MIEPEI 2021 2022 Artigo 1

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Research in Veterinary Science 118 (2018) 317–323

Contents lists available at ScienceDirect

Research in Veterinary Science


journal homepage: www.elsevier.com/locate/rvsc

Review

Concepts and challenges in the use of mesenchymal stem cells as a treatment T


for cartilage damage in the horse
Mohammed Zayeda,1, Steve Adaira, Tena Ursinia, James Schumachera, Nabil Miskb,

Madhu Dhara,
a
Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
b
Department of Animal Surgery, College of Veterinary Medicine, Assuit University, 71526 Assuit, Egypt

A R T I C LE I N FO A B S T R A C T

Keywords: Osteoarthritis (OA), the most common form of joint disease affecting humans and horses, is characterized by the
Regenerative medicine advance and decline of cartilage and loss of function of the affected joint. The progression of OA is steadily
Mesenchymal stem cell accompanied with biochemical events, which interfere with the cytokines and proteolytic enzymes responsible
Cartilage for progress of the disease. Recently, regenerative therapies have been used with an assumption that me-
Osteoarthritis
senchymal stem cells (MSCs) possess the potential to prevent the advancement of cartilage damage and po-
Horse
Review
tentially regenerate the injured tissue with an ultimate goal of preventing OA. We believe that despite various
challenges, the use of allogenic versus autologous MSCs in cartilage regeneration, is a major issue which can
directly or indirectly affect the other factors including, the timing of implantation, dose or cell numbers for
implantation, and the source of MSCs. Current knowledge reporting some of these challenges that the clinicians
might face in the treatment of cartilage damage in horses are presented. In this regard we conducted two
independent studies. In the first study we compared donor matched bone marrow and synovial fluid – derived
equine MSCs in vitro, and showed that the SFMSCs were similar to the BMMSCs in their proliferation, expression
of CD29, CD44 and CD90, but, exhibited a significantly different chondrogenesis. Additionally, 3.2–21% of all
SFMSCs were positive for MHC II, whereas, BMMSCs were negative. In the second study we observed that
injection of both the autologous and allogenic SFMSCs into the tarsocrural joint resulted in elevated levels of
total protein and total nucleated cell counts. Further experiments to evaluate the in vivo acute or chronic re-
sponse to allogenic or autologous MSCs are imperative.

1. Introduction the regeneration of cartilage, the outcome is not very effective. The
defects characteristically heal with fibrous tissue, rather than hyaline
The inflammation of the synovial membrane leads to synovial cell cartilage, and hence, the biochemical and biomechanical qualities of
proliferation, pain, chronic dysfunction, and it initiates the deteriora- the repaired cartilage are suboptimal (Jackson et al., 2001; Buckwalter,
tion of the cartilage in horses (McIlwraith CW, 1996). If not treated or 2002). As a result, there is still an unmet need to identify an efficient
controlled, cartilage degeneration progresses and results in osteoar- and efficacious strategy to control the progression of cartilage de-
thritis (OA). Currently, there are many options for treating OA in gradation and the subsequent treatment of OA in horses.
equids. These include systemic or intra-articular administration of non-
steroidal anti-inflammatory drugs (NSAIDs), hyaluronan (HA), corti- 2. Articular cartilage
costeroids (CSs), polysulphated glycosaminoglycan, articular resurfa-
cing techniques, including microfracture, autologous chondrocytes Articular cartilage, the most abundant type of cartilage found in
implantation, and osteochondral autologous graft procedures (Frisbie horses, is composed of flexible highly specified connective tissue. This
et al., 2003; Litzke et al., 2004). The poor intrinsic capability of ar- connective tissue composed of a dense matrix of collagen fibers and
ticular cartilage to repair makes the effective treatment of cartilaginous elastic fibers embedded in a rubbery ground substance makes the car-
defects challenging (Buckwalter, 1998). Even though various cellular tilage perfect for bearing mechanical stresses without permanent de-
transplant techniques and the use of drugs have been intended to aid in formation. Articular cartilage gets nourishment through diffusion of the


Corresponding author at: Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive Knoxville, TN 37996-4500, USA.
E-mail address: mdhar@utk.edu (M. Dhar).
1
Department of Animal Surgery, College of Veterinary Medicine, South Valley University, 83,523 Qena, Egypt.

https://doi.org/10.1016/j.rvsc.2018.03.011
Received 18 December 2017; Received in revised form 13 March 2018; Accepted 18 March 2018
0034-5288/ © 2018 Elsevier Ltd. All rights reserved.
M. Zayed et al. Research in Veterinary Science 118 (2018) 317–323

synovial fluid (Brama et al., 2000). The chondrocyte is the inhabitant using PRP have limitations due to small sample sizes or nonrandomized
cell type within articular cartilage and is responsible for synthesizing methodology (Sampson et al., 2008; Mishra et al., 2009; Rowden et al.,
and controlling the extracellular matrix. Chondrocytes depend on 2015). Hence, the use of PRP in the clinic has been limited (Sheth et al.,
anaerobic metabolism because they receive no direct supply of nu- 2012). It is difficult, however, to identify if the failure is due to the
trients from blood vessels or lymphatics (Buckwalter, 1998); hence, it actual treatment or the lack of standardized protocols, platelet se-
has minimal potential to regenerate. It is suggested that injured carti- paration techniques, and consequence measures.
lage and the associated synovial tissues cannot produce cartilaginous
tissue with the same morphologic and biomechanical properties of 4. MSCs
healthy articular cartilage (McIlwraith, 1996; Buckwalter, 1998). The
development of a joint disease, such as OA, is associated with sig- MSCs are plastic-adherent cells with a fibroblast-like morphology,
nificant alterations in the metabolism of cartilage, physiological im- have the capability of renewing themselves through cell division, and
balance of degradation, and synthesis factors of chondrocytes have ability to differentiate into bone, cartilage, and fat in vitro. MSCs
(Todhunter, 1996). originally are considered to be a type of pericyte or adventitial cells
(Krampera et al., 2007; Caplan, 2017). When implanted in vivo, MSCs
3. Treatment options for OA provide an immunomodulatory and anti-inflammatory effects on the
injured tissue (Asari et al., 2009). MSCs supply growth factors, re-
The use of NSAIDs and CSs as a conservative treatment option can generative cells, and the extra-cellular matrix necessary for cartilage
be used to inhibit the inflammation but cannot prevent the progression repair. When MSCs are administered intra-articularly, MSCs will adhere
of OA or regenerate the lost tissues (Goodrich and Nixon, 2006). The to injured tissues, where they may potentially undergo differentiation
healing of cartilage through the regeneration of hyaline tissue, rather or may induce the endogenous progenitor cells to differentiate, thereby,
than the production of fibrocartilage, should keep the damaged joint regenerating the injured articular cartilage (Koch et al., 2008).
with the best strength. Surgical approaches currently used to regenerate Bone marrow is a common source of MSCs (BM) (Govoni, 2015), but
damaged articular cartilage are complex and not very efficacious. One MSCs have also been derived from adipose tissue (AT) (Vidal and Lopez,
of these techniques is bone marrow stimulation, which includes tech- 2011), umbilical cord blood (UCB) (Tessier et al., 2015), peripheral
niques of abrasion chondroplasty (Edward et al., 2007), debridement blood (Dhar et al., 2012), synovium and synovial fluid (Murata et al.,
down into subchondral bone (Story and Bramlage, 2004), and sub- 2014). Synovial fluid-derived MSCs (SFMSCs) have been reported to
chondral drilling (Lang et al., 2009). The clinical efficiency of the ar- have features similar to the MSCs isolated from the bone marrow and
throscopy of microfracture for articular cartilage regeneration in horses the adipose tissue, and have become a valuable source of MSCs for the
has also been evaluated in an equine model of cartilage healing (Frisbie treatment of cartilage injuries (Murata et al., 2014; Prado et al., 2015;
et al., 2003). Although the clinical functionality was improved, the Mak et al., 2016; Zayed et al., 2017; Yao et al., 2018).
hyaline cartilage content was not optimal and methods to further en- Synovial fluid lubricates and nourishes articular cartilage. A de-
hance the quality of repaired cartilage are needed. crease in the quantity of synovial fluid within a joint is attributed to
Autologous chondrocyte implantation is another procedure used to inflammation, especially at the early stage of OA, which leads to the
treat full-thickness defects of articular cartilage. In an equine model, wearing of cartilage (Elsaid et al., 2005; Antonacci et al., 2012). Ad-
this procedure was used to evaluate the effect of fixed chondrocytes ditionally, synovial fluid contains important growth factors, such as
using a periosteal flap in a partial- and full-thickness cartilage defects. TGF-β1 and fibroblastic growth factor, which help in chondrogenesis
Results indicated that the cartilage defects recovered with better car- (Okazaki et al., 2001). Studies have illustrated that ovine BMMSCs co-
tilage healing scores (Nixon et al., 2011). Additionally, autologous os- cultured with normal synovial fluid express chondrogenic markers after
teochondral autografts may be substituted for small and medium-sized 2 weeks (Chen et al., 2005). Synovial fluid from injured human knee
focal chondral and osteochondral defects (Hangody et al., 2008). joints significantly affects MSCs in an in vitro model of chondrogenesis
However, clinical outcomes of regenerating fibrous tissue rather than (Zhang et al., 2008), indicating that synovial fluid is important for MSC
hyaline cartilage, the reduction in biochemical and biomechanical differentiation.
qualities, the complex and invasive nature of those procedures, together Primary cultures of cells are classified as MSCs based on the prop-
with the high costs are serious drawbacks for these procedures erties displayed after they are cultured and expanded in vitro (Dominici
(Buckwalter, 2002; Cokelaere et al., 2016). et al., 2006). One of these properties is the ability to adhere to poly-
Interleukin 1 receptor antagonist protein (IRAP) that neutralizes the styrene surfaces and establish colonies of cells (Friedenstein et al.,
destructive effects of IL-1 (DeForge et al., 1992). IRAP was developed to 1970). Another is their capacity to self-renew through mitosis (Qiu
counteract IL-1 that is produced in the traumatized joint. IRAP works by et al., 2016). Finally, under specific cell culture medium conditions,
preventing IL-1 binding to the IL-1 receptors, therefore blocking the MSCs can differentiate into cell types of the mesenchymal origin (bone,
damage and inflammation caused by IL-1 (Hannum et al., 1990; fat, and cartilage) (Vidal et al., 2006; Barberini et al., 2014). At the
Eisenberg et al., 1990). Frisbie et al. injected equine IRAP gene into same time, MSCs may differentiate into a cell type of a non-mesodermal
experimentally-induced OA, resulting in significant improvement of origin (Song et al., 2006). Because MSCs are able to differentiate into
clinical and histological signs, whereas placebo-treated horses did not chondrocytes, they are a potential source of cells for regenerating in-
(Frisbie et al., 2002). IRAP however, cannot regenerate lost tissues. jured cartilage tissue in clinical cases (Seo et al., 2014).
Platelet-rich plasma (PRP) is plasma enriched with platelets relative Differentiation of MSCs into chondrocytes is a complex process.
to the whole blood. It has been demonstrated in vitro that when platelets After MSCs are subjected to conditions that induce chondrogenesis,
within the PRP are stimulated by collagen (Harrison et al., 2011), they they begin to condense and form high-density, cellular aggregates,
release growth factors including, transforming growth factor beta (TGF- which progress into the deposition of cartilaginous extracellular matrix
β), platelet-derived growth factor, epidermal growth factor, and in- and cartilage formation (Yamashita et al., 2010) (Fig. 1). TGF-β1, β2,
sulin-like growth factor, all of which can significantly affect cartilage and β3 are the most commonly used isoforms, which serve as inducers
regeneration (Amable et al., 2013). These growth factors act synergis- of chondrocytes, and exposing MSCs to these growth factors leads to
tically to enhance the access of healthy inflammatory cells to the area of differentiation i.e. accumulation of proteoglycan and collagen type II
injury and speed the formation of connective tissue at the site of injury (Tuli et al., 2003; Wang et al., 2014). Other inducers of chondrogenesis
(van Buul et al., 2011). PRP has also been suggested to regulate the include BMP7 or BMP9 and IGF1 (Murphy et al., 2015). SRY-type HMG
action of matrix metalloproteinases to activate signal transduction box9 (Sox9) is a master regulator for chondrogenesis and stimulates
pathways (Patel et al., 2013). Most of the animal or human studies transcriptional activation of genes that develop the components of the

318
M. Zayed et al. Research in Veterinary Science 118 (2018) 317–323

Fig. 1. The phases of chondrogenesis. Mesenchymal stem cells (MSCs) were stimulated to undergo chondrogenesis in monolayer culture for 14 days in chondrogenic
medium containing TGF-β1. (A) The diagram shows the principal stages of chondrogenesis and indicates the role of Sox9, TGF-β1, FGF-2 and BMP-2 as a stimulant at
different stages during differentiation. (B) Expression of cartilage-specific marker (collagen type II) was detected by immunofluorescence in cultured MSCs; the image
demonstrates that collagen II is present throughout the section (green) and is merged with DAPI. The images illustrate the different phases of chondrogenesis
described in the referenced paper (Yamashita et al., 2010), are generated from our laboratory work. Scale bar: 100 μm. (For interpretation of the references to colour
in this figure legend, the reader is referred to the web version of this article.)

cartilage matrix, including collagen type II (Dy et al., 2012). It has been autologous cells normally takes roughly three weeks; however, it may
shown that the relative expression of collagen II, a specific marker of take longer in some patients (Watts, 2014). Due to the wide variety of
hyaline cartilage, depends on the expression of the Sox9 gene (Bi et al., patients and injuries being treated with stem cells, it is impossible to
1999; DeChiara et al., 2000). compare optimal treatment protocols. There is no appreciable literature
determining the ideal time, number of doses, or number of cells per
injection. Twenty million stem cells have become the current “cell
5. Clinical application of MSCs
dose” based on evidence of improved results (Smith, 2014); however,
doses can range from 10 to 50 million cells (Frisbie, 2014). Recently, it
Clinical application or implantation of MSCs for the treatment of
has been suggested to inject joints four to six weeks post injury or
cartilage damage can be determined by a number of factors, including
surgery to allow the inflammatory phase to pass (Frisbie, 2014); which
the dose, time of implantation, source, and the number of injections
is dependent on the clinician's judgement. More work must be done in
required (Fortier et al., 2011). Since controlled studies investigating
controlled models to determine the ideal dose and timing of injections.
each of these parameters is lacking, the clinicians use their judgement
Two studies showed that administration of allogeneic UCB or AT-
with the hope of obtaining a positive clinical outcome. Using the pa-
derived MSCs into normal joints did not cause a greater local in-
tient's own MSCs (autologous) to treat an injury or disease is believed to
flammatory response than autologous administration (Carrade et al.,
be safer than using MSCs from other donors (allogenic). One of the
2011; Pigott et al., 2013b). In contrast, Pezzanite et al., 2015 showed
major challenges in using autologous MSCs, however, is the timing.
that intradermal injection of allogenic MSCs produced antibody re-
Roughly 3 weeks are needed to expand and generate sufficient cell
sponses and could reduce the effectiveness of allogeneic MSCs injected
numbers for implantation. This is highly dependent on the tissue har-
repeatedly into the same horse (Pezzanite et al., 2015). In view of these
vest, isolation, characterization, and cell culture propagation cap-
conflicting reports, the specific in vitro and in vivo immunomodulatory
abilities. The time necessary to isolate the cells and implant them
effects of equine MSCs should be more thoroughly evaluated before
clinically might hinder the use of autologous MSCs. In addition, age,
allogeneic MSCs are used to treat horses for OA or other diseases. In
gender, and disease can also significantly affect the biological proper-
summary, several restrictions must be overcome before MSCs can be
ties and the function and thus, the efficacy of MSCs derived from spe-
used extensively for the treatment of articular cartilage damage. Vari-
cific tissue sources (Siegel et al., 2013). Using allogeneic, “off-the-
able reports regarding safety, limited clinical data to support efficacy,
shelf,” in vitro characterized MSCs alleviates some of these challenges.
and excessive differences in patient reaction must be addressed (Fink,
Using allogenic MSCs that have been expanded in culture, proven to
2009).
undergo tri- lineage differentiation, and immunophenotyped to express
specific protein markers (CD29, 44, 90, 105) may be more efficacious
than using autologous MSCs, which are rarely characterized prior to 6. Potential mechanism(s) of action of MSCs in cartilage repair
therapeutic application. MSCs have immunosuppressive properties,
which primarily result from the lack of major histocompatibility class II As described above, MSCs can differentiate into multiple cell-
(MHC II) antigens and the inhibition of T helper cytokines (Machado lineages and strategies exist to grow them in culture; hence, the interest
et al., 2013). Even though it has been shown that MSCs lack MHC II, a in a cell-based therapeutic approach for regenerating equine cartilage
slight increase in MHC II has been observed when MSCs undergo using MSCs has increased in the past few years (Wilke et al., 2007; Koch
chondrogenesis, in vitro (Le Blanc et al., 2003), which may or may not et al., 2008; De Schauwer et al., 2013). Despite the increase in the use of
affect the in vivo function. It has been demonstrated that equine MSCs MSCs in the clinic, the exact mechanism(s) by which MSCs speed the
from different tissues modulate the function of immune cells by unique healing of the damaged tissue has not been determined. Several the-
mechanisms (Paterson et al., 2014; Carrade et al., 2014) and hence, if ories have been put forward. Research suggests that MSCs are attracted
MSCs lack the MHC II proteins and can inhibit T cell activation and to the site of the injury, adhere to the damaged tissue, and differentiate
proliferation, they can potentially be implanted allogenically. into specific cellular phenotypes, such as chondrocytes, osteocytes, or
Despite the potential for increased immunogenicity, certain limita- tenocytes, (Gruh and Martin, 2009; Prockop and Oh, 2012). Alter-
tions of autologous cells have allowed allogenic cells to remain as a natively, MSCs might function by recruitment of anti-apoptotic factors
treatment option. As an animal or a human ages, the differentiation to inhibit cell death, formation of new blood vessels, recruiting growth
potential, expansion potential, and the overall number of stem cells will factors, and anti-inflammatory proteins to the site of injury to re-
decrease (Fafián-Labora et al. 2015). Isolation and expansion of generate and suppress inflammation in the injured tissue (Yagi et al.,

319
M. Zayed et al. Research in Veterinary Science 118 (2018) 317–323

Fig. 2. Outline of the main molecular, synovial and chondral changes that occur in the synovial joint during osteoarthritis (OA). This schematic underlines the
consequence of actions which direct to chondral degradation. Due to external stimuli on the synovial joint, pro- inflammatory cytokines, inflammatory meditators
and proteolytic enzymes will be released. Subsequently, chondral changes include cartilage degradation and loss of Aggrecan, COMP and collagen type II (Col II). The
figure was generated using the information provided in the referenced paper (Bonnet and Walsh, 2005).

2010; English, 2013). concentration of aggrecan in repaired tissue treated with BMMSCs
Synovitis, inflammation of the synovial membrane, is one of the (McIlwraith et al., 2011).
earliest changes in the pathophysiology of OA of horses and is asso- Even though intra-articular injection of cells is the implantation
ciated with changes in cartilage (McIlwraith, 1996). Pro-inflammatory method, it may not be effective. Implantation of cells loaded onto
cytokines, such as IL-β1 and TNF-α, and extracellular matrix–degrading scaffolds can be considered a more realistic way to deliver and “hold”
enzymes, such as matric metalloproteinases, aggrecanases, and neuro- cells within cartilage defects. Natural or synthetic materials can be used
peptides,are generated by the inflamed synovium. These substances to generate scaffolds (Moutos and Guilak, 2008; Nöth et al., 2008).
modify the balance of catabolic and anabolic factors, leading to the Wilke et al. (2007) implanted MSCs suspended in autogenous fibrin into
breakdown of cartilage (Bonnet and Walsh, 2005) (Fig. 2). Therefore, 15-mm experimentally created osteochondral defects in the femor-
controlling or resolving synovitis may inhibit the catabolic processes opatellar joints of six young mature horses. The joints were examined
within the joint, subsequently protecting cartilage damage. Therapy to arthroscopically, and the defect was biopsied at 30 days. Repaired
resolve non-infectious synovitis of horses involves systemic adminis- cartilaginous tissue and surrounding cartilage were evaluated by his-
tration of NSAIDs, PSGAGs, and intra-articular administration of CSs, tochemical examinations, immunohistochemical analysis of collagen
HA, or ACS (Doucet et al., 2008; Frisbie et al., 2007). Suppression of type I and type II, and biochemical assay of the cartilage's matrix at
lymphocytes by intra-articularly implanted MSCs has been suggested to 8 months. Results showed that arthroscopic scores for defects implanted
be one of the mechanisms by which MSCs act to suppress inflammation. with MSCs had improved significantly after one month, and biopsies
MSCs were found to secrete soluble factors that suppress proliferation revealed that several of the defects predominantly contained collagen
of mononuclear cells in the peripheral blood, alter expression of cyto- type II. Goodrich et al., (2016) recently compared the effects of intra-
kines, and immunosuppress lymphocytes (Carrade Holt et al., 2014; articular administration of BMMSCs delivered in a scaffold of auto-
Paterson et al., 2014; Ranera et al., 2016). Recently, intra-articular logous platelet-enriched fibrin (APEF) versus administration of APEF
implantation of cord blood MSCs in an equine in vivo model of synovitis alone on healing of full-thickness chondral defects. When the experi-
resulted in a significant decrease in the concentration of neutrophils mentally created defects were evaluated one year later, the arthroscopic
and mononuclear cells within the synovial fluid, supporting the concept findings, MRI scores, histologic scores, and scores for material stiffness
that MSCs may have therapeutic potential for treating horses for OA were found to be similar between the groups. The authors concluded
(Williams et al., 2015). that adding BMMSCs to APEF did not improve repair of cartilage and
had an unexpected effect of stimulating bone formation in cartilaginous
defects. In summary, the method of implantation and the source of
7. Delivery and source of MSCs MSCs are debatable and are dependent on the clinician's assessment.

Two other factors that might be important in using MSCs in the


clinic are the method of implantation and the source of the tissue from 8. Challenges facing the clinical use of MSCs in horses
which MSCs are isolated. Frisbie et al. assessed the clinical, biochem-
ical, and histologic effects of intra-articularly administered BMMSCs Some specific safety concerns of MSCs include teratoma-like masses,
and AT stromal vascular fraction for treatment of horses with OA. A aberrant migration of cells to other parts of the body, and im-
greater improvement was seen in joints with BMMSCs relative to the AT munogenicity against implanted cells (Fink, 2009). When treating the
stromal vascular fraction or placebo treated joints (Frisbie et al., 2009). equine synovial joint, immune responses are of significant concern.
Fortier et al., using an equine model, showed that delivery of a con- Reports have shown both immunomodulatory and immunogenic re-
centrated BM aspirate improves 15-mm diameter full-thickness carti- sponses in equine synovial joints (Pigott et al., 2013a; Pigott et al.,
laginous defects and is better than the microfracture technique (Fortier 2013b). Fifteen million allogenic, autologous, or xenogeneic BMMSCs
et al., 2010). In another study, intra-articular injection of BMMSCs with injected into normal fetlock joints caused acute synovitis in all horses
HA in chondral defects of horses, created experimentally, was eval- tested (Pigott et al., 2013a). Evaluation of synovial fluid from these
uated. Results confirmed a significant increase in firmness and a trend joints revealed an increased nucleated cell count, increased total pro-
for better overall quality of repaired tissue after 12 months. Im- tein, and increased IL-6 concentrations. Allogenic and xenogeneic
munohistochemical analysis showed a significantly greater treated joints showed a 25% greater degree of inflammation than

320
M. Zayed et al. Research in Veterinary Science 118 (2018) 317–323

Table 1
Physical joint parameters, nucleated cell counts (cells/μl) (TNCC), total protein (TP), differential cell count 0–24 h after intra-articular administration of allogeneic
SFMSCs. Total is the mean ± SD.
Hours Lameness Circumference TP TNCC Monocytes Neutrophils

0 0 35 ± 1.5 < 2.5 100 ± 0 46 ± 12 9.5 ± 0.5


8 0 37 ± 1.5
24 3–4 39 ± 0.5 < 3.5 28,050 ± 5750 27 ± 9 71 ± 8.5
7.47 Fold

autologous treated joints; however, all BMMSCs treated joints exhibited Lameness examination and SF analysis were assessed before and
multiple clinical signs of synovitis. The same group reported sig- after injection of three millions of cells (in 2 mL PBS) inside the tarso-
nificantly greater numbers and distribution of mononuclear in- crural (TC) joint for 72 h. Lameness examinations were performed at
flammatory cells in BMMSCs treated fetlocks compared to the control the walk and trot on hard ground every 24 h. Lameness was subjectively
synovium after 60 days (Pigott et al., 2013b). This brings into question graded (0–5) according to the AAEP lameness scale. Joints were sub-
the use of MSCs in diseased osteoarthritic joints, which are by definition jectively graded every 12 h for effusion (0 = none, 1 = mild,
already inflamed. Despite the use of multiple sensitive techniques no 2 = moderate 3 = severe). Joint circumference (cm2) was measured
BMMSCs were detected 60 days post injection. In another study, when every 12 h. Each SF sample was examined for routine cytological ana-
horses with stifle injuries were injected with autologous BMMSCs, 75% lysis, which included total nucleated cell count (TNCC) (Coulter Z2
returned to some level of athletic performance, reportedly higher than nucleated cell counter), total protein (TP), and differential cell count.
previous reports of 60–63% after arthroscopy alone. However, this in- From these numbers, absolute numbers of neutrophils and mononuclear
cluded only 33 horses of which almost 10% showed evidence of post cells were calculated.
injection synovitis (Ferris et al., 2014). Following injection of autologous and allogenic SFMSCs into the TC
Intra-articular injections of culture-expanded BMMSCs to a normal joint, there was a moderate increase in lameness (range 3–4). Compared
horse joint were performed at week 0 and week 4 by Joswig's group. to control joints, injection of both the autologous and allogenic SFMSCs
Results showed that repeated intra-articular injections of allogeneic into the TC joint resulted in elevated levels of TP and TNCC. Most
MSCs resulted in an undesirable clinical response, suggesting there is importantly, there was a 7.47 fold significant increase in neutrophil
immune recognition of allogeneic MSCs upon a second exposure count in the allogenic cells (Table 1) and a 2.1 fold increase in the joints
(Joswig et al., 2017). In contrast, Ardanaz et al. administrated single that received autologous cells (Table 2). This result was unexpected,
and repeat doses of autologous or of pooled allogeneic MSCs in healthy and the results of this study didn't support our hypotheses that intra-
equine joints, and showed an absence of hypersensitivity response to articular administration of equine SFMSCs could be implanted in an
the second allogeneic BMMSCs injection (Ardanaz et al., 2016). Re- autologous manner and would result in no adverse reactions, either at
cently, Colbath and colleagues showed that allogeneic and autologous the injection site or systemically in the animal. In addition, This pre-
BMMSCs appear to modulate inflammatory processes related to acute or liminary study reinforces the fact that it is preferable to characterize
chronic musculoskeletal injuries in the horse with equal potency MSCs in vitro before implantation. Even with the in vitro knowledge,
(Colbath et al., 2017). Thus, the use of autologous or allogenic MSCs is however, further work is needed to investigate allogenic or autologous
still under debate and more studies are required. administration of equine SFMSCs in OA joints.

10. Conclusion
9. Intra-articular administration of autologous versus allogenic
SFMSCs Despite recent advances in MSC biology in basic scientific and
clinical societies, there are still a lot of unanswered questions.
As described above, allogeneic versus autologous MSCs is a hot topic Optimization of these cell-based therapies will concentrate on cellular
in veterinary regenerative medicine, debating whether there is a dif- origin, isolation, enrichment, and processing as well as on the timing,
ference in efficacy for tissue healing and if there is a correlation with route of administration, formulation, and dosing. The issue of safety of
the immune system. It has been recently suggested that assays to allogeneic or even autologous MSCs is also very important.
evaluate cell-mediated and humoral immune responses against allo- Development of sound controlled clinical trials will be principal in in-
geneic MSCs should be performed to improve the efficacy of MSCs terpreting and identifying the ideal treatment regimens of MSC therapy
while maintaining their safety (Berglund et al. 2017). While these as- for joint disorders.
says are important, it defeats one of the main objectives of having an
allogenic source of cells, which is to provide the clinic with char- Acknowledgements
acterized usable MSCs in a timely manner. Hence, further investigations
are required to fully understand the allogenicity of MSCs. We acknowledge the assistance of Ms. Amanda Kelli Hand in
We recently carried out two independent studies to understand the proofreading the article for any grammatical, typographical, or for-
biological role of MSCs by evaluating some of the challenges described matting errors. This study was funded, in a part, by the Egyptian
in the above sections. In the first study, we carried out a donor-matched
comparison of chondrogenic potential of BM and SFMSCs (Zayed et al., Table 2
2017). MSCs were evaluated in vitro for their proliferation, trilineage Physical joint parameters, nucleated cell counts (cells/μl) (TNCC), total protein
differentiation, and expression of cell surface markers. Results showed (TP), differential cell count 0–24 h after intra-articular administration of au-
that even though the SFMSCs are considered more chondrogenic re- tologous SFMSCs. Total is the mean ± SD.
lative to BMMSCs, there was considerable donor-to-donor variation in Hours Lameness Circumference TP TNCC Monocytes Neutrophils
the expression of MHC II. In the second study, we compared the in vivo
response when the SFMSCs were administered in an autologous or an 0 0 36 < 2.5 200 16 32
8 0 37
allogenic manner in normal joints. Three horses free of significant la-
24 3 38 < 4.4 17,700 30 70
menesswere involved in the study. One horse received autologous 2.1 Fold
MSCs, while the remaining two horses received allogenic MSCs.

321
M. Zayed et al. Research in Veterinary Science 118 (2018) 317–323

Cultural and Educational Bureau (ECEB) and the University of Thompson, R.C., 1990. Primary structure and functional expression from com-
Tennessee's (R181710143) Center of Excellence in Livestock Diseases plementary DNA of a human interleukin-1 receptor antagonist. Nature 343, 341–346.
Elsaid, K.A., Jay, G.D., Warman, M.L., Rhee, D.K., Chichester, C.O., 2005. Association of
and Human Health. articular cartilage degradation and loss of boundary-lubricating ability of synovial
fluid following injury and inflammatory arthritis. Arthritis Rheum. 52, 1746–1755.
References English, K., 2013. Mechanisms of mesenchymal stromal cell immunomodulation.
Immunol. Cell Biol. 91, 19–26.
Fafián-Labora, J., Fernández-Pernas, P., Fuentes, I., De Toro, J., Oreiro, N., Sangiao-
Amable, P.R., Carias, R.B., Teixeira, M.V., da Cruz Pacheco, Í., Corrêa do Amaral, R.J., Alvarellos, S., et al., 2015. Influence of age on rat bone-marrow mesenchymal stem
Granjeiro, J.M., Borojevic, R., 2013. Platelet-rich plasma preparation for regenerative cells potential. Sci. Rep. 19, 16765.
medicine: optimization and quantification of cytokines and growth factors. Stem Cell Ferris, D.J., Frisbie, D.D., Kisiday, J.D., McIlwraith, C.W., Hague, B.A., Major, M.D.,
Res Ther 4, 67. Schneider, R.K., Zubrod, C.J., Kawcak, C.E., Goodrich, L.R., 2014. Clinical outcome
Antonacci, J.M., Schmidt, T.A., Serventi, L.A., Cai, M.Z., Shu, Y.L., Schumacher, B.L., after intra-articular administration of bone marrow derived mesenchymal stem cells
McIlwraith, C.W., Sah, R.L., 2012. Effects of equine joint injury on boundary lu- in 33 horses with stifle injury. Vet. Surg. 43, 255–265.
brication of articular cartilage by synovial fluid: role of hyaluronan. Arthritis Rheum. Fink, D.W., 2009. FDA regulation of stem cell–based products. Science 324, 1662.
64, 2917–2926. Fortier, L.A., Travis, A.J., 2011. Stem cells in veterinary medicine. Stem Cell Res Ther 2,
Ardanaz, N., Vázquez, F.J., Romero, A., Remacha, A.R., Barrachina, L., Sanz, A., Ranera, 9–14.
B., Vitoria, A., Albareda, J., Prades, M., Zaragoza, P., Martín-Burriel, I., Rodellar, C., Fortier, L.A., Potter, H.G., Rickey, E.J., Schnabel, L.V., Foo, L.F., Chong, L.R., Stokol, T.,
2016. Inflammatory response to the administration of mesenchymal stem cells in an Cheetham, J., Nixon, A.J., 2010. Concentrated bone marrow aspirate improves full-
equine experimental model: effect of autologous, and single and repeat doses of thickness cartilage repair compared with microfracture in the equine model. J. Bone
pooled allogeneic cells in healthy joints. BMC Vet. Res. 12, 65. Joint Surg. 92, 1927–1937.
Asari, S., Itakura, S., Ferreri, K., Liu, C., Kuroda, Y., Kandeel, F., 2009. Mesenchymal stem Friedenstein, A.J., Chailakhjan, R.K., Lalykina, K.S., 1970. The development of fibroblast
cells suppress B-cell terminal differentiation. Exp. Hematol. 37, 604–615. colonies in monolayer cultures of Guinea-Pig bone marrow and spleen cells. Cell
Barberini, D.J., Freitas, N.P., Magnoni, M.S., Maia, L., Listoni, A.J., Heckler, M.C., Prolif. 3, 393–403.
Sudano, M.J., Golim, M.A., da Cruz Landim-Alvarenga, F., Amorim, R.M., 2014. Frisbie, D.D., 2014. How to choose cases and utilize mesenchymal stem cells in joint
Equine mesenchymal stem cells from bone marrow, adipose tissue and umbilical injury, 1Proceedings of the 60th annual convention of the American Association of
cord: immunophenotypic characterization and differentiation potential. Stem Cell Equine Practitioners, Salt Lake City, Utah, USA, 2014, (December 6-10).
Res Ther 5, 25. Frisbie, D., Ghivizzani, S., Robbins, P., Evans, C., McIlwraith, C., 2002. Treatment of
Berglund, A., Fortier, L., Antczak, D., Schnabel, L., 2017. Immunoprivileged no more: experimental equine osteoarthritis by in vivo delivery of the equine interleukin-1
measuring the immunogenicity of allogeneic adult mesenchymal stem cells. Stem Cell receptor antagonist gene. Gene Ther. 9, 12–20.
Res Ther 8, 288. Frisbie, D.D., Oxford, J.T., Southwood, L., Trotter, G.W., Rodkey, W.G., Steadman, J.R.,
Bi, W., Deng, J.M., Zhang, Z., Behringer, R.R., de Crombrugghe, B., 1999. Sox9 is required Goodnight, J.L., McIlwraith, C.W., 2003. Early events in cartilage repair after sub-
for cartilage formation. Nat. Genet. 22, 85–89. chondral bone microfracture. Clin. Orthop. Relat. Res. 407, 215–227.
Bonnet, C.S., Walsh, D.A., 2005. Osteoarthritis, angiogenesis and inflammation. Frisbie, D.D., Kawcak, C.E., Werpy, N.M., Park, R.D., McIlwraith, C.W., 2007. Clinical,
Rheumatology 44, 7–16. biochemical, and histologic effects of intra-articular administration of autologous
Brama, P.A., Tekoppele, J.M., Bank, R.A, Barneveld, A., van Weeren, P.R., 2000. conditioned serum in horses with experimentally induced osteoarthritis. Am. J. Vet.
Functional adaptation of equine articular cartilage: the formation of regional bio- Res. 68, 290–296.
chemical characteristics up to age one year. Equine Vet. J. 32 (3), 217–221. Frisbie, D.D., Kisiday, J.D., Kawcak, C.E., et al., 2009. Evaluation of adipose-derived
Buckwalter, J.A., 1998. Articular cartilage: injuries and potential for healing. J. Orthop. stromal vascular fraction or bone marrow-derived mesenchymal stem cells for
Sports Phys. Ther. 28, 192–202. treatment of osteoarthritis. J. Orthop. Res. 27, 1675–1680.
Buckwalter, J.A., 2002. Articular cartilage injuries. Clin. Orthop. Relat. Res. 21–37. Goodrich, L.R., Nixon, A.J., 2006. Medical treatment of osteoarthritis in the horse - a
Caplan, A.I., 2017. Mesenchymal stem cells: time to change the name!. Stem Cells Transl. review. Vet. J. 171 (1), 51–69.
Med. 6 (6), 1445–1451. Goodrich, L.R., Chen, A.C., Werpy, N.M., Williams, A.A., Kisiday, J.D., Su, A.W., Cory, E.,
Carrade, D.D., Owens, S.D., Galuppo, L.D., Vidal, M.A., Ferraro, G.L., Librach, F., Morley, P.S., McIlwraith, C.W., Sah, R.L., Chu, C.R., 2016. Addition of mesenchymal
Buerchler, S., Friedman, M.S., Walker, N.J., Borjesson, D.L., 2011. Clinicopathologic stem cells to autologous platelet-enhanced fibrin scaffolds in chondral defects: does it
findings following intra-articular injection of autologous and allogeneic placentally enhance repair? J. Bone Joint Surg. 98, 23–34.
derived equine mesenchymal stem cells in horses. Cytotherapy 13. Govoni, K.E., 2015. Horses species symposium: use of mesenchymal stem cells in fracture
Carrade, D.D., Wood, J.A., Granick, J.L., Walker, N.J., Clark, K.C., Borjesson, D.L., 2014. repair in horses. J. Anim. Sci. 93, 871–878.
Equine mesenchymal stem cells inhibit T cell proliferation through different me- Gruh, I., Martin, U., 2009. Transdifferentiation of stem cells: a critical view. Adv.
chanisms depending on tissue source. Stem Cells Dev. 23, 1258–1265. Biochem. Eng. Biotechnol. 114, 73–106.
Chen, J., Wang, C., Lu, S., Wu, J., Guo, X., Duan, C., Dong, L., Song, Y., Zhang, J., Jing, D., Hangody, L., Vasarhelyi, G., Hangody, L.R., Sukosd, Z., Tibay, G., Bartha, L., Bodo, G.,
Wu, L., Ding, J., Li, D., 2005. In vivo chondrogenesis of adult bone-marrow-derived 2008. Autologous osteochondral grafting—technique and long-term results. Injury
autologous mesenchymal stem cells. Cell Tissue Res. 319, 429–438. 39, 32–39.
Cokelaere, S., Malda, J., Weeren, R., 2016. Cartilage defect repair in horses: current Hannum, C.H., Wilcox, C.W., Arend, W.P., Joslin, F.G., Dripps, D.J., Heimdal, P.L., Armes,
strategies and recent developments in regenerative medicine of the equine joint with L.G., Sommer, A., Eisenberg, S.P., Thompson, R.C., 1990. Interleukin-1 receptor an-
emphasis on the surgical approach. Vet. J. 214, 61–71. tagonist activity of a human interleukin-1 inhibitor. Nature 343, 336–340.
Colbath, A.C., Dow, S.W., Phillips, J.N., McIlwraith, C.W., Goodrich, L.R., 2017. Harrison, S., Vavken, P., Kevy, S., Jacobson, M., Zurakowski, D., Murray, M.M., 2011.
Autologous and allogeneic equine mesenchymal stem cells exhibit equivalent im- Platelet activation by collagen provides sustained release of anabolic cytokines. Am.
munomodulatory properties in vitro. Stem Cells Dev. 26, 503–511. J. Sports Med. 39, 729–734.
De Schauwer, C., Van de Walle, G.R., Van Soom, A., Meyer, E., 2013. Mesenchymal stem Jackson, D.W., Lalor, P.A., Aberman, H.M., Simon, T.M., 2001. Spontaneous repair of full-
cell therapy in horses: useful beyond orthopedic injuries? Vet Q 33, 234–241. thickness defects of articular cartilage in a goat model. A preliminary study. J. Bone
DeChiara, T.M., Kimble, R.B., Poueymirou, W.T., et al., 2000. Ror2, encoding a receptor- Joint Surg. 83, 53–64.
like tyrosine kinase, is required for cartilage and growth plate development. Nat. Joswig, A.-J., Mitchell, A., Cummings, K.J., Levine, G.J., Gregory, C.A., Smith, R., et al.,
Genet. 24, 271–274. 2017. Repeated intra-articular injection of allogeneic mesenchymal stem cells causes
DeForge, L.E., Tracey, D.E., Kenney, J.S., Remick, D.G., 1992. Interleukin-1 receptor an adverse response compared to autologous cells in the equine model. Stem Cell Res
antagonist protein inhibits interleukin-8 expression in lipopolysaccharide-stimulated Ther 8, 42.
human whole blood. Am. J. Pathol. 140 (5), 1045–1054. Koch, T.G., Berg, L.C., Betts, D.H., 2008. Concepts for the clinical use of stem cells in
Dhar, M., Neilsen, N., Beatty, K., Eaker, S., Adair, H., Geiser, D., 2012. Equine peripheral equine medicine. Can. Vet. J. 49, 1009–1017.
blood-derived mesenchymal stem cells: isolation, identification, trilineage differ- Krampera, M., Franchini, M., Pizzolo, G., Aprili, G., 2007. Mesenchymal stem cells: from
entiation and effect of hyperbaric oxygen treatment. Equine Vet. J. 44, 600–605. biology to clinical use. Blood Transfus. 5, 120–129.
Dominici, M., Le, B.K., Mueller, I., Slaper-Cortenbach, I., Marini, F., et al., 2006. Minimal Lang, H.M., Panizzi, L., Allen, A.L., Woodbury, M.R., Barber, S.M., 2009. Comparison of
criteria for defining multipotent mesenchymal stromal cells. The International three drilling techniques for carpometacarpal joint arthrodesis in horses. Vet. Surg.
Society for Cellular Therapy position statement. Cytotherapy 8, 315–317. 38 (8), 990–997.
Doucet, M.Y., Bertone, A.L., Hendrickson, D., Hughes, F., Macallister, C., McClure, S., Le Blanc, K., Tammik, C., Rosendahl, Zetterberg, Ringén, O., 2003. HLA expression and
Reinemeyer, C., Rossier, Y., Sifferman, R., Vrins, A.A., White, G., Kunkle, B., Alva, R., immunologic properties of differentiated and undifferentiated mesenchymal stem
Romano, D., Hanson, P.D., 2008. Comparison of efficacy and safety of paste for- cells. Exp. Hematol. 31 (10), 890–896.
mulations of firocoxib and phenylbutazone in horses with naturally occurring os- Litzke, L.E., Wagner, E., Baumgaertner, W., Hetzel, U., Josimovic-Alasevic, O., Libera, J.,
teoarthritis. J. Am. Vet. Med. Assoc. 232, 91–97. 2004. Repair of extensive articular cartilage defects in horses by autologous chon-
Dy, P., Wang, W., Bhattaram, P., Wang, Q., Wang, L., Ballock, R.T., Lefebvre, V., 2012. drocyte transplantation. Ann. Biomed. Eng. 32, 57–69.
Sox9 directs hypertrophic maturation and blocks osteoblast differentiation of growth Machado, C.D., Telles, P.D., Nascimento, I.L.O., 2013. Immunological characteristics of
plate chondrocytes. Dev. Cell 22, 597–609. mesenchymal stem cells. Rev. Bras. Hematol. Hemoter. 35, 62–67.
Edwards, R.B., Lu, Y., Uthamanthil, R.K., Bogdanske, J.J., Muir, P., Athanasiou, K.A., Mak, J., Jablonski, C.L., Leonard, C.A., Dunn, J.F., Raharjo, E., Matyas, J.R., et al., 2016.
2007. Comparison of mechanical débridement and radiofrequency energy for chon- Intra-articular injection of synovial mesenchymal stem cells improves cartilage repair
droplasty in an in vivo equine model of partial thickness cartilage injury. Osteoarthr. in a mouse injury model. Sci. Rep. 6, 23076.
Cartil. 15, 169–178. McIlwraith, C.W., 1996. General pathobiology of the joint and response to injury. In:
Eisenberg, S.P., Evans, R.J., Arend, W.P., Verderber, E., Brewer, M.T., Hannum, C.H., McIlwraith, C.W., Trotter, G.W. (Eds.), Joint Disease in the Horse. WB Saunders,

322
M. Zayed et al. Research in Veterinary Science 118 (2018) 317–323

Philadelphia, pp. 40–70. Surg. Am. 94, 298–307.


McIlwraith, C.W., Frisbie, D.D., Rodkey, W.G., Kisiday, J.D., Werpy, N.M., Kawcak, C.E., Siegel, G., Kluba, T., Hermanutz-Klein, U., Bieback, K., Northoff, H., et al., 2013.
Steadman, J.R., 2011. Evaluation of intra-articular mesenchymal stem cells to aug- Phenotype, donor age and gender affect function of human bone marrow-derived
ment healing of microfractured chondral defects. Arthroscopy 27, 1552–1561. mesenchymal stromal cells. BMC Med. 11.
Mishra, A., Jr, Woodall J., Vieira, A., 2009. Treatment of tendon and muscle using pla- Smith, R.K.W., 2014. How to select cases and utilize stem cells in tendon injury. In:
telet-rich plasma. Clin. Sports Med. 28, 113–125. Proceedings of the 60th Annual Convention of the American Association of Equine
Moutos, F.T., Guilak, F., 2008. Composite scaffolds for cartilage tissue engineering. Practitioners, Salt Lake City, Utah, USA. Lexington, American Association of Equine
Biorheology 45, 501–512. Practitioners (AAEP) (December 6–10).
Murata, D., Miyakoshi, D., Hatazoe, T., Miura, N., Tokunaga, S., Fujiki, M., Nakayama, K., Song, L., Webb, N.E., Song, Y., Tuan, R.S., 2006. Identification and functional analysis of
Misumi, K., 2014. Multipotency of equine mesenchymal stem cells derived from sy- candidate genes regulating mesenchymal stem cell self-renewal and multipotency.
novial fluid. Vet. J. 202, 53–61. Stem Cells 24, 1707–1718.
Murphy, M.K., Huey, D.J., Hu, J.C., Athanasiou, K.A., 2015. TGF-beta1, GDF-5, and BMP- Story, M.R., Bramlage, L.R., 2004. Arthroscopic debridement of subchondral bone cysts in
2 stimulation induces chondrogenesis in expanded human articular chondrocytes and the distal phalanx of 11 horses (1994–2000). Equine Vet. J. 36 (4), 356–360.
marrow-derived stromal cells. Stem Cells 33, 762–773. Tessier, L., Bienzle, D., Williams, L.B., Koch, T.G., 2015. Phenotypic and im-
Nixon, A.J., Begum, L., Mohammed, H.O., Huibregtse, B., O'Callaghan, M.M., Matthews, munomodulatory properties of equine cord blood-derived mesenchymal stromal
G.L., 2011. Autologous chondrocyte implantation drives early chondrogenesis and cells. PLoS One 10, e0122954.
organized repair in extensive full- and partial-thickness cartilage defects in an equine Todhunter, R.J., 1996. Anatomy and physiology of synovial joints. In: CW, McIlwraith,
model. J. Orthop. Res. 29, 1121–1130. Trotter, G.W. (Eds.), Joint Disease in the Horse. 1996. WB Saunders, Philadelphia,
Nöth, U., Steinert, A.F., Tuan, R.S., 2008. Technology insight: adult mesenchymal stem pp. 1–28.
cells for osteoarthritis therapy. Nat. Clin. Pract. Rheumatol. 4, 371–380. Tuli, R., Tuli, S., Nandi, S., Huang, X., Manner, P.A., Hozack, W.J., Danielson, K.G., Hall,
Okazaki, R., Sakai, A., Uezono, Y., Ootsuyama, A., Kunugita, N., Nakamura, T., Norimura, D.J., Tuan, R.S., 2003. Transforming growth factor-beta-mediated chondrogenesis of
T., 2001. Sequential changes in transforming growth factor (TGF)-beta1 concentra- human mesenchymal progenitor cells involves N-cadherin and mitogen-activated
tion in synovial fluid and mRNA expression of TGF-beta1 receptors in chondrocytes protein kinase and Wnt signaling cross-talk. J. Biol. Chem. 278, 41227–41236.
after immobilization of rabbit knees. J. Bone Miner. Metab. 19, 228–235. van Buul, G.M., Koevoet, W.L., Kops, N., Bos, P.K., Verhaar, J.A., Weinans, H., Bernsen,
Patel, S., Dhillon, M.S., Aggarwal, S., Marwaha, N., Jain, A., 2013. Treatment with pla- M.R., van Osch, G.J., 2011. Platelet-rich plasma releasate inhibits inflammatory
telet-rich plasma is more effective than placebo for knee osteoarthritis a prospective, processes in osteoarthritic chondrocytes. Am. J. Sports Med. 39, 2362–2370.
double-blind, randomized trial. Am. J. Sports Med. 41, 356–364. Vidal, M.A., Lopez, M.J., 2011. Adipogenic differentiation of adult equine mesenchymal
Paterson, Y.Z., Rash, N., Garvican, E.R., Paillot, R., Guest, D.J., 2014. Equine mesench- stromal cells. Methods Mol. Biol. 702, 61–75.
ymal stromal cells and embryo-derived stem cells are immune privileged in vitro. Vidal, M.A., Kilroy, G.E., Johnson, J.R., Lopez, M.J., Moore, R.M., Gimble, J.M., 2006.
Stem Cell Res Ther 5, 1–13. Cell growth characteristics and differentiation frequency of adherent equine bone
Pezzanite, L.M., Fortier, L.A., Antczak, D.F., Cassano, J.M., Brosnahan, M.M., Miller, D., marrow–derived mesenchymal stromal cells: adipogenic and osteogenic capacity.
Schnabel, L.V., 2015. Equine allogeneic bone marrow-derived mesenchymal stromal Vet. Surg. 35, 601–610.
cells elicit antibody responses in vivo. Stem Cell Res Ther 6, 54. Wang, X., Li, Y., Han, R., He, C., Wang, G., Wang, J., Zheng, J., Pei, M., Wei, L., 2014.
Pigott, J.H., Ishihara, A., Wellman, M.L., Russell, D.S., Bertone, A.L., 2013a. Demineralized bone matrix combined bone marrow mesenchymal stem cells, bone
Inflammatory effects of autologous genetically modified autologous, allogeneic, and morphogenetic protein-2 and transforming growth factor-beta3 gene promoted pig
xenogeneic mesenchymal stem cells after intra-articular injection in horses. Vet. cartilage defect repair. PLoS One 9, e116061.
Comp. Orthop. Traumatol. 26, 453–460. Watts, A.E., 2014. How to understand regenerative medicine - what is it? Proceedings of
Pigott, J.H., Ishihara, A., Wellman, M.L., Russell, D.S., Bertone, A.L., 2013b. Investigation the 60th Annual Convention of the American Association of Equine Practitioners, Salt
of the immune response to autologous, allogeneic, and xenogeneic mesenchymal Lake City, Utah, USA. pp. 494–498 (December 6-10).
stem cells after intra-articular injection in horses. Vet. Immunol. Immunopathol. 156, Wilke, M.M., Nydam, D.V., Nixon, A.J., 2007. Enhanced early chondrogenesis in articular
99–106. defects following arthroscopic mesenchymal stem cell implantation in an equine
Prado, A.A., Favaron, P.O., da Silva, L.C., Baccarin, R.Y., Miglino, M.A., Maria, D.A., model. J. Orthop. Res. 25, 913–925.
2015. Characterization of mesenchymal stem cells derived from the equine synovial Williams, L.B., Koenig, J.B., Black, B., Gibson, T.W., Sharif, S., Koch, T.G., 2015. Equine
fluid and membrane. BMC Vet. Res. 11, 281. allogeneic umbilical cord blood derived mesenchymal stromal cells reduce synovial
Prockop, D.J., Oh, J.Y., 2012. Medical therapies with adult stem/progenitor cells (MSCs): fluid nucleated cell count and induce mild self-limiting inflammation when evaluated
a backward journey from dramatic results in vivo to the cellular and molecular ex- in an LPS induced synovitis model. Equine Vet. J. 48 (5), 619–625.
planations. J. Cell. Biochem. 113 (5), 1460–1469. Yagi, H., Soto-Gutierrez, A., Parekkadan, B., Kitagawa, Y., Tompkins, R.G., Kobayashi, N.,
Qiu, J., Li, D., Mou, X., Li, J., Guo, W., Wang, S., Yu, X., Ma, B., Zhang, S., Tang, W., Sang, Yarmush, M.L., 2010. Mesenchymal stem cells: mechanisms of immunomodulation
Y., Gil, P.R., Liu, H., 2016. Effects of graphene quantum dots on the self-renewal and and homing. Cell Transplant. 19, 667–679.
differentiation of mesenchymal stem cells. Adv. Healthc. Mater 5, 702–710. Yamashita, A., Nishikawa, S., Rancourt, D.E., 2010. Identification of five developmental
Ranera, B., Antczak, D., Miller, D., Doroshenkova, T., Ryan, A., McIlwraith, C.W., Barry, processes during chondrogenic differentiation of embryonic stem cells. PLoS One 5,
F., 2016. Donor-derived equine mesenchymal stem cells suppress proliferation of e10998.
mismatched lymphocytes. Equine Vet. J. 48, 253–260. Yao, Y., Yu Li, Z., Zhang, H., Zheng, Y., Mai, L., Liu, W., Zhang, Z., Sun, Y., 2018. Synovial
Rowden, A., Dominici, P., D'Orazio, J., et al., 2015. Double-blind, randomized, placebo- fluid-derived synovial fragments represent an improved source of synovial me-
controlled study evaluating the use of platelet-rich plasma therapy (PRP) for acute senchymal stem cells in the temporomandibular joint. Int. J. Mol. Med. 41 (1),
ankle sprains in the emergency department. J. Emerg. Med. 49, 546–551. 173–183.
Sampson, S., Gerhardt, M., Mandelbaum, B., 2008. Platelet rich plasma injection grafts for Zayed, M., Caniglia, C., Misk, N., Dhar, M.S., 2017. Donor-matched comparison of
musculoskeletal injuries: a review. Curr. Rev. Musculoskelet Med. 1, 165–174. Chondrogenic potential of equine bone marrow- and synovial fluid-derived me-
Seo, J.P., Tsuzuki, N., Haneda, S., Yamada, K., Furuoka, H., Tabata, Y., Sasaki, N., 2014. senchymal stem cells: implications for cartilage tissue regeneration. Front Vet Sci 3,
Osteoinductivity of gelatin/beta-tricalcium phosphate sponges loaded with different 121.
concentrations of mesenchymal stem cells and bone morphogenetic protein-2 in an Zhang, S., Muneta, T., Morito, T., Mochizuki, T., Sekiya, I., 2008. Autologous synovial
equine bone defect model. Vet. Res. Commun. 38, 73–80. fluid enhances migration of mesenchymal stem cells from synovium of osteoarthritis
Sheth, N. Simunovic, Klein, G., Fu, F., Einhorn, T.A., et al., 2012. Efficacy of autologous patients in tissue culture system. J. Orthop. Res. 26, 1413–1418.
platelet-rich plasma use for orthopaedic indications: a meta-analysis. J. Bone Joint

323

You might also like