for the isolation, characterization, expansion and dif- [17] P.A. Zuk, M. Zhu, P. Ashjian, D.A. De Ugarte, J.I. Huang, H.
Mizuno, Z.C. Alfonso, J.K. Fraser, P. Benhaim, M.H. Hedrick, Mol.
ferentiation of ASCs has been provided. These protocols Biol. Cell 13 (2002) 4279–4295.
can readily be adapted to adjust for differences in the size [18] F. Guilak, K.E. Lott, H.A. Awad, Q. Cao, K.C. Hicok, B. Fermor,
of the adipose tissue sample. Many important scientific J.M. Gimble, J. Cell. Physiol. 206 (2006) 229–237.
and medical questions remain before the clinical applica- [19] J.M. Gimble, F. Guilak, Curr. Top. Dev. Biol. 58 (2003) 137–160.
tion of ASCs in humans. These will include the develop- [20] M. Rodbell, J. Biol. Chem. 241 (1966) 130–139.
[21] M. Rodbell, J. Biol. Chem. 241 (1966) 3909–3917.
ment of large scale manufacturing methods with [22] M. Rodbell, A.B. Jones, J. Biol. Chem. 241 (1966) 40–142.
appropriate quality assurance and quality control to gener- [23] R.L. Van, C.E. Bayliss, D.A. Roncari, J. Clin. Invest. 58 (1976) 699–
ate cells in compliance with current good manufacturing 704.
practices (cGMP). [24] P. Bjorntorp, M. Karlsson, H. Pertoft, P. Pettersson, L. Sjostrom, U.
Smith, J. Lipid Res. 19 (1978) 316–324.
[25] H. Hauner, G. Entenmann, M. Wabitsch, D. Gaillard, G.
References Ailhaud, R. Negrel, E.F. Pfeiffer, J. Clin. Invest. 84 (1989)
1663–1670.
[1] J.M. Gimble, Expert Opin. Biol. Ther. 3 (2003) 705–713. [26] H. Hauner, M. Wabitsch, E.F. Pfeiffer, Horm. Metab. Res. Suppl. 19
[2] G. Wei, G. Schubiger, F. Harder, A.M. Muller, Stem cells 18 (2000) (1988) 35–39.
409–414. [27] Y.G. Illouz, Plast. Reconstr. Surg. 72 (1983) 591–597.
[3] D. Woodbury, K. Reynolds, I.B. Black, J. Neurosci. Res. 96 (2002) [28] J.H. Moore Jr., J.W. Kolaczynski, L.M. Morales, R.V. Considine, Z.
908–917. Pietrzkowski, P.F. Noto, J.F. Caro, Aesthetic Plast. Surg. 19 (1995)
[4] A.P. Beltrami, K. Urbanek, J. Kajstura, S.M. Yan, N. Finato, R. 335–339.
Bussani, B. Nadal Ginard, F. Silvestri, A. Leri, C.A. Beltrami, New [29] J.F. Lalikos, Y.Q. Li, T.P. Roth, J.W. Doyle, W.E. Matory, W.T.
Engl. J. Med. 344 (2001) 1750–1757. Lawrence, J. Surg. Res. 70 (1997) 95–100.
[5] E. Gussoni, Y. Soneoka, C.D. Strickland, E.A. Buzney, M.K. Khan, [30] M.J. Oedayrajsingh-Varma, S.M. van Ham, M. Knippenberg, M.N.
A.F. Flint, L.M. Kunkel, R.C. Mulligan, Nature 401 (1999) 390–394. Helder, J. Klein-Nulend, T.E. Schouten, M.J. Ritt, F.J. van Milligen,
[6] D.N. Kotton, A. Fine, Cytotherapy 5 (2003) 169–173. Cytotherapy 8 (2006) 166–177.
[7] B.E. Petersen, W.C. Bowen, K.D. Patrene, W.M. Mars, A.K. [31] K. Yoshimura, T. Shigeura, D. Matsumoto, T. Sato, Y. Takaki, E.
Sullivan, N. Murase, S.S. Boggs, J.S. Greenberger, J.P. Goff, Science Aiba-Kojima, K. Sato, K. Inoue, T. Nagase, I. Koshima, K. Gonda,
284 (1999) 1168–1170. J. Cell. Physiol. 208 (2006) 64–76.
[8] M.F. Pittenger, A.M. Mackay, S.C. Beck, R.K. Jaiswal, R. Douglas, [32] D.D. Diascro, R.L. Vogel, T.E. Johnson, K.M. Witherup, S.M.
J.D. Mosca, M.A. Moorman, D.W. Simonetti, S. Craig, D.R. Pitzenberger, S.J. Rutledge, D.J. Prescott, G.A. Rodan, A. Schmidt,
Marshak, Science 284 (1999) 143–147. J. Bone Miner. Res. 13 (1998) 96–106.
[9] D.J. Prockop, Science 276 (1997) 71–74. [33] I. Sekiya, D.C. Colter, D.J. Prockop, Biochem. Biophys. Res. Comm.
[10] A.V. Terskikh, M.C. Easterday, L. Li, L. Hood, M.K. Kornblum, 284 (2001) 411–418.
D.H. Geschwind, I.L. Weissman, Proc. Natl. Acad. Sci. USA 98 [34] S.K. Kang, L.A. Putnam, J. Ylostalo, I.R. Popescu, J. Dufour, A.
(2001) 7934–7939. Belousov, B.A. Bunnell, J. Cell Sci. 117 (2004) 4289–4299.

Das könnte Ihnen auch gefallen