ArticlePDF Available

Effect of extracorporeal shock wave on proliferation and differentiation of equine adipose tissue-derived mesenchymal stem cells in vitro

Authors:

Abstract and Figures

Mesenchymal stem cells are regarded as common cellular precursors of the musculoskeletal tissue and are responsible for tissue regeneration in the course of musculoskeletal disorders. In equine veterinary medicine extracorporeal shock wave therapy (ESWT) is used to optimize healing processes of bone, tendon and cartilage. Nevertheless, little is known about the effects of the shock waves on cells and tissues. Thus, the aim of this study was to investigate the influence of focused ESWT on the viability, proliferation, and differentiation capacity of adipose tissue-derived mesenchymal stem cells (ASCs) and to explore its effects on gap junctional communication and the activation of signalling cascades associated with cell proliferation and differentiation. ASCs were treated with different pulses of focused ESWT. Treated cells showed increased proliferation and expression of Cx43, as detected by means of qRT-PCR, histological staining, immunocytochemistry and western blot. At the same time, cells responded to ESWT by significant activation (phosphorylation) of Erk1/2, detected in western blots. No significant effects on the differentiation potential of the ASCs were evident. Taken together, the present results show significant effects of shock waves on stem cells in vitro.
Content may be subject to copyright.
Am J Stem Cell 2013;2(1):62-73
www.AJSC.us /ISSN:2160-4150/AJSC1301001
Original Article
Effect of extracorporeal shock wave on proliferation and
differentiation of equine adipose tissue-derived
mesenchymal stem cells in vitro
O Raabe1*, K Shell1*, A Goessl1, C Crispens1, Y Delhasse2, A Eva3, G Scheiner-Bobis3, S Wenisch4, S Arnhold1
1Institute of Veterinary -Anatomy, -Histology, and -Embryology, Justus-Liebig University of Giessen, Germany;
2Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine,
German Sport University Cologne, Germany; 3Institute for Veterinary Physiology and Biochemistry, Justus-Liebig
University of Giessen, Germany; 4Department of Veterinary Surgery, Justus-Liebig University of Giessen, Germany.
*Raabe O. and Shell K. contributed equally to this work.
Received January 7, 2013; Accepted February 22, 2013; Epub March 8, 2013; Published March 18, 2013
Abstract: Mesenchymal stem cells are regarded as common cellular precursors of the musculoskeletal tissue and
are responsible for tissue regeneration in the course of musculoskeletal disorders. In equine veterinary medicine
extracorporeal shock wave therapy (ESWT) is used to optimize healing processes of bone, tendon and cartilage. Nev-
ertheless, little is known about the effects of the shock waves on cells and tissues. Thus, the aim of this study was
to investigate the inuence of focused ESWT on the viability, proliferation, and differentiation capacity of adipose
tissue-derived mesenchymal stem cells (ASCs) and to explore its effects on gap junctional communication and the
activation of signalling cascades associated with cell proliferation and differentiation. ASCs were treated with differ-
ent pulses of focused ESWT. Treated cells showed increased proliferation and expression of Cx43, as detected by
means of qRT-PCR, histological staining, immunocytochemistry and western blot. At the same time, cells responded
to ESWT by signicant activation (phosphorylation) of Erk1/2, detected in western blots. No signicant effects on
the differentiation potential of the ASCs were evident. Taken together, the present results show signicant effects
of shock waves on stem cells in vitro.
Keywords: Horse, shock wave, stem cell, proliferation, differentiation
Introduction
Disorders of the musculoskeletal system, such
as osteoarthritis, tendinopathies, and frac-
tures, are the main reasons for the early retire-
ment and euthanasia of horses [1, 2]. A con-
ventional treatment option for these disorders
consists of a combination of stall rest, physio-
therapy, hoof care, and long-term pain manage-
ment using nonsteroidal anti-inammatory
drugs. Other, more recent treatment options
include implantation of cells and tissue-engi-
neered constructs in bone, cartilage and ten-
don [3, 4]. Mesenchymal stem cells are ideal
cells for implantation. Equine ASCs possess
excellent therapeutic potential for tissue regen-
eration, have self-regenerative abilities [5], and
have the potential to differentiate into several
musculoskeletal-related cell lineages [6-8]. In
recent years, several clinical investigations
have revealed promising results in the stem cell
treatment of a variety of orthopedic disorders in
horses [9-13].
An alternative treatment for musculoskeletal
disorders is the application of ESWT. The shock
wave is an acoustic wave that is characterized
by an extremely high amplitude and short rise
time and is followed by a longer, low-magnitude
negative wave [14]. Originally, ESWT was used
for lithotripsy of uroliths and gall stones in
humans and animals [15-18]. Since the early
1990s, ESWT has been commonly applied in
the treatment of musculoskeletal disorders,
such as tendinopathies and osteoarthritis, in
equine medicine [19-26].
The precise therapeutic and biological effects
of ESWT on tissues or cells, however, are not
yet completely understood. It has been sug-
Equine adipose tissue-derived mesenchymal stem cells
63 Am J Stem Cell 2013;2(1):62-73
gested that ESWT induces cell growth and
enzymatic activities [19], leads to an induction
of neovascularisation [27] has an osteostimu-
latory effect [28], and has a direct effect on
membrane permeability and viability of chon-
drocytes and on the structure of cartilage [29].
It is assumed that ESWT could possibly lead to
an activation of endogenous stem cells.
Implantation of stem cells in combination with
shock wave therapy could be considered as a
new, alternative therapy for orthopaedic dis-
eases. In order to evaluate this possibility, we
investigate in the work presented here whether
various doses of ESWT might affect the mor-
phological and biochemical properties of mes-
enchymal stem cells by analysing its effects on
proliferation, apoptosis, and differentiation of
equine ASCs.
Material and methods
The chemical reagents were obtained from PAA
(PAA, Germany) or Sigma (Sigma, Germany)
unless otherwise noted.
Tissue harvest and isolation of ASCs
Subcutaneous adipose tissue was collected
from the region localized above the dorsal glu-
teal muscles of three mixed-breed horses
(aged, mean ± SD, 7.8 ± 2.3 years) as previ-
ously described [7]. The samples were obtained
from horses being slaughtered at the local
abattoir. All samples were collected within 20
min post mortem. ASCs were isolated by colla-
genase type I (Biochrom AG, Germany) diges-
tion as previously described [7].
ESWT
ASCs from passages 1 to 4 were used for ESWT.
Since shock wave conditions have a strong
effect on the behaviour and the intercellular
contacts of the cells [30], adherent cells were
treated after reaching 80% conuence. Cells
with an optimal morphology (spindle-shaped)
and vitality were used for the experiments.
Hypertrophic cells were excluded from the
experiment, because an increased cell size can
be taken as evidence of senescence. The ASCs
with visible intracellular granules were exclud-
ed as well, because this can be a sign for dedif-
ferentiation. The proliferation capacities and
the differentiation potential were randomly
tested.
The control group did not receive any extracor-
poreal shock wave treatment, while
Experimental Group 1 (EG 1000/9) received 9
pulses of 1000 shock waves and Experimental
Group 2 (EG 2000/3) received 3 pulses of
2000 shock waves, according to standard
means of delivery in horse patients [31, 32].
Cell morphology
ASCs were examined using a phase contrast
microscope and the Axiovision image analysis
system (Carl Zeiss, Germany).
MTT assay
Cell viability was evaluated using a colorimetric
MTT assay measuring reduction power
(AppliChem, Germany). The cells (15 x 103 cell/
well) were cultured for 24 h and 48 h at 37°C
with 5% CO2. Thereafter, 0.5 mg/ml of MTT
reagent was added to the Dulbecco’s Modied
Eagle’s Medium (DMEM with 1000 mg/L
Glukose) and the cells were incubated for 4 h at
37°C with 5% CO2. DMSO (200 µl) was then
added to dissolve the water-insoluble formazan
salt. Quantication was performed with a spec-
trophotometer at 570 nm (ELISA reader; Tecan,
Germany).
Senescence assessment (β-galactosidase
assay)
Processes of cell aging can be indicated by
assessing β-galactosidase (β-Gal) activity. For
the determination of changes caused by senes-
cence, cells (15 x 103 cell/well) were harvested
immediately after shock wave application and
incubated for 24 h at 37°C with 5% CO2. Cells
were then washed in 0.1 M phosphate-buffered
saline (PBS) and subsequently lysed in 0.1%
Triton in PBS. After centrifugation at 4°C and
13.000 g for 15 min, samples were incubated
with β-Gal solution for 16 h at 37°C. The absor-
bance of the dye at 570 nm was measured with
a spectrophotometer.
Immunouorescence staining
The immunouorescence staining was per-
formed as previously described [8]. To visualize
the actin cytoskeleton, cells were incubated
with Alexa Fluor 594 phalloidin (6.6 µM nal
concentration). Cell nuclei were counterstained
using the Hoechst nuclear stain H3334. Cell
proliferation was assessed by the detection of
Equine adipose tissue-derived mesenchymal stem cells
64 Am J Stem Cell 2013;2(1):62-73
Ki67 antigen. For the detection of apoptotic
cells, cells were stained with propidium iodide
(1.5 mM nal concentration).
The method of detection of connexin 43 (Cx43)
was similar to that for the detection of Ki67
antigen [8]. The cells were incubated with the
primary antibody (mouse anti-connexin43,
diluted 1:100 by vol.; DAKO, Germany) over-
night at 4°C and were then exposed to the sec-
ondary antibody (goat anti-mouse, diluted
1:200 by vol.; DAKO, Germany) for 30 min.
Fluorescence images of ASCs were obtained
using the Axiovision image analysis system
(Carl Zeiss, Germany).
Preparation of cell lysates
Cells were lysed using a commercially available
cell lysis buffer according to the manufacturer’s
protocol (Cell Signaling Technology, Germany).
All lysis steps were carried out on ice. After cen-
trifugation at 4°C and 13.000 for 15 min, pro-
tein content in the supernatant was determined
using the bicinchoninic acid (BCA) protein assay
reagent kit (Pierce, IL, USA).
SDS-PAGE and immunodetection of Cx43 and
total and activated Erk1/2
A total of 10 µg of proteins from cell lysates
were separated by SDS-PAGE using slab gels
containing 10% acrylamide and 0.3% N,
N′-methylene-bis-acrylamide. Biotinylated
molecular weight markers (Cell Signaling
Technology, Germany) were run parallel. After
electrophoresis, the proteins were electro-blot-
ted at 500 mA for 30 min onto nitrocellulose
membranes (Schleicher & Schuell, Germany).
After blotting, the membranes were incubated
with the appropriate primary antibody in a dilu-
tion of 1:500 (phosphorylated (p-Erk1/2) or
total Erk1/2 (t-Erk1/2) or anti-Cx43 (mouse
monoclonal anti-Cx43, Invitrogen) overnight at
4°C. The visualization of the ERK1/2 proteins
was performed using the appropriate horserad-
ish peroxidase-conjugated IgG (1:2000 in
TBS-T containing 5% nonfat milk) provided by
the enhanced chemiluminescence (ECL) kit
(Amersham-Pharmacia, Germany). The mem-
branes incubated with the C43 antibody were
further incubated with a biotinylated secondary
antibody (goat-anti-mouse IgG, in a dilution of
1:1000, DAKO Hamburg, Germany). Then the
peroxidase conjugated avidine (ABC)-complex
(Vector, Burlinghame, CA, USA) was applied. A
horseradish peroxidase-conjugated anti-biotin
IgG (Cell Signaling Technology, Germany) to
detect the biotinylated molecular weight mark-
ers was included in the incubation medium at a
dilution of 1:2000. After lm exposure, the den-
sity of the resultant bands was analyzed using
a digital documentation system (Adobe
Photoshop CS5, Ireland). As a negative control,
the primary antibody was replaced by non-
immune serum.
In vitro differentiation
Adipogenic and osteogenic differentiation was
performed in a monolayer as described previ-
ously [8]. Cells were plated at a density of 3 x
103 cells/cm2. All cells were incubated in a
humidied atmosphere at 37°C with 5% CO2 for
various periods. The medium was changed
three times a week.
Adipogenic differentiation
ASCs were cultured in DMEM supplemented
with 10% fetal calf serum (FCS), 1% antibiotics
(penicillin 100 U/ml, streptomycin 0.1 mg/ml;
P/S), 1 μM dexamethasone, 10 μM insulin, 0.5
mM 3-isobutyl-1-methylxanthine, and 100 μM
indomethacin. Adipogenic differentiation was
conrmed on day 14 using an Oil Red O stain as
an indicator of intracellular lipid accumulation
and adipocyte-specic gene expression.
Osteogenic differentiation
The cells used for osteogenic differentiation
were cultured in osteogenic medium. DMEM
was supplemented with 10% FCS, 1% P/S, 0.05
mM ascorbic acid-2-phosphate, 10 mM
β-glycerophosphate and 0.1 μM dexamethasone
for 3 weeks. Osteogenesis was demonstrated
by accumulation of mineralized calcium phos-
phate (calcied extracellular matrix, ECM)
assessed by von Kossa stain.
Chondrogenic differentiation
Chondrogenic differentiation was performed in
a 3D culture with a cell density of 3 x 105 cells/
pellet. For determination of chondrogenesis,
the cells were centrifuged in 15 ml Falcon tubes
at 200 g for 5 min. Cells were incubated over-
night at 37°C, 5% CO2 in a humidied atmo-
sphere. Formed cell pellets were cultivated in
chondrogenic differentiation medium supple-
Equine adipose tissue-derived mesenchymal stem cells
65 Am J Stem Cell 2013;2(1):62-73
mented with DMEM, 1% FCS, 1% P/S, 0.05%
ITSx100, 50 µM ascorbic acid, 100 nM dexa-
methasone and 10 ng/ml TGF β1 for 3 weeks.
After culture, pellets were xed in 4% parafor-
maldehyde, embedded in parafn, and cut into
5 µm thin sections. The sections were deparaf-
nised with xylene and ethanol. To detect pro-
teoglycan synthesis as an indicator of cartilage
production, sections were stained with Alcian
blue. Counterstaining was performed with
nuclear fast red.
Quantitative realtime-RT-PCR (qRT-PCR)
Total RNA was extracted using TRI® Reagent
according to the manufacturer’s protocol
(Sigma, Germany). The RNA concentration was
adjusted to 200 ng/µl, treated with a recombi-
nant DNAse I (Roche, Germany), and subse-
quently reverse-transcribed using GeneAmp®
Gold RNA PCR Core Kit according to the
manufacturer’s protocol (Applied Biosystems,
Germany). The PCR primers and annealing con-
ditions are listed in Table 1. Real-time RT-PCR
was carried out on a CFX96 Realtime Cycler
(Bio-Rad, Germany) using IQ SybrGreen
Supermix (Bio-Rad) according to the following
protocol: after an initial 3 min at 90°C ampli-
cation was accomplished by 40 cycles of heat-
ing for 15 sec at 90°C followed by 1 min 60°C,
with a subsequent melting curve. Expression of
gene of interest was mormalized to GAPDH
expression. Data were analyzed using the CFX
Manager software 1.6 (Bio-Rad) applying the
ΔΔCT-method for relative gene expression.
Statistical analysis of data
One-way ANOVA was performed for comparison
of groups. Subsequently, the groups were com-
pared pairwise with the control by the Mann-
Whitney U test followed by Dunnett’s test. For
all tests the statistical software program,
SPSS19.0, was used (IBM, Germany).
Results
Cell morphology
The ASCs showed elongated, spindle-shaped
cell bodies with 2-3 long, slim processes (Figure
1). Only a minor percentage of the shock wave-
treated cells changed their morphology (approx-
imately 10% within EG 1000/9 and approxi-
mately 20% within EG 2000/9). The cell
processes shortened and the cell bodies
gained volume, which may have been caused
by a swelling process (Figure 1).
The visualization of the f-actin cytoskeleton
was accomplished by phalloidin staining, where
the alignment of directed and undirected bers
was assessed. A slightly increased alignment of
Figure 1. Cell morphology of ASCs. Phase-contrast microscopy of untreated ASCs revealed highly spindle-shaped,
broblastic cell morphology. The cells of EG 1000/9 and EG 2000/3 were less spindle-shaped and the cell exten-
sions were shortened. The cytoskeleton of the ASCs was stained with phalloidin.
Equine adipose tissue-derived mesenchymal stem cells
66 Am J Stem Cell 2013;2(1):62-73
the f-actin bers in the shock wave-treated
cells was observed. However, no signicant
alterations could be detected (Figure 1).
Cell proliferation and viability
There was a signicantly increased prolifera-
tion of the stem cells, which was determined
utilizing the proliferation marker Ki67. Thus, the
proliferation in cells of the control group it was
about 25.01 ± 2.5%, in the cells of EG 1000/9
at 33.08 ± 2.4% and in EG 2000/3 at 40.95 ±
4.3% (Figures 2, 3A). The results of the prolif-
eration were conrmed by the MTT assay, so
that a signicant increase of the number of
shock wave-treated cells compared with the
untreated cells was clearly recognizable. Thus,
in the cells of the control group, the absorbance
after 24 h was about 0.45 ± 0.06 and after 48
h 0.98 ± 0.14; in the cells of EG 1000/9 after
24 h 0.73 ± 0.16 and after 48 h 1.28 ± 0.24; in
EG 2000/3 after 24 h 0.84 ± 0.25 and after 48
h 1.92 ± 0.61 (Figure 3B).
Cell senescence and apoptosis
The effect of mechanotransduction of the
shock wave on cell apoptosis was also exam-
ined. The cells of EG 2000/3 showed a signi-
cantly higher amount of cell apoptosis (12 ±
4.7%) than the cells of EG 1000/9 (7.72 ± 2.2%)
or the untreated cells (3.34 ± 0.28%) (Figures
2, 3C).
Cell senescence can be assessed by measur-
ing β-Gal activity. Following, the density of the
dye was measured. Relating to the untreated
cells, the results of the β-Gal assay revealed no
changes for the shock wave application at any
time. Thus, the senescence in cells of the con-
trol group was 0.19 ± 0.01, in the cells of EG
1000/9 at 0.19 ± 0.03 and in EG 2000/3 at
0.2 ± 0.04 (Figure 3D).
In vitro differentiation
The analysis of the differentiation potential
indicates to what extent stem cells can be
directed towards the adipogenic, osteogenic, or
chondrogenic lineage; this can be determined
by applying specic media and factors. Shock
wave-treated cells showed a slightly higher
potential for differentiation than untreated
cells. The adipogenic differentiation in the
treated groups was more pronounced in terms
of both numbers of differentiated cells and size
of fat vacuoles. Fat vacuoles were noted within
3-4 days of shock wave treatment in treated
cells and within 5 days in untreated cells (Figure
4). Following induction of osteogenic differenti-
ation, we observed a clear change according to
the morphology. The shock wave-treated cells
Figure 2. Cell proliferation and apoptosis. Cell proliferation was assessed by immunouorescence with the Ki67
antibody. Upper panels: Turquoise-stained nuclei indicate proliferation. Lower panels: apoptosis was stained with
propidium iodide; pink stained nuclei indicate apoptosis.
Equine adipose tissue-derived mesenchymal stem cells
67 Am J Stem Cell 2013;2(1):62-73
of both treated groups acquired a polygonal
shape within three days and cell aggregates
and nodule formation were observed after 3-5
days of culture; in contrast, this was observed
in the untreated cells after 5-7 days. Matrix
mineralization and calcium accumulation were
also more prominent in the treated groups
(Figure 4). Within a day after seeding the ASCs
in culture for chondrogenesis, three-dimension-
al aggregates were observed in all groups. Both
treated and untreated cell pellets stained posi-
tive for alcian blue, indicating the presence of
sulfated proteoglycans (Figure 4). No positive
staining (for all three differentiation lineages)
was observed in the cells cultured in growth
medium (data not shown). However, the analy-
sis of the adipo-, osteo- and chondrogenesis-
relevant mRNA expression did not show any
signicant differences between the samples
(Figure 5).
Effect of ESWT on Erk1/2
ESW treatment did not affect the expression of
total Erk1/2 (Figure 6; t-Erk1/2). The expres-
sion of p-Erk1/2 (activated Erk1/2), however,
was signicantly different between EG 2000/3
and EG 1000/9 and between treated cells of
the both groups and the untreated cells (Figure
6). In EG 1000/9 cells, p-Erk1/2 was signi-
cantly higher than p-Erk1/2 in untreated or in
EG 2000/3 cells.
Cx43 expression
Cx43 was detected by immunouorescence
staining in the untreated and both treated
groups (Figure 7A). This can be conrmed by
Western blot experiments (Figure 7C) showing
a marginally higher expression of Cx43 in both
treated groups (48.76% and 49.29%) com-
pared to the control group (46.01%). However,
looking at Cx43 mRNA expression a signicant
increase in Cx43 expression was observed in
EG 1000/9 in comparison with the untreated
cells and cells treated by EG 2000/3 (Figure
7B).
Discussion
ASCs have a high therapeutic potential and are
therefore used in various areas of regenerative
Figure 3. Quantitative analysis of cell proliferation, viability, apoptosis, and senescence. The treated cells showed
a better proliferative capability as well as a higher apoptotic rate than the untreated cells. The data were compiled
from triplicate determinations. The MTT assay showed a signicantly higher cell proliferation of the treated cells
than the untreated cells. The β-Gal assay showed that the cells of the three groups had nearly the same level of
senescence. All values reect the arithmetic mean ± standard deviation; a: signicantly higher than control, b: sig-
nicantly higher than EG 1000/9, a, b, c: all samples are signicant with each other.
Equine adipose tissue-derived mesenchymal stem cells
68 Am J Stem Cell 2013;2(1):62-73
medicine. However, the success of this therapy
is limited by the small number of stem cells that
reach the target area. Therefore, the cells have
to be conditioned for their in vivo application or
to be modied for the transplantation niche.
Recently, various forms of shock wave therapy
have been applied in addition to a stem cell
therapy. The mechanisms involved in shock
wave therapy, however, are not yet completely
understood and are often the subject of contro-
versy. Thus, the aim of the present study was to
examine whether behaviour and characteris-
tics of equine ASCs can be modied under the
inuence of shock wave therapy. For that pur-
pose the effects of ESWT on the cytoskeleton
of ASC and on their proliferation and differenti-
ation potential were analyzed. The results
obtained show a distinct mechano-sensitive
reaction of the equine ASCs to the application
of shock wave treatment. Signicantly different
effects were observed between the treated
groups and the untreated group, which were
consistent with results reported by other
researchers [33-35]. Additionally, there are
numerous reports about the effect of ESWT on
different cells (chondrocytes, marrow stromal
cells, tumor cells etc) [36-40]. Similar to our
ndings the authors of these papers report
about an increase in the proliferation rate as
well as about a dose-and impulse dependent
cytotoxic effect. However, the increased prolif-
eration rate especially of stem cells observed
after shock wave application, as investigated in
our study, suggests that the transplantation
results might be inuenced in a positive way if
division- and reproduction speed of stem cells
(proliferation rate) can be stimulated by ESWT
ex vivo prior to transplantation.
It is well known that the cytoskeleton plays an
active role in the process of apoptosis [41].
Thus, loosening the cells from their focal adhe-
Figure 4. Cell differentiation. The adipogenic differentiation was detected by the formation of lipid droplets in the cy-
toplasm of cells stained with Oil Red O. Osteogenic differentiation was demonstrated by calcium deposition stained
with von Kossa stain. Chondrogenic differentiation of ASCs was determined by a transformation from the broblastic
to a chondrocyte-like appearance; formation of GAG-rich matrix and numerous chondrone-like areas were observed.
The center of the pellet from EG 2000/3 became increasingly subjected to necrosis.
Equine adipose tissue-derived mesenchymal stem cells
69 Am J Stem Cell 2013;2(1):62-73
sion complexes as well as reorganization of the
f-actin bers and inhibition of actin dynamics
following cell contraction are some of the rst
signs of apoptosis [18]. Our investigation
showed signicant effects of the application of
shock wave treatment on cell apoptosis with
increasing severity of the shock wave condi-
tions. Furthermore, an alignment of the f-actin
bers was demonstrated by phalloidin staining
after shock wave application. Therefore, a cor-
relation between the dynamics of the f-actin
bre formation and apoptosis can be assumed
that is in accordance with the observations of
other research groups [18]. Furthermore, differ-
ent structures of the ASCs are altered during
shock wave application, and these ndings con-
rm that the use of the wrong shock wave con-
ditions can result in cell death.
Figure 5. qRT-PCR after differentiation. mRNA expression of adipogenic (Pparγ2), osteogenic (AP, OC, SSP1, Runx2),
and chondrogenic markers (Col I, Col III, and Col X). All values reect the arithmetic mean ± standard deviation. RGE:
relative gene expression.
Equine adipose tissue-derived mesenchymal stem cells
70 Am J Stem Cell 2013;2(1):62-73
Aging processes are reected in the morpholo-
gy of cells as well as in the activity of
β-galactosidase [42]; therefore, we measured
β-Gal activity to assess effects of the shock
wave treatment on cellular aging. There were
no differences in β-Gal activity between control
and treated cells, thus allowing to assume that
the treatment did not cause any senescence on
ASCs.
A better differentiation potential of the cells
after shock wave treatment was demonstrated
for the adipogenic, osteogenic, and chondro-
genic lineages (Figure 4) without any detect-
able changes in the mRNA expression of corre-
sponding markers (Figure 5). The shock waves
possibly stimulate the translation of the marker
mRNAs into protein without any direct effect on
mRNA synthesis. These results are strength-
ened by the fact that shock wave treatment
caused an almost 2-fold stimulation of Erk1/2
in the EG 1000/9 group when compared with
the untreated group. This effect was not
observed in the EG 2000/3 group, however,
demonstrating once again the importance of
the correct choice of treatment. Since p-Erk1/2
is involved in the regulation of cell growth and
differentiation, it is justied to assume that the
observed activation of the kinase might be of
signicance for the mitogenic and differentia-
tion responses of the ASCs. Activation of
Erk1/2 in connection with differentiation and
proliferation of mesenchymal stem cells has
been described previously [43, 44].
The analysis of the cell-cell contacts was per-
formed by examining the presence of Cx43, the
most common Cx in mammals [45]. Cx43 has a
Figure 6. Erk1/2 expression. A: Western blot analysis of total- and phosphorylated-Erk1/2 expression in untreated
(1), EG 1000/9 (2), and EG 2000/3 (3) groups. The p-Erk1/2 expression is signicantly higher in the EG 1000/9
than in the control or EG 2000/3 group (n=6; mean ± SEM). The value for the EG 2000/3 group is signicantly
smaller than that of the control group (n=6; mean ± SEM); a: signicantly higher than control, b: signicantly higher
than EG 2000/3.
Equine adipose tissue-derived mesenchymal stem cells
71 Am J Stem Cell 2013;2(1):62-73
role in the regulation of cell differentiation and
cell proliferation [46, 47]. On the protein level,
Cx43 was detected in the cells of untreated as
well as slightly increased in both shock wave-
treated groups, with a slightly higher expres-
sion in the EG 2000/3 group. Cx43-specic
mRNA expression, however, was higher in the
EG 1000/9 group than in EG 2000/3 or in
untreated cells. These ndings actually indicate
that shock wave treatment has the potency to
alter the expression of cell-cell contact proteins
as shown here for the gap junction proteins.
Furthermore, these ndings also conrm the
results for proliferation, and differentiation,
and reveal new aspects for fundamental
research, which might be translated to clinical
applications. Since the multiplication and culti-
vation of ASCs ex vivo allow for the production
of a high number of stem cells that can be sub-
jected to shock waves under ideal conditions
without affecting other cells in the surrounding
tissue, ex vivo pre-conditioning of equine stem
cells by shock wave application followed by the
re-implantation of these cells into tissue lesions
might help to improve the treatment of ortho-
paedic disorders.
Acknowledgments
This work was supported by Richard and
Annemarie Wolf-Foundation.
Address correspondence to: Dr. Stefan Arnhold,
Institute of Veterinary -Anatomy, -Histolog y and –
Embryology, Justus-Liebig University of Giessen,
Frankfurterstrasse 98, 35392 Giessen, Germany.
E-mail: stefan.arnhold@vetmed.uni-giessen.de
References
[1] Silver IA, Brown PN, Goodship AE, Lanyon LE,
McCullagh KG, Perry GC and Williams IF. A clin-
ical and experimental study of tendon injury,
healing and treatment in the horse. Equine Vet
J Suppl 1983 Jul; 1-43.
[2] Marr CM, Love S, Boyd JS and McKellar Q. Fac-
tors affecting the clinical outcome of injuries to
the supercial digital exor tendon in National
Hunt and point-to-point racehorses. Vet Rec
1993; 132: 476-479.
[3] Carrade DD, Owens SD, Galuppo LD, Vidal MA,
Ferraro GL, Librach F, Buerchler S, Friedman
MS, Walker NJ and Borjesson DL. Clinicopatho-
logic ndings following intra-articular injection
of autologous and allogeneic placentally de-
Figure 7. mRNA expression, immunouorescence staining and Western blot analysisof Cx43. Cx43 protein expres-
sion was detected immunocytochemically in all three groups (arrows) (A). The Cx43 mRNA expression is signicantly
higher in the EG 1000/9 than in the control or EG 2000/3 group. All values reect the arithmetic mean ± standard
deviation. RGE: relative gene expression; a: signicantly higher than control, b: signicantly higher than EG 2000/3
(B). Western blot analysis reveals an expression of Cx43 in both experimental groups as well as in the control group
with a slightly higher expression in the ESWG treated groups (C). Lanes 1-3 as in A, lane 4 = equine heart as posi-
tive control.
Equine adipose tissue-derived mesenchymal stem cells
72 Am J Stem Cell 2013;2(1):62-73
rived equine mesenchymal stem cells in hors-
es. Cytotherapy 2011; 13: 419-430.
[4] Nixon AJ, Begum L, Mohammed HO, Huibreg-
tse B, O’Callaghan MM and Matthews GL. Au-
tologous chondrocyte implantation drives early
chondrogenesis and organized repair in exten-
sive full- and partial-thickness cartilage de-
fects in an equine model. J Orthop Res 2011;
29: 1121-1130.
[5] Koch TG, Berg LC and Betts DH. Current and
future regenerative medicine - principles, con-
cepts, and therapeutic use of stem cell therapy
and tissue engineering in equine medicine.
Can Vet J 2009; 50: 155-165.
[6] Vidal MA, Kilroy GE, Lopez MJ, Johnson JR,
Moore RM and Gimble JM. Characterization of
equine adipose tissue-derived stromal cells:
adipogenic and osteogenic capacity and com-
parison with bone marrow-derived mesenchy-
mal stromal cells. Vet Surg 2007; 36: 613-
622.
[7] Raabe O, Reich C, Wenisch S, Hild A, Burg-
Roderfeld M, Siebert HC and Arnhold S. Hydro-
lyzed sh collagen induced chondrogenic dif-
ferentiation of equine adipose tissue-derived
stromal cells. Histochem Cell Biol 2010; 134:
545-554.
[8] Raabe O, Shell K, Wurtz A, Reich CM, Wenisch
S and Arnhold S. Further insights into the char-
acterization of equine adipose tissue-derived
mesenchymal stem cells. Vet Res Commun
2011; 35: 355-365.
[9] Smith RK, Korda M, Blunn GW and Goodship
AE. Isolation and implantation of autologous
equine mesenchymal stem cells from bone
marrow into the supercial digital exor tendon
as a potential novel treatment. Equine Vet J
2003; 35: 99-102.
[10] Wilke MM, Nydam DV and Nixon AJ. Enhanced
early chondrogenesis in articular defects fol-
lowing arthroscopic mesenchymal stem cell
implantation in an equine model. J Orthop Res
2007; 25: 913-925.
[11] Guest DJ, Smith MR and Allen WR. Monitoring
the fate of autologous and allogeneic mesen-
chymal progenitor cells injected into the super-
cial digital exor tendon of horses: prelimi-
nary study. Equine Vet J 2008; 40: 178-181.
[12] Watts AE, Yeager AE, Kopyov OV and Nixon AJ.
Fetal derived embryonic-like stem cells im-
prove healing in a large animal exor tendon-
itis model. Stem Cell Res Ther 2011; 2: 4.
[13] Godwin EE, Young NJ, Dudhia J, Beamish IC
and Smith RK. Implantation of bone marrow-
derived mesenchymal stem cells demon-
strates improved outcome in horses with over-
strain injury of the supercial digital exor
tendon. Equine Vet J 2012; 44: 25-32.
[14] Ogden JA, Toth-Kischkat A and Schultheiss R.
Principles of shock wave therapy. Clin Orthop
Relat Res 2001; 8-17.
[15] Pettersson B and Tiselius HG. Extracorporeal
shock wave lithotripsy of proximal and distal
ureteral stones. Eur Urol 1988; 14: 184-188.
[16] Block G, Adams LG, Widmer WR and Lingeman
JE. Use of extracorporeal shock wave lithotrip-
sy for treatment of nephrolithiasis and uretero-
lithiasis in ve dogs. J Am Vet Med Assoc 1996;
208: 531-536.
[17] Adams LG and Senior DF. Electrohydraulic and
extracorporeal shock-wave lithotripsy. Vet Clin
North Am Small Anim Pract 1999; 29: 293-
302, xv.
[18] Vetterkind S, Illenberger S, Kubicek J, Boosen
M, Appel S, Naim HY, Scheidtmann KH and
Preuss U. Binding of Par-4 to the actin cytoskel-
eton is essential for Par-4/Dlk-mediated apop-
tosis. Exp Cell Res 2005; 305: 392-408.
[19] Haupt G. Use of extracorporeal shock waves in
the treatment of pseudarthrosis, tendinopathy
and other orthopedic diseases. J Urol 1997;
158: 4-11.
[20] Bolt DM, Burba DJ, Hubert JD, Pettifer GR and
Hosgood GL. Evaluation of cutaneous analge-
sia after non-focused extracorporeal shock
wave application over the 3rd metacarpal bone
in horses. Can J Vet Res 2004; 68: 288-292.
[21] Crowe OM, Dyson SJ, Wright IM, Schramme MC
and Smith RK. Treatment of chronic or recur-
rent proximal suspensory desmitis using radial
pressure wave therapy in the horse. Equine Vet
J 2004; 36: 313-316.
[22] McClure SR, Van Sickle D and White MR. Ef-
fects of extracorporeal shock wave therapy on
bone. Vet Surg 2004; 33: 40-48.
[23] McClure SR, VanSickle D, Evans R, Reinertson
EL and Moran L. The effects of extracorporeal
shock-wave therapy on the ultrasonographic
and histologic appearance of collagenase-in-
duced equine forelimb suspensory ligament
desmitis. Ultrasound Med Biol 2004; 30: 461-
467.
[24] Pauwels FE, McClure SR, Amin V, Van Sickle D
and Evans RB. Effects of extracorporeal shock
wave therapy and radial pressure wave thera-
py on elasticity and microstructure of equine
cortical bone. Am J Vet Res 2004; 65: 207-
212.
[25] Revenaugh MS. Extracorporeal shock wave
therapy for treatment of osteoarthritis in the
horse: clinical applications. Vet Clin North Am
Equine Pract 2005; 21: 609-625, vi.
[26] Benson BM, Byron CR, Pondenis H and Stew-
art AA. The effects of radial shock waves on
the metabolism of equine cartilage explants in
vitro. N Z Vet J 2007; 55: 40-44.
Equine adipose tissue-derived mesenchymal stem cells
73 Am J Stem Cell 2013;2(1):62-73
[27] Wang CJ, Wang FS, Yang KD, Weng LH, Hsu CC,
Huang CS and Yang LC. Shock wave therapy
induces neovascularization at the tendon-
bone junction. A study in rabbits. J Orthop Res
2003; 21: 984-989.
[28] Wang FS, Yang KD, Chen RF, Wang CJ and
Sheen-Chen SM. Extracorporeal shock wave
promotes growth and differentiation of bone-
marrow stromal cells towards osteoprogenitors
associated with induction of TGF-beta1. J Bone
Joint Surg Br 2002; 84: 457-461.
[29] Byron CR, Benson BM, Stewart AA and Stewart
MC. Effects of radial shock waves on mem-
brane permeability and viability of chondro-
cytes and structure of articular cartilage in
equine cartilage explants. Am J Vet Res 2005;
66: 1757-1763.
[30] Tarbell JM, Weinbaum S and Kamm RD. Cellu-Cellu-
lar uid mechanics and mechanotransduction.
Ann Biomed Eng 2005; 33: 1719-1723.
[31] Lischer CJ, Ringer SK, Schnewlin M, Imboden I,
Furst A, Stockli M and Auer J. Treatment of
chronic proximal suspensory desmitis in hors-
es using focused electrohydraulic shockwave
therapy. Schweiz Arch Tierheilkd 2006; 148:
561-568.
[32] Waguespack RW, Burba DJ, Hubert JD, Vidal
MA, Lomax LG, Chirgwin SR and Lopez MJ. Ef-
fects of extracorporeal shock wave therapy on
desmitis of the accessory ligament of the deep
digital exor tendon in the horse. Vet Surg
2011; 40: 450-456.
[33] Smith FL, Carper SW, Hall JS, Gilligan BJ, Mad-
sen EL and Storm FK. Cellular effects of piezo-
electric versus electrohydraulic high energy
shock waves. J Urol 1992; 147: 486-490.
[34] Martini L, Giavaresi G, Fini M, Borsari V, Torri-
celli P and Giardino R. Early effects of extracor-
poreal shock wave treatment on osteoblast-
like cells: a comparative study between
electromagnetic and electrohydraulic devices.
J Trauma 2006; 61: 1198-1206.
[35] Lohrer H, Nauck T, Dorn-Lange NV, Scholl J and
Vester JC. Comparison of radial versus focused
extracorporeal shock waves in plantar fasciitis
using functional measures. Foot Ankle Int
2010; 31: 1-9.
[36] Brauner T, Brummer F and Hulser DF. Histopa-
thology of shock wave treated tumor cell sus-
pensions and multicell tumor spheroids. Ultra-Ultra-
sound Med Biol 1989; 15: 451-460.
[37] Brummer F, Brenner J, Brauner T and Hulser
DF. Effect of shock waves on suspended and
immobilized L1210 cells. Ultrasound Med Biol
1989; 15: 229-239.
[38] Haake M, Wessel C and Wilke A. [Effects of ex-
tracorporeal shock waves (ESW) on human
bone marrow cell cultures]. Biomed Tech (Berl)
1999; 44: 278-282.
[39] Dorotka R, Kubista B, Schatz KD and Trieb K.
Effects of extracorporeal shock waves on hu-
man articular chondrocytes and ovine bone
marrow stromal cells in vitro. Arch Orthop Trau-
ma Surg 2003; 123: 345-348.
[40] Leone L, Vetrano M, Ranieri D, Raffa S, Vulpia-
ni MC, Ferretti A, Torrisi MR and Visco V. Extra-
corporeal Shock Wave Treatment (ESWT) im-
proves in vitro functional activities of ruptured
human tendon-derived tenocytes. PLoS One
2012; 7: e49759.
[41] Mills JC, Stone NL and Pittman RN. Extranucle-
ar apoptosis. The role of the cytoplasm in the
execution phase. J Cell Biol 1999; 146: 703-
708.
[42] Stenderup K, Justesen J, Clausen C and Kas-
sem M. Aging is associated with decreased
maximal life span and accelerated senescence
of bone marrow stromal cells. Bone 2003; 33:
919-926.
[43] Risbud MV, Albert TJ, Guttapalli A, Vresilovic EJ,
Hillibrand AS, Vaccaro AR and Shapiro IM. Dif-
ferentiation of mesenchymal stem cells to-
wards a nucleus pulposus-like phenotype in
vitro: implications for cell-based transplanta-
tion therapy. Spine (Phila Pa 1976) 2004; 29:
2627-2632.
[44] Longobardi L, O‘Rear L, Aakula S, Johnstone B,
Shimer K, Chytil A, Horton WA, Moses HL and
Spagnoli A. Effect of IGF-I in the chondrogene-
sis of bone marrow mesenchymal stem cells in
the presence or absence of TGF-beta signal-
ing. J Bone Miner Res 2006; 21: 626-636.
[45] Laird DW. Life cycle of connexins in health and
disease. Biochem J 2006; 394: 527-543.
[46] Bruzzone R, White TW and Paul DL. Connec-
tions with connexins: the molecular basis of
direct intercellular signaling. Eur J Biochem
1996; 238: 1-27.
[47] Krutovskikh V and Yamasaki H. The role of gap
junctional intercellular communication (GJIC)
disorders in experimental and human carcino-
genesis. Histol Histopathol 1997; 12: 761-
768.
... Frontiers in Bioengineering and Biotechnology frontiersin.org 02 affected (Raabe et al., 2013). Interestingly, this study showed that SWA on CB-MSCs maintained their multilineage differentiation potential and even increased their potency toward the adipogenic and osteogenic lineages but not the chondrogenic lineage (Raabe et al., 2013). ...
... 02 affected (Raabe et al., 2013). Interestingly, this study showed that SWA on CB-MSCs maintained their multilineage differentiation potential and even increased their potency toward the adipogenic and osteogenic lineages but not the chondrogenic lineage (Raabe et al., 2013). Recently, cell biological effects of mechanical SWA on human bone marrow stromal cells (hBMSCs) cultured as a monolayer were also reported. ...
... Several reported studies have applied shock waves to cultured cells using a 2D monolayer in flasks (Raabe et al., 2013;Pemberton et al., 2015;Berebichez-Fridman et al., 2017), whereas in our approach, we applied shock waves to a 3D pellet structure of primary MSCs and the MG63 cell line. In our opinion, a 3D pellet cluster of cells is closer to the in vivo situation than adherent 2D monolayer cultured cells. ...
Article
Full-text available
Introduction: Extracorporeal shock wave therapy is a non-invasive and effective option for treating various musculoskeletal disorders. Recent literature indicates that the parameters for extracorporeal shock wave therapy, such as the optimal intensity, treatment frequency, and localization, are yet to be determined. Studies reporting on the effects of shock wave application on primary mesenchymal stromal cells (MSCs) as well as osteoblastic cell lines in vitro are barely available and not standardized. Methods: In this study, we designed a special setup to precisely expose primary MSCs and the osteoblastic cell line MG63 to shock waves and subsequently analyzed the resulting cellular responses using standardized protocols to investigate their viability, proliferation behavior, cytokine secretion, and osteogenic differentiation potential in vitro. The shock wave transducer was coupled to a specifically designed water bath containing a 5 mL tube holder. Primary human MSCs and MG63 cells were trypsinated and centrifuged in a 5 mL tube and exposed to single and repeated shock wave application using different intensities and numbers of pulses. Results: Single treatment of MSCs using intensities 5, 10, 15, and 20 and pulse numbers 100, 250, 500, 750, and 1,000 at a constant pulse repetition frequency of 1 Hz resulted in a decreased viability and proliferation of both cell types with an increase in the intensity and number of pulses compared to controls. No significant difference in the osteogenic differentiation was observed at different time intervals in both cell types when a single shock wave application was performed. However, repeated shock wave sessions over three consecutive days of primary MSCs using low intensity levels 0.1 and 1 showed significant osteogenic differentiation 4-fold higher than that of the extracted Alizarin Red S at day 14, whereas MG63 cells showed no significant osteogenic differentiation compared to their corresponding controls. More specifically, repeated shock wave application triggered a significant downregulation of COL1A1, upregulation of RUNX2, and sustained increase of OCN in primary MSCs but not in the cell line MG63 when induced toward the osteogenic differentiation. Discussion: The effects of shock wave application on MSCs make it an effective therapy in regenerative medicine. We established a protocol to analyze a standardized shock wave application on MSCs and were able to determine conditions that enhance the osteogenic differentiation of MSCs in vitro.
... They found that ADSCs treated with ESWT showed increased proliferation and expression of Cx43 and significant activation (phosphorylation) of Erk1/2. The results demonstrated that ESWT had significant effects on stem cells in vitro [38]. One study revealed that ESWT combined with human or rat ADSCs responded strongly to treatment in vitro. ...
... Growing evidence suggests that shockwave therapy modulates the differentiation potential of stem cells by promoting the proliferation and self-renewal of MSCs in vitro and safely accelerating the cartilage repair process in vivo [38,45,46]. In addition, the degeneration of hip articular cartilage with ONFH promotes the process of ONFH. ...
Article
Full-text available
Background Extracorporeal shockwave therapy (ESWT) and adipose-derived mesenchymal stem cells (ADSCs) have been used clinically for the treatment of osteonecrosis of the femoral head (ONFH). The study elucidated that ESWT, ADSCs, and combination therapy modulated pro-inflammatory cytokines in the articular cartilage and subchondral bone of early rat ONFH. Material and methods ESWT and ADSCs were prepared and isolated for treatment. Micro-CT, pathological analysis, and immunohistochemistry were performed and analysed. Results After treatments, subchondral bone of ONFH was improved in trabecular bone volume (BV/TV) (P < 0.001), thickness (Tb.Th) (P < 0.01 and 0.001), and separation (Tb.Sp) (P < 0.001) and bone mineral density (BMD) (P < 0.001) using micro-CT analysis. The articular cartilage was protected and decreased apoptosis markers after all the treatments. The expression of IL33 (P < 0.001), IL5 (P < 0.001), IL6 (P < 0.001), and IL17A (P < 0.01) was significantly decreased in the ESWT, ADSCs, and Combination groups as compared with ONFH group. The IL33 receptor ST2 was significantly increased after treatment (P < 0.001) as compared with ONFH group. The Combination group (P < 0.01) decreased the expression of IL6 better than the ESWT and ADSCs groups. Conclusion ESWT, ADSCs and combination therapy significantly protected articular cartilage and subchondral bone of early rat ONFH by modulating the expression of pro-inflammatory cytokines including, IL33 and its receptor ST2, IL5, IL6, and IL17A.
... However, the role of Li-ESWT in promoting stem/progenitor cell differentiation, proliferation and traction is unquestionable [94]. Many studies showed that human ADSCs, bone marrow-derived MSCs and human MSCs proliferation and differentiation were significantly activated by Li-ESWT treatment [95,96]. Furthermore, a study by Catalano et al. [97] demonstrated that combination of autologous human ADSCs and Li-ESWT therapy was shown to improve bone tissue repair in tissue engineering procedures. ...
Article
Full-text available
Erectile dysfunction (ED) is the most common sexual dysfunction disease in adult males. ED can be caused by many factors, such as vascular disease, neuropathy, metabolic disturbances, psychosocial causes, and side effects of medications. Although current oral phosphodiesterase type 5 inhibitors can achieve a certain effect, they cause temporary dilatation of blood vessels with no curative treatment effects. Emerging targeted technologies, such as stem cell therapy, protein therapy, and low-intensity extracorporeal shock wave therapy (Li-ESWT), are being used to achieve more natural and long-lasting effects in treating ED. However, the development and application of these therapeutic methods are still in their infancy, and their pharmacological pathways and specific mechanisms have not been fully discovered. This article reviews the preclinical basic research progress of stem cells, proteins, and Li-ESWT therapy, as well as the current status of clinical application of Li-ESWT therapy.
... In a study on the improvement of ACL reconstruction by extracorporeal shockwave, tibial tunnel enlargement was significantly reduced in the EWST group and there was reason to believe that ESWT could improve ACL reconstruction through vascular and tissue regeneration (Wang et al., 2014). Additionally, ESWT has an effect on stem cells in vitro, giving better potential for the differentiation of stem cells into lipogenic, osteogenic and fibrogenic cartilage (Raabe et al., 2013). Although the biological role of ESWT has been demonstrated in in vitro studies, the clinical value of ESWT in human tendinopathy awaits further clinical trials. ...
Article
Full-text available
Injuries at the tendon-bone interface are very common in the field of sports medicine, and healing at the tendon-bone interface is complex. Injuries to the tendon-bone interface can seriously affect a patient’s quality of life, so it is essential to restore stability and promote healing of the tendon-bone interface. In addition to surgical treatment, the healing of tendons and bones can also be properly combined with extracorporeal stimulation therapy during the recovery process. In this review, we discuss the effects of extracorporeal shock waves (ESWs), low-intensity pulsed ultrasound (LIPUS), and mechanical stress on tendon-bone healing, focusing on the possible mechanisms of action of mechanical stress on tendon-bone healing in terms of transcription factors and biomolecules. The aim is to provide possible therapeutic approaches for subsequent clinical treatment.
Article
Biologic therapies are becoming increasingly utilized by veterinarians. The literature regarding the interaction of biologic therapies with other therapeutics is still in its infancy. Initial studies have examined the effects of exercise, stress, various pharmaceutical interventions, extracorporeal shockwave, therapeutic laser, and hyperbaric oxygen on biologic therapies. Continued research is imperative as owners and veterinarians increasingly choose a multimodal approach to injury and illness. Further, understanding the effects of concurrently administered treatments and pharmaceuticals as well as the health status of the horse is imperative to providing the optimal therapeutic outcome.
Article
Regenerative medicine is used in the canine to optimize tissue healing and treat osteoarthritis and soft tissue injuries. Rehabilitation therapy is also often implemented in the treatment and management of musculoskeletal conditions in the canine. Initial experimental studies have shown that regenerative medicine and rehabilitation therapy may work safely and synergistically to enhance tissue healing. Although additional study is required to define optional rehabilitation therapy protocols after regenerative medicine therapy in the canine, certain fundamental principles of rehabilitation therapy still apply to patients treated with regenerative medicine.
Article
Full-text available
Over the past three decades, electrohydraulic extracorporeal shock wave therapy (ESWT) as a treatment modality for equine orthopaedic disorders has sparked exponential interest among practitioners, but its clinical applications are quickly evolving and a current review highlighting modernised equine clinical use is lacking. The objective of this review is to summarise the most current ESWT technology, evidence for its use, proposed mechanisms of action and clinical applications in horses while also highlighting the areas requiring further investigation. The three ways to generate a shock wave are through electrohydraulic, electromagnetic or piezoelectric mechanisms, but over the last decade, electrohydraulic systems have predominated due to the ability to focus and control a therapeutic waveform. Shock waves' primary physical effect is believed to be via mechanotransduction leading to cellular activation and downstream signalling. Experimentally, ESWT's effects on osseous, connective tissue and wound healing via various mechanisms of action have been reported both in the human and veterinary literature. Clinical trials have investigated ESWT's orthopaedic application including osteoarthritis, thoracolumbar pain, navicular syndrome, tendinopathy and proximal suspensory desmopathy, with its concomitant use with biologics representing an area of active research. Direct ESWT protocol comparisons in terms of long‐term efficacy with variables of energy, depth and exposed tissue types are still lacking with evidence‐based recommendations being largely anecdotal. Technical advancements to facilitate the safe and judicious use of ESWT include human and equine hearing protection, light sedation and/or patient restraint. Efforts to ensure the safe and judicious use of ESWT and its analgesic effects are ongoing.
Article
Full-text available
Zusammenfassung Die extrakorporale Stoßwellentherapie (ESWT) bedient sich akustischer Wellen, um biologische Wirkungen im Zielgewebe zu erreichen. Seit ihrer ersten klinischen Anwendung im Jahr 1980 ist eine Reihe an biologischen Antworten der ESWT mechanistisch aufgeklärt worden, so u. a. eine Stammzellaktivierung, antifibrotische Effekte, Effekte auf den verbesserten Lymphabfluss und weitere mehr. Zur Cellulite liegen mithin 12 klinische kontrollierte Studien vor, die positive Effekte der ESWT mit radialen wie auch fokussierten ESWT-Technologien bestätigen. Auch die Stimulation von autologem Fettgewebe mit „adipose-derived stem cells“ durch die ESWT ist mit einigen experimentellen Studien belegt. Insofern erscheint die ESWT als nebenwirkungsfreie Therapieform in der Hand des sachkundigen ESWT-Anwenders eine hochattraktive Therapieform zu sein, die auch in der Kombination mit anderen Therapieverfahren sinnvoll ist.
Article
Objective: To determine the outcomes and mechanisms of microenergy acoustic pulse (MAP) therapy in an irreversible rat model of female stress urinary incontinence. Materials and methods: Twenty-four female Sprague-Dawley rats were randomly assigned into four groups: sham control (sham), vaginal balloon dilation and ovariectomy (VBDO), VBDO + β-aminopropionitrile (BAPN), and VBDO + β-aminopropionitrile treated with MAP (MAP). MAP therapy was administered twice per week for 4 weeks. After a 1-week washout period, all 24 rats were evaluated with functional and histological studies. The urethral vascular plexus was examined by immunofluorescence staining with antibodies against collagen IV and von Willebrand factor (vWF). The urethral smooth muscle stem/progenitor cells (uSMPCs) were isolated and functionally studied in vivo and in vitro. Results: Functional study with leak point pressure (LPP) measurement showed that the MAP group had significantly higher LPPs compared to VBDO and BAPN groups. MAP ameliorated the decline in urethral wall thickness and increased the amount of extracellular matrix within the urethral wall, especially in the urethral and vaginal elastic fibers. MAP also improved the disruption of the urethral vascular plexus in the treated animals. In addition, MAP enhanced the regeneration of urethral and vaginal smooth muscle, and uSMPCs could be induced by MAP to differentiate into smooth muscle and neuron-like cells in vitro. Conclusion: MAP appears to restore urethral wall integrity by increasing muscle content in the urethra and the vagina and by improving the urethral vascular plexus and the extracellular matrix.
Article
Full-text available
Adjacent cells share ions, second messengers and small metabolites through intercellular channels which are present in gap junctions. This type of intercellular communication permits coordinated cellular activity, a critical feature for organ homeostasis during development and adult life of multicellular organisms. Intercellular channels are structurally more complex than other ion channels, because a complete cell-to-cell channel spans two plasma membranes and results from the association of two half channels, or connexons, contributed separately by each of the two participating cells. Each connexon, in turn, is a multimenc assembly of protein subunits. The structural proteins comprising these channels, collectively called connexins, are members of a highly related multigene family consisting of at least 13 members. Since the cloning of the first connexin in 1986, considerable progress has been made in our understanding of the complex molecular switches that control the formation and permeability of intercellular channels. Analysis of the mechanisms of channel assembly has revealed the selectivity of inter-connexin interactions and uncovered novel characteristics of the channel permeability and gating behavior. Structure/function studies have begun to provide a molecular understanding of the significance of connexin diversity and demonstrated the unique regulation of connexins by tyrosine kinases and oncogenes. Finally, mutations in two connexin genes have been linked to human diseases. The development of more specific approaches (dominant negative mutants, knockouts, transgenes) to study the functional role of connexins in organ homeostasis is providing a new perception about the significance of connexin diversity and the regulation of intercellular communication.
Article
Full-text available
The execution phase is the “active” phase of apoptosis occurring immediately after a cell commits to the death program. It lasts about an hour and is characterized by the hallmark morphologic features of apoptosis (e.g., membrane blebbing, chromatin condensation, and DNA fragmentation) culminating with disassembly and packaging of the cell for phagocytosis. Most studies of the execution phase have focused on elucidating nuclear events with the identification of important mechanisms responsible for nuclear execution such as the link between release of cytochrome c, activation of caspase 9/3, and DNA fragmentation (Li et al. 1997; Liu et al. 1997; Enari et al. 1998). Study of cytoplasmic or extranuclear events, on the other hand, has lagged. However, within the past two years, insights into underlying mechanisms and biochemical regulation have led to greatly increased interest in execution phase events occurring outside the nucleus. Despite the recent interest in the extranuclear execution phase, there has been no previous review of the literature, and, at first glance, the various studies appear relatively disparate. However, by subdividing the execution phase into three sequential phases, almost all the data obtained on extranuclear execution phase events can be organized into a relatively coherent paradigm (Fig. 1). In the model we propose, the first stage is release. As most cells enter the execution phase, they release extracellular matrix (ECM)1 attachments and reorganize focal adhesions (FA), adopting a more “rounded” morphology. This outward change correlates with loss of stress fibers (if present) and a reorganization of actin into a peripheral (cortical), membrane-associated ring. Microtubule disassembly also occurs in this stage. The blebbing stage begins with myosin II–dependent contraction of the actin ring followed by a period of sustained, dynamic plasma membrane protrusion and retraction. It continues until finally the cell enters the condensation stage, which is characterized by condensation into small apoptotic bodies or into a single, shrunken ball, correlating with dissolution of polymerized actin. This article will review advances in extranuclear execution phase events within the context of these stages. The goal of the proposed organizational scheme is not to be all-inclusive but, rather, to offer a temporal and structural framework within which nonnuclear execution phase events can be classified. Figure 1 Scheme for temporal and mechanistic organization of the cytoplasmic execution phase. (Top) Morphological view. (Bottom) Corresponding actin rearrangements.
Article
Full-text available
In vitro models of human tenocytes derived from healthy as well as from ruptured tendons were established, characterized and used at very early passage (P1) to evaluate the effects of Extracorporeal Shock Wave Treatment (ESWT). The molecular analysis of traditional tenocytic markers, including Scleraxis (Scx), Tenomodulin (Tnm), Tenascin-C (Tn-C) and Type I and III Collagens (Col I and Col III), permitted us to detect in our samples the simultaneous expression of all these genes and allowed us to compare their levels of expression in relationship to the source of the cells and treatments. In untreated conditions, higher molecular levels of Scx and Col I in tenocytes from pathological compared to healthy samples have been detected, suggesting - in the cells from injured tendon - the natural trigger of an early differentiation and repairing program, which depends by Scx and requires an increase in collagen expression. When ESWT (at the dose of 0.14 mJ/mm(2)) was applied to cultured tenocytes explanted from injured source, Scx and Col I were significantly diminished compared to healthy counterpart, indicating that such natural trigger maybe delayed by the treatment, in order to promote cellular repair. Herein, we show for the first time that ESWT enhances in vitro functional activities of ruptured tendon-derived tenocytes, such as proliferation and migration, which could probably contributes to tendon healing in vivo.
Article
Extracorporeal shock-wave (ESW) treatment hasbeen shown to be effective in promoting the healing of fractures. We aimed to determine whether ESW could enhance the growth of bone-marrow osteoprogenitor cells. We applied ESW to the left femur of rats 10 mm above the knee at 0.16 mJ/mm ² in a range of between 250 and 2000 impulses. Bone-marrow cells were harvested after ESW for one day and subjected to assessment of colony-forming unit (CFU) granulocytes, monocytes, erythocytes, megakaryocytes (CFU-Mix), CFU-stromal cells (CFU-S) and CFU-osteoprogenitors (CFU-O). We found that the mean value for the CFU-O colonies after treatment with 500 impulses of ESW was 168.2 CFU-O/well (sem 11.3) compared with 88.2 CFU-O/well (sem 7.2) in the control group. By contrast, ESW treatment did not affect haematopoiesis as shown by the CFU-Mix (p = 0.557). Treatment with 250 and 500 impulses promoted CFU-O, but not CFU-Mix formations whereas treatment with more than 750 impulses had an inhibiting effect. Treatment with 500 impulses also enhanced the activity of bone alkaline phosphatase in the subculture of CFU-O (p< 0.01), indicating a selective promotion of growth of osteoprogenitor cells. Similarly, formation of bone nodules in the long-term culture of bone-marrow osteoprogenitor cells was also significantly enhanced by ESW treatment with 500 impulses. The mean production of TGF-β1 was 610 pg/ml (sem 84.6) in culture supernatants from ESW-treated rats compared with 283 pg/ml (sem 36.8) in the control group. Our findings suggest that optimal treatment with ESW could enhance rat bone-marrow stromal growth and differentiation towards osteoprogenitors presumably by induction of TGF-β1.
Article
Objective: To characterize equine adipose tissue-derived stromal cell (ASC) frequency and growth characteristics and assess of their adipogenic and osteogenic differentiation potential. Study design: In vitro experimental study. Animals: Horses (n=5; aged, 9 months to 5 years). Methods: Cell doubling characteristics of ASCs harvested from supragluteal subcutaneous adipose tissue were evaluated over 10 passages. Primary, second (P2), and fourth (P4) passage ASCs were induced under appropriate conditions to undergo adipogenesis and osteogenesis. Limit dilution assays were performed on each passage to determine the frequency of colony-forming units with a fibroblastic (CFU-F) phenotype and the frequency of ASC differentiation into the adipocyte (CFU-Ad) and osteoblast (CFU-Ob) phenotype. Results: ASC isolates exhibited an average cell-doubling time of 2.1+/-0.9 days during the first 10 cell doublings. Approximately 1 in 2.3+/-0.4 of the total stromal vascular fraction nucleated cells were ASCs, based on the CFU-F assays, and 1 in 3.6+/-1.3 expressed alkaline phosphatase, an osteogenic marker. Primary ASCs differentiated in response to adipogenic (1 in 4.9+/-5.4, CFU-Ad) and osteogenic (1 in <2.44, CFU-Ob) inductive conditions and maintained their differentiation potential during subsequent passages (P2 and P4). Conclusion: The frequency, in vitro growth rate, and adipogenic and osteogenic differentiation potential of equine ASCs show some differences to those documented for ASCs in other mammalian species. Clinical relevance: Adipose tissue is a potential source of adult stem cells for tissue engineering applications in equine veterinary medicine.
Article
Objective: To evaluate the effects of extracorporeal shock wave therapy (ESWT) on collagenase-induced lesions in the accessory ligament of the deep digital flexor tendon (ALDDFT) of horses. Study Design: Paired, blinded controlled study. Animals: Eight Thoroughbred horses (3 mares, 5 geldings; mean±SD weight, 464±26 kg, mean age, 8±1.7 years). Methods: Lesions were created in both ALDDFTs of all horses by injection of 2 × 103 IU of collagenase type I. Percent lesion and structure (fiber alignment and echogenicity) were quantified with ultrasonographic imaging 3, 6, and 9 weeks after collagenase injection. After ultrasound examinations, ESWT (1000 shocks at 0.15 mJ/mm2) was applied to 1 ALDDFT in each horse. ALDDFT were harvested 15 weeks after collagenase injection and the microstructure, mRNA levels of collagen types I and III, and collagen and glycosaminoglycan content were evaluated. Results: There were no differences in percent lesion, echogenicity, or fiber alignment between control- and ESWT-treated ligaments at each evaluation time; however, compared with 3-week values, there was a significant increase in percent lesion and echogenicity for EWST treated ligaments at 6 weeks and significant decrease in both variables for treated and control ligaments at 12 weeks. Fiber alignment improved significantly at 9 weeks in controls and at 12 weeks in treated and control ligaments. Collagen type I mRNA levels were significantly higher in the ESWT treatment group compared with the control group 15 weeks after collagenase injection though differences in other mRNA levels, microstructure, and composition were not significant. Conclusions: Our results do not support an effect of ESWT on collagenase-induced lesions in the equine ALDDFT.
Article
Mesenchymal stem cells (MSCs) provide an important source of pluripotent cells for musculoskeletal tissue repair. This study examined the impact of MSC implantation on cartilage healing characteristics in a large animal model. Twelve full-thickness 15-mm cartilage lesions in the femoropatellar articulations of six young mature horses were repaired by injection of a self-polymerizing autogenous fibrin vehicle containing mesenchymal stem cells, or autogenous fibrin alone in control joints. Arthroscopic second look and defect biopsy was obtained at 30 days, and all animals were euthanized 8 months after repair. Cartilage repair tissue and surrounding cartilage were assessed by histology, histochemistry, collagen type I and type II immunohistochemistry, collagen type II in situ hybridization, and matrix biochemical assays. Arthroscopic scores for MSC-implanted defects were significantly improved at the 30-day arthroscopic assessment. Biopsy showed MSC-implanted defects contained increased fibrous tissue with several defects containing predominantly type II collagen. Long-term assessment revealed repair tissue filled grafted and control lesions at 8 months, with no significant difference between stem cell-treated and control defects. Collagen type II and proteoglycan content in MSC-implanted and control defects were similar. Mesenchymal stem cell grafts improved the early healing response, but did not significantly enhance the long-term histologic appearance or biochemical composition of full-thickness cartilage lesions.
Article
Mesenchymal stem (progenitor; stromal) cell (MSC) therapy has gained popularity for the treatment of equine tendon injuries but without reports of long-term follow-up. To evaluate the safety and reinjury rate of racehorses after intralesional MSC injection in a large study of naturally occurring superficial digital flexor tendinopathy and to compare these data with those published for other treatments. Safety was assessed clinically, ultrasonographically, scintigraphically and histologically in a cohort of treated cases: 141 client-owned treated racehorses followed-up for a minimum of 2 years after return to full work. Reinjury percentages were compared to 2 published studies of other treatments with similar selection criteria and follow-up. The number of race starts, discipline, age, number of MSCs injected and interval between injury and treatment were analysed. There were no adverse effects of the treatment with no aberrant tissue on histological examination. The reinjury percentage of all racehorses with follow-up (n = 113) undergoing MSC treatment was 27.4%, with the rate for flat (n = 8) and National Hunt (n = 105) racehorses being 50 and 25.7%, respectively. This was significantly less than published for National Hunt racehorses treated in other ways. No relationship between outcome and age, discipline, number of MSCs injected or injury to implantation interval was found. Whilst recognising the limitations of historical controls, this study has shown that MPC implantation is safe and appears to reduce the reinjury rate after superficial digital flexor tendinopathy, especially in National Hunt racehorses. This study has provided evidence for the long-term efficacy of MSC treatment for tendinopathy in racehorses and provides support for translation to human tendon injuries.