ArticlePDF Available

Isolation, characterization and differentiation of mesenchymal stem cells from amniotic fluid, umbilical cord blood and Wharton's jelly in the horse

Authors:

Abstract and Figures

Mesenchymal stem cells (MSCs) have been derived from multiple sources of the horse including umbilical cord blood (UCB) and amnion. This work aimed to identify and characterize stem cells from equine amniotic fluid (AF), CB and Wharton's Jelly (WJ). Samples were obtained from 13 mares at labour. AF and CB cells were isolated by centrifugation, while WJ was prepared by incubating with an enzymatic solution for 2  h. All cell lines were cultured in DMEM/TCM199 plus fetal bovine serum. Fibroblast-like cells were observed in 7/10 (70%) AF, 6/8 (75%) CB and 8/12 (66.7%) WJ samples. Statistically significant differences were found between cell-doubling times (DTs): cells isolated from WJ expanded more rapidly (2.0±0.6 days) than those isolated from CB (2.6±1.3 days) and AF (2.3±1.0 days) (P<0.05). Positive von Kossa and Alizarin Red S staining confirmed osteogenesis. Alcian Blue staining of matrix glycosaminoglycans illustrated chondrogenesis and positive Oil Red O lipid droplets staining suggested adipogenesis. All cell lines isolated were positive for CD90, CD44, CD105; and negative for CD34, CD14 and CD45. These findings suggest that equine MSCs from AF, UCB and WJ appeared to be a readily obtainable and highly proliferative cell lines from a uninvasive source that may represent a good model system for stem cell biology and cellular therapy applications in horses. However, to assess their use as an allogenic cell source, further studies are needed for evaluating the expression of markers related to cell immunogenicity.
Content may be subject to copyright.
REPRODUCTION
RESEARCH
Isolation, characterization and differentiation of mesenchymal
stem cells from amniotic fluid, umbilical cord blood and
Wharton’s jelly in the horse
Eleonora Iacono, Lara Brunori, Alessandro Pirrone, Pasquale Paolo Pagliaro
1
, Francesca Ricci
1
,
Pier Luigi Tazzari
1
and Barbara Merlo
Department of Veterinary Medical Sciences, University of Bologna, via Tolara di Sopra 50, 40064 Ozzano Emilia,
Bologna, Italy and
1
Immunohaematology and Transfusion Center, Policlinico S.Orsola-Malpighi, Bologna, Italy
Correspondence should be addressed to E Iacono; Email: eleonora.iacono2@unibo.it
Abstract
Mesenchymal stem cells (MSCs) have been derived from multiple sources of the horse including umbilical cord blood (UCB) and amnion.
This work aimed to identify and characterize stem cells from equine amniotic fluid (AF), CB and Wharton’s Jelly (WJ). Samples were
obtained from 13 mares at labour. AF and CB cells were isolated by centrifugation, while WJ was prepared by incubating with an
enzymatic solution for 2 h. All cell lines were cultured in DMEM/TCM199 plus fetal bovine serum. Fibroblast-like cells were observed in
7/10 (70%) AF, 6/8 (75%) CB and 8/12 (66.7%) WJ samples. Statistically significant differences were found between cell-doubling times
(DTs): cells isolated from WJ expanded more rapidly (2.0G0.6 days) than those isolated from CB (2.6G1.3 days) and AF (2.3G1.0 days)
(P!0.05). Positive von Kossa and Alizarin Red S staining confirmed osteogenesis. Alcian Blue staining of matrix glycosaminoglycans
illustrated chondrogenesis and positive Oil Red O lipid droplets staining suggested adipogenesis. All cell lines isolated were positive for
CD90, CD44, CD105; and negative for CD34, CD14 and CD45. These findings suggest that equine MSCs from AF, UCB and WJ appeared
to be a readily obtainable and highly proliferative cell lines from a uninvasive source that may represent a good model system for stem cell
biology and cellular therapy applications in horses. However, to assess their use as an allogenic cell source, further studies are needed for
evaluating the expression of markers related to cell immunogenicity.
Reproduction (2012) 143 455–468
Introduction
Over the past decade, stem cell research has emerged
as an area of major interest due to its potential in
regenerative medicine applications (Stocum 2001). The
three alternative stem cells are embryonic stem cells
(ESCs), adult stem cells (ASCs) and induced pluripotent
stem (IPS) cells.
ESCs are derived from blastocysts that propagate
readily (Thomson et al. 1998) and are capable of forming
embryoid bodies that generate a variety of specialized
cells including neural, cardiac and pancreatic cells
(Reubinoff et al. 2000). The establishment of horse ESC
lines is of interest for creation of models of human
genetic diseases and cell transplantation therapies.
Horse ESC lines would also be useful in the genetic
engineering of these animals for improved production
traits and products for disease resistance and biopharm-
ing. However, horses have the disadvantages of low
embryo numbers harvested during in vivo blastocysts
collection or in vitro embryo production (HacKett &
Fortier 2011). Saito et al. (2002) isolated for the first time
ES-like cells from equine blastocysts and induced their
in vitro differentiation. In human, different authors
observed that after ESCs in vivo implantation teratomas
have been developed. Li et al. (2006) injected equine
ES-like cells into several combined immune deficient
mice and observed no teratoma formation. However,
tumour formation by stem cells is more common
when they are injected into the same species from
which they were derived (Erdo et al. 2003). Recently,
the injection of undifferentiated equine ES-like cells
into experimentally induced tendon lesions has been
reported by Guest et al. (2010). In the study, at no time
was there any evidence of teratoma formation by the
horse ESCs, or differentiation into unwanted cell types;
however, longer follow-up periods in larger numbers
of horses are required to demonstrate full safety.
Accordingly, many scientists have focused on alterna-
tive sources of stem cells from various mesenchymal
tissues, such as bone marrow mesenchymal stem cells
(BMMSCs; Horwitz et al. 1999), adipose tissue (Seo
et al. 2005) and umbilical cord (UC; Romanov et al.
2003). MSCs have distinct cell–surface antigen markers.
q2012 Society for Reproduction and Fertility DOI: 10.1530/REP-10-0408
ISSN 1470–1626 (paper) 1741–7899 (online) Online version via www.reproduction-online.org
As reported by the International Society of Cellular
Therapy, MSC populations express CD73, CD90 and
CD105. CD73 is a purine catabolic enzyme with broad
substrate specificity that catalyses the dephosphorylation
of purine and pyrimidine ribo- and deoxyribonucleoside
monophosphates to their corresponding nucleosides
(Naito & Lowenstein 1981); furthermore, CD73 has
been postulated to play a role in cell adhesion (Airas
et al. 1995). CD90, also called Thy-1, is originally
discovered as a thymocyte antigen. Thy-1 can be used as
a marker for a variety of stem cells and for the axonal
processes of mature neurons, even if its function has not
yet been fully elucidated: it has speculated its roles in
cell–cell and cell–matrix interactions, with implication
in neurite outgrowth, nerve regeneration, apoptosis,
metastasis, inflammation and fibrosis (Nakamura et al.
2006). CD105, or endoglobin, is a well-known MSC
marker: it is a part of TGFbreceptor complex. It thus may
be involved in the binding of TGFb1, 3, etc. but it is also
involved in cytoskeletal organization affecting cell
morphology and migration (Dominici et al. 2006).
Additionally, MSCs lack expression of haematopoietic
markers, such as CD14, CD34 and CD45. CD45 is a
pan-leukocyte marker; CD34 markers are primitive
haematopoietic progenitors and endothelial cells; and
CD14 is prominently expressed in monocytes and
macrophages (Dominici et al. 2006).
The difficulties with bone marrow are obtaining tissue
samples from donors and finding a significant decrease
in both quality and differentiation potential of BMMSCs
with age donor. Widely disparate results have been
reported with regard to MSC numbers, differentiation
potency and aging. A negative correlation between
donor age, number and proliferative capacity of MSCs
isolated from young and old donors has been demon-
strated by several authors (Martin et al. 1970,Schneider
& Mitsui 1976,Majors et al. 1997,D’Ippolito et al.
1999,Stenderup et al. 2003,Mareschi et al. 2006), but
widely different results have been obtained with regard
to MSC antigen expression. Recently, Stolzing et al.
(2008) confirmed those already demonstrated in
previous studies (D’Ippolito et al. 1999,Mendes et al.
2002,Chen et al. 2005,Zhang et al. 2005,Zheng
et al. 2007,Kretlow et al. 2008), observing a significant
age-related change in membrane markers expression
levels, an alkaline phosphatase activity, chondrogenic
and myogenic differentiation potency decline with
donor age in contrast with adipogenic differentiation.
However, these data are in contrast with those reported
by other research groups (Bergman et al.1996,
Stenderup et al. 2001,2003,Bellows et al. 2003),
which showed no age-related differences in differen-
tiation potency. Different results obtained are with regard
to the choice of age groups, group size, gender, inclusion
of pathological states, isolation and cultivation con-
ditions (Sethe et al. 2006). In equine medicine, only
Colleoni et al. (2009) reported a significant decline in the
yield of cells from samples of two old horses (8 and 9
years old); however, due to the low number of samples
examined in this study, data need to be further
confirmed.
Beyond natural sources that are limited by stem cell
availability, immune intolerance and lineage specifi-
cation, the latest platform for recently developed
bioengineered stem cells are IPS cells (Nelson et al.
2010). IPS cells generation has been reported for mouse
(Takahashi & Yamanaka 2006), human (Park et al. 2008),
rat (Liao et al. 2009), monkey (Liu et al. 2008)and
recently in the horse (Nagy et al. 2011), suggesting that
virtually any mammalian species can be used for IPS cell
derivation. With the advent of IPS technology, the
limitations related to the use of ESCs and MSCs could
be addressed by the pluripotent potential of bioengi-
neered stem cells that are derived from autologous
sources. Utilizing IPS-based technology, all lineages of
the adult body have become viable targets for replace-
ment. IPS cells enable the ability to genetically repair
sequence defects through homologous recombination,
which then produces healthy stem cells devoid of the
original disease causing genetic impairment. Thereby,
the ability to reproducibly generate unlimited self-
derived progenitors that avoid immune intolerance is a
unique feature. However, the unlimited differentiation
potential of IPS is similar to ESCs, and thus the risk of
dysregulated growth and teratoma formation requires
stringent safeguards (Nelson et al. 2008). Moreover,
beyond the common challenges of natural pluripotent
stem cells, IPS cells also contain genetic modification as
a consequence of the strategy used for reprogramming or
spontaneously acquired cytogenetic abnormalities due
to extensive in vitro manipulation. The long-term
implications of nuclear reprogramming are yet to be
determined as this technology is in the early stages of
development.
The disadvantages of ESCs, BMMSCs and IPS have
accelerated the search for alternative sources of stem
cells as well as UC blood (UCB), Wharton’s jelly (WJ)
and amniotic fluid (AF). In human medicine, the use of
UCB tissue as a source of MSCs can be traced back to
2000: in this year Erices et al. (2000) showed that cells
from UCB were able to give rise to two types of adherent
cells, one of which expressed antigens typical of MSCs.
Equine UCBMSCs have been isolated for the first time by
Koch et al. (2007). Authors observed that isolated cells
were able to differentiate in vitro into osteocyte,
adipocyte and chondrocyte (Koch et al. 2007), but no
studies on their molecular characterization have been
performed. Only recently, Lovati et al. (2011), using
RT-PCR, observed that MSCs from UCB expressed
CD105, CD29 and CD44, and were negative for
haematopoietic marker CD34. No further investigations
have been carried out on equine UCBMSCs molecular
characterization.
456 E Iacono and others
Reproduction (2012) 143 455–468 www.reproduction-online.org
The UC intervascular stroma consists of so-called
‘mucous’ or mesenchymal connective tissue, also
called Wharton’s Jelly (WJ). WJ develops from extra-
embryonic mesoderm, binds and encases the umbilical
vessels, protecting them from twisting and compression
during pregnancy. It also has angiogenic and metabolic
roles for the umbilical circulation. Since 1990, stromal
cells, which basically resemble mesenchymal fibroblasts
found elsewhere during in utero development, were
identified in human WJ (Takechi et al. 1993,Nanaev
et al.1997). Stemness of these cells has been
demonstrated by in vitro differentiation (Lu et al. 2006)
and identification of MSCs membrane-specific markers.
Furthermore, these cells did not express haematopoietic
lineage markers such as CD34, CD45 and CD14
(Wang et al. 2004,Weiss et al. 2006,Secco et al.
2008). In the equine species, WJMSCs have been
isolated in 2007 by Hoynowski et al. Authors identified
isolated cells like MSCs due to their osteogenic,
chondrogenic and adipogenic potency when cultured
in induction media. As suggested by Dominici et al.
(2006), the expression of markers associated with stem
cells from embryonic and ASCs has been observed by
flow cytometry (Hoynowski et al. 2007).
Since 1980, multiple cell types from embryonic and
extra-embryonic tissues were identified in AF (Gosden
1983), but only in 2000 cells with an immunocyto-
chemical profile of mesenchymal, fibroblast/myofibro-
blast cell lineages have been isolated and expanded in
vitro (Kaviani et al. 2001). Prusa et al. (2004) observed
the presence of a cell distinct population expressing
POU5F1 (Oct4), a marker for pluripotent stem cells
known to be expressed in ESCs, in human AF (Pesce &
Scholer 2001). The mechanisms involved in the POU5F1
regulation in ESC have been widely investigated, but in
the adult stage (including post-natal and fetus stages)
POU5F1 function has not been identified even though it
has been associated with the undifferentiated pluripotent
state of stem cell populations derived from various adult
human tissues or organs such as bone marrow-derived
multipotent adult progenitor cells. Furthermore, positive
expression of mesenchymal markers such as CD90,
CD105 and CD73; and negative expression of haema-
topoietic markers such as CD45, CD34 and CD14 have
been observed (In’t Aker et al. 2003,Tsai et al. 2004,De
Coppi et al. 2007). Recently, De Coppi et al. (2007)
showed that human and rodent AF-derived stem cells
(AFMSCs) are positive for c-kit (CD117), that is also
expressed in the heart (Beltrami et al. 2003) and retina
stem cells (Koso et al. 2007), indicating that the c-kit
receptor can identify a stem cell population within
different organs. The c-kit receptor is a protein tyrosine
kinase that binds a cytokine named stem cell factor.
This cytokine and its receptor have been involved
in embryogenesis, carcinogenesis, spermatogenesis,
melanogenesis and play an important role in the
haematopoiesis during embryo development. AFMSCs
are thought to originate from the developing fetus and be
in an intermediate stage between ESCs and lineage-
restricted ASCs (De Coppi et al. 2007), but the exact
origin of these cells is unclear. However, due to their
adipogenic, osteogenic, myogenic, endothelial, neuro-
genic, hepatic, chondrogenic and renal differentiation
potency (Tsai et al. 2004,Perin et al. 2007), AFMSCs
may be a readily available source similar to ESCs for
large numbers of different cells progenitors (De Coppi
et al. 2007). In addition, AFMSCs have not shown
tumorigenicity, thus making them an exciting new
source of regenerative cells (Yo u et al. 2008).
Although there are promising results in human field,
few studies are available on characterization and
differentiation potential of these cells in domestic
animals. The aim of this study was to optimize the
isolation and culture of equine MSCs from AF, UCB and
WJ. Self-renewal ability and multilineage differentiation
capability have been further evaluated. We herein aimed
to widen the knowledge of equine MSCs from fetal fluid
and membranes characterization by testing several
molecular markers by flow cytometry. The markers
tested in the present study were chosen among those
suggested by the International Society for Cytotherapy
(ISC; Dominici et al. 2006). In addition, CD44 was
screened because of its role in MSC migration (Sackstein
et al. 2008) and its correlation with ‘stemness’ in other
MSC populations (Strem et al. 2005). Due to no
commercially available species-specific antibodies exist-
ence to characterize equine stem cells, in the present
study antibodies for human markers were involved using
cross-reactivity of the antibodies among different
species, such as recently reported by Park et al. (2011).
By an increasingly more detailed analysis of the equine
AFMSC phenotypic profile we hope that, as has been
done for human MSCs, a list of minimum cultural and
phenotypic criteria can be drawn up as soon as possible.
Results
Animals
In the equine species, the fetus was normally born in
anterior presentation, dorsal position and extended
(head, neck and forelimbs) posture. Failure to observe
the fluid-filled amnion (which may be visible only during
contractions) protruding from the vulva after 5 min from
the beginning of the second stage of parturition indicates
that we are probably facing a dystocia. In the present
work, AF, UCB and WJ have been recovered from mares
that underwent an eutocic (normal) parturition, without
induction, and which delivered healthy, viable foals, as
showed by an APGAR score R9(Vaala 2006). Samples
were obtained from 13 mares and no complications for
both mares and foals were encountered upon AF, CB and
UC sampling at delivery.
MSCs from equine amniotic fluid 457
www.reproduction-online.org Reproduction (2012) 143 455–468
Sampling and cell culture
AF samples from 10/13 (77.0%) donor mares have
been recovered: on average, we obtained 35.0
G23.5 ml/mare (range: 15–50 ml). We isolated cells
using a Percoll density-gradient centrifugation for elim-
inating urinary crystals, deposits of minerals and small
debris, which are present in AF during last gestation period
and can damage cells during in vitro culture. To obtain
the clear buffy coat layer, AF was diluted with PBS.
After washing pellets were seeded in culture dishes.
Adherent fibroblastoid spindle-shaped cells growing in
monolayer were isolated in 7/10 (70.0%) samples (Fig. 1A).
UCB samples (15–55 ml; mean 36.7G13.9 ml) were
recovered from 8/13 mares (61.5%), due to the early
umbilical vein collapse. From equine UCB, cells have
been isolated by Percoll density-gradient: to obtain a
clear buffy coat, UCB was diluted with PBS and
harvested mononuclear cells were washed several
times to remove platelets and red blood cells. Adherent
spindle-shaped cells have been isolated in 6/8 (75.0%)
samples (Fig. 1B); and in one sample (1/8: 12.5%) no
adherent cells were observed.
Immediately after foal detachment, UC samples have
been recovered (length w15 cm), after 13/13 (100%)
deliveries. UC samples were rinsed in antibiotic and
ethanol solutions and WJ has been isolated: the mean
weight of recovered jelly was 5.0G3.7 g (range: 1.8–
11 g). The jelly was minced and digested using an
enzymatic solution; after washing in PBS plus fetal
bovine serum (FBS), mononuclear cells were seeded in
culture dishes. Adherent cells with MSC morphology
have been isolated in 8/12 (66.7%) samples (Fig. 1C);
and 1/13 (7.7%) of collected UC was not processed for
cell isolation due to the absence of WJ.
The partum canal and delivery environment rep-
resent a non-sterile condition for horses. As a
consequence UC and amniotic tissue are exposed to
significant bacterial and yeasts contaminations. To
prevent from these contaminations, for AF and UCB
sampling, we used sterilized syringes and gloves and
pulled down AF quickly. Despite the precautions used,
in 3/10 (30.0%) AF samples no adherent cells have
been observed due to bacteria (1/3) and yeasts (2/3)
contaminations. On the contrary, only in 1/8 (12.5%)
UCB samples no adherent cells have been observed.
Lower isolation rate due to external contaminations has
been observed in WJ (4/12 samples: 33.3%). In this
study, to avoid contaminations without killing cells,
UC samples were immersed in a 70% ethanol solution
for 10 min but it was not sufficient to completely
eliminate contaminations.
Undifferentiated cells of all lines have been passaged
up to eight times and population-doubling times (DTs)
have been calculated. DT assay showed that AF, UCB
and WJMSCs were able to divide for an extensive period
in vitro. During P0 to P8, AF cells showed a mean DT of
2.3G1.0 days/CD (range: 0.6–4.5 days; Fig. 2A). By P8,
total mean CD was 37.3G3.0 (Fig. 2B). One line of
equine AFMSCs was cultured up to 118 days (15
passages) during which the cells reached 58.7 CD.
Considering the same culture passages (P0–P8), the
mean DT showed by equine UCBMSCs was 2.6
G1.3 days/CD (range: 1.1–5 days; Fig. 2C) and it was
statistically higher (P!0.05) than that showed by equine
AFMSCs (2.3G1.0 days/CD). By P8, UCBMSCs cell-
doubling number (CD) was 34.4G2.3 (Fig. 2D). Adher-
ent spindle-shaped cells isolated from equine WJ
showed a mean DT (2.0G0.6; range: 1.2–4 days;
Fig. 2E) significantly lower than those observed in
A
B
C
Figure 1 Monolayer of rapidly expanding adherent spindle-shaped
fibroblastoid cells compatible with undifferentiated MSC. (A) AF,
(B) UCB and (C) WJ. Magnification !10.
458 E Iacono and others
Reproduction (2012) 143 455–468 www.reproduction-online.org
AFMSCs and UCBMSCs culture (2.0G0.6 vs 2.6
G1.3 days/CD vs 2.3G1.0 days/CD respectively;
P!0.05). In this cell line, by P8, CD was 37.4G2.0
(Fig. 2F). Despite different DTs, no statistically significant
differences have been found in CD among the three
different cell lines. Furthermore, no lag phase has been
observed during in vitro culture of AF, UCB and
WJMSCs: in fact no statistically significant differences
in the number of CD have been found among different
culture passages (PO0.05). The results obtained indicate
that equine AFMSCs, UCBMSCs and WJMSCs had self-
renewal capacity.
5.0
4.5
4.0
3.5
3.0
2.5
2.0
1.5
1.0
0.5
0
4.0
3.5
3.0
2.5
2.0
1.5
1.0
0.5
0
01234
Passage
CD time (days/CD)
5678
45
40
35
30
25
20
15
10
5
0
01234
Passage
Total CD number
5678
6.5
6.0
5.5
5.0
4.5
4.0
3.5
3.0
2.5
2.0
1.5
1.0
0.5
0
01234
Passage
CD time (days/CD)
5678
40
35
30
25
20
15
10
5
0
01234
Passage
Total CD number
5678
01234
Passa
g
e
CD time (days/CD)
Cell doubling time equine
wharton's jelly MSCs (
n
=6)
Total cell doubling number equine
wharton's jelly MSCs (
n
=6)
Cell doubling time equine
wharton's jelly MSCs (
n
=4)
Cell doubling time amniotic
fluid MSCs (
n
=4)
AB
CD
EF
Total cell doubling number equine
equine cord blood MSCs (
n
=4)
Total cell doubling number equine
amniotic fluid MSCs (
n
=4)
5678
45
40
35
30
25
20
15
10
5
0
01234
Passa
g
e
Total CD number
5678
Figure 2 CT and CD of cultured primary and passaged MSCs. All values reflect the meanGS.D. (A and B) AFMSCs: data were compiled from seven
cell cultures. DT of primary cells and subsequent passages was not significantly different (PO0.05). The mean MSC doubling number at P8 was
37.3G3.0. (C and D) UCBMSCs: data were obtained from four cell cultures. DT of primary cells and subsequent passages was not significantly
different (PO0.05). The mean MSC doubling number at P8 was 34.4G2.3. (E and F) WJMSCs: data were compiled from six cell cultures. DT of
primary cells and subsequent passages was not significantly different (PO0.05). The mean MSC doubling number at P8 was 37.4G2.0.
MSCs from equine amniotic fluid 459
www.reproduction-online.org Reproduction (2012) 143 455–468
Immunophenotypic characterization by flow cytometry
ISC postulated that MSCs show positive expression for
CD73, CD90 and CD105; and negative expression for
CD14, CD34 and CD45 (Dominici et al. 2006). As there
are no equine-specific antibodies for flow cytometry, in
the present study we used anti-human antibodies,
routinely used by Immunohaematology and Transfusion
Center equipe, using cross-reactivity of antibodies
among different species. The antibodies efficiency was
verified by performing a control on circulating equine
lymphocytes. As expected, considering the results
obtained with human lymphocytes, adult and haemato-
poietic markers used have not been expressed by these
cells. Unexpected data have been registered for CD45
and CD73 that were negative also for lymphocytes (data
not shown). Furthermore, we compared amino acid
sequences using Basic Local Alignment Search Tool
(Blast). Our results, as summarized in Ta b le 1, showed a
similar expression pattern of MSC phenotype, but
different levels of reactivity of antibodies tested have
been observed. In particular, cells were reactive to
surface markers CD90 and CD105 in all cell lines
evaluated (Fig. 3). MSCs also demonstrated a marker
reaction with CD44 (Table 1 and Fig. 3), a cell–surface
glycoprotein having a role in MSCs migration. Others
markers for haematopoietic cell were not expressed
(Table 1 and Fig. 3). Due to negative lymphocytes CD45
and CD73 expression and the lack of horse CD45 and
CD73 sequence, for these markers cross-reactivity could
not be confirmed, as well as its negative expression by
equine MSCs.
In vitro differentiation
To characterize isolated cells, we further performed
trilineage differentiation assay. According to ISC, we
induced chondrogenic, osteogenic and adipogenic
differentiation culturing each lineage for several days in
induction media.
Chondrogenic induction has been performed in
monolayer culture and after 21 days cell Alcian Blue
staining has been performed to assess differentiation.
Alcian Blue staining is usually used to confirm
mucosubstances and acetic mucins deposition. We
found that induced cells were positive for Alcian Blue
(Fig. 4A). The negative controls kept in regular culture
medium showed no change in their morphology, no cells
stained positive for Alcian Blue (Fig. 4B).
Table 1 Flow cytometry analysis of P3 MSCs from AF, UCB and WJ for the surface markers CD90, CD105, CD73, CD44, CD14, CD34 and CD45.
Tissue CD90 (%) CD105 (%) CD73 (%) CD44 (%) CD14 (%) CD34 (%) CD45 (%)
AF 82.2 78.1 4.0 98.4 0.9 1.8 4.2
UCB 90.3 83.2 0.1 96.9 0.7 7.5 7.9
WJ 93.4 93.2 0.0 97.2 0.2 0.4 0.4
Isotype PE
CD105 CD44
AF CB WJ
CD90
CD90
CD90
CD105
CD105 CD44
CD44
CD34
CD45
CD73 CD73
CD45
CD34
CD73
CD34
CD45
Isotype APC Isotype
Figure 3 Overlay histograms of cytometry
analysis. In black isotypic controls are
represented. Empty histograms represent
the analysis with mAbs on mesenchymal
cell culture.
460 E Iacono and others
Reproduction (2012) 143 455–468 www.reproduction-online.org
To determine osteogenesis, isolated cells were
cultured in osteogenic induction medium and growth
medium was used as a control. Cells were maintained
in induction condition for 3 weeks. Significant calcium
depositions were detected in induced cells compared to
control using Von Kossa staining and Alizarin Red
(Fig. 4C and D).
To show adipogenic potency, cells were cultured
in adipogenic induction medium for 3 weeks. To detect
fat droplets and quantify adipogenesis, we stained cells
with Oil Red O, a fat-soluble dye. Cells maintained
in induction medium resulted positive for Oil Red O, while
undifferentiated control cells did not stain for fat droplets.
UCBMSCs showed a greater adipogenic potential than
AFMSCs and WJMSCs, characterized by a greater
accumulation of lipid vacuoles. Further investigation
using RT-PCR is needed to verify if different differentiation
capacities exist among these three cell lines.
AB
C
EF
GH
D
Figure 4 In vitro differentiation studies.
(A) Chondrogenic induction in equine AF
progenitors over 3 weeks: Alcian Blue
staining of glycosaminoglycans in cartilage
matrix. Magnification !10. (B) Chondro-
genic control: equine AF cells cultured in
regular medium for 21 days maintained
normal morphology and stained negative
for Alcian Blue. Magnification !10.
(C) Osteogenic induction in equine WJ
progenitors over 3 weeks: von Kossa
staining of extensive extracellular calcium
deposition. Magnification !20.
(D) Osteogenic control: equine WJ cells
cultured in standard medium for 21 days
maintained normal morphology and
stained negative for von Kossa staining.
Magnification !10. (E) Osteogenic induction
in equine AF progenitors over 3 weeks:
Alizarin Red S staining of extensive extra-
cellular calcium deposition. Magnification
!10. (F) Osteogenic control: equine AF cells
cultured in regular medium, after 21 days
presented normal morphology and stained
negative for Alizarin Red S staining.
Magnification !10. (G) Adipogenic induction
in equine CB progenitors over 3 weeks: Oil red
O staining of extensive intra-cellular lipid
droplet accumulation. Magnification !20.
(H) Adipogenic control: equine CB cells,
after 21 days of culture in standard
medium presented normal morphology and
stained negative for Oil red O staining.
Magnification !10.
MSCs from equine amniotic fluid 461
www.reproduction-online.org Reproduction (2012) 143 455–468
Discussion
Isolation and characterization of stem cells derived from
various tissues and sources are important issues in stem
cell field. WJ is the mature connective tissue surrounding
umbilical vessels: it is composed of myofibroblast-like
stromal cells, collagen fibres and proteoglycans
(Kobayashi et al. 1998). Recently, researchers have
focused on it as a potential new source of MSCs both
in humans and horses (Hoynowski et al. 2007,
Karahuseyinoglu et al. 2007,Passeri et al. 2009), but
the efficient isolation of cells that truly express MSC
characteristics has been somewhat controversial. Con-
troversial results have also been obtained for cells
isolated from equine UCB (Kock et al. 2007, Cremonesi
et al. 2008), and no report has been found on their
characterization using criteria postulated by ISC in this
species. Due to its contact with the developing fetus, AF
contains large numbers of suspended cells including
stem cells (In’t Anker et al. 2003,De Coppi et al. 2007).
To our knowledge, recently, only two studies have been
published on horse AF-derived progenitor cells, whereas
it is considered a rich source of MSCs in human beings.
We compared WJ, AF and UCB as viable, accessible and
not risky sources of MSCs in equine. In the present study
high initial sample volumes of AF, UCB and tissue have
been easily collected at the delivery, without any
invasiveness for the mare or for the foal, and high
numbers of viable nucleated cells from these sources
have been isolated, except for some failure samples
occurring in yeasts/bacterial contaminations as pre-
viously reported by other authors (Passeri et al. 2009,
Lovati et al. 2011). The isolation rate obtained in the
present study by centrifuging UCB on a Percoll density-
gradient was lower than that obtained by Koch et al.
(2009) using a PrepaCyte-EQ density gradient (100%).
Given the influence that laboratory conditions may
have on the outcome of projects, in the future it would
be appropriate to compare, under the same conditions,
the isolation method used in this study with those
reported by other authors. On the other hand, data
reported for human UCB (Kern et al. 2006) are lower
than that of ours. This difference could be due to
various factors, such as duration of parturition,
recovery protocol, time between sample collection
and processing in the laboratory: these factors
determined viable cells reduction because of their
transition to the fetus or their death.
We cultured cells isolated from equine AF, WJ and
UCB for several days, from P0 to P8, and they showed
a typical MSC morphology, having spindle shape
and adherent characteristics, as postulated by ISC
(Dominici et al. 2006). Regarding cells proliferation,
WJMSCs showed a higher expansion rate compared to
UCBMSCs and AFMSCs: results observed are similar
to those reported in human medicine for UCBMSCs
and WJMSCs (Sarugaser et al. 2005,Karahuseyinoglu
et al. 2007), but not for AFMSCs (Tsai et al. 2004).
Different DTs between human and horse AFMSCs
could be determined during the period of pregnancy in
which the drawing was made. Human AF samples
were recovered by amniocentesis, at the beginning of
the second trimester of pregnancy, therefore, human
AFMSCs could show DT more similar to ESCs.
UCBMSCs DT registered in our laboratory was higher
than that reported by Koch Research Group (Koch
et al. 2007,2009). The difference could be determined
by the higher concentration of FBS used by the authors
during in vitro culture (10 vs 30%). FBS is a mixture
of high-molecular-weight molecules that may promote
or inhibit cell growth and differentiation (Barnes et al.
1987,van der Valk et al. 2004); however, this
composition is largely unknown in both qualitative
and quantitative terms, and moreover FBS may be a
vehicle of known pathogens and unknown infectious
agents (Schallmoser et al. 2007). Different from that
observed by Lovati et al. (2011) and Eloit (1999),inour
study all cell lineages showed a similar fold expansion
during all passages (P0–P8) and WJMSCs were not
affected by the trypsin detachment as reported by the
other authors. The discrepancy in WJMSCs expansion
is likely due not only to different culture conditions,
but also to the different selections of cord tissue to be
processed. The immature cells that retain the ability to
proliferate were located close to the amniotic surface,
whereas highly differentiated, non-proliferating fibro-
blasts were located in closer proximity to the umbilical
vessels (Nanaev et al. 1997): in our study, only cord
jelly, closer to amniotic surface, was digested, while
Lovati et al. (2011) did not distinguish between near or
far from umbilical vessel portion thus the cell
population cultured was very heterogeneous and this
could explain the lower cell doubling.
In the present study, we also demonstrated the
multipotent capability of equine AFMSCs, UCBMSCs
and WJMSCs. The choice of lineages differentiation
was made based on the principles established by the
ISC (Dominici et al.2006). Chondrogenic and
osteogenic differentiations were carried out with a
view to future clinical application of these cells in
injured horses, as previously supported by Berg et al.
(2009). All three cell lines differentiated in chondro-
blasts, osteoblasts and adipocytes, as demonstrated by
staining, performed after 21 days of culture in
appropriate medium; control cells did not stain at all
and only the background colour remained. While a
lack, also testing different protocol, in adipogenic
efficiency of equine AFMSCs has been reported in a
previous study (Park et al. 2011), in our experiment
equine AFMSCs showed an adipogenic potential
similar to that observed in equine WJMSCs, while
UCBMSCs seem to show a greater adipogenic potential
than AFMSCs and WJMSCs, characterized by a
greater accumulation of lipid vacuoles. Further studies,
462 E Iacono and others
Reproduction (2012) 143 455–468 www.reproduction-online.org
using different induction protocols and evaluating
differentiation assay by cytology and RT-PCR, are
necessary to verify if these different findings are due
to different laboratory conditions or if in equine the
conditions for AF, WJ and UCBMSCs promote one type
of differentiation lineage rather than other, as suggested
by Park et al. (2011).
In the present work, phenotypic characterization of
equine AF, UCB and WJMSCs was carried out by the
expression analysis of several surface MSC markers. We
choose to investigate cells at the third culture passage,
as they have reached a sufficient level of homogeneity
at this stage. As there are no equine-specific antibodies
for flow cytometry, we used antibodies for human
markers. To provide evidence for inter-species cross-
reactivity, the similarity of CD markers was identified
comparing the amino acid sequence, as already been
reported by de Mattos Carvalho et al. (2009).
Furthermore, as suggested recently by Pascucci et al.
(2011) we used equine circulating lymphocytes as
control. In the present study, high percentage of equine
AF, UCB and WJ cells population expressed MSC
surface markers CD90 and CD105. In addition, flow
cytometry revealed that the majority of analysed cells
were positive also for CD44. Our data are similar to
those reported by Park et al. (2011) for equine AFMSCs
tested using anti-human antibodies; moreover, these
markers are present in human MSCs and have also
been found in equine MSCs isolated from adipose
tissue (de Mattos Carvalho et al. 2009,Pascucci et al.
2011). These findings confirm that concerning CD90
and CD44 a high phenotypic similarity do exist among
MSCs isolated from different species and sources (Strem
et al. 2005).
A lack of reactivity with haematopoietic markers
CD34 and CD14, which cross-reaction was confirmed
by lymphocytes investigation, indicates that isolated
cells are negative for haematopoietic progenitors. On the
other hand, the lack of reactivity of equine cells and
lymphocytes with the haematopoietic markers CD45
and MSC with marker CD73 probably indicates that the
human-directed reagents do not cross-react with their
corresponding equine epitopes. These findings need
further investigation to assess if, in particularly, the lack
of CD73 expression is due to the lack of cross-reactivity
or is a species-specific feature as interpreted by Pascucci
et al. (2011).
From results obtained in this study, we suggest the
existence of alternative sources of multipotent cells in
equine. Differentiation of AF, UCB and WJMSCs into
adipocytes and, above all, into osteocytes and chon-
drocytes underlies their possible use for regenerative
medicine in injured horses. Further study will be
necessary to evaluate the expression of cell antigens
and the possible use of these tissues as allogenic source
of MSCs.
Materials and Methods
Materials
All chemicals were obtained from Sigma–Aldrich, plastic
dishes and tubes from Sarstedt, Inc. (Newton, NC, USA) unless
otherwise noted.
Animals
Samples were obtained from 13 standardbred mares, between
6 and 15 years of age, belonging to one stable location near
the Faculty of Veterinary Medicine of Bologna University, or
housed at the Equine Perinatology Unit ‘Stefano Belluzzi’,
University of Bologna, for attending delivery. Experimental
procedures were carried out in accordance with DL 116/92
and were approved by the Ethics Committee at the University
of Bologna and by the Ministry of Health.
Sampling and MSCs isolation
Amniotic fluid
AF samples were taken soon after foal or placenta membranes
passed through vulva, using a sterile 18 gauge needle mounted
on a 60 ml sterile syringe (IMI, Padua, Italy), containing 1 ml
of EDTA solution to prevent clot formation that could affect
cell isolation. Samples were stored at 4 8Candfurther
processed within 12 h. Briefly, each sample was diluted 1:1
with Dulbecco’s Phosphate Buffer Solution (DPBS) containing
100 IU/ml penicillin and 100 mg/ml streptomycin. The
obtained solution was centrifuged (Heraeus Megafuge 1.0R;
rotor: Heraeus #2704; ThermoFisher Scientific Inc.) for 15 min
at 470 g. Supernatant was carefully removed and the pellet was
re-suspended in 5 ml of culture medium containing DMEM and
TCM199 (1:1), 10% (v/v) FBS (Gibco; Invitrogen Corporation),
100 IU/ml penicillin and 100 mg/ml streptomycin. Cells were
isolated by carefully loading sample on 5 ml of 70% Percoll
solution in a 50 ml polypropylene tube, centrifuging for 30 min
at 25 8C at 1880 g; the interphase was collected after aspirating
and discarding the supernatant, washed with 20 ml of culture
medium and centrifuged at 470 gfor 10 min at 25 8C. The
supernatant was aspirated and cells were washed with culture
medium a second and third time. Cells were then re-suspended
in 1 ml of culture medium and counted by haemocytometer.
Umbilical cord blood
CB was collected immediately after foaling and before the
UC breaks spontaneously or was broken according to
management protocol. Venipuncture of the umbilical vein
was performed with a 21 guage hypodermic needle attached
to a 60 ml sterile syringe (IMI), containing 1 ml of heparin
(Eparina Vister 5000 IU/ml; Marvecs Pharma, Milan, Italy) as
anti-coagulant. Blood samples were stored at 4 8C for at the
least 12 h. Each sample was diluted 1:1 with DPBS
containing 100 IU/ml penicillin and 100 mg/ml streptomycin.
The solution was centrifuged (Heraeus Megafuge 1.0R; rotor:
Heraeus #2704) for 15 min at 470 g. Supernatant was
removed and the pellet was re-suspended in 5 ml of culture
medium. The mononuclear cell fraction was isolated by
MSCs from equine amniotic fluid 463
www.reproduction-online.org Reproduction (2012) 143 455–468
carefully loading sample on 5 ml of 70% Percoll solution in a
50 ml tube, centrifuging for 30 min at 1880 gat 25 8C. The
interphase was then collected after aspirating and discarding
the supernatant washed with 20 ml of culture medium by
centrifuging at 470 gfor 10 min at 25 8C. The supernatant
was aspirated and cells were washed twice with culture
medium. Cells were then re-suspended in 1 ml of culture
medium and counted by haemocytometer.
Wharton’s Jelly
Immediately after breaking the UC, the part closest to the
Colt, characterized by an abundant amount of WJ, was
severed. Samples were stored in DPBS containing 100 IU/ml
penicillin and 100 mg/ml streptomycin, at 4 8C for at the latest
12 h. UC was disinfected by immersing for 10 min in 70%
ethanol. In a laminar flow hood, tissue was rinsed by
repeated immersion in DPBS and WJ was isolated, weighed
and minced finely (0.5 cm) by sterile scissors. Minced WJ
was transferred into a 50 ml polypropylene tube, to which
was added 1 ml/1 g sample of a digestion solution (0.1%
(w/v) collagenase type I (Gibco, Invitrogen Corporation),
dissolved in DPBS solution and sterilely filtered). The tissue
and digestion solution were mixed thoroughly, incubated in a
37 8C water bath for 1–2 h, and mixed every 15 min. After
incubation, collagenase was inactivated by diluting 1:1 with
DPBS plus 10% (v/v) FBS. The solution obtained was filtered
and undigested tissue was discarded. Nucleated cells were
pelleted at 470 gfor 10 min. The supernatant was discarded,
pellet was re-suspended in 5 ml of culture medium and spun
at 470 gfor 10 min to wash cells. This operation was
repeated three times. After the last wash, cell pellet was
re-suspended in 1 ml of culture medium and cell concen-
tration was counted by haemocytometer.
Cell doubling method
Primary cells were plated in a 25 cm
2
flask at a maximum
density of 5!10
4
cells/cm
2
and incubated in a 5% CO
2
humidified atmosphere at 38.5 8C. The medium was
completely replaced after 48 h in order to remove non-
adherent cells. Next the medium was completely replaced
every 3 days until the adherent cell population reached w80%
confluence. At this point, the adherent primary MSCs were
passaged by digestion with 0.05% (w/v) trypsin, counted
with a haemocytometer, and re-seeded as ‘Passage 1’ (P1) at
25!10
3
cells/cm
2
. For the subsequent passages, cells were
inoculated in 5 cm
2
flasks at 25!10
3
cells/cm
2
and allowed
to multiply for 6–7 days to 90% confluence before trypsini-
zation and successive passage. DT and CD were calculated
from haemocytometer counts and cell culture time (CT) for
each passage according to the following two formulae
(Vidal et al. 2007):
CD ZlnðNf=NiÞ=lnð2Þ(1)
DT ZCT=CD (2)
where N
f
is the final number of cells and N
i
the initial
number of cells.
Chondrogenic in vitro differentiation
Expanded cells were harvested after three passages, washed
in DPBS, and placed in a six-well plate at a density of 5!
10
3
cells/cm
2
in chondrogenic induction medium, consisting
of DMEM/TCM199, 100 IU/ml penicillin, 100 mg/ml strepto-
mycin, 6.25 mg/ml insulin, 50 nM ascorbate-2-phosphate,
0.1 mM dexamethasone, 10 ng/ml human transforming
growth factor (hTGF)-b1 and 1% (v/v) FBS. Cells in
monolayer were incubated for 3 weeks. As a negative
control an equal number of cells were cultured in expansion
media for 21 days. In both groups the medium was
completely replaced every 3 days. After 3 weeks of culture,
cells were fixed with 10% (v/v) formalin for 1 h at room
temperature (RT), then stained with Alcian Blue solution (1%
in 3% acetic acid (v/v), pH 2.5) for 15 min at RT. Alcian Blue
staining acid mucosubstances and acetic mucins confirmed
chondrogenic differentiation cytologically.
Osteogenic in vitro differentiation
Undifferentiated cells were induced towards the osteogenic
lineage using the protocol described by Mizuno & Hyaku-
soku (2003). Briefly, at passage 3 putative cells were seeded
in a six-well plate at a density of 5!10
3
cells/cm
2
and
cultured in base medium for 24 h to allow cell adhesion.
After cells had adhered to the plate, they were treated with
an osteogenic induction medium consisting of 10 mM
b-glycerophosphate, 0.1 mM dexamethasone, 50 mM ascor-
bate-2-phosphate and 10% (v/v) FBS in DMEM/TCM199.
DMEM/TCM199 supplemented with 10% FBS was used as a
control medium. Media in both groups were completely
changed twice a week. After 21 days of induction, cells were
stained to confirm osteogenic differentiation.
von Kossa staining
In the osteogenic assay, later stage of osteogenesis,
characterized by calcium deposition, was assessed 3 weeks
after induction via von Kossa and Alizarin Red S stainings
for detection of deposits of calcium or calcium salt.
For von Kossa staining, cells were washed three times
with PBS before cell fixation with 10% (v/v) formalin for
1 h at RT. Cells were washed five times with distilled water
before adding 1 ml of 5% (w/v) silver nitrate and exposing
to yellow light for 15 min. The wells were washed five times
with distilled water. Calcium phosphate deposits stained
black.
Alizarin Red S staining
To confirm osteogenic differentiation, Alizarin Red S staining
was also performed for detecting calcium deposition. In brief,
cells were rinsed with DPBS and fixed incubating in ice-cold
ethanol 70% (v/v) for 1 h at RT. After three washes with
distilled water 1 ml of 2% (w/v) Alizarin Red S (pH 4.1–4.3)
solution was added. The plate was incubated at RT for
30 min, then Alizarin Red S solution was removed and then
the cells were rinsed four times with distilled water.
464 E Iacono and others
Reproduction (2012) 143 455–468 www.reproduction-online.org
Adipogenic in vitro differentiation
Adipogenesis was induced using protocol described by Mizuno
& Hyakusoku (2003) and Koch et al. (2007). Six-well culture
plates were seeded at a density of 5!10
3
cells undifferentiated
MSCs per cm
2
of tissue culture surface area in 0.5 ml of
expansion medium per tissue culture surface. After 24 h,
medium was completely replaced with adipogenic induction
medium consisting of 15% (v/v) rabbit serum, 1 mM dexa-
methasone, 0.5 mM 3-isobutyl-1-methylxanthine (IBMX),
10 mg/ml bovine insulin, 0.2 mM indomethacin in DMEM/
TCM199. After 72 h of culture, the IBMX supplement was
removed from the medium. After 3 additional days, the
dexamethasone supplement was removed from the medium,
and cells were cultured for additional 15 days. Equal cell
number was cultured in expansion medium for negative
control. Both groups were maintained in culture for a total of
21 days and media were completely exchanged every 3 days.
To asses adipogenic differentiation, cells were stained to
evaluate the baseline formation of neutral lipid vacuoles
stainable with Oil Red O. Medium was aspirated, cells were
washed once with PBS then fixed with 10% (v/v) formalin for
1 h at RT. The formalin was then replaced with 2 ml of sterile
water for a few minutes. Water was aspirated and replaced with
60% (v/v) isopropanol for 5 min and then cells were covered
with Oil Red O solution (0.3% in 60% isopropanol (v/v)). After
5 min, cells were rinsed with distilled water and lipid vacuoles
appeared red.
Characterization of MSCs
Cytofluorimetric analysis was performed to identify cell surface
marker expression of equine MSCs. At passage 3, cells were
labelled with the following mAbs: CD105, CD45, CD90,
CD44, CD34, CD14 and CD73 (all from Beckman Coulter,
Fullerton, CA, USA). Cells were also labelled with isotype
control antibodies. Briefly, at 80% confluence, cells at P3 were
washed twice with DPBS, harvested using 0.05% (w/v) trypsin
solution and aliquoted at a concentration of 0.5–10
6
cells/ml.
Each aliquot was fixed and permeabilized using Reagent 1 of
Intraprep Kit (Beckman Coulter, Miami, FL, USA) according to
the manufacturer’s instructions. Cells were stained for 30 min
with either conjugated-specific antibodies or istotype-matched
control mouse IgG (Table 2) at recommended concentrations.
Labelled cells were washed twice in DPBS and fluorescence
intensity was evaluated using an FC500 two-laser equipped
cytometer (Beckman Coulter). All analyses were based on
control cells incubated with isotype-specific IgGs to establish
the background signal. Cross-reactivity of the antibodies used
was screened using cultured human and horse MSCs.
Furthermore, to verify cross-reactivity, control of circulating
equine lymphocytes was carried out. The similarity of CD
markers was also identified by comparing the amino acid
sequences using Basic Local Alignment Search Tool (BLAST).
Results were further analysed with the CXP dedicated program.
Statistical analysis
CT and CD are expressed as meanGS.D. Statistical analysis was
performed using Statistics for Windows (Stat Soft, Inc., Tulsa,
OK, USA). Data were analysed using one-way ANOVA for
multiple comparisons. Significance has been assessed for
P!0.05.
Declaration of interest
The authors declare that there is no conflict of interest that
could be perceived as prejudicing the impartiality of the
reported research.
Funding
This research was supported by University of Bologna (RFO:
Ricerca Fondamentale Orientata).
Acknowledgements
The authors wish to thank Prof. Gaetano Mari (Department of
Veterinary Medical Sciences, University of Bologna, Ozzano
Emilia (BO), Italy), Dr Carolina Castagnetti (Equine Perinatol-
ogy Unit, Department of Veterinary Medical Sciences,
University of Bologna, Ozzano Emilia (BO) Italy) and Scuderia
Trio Srl (Ozzano Emilia (BO), Italy), for the agreement of the
sample collection. Thanks are also due to Dr Silvia Colleoni
(AVANTEA Srl, Cremona, Italy) and Prof. Cesare Galli
(Department of Veterinary Medical Scineces, University of
Bologna, Ozzano Emilia (BO), Italy) for their advice on cell
culture protocols.
References
Airas L, Hellman J, Salmi M, Bono P, Puurunen T, Smith DJ & Jalkanen S
1995 CD73 is involved in lymphocyte binding to the endothelium:
characterization of lymphocyte-vascular adhesion protein 2 identifies it
as CD73. Journal of Experimental Medicine 182 1603–1608. (doi:10.
1084/jem.182.5.1603)
Barnes D, McKeehan WL & Sato GH 1987 Cellular endocrinology:
integrated physiology in vitro.In Vitro Cellular & Developmental Biology
23 659–662. (doi:10.1007/BF02620978)
Bellows CG, Pei W, Jia Y & Heersche JNM 2003 Proliferation,
differentiation and self-renewal of osteoprogenitors in vertebral cell
populations from aged and young female rats. Mechanisms of Ageing
and Development 124 747–757. (doi:10.1016/S0047-6374(03)00088-5)
Table 2 Primary antibodies and isotype used for flow cytometry.
Markers Primary antibody Ig
CD44FITC Mouse monoclonal IgG1
CD90PC5 Mouse monoclonal IgG1
CD105PE Mouse monoclonal IgG2a
CD73PE Mouse monoclonal IgG1
CD14PC5 Mouse monoclonal IgG2a
CD45APC Mouse monoclonal IgG1
Isotype
PC5 Mouse monoclonal IgG2a
FITC Mouse monoclonal IgG1
PE Mouse monoclonal IgG1
APC Mouse monoclonal IgG1
MSCs from equine amniotic fluid 465
www.reproduction-online.org Reproduction (2012) 143 455–468
Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S,
Kasahara H, Rota M, Musso E, Urbanek K et al. 2003 Adult cardiac stem
cells are multipotent and support myocardial regeneration. Cell 114
763–776. (doi:10.1016/S0092-8674(03)00687-1)
Berg LC, Koch TG, Heerkens T, Besonov K, Thomsen PD & Betts DH 2009
Chondrogenic potential of mesenchymal stromal cells derived from
equine bone marrow and umbilical cord blood. Veterinary and
Comparative Orthopaedics and Traumatology 22 363–370.
Bergman RJ, Gazit D, Kahn AJ, Gruber H, McDougall S & Hahn TJ
1996 Age related changes in osteogenic stem cells in mice. Journal of
Bone and Mineral Research 11 568–577. (doi:10.1002/jbmr.
5650110504)
Chen J, Sotome S, Wang J, Orii H, Uemura T & Shinomiya K 2005
Correlation of in vivo bone formation capability and in vitro
differentiation of human bone marrow stromal cells. Journal of Medical
and Dental Sciences 52 27–34.
Colleoni S, Bottani E, Tessaro I, Mari M, Merlo B, Romagnoli N, Spadari A,
Galli C & Lazzari G 2009 Isolation, growth and differentiation of
equine mesenchymal stem cells: effect of donor, source, amount of tissue
and supplementation with basic fibroblast growth factor. Veterinary
Research Communications 33 811–821. (doi:10.1007/s11259-009-
9229-0)
Cremonesi F, Violini S, Lange Consiglio A, Ramelli P, Ranzenigo G &
Mariani P 2008 Isolation, in vitro culture and characterization of foal
umbilical cord stem cells at birth. Veterinary Research Communications
32 S139–S142. (doi:10.1007/s11259-008-9116-0)
De Coppi P, Bartsch G, Siddiqui MM, Xu T, Santos CC, Perin L,
Mostoslavsky G, Serre AC, Snyder EY, Yoo JJ et al. 2007 Isolation of
amniotic stem cell lines with potential for therapy. Nature Biotechnology
25 100–106. (doi:10.1038/nbt1274)
D’Ippolito G, Schiller P, Ricordi C, Roos B & Howard G 1999 Age-related
osteogenic potential of mesenchymal stromal stem cells from human
vertebral bone marrow. Journal of Bone and Mineral Research 14
1115–1122. (doi:10.1359/jbmr.1999.14.7.1115)
Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F,
Krause D, Deans R, Keating A, Prockop D & Horwitz E 2006
Minimal criteria for defining multipotent mesenchymal stromal cells.
The International Society for Cellular Therapy position statement.
Cytotherapy 8315–317. (doi:10.1080/14653240600855905)
Eloit M 1999 Risks of virus transmission associated with animal sera
or substitutes and methods of control. Developments in Biological
Standardization 99 9–16.
Erdo F, Buhrle C, Blunk J, Hoehn M, Xia Y, Flieschmann B, Focking M,
Kustermann E, Kolossov E, Heschler J et al. 2003 Host-dependent
tumorogenesis of embryonic stem cell transplantation in experimental
stroke. Journal of Cerebral Blood Flow and Metabolism 23 780–785.
(doi:10.1097/01.WCB.0000071886.63724.FB)
Erices A, Conget P & Minguell JJ 2000 Mesenchymal progenitor cells in
human umbilical cord blood. British Journal of Haematology 109
235–242. (doi:10.1046/j.1365-2141.2000.01986.x)
Gosden CM 1983 Amniotic fluid cell types and culture. British Medical
Bulletin 39 348–354.
Guest DJ, Smith MRW & Allen WR 2010 Equine embryonic stem-like
cells and mesenchymal stromal cells have different survival rates
and migration patterns following their injection into damaged super-
ficial digital flexor tendon. Equine Veterinary Journal 42 636–642.
(doi:10.1111/j.2042-3306.2010.00112.x)
Hackett CH & Fortier LA 2011 Embryonic stem cells and iPS cells: sources
and characteristics. Veterinary Clinics of North America Equine Practice
27 233–242. (doi:10.1016/j.cveq.2011.04.003)
Horwitz EM, Prockop DJ, Fitzpatrick LA, Koo WW, Gordon PL, Neel M,
Sussman M, Orchard P, Marx JC, Pyeritz RE & Brenner MK 1999
Transplantability and therapeutic effects of bone marrow-derived
mesenchymal cells in children with osteogenesis imperfecta. Nature
Medicine 5309–313.
Hoynowski SM, Fry MM, Gardner BM, Leming MT, Tucker JR, Black L,
Sand T & Mitchell KE 2007 Characterization and differentiation of
equine umbilical cord-derived matrix cells. Biochemical and Bio-
physical Research Communications 362 347–353. (doi:10.1016/j.bbrc.
2007.07.182)
In’t Anker PS, Scherjon SA, Kleijburg van der Keur C, Noort WA, Claas FH,
Willemze R, Fibbe WE & Kanhai HH 2003 Amniotic fluid as a novel
source of mesenchymal stem cells for therapeutic transplantation. Blood
102 1548–1549. (doi:10.1182/blood-2003-04-1291)
Karahuseyinoglu S, H Cinar O, Kilic E, Kara F, Akay GG, Demiralp DO,
Tukun A, Uckan D & Can A 2007 Biology of stem cells in human
umbilical cord stroma: in situ and in vitro surveys. Stem Cells 25
319–331.
Kaviani A, Perry TE, Dzakovic A, Jennings RW, Ziegler MM & Fauza DO
2001 The amniotic fluid as a source of cells for fetal tissue engineering.
Journal of Pediatric Surgery 36 1662–1665. (doi:10.1053/jpsu.2001.
27945)
Kern S, Eichler H, Stoeve J, Kluter H & Bieback K 2006 Comparative
analysis of mesenchymal stem cells from bone marrow, umbilical cord
blood, or adipose tissue. Stem Cells 24 1294–1301. (doi:10.1634/
stemcells.2005-0342)
Kobayashi K, Kubota T & Aso T 1998 Study on myofibroblast differentiation
in the stromal cells of Wharton’s jelly: expression and localization of
alpha-smooth muscle actin. Early Human Development 51 223–233.
(doi:10.1016/S0378-3782(97)00123-0)
Koch TG, Heerkens T, Thomsen PD & Betts DH 2007 Isolation of
mesenchymal stem cells from equine umbilical cord blood. BMC
Biotechnology 726–34. (doi:10.1186/1472-6750-7-26)
Koch TG, Thomsen PD & Betts DH 2009 Improved isolation protocol for
equine cord blood derived mesenchymal stromal cells. Cytotherapy 11
443–447. (doi:10.1080/14653240902887259)
Koso H, Satoh S & Watanabe S 2007 c-kit marks late retinal progenitor cells
and regulates their differentiation in developing mouse retina. Develop-
mental Biology 301 141–154. (doi:10.1016/j.ydbio.2006.09.027)
Kretlow JD, Jin Y, Liu W, Zhang WJ, Hong T, Zhou G, Baggett LS, Mikos AG
& Cao Y 2008 Donor age and cell passage affects differentiation potential
of murine bone marrow-derived stem cells. BMC Cell Biology 960.
(doi:10.1186/1471-2121-9-60)
Li X, Zhou SG, Imreh MP, Ahrlund-Richter L & Allen WR 2006 Horse
embryonic stem cell like cell lines from the proliferation of inner cell
mass cells. Stem Cells and Development 15 523–531.
Liao J, Cui C, Chen S, Ren J, Chen J, Gao Y, Li H, Jia N, Cheng L, Xiao H
et al. 2009 Generation of induced pluripotent stem cell lines from adult
rat cells. Cell Stem Cell 411–15. (doi:10.1016/j.stem.2008.11.013)
Liu H, Zhu F, Yong J, Zhang P, Hou P, Li H, Jiang W, Cai J, Liu M, Cui K et al.
2008 Generation of induced pluripotent stem cells from adult rhesus
monkey fibroblasts. Cell Stem Cell 3587–590. (doi:10.1016/j.stem.
2008.10.014)
Lovati AB, Corradetti B, Lange Consiglio A, Recordati C, Bonacina E,
Bizzaro D & Cremonesi F 2011 Comparison of equine bone marrow-,
umbilical cord matrix and amniotic fluid-derived progenitor cells.
Veterinary Research Communications 35 103–121. (doi:10.1007/
s11259-010-9457-3)
Lu LL, Liu YJ, Yang SG, Zhao QJ, Wang X, Gong W, Han ZB, Xu ZS, Lu YX,
Liu D et al. 2006 Isolation and characterization of human umbilical cord
mesenchymal stem cells with hematopoiesis-supportive function and
other potentials. Haematologica 91 1017–1026.
Majors AK, Boehm CA, Nitto H, Midura RJ & Muschler GF 1997
Characterization of human bone marrow stromal cells with respect to
osteoblastic differentiation. Journal of Orthopaedic Research 15
546–557. (doi:10.1002/jor.1100150410)
Mareschi K, Ferrero I, Rustichelli D, Aschero S, Gammaitoni L, Aglietta M,
Madon E & Fagioli F 2006 Expansion of mesenchymal stem cells isolated
from pediatric and adult donor bone marrow. Journal of Cellular
Biochemistry 97 744–754. (doi:10.1002/jcb.20681)
Martin GM, Sprague CA & Epstein CJ 1970 Replicative lifespan of
cultivated human cells. Effects of donor’s age, tissue, and genotype.
Laboratory Investigation 23 86–92.
de Mattos Carvalho A, Alves AL, Golim MA, Moroz A, Hussni CA,
de Oliveira PG & Deffune E 2009 Isolation and immunophenotypic
characterization of mesenchymal stem cells derived from equine species
adipose tissue. Veterinary Immunology and Immunopathology 132
303–306. (doi:10.1016/j.vetimm.2009.06.014)
Mendes SC, Tibbe JM, Veenhof M, Bakker K, Both S, Platenburg PP,
Oner FC, de Bruijn JD & van Blitterswijk CA 2002 Bone
466 E Iacono and others
Reproduction (2012) 143 455–468 www.reproduction-online.org
tissue-engineered implants using human bone marrow stromal cells:
effect of culture conditions and donor age. Tissue Engineering 8
911–920. (doi:10.1089/107632702320934010)
Mizuno H & Hyakusoku H 2003 Mesengenic potential and future clinical
perspective of human processed lipoaspirate cells. Journal of Nippon
Medical School 70 300–306. (doi:10.1272/jnms.70.300)
Nagy K, Sung HK, Zhang P, Laflamme S, Vincent P, Agha-Mohammadi S,
Woltjen K, Monetti C, Michael IP, Smith LC et al. 2011 Induced
pluripotent stem cell lines derived from equine fibroblasts. Stem Cell
Reviews 7693–702. (doi:10.1007/s12015-011-9239-5)
Naito Y & Lowenstein JM 1981 50-Nucleotidase from rat heart.
Biochemistry 20 5188–5194. (doi:10.1021/bi00521a014)
Nakamura Y, Muguruma Y, Yahata T, Miyatake H, Sakai D, Mochida J,
Hotta T & Ando K 2006 Expression of CD90 on keratinocyte
stem/progenitor cells. British Journal of Dermatology 154 1062–1070.
(doi:10.1111/j.1365-2133.2006.07209.x)
Nanaev AK, Kohnen G, Milovanov AP, Domogatsky SP & Kaufmann P 1997
Stromal differentiation and architecture of the human umbilical cord.
Placenta 18 53–64. (doi:10.1016/S0143-4004(97)90071-0)
Nelson TJ, Faustino RS, Chiriac A, Crespo-Diaz R, Behfar A & Terzic A
2008 CXCR4C/FLK-1Cbiomarkers select a cardiopoietic lineage
from embryonic stem cells. Stem Cells 26 1464–1473. (doi:10.1634/
stemcells.2007-0808)
Nelson TJ, Martinez-Fernandez A, Yamada S, Ikeda Y, Perez-Terzic C &
Terzic A 2010 Induced pluripotent stem cells: advantages to
applications. Stem Cells and Cloning: Advances and Applications 3
29–37. (doi:10.2147/SCCAA.S4954)
Park IH, Zhao R, West JA, Yabuuchi A, Huo H, Ince TA, Lerou PH,
Lensch MW & Daley GQ 2008 Reprogramming of human somatic cells
to pluripotency with defined factors. Nature 451 141–146. (doi:10.1038/
nature06534)
Park SB, Seo MS, Kang JG, Chae JS & Kang KS 2011 Isolation
and characterization of equine amniotic fluid-derived multipotent
stem cells. Cytotherapy 13 341–349. (doi:10.3109/14653249.2010.
520312)
Pascucci L, Curina G, Mercati F, Marini C, Dall’Aglio C, Paternesi B &
Ceccarelli P 2011 Flow cytometric characterization of culture expanded
multipotent mesenchymal stromal cells (MSCs) from horse adipose
tissue: towards the definition of minimal stemness criteria. Veterinary
Immunology and Immunopathology 144 499–1506.
Passeri S, Nocchi F, Lamanna R, Lapi S, Miragliotta V, Giannessi E,
Abramo F, Stornelli MR, Matarazzo M, Plenteda D et al. 2009 Isolation
and expansion of equine umbilical cord-derived matrix cells (EUCMCs).
Cell Biology International 33 100–105. (doi:10.1016/j.cellbi.2008.
10.012)
Perin L, Giuliani S, Jin D, Sedrakyan S, Carraro G, Habibian R,
Warburton D, Atala A & De Filippo RE 2007 Renal differentiation of
amniotic fluid stem cells. Cell Proliferation 40 936–948. (doi:10.1111/j.
1365-2184.2007.00478.x)
Pesce M & Scholer HR 2001 Oct-4: gatekeeper in the beginning of
mammalian development. Stem Cells 19 271–278. (doi:10.1634/
stemcells.19-4-271)
Prusa AR, Marton E, Rosner M, Bernaschek G & Hengstschlager M 2004
Oct-4-expressing cells in human amniotic fluid: a new source for stem
cell research? Human Reproduction 18 1489–1493. (doi:10.1093/
humrep/deg279)
Reubinoff BE, Pera MF, Fong CY, Trounson A & Bongso A 2000 Embryonic
stem cell lines from human blastocytes: somatic differentiation in vitro.
Nature Biotechnology 18 399–404. (doi:10.1038/74447)
Romanov YA, Svintsitskaya VA & Smirnov VN 2003 Searching for
alternative sources of postnatal human mesenchymal stem cells:
candidate MSC like cells from umbilical cord. Stem Cells 21 105–110.
(doi:10.1634/stemcells.21-1-105)
Sackstein R, Merzaban JS, Cain DW, Dagia NM, Spencer JA, Lin CP &
Wohlgemuth R 2008 Ex vivo glycan engineering of CD44 programs
human multipotent mesenchymal stromal cell trafficking to bone. Nature
Medicine 14 181–187. (doi:10.1038/nm1703)
Saito S, Ugai H, Sawaid K, Yamamoto Y, Minamihashi A, Kurosaka K,
Kobayashi Y, Murata T, Obata Y & Yokoyama K 2002 Isolation of
embryonic stem-like cells from equine blastocysts and their differen-
tiation in vitro.FEBS Letters 531 389–396.
Sarugaser R, Lickorish D, Baksh D, Hosseini MM & Davies JE 2005 Human
umbilical cord perivascular (HUCPV) cells: a source of mesenchymal
progenitors. Stem Cells 23 220–229. (doi:10.1634/stemcells.2004-0166)
Schallmoser K, Bartmann C, Rohde E, Reinisch A, Kashofer K,
Stadelmeyer E, Drexler C, Lanzer G, Linkesch W & Strunk D 2007
Human platelet lysate can replace fetal bovine serum for clinical-scale
expansion of functional mesenchymal stromal cells. Transfusion 47
1436–1446. (doi:10.1111/j.1537-2995.2007.01220.x)
Schneider EL & Mitsui Y 1976 The relationship between in vitro cellular
aging and in vivo human age. PNAS 73 584–588. (doi:10.1073/pnas.73.
10.3584)
Secco M, Zucconi E, Vieira NM, Fogac LLQ, Cerqueira A, Carvalho MDF,
Jazeidje T, Okamoto OK, Muotri AR & Zatz M 2008 Multipotent stem
cells from umbilical cord: cord is richer than blood!. Stem Cells 26
146–150. (doi:10.1634/stemcells.2007-0381)
Seo MJ, Suh SY, Bae YC & Jung JS 2005 Differentiation of human adipose
stromal cells into hepatic lineage in vitro and in vivo.Biochemical and
Biophysical Research Communications 328 258–264. (doi:10.1016/j.
bbrc.2004.12.158)
Sethe S, Scutt A & Stolzing A 2006 Aging of mesenchymal stem cells.
Ageing Research Reviews 591–116. (doi:10.1016/j.arr.2005.10.001)
Stenderup K, Justesen J, Eriksen EF, Rattan SI & Kassem M 2001 Number
and proliferative capacity of osteogenic stem cells are maintained during
aging and in patients with osteoporosis. Journal of Bone and Mineral
Research 16 1120–1129. (doi:10.1359/jbmr.2001.16.6.1120)
Stenderup K, Justesen J, Clausen C & Kassem IM 2003 Aging is associated
with decreased maximal life span and accelerated senescence of bone
marrow stromal cells. Bone 33 919–926. (doi:10.1016/j.bone.2003.07.
005)
Stocum DL 2001 Stem cell in regenerative biology and medicine. Wound
Repair and Regeneration 9429–442. (doi:10.1046/j.1524-475x.2001.
00429.x)
Stolzing A, Jones E, McGonagle D & Scutt A 2008 Age-related changes in
human bone marrow derived mesenchymal stem cells: consequences for
cell therapies. Mechanisms of Ageing and Development 129 163–173.
(doi:10.1016/j.mad.2007.12.002)
Strem BM, Hicok KC, Zhu M, Wulur I, Zeni A, Schreiber RE, Fraser JK &
Hedrick MH 2005 Multipotential differentiation of adipose tissue-
derived stem cells. Keio Journal of Medicine 54 132–141. (doi:10.
2302/kjm.54.132)
Takahashi K & Yamanaka S 2006 Induction of pluripotent stem cells from
mouse embryonic and adult fibroblast cultures by defined factors. Cell 26
663–676. (doi:10.1016/j.cell.2006.07.024)
Takechi K, Kuwabara Y & Mizuno M 1993 Ultrastructural and immunohis-
tochemical studies of Wharton’s jelly umbilical cord cells. Placenta 14
235–245. (doi:10.1016/S0143-4004(05)80264-4)
Thomson JA, Istkovitz Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ &
Marshall VS 1998 Embryonic stem cell lines derived from human
blastocysts. Science 282 1145–1147. (doi:10.1126/science.282.5391.
1145)
Tsai MS, Lee JL, Chang YJ & Hwang SM 2004 Isolation of human
multipotent mesenchymal stem cells from second-trimester amniotic
fluid using a novel two-stage culture protocol. Human Reproduction 19
1450–1456. (doi:10.1093/humrep/deh279)
Vaala WE 2006 Perinatology. In The Equine Manual, 2nd edn, pp 803–804.
Eds AJ Higgins & JR Snyder. Elsevier Press.
van der Valk J, Mellor D, Brands R, Fischer R, Gruber F, Gstraunthaler G,
Hellebrekers L, Hyllner J, Jonker FH, Prieto P et al. 2004 The humane
collection of fetal bovine serum and possibilities for serum-free cell and
tissue culture. Toxicology In Vitro 18 1–12. (doi:10.1016/j.tiv.2003.08.
009)
Vidal MA, Kilroy GE, Lopez ME, Johnson JR, Moore RM & Gimble JM 2007
Characterization of equine adipose tissue-derived stromal cells:
adipogenic and osteogenic capacity and comparison with bone
marrow-derived mesenchymal stromal cells. Veterinary Surgery 36
613–622. (doi:10.1111/j.1532-950X.2007.00313.x)
Wang HS, Hung SC, PengST, Huang CC, Wei HM, Guo YJ, Fu YS, Lai MC &
Chen CC 2004 Mesenchymal stem cells in the Wharton’s jelly of the
human umbilical cord. Stem Cells 22 1330–1337. (doi:10.1634/
stemcells.2004-0013)
Weiss ML, Medicetty S, Bledsoe AR, Rachakatla RS, Choi M, Merchav S,
Luo Y, Rao MS, Velagaleti G & Troyer D 2006 Human umbilical cord
MSCs from equine amniotic fluid 467
www.reproduction-online.org Reproduction (2012) 143 455–468
matrix stem cells: preliminary characterization and effect of trans-
plantation in a rodent model of Parkinson’s disease. Stem Cells 24
781–792. (doi:10.1634/stemcells.2005-0330)
You Q, Cai L, Zheng J, Tong X, Zhang D & Zhang Y 2008 Isolation of human
mesenchymal stem cells from third-trimester amniotic fluid. International
Journal of Gynaecology and Obstetrics 103 149–152. (doi:10.1016/j.
ijgo.2008.06.012)
Zhang H, Fazel S, Tian H, Mickle DAG, Weisel RD, Fujii T & Li RK 2005
Increasing donor age adversely impacts beneficial effects of bone
marrow but not smooth muscle myocardial cell therapy. American
Journal of Physiology. Heart and Circulatory Physiology 289
H2089–H2096. (doi:10.1152/ajpheart.00019.2005)
Zheng H, Martin JA, Duwayri Y, Falcon G & Buckwalter JA 2007 Impact of
aging on rat bone marrow-derived stem cell chondrogenesis. Journals of
Gerontology. Series A, Biological Sciences and Medical Sciences 62
136–148. (doi:10.1093/gerona/62.2.136)
Received 28 September 2010
First decision 12 November 2010
Revised manuscript received 2 December 2011
Accepted 20 January 2012
468 E Iacono and others
Reproduction (2012) 143 455–468 www.reproduction-online.org
... Number of CFU formed by eATMSCs after storage for 6 (T6), 24 (T24) and 48 (T48) hrs in saline solution (S) and plasma (P) at 4 °C (4) and room temperature (20): a vs. b; c vs. d; e vs. f: eWJMSCs viability after storage for 6 (T6), 24 (T24) and 48 (T48) hrs in saline solution (S)and plasma (P) at 4 • C (4) and room temperature(20). No statistical differences have been found among groups.Stored eATMSCs and eWJMSCs also preserved the ability to form CFU when cultured in vitro; in both cell lines this statistically decreased as storage hrs increased, except for eATMSCs stored for 6 hrs in saline solution at 4 • C. Data are shown inFigures 3 and 4. ...
... eWJMSCs viability after storage for 6 (T6), 24 (T24) and 48 (T48) hrs in saline solution (S) and plasma (P) at 4 °C (4) and room temperature(20). No statistical differences have been found among groups.Stored eATMSCs and eWJMSCs also preserved the ability to form CFU when cultured in vitro; in both cell lines this statistically decreased as storage hrs increased, except for eATMSCs stored for 6 hrs in saline solution at 4 °C. ...
... Composition of induction media[19,20]. ...
Article
Full-text available
To use Mesenchymal Stromal Cells (MSCs) in equine patients, isolation and expansion are performed in a laboratory. Cells are then sent back to the veterinary clinic. The main goal of storage conditions during cell transport is to preserve their biological properties and viability. The aim of this study was to evaluate the effects of storage solutions, temperature and time on the characteristics of equine adipose tissue and Wharton’s jelly-derived MSCs. We compared two different storage solutions (plasma and 0.9% NaCl), two different temperatures (4 °C and room temperature) and three time frames (6, 24, 48 h). Cell viability, colony-forming units, trilineage differentiation, the expression of CD45 and CD90 antigens and adhesion potentials were evaluated. Despite the molecular characterization and differentiation potential were not influenced by storage conditions, viability, colony-forming units and adhesion potential are influenced in different way, depending on MSCs sources. Overall, this study found that, despite equine adipose tissue MSCs being usable after 24 h of storage, cells derived from Wharton’s jelly need to be used within 6 h. Moreover, while for adipose cells the best conservation solutions seems to be plasma, the cell viability of Wharton’s jelly MSCs declined in both saline and plasma solution, confirming their reduced resistance to conservation.
... AF-derived MSCs (AF-MSCs) have several advantages, including a reduced risk of immunological rejection and oncogenesis, pluripotency suggested by their origin from the proximity of the embryonic inner cell mass, and a less invasive collection procedure (19). AF-MSCs are used in regenerative therapy as the cells can be used to obtain tissues in large quantities and have a low risk of immune rejection (20,21). ...
Article
Full-text available
Amniotic fluid mesenchymal stem cells (AF-MSCs), which can be obtained from fetal tissue, reportedly have self-renewal capacity and multi-lineage differentiation potential. The aim of this study was to identify the biological characteristics of AF-MSCs and evaluate their stability and safety in long-term culture. To confirm the biological characteristics of AF-MSCs, morphology, proliferation capacity, karyotype, differentiation capacity, gene expression level, and immunophenotype were analyzed after isolating AF-MSCs from equine amniotic fluid. AF-MSCs were differentiated into adipocytes, chondrocytes, and osteocytes. Immunophenotype analyses revealed expression levels of ≥95% and ≤ 2% of cells for a positive and negative marker, respectively. Analysis of the MSCs relative gene expression levels of AF-MSCs was approximately at least twice that of the control. The endotoxin level was measured to verify the safety of AF-MSCs and was found to be less than the standard value of 0.5 EU/ml. AF-MSCs were cultured for a long time without any evidence of abnormalities in morphology, proliferation ability, and karyotype. These results suggest that amniotic fluid is a competent source for acquiring equine MSCs and that it is valuable as a cell therapy due to its high stability.
... MSCs are reliably available due to their defined localisation in stem cell niches. To this day, MSCs used for clinical trials in horses have typically been isolated from bone marrow and, to a lesser extent, from adipose tissue or umbilical cord blood, umbilical cord matrix (Wharton's Jelly), placental tissue, and amniotic fluid [6][7][8][9][10][11][12]. The main therapeutic applications of these equine MSCs included primarily horse tendon injuries and osteoarthritis, as well as other cartilage and ligament injuries [1]. ...
Article
Full-text available
Equine mesenchymal stem cells (MSC) of various origins have been identified in horses, including MSCs from the bone marrow and adipose tissue. However, these stem cell sources are highly invasive in sampling, which thereby limits their clinical application in equine veterinary medicine. This study presents a novel method using an air-liquid interface to isolate stem cells from the hair follicle outer root sheath of the equine forehead skin. These stem cells cultured herewith showed high proliferation and asumed MSC phenotype by expressing MSC positive biomarkers (CD29, CD44 CD90) while not expressing negative markers (CD14, CD34 and CD45). They were capable of differentiating towards chondrogenic, osteogenic and adipogenic lineages, which was comparable with MSCs from adipose tissue. Due to their proliferative phenotype in vitro, MSC-like profile and differentiation capacities, we named them equine mesenchymal stem cells from the hair follicle outer root sheath (eMSCORS). eMSCORS present a promising alternative stem cell source for the equine veterinary medicine. Graphical abstract
... Foetal fluids (amniotic fluid, umbilical cord blood), and foetal adnexa (Wharton's jelly, amniotic membrane) have been identified as ideal alternative sources of MSCs in different animal species, such as horse [17][18][19], cattle [20,21], goat [22,23], and others. The benefits of these cells compared to adult MSCs are due to their origin from extraembryonic tissues; in fact, because they are at the maternal-foetal interface, these cells present low immunogenicity and immunomodulatory properties, making them a good candidate for allo-and xenotransplantation [24]. ...
Article
Full-text available
In the last decade, researchers described Mesenchymal Stem/stromal cells (MSCs) as a possible population of cells for cell-based therapies in regenerative medicine, both for humans and animals [...]
... All the four types of oMSCs were CD73 + , CD90 + and CD105 + but lack hematopoietic markers. The lack of species-speci c antibodies, as well as variances in the types of tissue sources and harvesting methods, are blamed for the heterogeneity in marker expression (Gugjoo et The MSCs in culture showed a typical growth curve with initial lag phase, an exponential phase, a plateau phase and a decline phase which determines the general behavior of cellular growth (Colter et al. 2001 (2007) and Iacono et al (2012) who conducted the study on humans and equines, respectively. Thus, the proliferation potential of MSCs reveals a species-speci c growth rate. ...
Preprint
Full-text available
Fetal adnexa derived Mesenchymal Stem Cells (MSC) offer all in one solution in therapeutics. The present study was designed to isolate, cultivate, characterize and to evaluate the growth kinetics of ovine fetal adnexa-derived Mesenchymal Stem Cells (MSCs). The gravid uteri of ewes were collected from a local abattoir. The four cell lines viz. Wharton’s Jelly (oWJ), Cord Blood (oCB), Amniotic Fluid (oAF), and Amniotic Sac (oAS) were isolated and expanded in vitro and characterized for surface markers (CD73, CD90 & CD105) and pluripotency markers (SOX2 & OCT4) at 3rd passage. The growth kinetics of MSCs was investigated at 3rd and 5th passages. Similarly, the colony forming unit (CFU) Assay was performed at 3rd passage. The fetal adnexa derived Ovine MSCs showed the amplification of CD73, CD90 & CD105, whereas, negative marker CD34 was not amplified. Similarly, the MSCs also qualified for the amplification of Pluripotency markers (OCT4, SOX2). The MSCs in culture showed a typical growth curve with initial lag phase, an exponential phase, a plateau phase and a decline phase. The proliferation potential of MSCs was higher at P5 than P3. The overall growth rate was significantly (p < 0.05) highest in oAF, followed by oCB, oWJ and oAS. The population doubling time (PDT) was significantly (p < 0.05) highest in oAS (87.28 ± 3.24 hr) and least in oAF (39.75 ± 1.09 hr). The MSCs showed a significant (p < 0.05) difference in their clonogenic potential. The colony number was highest in oAF (53.67 ± 4.06) and least in oAS (22.0 ± 2.08). The study reveals that in terms of growth kinetics and clonogenic potential, oAF-MSCs were superior which outperformed other MSCs indicating that oAF derived MSCs could be utilized for regenerative medicine.
... Even cell detaching agent (like trypsin) may impair receptors on cell surface. MSCs dynamic immunophenotype may too impart alterations in their biological features (Colleoni et al. 2009, Ranera et al. 2011, De Schauwer et al. 2012, Iacono et al. 2012, Kang et al. 2013, Paebst et al. 2014, Tessier et al. 2015, Gugjoo et al. 2020c. ...
Article
Full-text available
Stem cell, a wonder cell, acts as a basic unit for an individual development in early prenatal life, and repairs and regenerates the tissue and/ organ in post-natal life. The stem cell research although conducted extensively is still in its infancy for standardized therapeutics. Among various stem cells types, multi-potential mesenchymal stem cell (MSC) is mainly evaluated for therapeutic applications. These cells have been isolated from almost all the body organs/ tissues and fetal membranes and are culture expanded for higher concentrations. Like human, MSCs harvested from veterinary species are characterized on the basis of International Society for Cellular Therapy (ISCT). Extensive literature on their therapeutic applications in musculoskeletal and non-musculoskeletal systems evidences their potential utility under clinical settings. Currently, limited understanding in their hysiological mechanisms and availability of limited non-uniform in vivo studies restrict their definitive therapeutic applications. Lack of regulatory set up in India makes MSCs research in veterinary medicine a more complicated field. This review details the current status and possible ways to improve MSCs therapeutic applications in veterinary medicine, in general and in Indian system, in particular.
... Given the limited number of published preclinical studies, the variability between the animal models used, the specific injury model, and route of administration, it was not feasible to perform a comparative analysis. However, another review may be completed in the near future with an emphasis on preclinical models as the data seem to be positive in several studies [38,39]. ...
Article
Full-text available
Musculoskeletal ailments affect millions of people around the world and place a high burden on healthcare. Traditional treatment modalities are limited and do not address underlying pathologies. Mesenchymal stem cells (MSCs) have emerged as an exciting therapeutic alternative and Wharton’s jelly-derived mesenchymal stem cells (WJSCs) are some of these. This review reports the clinical and functional outcomes of the applications of WJSCs in orthopedic surgery. A systematic review was conducted utilizing the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. The studies that used culture-expanded, mesenchymal stem or stromal cells, MSCs and/or connective tissues procured from Wharton’s jelly (WJ), from January 2010 to October 2021, were included. Conventional non-operative therapies and placebos were used as comparisons. Six studies that directly discussed WJSCs use in an animal model or the basic scientific testing using an injury model were identified. Five publications studied cartilage injury, three studied degenerative disc disease, one was related to osteoarthritis, and one was related to osteochondral defects. The results of these studies suggested the benefits of WJSCs in the management of these orthopedic pathologies. To adequately assess the safety and efficacy of WJSCs in orthopedic surgery, further randomized controlled clinical studies are necessary.
Article
Full-text available
This study was designed to isolate, cultivate, characterize and evaluate the growth kinetics of mesenchymal stem cells (MSCs) derived from fetal adnexa of sheep. The gravid uteri of ewes were collected from a local abattoir. The MSCs isolated from different fetal regions (Wharton's Jelly [oWJ], cord blood [oCB], amniotic fluid [oAF] and amniotic Sac [oAS]) were expanded in vitro and characterized for surface and pluripotency markers. The growth kinetics of MSCs was compared at 3rd and 5th passages. Similarly, the colony-forming efficiency (CFE) assay was performed at 3rd passage. The fetal adnexa-derived ovine MSCs showed the expression of CD73, CD90 and CD105. Similarly, the MSCs also expressed pluripotency markers, OCT4 and SOX2. Besides, cells also differentiated into osteogenic, chondrogenic and adipogenic lineages. The MSCs in culture showed a typical growth curve with initial lag phase, an exponential phase, a plateau phase and a decline phase. The growth rate was highest in oAF-MSCs at P5. The population doubling time (PDT) was highest in oAS-MSCs (87.28 ± 3.24 h), whereas the colony number was highest in oAF-MSCs (53.67 ± 4.06). The study reveals that oAF-MSCs were superior which outperformed other MSCs indicating that oAF-derived MSCs could be utilized for regenerative medicine.
Article
Full-text available
The paper summarizes modern concepts of stem cells, considers their classification, the importance for the fundamental purposes of cell biology and clinical medicine, and shows the contradictions between the concepts of classical histology and modern information about stem cells. It also discusses the role and significance of the basic concepts of stem cells for the development of regenerative medicine. The question is raised on necessity of arrangement in the medical institutes of the course on stem cells and cellular technologies.
Article
Full-text available
The proximate obstacle to realizing the potential benefits of stem cell-based regenerative therapeutics is to safely and efficiently deliver the cells where they are needed/required. Though transplantation of culture-expanded human mesenchymal stem cells (MSC) holds the potential for cure of generalized skeletal diseases such as osteogenesis imperfecta, clinical effectiveness has been constrained to date by the poor osteotropism (migration to bone) of infused MSCs. Tissue-specific migration is initiated by adhesive interactions mediated by molecules on the surface of blood-borne cells that are specialized to resist the shear forces of blood flow, the "homing receptors". The recruitment of circulating cells to bone occurs within the marrow, at specialized marrow vessels that constitutively express E-selectin, a lectin that recognizes sialofucosylated determinants on its respective ligand(s). To gain insight on the expression and function of homing receptors on human MSC, we performed flow cytometry and Western blot analysis, together with parallel plate flow chamber studies under physiologic shear conditions. Whereas human hematopoietic progenitor cells (HPC) characteristically display the E-selectin ligands CLA and HCELL, which are (2,3)-sialyl-(1,3)-fucosylated glycoforms of PSGL-1 and CD44, respectively, biochemical and functional studies showed that human MSC express no E-selectin ligands and are devoid of PSGL-1. We also found that human MSC did not express many effectors of cellular trafficking to marrow present on HPC, such as LFA-1, LPAM-1 (4β7), nor the chemokine receptor CXCR4, but did express VLA-4 and, notably, display a CD44 glycoform bearing (2,3)-sialyllactosamine modifications that can serve as an acceptor for exogenous fucosylation. Employing an (1,3)-fucosyltransferase preparation and enzymatic conditions specifically designed for treating live cells, the native human MSC surface CD44 glycoform was converted to the E-selectin ligand HCELL with commensurate induction of potent E-selectin-dependent shear-resistant rolling interactions on activated endothelium, without effects on MSC viability or multipotency. As observed by real-time intravital microscopy in immunocompromised (NOD/SCID) mouse hosts, intravenously infused HCELL+ human MSCs homed robustly to bone with marrow infiltrates evident within hours of infusion, whereas unmodified MSC and HCELL+ MSC treated with sialidase (which eliminates E-selectin binding) showed negligible rolling on marrow vessels and did not extravasate. Alignment of extravasated HCELL+ human MSC along the endosteal surface was observed, with rare differentiation to osteoblasts as evidenced by immunohistochemical staining for human osteocalcin. These results indicate that despite absence of CXCR4 and many other effectors of cellular trafficking to marrow, osteotropism of systemically administered human MSC was conferred by ex vivo cell surface carbohydrate modification of a single glycoprotein, CD44, rendering the potent E-selectin ligand HCELL. Our findings unveil a readily translatable roadmap for programming MSC trafficking, thus overcoming a critical barrier in use of these cells for regenerative therapeutics of systemic skeletal diseases.
Article
Full-text available
The domesticated horse represents substantial value for the related sports and recreational fields, and holds enormous potential as a model for a range of medical conditions commonly found in humans. Most notable of these are injuries to muscles, tendons, ligaments and joints. Induced pluripotent stem (iPS) cells have sparked tremendous hopes for future regenerative therapies of conditions that today are not possible to cure. Equine iPS (EiPS) cells, in addition to bringing promises to the veterinary field, open up the opportunity to utilize horses for the validation of stem cell based therapies before moving into the human clinical setting. In this study, we report the generation of iPS cells from equine fibroblasts using a piggyBac (PB) transposon-based method to deliver transgenes containing the reprogramming factors Oct4, Sox2, Klf4 and c-Myc, expressed in a temporally regulated fashion. The established iPS cell lines express hallmark pluripotency markers, display a stable karyotype even during long-term culture, and readily form complex teratomas containing all three embryonic germ layer derived tissues upon in vivo grafting into immunocompromised mice. Our EiPS cell lines hold the promise to enable the development of a whole new range of stem cell-based regenerative therapies in veterinary medicine, as well as aid the development of preclinical models for human applications. EiPS cell could also potentially be used to revive recently extinct or currently threatened equine species.
Article
Full-text available
The aim of the study was to compare in vitro the stemness features of horse progenitor cells derived from bone marrow (BM-MSCs), amniotic fluid (AF-MSCs) and umbilical cord matrix (EUC-MSCs). It has been suggested that there may be a stem cell population within both umbilical cord matrix and amniotic fluid. However, little knowledge exists about the characteristics of these progenitor cells within these sources in the equine species. This study wanted to investigate an alternative and non-invasive stem cell source for the equine tissue engineering and to learn more about the properties of these cells for future cell banking. Bone marrow, umbilical cord and amniotic fluid samples were harvested from different horses. Cells were analyzed for proliferation, immunocytochemical, stem cell gene expression and multilineage plasticity. BM- and AF-MSCs took similar time to reach confluence and showed comparable plating efficiency. All cell lines expressed identical stem cell markers and capability to differentiate towards osteogenic lineage. Almost all cell lines differentiated into the adipogenic lineage as demonstrated by cytochemical staining, even if no adipose gene expression was detectable for AF-MSCs. AF- and EUC-MSCs showed a limited chondrogenic differentiation compared with BM-MSCs as demonstrated by histological and biochemical analyses. These findings suggest that AF-MSCs appeared to be a readily obtainable and highly proliferative cell line from an uninvasive source that may represent a good model system for stem cell biology. More studies are needed to investigate their multilineage potential. EUC-MSCs need to be further investigated regarding their particular behavior in vitro represented by spheroid formation.
Article
Full-text available
Induced pluripotent stem cell (iPS) technology has enriched the armamentarium of regenerative medicine by introducing autologous pluripotent progenitor pools bioengineered from ordinary somatic tissue. Through nuclear reprogramming, patient-specific iPS cells have been derived and validated. Optimizing iPS-based methodology will ensure robust applications across discovery science, offering opportunities for the development of personalized diagnostics and targeted therapeutics. Here, we highlight the process of nuclear reprogramming of somatic tissues that, when forced to ectopically express stemness factors, are converted into bona fide pluripotent stem cells. Bioengineered stem cells acquire the genuine ability to generate replacement tissues for a wide-spectrum of diseased conditions, and have so far demonstrated therapeutic benefit upon transplantation in model systems of sickle cell anemia, Parkinson's disease, hemophilia A, and ischemic heart disease. The field of regenerative medicine is therefore primed to adopt and incorporate iPS cell-based advancements as a next generation stem cell platforms.
Article
To confirm the characteristics of the stromal cells of Wharton's jelly, we investigated the morphological changes in these cells during each trimester of pregnancy. We evaluated the cytoskeletal features of these cells by examining immunohistochemically the localization of one of the contractile proteins, α-smooth muscle actin (ASMA). After the second trimester, the stromal cells of Wharton's jelly were stained with ASMA antibody, exhibited the ultrastructural characteristics of the myofibroblasts, and began to express numerous microfilaments in the cytoplasm. Postembedding immunogold labeling detected immunoreactivity for ASMA on these microfilaments. The finding indicated that the stromal cells of Wharton's jelly undergo a time-dependent maturation involving the differentiation of myofibroblasts during the last 6 months of pregnancy. These cells possess a contractile function that may help to protect the umbilical vessels from compression, considering that ASMA was detected in the microfilamentous bundles.
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
The field of regenerative medicine research is rapidly expanding. One area of interest to equine researchers is the possibility of isolating or generating pluripotent cells, capable of producing differentiated cell types derived from all 3 primary germ layers. Reports of equine embryonic stem-like (ES) cell isolation can be found in the literature. Other groups are working to produce equine-induced pluripotent stem (iPS) cells. This article summarizes the essential features needed to characterize a cell type as pluripotent, specific challenges in using the horse as a model organism for pluripotent cell generation, and current and upcoming clinical trials using ES/iPS cells.
Article
In the last decades, multipotent mesenchymal progenitor cells have been isolated from many adult tissues of different species. The International Society for Cellular Therapy (ISCT) has recently established that multipotent mesenchymal stromal cells (MSCs) is the currently recommended designation. In this study, we used flow cytometry to evaluate the expression of several molecules related to stemness (CD90, CD44, CD73 and STRO-1) in undifferentiated, early-passaged MSCs isolated from adipose tissue of four donor horses (AdMSCs). The four populations unanimously expressed high levels of CD90 and CD44. On the contrary, they were unexpectedly negative to CD73. A small percentage of the cells, finally, showed the expression of STRO-1. This last result might be due to the existence of a small subpopulation of STRO-1+ cells or to a poor cross-reactivity of the antibody. A remarkable donor-to-donor consistency and reproducibility of these findings was demonstrated. The data presented herein support the idea that equine AdMSCs may be easily isolated and selected by adherence to tissue culture plastic and exhibit a surface profile characterized by some peculiar differences in comparison to those described in other species. Continued characterization of these cells will help to clarify several aspects of their biology and may ultimately enable the isolation of specific, purified subpopulations.