SlideShare uma empresa Scribd logo
1 de 18
Baixar para ler offline
Mammalian MSC from Selected Species:
Features and Applications
Christiane Uder, Sandra Br€uckner, Sandra Winkler, Hans-Michael Tautenhahn,†‡
Bruno Christ†
*
 Abstract
Mesenchymal stromal/stem cells (MSC) are promising candidates for cellular therapy
of different diseases in humans and in animals. Following the guidelines of the Interna-
tional Society for Cell Therapy, human MSC may be identified by expression of a spe-
cific panel of cell surface markers (CD1051, CD731, CD901, CD34-, CD14-, or
CD11b-, CD79- or CD19-, HLA-DR-). In addition, multiple differentiation potential
into at least the osteogenic, adipogenic, and chondrogenic lineage is a main criterion
for MSC definition. Human MSC and MSC of a variety of mammals isolated from dif-
ferent tissues meet these criteria. In addition to the abovementioned, they express
many more cell surface markers. Yet, these are not uniquely expressed by MSC. The
gross phenotypic appearance like marker expression and differentiation potential is
similar albeit not identical for MSC from different tissues and species. Similarly, MSC
may feature different biological characteristics depending on the tissue source and the
isolation and culture procedures. Their versatile biological qualities comprising immu-
nomodulatory, anti-inflammatory, and proregenerative capacities rely largely on the
migratory and secretory capabilities of MSC. They are attracted to sites of tissue lesion
and secrete factors to promote self-repair of the injured tissue. This is a big perspective
for clinical MSC applications in both veterinary and human medicine. Phase I/II clini-
cal trials have been initiated to assess safety and feasibility of MSC therapies in acute
and chronic disease settings. Yet, since the mode of MSC action in a specific disease
environment is still unknown at large, it is mandatory to unravel the response of MSC
from a given source onto a specific disease environment in suitable animal models
prior to clinical applications. VC 2017 International Society for Advancement of Cytometry
 Key terms
cell therapy; clinical trials; differentiation; mammals; mesenchymal stem cells; surface
marker
BASED on his experiments with frogs, the German pathologist Cohnheim
(1839–1884) already hypothesized that cells especially from bone marrow may
migrate and repair tissue damage after injury. His observations were substantiated by
the work of Friedenstein et al. nearly 100 years later. They demonstrated that bone
marrow cells and their descendants of non-hematopoietic origin may differentiate in
vitro into cells of other tissues of mesenchymal origin (1). They termed these cells
“bone marrow fibroblasts,” which were renamed in the following years by various
authors into bone marrow stromal cells, mesenchymal stem cells, or skeletal stem
cells (2). The International Society for Cell Therapy published a position article
addressing the usage of the term “mesenchymal stem cell” (MSC). According to this
statement, it is recommended strictly to discriminate between “multipotent mesen-
chymal stromal cells” and “mesenchymal stem cells.” The first term was suggested to
be used for fibroblast-like plastic adherent cells irrespective of their tissue origin. The
latter designated stem cells, which meet particular stem cell definition criteria (3).
Today, the term “mesenchymal stromal cell” is often synonymously used
with “mesenchymal stem cell” or even preferred due to the fact that many cell
Department of Visceral, Transplantation,
Thoracic and Vascular Surgery, Applied
Molecular Hepatology Laboratory,
University Hospital of Leipzig,
Liebigstraße 21, Leipzig D-04103,
Germany
*Correspondence to: Bruno Christ, PhD,
Department of Visceral, Transplantation,
Thoracic and Vascular Surgery, Applied
Molecular Hepatology Lab, University
Hospital Leipzig, Liebigstraße 21, D-04103
Leipzig, Germany. Email: bruno.christ@
medizin.uni-leipzig.de
†
Equal contributions as senior authors.
‡
Present address: Division of General,
Visceral and Vascular Surgery, University
Hospital of Jena, Am Klinikum 1, Jena
07747, Germany
Published online 00 Month 2017 in Wiley
Online Library (wileyonlinelibrary.com)
DOI: 10.1002/cyto.a.23239
VC 2017 International Society for
Advancement of Cytometry
Cytometry Part A  00A: 00À00, 2017
Review Article
preparations do not necessarily feature true stem cell charac-
teristics like self-renewal and differentiation potential. Mini-
mal criteria have been postulated by the International Society
of Cell Therapy to define human MSC unequivocally. The first
criterion to be met by MSC is their ability to adhere to plastic
surfaces under standardized culture conditions. Second, a
minimum percentage of cells must feature a specified surface
marker profile, that is, 95% of the cells must express CD105,
CD73, and CD90, but 2% may express CD45, CD34, CD14
or CD11b, CD79 or CD19 and HLA-DR (4). Finally, MSC
must be able to differentiate at least into three different line-
ages like osteoblasts, adipocytes, and chondrocytes in vitro.
However, since these criteria do not take into account the vast
variety of MSC preparations from different sources and spe-
cies as well as the different isolation and culture conditions
yielding cells with different characteristics, it has been sug-
gested that a new description of MSC is necessary. Accord-
ingly, the term “mesenchymal stromal cell” should be used for
the general population of MSC, which is mostly a heteroge-
neous mix of different cell populations. The term
“mesenchymal stem cell” should be restricted to a cell popula-
tion featuring stem cell characteristics like self-renewal and
differentiation potential. In addition, description of cells
should include species, tissue resource, the information on
whether cells are primary or cultured, and culture passage
numbers besides others (5). Making up only 0.001 to 0.01%
of a tissue mass, the quantity of MSC in tissues is rare, their
morphology fibroblast-like and spindle-shaped (2,6,7). In
respect of their growth characteristics, Conget et al. observed
a doubling time of 33 h as well as an immense expansion
potential (8). Beyond these specific cell cycle characteristics,
the isolated cell population contained a subgroup of about
20% resting cells, presumably needed to replenish the stem
cell pool after growth stimulation (7).
The therapeutic use of MSC has been addressed in animal
models in a variety of applications comprising in general the
local implantation for regional tissue repair, systemic transplan-
tation for diseases affecting the entire organism, stem cell ther-
apy combined with gene therapy, and the generation/
development of engineered tissues (2). Clinically, MSC have
been established in clinical phase I/II trials to treat a wide range
of diseases comprising cardiovascular repair, therapy of lung
fibrosis and spinal cord injury as well as bone and cartilage
replacement besides others. In this context, it has been empha-
sized that the clinical use of MSC warrants extensive characteri-
zation of the cell product comprising identity, purity, safety,
and biological feasibility. Tissue-based cytometry and/or flow
cytometry were proposed as methodology of choice to unequiv-
ocally identify MSC and their subpopulations (9) (Fig. 1).
There is an overwhelming literature available dealing with MSC
from a great variety of organisms and their organs and tissues
including humans. It is nearly impossible to completely review
this vast magnitude of studies and to compare their outcomes
to each other. Therefore, in this review we focus on humans,
selected large animals as well as rodents and try to give a more
generalized overview on the organ and tissue resources of MSC
and their biological features in the context of the mode of MSC
actions and potential use of MSC in clinical applications.
Figure 1. Flow chart of current MSC therapeutic applications. In principle, the therapeutic use of MSC in humans or animals follows the
same rational. Cells are isolated most commonly from either bone marrow or adipose tissue because of the versatile availability. Cells
must be characterized unequivocally in terms of their phenotypic features and biological actions before delivery into patients, either
human or animal. Since MSC are classified Advanced Therapy Medicinal Products (ATMP), their safety, feasibility and therapeutic poten-
tial must be confirmed in humans by clinical trials according to regulatory guidelines. [Color figure can be viewed at wileyonlinelibrary.
com]
Review Article
2 Cross-species MSC
MSC AVAILABILITY AND MAINTENANCE
It seems that MSC are present in virtually all kinds of tis-
sues, organs, and organisms investigated so far. The methods
of isolation rely on the enrichment from the mononuclear cell
fraction by a number of different methods described. Because
of the ease of access and sufficient availability, the most prom-
inent sources for the isolation of MSC are bone marrow and
adipose tissue, which has been described in hominids, large,
and small animals. In the following chapter, both prominent
and some rare sources will be outlined representatively.
Tissue Sources
Principal steps in the isolation procedure to gain MSC
are similar in studies across species. In humans, large and
small animals, the MSC isolation procedure comprised tissue
digestion by collagenase, removal of debris by filtration of the
cell suspension, and density gradient centrifugation to obtain
the mononuclear cell fraction. Mononuclear cells were then
cultured on plastic dishes in simple Modified Eagle’s Medium
containing fetal calf serum as appropriate. After 24 h, floating
cells were removed to enrich for the plastic-adherent MSC.
With culture progressing, cells proliferated and grew to con-
fluency after a period in time (10–16). This method, mostly
used for isolation of MSC from soft tissues, is modified for
the isolation of MSC from human and small and large animal
bone marrow. In pigs, MSC were isolated from the substantia
spongiosa of the os femoris, which was scraped off and digested
by collagenase (17). After the subsequent density gradient cen-
trifugation to gain the mononuclear cell fraction, cells were
seeded onto plastic dishes and the MSC enriched following
protocols according to those described for the isolation of
MSC from soft tissues (9). Similar protocols have also been
used for the isolation of MSC from human bone marrow
(7,18–20). Bone marrow waste material is available from elec-
tive knee and hip surgery or major amputations and moreover
could be gained from voluntary crista iliaca punctures. Isola-
tion procedures are similar as compared to methods from
other species like dog (21,22), horse (23), or sheep (24) and
aim at enrichment of MSC from the mononuclear cell frac-
tion. In rodents, the isolation procedure is modified, taking
into account the low amount of bone marrow available at all.
Generally, mouse and rat bone marrow was completely
flushed out of the femur and tibia with an appropriate
medium and density gradient centrifugation was either per-
formed to enrich for mononuclear cells (25), or was omitted
for use of whole bone marrow cell populations (26). Methods
applying magnetic bead technology have been established for
example for positive selection using CD49a as a capture anti-
gen for the isolation of human MSC (27), or for negative
selection using CD45/glycophorin A to remove hematopoietic
cells from the mononuclear cell fraction derived from human
bone marrow (28), just to mention two examples out of a
plenty of methods describing positive or negative selection of
MSC from different sources and species.
A previous review highlighted the need for unequivocal
characterization of MSC from various sources with the goal to
identify subpopulations of cells especially in preparations
from adipose tissue sources, which apart from MSC contained
endothelial progenitors, pericytes, and others. This is relevant,
since flow cytometry analyses revealed different marker pro-
files on subpopulations of MSC and on MSC from different
sources, which might correlate to different functional biologi-
cal features of the cells (29). Accordingly, cross-species com-
parisons of MSC corroborated that markers and functions of
MSC might be similar between different sources and species,
but they are not identical. Human adipose tissue is collected
as waste material from elective abdominal surgery from
patients of different age and gender, and thus represents a
source providing MSC from different biological backgrounds.
While the gross biological features of MSC from adipose tissue
like expression of surface markers, morphology and differenti-
ation potential was similar to MSC from other sources, vari-
ability was detected in terms of isolation yield, proliferation
rates, and expandability (10,30–33). In line, adipose tissue-
derived MSC from non-human primates (rhesus monkey)
(34) and from horse subcutaneous adipose tissue adjacent to
the dorsal gluteal muscle (35) showed similar biological fea-
tures to human MSC from different sources in principle, but
varied in specific details like expression of distinct surface
markers, or doubling times and number of population dou-
blings during passaging. Porcine MSC from subcutaneous and
visceral adipose tissue with similar biological features were
explored in order to establish a large animal model of MSC
transplantation to investigate proof-of-concept as well as
safety aspects, relevant regulatory issues on the way to clinical
translation (11,36). In most experimental settings in rodents
(rat, mouse), MSC were isolated from bone marrow or adi-
pose tissue (37,38,123). Again, principal biological features
like morphology, multi-lineage differentiation potential, and
surface marker expression were similar, but not identical to
characteristics of MSC from other species and organs. Com-
paring characteristics of MSC from different species, tissues
and organs remains incomplete, since isolation procedures
and culture conditions varied between studies, which surely
contributes to differences in biological features and functions
of MSC as addressed below. However, it has been shown that
nearly all post-natal organs and tissues in the mouse harbored
MSC with similar features indicating a common precursor or
origin. Indeed, since MSC cultures could be grown from large
and small blood vessels all over the body, it was suggested that
MSC might reside in common perivascular niches in the dif-
ferent organs (39). A comprehensive overview on different
sources of MSC from hominids as well as large and rodent
mammals is given in Table 1.
In Vitro Expansion
It must be anticipated that culturing MSC changes their
biological features as discussed above. However, one promi-
nent quality of stem cells is their self-renewal potential, which
allows for the propagation of the MSC in vitro. This is of
utmost importance, since clinical application of MSC requires
cell numbers far higher than those originally obtained in a
na€ıve state from isolation procedures described in the
Review Article
Cytometry Part A  00A: 00À00, 2017 3
previous paragraph. Therefore, in vitro expansion of MSC was
extensively studied and revealed inconsistency concerning the
impact of isolation, culture and expansion procedures on the
biological characteristics of MSC. On the one hand, the multi-
ple differentiation potential of MSC has been documented
both in vitro and in vivo in various tissues confirming their
consistent functional performance even after extensive manip-
ulation by rigid isolation and cultivation procedures. This was
shown by applying various culture conditions to human bone
marrow-derived MSC (20): irrespective of whether the serum-
free culture medium was supplemented with basic fibroblast
growth factor (bFGF), with 2% fetal bovine serum (FBS)
together with epidermal growth factor and platelet-derived
growth factor (PDGF), or with 10% FBS, proliferation rates
or expression of surface molecules were not affected. From
these findings it was inferred that biological functions of MSC
were largely independent of and resistant to the method of
isolation and cultivation (102,103). In contrast, confluent
growth of human bone marrow-derived MSC changed the
expression of several cell surface markers and proteins secreted
into the culture supernatants, but did not impact on immu-
nomodulatory features (104). Because of the high clinical rele-
vance, these studies on the impact of culture conditions on
MSC characteristics have mostly been performed with human
MSC, and to a less extent with MSC from other species. Sheep
are often used as pre-clinical large animal models to test the
therapeutic potential of MSC or MSC engineered by scaffold-
ing for the treatment of degenerative joint diseases (105,106).
In this setting, ovine MSC changed their proliferative and dif-
ferentiation behavior as well as surface epitope expression
depending on growth factors or serum present in the culture
media (107). Similarly, the differentiation capabilities of MSC
decreased at later passages (40–50 population doublings)
(108) suggesting that frequent iterated passaging resulted in
aging and loss of stemness. Unrestricted differentiation poten-
tial of human MSC was confined up to the 6th passage.
Beyond this passage number, the loss of stem cell features
coincided with the decrease of the mean telomere length from
9.19 kb to 8.7 kb at the 9th passage (109). Not human, but
MSC derived from rhesus monkey developed a distinct popu-
lation of polyploid cells at passage 20, which progressed to
aneuploidy at passage 30 (110). In addition, chromosomal
instability arising at higher passage numbers was described
across several species including human (111), mouse (112),
and rat (113). In contrast, no chromosomal instability was
observed in endometrium-derived goat MSC up to passage
Table 1. Overview of the different tissue sources for MSC isolation in mammals (selected publications); 1 published
HOMINIDS LARGE ANIMALS SMALL ANIMALS
TISSUE REFERENCES HUMAN
RHESUS
MONKEY HORSE SHEEP PIG CATTLE DOG CAT RABBIT RAT MOUSE
Bone marrow (17,18,34,39–46) 1 1 1 1 1 1 1 1 1 1 1
Adipose tissue (10,11,34,35,37,38,
41,42,46–49)
1 1 1 1 1 1 1 1 1 1 1
Peripheral blood (50–57) 1 1 1 1 1 1
Umbilical cord blood (42,58–61) 1 1 1 1 1
Embryonic tissue (62–64) 1 1 1
Fetal blood (62) 1
Fetal heart (65) 1
Amnion fluid (66–69) 1 1 1 1
Amniotic membrane (67,70–72) 1 1 1 1
Placenta (73) 1
Synovia (44,74–77) 1 1 1 1 1 1
Trabecular bone (78–81) 1 1
Periosteum (75,82–84) 1 1 1 1
Tendon (42,85) 1 1 1
Dermis (86–88) 1 1 1
Teeth (89,90) 1 1
Muscle (39,75,84,91) 1 1 1 1
Brain (39) 1
Thymus (39,92) 1 1
Kidney (39) 1
Liver (39,41) 1 1
Fetal liver (64,93–95) 1 1 1 1
Fetal pancreas (96) 1
Pancreas (97–99) 1 1 1
Spleen (39,92) 1 1
Lung (39,100,101) 1 1
Review Article
4 Cross-species MSC
number 60 (114). In summary, the basic features of MSC from
different species comprising multi-lineage differentiation, sur-
face marker expression besides others seem to be very similar,
though not identical, at low passage numbers. Expansion
beyond changes characteristics mainly associated with general
signs of ageing.
PHENOTYPIC AND FUNCTIONAL FEATURES
Surface Markers
The list of surface antigens detected on MSC has grown
enormously in recent years. Surface markers expressed on
MSC across all species considered here are CD29 and CD44.
In contrast, the hematopoietic marker CD45 is not expressed,
discriminating MSC from hematopoietic stem cells unequivo-
cally. Some antigens like CD166 are only expressed in human
(116), rat (117), and sheep (107) MSC. CD166 is expressed on
osteoprogenitor cells and not on terminally differentiated
osteocytes, which supports the idea of dynamic regulation of
antigen expression during differentiation (117). While the
functional relevance of surface antigens on MSC is still mostly
elusive, the expression of cell adhesion molecules like integrin
a1, a2, a3, a5, a6, aV as well as b1, b3, and b4 may play an
important role in cell migration, homing and tissue integra-
tion of MSC (118). Most integrins were investigated in human
MSC (119), while integrin aV (CD51) was also detected in
mice (120). A comparison of investigated surface markers of
selected mammals is given in Table 2. It is noteworthy that
this list of markers is not specific or unique to identify MSC
definitely, because most proteins are expressed on other cells
as well. In addition, as discussed in the previous section, the
marker profiles on MSC might change due to different isola-
tion, culture, and expansion conditions or upon lineage differ-
entiation. Vice versa, it has been shown recently that, albeit
marker expression, telomere lengths and mitochondrial activ-
ity remained unchanged during long-term culture of human
bone marrow- and umbilical cord-derived MSC, functional
features like proliferative and immunosuppressive capacity
decreased over time (142). Therefore, expression of certain
MSC markers does not necessarily reflect the functional
capacities of MSC and vice versa, which in addition might dif-
fer in MSC from different species.
Immunomodulation
Immunomodulatory properties are an important feature
of MSC. From in vitro experiments it is known that MSC do
not trigger an immune response comprising allogeneic recog-
nition and elimination (143). Thus, the application of MSC
regarding cell therapy in allogeneic or even xenogeneic scenar-
ios seems to be feasible (144). In humans, anti-inflammatory
and immunomodulatory effects were described particularly in
graft-versus-host disease patients (143,144). In a xenogeneic
model crossing species borders, human MSC were trans-
planted in utero into fetal sheep (145). Effective engraftment
and persistence in multiple tissues was recorded as well as dif-
ferentiation into many cell or tissue types like chondrocytes,
myocytes, or thymic stroma. Surprisingly, the engraftment
persisted long-term suggesting unique immunological
acceptance, which possibly was attributable to the deficiency
of HLA class II surface antigens (9,78). Similarly, cross-species
immunotolerance was obtained using human placenta-
derived MSC to treat myelomeningocele in utero in a fetal
ovine model (146), or canine placenta-derived MSC to treat
neurological disorders in dogs (147). Horses, however, which
were transplanted with allogeneic MHC-mismatched MSC,
contained antibodies that killed donor MSC in an in vitro
cytotoxicity assay indicating that MHC compatibility seemed
required for acceptance of MSC transplants (148,149). In line,
MSC injection into healthy horses caused a mild increase of
blood CD8 T-cells and regulatory T cells in the spleen, which
might indicate a cytotoxic response to the allogeneic cell
transplants. However, no host tissue cytotoxic or inflamma-
tory was observed (150).
Generally, alloreactivity may be measured in vitro by the
secretion of IFNc by activated lymphocytes in the mixed lym-
phocyte reaction (MLR) assay. Using this assay, MSC sup-
pressed lymphocyte reactions using both autologous and
allogeneic T-cells or dendritic cells (9), and initiated the acti-
vation of alloreactive T-cells without inducing the T-cell pro-
liferation response (78). Even high numbers of human MSC
did not cause an alloreactive lymphocyte proliferation (151).
Allogeneic human bone marrow-derived MSC expressing
MHC molecules activated T-cells only in the presence of the
co-stimulatory factors like CD80 or CD86, which are not
expressed on MSC, even not after stimulation with interferon
c (IFNc) (152). Additionally, allogeneic MSC may stimulate
T-cells indirectly via antigen-presenting cells (APC) presenting
their MHC-antigens to T-cells. Yet, stimulation of prolifera-
tion of allogeneic lymphocytes by baboon MSC failed (121).
Also, human placenta-derived MCS were unable to stimulate
resting T cells, but inhibited phytohemagglutinin (PHA)-
stimulated T cells (73). Neither human nor rat MSC could
induce any IFNc output by PBMC (Peripheral Blood Mono-
nuclear Cells) indicating the lack of immunogenicity of MSC
(115). Also direct cell-cell interactions were involved in the
immune regulation by MSC. Human MSC bound activated
T-lymphocytes with higher affinity than resting cells,
T-lymphocytes better than B-lymphocytes and preferred cells
of the lymphatic lineage over the myelocyte cell lineage
(9,118). Along this line, human MSC minimized the T-cell
response indirectly via attenuation of dendritic cell matura-
tion from monocytes (153). After application of MSC into
injured mouse livers, more phagocytic cells were recruited
than to livers without MSC suggesting that the MSC boosted
cell recruitment (154). These data summarize that in all spe-
cies investigated so far, MSC modulated the immune response
by affecting various pathways and cells of the immune system
either directly or by communicating with other cells of the
immune defence, both the innate and the adaptive immune
system.
The immunomodulating features of MSC are mainly
mediated by paracrine mechanisms. Human MSC decreased
the secretion of the proinflammatory cytokines IFN-c (155),
IL-12 (156), and TNFa (155), while they elevated the anti-
inflammatory cytokine IL-10 (155,157,158). Human bone
Review Article
Cytometry Part A  00A: 00À00, 2017 5
Table 2. Overview of the expression of different surface antigens (1 positive, 6 heterogeneous, – negative) on human MSC compared
with mice, rats, pigs, and horses and their biological meaning or term
HOMINID
LARGE ANIMALS SMALL ANIMALS
CLUSTER OF
DIFFERENTIATION FUNCTION/LOCALIZATION HUMAN HORSE SHEEP PIG RAT MOUSE
CD1a Presentation of Lipid-Antigens
to T-Cells
–
CD9 TSPAN-29 (Tetraspanin 29) 1
CD10 MME (membrane
metallo-endopeptidase)
1
CD11b Integrin a M – – –
CD13 ANPEP (alanylaminopeptidase) 1 1
CD14 Monocytes/Macrophages – – –
CD18 Integrin b2 –
CD19 B-Lymphocytes –
CD24 HSA (Heat Stable Antigen) 1
CD25 Interleukin 2 Receptor a –
CD29 Integrin b1 1 1 1 1 1 1
CD31 PECAM 1 (platelet/endothelial
cell adhesion molecule 1)
– –
CD34 Hematopoietic stem cells – – – – –
CD40 TNF receptor superfamily
member 5
–
CD44 Hematopoietic cell E-/L-Selectin
ligand
1 1 1 1 1 1
CD45 PTPRC (protein tyrosine phos-
phatase, receptor type, C)
– – – – – –
CD46 Membrane co-factor protein 1
CD49a Integrin a1 1
CD49b Integrin a2 1
CD49c Integrin a3 1
CD49d Integrin a4 –
CD49e Integrin a5 1 1
CD49f Integrin a6 1
CD50 ICAM-3 (intercellular adhesion
molecule 3)
–
CD51 Integrin alpha-v 1 1
CD54 ICAM-1 (intercellular adhesion
molecule 1)
–
CD55 DAF (decay-accelerating factor) 1
CD56 NCAM 1 (neural cell adhesion
molecule 1)
1
CD58 LFA-3 (lymphocyte
function-associated antigen 3)
1 1
CD59 MAC-inhibitory protein
(MAC-IP)
1
CD61 Integrin b3 1 1 1
CD62 SELP (Selectin P) –
CD68 Lysosomal/endosomal-associated
membrane glycoprotein 4
(LAMP-4)
–
CD71 TFRC (transferrin receptor) 1 1
CD72 LYB-2 1
CD73 Ecto-5’-nucleotidase 1 – 6 1
Review Article
6 Cross-species MSC
marrow-derived MSC suppressed the proliferation of CD41
and CD81 T cells, which were stimulated by dendritic cells
(DC) and peripheral blood lymphocytes (PBL). Inhibition of
growth was not due to apoptosis and T cell proliferation could
be re-stimulated. This phenomenon had no immunologic
restriction and was triggered by both cellular and non-specific
mitogenic stimuli based on the production of soluble
paracrine factors (159). In a similar finding, human bone
marrow-derived MSC actively suppressed the proliferation of
T-cells stimulated by anti-CD3 and anti-CD28 antibodies as
well as the proliferation of responder PBMC stimulated by
third-party allogeneic PBMC. Separation of cultured MSC
and PBMC by a semi-permeable membrane did not prevent
suppression indicating the involvement of paracrine
TABLE 2. Continued
HOMINID
LARGE ANIMALS SMALL ANIMALS
CLUSTER OF
DIFFERENTIATION FUNCTION/LOCALIZATION HUMAN HORSE SHEEP PIG RAT MOUSE
CD80 B7-1 (ligand of CD28 and
CTLA-4);
T-lymphocyte activation
antigen
– 1
CD81 Tetraspanin 28 1
CD86 B7-2 (ligand of CD28 and
CTLA-5)
– –l 1
CD90 THY1 (thy-1 cell surface
antigen)
1 6 6 1 1 –
CD90.2 THY1.2 1
CD95 Fas 1
CD102 ICAM-2 (intercellular adhesion
molecule 2)
1
CD104 Integrin b4 1
CD105 Endoglin5TGFß receptor III 1 6 6 1
CD106 VCAM 1 (vascular cell adhesion
molecule 1)
6 1 1
CD117 c-Kit – –
CD119 Interferon c receptor alpha 1
CD120a Tumor necrosis factor receptor
superfamily, member 1A
1
CD123 Interleukin 3 receptor a –
CD124 Interleukin 4 receptor 1
CD127 Interleukin 7 receptor –
CD133 Prominin 1 –
CD146 MCAM (melanoma cell
adhesion molecule)
1 1
CD166 ALCAM 5SB-10; (activated leu-
kocyte cell adhesion molecule)
1 1
CD184 CXCR4, SDF-1alpha receptor 1 1
HLA-ABC HLA Class I/MHC I 1 1
HLA-DR HLA Class II/MHC II – – –
Sca-1 Stem cell antigen-1 1
SH-2 Antibody against epitope of
CD105
1
SH-3 Antibody against epitope of
CD73
1
SH-4 Antibody against another
epitope of CD73
1 1
References (7,8,62,97,98,
115,118,119,121,
122,124–126)
(42,127,128) (41,105,107,
129–132)
(17,133,134) (13,135) (136–141)
It appears that human profiles have been characterized best as compared to other species among mammals. This might be due to
the lack of suitable antibodies for FACS analyses but will for sure be part of future research in terms of functional characterization of MSC.
Review Article
Cytometry Part A  00A: 00À00, 2017 7
mechanisms mediated by secretion of IL-10, TGF-b1, prosta-
glandin E2 or tryptophane (152). In rodents with lung and
kidney injury, MSC improved the outcome by paracrine
effects mediated by a shift from a proinflammatory to an anti-
inflammatory cytokine milieu (115). Today, it is quite obvious
that MSC exert their pleiotropic actions on both the innate
and adaptive immune system via a great variety of different
factors and mediators comprising prostaglandins, cytokines
and growth factors, indoleamine 2,3-dioxygenase (IDO),
tumor necrosis factor a-stimulated gene 6 (TSG6), heme oxy-
genase 1 (HO-1), interleukin-1 receptor antagonist (IL-1RA)
besides others (cf. (160–162) for comprehensive reviews).
The immune suppression mediated by MSC may cross
species barriers: porcine, rodent, and human MSC suppressed
xenogeneic lymphocytes activated by lymphocytes of another
species. However, mechanistically there seem to be differences
between species, because human MSC were able to act onto
lymphocytes even if separated by a permeable membrane, but
the action of rodent MSC required cell-cell-contacts (115).
Corroborating these differences, human MSC induced no sus-
tained tolerance, anergy or apoptosis of activated lympho-
cytes, because they recovered their ability to be stimulated
after suppression of allogeneic stimulation of the lymphocytes
by human or baboon MSC. In contrast, lymphocytes co-
cultured with rodent MSC for 24 h, still produced IFNc, but
rested in G1 of the cell cycle and stopped proliferating, which
uncovers another difference of immunological mode of action
between the species (78,121,163). It was stated by Le Blanc
and Ringden that the suppression of T-cell activity by MSC
was not efficient enough to avoid xenotransplant rejection.
The natural immune response after xenotransplantation indi-
cates activation of the innate and acquired immune system,
involving humoral factors like antibodies and the complement
system, as well as cellular mechanisms involving for example,
natural killer cells and macrophages (115). It is obvious that
MSC from most species investigated may interfere with all
these levels of immune activation. Yet, a critical review of the
single factors is quite mandatory to understand the overarch-
ing coherences (121,164).
Migratory Potential
The fact that MSC have been found in almost all tissues
investigated so far might imply that they represent a stem cell
pool in the tissue to replenish tissue loss on demand, either
after injury, or in the context of physiological cell turnover.
However, it became obvious that MSC, when transplanted sys-
temically into injured hosts, migrated to the site of injury and,
under the influence of local damage signals, differentiated
into cells displaying the phenotype of the corresponding target
tissue. Thus, they contributed to repair of the impacted tissue
by functional replacement (9). Tracking of MSC migration
in vivo after transplantation in order to evaluate feasibility of
the site of application, homing, and tissue distribution as well
as functionality is of great interest. An elegant method has
been described by Ribot et al. who applied magnetic resonance
imaging (MRI) of human MSC transplanted into the hind
limb of nude mice after pre-loading of the cells with the
fluorophore fluorine or ultra-small iron-loaded nanoparticles
(165). Homing of MSC has been described for several species.
For example, bone marrow-derived MSC were transplanted in
a mouse model of acute hepatic injury and contributed to the
functional recovery of the liver (166). Also, in a rat model of
myocardial infarction, MSC administered systemically
migrated to the injured heart (167) again indicating attraction
of MSC by injured tissues. Migration might be provoked by
chemotactic signals produced by the tissue at the site of injury
(168). Stromal-derived factor-1 (SDF-1) seems an important
chemoattractant for MSC recruitment. Abbott et al. provided
evidence for SDF-1/CXCR4 interactions in the recruitment of
murine bone marrow-derived MSC to myocardial infarction
(169). CXCR4, also known as CD184 or SDF-1 alpha receptor,
is expressed by MSC from several species including canine
(170), human (122), and mice (136). CXCR4 expression
seems not only to be important for MSC migration to sites of
tissue injury, but also to the bone marrow as shown for
human MSC from bone marrow (137). Besides CXCR4, a row
of other surface proteins is likely to be involved in MSC hom-
ing. Ip et al. presented data showing that integrin1 ß (CD29)
but not CXCR4 on murine MSC was important for MSC
migration and engraftment into the infarcted myocardium
(171). In line, Ponte et al. identified several cytokines includ-
ing PDGF-AB, IGF-1, and RANTES, which were more effec-
tive in driving MSC homing than SDF-1. TNFa enhanced the
migratory response to these factors (172). A set of several che-
mokine receptors (CXCR4, CX3CR1, CXCR6, CCR1, and
CCR7) expressed on human bone marrow-derived MSC
seemed to be involved in the chemotactic migration in
response to CX3CL1 and CXCL12 as shown in a human pan-
creatic islet in vitro model (97). Monocyte chemoattractant
protein-1 (MCP-1), usually not expressed in healthy rat
brains, was rapidly up-regulated after middle cerebral artery
occlusion. In these ischemic rat brains, MCP-1 promoted the
migration of intravenously infused bone marrow-derived-
MSC to the site of injury. Migration was reduced significantly,
when brain tissue extracts were incubated with an antibody
against MCP-1 suggesting the chemotactic impact on MSC
migration (173). Corroborating the migratory potential of
MSC, native murine MSC transplanted into sublethally irradi-
ated mice could be recovered from the bone marrow and the
spleen already 24 h after systemic application showing their
rapid and efficient homing. This again underlined the
assumption that the mobilization of MSC and their subse-
quent homing to injured tissues depended on the systemic
and local inflammatory state of the organism (138,154).
Finally, matrix metalloproteinases (MMP) secreted by MSC
were required for the crossing of the basement membrane and
MSC tissue homing. The inflammatory cytokine-dependent
expression of MMP-2, MT1-MMP, and MMP-9 by MSC
strongly stimulated the chemotactic migration of human
MSC through basement membranes (174). Propagation of rat
bone marrow-derived MSC in vitro decreased their ability to
home to the bone marrow demonstrating that manipulation
of MSC in vitro prior to transplantation might severely
Review Article
8 Cross-species MSC
impact on their migratory potential and hence their therapeu-
tic potential (175).
Proregenerative Mode of Action
The regenerative potential of MSC, differentiated or not,
may principally be based on two mechanisms. One is character-
ized by functional tissue replacement of injured tissue, which
requires the MSC to differentiate and adopt the functional
properties of the host tissue after engraftment. The second fea-
ture is based on paracrine mechanisms promoting self-
regeneration of the injured tissue. The first may be achieved by
integration into the damaged tissue and proliferation of MSC
transplants at the site of tissue damage until complete tissue
repair, the second by the prevention of injury progress followed
by stimulation of self-repair of the tissue affected. It is quite
obvious that tissue integration needs direct cell–cell interac-
tions, while paracrine mechanisms may act locally after recruit-
ment of MSC transplants to the site of injury. This
consideration is of high relevance in clinical applications. Acute
organ injury needs temporary therapeutic intervention com-
prising rapid local actions of MSC to promote self-repair of the
injured tissue/organ. Chronic diseases, however, require sus-
tained tissue support preferably by tissue integration and differ-
entiation of MSC into the cell type of the injured tissue in order
to ameliorate organ/tissue dysfunction long-term by functional
replacement of the diseased host tissue.
In a xenogeneic mouse model of partial hepatectomy,
human subcutaneous adipose tissue-derived MSC, which had
been differentiated into hepatocyte-like cells prior to trans-
plantation, re-populated the liver of immune-deficient mice
by about 25% ten weeks after transplantation. This was a 10-
times higher rate as compared to the use of undifferentiated
MSC (176). While in this experiment an integration of cell
transplants into the liver was achieved indicating the func-
tional substitution of the recipient liver by donor MSC, a
mixed direct and paracrine effect was described in mice and
rats after intoxication with acetaminophen. The drug induces
regional necrosis in the liver, which was occupied by trans-
planted human bone marrow-derived and rat adipose tissue-
derived MSC in immune-deficient mice and rats, respectively.
They prevented disease progress by inhibition of apoptosis
and necrosis, and supported liver regeneration by stimulating
proliferation of host hepatocytes. Functional cell transplants
were found in the host livers for up to seven weeks after trans-
plantation (154,164). This indicates that MSC are capable of
liver support by functional tissue substitution and paracrine
stimulation of tissue regeneration after acute inflammatory
insult. Similar results were obtained in rats, in which acute
liver failure was induced by D-galactosamine. Here, systemic
injection of MSC reduced both the apoptotic rate and aug-
mented the proliferation of host liver hepatocytes indicative
for the stimulation of cell cycle progression as verified by an
increase in the expression of cell cycle proteins. This clearly
demonstrated the paracrine hepatotropic action of MSC
transplants (177). Human MSC from fetal blood also stimu-
lated the proliferation and differentiation of CD341 hemato-
poietic stem cells from umbilical cord blood (62). These
proproliferative features of MSC were mediated by secretion
of growth factors. In vitro chondrogenesis of periost-derived
MSC from young rabbits was strongly stimulated by a combi-
nation of growth factors IGF-1 and TGF-b1 (178). A caprine
model of osteoarthritis combined with local application of
autologous bone marrow-derived MSC corroborated the
pleiotropic regenerative potential of MSC. In this model,
autologous MSC labeled with green fluorescent protein (GFP)
were injected intra-articularly six weeks after unilateral induc-
tion of osteoarthritis in the knees of goats. GFP-labeled cells
were detected in the treated joints and successfully regenerated
the meniscus (179). Together these data show that MSC
possess a proregenerative potential, which makes them
ideal candidates for regenerative medicine in a wide variety
of degenerative disease applications both in humans and
animals.
CLINICAL USE
Originating from the versatile successful therapeutic use
in animal experiments, hope grew that MSC therapy might be
feasible clinically. However, clinical animal trials have only
been performed in dogs, cats and horses. In humans, a bulk of
phase I/II clinical trials were initiated to assess safety and feasi-
bility of MSC therapy under various disease conditions.
Animal Trials
Only few clinical trials in pets like cats and dogs are avail-
able. Intraperitoneal autologous adipose tissue-derived MSC
injection was evaluated in 10 healthy adult cats. The injection
was well tolerated with only mild adverse events in two cats
(180). Quimby et al. described a randomized, placebo-
controlled trial in eight cats evaluating intravenous applica-
tion of adipose-derived allogeneic MSC in feline chronic kid-
ney disease. The administration of MSC was not associated
with adverse events, however, significant improvement of
renal function was not achieved by MSC administration
(181). The safety of intravenous application of allogeneic adi-
pose tissue-derived MSC was assessed in 11 dogs suffering
from inflammatory bowel disease. The single injection of
MSC was safe and improved gastrointestinal lesions by reduc-
ing gastrointestinal inflammation (182).
In large animal clinics, MSC are used mainly to treat
musculoskeletal diseases in horses like tendon injuries or bone
and cartilage damage (183). Feasibility of autologous, geneti-
cally modified autologous, allogeneic, and xenogeneic MSC
was tested by joint injection in six healthy horses. A total
number of 15 3 106
MSC induced a moderate acute inflam-
matory response in the joint, which was greater using alloge-
neic and xenogeneic as compared to autologous MSC (184).
Thus, both autologous and allogenic MSC application in
horses seems to be safe and feasible. Accordingly, in most
equine clinical trials autologous bone marrow- or adipose
tissue-derived MSC were used. The most prominent diseases
treated represented tendon lesions, which showed improve-
ment after injection of 2 3 106
–10 3 106
autologous MSC
directly into the site of the lesion. No study reported of
adverse effects provoked by MSC application. Thirteen out of
Review Article
Cytometry Part A  00A: 00À00, 2017 9
18 horses returned to race competitions after autologous bone
marrow-derived MSC injection into sites of ligament and ten-
don lesions (185). Similar findings demonstrated recovery
from lesions of the superficial digital flexor tendon in race-
horses in 9 out of 11 horses by MSC treatment (186). Bone
marrow-derived MSC improved superficial digital flexor ten-
dinopathy and reduced the re-injury rate in national hunt
racehorses (187). Therapy of tendonitis with MSC was
assessed in horses in a randomized controlled trial by direct
injection of 10 3 106
autologous adipose tissue-derived MSC.
These prevented progression of tendon lesion and decreased
inflammation (188). Albeit no significant difference was
achieved clinically or histologically in horses after treatment
of intra-articular cartilage lesions with bone marrow-derived
MSC, it seemed that repair quality was improved by augmen-
tation of aggrecan content and tissue firmness (189). Only
partial repair was described treating equine bone spavin with
adipose tissue-derived MSC (190). In summary, equine MSC
might thus be clinically established to treat inflammatory
acute and chronic degenerative diseases of tendon, cartilage
and bone. Overall, the clinical application of both autologous
and allogenic MSC in several animal species including horse,
dog and cat turned out safe and without adverse events.
Human Trials
The ClinicalTrials.gov register provides a plenty of clini-
cal trials using human MSC administration for treatment of
different diseases. Studies not yet recruiting patients will not
be discussed here. A summary of studies reporting
“recruitment completed” and comprising the most relevant
diseases is given in Table 3. Most of the studies involving MSC
registered on ClinicalTrials.gov do not present results so far.
Therefore, we retrieved the PubMed database using the MeSH
terms “clinical trials” and “mesenchymal stromal cells.”
Results are summarized in Table 4 comprising type of disease,
source of MSC, route and dose of application, number of
patients, study type and outcome of both human and equine
clinical trials. Most study results are available for MSC appli-
cation in degenerative neuronal diseases like amyotrophic lat-
eral sclerosis (ALS), an as yet untreatable, progressive,
neurodegenerative disorder resulting in death of
motoneurons, or multiple sclerosis (MS), a chronic inflamma-
tion of the nervous system. Except for one study using adipose
tissue-derived MSC (191) to treat ALS or MS, all other clinical
trials used autologous bone marrow-derived MSC (192–199).
For the treatment of MS, intravenous application was chosen,
whereas for ALS cells were mostly injected intrathecally. Cell
numbers injected ranged from 1 3 106
–1 3 108
cells per kg
body weight. In all phase I/II clinical trials reporting treatment
of ALS or MS with MSC, cell application provoked no adverse
events, retarded ALS progression (194,196), induced immu-
nomodulatory effects (196,199) and improved visual acuity in
MS (198). Autologous bone marrow- or adipose tissue-
derived MSC application showed no adverse events in spinal
cord injury (200–202) or in osteoarthritis (203,204). For the
treatment of graft-versus-host disease (GvHD), mainly alloge-
neic bone marrow-derived stem cells were administrated
intravenously causing also no adverse events and featuring
immunosuppressive properties (205,206). This was corrobo-
rated in a pediatric study of GvHD, which confirmed the
immunomodulatory potential of allogenic MSC (207). The
anti-inflammatory action of both autologous (208) and
allogenic (209) MSC was demonstrated in Crohn’s disease
after intravenous application of bone marrow-derived MSC.
Treatment of several liver diseases including liver transplanta-
tion with MSC has shown safety and feasibility (210). In
acute-on-chronic liver failure, allogeneic umbilical cord-
derived MSC lowered the MELD (Modell of End stage Liver
Diseases) score indicating improvement of liver function
(211). In liver cirrhosis, representing another serious impair-
ment of liver function, autologous bone marrow-derived MSC
also reduced the MELD score (212). Corroborating the benefi-
cial impact on acute diseases, the application of autologous
(213) or allogeneic bone marrow-derived MSC (214) was safe
and improved the NYHA (New York Heart Association) score
in acute myocardial infarction.
In summary, the outcomes of clinical trials using human
MSC for therapy of acute and chronic inflammatory diseases
of different etiologies show that the treatment is bona fide safe
and may encompass cells of autologous or allogeneic origin.
The major mode of MSC action is probably the modulation
of inflammatory processes attenuating tissue damage and fos-
tering self-repair of the injured tissue. Since in most studies
cells were administered systemically, soluble or, after migra-
tion of MSC to the site of injury, even paracrine mechanisms
must be anticipated, thus substantiating findings, which have
been prior proven in animal models as discussed above.
Safety Issues
Despite their well acknowledged and unique proregener-
ative and immunomodulatory features, which make MSC
highly attractive for numerous applications in regenerative
medicine as shown above, some critical issues potentially limit
the clinical use of MSC. This section will finally give a short
insight into the contrary views on special safety concerns.
A serious constraint of the therapeutic use of MSC is
their putative profibrogenic potential (216–218). Human
bone marrow-derived MSC contributed to lung fibrosis (219)
Table 3. Overview of human clinical trials involving MSC therapy
with patient recruitment completed
DISEASE
NO. OF REGISTERED
STUDIES
ALS (amyotrophic lateral sclerosis) 6
MS (multiple sclerosis) 8
Spinal cord injury 9
Osteoarthritis 30
Graft-versus-host disease 11
Crohns disease 4
Liver diseases 8
Kidney disease 2
Acute myocardial infarction 6
Source: ClinicalTrials.gov
Review Article
10 Cross-species MSC
Table4.Compilationofclinicaltrialsusingmesenchymalstemcellsinhumansandhorsesandstudyoutcome
SPECIESDISEASEORIGINOFMSC
ROUTEOFAPPLICATIONCELL
NUMBERSINFUSEDNO.PATIENTSTUDYTYPEOUTCOMEREF
HumanAmyotrophic
lateralsclerosis
(ALS)
AutologousbonemarrowIntrathecally2x1*106
/kg8PhaseINoadverseevents(192)
Autologousbonemarrow37Investigator-
initiatedtrial
VEGF,ANG,and
TGF-blevelsinMSCidentifiedaspotential
biologicalmarkerstopredicttheeffective-
nessofautologousMSCtherapy
(193)
AutologousbonemarrowIntramuscularly
intrathecally
1*106
/kg
6
6
PhaseI/IINoadverseevents
reductioninALSFunctionalRatingScale
Revised(ALS-FRS-R)slope
(149)
AutologousadiposetissueIntrathecally107
-108
/kg27PhaseINoadverseevents(191)
Autologousbonemarrowintrathecally1564.5*106
26PhaseI/IIaNoadverseevents
retardeddiseaseprogression
(195)
ALS/MSAutologousbonemar-rowintrathecally
intravenously2.5*106
34
14
PhaseI/IIClinicallyfeasible/noadverseevents
inducesimmediateimmunomodulatory
effects
(196)
Multiplesclerosis
(MS)
AutologousbonemarrowIntravenously1–2*106
/kg11PhaseIIaInitialsafetyprofileandfeasibilityofthe
intervention
(197)
AutologousbonemarrowIntravenously1.6*106
/kg10PhaseIIaNoseriousadverseevents
improvementofvisualacuity
(198)
AutologousbonemarrowIntravenously1–2*106
/kg8PhaseIINoadverseevents
reductionofinflammation
(199)
SpinalcordinjuryAutologousadiposetissueIntravenously4*108
12PhaseINoadverseevents
notumordevelopment
(200)
BonemarrowLocally107
/ml14PhaseINoadverseevents
variableimprovementsintactilesensitivity
(201)
Autologousbonemarrowintramedullary1.6*106
subdurally3.23106
16PhaseIIINoadverseevents
veryweaktherapeuticeffectcomparedwith
multipleMSCsinjection
(202)
OsteoarthritisAutologousadiposetissueintraarticularly
lowdose1.0*107
middose5.0*107
highdose1.0*108
9PhaseINoadverseevents
highdoseimprovedfunctionandpain
(203)
Autologousadiposetissueintraarticularly
lowdose
2*106
middose10*106
highdose50*106
10PhaseINoadverseevents
patientstreatedwithlow-doseMSCexperi-
encedsignificantimprovementsinpain
levels
(204)
Graft-versus-host
disease(GvHD)
Allogeneicbonemarrowintravenously
1.1*106
/kg
25PhaseI/IINoadverseeventsrelatedtotheMSC
17responded
(205)
Review Article
Cytometry Part A  00A: 00À00, 2017 11
TABLE4.Continued
SPECIESDISEASEORIGINOFMSC
ROUTEOFAPPLICATIONCELL
NUMBERSINFUSEDNO.PATIENTSTUDYTYPEOUTCOMEREF
7patientsdidnotrespondtoMSCinfusion
Allogeneicbonemarrow0.9-9*106
/kg8PhaseIimmunosuppressive
healdamagedintestinalepithelium
(206)
Allogeneicbonemarrowintravenously
2*106
/kg
severalapplications
every4days
12
(pediatric)
PhaseI/IINoinfusionalorotheridentifiableacutetox-
icityadverseevents
multipleinfusionswerewelltoleratedand
weresafeinchildren
clinicalresponses,particularlyintheGIsys-
tem,wereseeninthemajorityofchildren
withsevererefractoryaGvHD
(207)
Crohn’sdiseaseAutologousbonemarrowintravenouslyupto106
/kg12PhaseInoadverseeventsandfeasible(208)
Allogeneicbonemarrowintravenously2*106
/kg15PhaseIIreducedCrohn’sDiseaseActivityIndex
(CDAI)andCrohn’sDiseaseEndoscopic
IndexofSeverity(CDEIS)
(209)
Livertransplantation
Acute-on-chronic
liverfailure
Livercirrhosis
n.k.portalvein(0d)centralvein
(2d)1.5*108
1Casestudynoevidenceofacutetoxicity
administrationtechnicallyfeasible
(210)
Allogeneicumbilicalcordintravenously0.5*106
24PhaseI/IInoadverseevents
reductionofMELDscore
(211)
Autologousbonemarrowintravenouslyorportalvein
3–5*106
/kg
8PhaseInoadverseevents
improvementofModelforEnd-stageLiver
Disease(MELD)score
(212)
KidneydiseaseAutologousbonemarrowintravenously1–2*106
/kg6PhaseINoadverseevents
anticipatedsystemicimmunosuppression
(215)
Acute
myocardial
infarction
Autologousbonemarrow
Allogeneicbonemarrow
10infarctedsites
20–200*106
15
15
PhaseI/II
randomized
comparison
Noadverseeventswithautologousandallo-
genicMSC
improvementofNewYorkHeartAssociation
(NYHA)score
(214)
AutologousBoneMarrowInfarct-relatedartery
1*106
cells
80PhaseII/IIINoadverseevents
modestimprovementinleftventricularejec-
tionfraction(LVEF)at6-monthfollow-up
(213)
HorseTendonlesionAutologousbonemarrowintolesion
2*106
/cm2
18PhaseI/IINoadverseevents
13horsesreturnedtoracecompetitions
(185)
Autologousbonemarrowintolesion
10*106
141PhaseI/IINoadverseeffects
reductionofthere-injuryrateaftersuperficial
digitalflexortendinopathy,esp.inNational
Huntracehorses.
(187)
Autologousadiposetissueintolesion
10*106
8phaseI/IIPreventionoftendonlesionprogressionas
observedbyultrasoundexamination
improvementoftissueorganization
attenuationofinflammation
(188)
Review Article
12 Cross-species MSC
and the fibrotic action of ovarian endometrial stromal cells
was enhanced by paracrine production of TGF-b1 and Wnt1
after treatment with human endometrium-derived MSC
(220). However, studies in rats and mice demonstrated that
MSC did not contribute to but rather improved liver fibrosis
(221–224). In rodent models of liver regeneration after partial
hepatectomy in rats and mice using either syngeneic or xeno-
geneic MSC, no signs of profibrotic impact for up to ten
weeks after transplantation were observed (13,176). The anti-
fibrotic effects of human MSC transplanted into xenogeneic
rodent models were also obvious under acute and chronic
liver (154,225), lung (138,226,227), or kidney (228,229) dam-
age conditions. For instance, in a murine bleomycin-induced
lung fibrosis model, MSC lowered type I pro-collagen, alpha-
smooth muscle actin (a-SMA), TGF-ß and the oncogenic
transcription factor c-Myc (230). Furthermore, transplanta-
tion of MSC into heart, muscle or lung did not result in any
extracellular matrix changes indicative for fibrosis or emer-
gence of malignant cells (47). Human bone marrow-derived
MSC suppressed abnormal collagen I deposition in the
nucleus pulposus by modulating MMP12 and HSP47 (231).
In several rodent models of liver fibrosis, the antifibrotic effect
of human bone marrow- and placenta-derived MSC on the
resolution of fibrosis depended on the balance between the
matrix metalloproteinases (MMP) and their inhibitors
(TIMP) (223,232,233). Especially MMP2 (234) and MMP9
(232) were activated by MSC. Furthermore, the expression of
a-SMA was inhibited and apoptosis of hepatic stellate cells
augmented, all contributing to regression of fibrosis
(235,236). TGF-b1, the major mediator of fibrosis, was down-
regulated by human MSC in fibrotic mouse livers (237), and
its signaling through the phosphorylation of Smad2 was atten-
uated (238). Transplantation of human umbilical cord blood-
derived MSC increased IL-4, which promoted mobilization of
Kupffer cells alleviating liver fibrosis in rats (239).The contro-
versial results showing either fibrotic or antifibrotic features of
MSC might also depend on the use of different tissue sources
and methods used for the isolation of MSC as well as on the
different animal models or fibrotic organs used. Accordingly,
it has been demonstrated in mice that adipose tissue-derived
human MSC were significantly more efficient in reducing skin
fibrosis as compared to bone marrow-derived MSC. This dif-
ference was related to a stronger reduction of TNFa and IL-1b
as well as an enhanced ratio of MMP1/TIMP1 in skin and
lung tissues when treated with adipose tissue- as compared to
treatment with bone marrow-derived MSC (240).
CONCLUSION/SUMMARY
In mammals a big variety of different tissue and organ
sources harboring MSC exists. Nearly every tissue investigated
so far contained MSC with similar biological characteristics
such as surface marker expression indicating conserved fea-
tures and functions in mammals at least. Carrying a low ethi-
cal burden, and considering their high regenerative properties,
MSC are good candidates for the use of cellular therapy in
both human clinical and in veterinary medical settings. The
TABLE4.Continued
SPECIESDISEASEORIGINOFMSC
ROUTEOFAPPLICATIONCELL
NUMBERSINFUSEDNO.PATIENTSTUDYTYPEOUTCOMEREF
Autologousn.k.n.k.11PhaseI/IIClinicalrecoveryfromtendonlesionsin9out
of11horses
(186)
BonespavinAutologousadiposetissueintraarticularly
n.k.
n.k.PhaseI/IILong-termbeneficialinfluence(190)
CartilagelesionAutologousbonemarrowIntraarticularly
20*106
10PhaseI/IIIncreaseofaggrecanlevelsofinrepairedtissue
noimprovementoflesions.
(189)
HealthyAutologous
Autologous,genetically
modified
allogeneic
xenogeneic
Intojoint
15*106
6PhaseIModerateacuteinflammatoryjointresponse
thatwasgreaterforallogeneicandxenoge-
neicMSC
(184)
Theclassificationoftrialsinhorsesdoesnotrepresentanofficialterminologyasusedinhumanclinicaltrials.Ithasbeenintroducedinordertodemonstratethatsafetyandfeasi-
bilityaswellasinthesecondlinetherapeuticeffectivenesshasbeentestedasusuallyinhumanclinicalphaseI/IItrials.n.k.5notknown.
Review Article
Cytometry Part A  00A: 00À00, 2017 13
regeneration of tendon, bone and cartilage seems to be the
most promising clinical application at present in large animals
like horses but in humans as well. Nevertheless, their anti-
inflammatory and immunomodulatory features make MSC
highly attractive for clinical use in the setting of a huge range
of diseases comprising both acute and chronic inflammatory
tissue deteriorations in man and animals. Yet, it is mandatory
prior to clinical applications to unravel the response of MSC
from a given source onto a specific disease environment in
suitable animal models.
ACKNOWLEDGMENTS
All authors declare that they have no conflicts of interest.
AUTHOR CONTRIBUTIONS
All authors substantially contributed to conception and
design, drafting and writing the article and revising it critically
for important intellectual content.
LITERATURE CITED
1. Friedenstein AJ, Chailakhyan RK, Gerasimov UV. Bone marrow osteogenic stem
cells: In vitro cultivation and transplantation in diffusion chambers. Cell Tissue
Kinet 1987;20:263–272.
2. Kassem M, Kristiansen M, Abdallah BM. Mesenchymal stem cells: Cell biology and
potential use in therapy. Basic Clin Pharmacol Toxicol 2004;95:209–214.
3. Horwitz EM, Le Blanc K, Dominici M, Mueller I, Slaper-Cortenbach I, Marini FC,
Deans RJ, Krause DS, Keating A. Clarification of the nomenclature for MSC: The
International Society for Cellular Therapy position statement. Cytotherapy 2005;7:
393–395.
4. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans
R, Keating A, Prockop D, Horwitz E. Minimal criteria for defining multipotent mes-
enchymal stromal cells. The International Society for Cellular Therapy position
statement. Cytotherapy 2006;8:315–317.
5. Keating A. Mesenchymal stromal cells: New directions. Cell Stem Cell 2012;10:
709–716.
6. Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: Mesenchymal stem
cells: Their phenotype, differentiation capacity, immunological features, and poten-
tial for homing. Stem Cells 2007;25:2739–2749.
7. Pittenger MF. Multilineage potential of adult human mesenchymal stem cells.
Science 1999;284:143–147.
8. Conget PA, Minguell JJ. Phenotypical and functional properties of human bone mar-
row mesenchymal progenitor cells. J Cell Physiol 1999;181:67–73.
9. Barry FP, Murphy JM. Mesenchymal stem cells: Clinical applications and biological
characterization. Int J Biochem Cell Biol 2004;36:568–584.
10. Seo MJ, Suh SY, Bae YC, Jung JS. Differentiation of human adipose stromal cells into
hepatic lineage in vitro and in vivo. Biochem Biophys Res Commun 2005;328:
258–264.
11. Bruckner S, Tautenhahn HM, Winkler S, Stock P, Dollinger M, Christ B. A fat option
for the pig: Hepatocytic differentiated mesenchymal stem cells for translational
research. Exp Cell Res 2014;321:267–275.
12. Kim D, Monaco E, Maki A, de Lima AS, Kong HJ, Hurley WL, Wheeler MB.
Morphologic and transcriptomic comparison of adipose- and bone-marrow-derived
porcine stem cells cultured in alginate hydrogels. Cell Tissue Res 2010;341:359–370.
13. Sgodda M, Aurich H, Kleist S, Aurich I, Konig S, Dollinger MM, Fleig WE, Christ B.
Hepatocyte differentiation of mesenchymal stem cells from rat peritoneal adipose
tissue in vitro and in vivo. Exp Cell Res 2007;313:2875–2886.
14. Laschke MW, Schank TE, Scheuer C, Kleer S, Shadmanov T, Eglin D, Alini M,
Menger MD. In vitro osteogenic differentiation of adipose-derived mesenchymal
stem cell spheroids impairs their in vivo vascularization capacity inside implanted
porous polyurethane scaffolds. Acta Biomater 2014;10:4226–4235.
15. Tholpady SS, Katz AJ, Ogle RC. Mesenchymal stem cells from rat visceral fat exhibit
multipotential differentiation in vitro. Anat Rec A Discov Mol Cell Evol Biol 2003;
272:398–402.
16. Lin Y, Chen X, Yan Z, Liu L, Tang W, Zheng X, Li Z, Qiao J, Li S, Tian W. Multiline-
age differentiation of adipose-derived stromal cells from GFP transgenic mice. Mol
Cell Biochem 2006;285:69–78.
17. Bruckner S, Tautenhahn HM, Winkler S, Stock P, Jonas S, Dollinger M, Christ B.
Isolation and hepatocyte differentiation of mesenchymal stem cells from porcine
bone marrow–”surgical waste” as a novel MSC source. Transplant Proc 2013;45:
2056–2058.
18. Aurich I, Mueller LP, Aurich H, Luetzkendorf J, Tisljar K, Dollinger MM,
Schormann W, Walldorf J, Hengstler JG, Fleig WE, et al. Functional integration of
hepatocytes derived from human mesenchymal stem cells into mouse livers. Gut
2007;56:405–415.
19. Pelz S, Stock P, Bruckner S, Christ B. A methionine-choline-deficient diet elicits
NASH in the immunodeficient mouse featuring a model for hepatic cell transplanta-
tion. Exp Cell Res 2012;318:276–287.
20. Lodie TA, Blickarz CE, Devarakonda TJ, He C, Dash AB, Clarke J, Gleneck K,
Shihabuddin L, Tubo R. Systematic analysis of reportedly distinct populations of
multipotent bone marrow-derived stem cells reveals a lack of distinction. Tissue Eng
2002;8:739–751.
21. Kazemi D, Shams Asenjan K, Dehdilani N, Parsa H. Canine articular cartilage regen-
eration using mesenchymal stem cells seeded on platelet rich fibrin: Macroscopic
and histological assessments. Bone Joint Res 2017;6:98–107.
22. Kadiyala S, Young RG, Thiede MA, Bruder SP. Culture expanded canine mesenchy-
mal stem cells possess osteochondrogenic potential in vivo and in vitro. Cell Trans-
plant 1997;6:125–134.
23. Fortier LA, Nixon AJ, Williams J, Cable CS. Isolation and chondrocytic differentia-
tion of equine bone marrow-derived mesenchymal stem cells. Am J Vet Res 1998;59:
1182–1187.
24. Caminal M, Velez R, Rabanal RM, Vivas D, Batlle-Morera L, Aguirre M, Barquinero
J, Garcia J, Vives J. A reproducible method for the isolation and expansion of ovine
mesenchymal stromal cells from bone marrow for use in regenerative medicine pre-
clinical studies. J Tissue Eng Regen Med 2016; Nov 18. doi: 10.1002/term.2254.
[Epub ahead of print]
25. Gao LR, Zhang NK, Bai J, Ding QA, Wang ZG, Zhu ZM, Fei YX, Yang Y, Xu
RY, Chen Y. The apelin-APJ pathway exists in cardiomyogenic cells derived
from mesenchymal stem cells in vitro and in vivo. Cell Transplant 2010;19:
949–958.
26. Tropel P, Noel D, Platet N, Legrand P, Benabid AL, Berger F. Isolation and characteri-
sation of mesenchymal stem cells from adult mouse bone marrow. Exp Cell Res
2004;295:395–406.
27. Deschaseaux F, Gindraux F, Saadi R, Obert L, Chalmers D, Herve P. Direct
selection of human bone marrow mesenchymal stem cells using an anti-CD49a
antibody reveals their CD45med,low phenotype. Br J Haematol 2003;122:506–
517.
28. Tondreau T, Lagneaux L, Dejeneffe M, Massy M, Mortier C, Delforge A, Bron D.
Bone marrow-derived mesenchymal stem cells already express specific neural pro-
teins before any differentiation. Differentiation 2004;72:319–326.
29. Donnenberg VS, Ulrich H. Mesenchymal stem cells, therapy, and cytometry. Cytom-
etry A 2013;83:8–10.
30. Lin Y, Liu L, Li Z, Qiao J, Wu L, Tang W, Zheng X, Chen X, Yan Z, Tian W. Pluripo-
tency potential of human adipose-derived stem cells marked with exogenous green
fluorescent protein. Mol Cell Biochem 2006;291:1–10.
31. Oedayrajsingh-Varma MJ, van Ham SM, Knippenberg M, Helder MN, Klein-Nulend
J, Schouten TE, Ritt MJ, van Milligen FJ. Adipose tissue-derived mesenchymal stem
cell yield and growth characteristics are affected by the tissue-harvesting procedure.
Cytotherapy 2006;8:166–177.
32. Kern S, Eichler H, Stoeve J, Kluter H, Bieback K. Comparative analysis of mesenchy-
mal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells
2006;24:1294–1301.
33. Wagner W, Wein F, Seckinger A, Frankhauser M, Wirkner U, Krause U, Blake J,
Schwager C, Eckstein V, Ansorge W, et al. Comparative characteristics of mesenchy-
mal stem cells from human bone marrow, adipose tissue, and umbilical cord blood.
Exp Hematol 2005;33:1402–1416.
34. Izadpanah R, Trygg C, Patel B, Kriedt C, Dufour J, Gimble JM, Bunnell BA. Biologic
properties of mesenchymal stem cells derived from bone marrow and adipose tissue.
J Cell Biochem 2006;99:1285–1297.
35. Ranera B, Lyahyai J, Romero A, Vazquez FJ, Remacha AR, Bernal ML, Zaragoza P,
Rodellar C, Martin-Burriel I. Immunophenotype and gene expression profiles of cell
surface markers of mesenchymal stem cells derived from equine bone marrow and
adipose tissue. Vet Immunol Immunopathol 2011;144:147–154.
36. Groth A, Ottinger S, Kleist C, Mohr E, Golriz M, Schultze D, Bruns H, Mehrabi A,
Schemmer P, Buchler MW, et al. Evaluation of porcine mesenchymal stem cells for
therapeutic use in human liver cancer. Int J Oncol 2012;40:391–401.
37. Jeong SH, Ji YH, Yoon ES. Immunosuppressive activity of adipose tissue-derived
mesenchymal stem cells in a rat model of hind limb allotransplantation. Transplant
Proc 2014;46:1606–1614.
38. Taha MF, Hedayati V. Isolation, identification and multipotential differentiation of
mouse adipose tissue-derived stem cells. Tissue Cell 2010;42:211–216.
39. da Silva Meirelles L, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in
virtually all post-natal organs and tissues. J Cell Sci 2006;119:2204–2213.
40. Schwartz RE, Reyes M, Koodie L, Jiang Y, Blackstad M, Lund T, Lenvik T,
Johnson S, Hu WS, Verfaillie CM. Multipotent adult progenitor cells from bone
marrow differentiate into functional hepatocyte-like cells. J Clin Invest 2002;109:
1291–1302.
41. Heidari B, Shirazi A, Akhondi MM, Hassanpour H, Behzadi B, Naderi MM, Sarvari
A, Borjian S. Comparison of proliferative and multilineage differentiation potential
of sheep mesenchymal stem cells derived from bone marrow, liver, and adipose tis-
sue. Avicenna J Med Biotechnol 2013;5:104–117.
42. Paebst F, Piehler D, Brehm W, Heller S, Schroeck C, Tarnok A, Burk J. Comparative
immunophenotyping of equine multipotent mesenchymal stromal cells: An
approach toward a standardized definition. Cytometry Part A 2014;85A:678–687.
43. Khojasteh A, Dashti SG, Dehghan MM, Behnia H, Abbasnia P, Morad G. The osteor-
egenerative effects of platelet-derived growth factor BB cotransplanted with mesen-
chymal stem cells, loaded on freeze-dried mineral bone block: A pilot study in dog
mandible. J Biomed Mater Res B Appl Biomater 2014;102:1771–1778.
44. Liu H, Wei X, Ding X, Li X, Zhou G, Li P, Fan Y. Comparison of cellular responses of
mesenchymal stem cells derived from bone marrow and synovium on combined silk
scaffolds. J Biomed Mater Res A 2015;103:115–125.
Review Article
14 Cross-species MSC
45. Lu T, Huang Y, Wang H, Ma Y, Guan W. Multi-lineage potential research of bone
marrow-derived stromal cells (BMSCs) from cattle. Appl Biochem Biotechnol 2014;
172:21–35.
46. Webb TL, Quimby JM, Dow SW. In vitro comparison of feline bone marrow-derived
and adipose tissue-derived mesenchymal stem cells. J Feline Med Surg 2012;14:
165–168.
47. Hui JH, Li L, Teo YH, Ouyang HW, Lee EH. Comparative study of the ability of mes-
enchymal stem cells derived from bone marrow, periosteum, and adipose tissue in
treatment of partial growth arrest in rabbit. Tissue Eng 2005;11:904–912.
48. Kang BJ, Lee SH, Kweon OK, Cho JY. Differentiation of canine adipose tissue-
derived mesenchymal stem cells towards endothelial progenitor cells. Am J Vet Res
2014;75:685–691.
49. Lu T, Xiong H, Wang K, Wang S, Ma Y, Guan W. Isolation and characterization of
adipose-derived mesenchymal stem cells (ADSCs) from cattle. Appl Biochem
Biotechnol 2014;174:719–728.
50. Smiler D, Soltan M, Albitar M. Toward the identification of mesenchymal stem cells
in bone marrow and peripheral blood for bone regeneration. Implant Dent 2008;17:
236–247.
51. Chong PP, Selvaratnam L, Abbas AA, Kamarul T. Human peripheral blood derived
mesenchymal stem cells demonstrate similar characteristics and chondrogenic differ-
entiation potential to bone marrow derived mesenchymal stem cells. J Orthop Res
2012;30:634–642.
52. Li S, Huang KJ, Wu JC, Hu MS, Sanyal M, Hu M, Longaker MT, Lorenz HP. Periph-
eral blood-derived mesenchymal stem cells: Candidate cells responsible for healing
critical-sized calvarial bone defects. Stem Cells Transl Med 2015;4:359–368.
53. Lyahyai J, Mediano DR, Ranera B, Sanz A, Remacha AR, Bolea R, Zaragoza P,
Rodellar C, Martin-Burriel I. Isolation and characterization of ovine mesenchymal
stem cells derived from peripheral blood. BMC Vet Res 2012;8:169.
54. Giovannini S, Brehm W, Mainil-Varlet P, Nesic D. Multilineage differentiation
potential of equine blood-derived fibroblast-like cells. Differentiation 2008;76:
118–129.
55. Dhar M, Neilsen N, Beatty K, Eaker S, Adair H, Geiser D. Equine peripheral
blood-derived mesenchymal stem cells: Isolation, identification, trilineage
differentiation and effect of hyperbaric oxygen treatment. Equine Vet J 2012;44:
600–605.
56. Wan C, He Q, Li G. Allogenic peripheral blood derived mesenchymal stem cells
(MSCs) enhance bone regeneration in rabbit ulna critical-sized bone defect model.
J Orthop Res 2006;24:610–618.
57. Sato K, Yamawaki-Ogata A, Kanemoto I, Usui A, Narita Y. Isolation and characteri-
sation of peripheral blood-derived feline mesenchymal stem cells. Vet J 2016;216:
183–188.
58. Lee KD, Kuo TK, Whang-Peng J, Chung YF, Lin CT, Chou SH, Chen JR, Chen YP,
Lee OK. In vitro hepatic differentiation of human mesenchymal stem cells. Hepatol-
ogy 2004;40:1275–1284.
59. Raoufi MF, Tajik P, Dehghan MM, Eini F, Barin A. Isolation and differentiation of
mesenchymal stem cells from bovine umbilical cord blood. Reprod Domest Anim
2011;46:95–99.
60. Li WW, Wei YH, Li H, Lai DM, Lin TN. Isolation and characterization of a novel
strain of mesenchymal stem cells from mouse umbilical cord: Potential application
in cell-based therapy. PLoS One 2013;8:e74478.
61. Seo MS, Jeong YH, Park JR, Park SB, Rho KH, Kim HS, Yu KR, Lee SH, Jung JW,
Lee YS, et al. Isolation and characterization of canine umbilical cord blood-derived
mesenchymal stem cells. J Vet Sci 2009;10:181–187.
62. Campagnoli C, Roberts IA, Kumar S, Bennett PR, Bellantuono I, Fisk NM. Identifi-
cation of mesenchymal stem/progenitor cells in human first-trimester fetal blood,
liver, and bone marrow. Blood 2001;98:2396–2402.
63. Mancanares CA, Oliveira VC, Oliveira LJ, Carvalho AF, Sampaio RV, Mancanares
AC, Souza AF, Perecin F, Meirelles FV, Miglino MA, et al. Isolation and characteriza-
tion of mesenchymal stem cells from the yolk sacs of bovine embryos. Theriogenol-
ogy 2015;84:887–898.
64. Wenceslau CV, Miglino MA, Martins DS, Ambrosio CE, Lizier NF, Pignatari GC,
Kerkis I. Mesenchymal progenitor cells from canine fetal tissues: Yolk sac, liver, and
bone marrow. Tissue Eng A 2011;17:2165–2176.
65. Garikipati VN, Jadhav S, Pal L, Prakash P, Dikshit M, Nityanand S. Mesenchymal
stem cells from fetal heart attenuate myocardial injury after infarction: An in vivo
serial pinhole gated SPECT-CT study in rats. PLoS One 2014;9:e100982.
66. In ’t Anker PS, Scherjon SA, Kleijburg-van der Keur C, Noort WA, Claas FH,
Willemze R, Fibbe WE, Kanhai HH. Amniotic fluid as a novel source of mesenchy-
mal stem cells for therapeutic transplantation. Blood 2003;102:1548–1549.
67. Filioli Uranio M, Valentini L, Lange-Consiglio A, Caira M, Guaricci AC, L’Abbate A,
Catacchio CR, Ventura M, Cremonesi F, Dell’Aquila ME. Isolation, proliferation,
cytogenetic, and molecular characterization and in vitro differentiation potency of
canine stem cells from foetal adnexa: A comparative study of amniotic fluid, amnion,
and umbilical cord matrix. Mol Reprod Dev 2011;78:361–373.
68. Weber B, Kehl D, Bleul U, Behr L, Sammut S, Frese L, Ksiazek A, Achermann J,
Stranzinger G, Robert J, et al. In vitro fabrication of autologous living tissue-
engineered vascular grafts based on prenatally harvested ovine amniotic fluid-
derived stem cells. J Tissue Eng Regen Med 2016;10:52–70.
69. Iacono E, Brunori L, Pirrone A, Pagliaro PP, Ricci F, Tazzari PL, Merlo B. Isolation,
characterization and differentiation of mesenchymal stem cells from amniotic fluid,
umbilical cord blood and Wharton’s jelly in the horse. Reproduction 2012;143:
455–468.
70. Marcus AJ, Coyne TM, Rauch J, Woodbury D, Black IB. Isolation, characterization,
and differentiation of stem cells derived from the rat amniotic membrane. Differenti-
ation 2008;76:130–144.
71. Seo MS, Park SB, Kim HS, Kang JG, Chae JS, Kang KS. Isolation and characterization
of equine amniotic membrane-derived mesenchymal stem cells. J Vet Sci 2013;14:
151–159.
72. Lange-Consiglio A, Corradetti B, Bertani S, Notarstefano V, Perrini C, Marini MG,
Arrighi S, Bosi G, Belloli A, Pravettoni D, et al. Peculiarity of porcine amniotic mem-
brane and its derived cells: A contribution to the study of cell therapy from a large
animal model. Cell Reprogram 2015;17:472–483.
73. Vellasamy S, Sandrasaigaran P, Vidyadaran S, George E, Ramasamy R. Isolation and
characterisation of mesenchymal stem cells derived from human placenta tissue.
World J Stem Cells 2012;4:53–61.
74. Futami I, Ishijima M, Kaneko H, Tsuji K, Ichikawa-Tomikawa N, Sadatsuki R,
Muneta T, Arikawa-Hirasawa E, Sekiya I, Kaneko K. Isolation and characterization
of multipotential mesenchymal cells from the mouse synovium. PLoS One 2012;7:
e45517.
75. Yoshimura H, Muneta T, Nimura A, Yokoyama A, Koga H, Sekiya I. Comparison of
rat mesenchymal stem cells derived from bone marrow, synovium, periosteum, adi-
pose tissue, and muscle. Cell Tissue Res 2007;327:449–462.
76. Prado AA, Favaron PO, da Silva LC, Baccarin RY, Miglino MA, Maria DA.
Characterization of mesenchymal stem cells derived from the equine synovial fluid
and membrane. BMC Vet Res 2015;11:281.
77. Shintani N, Hunziker EB. Chondrogenic differentiation of bovine synovium: Bone
morphogenetic proteins 2 and 7 and transforming growth factor beta1 induce the
formation of different types of cartilaginous tissue. Arthritis Rheum 2007;56:
1869–1879.
78. Klyushnenkova E, Mosca JD, Zernetkina V, Majumdar MK, Beggs KJ, Simonetti
DW, Deans RJ, McIntosh KR. T cell responses to allogeneic human mesenchymal
stem cells: Immunogenicity, tolerance, and suppression. J Biomed Sci 2005;12:
47–57.
79. Liu D, Wang Y, Ye Y, Yin G, Chen L. Distinct molecular basis for endothelial differen-
tiation: Gene expression profiles of human mesenchymal stem cells versus umbilical
vein endothelial cells. Cell Immunol 2014;289:7–14.
80. Alimperti S, You H, George T, Agarwal SK, Andreadis ST. Cadherin-11 regulates
both mesenchymal stem cell differentiation into smooth muscle cells and the devel-
opment of contractile function in vivo. J Cell Sci 2014;127:2627–2638.
81. Chang YP, Hong HP, Lee YH, Liu IH. The canine epiphyseal-derived mesenchymal
stem cells are comparable to bone marrow derived-mesenchymal stem cells. J Vet
Med Sci 2015;77:273–280.
82. O’Driscoll SW, Saris DB, Ito Y, Fitzimmons JS. The chondrogenic potential of perios-
teum decreases with age. J Orthop Res 2001;19:95–103.
83. Chang H, Docheva D, Knothe UR, Knothe Tate ML. Arthritic periosteal tissue from
joint replacement surgery: A novel, autologous source of stem cells. Stem Cells Transl
Med 2014;3:308–317.
84. Kisiel AH, McDuffee LA, Masaoud E, Bailey TR, Esparza Gonzalez BP, Nino-Fong R.
Isolation, characterization, and in vitro proliferation of canine mesenchymal stem
cells derived from bone marrow, adipose tissue, muscle, and periosteum. Am J Vet
Res 2012;73:1305–1317.
85. Rui YF, Lui PP, Li G, Fu SC, Lee YW, Chan KM. Isolation and characterization of
multipotent rat tendon-derived stem cells. Tissue Eng A 2010;16:1549–1558.
86. Feisst V, Brooks AE, Chen CJ, Dunbar PR. Characterization of mesenchymal progen-
itor cell populations directly derived from human dermis. Stem Cells Dev 2014;23:
631–642.
87. Forni MF, Ramos Maia Lobba A, Pereira Ferreira AH, Sogayar MC. Simultaneous
isolation of three different stem cell populations from murine skin. PLoS One 2015;
10:e0140143.
88. Jahroomishirazi R, Bader A, Ebert S, Schmidt C, Sedaghati B, Schulz-Siegmund M,
Zscharnack M. Isolation and characterization of CD271(1) stem cells derived from
sheep dermal skin. Cells Tissues Organs 2014;200:141–152.
89. Miura M, Gronthos S, Zhao M, Lu B, Fisher LW, Robey PG, Shi S. SHED: Stem cells
from human exfoliated deciduous teeth. Proc Natl Acad Sci USA 2003;100:5807–
5812.
90. Mensing N, Gasse H, Hambruch N, Haeger JD, Pfarrer C, Staszyk C. Isolation and
characterization of multipotent mesenchymal stromal cells from the gingiva and the
periodontal ligament of the horse. BMC Vet Res 2011;7:42.
91. Beane OS, Fonseca VC, Cooper LL, Koren G, Darling EM. Impact of aging on the
regenerative properties of bone marrow-, muscle-, and adipose-derived mesenchy-
mal stem/stromal cells. PLoS One 2014;9:e115963.
92. Krampera M, Sartoris S, Liotta F, Pasini A, Angeli R, Cosmi L, Andreini A, Mosna F,
Bonetti B, Rebellato E, et al. Immune regulation by mesenchymal stem cells derived
from adult spleen and thymus. Stem Cells Dev 2007;16:797–810.
93. Moreno R, Martinez-Gonzalez I, Rosal M, Farwati A, Gratacos E, Aran JM. Charac-
terization of mesenchymal stem cells isolated from the rabbit fetal liver. Stem Cells
Dev 2010;19:1579–1588.
94. Todorov P, Hristova E, Konakchieva R, Michova A, Dimitrov J. Comparative studies
of different cryopreservation methods for mesenchymal stem cells derived from
human fetal liver. Cell Biol Int 2010;34:455–462.
95. Lu T, Hu P, Su X, Li C, Ma Y, Guan W. Isolation and characterization of mes-
enchymal stem cells derived from fetal bovine liver. Cell Tissue Bank 2014;15:
439–450.
96. Hu Y, Liao L, Wang Q, Ma L, Ma G, Jiang X, Zhao RC. Isolation and identification
of mesenchymal stem cells from human fetal pancreas. J Lab Clin Med 2003;141:
342–349.
97. Sordi V, Malosio ML, Marchesi F, Mercalli A, Melzi R, Giordano T, Belmonte N,
Ferrari G, Leone BE, Bertuzzi F, et al. Bone marrow mesenchymal stem cells express
a restricted set of functionally active chemokine receptors capable of promoting
migration to pancreatic islets. Blood 2005;106:419–427.
Review Article
Cytometry Part A  00A: 00À00, 2017 15
98. Moniri MR, Sun XY, Rayat J, Dai D, Ao Z, He Z, Verchere CB, Dai LJ, Warnock GL.
TRAIL-engineered pancreas-derived mesenchymal stem cells: Characterization and
cytotoxic effects on pancreatic cancer cells. Cancer Gene Ther 2012;19:652–658.
99. Guo XR, Wang XL, Li MC, Yuan YH, Chen Y, Zou DD, Bian LJ, Li DS. PDX-1
mRNA-induced reprogramming of mouse pancreas-derived mesenchymal stem cells
into insulin-producing cells in vitro. Clin Exp Med 2015;15:501–509.
100. Collins JJ, Mobius MA, Thebaud B. Isolation of CD1461 Resident Lung Mesenchy-
mal Stromal Cells from Rat Lungs. J Visual Exp 2016.
101. Hu P, Pu Y, Li X, Zhu Z, Zhao Y, Guan W, Ma Y. Isolation, in vitro culture and
identification of a new type of mesenchymal stem cell derived from fetal bovine
lung tissues. Mol Med Rep 2015;12:3331–3338.
102. Eslaminejad MB, Nadri S. Murine mesenchymal stem cell isolated and expanded in
low and high density culture system: Surface antigen expression and osteogenic cul-
ture mineralization. In Vitro Cell Dev Biol Anim 2009;45:451–459.
103. Haack-Sorensen M, Hansen SK, Hansen L, Gaster M, Hyttel P, Ekblond A, Kastrup
J. Mesenchymal stromal cell phenotype is not influenced by confluence during cul-
ture expansion. Stem Cell Rev 2013;9:44–58.
104. Ren J, Wang H, Tran K, Civini S, Jin P, Castiello L, Feng J, Kuznetsov SA, Robey
PG, Sabatino M, et al. Human bone marrow stromal cell confluence: Effects on cell
characteristics and methods of assessment. Cytotherapy 2015;17:897–911.
105. Sanjurjo-Rodriguez C, Castro-Vinuelas R, Hermida-Gomez T, Fernandez-Vazquez
T, Fuentes-Boquete IM, de Toro-Santos FJ, Diaz-Prado SM, Blanco-Garcia FJ.
Ovine mesenchymal stromal cells: Morphologic, phenotypic and functional charac-
terization for osteochondral tissue engineering. PLoS One 2017;12:e0171231.
106. Zscharnack M, Hepp P, Richter R, Aigner T, Schulz R, Somerson J, Josten C, Bader
A, Marquass B. Repair of chronic osteochondral defects using predifferentiated
mesenchymal stem cells in an ovine model. Am J Sports Med 2010;38:1857–1869.
107. Adamzyk C, Emonds T, Falkenstein J, Tolba R, Jahnen-Dechent W, Lethaus B,
Neuss S. Different culture media affect proliferation, surface epitope expression,
and differentiation of ovine MSC. Stem Cells Int 2013;2013:387324.
108. Halfon S, Abramov N, Grinblat B, Ginis I. Markers distinguishing mesenchymal
stem cells from fibroblasts are downregulated with passaging. Stem Cells Dev 2011;
20:53–66.
109. Bonab MM, Alimoghaddam K, Talebian F, Ghaffari SH, Ghavamzadeh A, Nikbin B.
Aging of mesenchymal stem cell in vitro. BMC Cell Biol 2006;7:14.
110. Izadpanah R, Kaushal D, Kriedt C, Tsien F, Patel B, Dufour J, Bunnell BA.
Long-term in vitro expansion alters the biology of adult mesenchymal stem cells.
Cancer Res 2008;68:4229–4238.
111. Rosland GV, Svendsen A, Torsvik A, Sobala E, McCormack E, Immervoll H,
Mysliwietz J, Tonn JC, Goldbrunner R, Lonning PE, et al. Long-term cultures of
bone marrow-derived human mesenchymal stem cells frequently undergo sponta-
neous malignant transformation. Cancer Res 2009;69:5331–5339.
112. Miura M, Miura Y, Padilla-Nash HM, Molinolo AA, Fu B, Patel V, Seo BM,
Sonoyama W, Zheng JJ, Baker CC, et al. Accumulated chromosomal instability in
murine bone marrow mesenchymal stem cells leads to malignant transformation.
Stem Cells 2006;24:1095–1103.
113. Foudah D, Redaelli S, Donzelli E, Bentivegna A, Miloso M, Dalpra L, Tredici G.
Monitoring the genomic stability of in vitro cultured rat bone-marrow-derived
mesenchymal stem cells. Chromosome Res 2009;17:1025–1039.
114. Tamadon A, Mehrabani D, Zarezadeh Y, Rahmanifar F, Dianatpour M, Zare S.
Caprine endometrial mesenchymal stromal stem cell: Multilineage potential, char-
acterization, and growth kinetics in breeding and anestrous stages. Vet Med Int
2017;2017:5052801.
115. Le Blanc K, Ringden O. Immunomodulation by mesenchymal stem cells and clini-
cal experience. J Intern Med 2007;262:509–525.
116. Ozbey O, Sahin Z, Acar N, Ustunel I. Distribution of CD105 and CD166 positive
cells in the proximal epiphysis of developing rat humerus. Histol Histopathol 2010;
25:1437–1445.
117. Bruder SP, Horowitz MC, Mosca JD, Haynesworth SE. Monoclonal antibodies reac-
tive with human osteogenic cell surface antigens. Bone 1997;21:225–235.
118. Majumdar MK, Keane-Moore M, Buyaner D, Hardy WB, Moorman MA, McIntosh
KR, Mosca JD. Characterization and functionality of cell surface molecules on
human mesenchymal stem cells. J Biomed Sci 2003;10:228–241.
119. Wang HS, Hung SC, Peng ST, Huang CC, Wei HM, Guo YJ, Fu YS, Lai MC, Chen
CC. Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord.
Stem Cells 2004;22:1330–1337.
120. Nishikiori R, Watanabe K, Kato K. Antibody arrays for quality control of mesenchy-
mal stem cells. ACS Appl Mater Interfaces 2015;7:16828–16836.
121. Bartholomew A, Sturgeon C, Siatskas M, Ferrer K, McIntosh K, Patil S, Hardy W,
Devine S, Ucker D, Deans R, et al. Mesenchymal stem cells suppress lymphocyte
proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol 2002;30:
42–48.
122. Fazeli Z, Omrani MD, Ghaderian SM. CD29/CD184 expression analysis provides a
signature for identification of neuronal like cells differentiated from PBMSCs.
Neurosci Lett 2016;630:189–193.
123. Stock P, Bruckner S, Ebensing S, Hempel M, Dollinger MM, Christ B. The genera-
tion of hepatocytes from mesenchymal stem cells and engraftment into murine
liver. Nat Protoc 2010;5:617–627.
124. Sousa BR, Parreira RC, Fonseca EA, Amaya MJ, Tonelli FM, Lacerda SM, Lalwani P,
Santos AK, Gomes KN, Ulrich H, et al. Human adult stem cells from diverse
origins: An overview from multiparametric immunophenotyping to clinical appli-
cations. Cytometry Part A 2014;85A:43–77.
125. Soland MA, Bego M, Colletti E, Zanjani ED, St Jeor S, Porada CD, Almeida-Porada
G. Mesenchymal stem cells engineered to inhibit complement-mediated damage.
PLoS One 2013;8:e60461.
126. Delorme B, Ringe J, Gallay N, Le Vern Y, Kerboeuf D, Jorgensen C, Rosset P,
Sensebe L, Layrolle P, Haupl T, et al. Specific plasma membrane protein phenotype
of culture-amplified and native human bone marrow mesenchymal stem cells.
Blood 2008;111:2631–2635.
127. Barberini DJ, Freitas NP, Magnoni MS, Maia L, Listoni AJ, Heckler MC, Sudano
MJ, Golim MA, da Cruz Landim-Alvarenga F, Amorim RM. Equine mesenchymal
stem cells from bone marrow, adipose tissue and umbilical cord: Immunopheno-
typic characterization and differentiation potential. Stem Cell Res Ther 2014;5:25.
128. Burk J, Badylak SF, Kelly J, Brehm W. Equine cellular therapy–from stall to bench to
bedside?. Cytometry Part A 2013;83A:103–113.
129. Godoy RF, Alves AL, Gibson AJ, Lima EM, Goodship AE. Do progenitor cells from
different tissue have the same phenotype? Res Vet Sci 2014;96:454–459.
130. Tian Y, Tao L, Zhao S, Tai D, Liu D, Liu P. Isolation and morphological characteri-
zation of ovine amniotic fluid mesenchymal stem cells. Exp Anim 2016;65:125–134.
131. McCarty RC, Gronthos S, Zannettino AC, Foster BK, Xian CJ. Characterisation and
developmental potential of ovine bone marrow derived mesenchymal stem cells.
J Cell Physiol 2009;219:324–333.
132. Desantis S, Accogli G, Zizza S, Mastrodonato M, Blasi A, Francioso E, Rossi R,
Crovace A, Resta L. Ultrastructural study of cultured ovine bone marrow-derived
mesenchymal stromal cells. Ann Anat 2015;201:43–49.
133. Noort WA, Oerlemans MI, Rozemuller H, Feyen D, Jaksani S, Stecher D, Naaijkens
B, Martens AC, Buhring HJ, Doevendans PA, et al. Human versus porcine mesen-
chymal stromal cells: Phenotype, differentiation potential, immunomodulation and
cardiac improvement after transplantation. J Cell Mol Med 2012;16:1827–1839.
134. Mohsin S, Troupes CD, Starosta T, Sharp TE, Agra EJ, Smith S, Duran JM,
Zalavadia N, Zhou Y, Kubo H, et al. Unique features of cortical bone stem cells
associated with repair of the injured heart. Circ Res 2015;117:1024–1033.
135. Wang PP, Wang JH, Yan ZP, Hu MY, Lau GK, Fan ST, Luk JM. Expression of
hepatocyte-like phenotypes in bone marrow stromal cells after HGF induction. Bio-
chem Biophys Res Commun 2004;320:712–716.
136. Shichinohe H, Kuroda S, Yano S, Hida K, Iwasaki Y. Role of SDF-1/CXCR4 system
in survival and migration of bone marrow stromal cells after transplantation into
mice cerebral infarct. Brain Res 2007;1183:138–147.
137. Wynn RF, Hart CA, Corradi-Perini C, O’Neill L, Evans CA, Wraith JE, Fairbairn LJ,
Bellantuono I. A small proportion of mesenchymal stem cells strongly expresses
functionally active CXCR4 receptor capable of promoting migration to bone mar-
row. Blood 2004;104:2643–2645.
138. Ortiz LA, Gambelli F, McBride C, Gaupp D, Baddoo M, Kaminski N, Phinney DG.
Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin
exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci USA 2003;100:
8407–8411.
139. Pelekanos RA, Li J, Gongora M, Chandrakanthan V, Scown J, Suhaimi N, Brooke G,
Christensen ME, Doan T, Rice AM, et al. Comprehensive transcriptome and immu-
nophenotype analysis of renal and cardiac MSC-like populations supports strong
congruence with bone marrow MSC despite maintenance of distinct identities.
Stem Cell Res 2012;8:58–73.
140. Hegyi B, Sagi B, Kovacs J, Kiss J, Urban VS, Meszaros G, Monostori E, Uher F.
Identical, similar or different? Learning about immunomodulatory function of
mesenchymal stem cells isolated from various mouse tissues: Bone marrow, spleen,
thymus and aorta wall. Int Immunol 2010;22:551–559.
141. Lei J, Hui D, Huang W, Liao Y, Yang L, Liu L, Zhang Q, Qi G, Song W, Zhang Y,
et al. Heterogeneity of the biological properties and gene expression profiles of
murine bone marrow stromal cells. Int J Biochem Cell Biol 2013;45:2431–2443.
142. de Witte SFH, Lambert EE, Merino A, Strini T, Douben H, O’Flynn L, Elliman SJ,
de Klein A, Newsome PN, Baan CC, et al. Aging of bone marrow- and umbilical
cord-derived mesenchymal stromal cells during expansion. Cytotherapy 2017;19:
798–807.
143. Gotherstrom C. Immunomodulation by multipotent mesenchymal stromal cells.
Transplantation 2007;84:S35–S37.
144. Le Blanc K, Ringden O. Immunobiology of human mesenchymal stem cells and
future use in hematopoietic stem cell transplantation. Biol Blood Marrow Transpl
2005;11:321–334.
145. Liechty KW, MacKenzie TC, Shaaban AF, Radu A, Moseley AM, Deans R, Marshak
DR, Flake AW. Human mesenchymal stem cells engraft and demonstrate site-
specific differentiation after in utero transplantation in sheep. Nat Med 2000;6:
1282–1286.
146. Wang A, Brown EG, Lankford L, Keller BA, Pivetti CD, Sitkin NA, Beattie MS,
Bresnahan JC, Farmer DL. Placental mesenchymal stromal cells rescue ambulation
in ovine myelomeningocele. Stem Cells Transl Med 2015;4:659–669.
147. Long C, Lankford L, Kumar P, Grahn R, Borjesson DL, Farmer D, Wang A. Isolation
and characterization of canine placenta-derived mesenchymal stromal cells for the
treatment of neurological disorders in dogs. Cytometry A; 2017 Jul 17. DOI:
10.1002/cyto.a.23171. [Epub ahead of print]
148. Berglund AK, Schnabel LV. Allogeneic major histocompatibility complex-
mismatched equine bone marrow-derived mesenchymal stem cells are targeted for
death by cytotoxic anti-major histocompatibility complex antibodies. Equine Vet J
2017;49:539–544.
149. Pezzanite LM, Fortier LA, Antczak DF, Cassano JM, Brosnahan MM, Miller D,
Schnabel LV. Equine allogeneic bone marrow-derived mesenchymal stromal cells
elicit antibody responses in vivo. Stem Cell Res Ther 2015;6:54.
150. Kol A, Wood JA, Carrade Holt DD, Gillette JA, Bohannon-Worsley LK, Puchalski
SM, Walker NJ, Clark KC, Watson JL, Borjesson DL. Multiple intravenous
injections of allogeneic equine mesenchymal stem cells do not induce a systemic
inflammatory response but do alter lymphocyte subsets in healthy horses. Stem
Cell Res Ther 2015;6:73.
Review Article
16 Cross-species MSC
Mammalian MSC from Selected Species: Features and Applications Christiane Uder, Sandra Br€uckner, Sandra Winkler, Hans-Michael Tautenhahn,†‡ Bruno Christ†*
Mammalian MSC from Selected Species: Features and Applications Christiane Uder, Sandra Br€uckner, Sandra Winkler, Hans-Michael Tautenhahn,†‡ Bruno Christ†*

Mais conteúdo relacionado

Mais procurados

Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...ComprehensiveBiologi
 
Ch 21 _regeneration_of_ischemic_cardiovascular_damage_using_whartons_jelly_as...
Ch 21 _regeneration_of_ischemic_cardiovascular_damage_using_whartons_jelly_as...Ch 21 _regeneration_of_ischemic_cardiovascular_damage_using_whartons_jelly_as...
Ch 21 _regeneration_of_ischemic_cardiovascular_damage_using_whartons_jelly_as...ComprehensiveBiologi
 
Umbilical cord vs_blood_msc_source.zeddou2010_(1)
Umbilical cord vs_blood_msc_source.zeddou2010_(1)Umbilical cord vs_blood_msc_source.zeddou2010_(1)
Umbilical cord vs_blood_msc_source.zeddou2010_(1)ComprehensiveBiologi
 
Ch 24 _clinical_trials_and_family_banking_of_perinatal_stem_cells
Ch 24 _clinical_trials_and_family_banking_of_perinatal_stem_cellsCh 24 _clinical_trials_and_family_banking_of_perinatal_stem_cells
Ch 24 _clinical_trials_and_family_banking_of_perinatal_stem_cellsComprehensiveBiologi
 
Peripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordPeripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordComprehensiveBiologi
 
Excellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterialsExcellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterialsComprehensiveBiologi
 
Journal of stem cells research
Journal of stem cells researchJournal of stem cells research
Journal of stem cells researchScidoc Publishers
 
Cancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson Publishers
Cancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson PublishersCancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson Publishers
Cancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson PublishersCrimsonpublishersCancer
 
Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...
Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...
Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...raditio ghifiardi
 
Organoid culture in cancer
Organoid culture in cancerOrganoid culture in cancer
Organoid culture in cancerSamieh Asadian
 
Femoral Head Bone vs Acetabular Subchondral Bone: Selecting the Optimal Anat...
Femoral Head Bone vs Acetabular Subchondral Bone:  Selecting the Optimal Anat...Femoral Head Bone vs Acetabular Subchondral Bone:  Selecting the Optimal Anat...
Femoral Head Bone vs Acetabular Subchondral Bone: Selecting the Optimal Anat...remedypublications2
 
Exosomes - Diagnostics and Therapeutics
Exosomes - Diagnostics and TherapeuticsExosomes - Diagnostics and Therapeutics
Exosomes - Diagnostics and TherapeuticsSumedhaBobade
 

Mais procurados (20)

Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
Decellularized whartons jelly_from_human_umbilical_cord_as_a_novel_3_d_scaffo...
 
Ch 21 _regeneration_of_ischemic_cardiovascular_damage_using_whartons_jelly_as...
Ch 21 _regeneration_of_ischemic_cardiovascular_damage_using_whartons_jelly_as...Ch 21 _regeneration_of_ischemic_cardiovascular_damage_using_whartons_jelly_as...
Ch 21 _regeneration_of_ischemic_cardiovascular_damage_using_whartons_jelly_as...
 
Umbilical cord vs_blood_msc_source.zeddou2010_(1)
Umbilical cord vs_blood_msc_source.zeddou2010_(1)Umbilical cord vs_blood_msc_source.zeddou2010_(1)
Umbilical cord vs_blood_msc_source.zeddou2010_(1)
 
Whartons jelly ms_cs_a4_m
Whartons jelly ms_cs_a4_mWhartons jelly ms_cs_a4_m
Whartons jelly ms_cs_a4_m
 
Ch 24 _clinical_trials_and_family_banking_of_perinatal_stem_cells
Ch 24 _clinical_trials_and_family_banking_of_perinatal_stem_cellsCh 24 _clinical_trials_and_family_banking_of_perinatal_stem_cells
Ch 24 _clinical_trials_and_family_banking_of_perinatal_stem_cells
 
Ms cand cryopreservation
Ms cand cryopreservationMs cand cryopreservation
Ms cand cryopreservation
 
Peripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordPeripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcord
 
Application of Nanoscaffolds in Mesenchymal
Application of Nanoscaffolds in MesenchymalApplication of Nanoscaffolds in Mesenchymal
Application of Nanoscaffolds in Mesenchymal
 
Excellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterialsExcellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterials
 
Stem cells journal
Stem cells  journalStem cells  journal
Stem cells journal
 
Transplantation journal
Transplantation journalTransplantation journal
Transplantation journal
 
Journal of stem cells research
Journal of stem cells researchJournal of stem cells research
Journal of stem cells research
 
International Journal of Stem Cells & Research
International Journal of Stem Cells & ResearchInternational Journal of Stem Cells & Research
International Journal of Stem Cells & Research
 
Cancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson Publishers
Cancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson PublishersCancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson Publishers
Cancer Stem Cells and the Unicellular Life Cycle of Cancer_Crimson Publishers
 
Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...
Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...
Wharton’s jelly mesenchymal stem cells as off the-shelf cellular therapeutics...
 
Organoid culture in cancer
Organoid culture in cancerOrganoid culture in cancer
Organoid culture in cancer
 
Stem cell research
Stem cell researchStem cell research
Stem cell research
 
Femoral Head Bone vs Acetabular Subchondral Bone: Selecting the Optimal Anat...
Femoral Head Bone vs Acetabular Subchondral Bone:  Selecting the Optimal Anat...Femoral Head Bone vs Acetabular Subchondral Bone:  Selecting the Optimal Anat...
Femoral Head Bone vs Acetabular Subchondral Bone: Selecting the Optimal Anat...
 
PONE2013_VecslerM
PONE2013_VecslerMPONE2013_VecslerM
PONE2013_VecslerM
 
Exosomes - Diagnostics and Therapeutics
Exosomes - Diagnostics and TherapeuticsExosomes - Diagnostics and Therapeutics
Exosomes - Diagnostics and Therapeutics
 

Semelhante a Mammalian MSC from Selected Species: Features and Applications Christiane Uder, Sandra Br€uckner, Sandra Winkler, Hans-Michael Tautenhahn,†‡ Bruno Christ†*

A Leading Role of Stem Cells in Breast Malignant Cells
A Leading Role of Stem Cells in Breast Malignant CellsA Leading Role of Stem Cells in Breast Malignant Cells
A Leading Role of Stem Cells in Breast Malignant CellsBRNSSPublicationHubI
 
Mesenchymal stem cells in Orthopaedics
Mesenchymal stem cells in OrthopaedicsMesenchymal stem cells in Orthopaedics
Mesenchymal stem cells in OrthopaedicsDr. Bushu Harna
 
describe two recent scientific research studies and it therapies inv.pdf
describe two recent scientific research studies and it therapies inv.pdfdescribe two recent scientific research studies and it therapies inv.pdf
describe two recent scientific research studies and it therapies inv.pdffeelingcomputors
 
REVIEWCancer stem cells a new framework for the designo.docx
REVIEWCancer stem cells a new framework for the designo.docxREVIEWCancer stem cells a new framework for the designo.docx
REVIEWCancer stem cells a new framework for the designo.docxjoellemurphey
 
Fundamentals of Stem Cells
Fundamentals of Stem CellsFundamentals of Stem Cells
Fundamentals of Stem CellsDitoAnurogo1
 
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...Lipogems Equine & Lipogems Canine
 
Stem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionStem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionAnkita-rastogi
 
Stem cells for Bladder Dysfunction
Stem cells for Bladder DysfunctionStem cells for Bladder Dysfunction
Stem cells for Bladder DysfunctionAnkita-rastogi
 
Mesenchymal Stem Cells.pptx
Mesenchymal Stem Cells.pptxMesenchymal Stem Cells.pptx
Mesenchymal Stem Cells.pptxSwapnaliBehera
 
1 s2.0-s1742706114003110-main(4)
1 s2.0-s1742706114003110-main(4)1 s2.0-s1742706114003110-main(4)
1 s2.0-s1742706114003110-main(4)ayuprasiska
 
CHI's Targeting Stromal Cells in Cancer and Inflammatory Diseases Conference ...
CHI's Targeting Stromal Cells in Cancer and Inflammatory Diseases Conference ...CHI's Targeting Stromal Cells in Cancer and Inflammatory Diseases Conference ...
CHI's Targeting Stromal Cells in Cancer and Inflammatory Diseases Conference ...James Prudhomme
 
3D In Vitro Model for Drug Efficiency Testing
3D In Vitro Model for Drug Efficiency Testing3D In Vitro Model for Drug Efficiency Testing
3D In Vitro Model for Drug Efficiency Testingjudoublen
 
No adjust csc model 1
No adjust csc model 1No adjust csc model 1
No adjust csc model 1Juan Gomez
 
Minimal Criteria for Defining MSC's. The ISCT Position Statement
Minimal Criteria for Defining MSC's. The ISCT Position StatementMinimal Criteria for Defining MSC's. The ISCT Position Statement
Minimal Criteria for Defining MSC's. The ISCT Position StatementLipogems Equine & Lipogems Canine
 

Semelhante a Mammalian MSC from Selected Species: Features and Applications Christiane Uder, Sandra Br€uckner, Sandra Winkler, Hans-Michael Tautenhahn,†‡ Bruno Christ†* (20)

A Leading Role of Stem Cells in Breast Malignant Cells
A Leading Role of Stem Cells in Breast Malignant CellsA Leading Role of Stem Cells in Breast Malignant Cells
A Leading Role of Stem Cells in Breast Malignant Cells
 
Mesenchymal stem cells in Orthopaedics
Mesenchymal stem cells in OrthopaedicsMesenchymal stem cells in Orthopaedics
Mesenchymal stem cells in Orthopaedics
 
describe two recent scientific research studies and it therapies inv.pdf
describe two recent scientific research studies and it therapies inv.pdfdescribe two recent scientific research studies and it therapies inv.pdf
describe two recent scientific research studies and it therapies inv.pdf
 
Cancer stem cells Cscs
Cancer stem cells CscsCancer stem cells Cscs
Cancer stem cells Cscs
 
REVIEWCancer stem cells a new framework for the designo.docx
REVIEWCancer stem cells a new framework for the designo.docxREVIEWCancer stem cells a new framework for the designo.docx
REVIEWCancer stem cells a new framework for the designo.docx
 
Fundamentals of Stem Cells
Fundamentals of Stem CellsFundamentals of Stem Cells
Fundamentals of Stem Cells
 
Name .docx
Name                                                             .docxName                                                             .docx
Name .docx
 
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
Adipose Tissue and Mesenchymal Stem Cells: State of the Art and Lipogems® Tec...
 
Stem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionStem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder Dysfunction
 
Stem cells for Bladder Dysfunction
Stem cells for Bladder DysfunctionStem cells for Bladder Dysfunction
Stem cells for Bladder Dysfunction
 
E0351026032
E0351026032E0351026032
E0351026032
 
Mesenchymal Stem Cells.pptx
Mesenchymal Stem Cells.pptxMesenchymal Stem Cells.pptx
Mesenchymal Stem Cells.pptx
 
1 s2.0-s1742706114003110-main(4)
1 s2.0-s1742706114003110-main(4)1 s2.0-s1742706114003110-main(4)
1 s2.0-s1742706114003110-main(4)
 
CHI's Targeting Stromal Cells in Cancer and Inflammatory Diseases Conference ...
CHI's Targeting Stromal Cells in Cancer and Inflammatory Diseases Conference ...CHI's Targeting Stromal Cells in Cancer and Inflammatory Diseases Conference ...
CHI's Targeting Stromal Cells in Cancer and Inflammatory Diseases Conference ...
 
3D In Vitro Model for Drug Efficiency Testing
3D In Vitro Model for Drug Efficiency Testing3D In Vitro Model for Drug Efficiency Testing
3D In Vitro Model for Drug Efficiency Testing
 
Adult stem cells hope
Adult stem cells hopeAdult stem cells hope
Adult stem cells hope
 
Adult stem cells hope
Adult stem cells hopeAdult stem cells hope
Adult stem cells hope
 
Project: defining the endothelial hematopoietic symbiosis
Project: defining the endothelial hematopoietic symbiosisProject: defining the endothelial hematopoietic symbiosis
Project: defining the endothelial hematopoietic symbiosis
 
No adjust csc model 1
No adjust csc model 1No adjust csc model 1
No adjust csc model 1
 
Minimal Criteria for Defining MSC's. The ISCT Position Statement
Minimal Criteria for Defining MSC's. The ISCT Position StatementMinimal Criteria for Defining MSC's. The ISCT Position Statement
Minimal Criteria for Defining MSC's. The ISCT Position Statement
 

Mais de Lipogems Equine & Lipogems Canine

Intra-Articular Administration of Autologous Micro-Fragmented Adipose Tissue ...
Intra-Articular Administration of Autologous Micro-Fragmented Adipose Tissue ...Intra-Articular Administration of Autologous Micro-Fragmented Adipose Tissue ...
Intra-Articular Administration of Autologous Micro-Fragmented Adipose Tissue ...Lipogems Equine & Lipogems Canine
 
Lipogems regenerative medicine seminar by lipocast biotech uk itinerary for s...
Lipogems regenerative medicine seminar by lipocast biotech uk itinerary for s...Lipogems regenerative medicine seminar by lipocast biotech uk itinerary for s...
Lipogems regenerative medicine seminar by lipocast biotech uk itinerary for s...Lipogems Equine & Lipogems Canine
 
Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2 - With...
Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2 - With...Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2 - With...
Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2 - With...Lipogems Equine & Lipogems Canine
 
Lipocast Biotech UK Lipogems Canine Standard Operating Procedure v1
Lipocast Biotech UK Lipogems Canine Standard Operating Procedure v1Lipocast Biotech UK Lipogems Canine Standard Operating Procedure v1
Lipocast Biotech UK Lipogems Canine Standard Operating Procedure v1Lipogems Equine & Lipogems Canine
 
Lipocast Biotech UK Lipogems Equine Standard Operating Procedure 007 - withou...
Lipocast Biotech UK Lipogems Equine Standard Operating Procedure 007 - withou...Lipocast Biotech UK Lipogems Equine Standard Operating Procedure 007 - withou...
Lipocast Biotech UK Lipogems Equine Standard Operating Procedure 007 - withou...Lipogems Equine & Lipogems Canine
 
The Effect of Intra-articular Injection of Autologous Microfragmented Fat Tis...
The Effect of Intra-articular Injection of Autologous Microfragmented Fat Tis...The Effect of Intra-articular Injection of Autologous Microfragmented Fat Tis...
The Effect of Intra-articular Injection of Autologous Microfragmented Fat Tis...Lipogems Equine & Lipogems Canine
 
Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue an...
Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue an...Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue an...
Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue an...Lipogems Equine & Lipogems Canine
 
Lipocast Biotech UK Lipogems Canine Veterinary Presentation 2017 v.1.3 - With...
Lipocast Biotech UK Lipogems Canine Veterinary Presentation 2017 v.1.3 - With...Lipocast Biotech UK Lipogems Canine Veterinary Presentation 2017 v.1.3 - With...
Lipocast Biotech UK Lipogems Canine Veterinary Presentation 2017 v.1.3 - With...Lipogems Equine & Lipogems Canine
 
Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2
Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2 Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2
Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2 Lipogems Equine & Lipogems Canine
 
Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...
Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...
Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...Lipogems Equine & Lipogems Canine
 
Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...
Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...
Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...Lipogems Equine & Lipogems Canine
 
Veterinary Record - Lipogems story - Regenerative therapy for canine joint co...
Veterinary Record - Lipogems story - Regenerative therapy for canine joint co...Veterinary Record - Lipogems story - Regenerative therapy for canine joint co...
Veterinary Record - Lipogems story - Regenerative therapy for canine joint co...Lipogems Equine & Lipogems Canine
 
Vet Times 2017 Lipogems Canine Feature - Novel regeneration therapy available...
Vet Times 2017 Lipogems Canine Feature - Novel regeneration therapy available...Vet Times 2017 Lipogems Canine Feature - Novel regeneration therapy available...
Vet Times 2017 Lipogems Canine Feature - Novel regeneration therapy available...Lipogems Equine & Lipogems Canine
 

Mais de Lipogems Equine & Lipogems Canine (20)

Equestrian Life June 2019
Equestrian Life June 2019 Equestrian Life June 2019
Equestrian Life June 2019
 
A Selection of Lipogems Canine Case Studies v1.
A Selection of Lipogems Canine Case Studies v1.A Selection of Lipogems Canine Case Studies v1.
A Selection of Lipogems Canine Case Studies v1.
 
Intra-Articular Administration of Autologous Micro-Fragmented Adipose Tissue ...
Intra-Articular Administration of Autologous Micro-Fragmented Adipose Tissue ...Intra-Articular Administration of Autologous Micro-Fragmented Adipose Tissue ...
Intra-Articular Administration of Autologous Micro-Fragmented Adipose Tissue ...
 
Lipocast bitoech uk privacy and data protection policy
Lipocast bitoech uk privacy and data protection policyLipocast bitoech uk privacy and data protection policy
Lipocast bitoech uk privacy and data protection policy
 
Lipogems regenerative medicine seminar by lipocast biotech uk itinerary for s...
Lipogems regenerative medicine seminar by lipocast biotech uk itinerary for s...Lipogems regenerative medicine seminar by lipocast biotech uk itinerary for s...
Lipogems regenerative medicine seminar by lipocast biotech uk itinerary for s...
 
Vet Times February Issue 2018
Vet Times February Issue 2018Vet Times February Issue 2018
Vet Times February Issue 2018
 
Lipogems Equine Featured in Polo Times February 2018 Issue
Lipogems Equine Featured in Polo Times February 2018 IssueLipogems Equine Featured in Polo Times February 2018 Issue
Lipogems Equine Featured in Polo Times February 2018 Issue
 
Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2 - With...
Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2 - With...Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2 - With...
Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2 - With...
 
Lipocast Biotech UK Lipogems Canine Standard Operating Procedure v1
Lipocast Biotech UK Lipogems Canine Standard Operating Procedure v1Lipocast Biotech UK Lipogems Canine Standard Operating Procedure v1
Lipocast Biotech UK Lipogems Canine Standard Operating Procedure v1
 
Lipocast Biotech UK Lipogems Equine Standard Operating Procedure 007 - withou...
Lipocast Biotech UK Lipogems Equine Standard Operating Procedure 007 - withou...Lipocast Biotech UK Lipogems Equine Standard Operating Procedure 007 - withou...
Lipocast Biotech UK Lipogems Equine Standard Operating Procedure 007 - withou...
 
The Effect of Intra-articular Injection of Autologous Microfragmented Fat Tis...
The Effect of Intra-articular Injection of Autologous Microfragmented Fat Tis...The Effect of Intra-articular Injection of Autologous Microfragmented Fat Tis...
The Effect of Intra-articular Injection of Autologous Microfragmented Fat Tis...
 
Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue an...
Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue an...Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue an...
Angiogenic and anti-inflammatory properties of micro-fragmented fat tissue an...
 
IFATS Presentation by Dr offer Zeira - 2016 20.11.16
IFATS Presentation by Dr offer Zeira - 2016 20.11.16IFATS Presentation by Dr offer Zeira - 2016 20.11.16
IFATS Presentation by Dr offer Zeira - 2016 20.11.16
 
Lipocast Biotech UK Lipogems Canine Veterinary Presentation 2017 v.1.3 - With...
Lipocast Biotech UK Lipogems Canine Veterinary Presentation 2017 v.1.3 - With...Lipocast Biotech UK Lipogems Canine Veterinary Presentation 2017 v.1.3 - With...
Lipocast Biotech UK Lipogems Canine Veterinary Presentation 2017 v.1.3 - With...
 
Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2
Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2 Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2
Lipocast Biotech UK Lipogems Equine and Canine Presentation 2017 v.1.2
 
Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...
Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...
Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...
 
Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...
Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...
Lipocast Biotech UK Lipogems Equine Veterinary Presentation 2017 v.1.4 - With...
 
Veterinary Record - Lipogems story - Regenerative therapy for canine joint co...
Veterinary Record - Lipogems story - Regenerative therapy for canine joint co...Veterinary Record - Lipogems story - Regenerative therapy for canine joint co...
Veterinary Record - Lipogems story - Regenerative therapy for canine joint co...
 
Vet Times 2017 Lipogems Canine Feature - Novel regeneration therapy available...
Vet Times 2017 Lipogems Canine Feature - Novel regeneration therapy available...Vet Times 2017 Lipogems Canine Feature - Novel regeneration therapy available...
Vet Times 2017 Lipogems Canine Feature - Novel regeneration therapy available...
 
Vet Practice News January 2017 - News Story
Vet Practice News January 2017  - News StoryVet Practice News January 2017  - News Story
Vet Practice News January 2017 - News Story
 

Último

Reboulia: features, anatomy, morphology etc.
Reboulia: features, anatomy, morphology etc.Reboulia: features, anatomy, morphology etc.
Reboulia: features, anatomy, morphology etc.Silpa
 
biology HL practice questions IB BIOLOGY
biology HL practice questions IB BIOLOGYbiology HL practice questions IB BIOLOGY
biology HL practice questions IB BIOLOGY1301aanya
 
Grade 7 - Lesson 1 - Microscope and Its Functions
Grade 7 - Lesson 1 - Microscope and Its FunctionsGrade 7 - Lesson 1 - Microscope and Its Functions
Grade 7 - Lesson 1 - Microscope and Its FunctionsOrtegaSyrineMay
 
CYTOGENETIC MAP................ ppt.pptx
CYTOGENETIC MAP................ ppt.pptxCYTOGENETIC MAP................ ppt.pptx
CYTOGENETIC MAP................ ppt.pptxSilpa
 
Proteomics: types, protein profiling steps etc.
Proteomics: types, protein profiling steps etc.Proteomics: types, protein profiling steps etc.
Proteomics: types, protein profiling steps etc.Silpa
 
Atp synthase , Atp synthase complex 1 to 4.
Atp synthase , Atp synthase complex 1 to 4.Atp synthase , Atp synthase complex 1 to 4.
Atp synthase , Atp synthase complex 1 to 4.Silpa
 
(May 9, 2024) Enhanced Ultrafast Vector Flow Imaging (VFI) Using Multi-Angle ...
(May 9, 2024) Enhanced Ultrafast Vector Flow Imaging (VFI) Using Multi-Angle ...(May 9, 2024) Enhanced Ultrafast Vector Flow Imaging (VFI) Using Multi-Angle ...
(May 9, 2024) Enhanced Ultrafast Vector Flow Imaging (VFI) Using Multi-Angle ...Scintica Instrumentation
 
Dr. E. Muralinath_ Blood indices_clinical aspects
Dr. E. Muralinath_ Blood indices_clinical  aspectsDr. E. Muralinath_ Blood indices_clinical  aspects
Dr. E. Muralinath_ Blood indices_clinical aspectsmuralinath2
 
Human genetics..........................pptx
Human genetics..........................pptxHuman genetics..........................pptx
Human genetics..........................pptxSilpa
 
Asymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 b
Asymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 bAsymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 b
Asymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 bSérgio Sacani
 
Climate Change Impacts on Terrestrial and Aquatic Ecosystems.pptx
Climate Change Impacts on Terrestrial and Aquatic Ecosystems.pptxClimate Change Impacts on Terrestrial and Aquatic Ecosystems.pptx
Climate Change Impacts on Terrestrial and Aquatic Ecosystems.pptxDiariAli
 
Biogenic Sulfur Gases as Biosignatures on Temperate Sub-Neptune Waterworlds
Biogenic Sulfur Gases as Biosignatures on Temperate Sub-Neptune WaterworldsBiogenic Sulfur Gases as Biosignatures on Temperate Sub-Neptune Waterworlds
Biogenic Sulfur Gases as Biosignatures on Temperate Sub-Neptune WaterworldsSérgio Sacani
 
PSYCHOSOCIAL NEEDS. in nursing II sem pptx
PSYCHOSOCIAL NEEDS. in nursing II sem pptxPSYCHOSOCIAL NEEDS. in nursing II sem pptx
PSYCHOSOCIAL NEEDS. in nursing II sem pptxSuji236384
 
Genome sequencing,shotgun sequencing.pptx
Genome sequencing,shotgun sequencing.pptxGenome sequencing,shotgun sequencing.pptx
Genome sequencing,shotgun sequencing.pptxSilpa
 
Cyanide resistant respiration pathway.pptx
Cyanide resistant respiration pathway.pptxCyanide resistant respiration pathway.pptx
Cyanide resistant respiration pathway.pptxSilpa
 
Molecular markers- RFLP, RAPD, AFLP, SNP etc.
Molecular markers- RFLP, RAPD, AFLP, SNP etc.Molecular markers- RFLP, RAPD, AFLP, SNP etc.
Molecular markers- RFLP, RAPD, AFLP, SNP etc.Silpa
 
TransientOffsetin14CAftertheCarringtonEventRecordedbyPolarTreeRings
TransientOffsetin14CAftertheCarringtonEventRecordedbyPolarTreeRingsTransientOffsetin14CAftertheCarringtonEventRecordedbyPolarTreeRings
TransientOffsetin14CAftertheCarringtonEventRecordedbyPolarTreeRingsSérgio Sacani
 
Zoology 5th semester notes( Sumit_yadav).pdf
Zoology 5th semester notes( Sumit_yadav).pdfZoology 5th semester notes( Sumit_yadav).pdf
Zoology 5th semester notes( Sumit_yadav).pdfSumit Kumar yadav
 
Call Girls Ahmedabad +917728919243 call me Independent Escort Service
Call Girls Ahmedabad +917728919243 call me Independent Escort ServiceCall Girls Ahmedabad +917728919243 call me Independent Escort Service
Call Girls Ahmedabad +917728919243 call me Independent Escort Serviceshivanisharma5244
 

Último (20)

Reboulia: features, anatomy, morphology etc.
Reboulia: features, anatomy, morphology etc.Reboulia: features, anatomy, morphology etc.
Reboulia: features, anatomy, morphology etc.
 
biology HL practice questions IB BIOLOGY
biology HL practice questions IB BIOLOGYbiology HL practice questions IB BIOLOGY
biology HL practice questions IB BIOLOGY
 
Site Acceptance Test .
Site Acceptance Test                    .Site Acceptance Test                    .
Site Acceptance Test .
 
Grade 7 - Lesson 1 - Microscope and Its Functions
Grade 7 - Lesson 1 - Microscope and Its FunctionsGrade 7 - Lesson 1 - Microscope and Its Functions
Grade 7 - Lesson 1 - Microscope and Its Functions
 
CYTOGENETIC MAP................ ppt.pptx
CYTOGENETIC MAP................ ppt.pptxCYTOGENETIC MAP................ ppt.pptx
CYTOGENETIC MAP................ ppt.pptx
 
Proteomics: types, protein profiling steps etc.
Proteomics: types, protein profiling steps etc.Proteomics: types, protein profiling steps etc.
Proteomics: types, protein profiling steps etc.
 
Atp synthase , Atp synthase complex 1 to 4.
Atp synthase , Atp synthase complex 1 to 4.Atp synthase , Atp synthase complex 1 to 4.
Atp synthase , Atp synthase complex 1 to 4.
 
(May 9, 2024) Enhanced Ultrafast Vector Flow Imaging (VFI) Using Multi-Angle ...
(May 9, 2024) Enhanced Ultrafast Vector Flow Imaging (VFI) Using Multi-Angle ...(May 9, 2024) Enhanced Ultrafast Vector Flow Imaging (VFI) Using Multi-Angle ...
(May 9, 2024) Enhanced Ultrafast Vector Flow Imaging (VFI) Using Multi-Angle ...
 
Dr. E. Muralinath_ Blood indices_clinical aspects
Dr. E. Muralinath_ Blood indices_clinical  aspectsDr. E. Muralinath_ Blood indices_clinical  aspects
Dr. E. Muralinath_ Blood indices_clinical aspects
 
Human genetics..........................pptx
Human genetics..........................pptxHuman genetics..........................pptx
Human genetics..........................pptx
 
Asymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 b
Asymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 bAsymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 b
Asymmetry in the atmosphere of the ultra-hot Jupiter WASP-76 b
 
Climate Change Impacts on Terrestrial and Aquatic Ecosystems.pptx
Climate Change Impacts on Terrestrial and Aquatic Ecosystems.pptxClimate Change Impacts on Terrestrial and Aquatic Ecosystems.pptx
Climate Change Impacts on Terrestrial and Aquatic Ecosystems.pptx
 
Biogenic Sulfur Gases as Biosignatures on Temperate Sub-Neptune Waterworlds
Biogenic Sulfur Gases as Biosignatures on Temperate Sub-Neptune WaterworldsBiogenic Sulfur Gases as Biosignatures on Temperate Sub-Neptune Waterworlds
Biogenic Sulfur Gases as Biosignatures on Temperate Sub-Neptune Waterworlds
 
PSYCHOSOCIAL NEEDS. in nursing II sem pptx
PSYCHOSOCIAL NEEDS. in nursing II sem pptxPSYCHOSOCIAL NEEDS. in nursing II sem pptx
PSYCHOSOCIAL NEEDS. in nursing II sem pptx
 
Genome sequencing,shotgun sequencing.pptx
Genome sequencing,shotgun sequencing.pptxGenome sequencing,shotgun sequencing.pptx
Genome sequencing,shotgun sequencing.pptx
 
Cyanide resistant respiration pathway.pptx
Cyanide resistant respiration pathway.pptxCyanide resistant respiration pathway.pptx
Cyanide resistant respiration pathway.pptx
 
Molecular markers- RFLP, RAPD, AFLP, SNP etc.
Molecular markers- RFLP, RAPD, AFLP, SNP etc.Molecular markers- RFLP, RAPD, AFLP, SNP etc.
Molecular markers- RFLP, RAPD, AFLP, SNP etc.
 
TransientOffsetin14CAftertheCarringtonEventRecordedbyPolarTreeRings
TransientOffsetin14CAftertheCarringtonEventRecordedbyPolarTreeRingsTransientOffsetin14CAftertheCarringtonEventRecordedbyPolarTreeRings
TransientOffsetin14CAftertheCarringtonEventRecordedbyPolarTreeRings
 
Zoology 5th semester notes( Sumit_yadav).pdf
Zoology 5th semester notes( Sumit_yadav).pdfZoology 5th semester notes( Sumit_yadav).pdf
Zoology 5th semester notes( Sumit_yadav).pdf
 
Call Girls Ahmedabad +917728919243 call me Independent Escort Service
Call Girls Ahmedabad +917728919243 call me Independent Escort ServiceCall Girls Ahmedabad +917728919243 call me Independent Escort Service
Call Girls Ahmedabad +917728919243 call me Independent Escort Service
 

Mammalian MSC from Selected Species: Features and Applications Christiane Uder, Sandra Br€uckner, Sandra Winkler, Hans-Michael Tautenhahn,†‡ Bruno Christ†*

  • 1. Mammalian MSC from Selected Species: Features and Applications Christiane Uder, Sandra Br€uckner, Sandra Winkler, Hans-Michael Tautenhahn,†‡ Bruno Christ† * Abstract Mesenchymal stromal/stem cells (MSC) are promising candidates for cellular therapy of different diseases in humans and in animals. Following the guidelines of the Interna- tional Society for Cell Therapy, human MSC may be identified by expression of a spe- cific panel of cell surface markers (CD1051, CD731, CD901, CD34-, CD14-, or CD11b-, CD79- or CD19-, HLA-DR-). In addition, multiple differentiation potential into at least the osteogenic, adipogenic, and chondrogenic lineage is a main criterion for MSC definition. Human MSC and MSC of a variety of mammals isolated from dif- ferent tissues meet these criteria. In addition to the abovementioned, they express many more cell surface markers. Yet, these are not uniquely expressed by MSC. The gross phenotypic appearance like marker expression and differentiation potential is similar albeit not identical for MSC from different tissues and species. Similarly, MSC may feature different biological characteristics depending on the tissue source and the isolation and culture procedures. Their versatile biological qualities comprising immu- nomodulatory, anti-inflammatory, and proregenerative capacities rely largely on the migratory and secretory capabilities of MSC. They are attracted to sites of tissue lesion and secrete factors to promote self-repair of the injured tissue. This is a big perspective for clinical MSC applications in both veterinary and human medicine. Phase I/II clini- cal trials have been initiated to assess safety and feasibility of MSC therapies in acute and chronic disease settings. Yet, since the mode of MSC action in a specific disease environment is still unknown at large, it is mandatory to unravel the response of MSC from a given source onto a specific disease environment in suitable animal models prior to clinical applications. VC 2017 International Society for Advancement of Cytometry Key terms cell therapy; clinical trials; differentiation; mammals; mesenchymal stem cells; surface marker BASED on his experiments with frogs, the German pathologist Cohnheim (1839–1884) already hypothesized that cells especially from bone marrow may migrate and repair tissue damage after injury. His observations were substantiated by the work of Friedenstein et al. nearly 100 years later. They demonstrated that bone marrow cells and their descendants of non-hematopoietic origin may differentiate in vitro into cells of other tissues of mesenchymal origin (1). They termed these cells “bone marrow fibroblasts,” which were renamed in the following years by various authors into bone marrow stromal cells, mesenchymal stem cells, or skeletal stem cells (2). The International Society for Cell Therapy published a position article addressing the usage of the term “mesenchymal stem cell” (MSC). According to this statement, it is recommended strictly to discriminate between “multipotent mesen- chymal stromal cells” and “mesenchymal stem cells.” The first term was suggested to be used for fibroblast-like plastic adherent cells irrespective of their tissue origin. The latter designated stem cells, which meet particular stem cell definition criteria (3). Today, the term “mesenchymal stromal cell” is often synonymously used with “mesenchymal stem cell” or even preferred due to the fact that many cell Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany *Correspondence to: Bruno Christ, PhD, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Lab, University Hospital Leipzig, Liebigstraße 21, D-04103 Leipzig, Germany. Email: bruno.christ@ medizin.uni-leipzig.de † Equal contributions as senior authors. ‡ Present address: Division of General, Visceral and Vascular Surgery, University Hospital of Jena, Am Klinikum 1, Jena 07747, Germany Published online 00 Month 2017 in Wiley Online Library (wileyonlinelibrary.com) DOI: 10.1002/cyto.a.23239 VC 2017 International Society for Advancement of Cytometry Cytometry Part A 00A: 00À00, 2017 Review Article
  • 2. preparations do not necessarily feature true stem cell charac- teristics like self-renewal and differentiation potential. Mini- mal criteria have been postulated by the International Society of Cell Therapy to define human MSC unequivocally. The first criterion to be met by MSC is their ability to adhere to plastic surfaces under standardized culture conditions. Second, a minimum percentage of cells must feature a specified surface marker profile, that is, 95% of the cells must express CD105, CD73, and CD90, but 2% may express CD45, CD34, CD14 or CD11b, CD79 or CD19 and HLA-DR (4). Finally, MSC must be able to differentiate at least into three different line- ages like osteoblasts, adipocytes, and chondrocytes in vitro. However, since these criteria do not take into account the vast variety of MSC preparations from different sources and spe- cies as well as the different isolation and culture conditions yielding cells with different characteristics, it has been sug- gested that a new description of MSC is necessary. Accord- ingly, the term “mesenchymal stromal cell” should be used for the general population of MSC, which is mostly a heteroge- neous mix of different cell populations. The term “mesenchymal stem cell” should be restricted to a cell popula- tion featuring stem cell characteristics like self-renewal and differentiation potential. In addition, description of cells should include species, tissue resource, the information on whether cells are primary or cultured, and culture passage numbers besides others (5). Making up only 0.001 to 0.01% of a tissue mass, the quantity of MSC in tissues is rare, their morphology fibroblast-like and spindle-shaped (2,6,7). In respect of their growth characteristics, Conget et al. observed a doubling time of 33 h as well as an immense expansion potential (8). Beyond these specific cell cycle characteristics, the isolated cell population contained a subgroup of about 20% resting cells, presumably needed to replenish the stem cell pool after growth stimulation (7). The therapeutic use of MSC has been addressed in animal models in a variety of applications comprising in general the local implantation for regional tissue repair, systemic transplan- tation for diseases affecting the entire organism, stem cell ther- apy combined with gene therapy, and the generation/ development of engineered tissues (2). Clinically, MSC have been established in clinical phase I/II trials to treat a wide range of diseases comprising cardiovascular repair, therapy of lung fibrosis and spinal cord injury as well as bone and cartilage replacement besides others. In this context, it has been empha- sized that the clinical use of MSC warrants extensive characteri- zation of the cell product comprising identity, purity, safety, and biological feasibility. Tissue-based cytometry and/or flow cytometry were proposed as methodology of choice to unequiv- ocally identify MSC and their subpopulations (9) (Fig. 1). There is an overwhelming literature available dealing with MSC from a great variety of organisms and their organs and tissues including humans. It is nearly impossible to completely review this vast magnitude of studies and to compare their outcomes to each other. Therefore, in this review we focus on humans, selected large animals as well as rodents and try to give a more generalized overview on the organ and tissue resources of MSC and their biological features in the context of the mode of MSC actions and potential use of MSC in clinical applications. Figure 1. Flow chart of current MSC therapeutic applications. In principle, the therapeutic use of MSC in humans or animals follows the same rational. Cells are isolated most commonly from either bone marrow or adipose tissue because of the versatile availability. Cells must be characterized unequivocally in terms of their phenotypic features and biological actions before delivery into patients, either human or animal. Since MSC are classified Advanced Therapy Medicinal Products (ATMP), their safety, feasibility and therapeutic poten- tial must be confirmed in humans by clinical trials according to regulatory guidelines. [Color figure can be viewed at wileyonlinelibrary. com] Review Article 2 Cross-species MSC
  • 3. MSC AVAILABILITY AND MAINTENANCE It seems that MSC are present in virtually all kinds of tis- sues, organs, and organisms investigated so far. The methods of isolation rely on the enrichment from the mononuclear cell fraction by a number of different methods described. Because of the ease of access and sufficient availability, the most prom- inent sources for the isolation of MSC are bone marrow and adipose tissue, which has been described in hominids, large, and small animals. In the following chapter, both prominent and some rare sources will be outlined representatively. Tissue Sources Principal steps in the isolation procedure to gain MSC are similar in studies across species. In humans, large and small animals, the MSC isolation procedure comprised tissue digestion by collagenase, removal of debris by filtration of the cell suspension, and density gradient centrifugation to obtain the mononuclear cell fraction. Mononuclear cells were then cultured on plastic dishes in simple Modified Eagle’s Medium containing fetal calf serum as appropriate. After 24 h, floating cells were removed to enrich for the plastic-adherent MSC. With culture progressing, cells proliferated and grew to con- fluency after a period in time (10–16). This method, mostly used for isolation of MSC from soft tissues, is modified for the isolation of MSC from human and small and large animal bone marrow. In pigs, MSC were isolated from the substantia spongiosa of the os femoris, which was scraped off and digested by collagenase (17). After the subsequent density gradient cen- trifugation to gain the mononuclear cell fraction, cells were seeded onto plastic dishes and the MSC enriched following protocols according to those described for the isolation of MSC from soft tissues (9). Similar protocols have also been used for the isolation of MSC from human bone marrow (7,18–20). Bone marrow waste material is available from elec- tive knee and hip surgery or major amputations and moreover could be gained from voluntary crista iliaca punctures. Isola- tion procedures are similar as compared to methods from other species like dog (21,22), horse (23), or sheep (24) and aim at enrichment of MSC from the mononuclear cell frac- tion. In rodents, the isolation procedure is modified, taking into account the low amount of bone marrow available at all. Generally, mouse and rat bone marrow was completely flushed out of the femur and tibia with an appropriate medium and density gradient centrifugation was either per- formed to enrich for mononuclear cells (25), or was omitted for use of whole bone marrow cell populations (26). Methods applying magnetic bead technology have been established for example for positive selection using CD49a as a capture anti- gen for the isolation of human MSC (27), or for negative selection using CD45/glycophorin A to remove hematopoietic cells from the mononuclear cell fraction derived from human bone marrow (28), just to mention two examples out of a plenty of methods describing positive or negative selection of MSC from different sources and species. A previous review highlighted the need for unequivocal characterization of MSC from various sources with the goal to identify subpopulations of cells especially in preparations from adipose tissue sources, which apart from MSC contained endothelial progenitors, pericytes, and others. This is relevant, since flow cytometry analyses revealed different marker pro- files on subpopulations of MSC and on MSC from different sources, which might correlate to different functional biologi- cal features of the cells (29). Accordingly, cross-species com- parisons of MSC corroborated that markers and functions of MSC might be similar between different sources and species, but they are not identical. Human adipose tissue is collected as waste material from elective abdominal surgery from patients of different age and gender, and thus represents a source providing MSC from different biological backgrounds. While the gross biological features of MSC from adipose tissue like expression of surface markers, morphology and differenti- ation potential was similar to MSC from other sources, vari- ability was detected in terms of isolation yield, proliferation rates, and expandability (10,30–33). In line, adipose tissue- derived MSC from non-human primates (rhesus monkey) (34) and from horse subcutaneous adipose tissue adjacent to the dorsal gluteal muscle (35) showed similar biological fea- tures to human MSC from different sources in principle, but varied in specific details like expression of distinct surface markers, or doubling times and number of population dou- blings during passaging. Porcine MSC from subcutaneous and visceral adipose tissue with similar biological features were explored in order to establish a large animal model of MSC transplantation to investigate proof-of-concept as well as safety aspects, relevant regulatory issues on the way to clinical translation (11,36). In most experimental settings in rodents (rat, mouse), MSC were isolated from bone marrow or adi- pose tissue (37,38,123). Again, principal biological features like morphology, multi-lineage differentiation potential, and surface marker expression were similar, but not identical to characteristics of MSC from other species and organs. Com- paring characteristics of MSC from different species, tissues and organs remains incomplete, since isolation procedures and culture conditions varied between studies, which surely contributes to differences in biological features and functions of MSC as addressed below. However, it has been shown that nearly all post-natal organs and tissues in the mouse harbored MSC with similar features indicating a common precursor or origin. Indeed, since MSC cultures could be grown from large and small blood vessels all over the body, it was suggested that MSC might reside in common perivascular niches in the dif- ferent organs (39). A comprehensive overview on different sources of MSC from hominids as well as large and rodent mammals is given in Table 1. In Vitro Expansion It must be anticipated that culturing MSC changes their biological features as discussed above. However, one promi- nent quality of stem cells is their self-renewal potential, which allows for the propagation of the MSC in vitro. This is of utmost importance, since clinical application of MSC requires cell numbers far higher than those originally obtained in a na€ıve state from isolation procedures described in the Review Article Cytometry Part A 00A: 00À00, 2017 3
  • 4. previous paragraph. Therefore, in vitro expansion of MSC was extensively studied and revealed inconsistency concerning the impact of isolation, culture and expansion procedures on the biological characteristics of MSC. On the one hand, the multi- ple differentiation potential of MSC has been documented both in vitro and in vivo in various tissues confirming their consistent functional performance even after extensive manip- ulation by rigid isolation and cultivation procedures. This was shown by applying various culture conditions to human bone marrow-derived MSC (20): irrespective of whether the serum- free culture medium was supplemented with basic fibroblast growth factor (bFGF), with 2% fetal bovine serum (FBS) together with epidermal growth factor and platelet-derived growth factor (PDGF), or with 10% FBS, proliferation rates or expression of surface molecules were not affected. From these findings it was inferred that biological functions of MSC were largely independent of and resistant to the method of isolation and cultivation (102,103). In contrast, confluent growth of human bone marrow-derived MSC changed the expression of several cell surface markers and proteins secreted into the culture supernatants, but did not impact on immu- nomodulatory features (104). Because of the high clinical rele- vance, these studies on the impact of culture conditions on MSC characteristics have mostly been performed with human MSC, and to a less extent with MSC from other species. Sheep are often used as pre-clinical large animal models to test the therapeutic potential of MSC or MSC engineered by scaffold- ing for the treatment of degenerative joint diseases (105,106). In this setting, ovine MSC changed their proliferative and dif- ferentiation behavior as well as surface epitope expression depending on growth factors or serum present in the culture media (107). Similarly, the differentiation capabilities of MSC decreased at later passages (40–50 population doublings) (108) suggesting that frequent iterated passaging resulted in aging and loss of stemness. Unrestricted differentiation poten- tial of human MSC was confined up to the 6th passage. Beyond this passage number, the loss of stem cell features coincided with the decrease of the mean telomere length from 9.19 kb to 8.7 kb at the 9th passage (109). Not human, but MSC derived from rhesus monkey developed a distinct popu- lation of polyploid cells at passage 20, which progressed to aneuploidy at passage 30 (110). In addition, chromosomal instability arising at higher passage numbers was described across several species including human (111), mouse (112), and rat (113). In contrast, no chromosomal instability was observed in endometrium-derived goat MSC up to passage Table 1. Overview of the different tissue sources for MSC isolation in mammals (selected publications); 1 published HOMINIDS LARGE ANIMALS SMALL ANIMALS TISSUE REFERENCES HUMAN RHESUS MONKEY HORSE SHEEP PIG CATTLE DOG CAT RABBIT RAT MOUSE Bone marrow (17,18,34,39–46) 1 1 1 1 1 1 1 1 1 1 1 Adipose tissue (10,11,34,35,37,38, 41,42,46–49) 1 1 1 1 1 1 1 1 1 1 1 Peripheral blood (50–57) 1 1 1 1 1 1 Umbilical cord blood (42,58–61) 1 1 1 1 1 Embryonic tissue (62–64) 1 1 1 Fetal blood (62) 1 Fetal heart (65) 1 Amnion fluid (66–69) 1 1 1 1 Amniotic membrane (67,70–72) 1 1 1 1 Placenta (73) 1 Synovia (44,74–77) 1 1 1 1 1 1 Trabecular bone (78–81) 1 1 Periosteum (75,82–84) 1 1 1 1 Tendon (42,85) 1 1 1 Dermis (86–88) 1 1 1 Teeth (89,90) 1 1 Muscle (39,75,84,91) 1 1 1 1 Brain (39) 1 Thymus (39,92) 1 1 Kidney (39) 1 Liver (39,41) 1 1 Fetal liver (64,93–95) 1 1 1 1 Fetal pancreas (96) 1 Pancreas (97–99) 1 1 1 Spleen (39,92) 1 1 Lung (39,100,101) 1 1 Review Article 4 Cross-species MSC
  • 5. number 60 (114). In summary, the basic features of MSC from different species comprising multi-lineage differentiation, sur- face marker expression besides others seem to be very similar, though not identical, at low passage numbers. Expansion beyond changes characteristics mainly associated with general signs of ageing. PHENOTYPIC AND FUNCTIONAL FEATURES Surface Markers The list of surface antigens detected on MSC has grown enormously in recent years. Surface markers expressed on MSC across all species considered here are CD29 and CD44. In contrast, the hematopoietic marker CD45 is not expressed, discriminating MSC from hematopoietic stem cells unequivo- cally. Some antigens like CD166 are only expressed in human (116), rat (117), and sheep (107) MSC. CD166 is expressed on osteoprogenitor cells and not on terminally differentiated osteocytes, which supports the idea of dynamic regulation of antigen expression during differentiation (117). While the functional relevance of surface antigens on MSC is still mostly elusive, the expression of cell adhesion molecules like integrin a1, a2, a3, a5, a6, aV as well as b1, b3, and b4 may play an important role in cell migration, homing and tissue integra- tion of MSC (118). Most integrins were investigated in human MSC (119), while integrin aV (CD51) was also detected in mice (120). A comparison of investigated surface markers of selected mammals is given in Table 2. It is noteworthy that this list of markers is not specific or unique to identify MSC definitely, because most proteins are expressed on other cells as well. In addition, as discussed in the previous section, the marker profiles on MSC might change due to different isola- tion, culture, and expansion conditions or upon lineage differ- entiation. Vice versa, it has been shown recently that, albeit marker expression, telomere lengths and mitochondrial activ- ity remained unchanged during long-term culture of human bone marrow- and umbilical cord-derived MSC, functional features like proliferative and immunosuppressive capacity decreased over time (142). Therefore, expression of certain MSC markers does not necessarily reflect the functional capacities of MSC and vice versa, which in addition might dif- fer in MSC from different species. Immunomodulation Immunomodulatory properties are an important feature of MSC. From in vitro experiments it is known that MSC do not trigger an immune response comprising allogeneic recog- nition and elimination (143). Thus, the application of MSC regarding cell therapy in allogeneic or even xenogeneic scenar- ios seems to be feasible (144). In humans, anti-inflammatory and immunomodulatory effects were described particularly in graft-versus-host disease patients (143,144). In a xenogeneic model crossing species borders, human MSC were trans- planted in utero into fetal sheep (145). Effective engraftment and persistence in multiple tissues was recorded as well as dif- ferentiation into many cell or tissue types like chondrocytes, myocytes, or thymic stroma. Surprisingly, the engraftment persisted long-term suggesting unique immunological acceptance, which possibly was attributable to the deficiency of HLA class II surface antigens (9,78). Similarly, cross-species immunotolerance was obtained using human placenta- derived MSC to treat myelomeningocele in utero in a fetal ovine model (146), or canine placenta-derived MSC to treat neurological disorders in dogs (147). Horses, however, which were transplanted with allogeneic MHC-mismatched MSC, contained antibodies that killed donor MSC in an in vitro cytotoxicity assay indicating that MHC compatibility seemed required for acceptance of MSC transplants (148,149). In line, MSC injection into healthy horses caused a mild increase of blood CD8 T-cells and regulatory T cells in the spleen, which might indicate a cytotoxic response to the allogeneic cell transplants. However, no host tissue cytotoxic or inflamma- tory was observed (150). Generally, alloreactivity may be measured in vitro by the secretion of IFNc by activated lymphocytes in the mixed lym- phocyte reaction (MLR) assay. Using this assay, MSC sup- pressed lymphocyte reactions using both autologous and allogeneic T-cells or dendritic cells (9), and initiated the acti- vation of alloreactive T-cells without inducing the T-cell pro- liferation response (78). Even high numbers of human MSC did not cause an alloreactive lymphocyte proliferation (151). Allogeneic human bone marrow-derived MSC expressing MHC molecules activated T-cells only in the presence of the co-stimulatory factors like CD80 or CD86, which are not expressed on MSC, even not after stimulation with interferon c (IFNc) (152). Additionally, allogeneic MSC may stimulate T-cells indirectly via antigen-presenting cells (APC) presenting their MHC-antigens to T-cells. Yet, stimulation of prolifera- tion of allogeneic lymphocytes by baboon MSC failed (121). Also, human placenta-derived MCS were unable to stimulate resting T cells, but inhibited phytohemagglutinin (PHA)- stimulated T cells (73). Neither human nor rat MSC could induce any IFNc output by PBMC (Peripheral Blood Mono- nuclear Cells) indicating the lack of immunogenicity of MSC (115). Also direct cell-cell interactions were involved in the immune regulation by MSC. Human MSC bound activated T-lymphocytes with higher affinity than resting cells, T-lymphocytes better than B-lymphocytes and preferred cells of the lymphatic lineage over the myelocyte cell lineage (9,118). Along this line, human MSC minimized the T-cell response indirectly via attenuation of dendritic cell matura- tion from monocytes (153). After application of MSC into injured mouse livers, more phagocytic cells were recruited than to livers without MSC suggesting that the MSC boosted cell recruitment (154). These data summarize that in all spe- cies investigated so far, MSC modulated the immune response by affecting various pathways and cells of the immune system either directly or by communicating with other cells of the immune defence, both the innate and the adaptive immune system. The immunomodulating features of MSC are mainly mediated by paracrine mechanisms. Human MSC decreased the secretion of the proinflammatory cytokines IFN-c (155), IL-12 (156), and TNFa (155), while they elevated the anti- inflammatory cytokine IL-10 (155,157,158). Human bone Review Article Cytometry Part A 00A: 00À00, 2017 5
  • 6. Table 2. Overview of the expression of different surface antigens (1 positive, 6 heterogeneous, – negative) on human MSC compared with mice, rats, pigs, and horses and their biological meaning or term HOMINID LARGE ANIMALS SMALL ANIMALS CLUSTER OF DIFFERENTIATION FUNCTION/LOCALIZATION HUMAN HORSE SHEEP PIG RAT MOUSE CD1a Presentation of Lipid-Antigens to T-Cells – CD9 TSPAN-29 (Tetraspanin 29) 1 CD10 MME (membrane metallo-endopeptidase) 1 CD11b Integrin a M – – – CD13 ANPEP (alanylaminopeptidase) 1 1 CD14 Monocytes/Macrophages – – – CD18 Integrin b2 – CD19 B-Lymphocytes – CD24 HSA (Heat Stable Antigen) 1 CD25 Interleukin 2 Receptor a – CD29 Integrin b1 1 1 1 1 1 1 CD31 PECAM 1 (platelet/endothelial cell adhesion molecule 1) – – CD34 Hematopoietic stem cells – – – – – CD40 TNF receptor superfamily member 5 – CD44 Hematopoietic cell E-/L-Selectin ligand 1 1 1 1 1 1 CD45 PTPRC (protein tyrosine phos- phatase, receptor type, C) – – – – – – CD46 Membrane co-factor protein 1 CD49a Integrin a1 1 CD49b Integrin a2 1 CD49c Integrin a3 1 CD49d Integrin a4 – CD49e Integrin a5 1 1 CD49f Integrin a6 1 CD50 ICAM-3 (intercellular adhesion molecule 3) – CD51 Integrin alpha-v 1 1 CD54 ICAM-1 (intercellular adhesion molecule 1) – CD55 DAF (decay-accelerating factor) 1 CD56 NCAM 1 (neural cell adhesion molecule 1) 1 CD58 LFA-3 (lymphocyte function-associated antigen 3) 1 1 CD59 MAC-inhibitory protein (MAC-IP) 1 CD61 Integrin b3 1 1 1 CD62 SELP (Selectin P) – CD68 Lysosomal/endosomal-associated membrane glycoprotein 4 (LAMP-4) – CD71 TFRC (transferrin receptor) 1 1 CD72 LYB-2 1 CD73 Ecto-5’-nucleotidase 1 – 6 1 Review Article 6 Cross-species MSC
  • 7. marrow-derived MSC suppressed the proliferation of CD41 and CD81 T cells, which were stimulated by dendritic cells (DC) and peripheral blood lymphocytes (PBL). Inhibition of growth was not due to apoptosis and T cell proliferation could be re-stimulated. This phenomenon had no immunologic restriction and was triggered by both cellular and non-specific mitogenic stimuli based on the production of soluble paracrine factors (159). In a similar finding, human bone marrow-derived MSC actively suppressed the proliferation of T-cells stimulated by anti-CD3 and anti-CD28 antibodies as well as the proliferation of responder PBMC stimulated by third-party allogeneic PBMC. Separation of cultured MSC and PBMC by a semi-permeable membrane did not prevent suppression indicating the involvement of paracrine TABLE 2. Continued HOMINID LARGE ANIMALS SMALL ANIMALS CLUSTER OF DIFFERENTIATION FUNCTION/LOCALIZATION HUMAN HORSE SHEEP PIG RAT MOUSE CD80 B7-1 (ligand of CD28 and CTLA-4); T-lymphocyte activation antigen – 1 CD81 Tetraspanin 28 1 CD86 B7-2 (ligand of CD28 and CTLA-5) – –l 1 CD90 THY1 (thy-1 cell surface antigen) 1 6 6 1 1 – CD90.2 THY1.2 1 CD95 Fas 1 CD102 ICAM-2 (intercellular adhesion molecule 2) 1 CD104 Integrin b4 1 CD105 Endoglin5TGFß receptor III 1 6 6 1 CD106 VCAM 1 (vascular cell adhesion molecule 1) 6 1 1 CD117 c-Kit – – CD119 Interferon c receptor alpha 1 CD120a Tumor necrosis factor receptor superfamily, member 1A 1 CD123 Interleukin 3 receptor a – CD124 Interleukin 4 receptor 1 CD127 Interleukin 7 receptor – CD133 Prominin 1 – CD146 MCAM (melanoma cell adhesion molecule) 1 1 CD166 ALCAM 5SB-10; (activated leu- kocyte cell adhesion molecule) 1 1 CD184 CXCR4, SDF-1alpha receptor 1 1 HLA-ABC HLA Class I/MHC I 1 1 HLA-DR HLA Class II/MHC II – – – Sca-1 Stem cell antigen-1 1 SH-2 Antibody against epitope of CD105 1 SH-3 Antibody against epitope of CD73 1 SH-4 Antibody against another epitope of CD73 1 1 References (7,8,62,97,98, 115,118,119,121, 122,124–126) (42,127,128) (41,105,107, 129–132) (17,133,134) (13,135) (136–141) It appears that human profiles have been characterized best as compared to other species among mammals. This might be due to the lack of suitable antibodies for FACS analyses but will for sure be part of future research in terms of functional characterization of MSC. Review Article Cytometry Part A 00A: 00À00, 2017 7
  • 8. mechanisms mediated by secretion of IL-10, TGF-b1, prosta- glandin E2 or tryptophane (152). In rodents with lung and kidney injury, MSC improved the outcome by paracrine effects mediated by a shift from a proinflammatory to an anti- inflammatory cytokine milieu (115). Today, it is quite obvious that MSC exert their pleiotropic actions on both the innate and adaptive immune system via a great variety of different factors and mediators comprising prostaglandins, cytokines and growth factors, indoleamine 2,3-dioxygenase (IDO), tumor necrosis factor a-stimulated gene 6 (TSG6), heme oxy- genase 1 (HO-1), interleukin-1 receptor antagonist (IL-1RA) besides others (cf. (160–162) for comprehensive reviews). The immune suppression mediated by MSC may cross species barriers: porcine, rodent, and human MSC suppressed xenogeneic lymphocytes activated by lymphocytes of another species. However, mechanistically there seem to be differences between species, because human MSC were able to act onto lymphocytes even if separated by a permeable membrane, but the action of rodent MSC required cell-cell-contacts (115). Corroborating these differences, human MSC induced no sus- tained tolerance, anergy or apoptosis of activated lympho- cytes, because they recovered their ability to be stimulated after suppression of allogeneic stimulation of the lymphocytes by human or baboon MSC. In contrast, lymphocytes co- cultured with rodent MSC for 24 h, still produced IFNc, but rested in G1 of the cell cycle and stopped proliferating, which uncovers another difference of immunological mode of action between the species (78,121,163). It was stated by Le Blanc and Ringden that the suppression of T-cell activity by MSC was not efficient enough to avoid xenotransplant rejection. The natural immune response after xenotransplantation indi- cates activation of the innate and acquired immune system, involving humoral factors like antibodies and the complement system, as well as cellular mechanisms involving for example, natural killer cells and macrophages (115). It is obvious that MSC from most species investigated may interfere with all these levels of immune activation. Yet, a critical review of the single factors is quite mandatory to understand the overarch- ing coherences (121,164). Migratory Potential The fact that MSC have been found in almost all tissues investigated so far might imply that they represent a stem cell pool in the tissue to replenish tissue loss on demand, either after injury, or in the context of physiological cell turnover. However, it became obvious that MSC, when transplanted sys- temically into injured hosts, migrated to the site of injury and, under the influence of local damage signals, differentiated into cells displaying the phenotype of the corresponding target tissue. Thus, they contributed to repair of the impacted tissue by functional replacement (9). Tracking of MSC migration in vivo after transplantation in order to evaluate feasibility of the site of application, homing, and tissue distribution as well as functionality is of great interest. An elegant method has been described by Ribot et al. who applied magnetic resonance imaging (MRI) of human MSC transplanted into the hind limb of nude mice after pre-loading of the cells with the fluorophore fluorine or ultra-small iron-loaded nanoparticles (165). Homing of MSC has been described for several species. For example, bone marrow-derived MSC were transplanted in a mouse model of acute hepatic injury and contributed to the functional recovery of the liver (166). Also, in a rat model of myocardial infarction, MSC administered systemically migrated to the injured heart (167) again indicating attraction of MSC by injured tissues. Migration might be provoked by chemotactic signals produced by the tissue at the site of injury (168). Stromal-derived factor-1 (SDF-1) seems an important chemoattractant for MSC recruitment. Abbott et al. provided evidence for SDF-1/CXCR4 interactions in the recruitment of murine bone marrow-derived MSC to myocardial infarction (169). CXCR4, also known as CD184 or SDF-1 alpha receptor, is expressed by MSC from several species including canine (170), human (122), and mice (136). CXCR4 expression seems not only to be important for MSC migration to sites of tissue injury, but also to the bone marrow as shown for human MSC from bone marrow (137). Besides CXCR4, a row of other surface proteins is likely to be involved in MSC hom- ing. Ip et al. presented data showing that integrin1 ß (CD29) but not CXCR4 on murine MSC was important for MSC migration and engraftment into the infarcted myocardium (171). In line, Ponte et al. identified several cytokines includ- ing PDGF-AB, IGF-1, and RANTES, which were more effec- tive in driving MSC homing than SDF-1. TNFa enhanced the migratory response to these factors (172). A set of several che- mokine receptors (CXCR4, CX3CR1, CXCR6, CCR1, and CCR7) expressed on human bone marrow-derived MSC seemed to be involved in the chemotactic migration in response to CX3CL1 and CXCL12 as shown in a human pan- creatic islet in vitro model (97). Monocyte chemoattractant protein-1 (MCP-1), usually not expressed in healthy rat brains, was rapidly up-regulated after middle cerebral artery occlusion. In these ischemic rat brains, MCP-1 promoted the migration of intravenously infused bone marrow-derived- MSC to the site of injury. Migration was reduced significantly, when brain tissue extracts were incubated with an antibody against MCP-1 suggesting the chemotactic impact on MSC migration (173). Corroborating the migratory potential of MSC, native murine MSC transplanted into sublethally irradi- ated mice could be recovered from the bone marrow and the spleen already 24 h after systemic application showing their rapid and efficient homing. This again underlined the assumption that the mobilization of MSC and their subse- quent homing to injured tissues depended on the systemic and local inflammatory state of the organism (138,154). Finally, matrix metalloproteinases (MMP) secreted by MSC were required for the crossing of the basement membrane and MSC tissue homing. The inflammatory cytokine-dependent expression of MMP-2, MT1-MMP, and MMP-9 by MSC strongly stimulated the chemotactic migration of human MSC through basement membranes (174). Propagation of rat bone marrow-derived MSC in vitro decreased their ability to home to the bone marrow demonstrating that manipulation of MSC in vitro prior to transplantation might severely Review Article 8 Cross-species MSC
  • 9. impact on their migratory potential and hence their therapeu- tic potential (175). Proregenerative Mode of Action The regenerative potential of MSC, differentiated or not, may principally be based on two mechanisms. One is character- ized by functional tissue replacement of injured tissue, which requires the MSC to differentiate and adopt the functional properties of the host tissue after engraftment. The second fea- ture is based on paracrine mechanisms promoting self- regeneration of the injured tissue. The first may be achieved by integration into the damaged tissue and proliferation of MSC transplants at the site of tissue damage until complete tissue repair, the second by the prevention of injury progress followed by stimulation of self-repair of the tissue affected. It is quite obvious that tissue integration needs direct cell–cell interac- tions, while paracrine mechanisms may act locally after recruit- ment of MSC transplants to the site of injury. This consideration is of high relevance in clinical applications. Acute organ injury needs temporary therapeutic intervention com- prising rapid local actions of MSC to promote self-repair of the injured tissue/organ. Chronic diseases, however, require sus- tained tissue support preferably by tissue integration and differ- entiation of MSC into the cell type of the injured tissue in order to ameliorate organ/tissue dysfunction long-term by functional replacement of the diseased host tissue. In a xenogeneic mouse model of partial hepatectomy, human subcutaneous adipose tissue-derived MSC, which had been differentiated into hepatocyte-like cells prior to trans- plantation, re-populated the liver of immune-deficient mice by about 25% ten weeks after transplantation. This was a 10- times higher rate as compared to the use of undifferentiated MSC (176). While in this experiment an integration of cell transplants into the liver was achieved indicating the func- tional substitution of the recipient liver by donor MSC, a mixed direct and paracrine effect was described in mice and rats after intoxication with acetaminophen. The drug induces regional necrosis in the liver, which was occupied by trans- planted human bone marrow-derived and rat adipose tissue- derived MSC in immune-deficient mice and rats, respectively. They prevented disease progress by inhibition of apoptosis and necrosis, and supported liver regeneration by stimulating proliferation of host hepatocytes. Functional cell transplants were found in the host livers for up to seven weeks after trans- plantation (154,164). This indicates that MSC are capable of liver support by functional tissue substitution and paracrine stimulation of tissue regeneration after acute inflammatory insult. Similar results were obtained in rats, in which acute liver failure was induced by D-galactosamine. Here, systemic injection of MSC reduced both the apoptotic rate and aug- mented the proliferation of host liver hepatocytes indicative for the stimulation of cell cycle progression as verified by an increase in the expression of cell cycle proteins. This clearly demonstrated the paracrine hepatotropic action of MSC transplants (177). Human MSC from fetal blood also stimu- lated the proliferation and differentiation of CD341 hemato- poietic stem cells from umbilical cord blood (62). These proproliferative features of MSC were mediated by secretion of growth factors. In vitro chondrogenesis of periost-derived MSC from young rabbits was strongly stimulated by a combi- nation of growth factors IGF-1 and TGF-b1 (178). A caprine model of osteoarthritis combined with local application of autologous bone marrow-derived MSC corroborated the pleiotropic regenerative potential of MSC. In this model, autologous MSC labeled with green fluorescent protein (GFP) were injected intra-articularly six weeks after unilateral induc- tion of osteoarthritis in the knees of goats. GFP-labeled cells were detected in the treated joints and successfully regenerated the meniscus (179). Together these data show that MSC possess a proregenerative potential, which makes them ideal candidates for regenerative medicine in a wide variety of degenerative disease applications both in humans and animals. CLINICAL USE Originating from the versatile successful therapeutic use in animal experiments, hope grew that MSC therapy might be feasible clinically. However, clinical animal trials have only been performed in dogs, cats and horses. In humans, a bulk of phase I/II clinical trials were initiated to assess safety and feasi- bility of MSC therapy under various disease conditions. Animal Trials Only few clinical trials in pets like cats and dogs are avail- able. Intraperitoneal autologous adipose tissue-derived MSC injection was evaluated in 10 healthy adult cats. The injection was well tolerated with only mild adverse events in two cats (180). Quimby et al. described a randomized, placebo- controlled trial in eight cats evaluating intravenous applica- tion of adipose-derived allogeneic MSC in feline chronic kid- ney disease. The administration of MSC was not associated with adverse events, however, significant improvement of renal function was not achieved by MSC administration (181). The safety of intravenous application of allogeneic adi- pose tissue-derived MSC was assessed in 11 dogs suffering from inflammatory bowel disease. The single injection of MSC was safe and improved gastrointestinal lesions by reduc- ing gastrointestinal inflammation (182). In large animal clinics, MSC are used mainly to treat musculoskeletal diseases in horses like tendon injuries or bone and cartilage damage (183). Feasibility of autologous, geneti- cally modified autologous, allogeneic, and xenogeneic MSC was tested by joint injection in six healthy horses. A total number of 15 3 106 MSC induced a moderate acute inflam- matory response in the joint, which was greater using alloge- neic and xenogeneic as compared to autologous MSC (184). Thus, both autologous and allogenic MSC application in horses seems to be safe and feasible. Accordingly, in most equine clinical trials autologous bone marrow- or adipose tissue-derived MSC were used. The most prominent diseases treated represented tendon lesions, which showed improve- ment after injection of 2 3 106 –10 3 106 autologous MSC directly into the site of the lesion. No study reported of adverse effects provoked by MSC application. Thirteen out of Review Article Cytometry Part A 00A: 00À00, 2017 9
  • 10. 18 horses returned to race competitions after autologous bone marrow-derived MSC injection into sites of ligament and ten- don lesions (185). Similar findings demonstrated recovery from lesions of the superficial digital flexor tendon in race- horses in 9 out of 11 horses by MSC treatment (186). Bone marrow-derived MSC improved superficial digital flexor ten- dinopathy and reduced the re-injury rate in national hunt racehorses (187). Therapy of tendonitis with MSC was assessed in horses in a randomized controlled trial by direct injection of 10 3 106 autologous adipose tissue-derived MSC. These prevented progression of tendon lesion and decreased inflammation (188). Albeit no significant difference was achieved clinically or histologically in horses after treatment of intra-articular cartilage lesions with bone marrow-derived MSC, it seemed that repair quality was improved by augmen- tation of aggrecan content and tissue firmness (189). Only partial repair was described treating equine bone spavin with adipose tissue-derived MSC (190). In summary, equine MSC might thus be clinically established to treat inflammatory acute and chronic degenerative diseases of tendon, cartilage and bone. Overall, the clinical application of both autologous and allogenic MSC in several animal species including horse, dog and cat turned out safe and without adverse events. Human Trials The ClinicalTrials.gov register provides a plenty of clini- cal trials using human MSC administration for treatment of different diseases. Studies not yet recruiting patients will not be discussed here. A summary of studies reporting “recruitment completed” and comprising the most relevant diseases is given in Table 3. Most of the studies involving MSC registered on ClinicalTrials.gov do not present results so far. Therefore, we retrieved the PubMed database using the MeSH terms “clinical trials” and “mesenchymal stromal cells.” Results are summarized in Table 4 comprising type of disease, source of MSC, route and dose of application, number of patients, study type and outcome of both human and equine clinical trials. Most study results are available for MSC appli- cation in degenerative neuronal diseases like amyotrophic lat- eral sclerosis (ALS), an as yet untreatable, progressive, neurodegenerative disorder resulting in death of motoneurons, or multiple sclerosis (MS), a chronic inflamma- tion of the nervous system. Except for one study using adipose tissue-derived MSC (191) to treat ALS or MS, all other clinical trials used autologous bone marrow-derived MSC (192–199). For the treatment of MS, intravenous application was chosen, whereas for ALS cells were mostly injected intrathecally. Cell numbers injected ranged from 1 3 106 –1 3 108 cells per kg body weight. In all phase I/II clinical trials reporting treatment of ALS or MS with MSC, cell application provoked no adverse events, retarded ALS progression (194,196), induced immu- nomodulatory effects (196,199) and improved visual acuity in MS (198). Autologous bone marrow- or adipose tissue- derived MSC application showed no adverse events in spinal cord injury (200–202) or in osteoarthritis (203,204). For the treatment of graft-versus-host disease (GvHD), mainly alloge- neic bone marrow-derived stem cells were administrated intravenously causing also no adverse events and featuring immunosuppressive properties (205,206). This was corrobo- rated in a pediatric study of GvHD, which confirmed the immunomodulatory potential of allogenic MSC (207). The anti-inflammatory action of both autologous (208) and allogenic (209) MSC was demonstrated in Crohn’s disease after intravenous application of bone marrow-derived MSC. Treatment of several liver diseases including liver transplanta- tion with MSC has shown safety and feasibility (210). In acute-on-chronic liver failure, allogeneic umbilical cord- derived MSC lowered the MELD (Modell of End stage Liver Diseases) score indicating improvement of liver function (211). In liver cirrhosis, representing another serious impair- ment of liver function, autologous bone marrow-derived MSC also reduced the MELD score (212). Corroborating the benefi- cial impact on acute diseases, the application of autologous (213) or allogeneic bone marrow-derived MSC (214) was safe and improved the NYHA (New York Heart Association) score in acute myocardial infarction. In summary, the outcomes of clinical trials using human MSC for therapy of acute and chronic inflammatory diseases of different etiologies show that the treatment is bona fide safe and may encompass cells of autologous or allogeneic origin. The major mode of MSC action is probably the modulation of inflammatory processes attenuating tissue damage and fos- tering self-repair of the injured tissue. Since in most studies cells were administered systemically, soluble or, after migra- tion of MSC to the site of injury, even paracrine mechanisms must be anticipated, thus substantiating findings, which have been prior proven in animal models as discussed above. Safety Issues Despite their well acknowledged and unique proregener- ative and immunomodulatory features, which make MSC highly attractive for numerous applications in regenerative medicine as shown above, some critical issues potentially limit the clinical use of MSC. This section will finally give a short insight into the contrary views on special safety concerns. A serious constraint of the therapeutic use of MSC is their putative profibrogenic potential (216–218). Human bone marrow-derived MSC contributed to lung fibrosis (219) Table 3. Overview of human clinical trials involving MSC therapy with patient recruitment completed DISEASE NO. OF REGISTERED STUDIES ALS (amyotrophic lateral sclerosis) 6 MS (multiple sclerosis) 8 Spinal cord injury 9 Osteoarthritis 30 Graft-versus-host disease 11 Crohns disease 4 Liver diseases 8 Kidney disease 2 Acute myocardial infarction 6 Source: ClinicalTrials.gov Review Article 10 Cross-species MSC
  • 11. Table4.Compilationofclinicaltrialsusingmesenchymalstemcellsinhumansandhorsesandstudyoutcome SPECIESDISEASEORIGINOFMSC ROUTEOFAPPLICATIONCELL NUMBERSINFUSEDNO.PATIENTSTUDYTYPEOUTCOMEREF HumanAmyotrophic lateralsclerosis (ALS) AutologousbonemarrowIntrathecally2x1*106 /kg8PhaseINoadverseevents(192) Autologousbonemarrow37Investigator- initiatedtrial VEGF,ANG,and TGF-blevelsinMSCidentifiedaspotential biologicalmarkerstopredicttheeffective- nessofautologousMSCtherapy (193) AutologousbonemarrowIntramuscularly intrathecally 1*106 /kg 6 6 PhaseI/IINoadverseevents reductioninALSFunctionalRatingScale Revised(ALS-FRS-R)slope (149) AutologousadiposetissueIntrathecally107 -108 /kg27PhaseINoadverseevents(191) Autologousbonemarrowintrathecally1564.5*106 26PhaseI/IIaNoadverseevents retardeddiseaseprogression (195) ALS/MSAutologousbonemar-rowintrathecally intravenously2.5*106 34 14 PhaseI/IIClinicallyfeasible/noadverseevents inducesimmediateimmunomodulatory effects (196) Multiplesclerosis (MS) AutologousbonemarrowIntravenously1–2*106 /kg11PhaseIIaInitialsafetyprofileandfeasibilityofthe intervention (197) AutologousbonemarrowIntravenously1.6*106 /kg10PhaseIIaNoseriousadverseevents improvementofvisualacuity (198) AutologousbonemarrowIntravenously1–2*106 /kg8PhaseIINoadverseevents reductionofinflammation (199) SpinalcordinjuryAutologousadiposetissueIntravenously4*108 12PhaseINoadverseevents notumordevelopment (200) BonemarrowLocally107 /ml14PhaseINoadverseevents variableimprovementsintactilesensitivity (201) Autologousbonemarrowintramedullary1.6*106 subdurally3.23106 16PhaseIIINoadverseevents veryweaktherapeuticeffectcomparedwith multipleMSCsinjection (202) OsteoarthritisAutologousadiposetissueintraarticularly lowdose1.0*107 middose5.0*107 highdose1.0*108 9PhaseINoadverseevents highdoseimprovedfunctionandpain (203) Autologousadiposetissueintraarticularly lowdose 2*106 middose10*106 highdose50*106 10PhaseINoadverseevents patientstreatedwithlow-doseMSCexperi- encedsignificantimprovementsinpain levels (204) Graft-versus-host disease(GvHD) Allogeneicbonemarrowintravenously 1.1*106 /kg 25PhaseI/IINoadverseeventsrelatedtotheMSC 17responded (205) Review Article Cytometry Part A 00A: 00À00, 2017 11
  • 12. TABLE4.Continued SPECIESDISEASEORIGINOFMSC ROUTEOFAPPLICATIONCELL NUMBERSINFUSEDNO.PATIENTSTUDYTYPEOUTCOMEREF 7patientsdidnotrespondtoMSCinfusion Allogeneicbonemarrow0.9-9*106 /kg8PhaseIimmunosuppressive healdamagedintestinalepithelium (206) Allogeneicbonemarrowintravenously 2*106 /kg severalapplications every4days 12 (pediatric) PhaseI/IINoinfusionalorotheridentifiableacutetox- icityadverseevents multipleinfusionswerewelltoleratedand weresafeinchildren clinicalresponses,particularlyintheGIsys- tem,wereseeninthemajorityofchildren withsevererefractoryaGvHD (207) Crohn’sdiseaseAutologousbonemarrowintravenouslyupto106 /kg12PhaseInoadverseeventsandfeasible(208) Allogeneicbonemarrowintravenously2*106 /kg15PhaseIIreducedCrohn’sDiseaseActivityIndex (CDAI)andCrohn’sDiseaseEndoscopic IndexofSeverity(CDEIS) (209) Livertransplantation Acute-on-chronic liverfailure Livercirrhosis n.k.portalvein(0d)centralvein (2d)1.5*108 1Casestudynoevidenceofacutetoxicity administrationtechnicallyfeasible (210) Allogeneicumbilicalcordintravenously0.5*106 24PhaseI/IInoadverseevents reductionofMELDscore (211) Autologousbonemarrowintravenouslyorportalvein 3–5*106 /kg 8PhaseInoadverseevents improvementofModelforEnd-stageLiver Disease(MELD)score (212) KidneydiseaseAutologousbonemarrowintravenously1–2*106 /kg6PhaseINoadverseevents anticipatedsystemicimmunosuppression (215) Acute myocardial infarction Autologousbonemarrow Allogeneicbonemarrow 10infarctedsites 20–200*106 15 15 PhaseI/II randomized comparison Noadverseeventswithautologousandallo- genicMSC improvementofNewYorkHeartAssociation (NYHA)score (214) AutologousBoneMarrowInfarct-relatedartery 1*106 cells 80PhaseII/IIINoadverseevents modestimprovementinleftventricularejec- tionfraction(LVEF)at6-monthfollow-up (213) HorseTendonlesionAutologousbonemarrowintolesion 2*106 /cm2 18PhaseI/IINoadverseevents 13horsesreturnedtoracecompetitions (185) Autologousbonemarrowintolesion 10*106 141PhaseI/IINoadverseeffects reductionofthere-injuryrateaftersuperficial digitalflexortendinopathy,esp.inNational Huntracehorses. (187) Autologousadiposetissueintolesion 10*106 8phaseI/IIPreventionoftendonlesionprogressionas observedbyultrasoundexamination improvementoftissueorganization attenuationofinflammation (188) Review Article 12 Cross-species MSC
  • 13. and the fibrotic action of ovarian endometrial stromal cells was enhanced by paracrine production of TGF-b1 and Wnt1 after treatment with human endometrium-derived MSC (220). However, studies in rats and mice demonstrated that MSC did not contribute to but rather improved liver fibrosis (221–224). In rodent models of liver regeneration after partial hepatectomy in rats and mice using either syngeneic or xeno- geneic MSC, no signs of profibrotic impact for up to ten weeks after transplantation were observed (13,176). The anti- fibrotic effects of human MSC transplanted into xenogeneic rodent models were also obvious under acute and chronic liver (154,225), lung (138,226,227), or kidney (228,229) dam- age conditions. For instance, in a murine bleomycin-induced lung fibrosis model, MSC lowered type I pro-collagen, alpha- smooth muscle actin (a-SMA), TGF-ß and the oncogenic transcription factor c-Myc (230). Furthermore, transplanta- tion of MSC into heart, muscle or lung did not result in any extracellular matrix changes indicative for fibrosis or emer- gence of malignant cells (47). Human bone marrow-derived MSC suppressed abnormal collagen I deposition in the nucleus pulposus by modulating MMP12 and HSP47 (231). In several rodent models of liver fibrosis, the antifibrotic effect of human bone marrow- and placenta-derived MSC on the resolution of fibrosis depended on the balance between the matrix metalloproteinases (MMP) and their inhibitors (TIMP) (223,232,233). Especially MMP2 (234) and MMP9 (232) were activated by MSC. Furthermore, the expression of a-SMA was inhibited and apoptosis of hepatic stellate cells augmented, all contributing to regression of fibrosis (235,236). TGF-b1, the major mediator of fibrosis, was down- regulated by human MSC in fibrotic mouse livers (237), and its signaling through the phosphorylation of Smad2 was atten- uated (238). Transplantation of human umbilical cord blood- derived MSC increased IL-4, which promoted mobilization of Kupffer cells alleviating liver fibrosis in rats (239).The contro- versial results showing either fibrotic or antifibrotic features of MSC might also depend on the use of different tissue sources and methods used for the isolation of MSC as well as on the different animal models or fibrotic organs used. Accordingly, it has been demonstrated in mice that adipose tissue-derived human MSC were significantly more efficient in reducing skin fibrosis as compared to bone marrow-derived MSC. This dif- ference was related to a stronger reduction of TNFa and IL-1b as well as an enhanced ratio of MMP1/TIMP1 in skin and lung tissues when treated with adipose tissue- as compared to treatment with bone marrow-derived MSC (240). CONCLUSION/SUMMARY In mammals a big variety of different tissue and organ sources harboring MSC exists. Nearly every tissue investigated so far contained MSC with similar biological characteristics such as surface marker expression indicating conserved fea- tures and functions in mammals at least. Carrying a low ethi- cal burden, and considering their high regenerative properties, MSC are good candidates for the use of cellular therapy in both human clinical and in veterinary medical settings. The TABLE4.Continued SPECIESDISEASEORIGINOFMSC ROUTEOFAPPLICATIONCELL NUMBERSINFUSEDNO.PATIENTSTUDYTYPEOUTCOMEREF Autologousn.k.n.k.11PhaseI/IIClinicalrecoveryfromtendonlesionsin9out of11horses (186) BonespavinAutologousadiposetissueintraarticularly n.k. n.k.PhaseI/IILong-termbeneficialinfluence(190) CartilagelesionAutologousbonemarrowIntraarticularly 20*106 10PhaseI/IIIncreaseofaggrecanlevelsofinrepairedtissue noimprovementoflesions. (189) HealthyAutologous Autologous,genetically modified allogeneic xenogeneic Intojoint 15*106 6PhaseIModerateacuteinflammatoryjointresponse thatwasgreaterforallogeneicandxenoge- neicMSC (184) Theclassificationoftrialsinhorsesdoesnotrepresentanofficialterminologyasusedinhumanclinicaltrials.Ithasbeenintroducedinordertodemonstratethatsafetyandfeasi- bilityaswellasinthesecondlinetherapeuticeffectivenesshasbeentestedasusuallyinhumanclinicalphaseI/IItrials.n.k.5notknown. Review Article Cytometry Part A 00A: 00À00, 2017 13
  • 14. regeneration of tendon, bone and cartilage seems to be the most promising clinical application at present in large animals like horses but in humans as well. Nevertheless, their anti- inflammatory and immunomodulatory features make MSC highly attractive for clinical use in the setting of a huge range of diseases comprising both acute and chronic inflammatory tissue deteriorations in man and animals. Yet, it is mandatory prior to clinical applications to unravel the response of MSC from a given source onto a specific disease environment in suitable animal models. ACKNOWLEDGMENTS All authors declare that they have no conflicts of interest. AUTHOR CONTRIBUTIONS All authors substantially contributed to conception and design, drafting and writing the article and revising it critically for important intellectual content. LITERATURE CITED 1. Friedenstein AJ, Chailakhyan RK, Gerasimov UV. Bone marrow osteogenic stem cells: In vitro cultivation and transplantation in diffusion chambers. Cell Tissue Kinet 1987;20:263–272. 2. Kassem M, Kristiansen M, Abdallah BM. Mesenchymal stem cells: Cell biology and potential use in therapy. Basic Clin Pharmacol Toxicol 2004;95:209–214. 3. Horwitz EM, Le Blanc K, Dominici M, Mueller I, Slaper-Cortenbach I, Marini FC, Deans RJ, Krause DS, Keating A. Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement. Cytotherapy 2005;7: 393–395. 4. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop D, Horwitz E. Minimal criteria for defining multipotent mes- enchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006;8:315–317. 5. Keating A. Mesenchymal stromal cells: New directions. Cell Stem Cell 2012;10: 709–716. 6. Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: Mesenchymal stem cells: Their phenotype, differentiation capacity, immunological features, and poten- tial for homing. Stem Cells 2007;25:2739–2749. 7. Pittenger MF. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143–147. 8. Conget PA, Minguell JJ. Phenotypical and functional properties of human bone mar- row mesenchymal progenitor cells. J Cell Physiol 1999;181:67–73. 9. Barry FP, Murphy JM. Mesenchymal stem cells: Clinical applications and biological characterization. Int J Biochem Cell Biol 2004;36:568–584. 10. Seo MJ, Suh SY, Bae YC, Jung JS. Differentiation of human adipose stromal cells into hepatic lineage in vitro and in vivo. Biochem Biophys Res Commun 2005;328: 258–264. 11. Bruckner S, Tautenhahn HM, Winkler S, Stock P, Dollinger M, Christ B. A fat option for the pig: Hepatocytic differentiated mesenchymal stem cells for translational research. Exp Cell Res 2014;321:267–275. 12. Kim D, Monaco E, Maki A, de Lima AS, Kong HJ, Hurley WL, Wheeler MB. Morphologic and transcriptomic comparison of adipose- and bone-marrow-derived porcine stem cells cultured in alginate hydrogels. Cell Tissue Res 2010;341:359–370. 13. Sgodda M, Aurich H, Kleist S, Aurich I, Konig S, Dollinger MM, Fleig WE, Christ B. Hepatocyte differentiation of mesenchymal stem cells from rat peritoneal adipose tissue in vitro and in vivo. Exp Cell Res 2007;313:2875–2886. 14. Laschke MW, Schank TE, Scheuer C, Kleer S, Shadmanov T, Eglin D, Alini M, Menger MD. In vitro osteogenic differentiation of adipose-derived mesenchymal stem cell spheroids impairs their in vivo vascularization capacity inside implanted porous polyurethane scaffolds. Acta Biomater 2014;10:4226–4235. 15. Tholpady SS, Katz AJ, Ogle RC. Mesenchymal stem cells from rat visceral fat exhibit multipotential differentiation in vitro. Anat Rec A Discov Mol Cell Evol Biol 2003; 272:398–402. 16. Lin Y, Chen X, Yan Z, Liu L, Tang W, Zheng X, Li Z, Qiao J, Li S, Tian W. Multiline- age differentiation of adipose-derived stromal cells from GFP transgenic mice. Mol Cell Biochem 2006;285:69–78. 17. Bruckner S, Tautenhahn HM, Winkler S, Stock P, Jonas S, Dollinger M, Christ B. Isolation and hepatocyte differentiation of mesenchymal stem cells from porcine bone marrow–”surgical waste” as a novel MSC source. Transplant Proc 2013;45: 2056–2058. 18. Aurich I, Mueller LP, Aurich H, Luetzkendorf J, Tisljar K, Dollinger MM, Schormann W, Walldorf J, Hengstler JG, Fleig WE, et al. Functional integration of hepatocytes derived from human mesenchymal stem cells into mouse livers. Gut 2007;56:405–415. 19. Pelz S, Stock P, Bruckner S, Christ B. A methionine-choline-deficient diet elicits NASH in the immunodeficient mouse featuring a model for hepatic cell transplanta- tion. Exp Cell Res 2012;318:276–287. 20. Lodie TA, Blickarz CE, Devarakonda TJ, He C, Dash AB, Clarke J, Gleneck K, Shihabuddin L, Tubo R. Systematic analysis of reportedly distinct populations of multipotent bone marrow-derived stem cells reveals a lack of distinction. Tissue Eng 2002;8:739–751. 21. Kazemi D, Shams Asenjan K, Dehdilani N, Parsa H. Canine articular cartilage regen- eration using mesenchymal stem cells seeded on platelet rich fibrin: Macroscopic and histological assessments. Bone Joint Res 2017;6:98–107. 22. Kadiyala S, Young RG, Thiede MA, Bruder SP. Culture expanded canine mesenchy- mal stem cells possess osteochondrogenic potential in vivo and in vitro. Cell Trans- plant 1997;6:125–134. 23. Fortier LA, Nixon AJ, Williams J, Cable CS. Isolation and chondrocytic differentia- tion of equine bone marrow-derived mesenchymal stem cells. Am J Vet Res 1998;59: 1182–1187. 24. Caminal M, Velez R, Rabanal RM, Vivas D, Batlle-Morera L, Aguirre M, Barquinero J, Garcia J, Vives J. A reproducible method for the isolation and expansion of ovine mesenchymal stromal cells from bone marrow for use in regenerative medicine pre- clinical studies. J Tissue Eng Regen Med 2016; Nov 18. doi: 10.1002/term.2254. [Epub ahead of print] 25. Gao LR, Zhang NK, Bai J, Ding QA, Wang ZG, Zhu ZM, Fei YX, Yang Y, Xu RY, Chen Y. The apelin-APJ pathway exists in cardiomyogenic cells derived from mesenchymal stem cells in vitro and in vivo. Cell Transplant 2010;19: 949–958. 26. Tropel P, Noel D, Platet N, Legrand P, Benabid AL, Berger F. Isolation and characteri- sation of mesenchymal stem cells from adult mouse bone marrow. Exp Cell Res 2004;295:395–406. 27. Deschaseaux F, Gindraux F, Saadi R, Obert L, Chalmers D, Herve P. Direct selection of human bone marrow mesenchymal stem cells using an anti-CD49a antibody reveals their CD45med,low phenotype. Br J Haematol 2003;122:506– 517. 28. Tondreau T, Lagneaux L, Dejeneffe M, Massy M, Mortier C, Delforge A, Bron D. Bone marrow-derived mesenchymal stem cells already express specific neural pro- teins before any differentiation. Differentiation 2004;72:319–326. 29. Donnenberg VS, Ulrich H. Mesenchymal stem cells, therapy, and cytometry. Cytom- etry A 2013;83:8–10. 30. Lin Y, Liu L, Li Z, Qiao J, Wu L, Tang W, Zheng X, Chen X, Yan Z, Tian W. Pluripo- tency potential of human adipose-derived stem cells marked with exogenous green fluorescent protein. Mol Cell Biochem 2006;291:1–10. 31. Oedayrajsingh-Varma MJ, van Ham SM, Knippenberg M, Helder MN, Klein-Nulend J, Schouten TE, Ritt MJ, van Milligen FJ. Adipose tissue-derived mesenchymal stem cell yield and growth characteristics are affected by the tissue-harvesting procedure. Cytotherapy 2006;8:166–177. 32. Kern S, Eichler H, Stoeve J, Kluter H, Bieback K. Comparative analysis of mesenchy- mal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells 2006;24:1294–1301. 33. Wagner W, Wein F, Seckinger A, Frankhauser M, Wirkner U, Krause U, Blake J, Schwager C, Eckstein V, Ansorge W, et al. Comparative characteristics of mesenchy- mal stem cells from human bone marrow, adipose tissue, and umbilical cord blood. Exp Hematol 2005;33:1402–1416. 34. Izadpanah R, Trygg C, Patel B, Kriedt C, Dufour J, Gimble JM, Bunnell BA. Biologic properties of mesenchymal stem cells derived from bone marrow and adipose tissue. J Cell Biochem 2006;99:1285–1297. 35. Ranera B, Lyahyai J, Romero A, Vazquez FJ, Remacha AR, Bernal ML, Zaragoza P, Rodellar C, Martin-Burriel I. Immunophenotype and gene expression profiles of cell surface markers of mesenchymal stem cells derived from equine bone marrow and adipose tissue. Vet Immunol Immunopathol 2011;144:147–154. 36. Groth A, Ottinger S, Kleist C, Mohr E, Golriz M, Schultze D, Bruns H, Mehrabi A, Schemmer P, Buchler MW, et al. Evaluation of porcine mesenchymal stem cells for therapeutic use in human liver cancer. Int J Oncol 2012;40:391–401. 37. Jeong SH, Ji YH, Yoon ES. Immunosuppressive activity of adipose tissue-derived mesenchymal stem cells in a rat model of hind limb allotransplantation. Transplant Proc 2014;46:1606–1614. 38. Taha MF, Hedayati V. Isolation, identification and multipotential differentiation of mouse adipose tissue-derived stem cells. Tissue Cell 2010;42:211–216. 39. da Silva Meirelles L, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci 2006;119:2204–2213. 40. Schwartz RE, Reyes M, Koodie L, Jiang Y, Blackstad M, Lund T, Lenvik T, Johnson S, Hu WS, Verfaillie CM. Multipotent adult progenitor cells from bone marrow differentiate into functional hepatocyte-like cells. J Clin Invest 2002;109: 1291–1302. 41. Heidari B, Shirazi A, Akhondi MM, Hassanpour H, Behzadi B, Naderi MM, Sarvari A, Borjian S. Comparison of proliferative and multilineage differentiation potential of sheep mesenchymal stem cells derived from bone marrow, liver, and adipose tis- sue. Avicenna J Med Biotechnol 2013;5:104–117. 42. Paebst F, Piehler D, Brehm W, Heller S, Schroeck C, Tarnok A, Burk J. Comparative immunophenotyping of equine multipotent mesenchymal stromal cells: An approach toward a standardized definition. Cytometry Part A 2014;85A:678–687. 43. Khojasteh A, Dashti SG, Dehghan MM, Behnia H, Abbasnia P, Morad G. The osteor- egenerative effects of platelet-derived growth factor BB cotransplanted with mesen- chymal stem cells, loaded on freeze-dried mineral bone block: A pilot study in dog mandible. J Biomed Mater Res B Appl Biomater 2014;102:1771–1778. 44. Liu H, Wei X, Ding X, Li X, Zhou G, Li P, Fan Y. Comparison of cellular responses of mesenchymal stem cells derived from bone marrow and synovium on combined silk scaffolds. J Biomed Mater Res A 2015;103:115–125. Review Article 14 Cross-species MSC
  • 15. 45. Lu T, Huang Y, Wang H, Ma Y, Guan W. Multi-lineage potential research of bone marrow-derived stromal cells (BMSCs) from cattle. Appl Biochem Biotechnol 2014; 172:21–35. 46. Webb TL, Quimby JM, Dow SW. In vitro comparison of feline bone marrow-derived and adipose tissue-derived mesenchymal stem cells. J Feline Med Surg 2012;14: 165–168. 47. Hui JH, Li L, Teo YH, Ouyang HW, Lee EH. Comparative study of the ability of mes- enchymal stem cells derived from bone marrow, periosteum, and adipose tissue in treatment of partial growth arrest in rabbit. Tissue Eng 2005;11:904–912. 48. Kang BJ, Lee SH, Kweon OK, Cho JY. Differentiation of canine adipose tissue- derived mesenchymal stem cells towards endothelial progenitor cells. Am J Vet Res 2014;75:685–691. 49. Lu T, Xiong H, Wang K, Wang S, Ma Y, Guan W. Isolation and characterization of adipose-derived mesenchymal stem cells (ADSCs) from cattle. Appl Biochem Biotechnol 2014;174:719–728. 50. Smiler D, Soltan M, Albitar M. Toward the identification of mesenchymal stem cells in bone marrow and peripheral blood for bone regeneration. Implant Dent 2008;17: 236–247. 51. Chong PP, Selvaratnam L, Abbas AA, Kamarul T. Human peripheral blood derived mesenchymal stem cells demonstrate similar characteristics and chondrogenic differ- entiation potential to bone marrow derived mesenchymal stem cells. J Orthop Res 2012;30:634–642. 52. Li S, Huang KJ, Wu JC, Hu MS, Sanyal M, Hu M, Longaker MT, Lorenz HP. Periph- eral blood-derived mesenchymal stem cells: Candidate cells responsible for healing critical-sized calvarial bone defects. Stem Cells Transl Med 2015;4:359–368. 53. Lyahyai J, Mediano DR, Ranera B, Sanz A, Remacha AR, Bolea R, Zaragoza P, Rodellar C, Martin-Burriel I. Isolation and characterization of ovine mesenchymal stem cells derived from peripheral blood. BMC Vet Res 2012;8:169. 54. Giovannini S, Brehm W, Mainil-Varlet P, Nesic D. Multilineage differentiation potential of equine blood-derived fibroblast-like cells. Differentiation 2008;76: 118–129. 55. Dhar M, Neilsen N, Beatty K, Eaker S, Adair H, Geiser D. Equine peripheral blood-derived mesenchymal stem cells: Isolation, identification, trilineage differentiation and effect of hyperbaric oxygen treatment. Equine Vet J 2012;44: 600–605. 56. Wan C, He Q, Li G. Allogenic peripheral blood derived mesenchymal stem cells (MSCs) enhance bone regeneration in rabbit ulna critical-sized bone defect model. J Orthop Res 2006;24:610–618. 57. Sato K, Yamawaki-Ogata A, Kanemoto I, Usui A, Narita Y. Isolation and characteri- sation of peripheral blood-derived feline mesenchymal stem cells. Vet J 2016;216: 183–188. 58. Lee KD, Kuo TK, Whang-Peng J, Chung YF, Lin CT, Chou SH, Chen JR, Chen YP, Lee OK. In vitro hepatic differentiation of human mesenchymal stem cells. Hepatol- ogy 2004;40:1275–1284. 59. Raoufi MF, Tajik P, Dehghan MM, Eini F, Barin A. Isolation and differentiation of mesenchymal stem cells from bovine umbilical cord blood. Reprod Domest Anim 2011;46:95–99. 60. Li WW, Wei YH, Li H, Lai DM, Lin TN. Isolation and characterization of a novel strain of mesenchymal stem cells from mouse umbilical cord: Potential application in cell-based therapy. PLoS One 2013;8:e74478. 61. Seo MS, Jeong YH, Park JR, Park SB, Rho KH, Kim HS, Yu KR, Lee SH, Jung JW, Lee YS, et al. Isolation and characterization of canine umbilical cord blood-derived mesenchymal stem cells. J Vet Sci 2009;10:181–187. 62. Campagnoli C, Roberts IA, Kumar S, Bennett PR, Bellantuono I, Fisk NM. Identifi- cation of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood 2001;98:2396–2402. 63. Mancanares CA, Oliveira VC, Oliveira LJ, Carvalho AF, Sampaio RV, Mancanares AC, Souza AF, Perecin F, Meirelles FV, Miglino MA, et al. Isolation and characteriza- tion of mesenchymal stem cells from the yolk sacs of bovine embryos. Theriogenol- ogy 2015;84:887–898. 64. Wenceslau CV, Miglino MA, Martins DS, Ambrosio CE, Lizier NF, Pignatari GC, Kerkis I. Mesenchymal progenitor cells from canine fetal tissues: Yolk sac, liver, and bone marrow. Tissue Eng A 2011;17:2165–2176. 65. Garikipati VN, Jadhav S, Pal L, Prakash P, Dikshit M, Nityanand S. Mesenchymal stem cells from fetal heart attenuate myocardial injury after infarction: An in vivo serial pinhole gated SPECT-CT study in rats. PLoS One 2014;9:e100982. 66. In ’t Anker PS, Scherjon SA, Kleijburg-van der Keur C, Noort WA, Claas FH, Willemze R, Fibbe WE, Kanhai HH. Amniotic fluid as a novel source of mesenchy- mal stem cells for therapeutic transplantation. Blood 2003;102:1548–1549. 67. Filioli Uranio M, Valentini L, Lange-Consiglio A, Caira M, Guaricci AC, L’Abbate A, Catacchio CR, Ventura M, Cremonesi F, Dell’Aquila ME. Isolation, proliferation, cytogenetic, and molecular characterization and in vitro differentiation potency of canine stem cells from foetal adnexa: A comparative study of amniotic fluid, amnion, and umbilical cord matrix. Mol Reprod Dev 2011;78:361–373. 68. Weber B, Kehl D, Bleul U, Behr L, Sammut S, Frese L, Ksiazek A, Achermann J, Stranzinger G, Robert J, et al. In vitro fabrication of autologous living tissue- engineered vascular grafts based on prenatally harvested ovine amniotic fluid- derived stem cells. J Tissue Eng Regen Med 2016;10:52–70. 69. Iacono E, Brunori L, Pirrone A, Pagliaro PP, Ricci F, Tazzari PL, Merlo B. Isolation, characterization and differentiation of mesenchymal stem cells from amniotic fluid, umbilical cord blood and Wharton’s jelly in the horse. Reproduction 2012;143: 455–468. 70. Marcus AJ, Coyne TM, Rauch J, Woodbury D, Black IB. Isolation, characterization, and differentiation of stem cells derived from the rat amniotic membrane. Differenti- ation 2008;76:130–144. 71. Seo MS, Park SB, Kim HS, Kang JG, Chae JS, Kang KS. Isolation and characterization of equine amniotic membrane-derived mesenchymal stem cells. J Vet Sci 2013;14: 151–159. 72. Lange-Consiglio A, Corradetti B, Bertani S, Notarstefano V, Perrini C, Marini MG, Arrighi S, Bosi G, Belloli A, Pravettoni D, et al. Peculiarity of porcine amniotic mem- brane and its derived cells: A contribution to the study of cell therapy from a large animal model. Cell Reprogram 2015;17:472–483. 73. Vellasamy S, Sandrasaigaran P, Vidyadaran S, George E, Ramasamy R. Isolation and characterisation of mesenchymal stem cells derived from human placenta tissue. World J Stem Cells 2012;4:53–61. 74. Futami I, Ishijima M, Kaneko H, Tsuji K, Ichikawa-Tomikawa N, Sadatsuki R, Muneta T, Arikawa-Hirasawa E, Sekiya I, Kaneko K. Isolation and characterization of multipotential mesenchymal cells from the mouse synovium. PLoS One 2012;7: e45517. 75. Yoshimura H, Muneta T, Nimura A, Yokoyama A, Koga H, Sekiya I. Comparison of rat mesenchymal stem cells derived from bone marrow, synovium, periosteum, adi- pose tissue, and muscle. Cell Tissue Res 2007;327:449–462. 76. Prado AA, Favaron PO, da Silva LC, Baccarin RY, Miglino MA, Maria DA. Characterization of mesenchymal stem cells derived from the equine synovial fluid and membrane. BMC Vet Res 2015;11:281. 77. Shintani N, Hunziker EB. Chondrogenic differentiation of bovine synovium: Bone morphogenetic proteins 2 and 7 and transforming growth factor beta1 induce the formation of different types of cartilaginous tissue. Arthritis Rheum 2007;56: 1869–1879. 78. Klyushnenkova E, Mosca JD, Zernetkina V, Majumdar MK, Beggs KJ, Simonetti DW, Deans RJ, McIntosh KR. T cell responses to allogeneic human mesenchymal stem cells: Immunogenicity, tolerance, and suppression. J Biomed Sci 2005;12: 47–57. 79. Liu D, Wang Y, Ye Y, Yin G, Chen L. Distinct molecular basis for endothelial differen- tiation: Gene expression profiles of human mesenchymal stem cells versus umbilical vein endothelial cells. Cell Immunol 2014;289:7–14. 80. Alimperti S, You H, George T, Agarwal SK, Andreadis ST. Cadherin-11 regulates both mesenchymal stem cell differentiation into smooth muscle cells and the devel- opment of contractile function in vivo. J Cell Sci 2014;127:2627–2638. 81. Chang YP, Hong HP, Lee YH, Liu IH. The canine epiphyseal-derived mesenchymal stem cells are comparable to bone marrow derived-mesenchymal stem cells. J Vet Med Sci 2015;77:273–280. 82. O’Driscoll SW, Saris DB, Ito Y, Fitzimmons JS. The chondrogenic potential of perios- teum decreases with age. J Orthop Res 2001;19:95–103. 83. Chang H, Docheva D, Knothe UR, Knothe Tate ML. Arthritic periosteal tissue from joint replacement surgery: A novel, autologous source of stem cells. Stem Cells Transl Med 2014;3:308–317. 84. Kisiel AH, McDuffee LA, Masaoud E, Bailey TR, Esparza Gonzalez BP, Nino-Fong R. Isolation, characterization, and in vitro proliferation of canine mesenchymal stem cells derived from bone marrow, adipose tissue, muscle, and periosteum. Am J Vet Res 2012;73:1305–1317. 85. Rui YF, Lui PP, Li G, Fu SC, Lee YW, Chan KM. Isolation and characterization of multipotent rat tendon-derived stem cells. Tissue Eng A 2010;16:1549–1558. 86. Feisst V, Brooks AE, Chen CJ, Dunbar PR. Characterization of mesenchymal progen- itor cell populations directly derived from human dermis. Stem Cells Dev 2014;23: 631–642. 87. Forni MF, Ramos Maia Lobba A, Pereira Ferreira AH, Sogayar MC. Simultaneous isolation of three different stem cell populations from murine skin. PLoS One 2015; 10:e0140143. 88. Jahroomishirazi R, Bader A, Ebert S, Schmidt C, Sedaghati B, Schulz-Siegmund M, Zscharnack M. Isolation and characterization of CD271(1) stem cells derived from sheep dermal skin. Cells Tissues Organs 2014;200:141–152. 89. Miura M, Gronthos S, Zhao M, Lu B, Fisher LW, Robey PG, Shi S. SHED: Stem cells from human exfoliated deciduous teeth. Proc Natl Acad Sci USA 2003;100:5807– 5812. 90. Mensing N, Gasse H, Hambruch N, Haeger JD, Pfarrer C, Staszyk C. Isolation and characterization of multipotent mesenchymal stromal cells from the gingiva and the periodontal ligament of the horse. BMC Vet Res 2011;7:42. 91. Beane OS, Fonseca VC, Cooper LL, Koren G, Darling EM. Impact of aging on the regenerative properties of bone marrow-, muscle-, and adipose-derived mesenchy- mal stem/stromal cells. PLoS One 2014;9:e115963. 92. Krampera M, Sartoris S, Liotta F, Pasini A, Angeli R, Cosmi L, Andreini A, Mosna F, Bonetti B, Rebellato E, et al. Immune regulation by mesenchymal stem cells derived from adult spleen and thymus. Stem Cells Dev 2007;16:797–810. 93. Moreno R, Martinez-Gonzalez I, Rosal M, Farwati A, Gratacos E, Aran JM. Charac- terization of mesenchymal stem cells isolated from the rabbit fetal liver. Stem Cells Dev 2010;19:1579–1588. 94. Todorov P, Hristova E, Konakchieva R, Michova A, Dimitrov J. Comparative studies of different cryopreservation methods for mesenchymal stem cells derived from human fetal liver. Cell Biol Int 2010;34:455–462. 95. Lu T, Hu P, Su X, Li C, Ma Y, Guan W. Isolation and characterization of mes- enchymal stem cells derived from fetal bovine liver. Cell Tissue Bank 2014;15: 439–450. 96. Hu Y, Liao L, Wang Q, Ma L, Ma G, Jiang X, Zhao RC. Isolation and identification of mesenchymal stem cells from human fetal pancreas. J Lab Clin Med 2003;141: 342–349. 97. Sordi V, Malosio ML, Marchesi F, Mercalli A, Melzi R, Giordano T, Belmonte N, Ferrari G, Leone BE, Bertuzzi F, et al. Bone marrow mesenchymal stem cells express a restricted set of functionally active chemokine receptors capable of promoting migration to pancreatic islets. Blood 2005;106:419–427. Review Article Cytometry Part A 00A: 00À00, 2017 15
  • 16. 98. Moniri MR, Sun XY, Rayat J, Dai D, Ao Z, He Z, Verchere CB, Dai LJ, Warnock GL. TRAIL-engineered pancreas-derived mesenchymal stem cells: Characterization and cytotoxic effects on pancreatic cancer cells. Cancer Gene Ther 2012;19:652–658. 99. Guo XR, Wang XL, Li MC, Yuan YH, Chen Y, Zou DD, Bian LJ, Li DS. PDX-1 mRNA-induced reprogramming of mouse pancreas-derived mesenchymal stem cells into insulin-producing cells in vitro. Clin Exp Med 2015;15:501–509. 100. Collins JJ, Mobius MA, Thebaud B. Isolation of CD1461 Resident Lung Mesenchy- mal Stromal Cells from Rat Lungs. J Visual Exp 2016. 101. Hu P, Pu Y, Li X, Zhu Z, Zhao Y, Guan W, Ma Y. Isolation, in vitro culture and identification of a new type of mesenchymal stem cell derived from fetal bovine lung tissues. Mol Med Rep 2015;12:3331–3338. 102. Eslaminejad MB, Nadri S. Murine mesenchymal stem cell isolated and expanded in low and high density culture system: Surface antigen expression and osteogenic cul- ture mineralization. In Vitro Cell Dev Biol Anim 2009;45:451–459. 103. Haack-Sorensen M, Hansen SK, Hansen L, Gaster M, Hyttel P, Ekblond A, Kastrup J. Mesenchymal stromal cell phenotype is not influenced by confluence during cul- ture expansion. Stem Cell Rev 2013;9:44–58. 104. Ren J, Wang H, Tran K, Civini S, Jin P, Castiello L, Feng J, Kuznetsov SA, Robey PG, Sabatino M, et al. Human bone marrow stromal cell confluence: Effects on cell characteristics and methods of assessment. Cytotherapy 2015;17:897–911. 105. Sanjurjo-Rodriguez C, Castro-Vinuelas R, Hermida-Gomez T, Fernandez-Vazquez T, Fuentes-Boquete IM, de Toro-Santos FJ, Diaz-Prado SM, Blanco-Garcia FJ. Ovine mesenchymal stromal cells: Morphologic, phenotypic and functional charac- terization for osteochondral tissue engineering. PLoS One 2017;12:e0171231. 106. Zscharnack M, Hepp P, Richter R, Aigner T, Schulz R, Somerson J, Josten C, Bader A, Marquass B. Repair of chronic osteochondral defects using predifferentiated mesenchymal stem cells in an ovine model. Am J Sports Med 2010;38:1857–1869. 107. Adamzyk C, Emonds T, Falkenstein J, Tolba R, Jahnen-Dechent W, Lethaus B, Neuss S. Different culture media affect proliferation, surface epitope expression, and differentiation of ovine MSC. Stem Cells Int 2013;2013:387324. 108. Halfon S, Abramov N, Grinblat B, Ginis I. Markers distinguishing mesenchymal stem cells from fibroblasts are downregulated with passaging. Stem Cells Dev 2011; 20:53–66. 109. Bonab MM, Alimoghaddam K, Talebian F, Ghaffari SH, Ghavamzadeh A, Nikbin B. Aging of mesenchymal stem cell in vitro. BMC Cell Biol 2006;7:14. 110. Izadpanah R, Kaushal D, Kriedt C, Tsien F, Patel B, Dufour J, Bunnell BA. Long-term in vitro expansion alters the biology of adult mesenchymal stem cells. Cancer Res 2008;68:4229–4238. 111. Rosland GV, Svendsen A, Torsvik A, Sobala E, McCormack E, Immervoll H, Mysliwietz J, Tonn JC, Goldbrunner R, Lonning PE, et al. Long-term cultures of bone marrow-derived human mesenchymal stem cells frequently undergo sponta- neous malignant transformation. Cancer Res 2009;69:5331–5339. 112. Miura M, Miura Y, Padilla-Nash HM, Molinolo AA, Fu B, Patel V, Seo BM, Sonoyama W, Zheng JJ, Baker CC, et al. Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation. Stem Cells 2006;24:1095–1103. 113. Foudah D, Redaelli S, Donzelli E, Bentivegna A, Miloso M, Dalpra L, Tredici G. Monitoring the genomic stability of in vitro cultured rat bone-marrow-derived mesenchymal stem cells. Chromosome Res 2009;17:1025–1039. 114. Tamadon A, Mehrabani D, Zarezadeh Y, Rahmanifar F, Dianatpour M, Zare S. Caprine endometrial mesenchymal stromal stem cell: Multilineage potential, char- acterization, and growth kinetics in breeding and anestrous stages. Vet Med Int 2017;2017:5052801. 115. Le Blanc K, Ringden O. Immunomodulation by mesenchymal stem cells and clini- cal experience. J Intern Med 2007;262:509–525. 116. Ozbey O, Sahin Z, Acar N, Ustunel I. Distribution of CD105 and CD166 positive cells in the proximal epiphysis of developing rat humerus. Histol Histopathol 2010; 25:1437–1445. 117. Bruder SP, Horowitz MC, Mosca JD, Haynesworth SE. Monoclonal antibodies reac- tive with human osteogenic cell surface antigens. Bone 1997;21:225–235. 118. Majumdar MK, Keane-Moore M, Buyaner D, Hardy WB, Moorman MA, McIntosh KR, Mosca JD. Characterization and functionality of cell surface molecules on human mesenchymal stem cells. J Biomed Sci 2003;10:228–241. 119. Wang HS, Hung SC, Peng ST, Huang CC, Wei HM, Guo YJ, Fu YS, Lai MC, Chen CC. Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord. Stem Cells 2004;22:1330–1337. 120. Nishikiori R, Watanabe K, Kato K. Antibody arrays for quality control of mesenchy- mal stem cells. ACS Appl Mater Interfaces 2015;7:16828–16836. 121. Bartholomew A, Sturgeon C, Siatskas M, Ferrer K, McIntosh K, Patil S, Hardy W, Devine S, Ucker D, Deans R, et al. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol 2002;30: 42–48. 122. Fazeli Z, Omrani MD, Ghaderian SM. CD29/CD184 expression analysis provides a signature for identification of neuronal like cells differentiated from PBMSCs. Neurosci Lett 2016;630:189–193. 123. Stock P, Bruckner S, Ebensing S, Hempel M, Dollinger MM, Christ B. The genera- tion of hepatocytes from mesenchymal stem cells and engraftment into murine liver. Nat Protoc 2010;5:617–627. 124. Sousa BR, Parreira RC, Fonseca EA, Amaya MJ, Tonelli FM, Lacerda SM, Lalwani P, Santos AK, Gomes KN, Ulrich H, et al. Human adult stem cells from diverse origins: An overview from multiparametric immunophenotyping to clinical appli- cations. Cytometry Part A 2014;85A:43–77. 125. Soland MA, Bego M, Colletti E, Zanjani ED, St Jeor S, Porada CD, Almeida-Porada G. Mesenchymal stem cells engineered to inhibit complement-mediated damage. PLoS One 2013;8:e60461. 126. Delorme B, Ringe J, Gallay N, Le Vern Y, Kerboeuf D, Jorgensen C, Rosset P, Sensebe L, Layrolle P, Haupl T, et al. Specific plasma membrane protein phenotype of culture-amplified and native human bone marrow mesenchymal stem cells. Blood 2008;111:2631–2635. 127. Barberini DJ, Freitas NP, Magnoni MS, Maia L, Listoni AJ, Heckler MC, Sudano MJ, Golim MA, da Cruz Landim-Alvarenga F, Amorim RM. Equine mesenchymal stem cells from bone marrow, adipose tissue and umbilical cord: Immunopheno- typic characterization and differentiation potential. Stem Cell Res Ther 2014;5:25. 128. Burk J, Badylak SF, Kelly J, Brehm W. Equine cellular therapy–from stall to bench to bedside?. Cytometry Part A 2013;83A:103–113. 129. Godoy RF, Alves AL, Gibson AJ, Lima EM, Goodship AE. Do progenitor cells from different tissue have the same phenotype? Res Vet Sci 2014;96:454–459. 130. Tian Y, Tao L, Zhao S, Tai D, Liu D, Liu P. Isolation and morphological characteri- zation of ovine amniotic fluid mesenchymal stem cells. Exp Anim 2016;65:125–134. 131. McCarty RC, Gronthos S, Zannettino AC, Foster BK, Xian CJ. Characterisation and developmental potential of ovine bone marrow derived mesenchymal stem cells. J Cell Physiol 2009;219:324–333. 132. Desantis S, Accogli G, Zizza S, Mastrodonato M, Blasi A, Francioso E, Rossi R, Crovace A, Resta L. Ultrastructural study of cultured ovine bone marrow-derived mesenchymal stromal cells. Ann Anat 2015;201:43–49. 133. Noort WA, Oerlemans MI, Rozemuller H, Feyen D, Jaksani S, Stecher D, Naaijkens B, Martens AC, Buhring HJ, Doevendans PA, et al. Human versus porcine mesen- chymal stromal cells: Phenotype, differentiation potential, immunomodulation and cardiac improvement after transplantation. J Cell Mol Med 2012;16:1827–1839. 134. Mohsin S, Troupes CD, Starosta T, Sharp TE, Agra EJ, Smith S, Duran JM, Zalavadia N, Zhou Y, Kubo H, et al. Unique features of cortical bone stem cells associated with repair of the injured heart. Circ Res 2015;117:1024–1033. 135. Wang PP, Wang JH, Yan ZP, Hu MY, Lau GK, Fan ST, Luk JM. Expression of hepatocyte-like phenotypes in bone marrow stromal cells after HGF induction. Bio- chem Biophys Res Commun 2004;320:712–716. 136. Shichinohe H, Kuroda S, Yano S, Hida K, Iwasaki Y. Role of SDF-1/CXCR4 system in survival and migration of bone marrow stromal cells after transplantation into mice cerebral infarct. Brain Res 2007;1183:138–147. 137. Wynn RF, Hart CA, Corradi-Perini C, O’Neill L, Evans CA, Wraith JE, Fairbairn LJ, Bellantuono I. A small proportion of mesenchymal stem cells strongly expresses functionally active CXCR4 receptor capable of promoting migration to bone mar- row. Blood 2004;104:2643–2645. 138. Ortiz LA, Gambelli F, McBride C, Gaupp D, Baddoo M, Kaminski N, Phinney DG. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci USA 2003;100: 8407–8411. 139. Pelekanos RA, Li J, Gongora M, Chandrakanthan V, Scown J, Suhaimi N, Brooke G, Christensen ME, Doan T, Rice AM, et al. Comprehensive transcriptome and immu- nophenotype analysis of renal and cardiac MSC-like populations supports strong congruence with bone marrow MSC despite maintenance of distinct identities. Stem Cell Res 2012;8:58–73. 140. Hegyi B, Sagi B, Kovacs J, Kiss J, Urban VS, Meszaros G, Monostori E, Uher F. Identical, similar or different? Learning about immunomodulatory function of mesenchymal stem cells isolated from various mouse tissues: Bone marrow, spleen, thymus and aorta wall. Int Immunol 2010;22:551–559. 141. Lei J, Hui D, Huang W, Liao Y, Yang L, Liu L, Zhang Q, Qi G, Song W, Zhang Y, et al. Heterogeneity of the biological properties and gene expression profiles of murine bone marrow stromal cells. Int J Biochem Cell Biol 2013;45:2431–2443. 142. de Witte SFH, Lambert EE, Merino A, Strini T, Douben H, O’Flynn L, Elliman SJ, de Klein A, Newsome PN, Baan CC, et al. Aging of bone marrow- and umbilical cord-derived mesenchymal stromal cells during expansion. Cytotherapy 2017;19: 798–807. 143. Gotherstrom C. Immunomodulation by multipotent mesenchymal stromal cells. Transplantation 2007;84:S35–S37. 144. Le Blanc K, Ringden O. Immunobiology of human mesenchymal stem cells and future use in hematopoietic stem cell transplantation. Biol Blood Marrow Transpl 2005;11:321–334. 145. Liechty KW, MacKenzie TC, Shaaban AF, Radu A, Moseley AM, Deans R, Marshak DR, Flake AW. Human mesenchymal stem cells engraft and demonstrate site- specific differentiation after in utero transplantation in sheep. Nat Med 2000;6: 1282–1286. 146. Wang A, Brown EG, Lankford L, Keller BA, Pivetti CD, Sitkin NA, Beattie MS, Bresnahan JC, Farmer DL. Placental mesenchymal stromal cells rescue ambulation in ovine myelomeningocele. Stem Cells Transl Med 2015;4:659–669. 147. Long C, Lankford L, Kumar P, Grahn R, Borjesson DL, Farmer D, Wang A. Isolation and characterization of canine placenta-derived mesenchymal stromal cells for the treatment of neurological disorders in dogs. Cytometry A; 2017 Jul 17. DOI: 10.1002/cyto.a.23171. [Epub ahead of print] 148. Berglund AK, Schnabel LV. Allogeneic major histocompatibility complex- mismatched equine bone marrow-derived mesenchymal stem cells are targeted for death by cytotoxic anti-major histocompatibility complex antibodies. Equine Vet J 2017;49:539–544. 149. Pezzanite LM, Fortier LA, Antczak DF, Cassano JM, Brosnahan MM, Miller D, Schnabel LV. Equine allogeneic bone marrow-derived mesenchymal stromal cells elicit antibody responses in vivo. Stem Cell Res Ther 2015;6:54. 150. Kol A, Wood JA, Carrade Holt DD, Gillette JA, Bohannon-Worsley LK, Puchalski SM, Walker NJ, Clark KC, Watson JL, Borjesson DL. Multiple intravenous injections of allogeneic equine mesenchymal stem cells do not induce a systemic inflammatory response but do alter lymphocyte subsets in healthy horses. Stem Cell Res Ther 2015;6:73. Review Article 16 Cross-species MSC