Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article En | MEDLINE | ID: mdl-34884768

Fetal cartilage fully regenerates following injury, while in adult mammals cartilage injury leads to osteoarthritis (OA). Thus, in this study, we compared the in vivo injury response of fetal and adult ovine articular cartilage histologically and proteomically to identify key factors of fetal regeneration. In addition, we compared the secretome of fetal ovine mesenchymal stem cells (MSCs) in vitro with injured fetal cartilage to identify potential MSC-derived therapeutic factors. Cartilage injury caused massive cellular changes in the synovial membrane, with macrophages dominating the fetal, and neutrophils the adult, synovial cellular infiltrate. Correspondingly, proteomics revealed differential regulation of pro- and anti-inflammatory mediators and growth-factors between adult and fetal joints. Neutrophil-related proteins and acute phase proteins were the two major upregulated protein groups in adult compared to fetal cartilage following injury. In contrast, several immunomodulating proteins and growth factors were expressed significantly higher in the fetus than the adult. Comparison of the in vitro MSCs proteome with the in vivo fetal regenerative signature revealed shared upregulation of 17 proteins, suggesting their therapeutic potential. Biomimicry of the fetal paracrine signature to reprogram macrophages and modulate inflammation could be an important future research direction for developing novel therapeutics.


Cartilage, Articular/growth & development , Cartilage, Articular/injuries , Cell- and Tissue-Based Therapy/methods , Osteoarthritis/pathology , Regeneration/physiology , Acute-Phase Proteins/metabolism , Animals , Cells, Cultured , Fetus/physiology , Macrophages/cytology , Mesenchymal Stem Cells/metabolism , Neutrophils/cytology , Sheep , Synovial Membrane/cytology , Synovial Membrane/injuries , Synovial Membrane/metabolism
2.
Sci Rep ; 11(1): 12451, 2021 06 14.
Article En | MEDLINE | ID: mdl-34127759

For research on tendon injury, many different animal models are utilized; however, the extent to which these species simulate the clinical condition and disease pathophysiology has not yet been critically evaluated. Considering the importance of inflammation in tendon disease, this study compared the cellular and molecular features of inflammation in tenocytes of humans and four common model species (mouse, rat, sheep, and horse). While mouse and rat tenocytes most closely equalled human tenocytes' low proliferation capacity and the negligible effect of inflammation on proliferation, the wound closure speed of humans was best approximated by rats and horses. The overall gene expression of human tenocytes was most similar to mice under healthy, to horses under transient and to sheep under constant inflammatory conditions. Humans were best matched by mice and horses in their tendon marker and collagen expression, by horses in extracellular matrix remodelling genes, and by rats in inflammatory mediators. As no single animal model perfectly replicates the clinical condition and sufficiently emulates human tenocytes, fit-for-purpose selection of the model species for each specific research question and combination of data from multiple species will be essential to optimize translational predictive validity.


Tendon Injuries/immunology , Tendons/pathology , Tenocytes/immunology , Animals , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Extracellular Matrix/metabolism , Female , Horses , Humans , Inflammation/immunology , Inflammation/pathology , Mice , Primary Cell Culture , Rats , Sheep , Species Specificity , Tendon Injuries/pathology , Tendons/cytology , Tendons/immunology , Tenocytes/metabolism
3.
Int J Mol Sci ; 22(11)2021 May 25.
Article En | MEDLINE | ID: mdl-34070692

Tendinopathies are painful, disabling conditions that afflict 25% of the adult human population. Filling an unmet need for realistic large-animal models, we here present an ovine model of tendon injury for the comparative study of adult scarring repair and fetal regeneration. Complete regeneration of the fetal tendon within 28 days is demonstrated, while adult tendon defects remained macroscopically and histologically evident five months post-injury. In addition to a comprehensive histological assessment, proteome analyses of secretomes were performed. Confirming histological data, a specific and pronounced inflammation accompanied by activation of neutrophils in adult tendon defects was observed, corroborated by the significant up-regulation of pro-inflammatory factors, neutrophil attracting chemokines, the release of potentially tissue-damaging antimicrobial and extracellular matrix-degrading enzymes, and a response to oxidative stress. In contrast, secreted proteins of injured fetal tendons included proteins initiating the resolution of inflammation or promoting functional extracellular matrix production. These results demonstrate the power and relevance of our novel ovine fetal tendon regeneration model, which thus promises to accelerate research in the field. First insights from the model already support our molecular understanding of successful fetal tendon healing processes and may guide improved therapeutic strategies.


Extracellular Matrix/metabolism , Neutrophil Activation , Neutrophils/metabolism , Regeneration , Tendinopathy/metabolism , Tendons/physiology , Animals , Extracellular Matrix/pathology , Female , Fetus , Humans , Sheep , Tendinopathy/pathology
4.
Animals (Basel) ; 11(1)2021 Jan 19.
Article En | MEDLINE | ID: mdl-33477808

Musculoskeletal injuries and chronic degenerative diseases commonly affect both athletic and sedentary horses and can entail the end of their athletic careers. The ensuing repair processes frequently do not yield fully functional regeneration of the injured tissues but biomechanically inferior scar or replacement tissue, causing high reinjury rates, degenerative disease progression and chronic morbidity. Regenerative medicine is an emerging, rapidly evolving branch of translational medicine that aims to replace or regenerate cells, tissues, or organs to restore or establish normal function. It includes tissue engineering but also cell-based and cell-free stimulation of endogenous self-repair mechanisms. Some regenerative medicine therapies have made their way into equine clinical practice mainly to treat tendon injures, tendinopathies, cartilage injuries and degenerative joint disorders with promising results. However, the qualitative and quantitative spatiotemporal requirements for specific bioactive factors to trigger tissue regeneration in the injury response are still unknown, and consequently, therapeutic approaches and treatment results are diverse. To exploit the full potential of this burgeoning field of medicine, further research will be required and is ongoing. This review summarises the current knowledge of commonly used regenerative medicine treatments in equine patients and critically discusses their use.

5.
Article En | MEDLINE | ID: mdl-32903631

Rapid developments in Regenerative Medicine and Tissue Engineering has witnessed an increasing drive toward clinical translation of breakthrough technologies. However, the progression of promising preclinical data to achieve successful clinical market authorisation remains a bottleneck. One hurdle for progress to the clinic is the transition from small animal research to advanced preclinical studies in large animals to test safety and efficacy of products. Notwithstanding this, to draw meaningful and reliable conclusions from animal experiments it is critical that the species and disease model of choice is relevant to answer the research question as well as the clinical problem. Selecting the most appropriate animal model requires in-depth knowledge of specific species and breeds to ascertain the adequacy of the model and outcome measures that closely mirror the clinical situation. Traditional reductionist approaches in animal experiments, which often do not sufficiently reflect the studied disease, are still the norm and can result in a disconnect in outcomes observed between animal studies and clinical trials. To address these concerns a reconsideration in approach will be required. This should include a stepwise approach using in vitro and ex vivo experiments as well as in silico modeling to minimize the need for in vivo studies for screening and early development studies, followed by large animal models which more closely resemble human disease. Naturally occurring, or spontaneous diseases in large animals remain a largely untapped resource, and given the similarities in pathophysiology to humans they not only allow for studying new treatment strategies but also disease etiology and prevention. Naturally occurring disease models, particularly for longer lived large animal species, allow for studying disorders at an age when the disease is most prevalent. As these diseases are usually also a concern in the chosen veterinary species they would be beneficiaries of newly developed therapies. Improved awareness of the progress in animal models is mutually beneficial for animals, researchers, human and veterinary patients. In this overview we describe advantages and disadvantages of various animal models including domesticated and companion animals used in regenerative medicine and tissue engineering to provide an informed choice of disease-relevant animal models.

6.
Regen Med ; 15(6): 1775-1787, 2020 06.
Article En | MEDLINE | ID: mdl-32808582

Regenerative medicine is commonly used in human and equine athletes. Potential therapies include culture expanded stem cells, stromal vascular fraction of adipose tissue, platelet-rich plasma, bone marrow concentrate, or autologous conditioned serum. The purpose of this manuscript is to disseminate findings from a workshop on the development of translational regenerative medicine in the equine field. Five themes emerged: stem cell characterization and tenogenic differentiation; interactions between mesenchymal stem cells, other cells and the environment; scaffolds and cell packaging; blood- and bone marrow-based regenerative medicines; clinical use of regenerative therapies. Evidence gained through the use of regenerative medicine applications in the horse should continue to translate to the human patient, bringing novel regenerative therapies to both humans and horses.


Horse Diseases/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , One Health , Regenerative Medicine , Animals , Cell Differentiation , Congresses as Topic , Horses
7.
J Anat ; 236(4): 688-700, 2020 04.
Article En | MEDLINE | ID: mdl-31792963

Aging is hypothesized to be associated with changes in tendon matrix composition which may lead to alteration of tendon material properties and hence propensity to injury. Altered gene expression may offer insights into disease pathophysiology and thus open new perspectives toward designing pathophysiology-driven therapeutics. Therefore, the current study aimed at identifying naturally occurring differences in tendon micro-morphology and gene expression of newborn, young and old horses. Age-related differences in the distribution pattern of tendon fibril thickness and in the expression of the tendon relevant genes collagen type 1 (Col1), Col3, Col5, tenascin-C, decorin, tenomodulin, versican, scleraxis and cartilage oligomeric matrix protein were investigated. A qualitative and quantitative gene expression and collagen fibril diameter analysis was performed for the most frequently injured equine tendon, the superficial digital flexor tendon, in comparison with the deep digital flexor tendon. Most analyzed genes (Col1, Col3, Col5, tenascin-C, tenomodulin, scleraxis) were expressed at a higher level in foals (age ≤ 6 months) than in horses of 2.75 years (age at which flexor tendons become mature in structure) and older, decorin expression increased with age. Decorin was previously reported to inhibit the lateral fusion of collagen fibrils, causing a thinner fibril diameter with increased decorin concentration. The results of this study suggested that reduction of tendon fibril diameters commonly seen in equine tendons with increasing age might be a natural age-related phenomenon leading to greater fibril surface areas with increased fibrillar interaction and reduced sliding at the fascicular/fibrillar interface and hence a stiffer interfascicular/interfibrillar matrix. This may be a potential reason for the higher propensity to tendinopathies with increasing age.


Aging/physiology , Collagen/genetics , Decorin/genetics , Gene Expression , Membrane Proteins/genetics , Tendons/metabolism , Age Factors , Animals , Collagen/metabolism , Decorin/metabolism , Horses , Membrane Proteins/metabolism
8.
Animals (Basel) ; 9(8)2019 Jul 31.
Article En | MEDLINE | ID: mdl-31370196

We developed a finite element model (FEM) of the equine stifle joint to identify pressure peaks and simulate translocation and deformation of the menisci. A series of sectional magnetic resonance images (1.5 T) of the stifle joint of a 23 year old Shetland pony gelding served as basis for image segmentation. Based on the 3D polygon models of femur, tibia, articular cartilages, menisci, collateral ligaments and the meniscotibial ligaments, an FEM model was generated. Tissue material properties were assigned based on data from human (Open knee(s) project) and bovine femoro-tibial joint available in the literature. The FEM model was tested across a range of motion of approximately 30°. Pressure load was overall higher in the lateral meniscus than in the medial. Accordingly, the simulation showed higher translocation and deformation in the lateral compared to the medial meniscus. The results encourage further refinement of this model for studying loading patterns on menisci and articular cartilages as well as the resulting mechanical stress in the subchondral bone (femur and tibia). A functional FEM model can not only help identify segments in the stifle which are predisposed to injury, but also to better understand the progression of certain stifle disorders, simulate treatment/surgery effects and to optimize implant/transplant properties.

9.
PLoS One ; 14(4): e0214709, 2019.
Article En | MEDLINE | ID: mdl-30939166

The purpose of the current study was to establish an in vitro model for osteoarthritis (OA) by co-culture of osteochondral and synovial membrane explants. Osteochondral explants were cultured alone (control-1) or in co-culture with synovial membrane explants (control-2) in standard culture medium or with interleukin-1ß (IL1ß) and tumor necrosis factor (TNFα) added to the culture medium (OA-model-1 = osteochondral explant; OA-model-2 = osteochondroal-synovial explant). In addition, in OA-model groups a 2-mm partial-thickness defect was created in the centre of the cartilage explant. Changes in the expression of extracellular matrix (ECM) genes (collagen type-1 (Col1), Col2, Col10 and aggrecan) as well as presence and quantity of inflammatory marker genes (IL6, matrix metalloproteinase-1 (MMP1), MMP3, MMP13, a disintegrin and metalloproteinase with-thrombospondin-motif-5 (ADAMTS5) were analysed by immunohistochemistry, qPCR and ELISA. To monitor the activity of classically-activated pro-inflammatory (M1) versus alternatively-activated anti-inflammatory/repair (M2) synovial macrophages, the nitric oxide/urea ratio in the supernatant of osteochondral-synovial explant co-cultures was determined. In both OA-model groups immunohistochemistry and qPCR showed a significantly increased expression of MMPs and IL6 compared to their respective control group. ELISA results confirmed a statistically significant increase in MMP1and MMP3 production over the culturing period. In the osteochondral-synovial explant co-culture OA-model the nitric oxide/urea ratio was increased compared to the control group, indicating a shift toward M1 synovial macrophages. In summary, chemical damage (TNFα, IL1ß) in combination with a partial-thickness cartilage defect elicits an inflammatory response similar to naturally occurring OA in osteochondral explants with and without osteochondral-synovial explant co-cultures and OA-model-2 showing a closer approximation of OA due to the additional shift of synovial macrophages toward the pro-inflammatory M1 phenotype.


Chondrocytes/cytology , Models, Biological , Osteoarthritis/pathology , Synovial Membrane/cytology , Animals , Chondrocytes/metabolism , Chondrocytes/pathology , Coculture Techniques , Collagen Type I/genetics , Collagen Type I/metabolism , Horses , Interleukin-1beta/pharmacology , Interleukin-6/genetics , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Nitric Oxide/metabolism , Synovial Membrane/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Urea/metabolism
10.
Dis Model Mech ; 11(7)2018 07 06.
Article En | MEDLINE | ID: mdl-29991479

Osteoarthritis (OA), a degenerative joint disease characterized by progressive cartilage degeneration, is one of the leading causes of disability worldwide owing to the limited regenerative capacity of adult articular cartilage. Currently, there are no disease-modifying pharmacological or surgical therapies for OA. Fetal mammals, in contrast to adults, are capable of regenerating injured cartilage in the first two trimesters of gestation. A deeper understanding of the properties intrinsic to the response of fetal tissue to injury would allow us to modulate the way in which adult tissue responds to injury. In this study, we employed secretome proteomics to compare fetal and adult protein regulation in response to cartilage injury using an ovine cartilage defect model. The most relevant events comprised proteins associated with the immune response and inflammation, proteins specific for cartilage tissue and cartilage development, and proteins involved in cell growth and proliferation. Alarmins S100A8, S100A9 and S100A12 and coiled-coil domain containing 88A (CCDC88A), which are associated with inflammatory processes, were found to be significantly upregulated following injury in adult, but not in fetal animals. By contrast, cartilage-specific proteins like proteoglycan 4 were upregulated in response to injury only in fetal sheep postinjury. Our results demonstrate the power and relevance of the ovine fetal cartilage regeneration model presented here for the first time. The identification of previously unrecognized modulatory proteins that plausibly affect the healing process holds great promise for potential therapeutic interventions.


Aging/pathology , Cartilage, Articular/pathology , Fetus/pathology , Fibrocartilage/pathology , Proteins/metabolism , Proteomics , Regeneration , Animals , Cartilage, Articular/injuries , Cartilage, Articular/surgery , Disease Models, Animal , Extracellular Matrix/metabolism , Female , Ki-67 Antigen/metabolism , Mass Spectrometry , Matrix Metalloproteinases/metabolism , Sheep
11.
PLoS One ; 13(3): e0194052, 2018.
Article En | MEDLINE | ID: mdl-29522550

Meniscal pathologies are among the most common injuries of the femorotibial joint in both human and equine patients. Pathological forces and ensuing injuries of the cranial horn of the equine medial meniscus are considered analogous to those observed in the human posterior medial horn. Biomechanical properties of human menisci are site- and depth- specific. However, the influence of equine meniscus topography and composition on its biomechanical properties is yet unknown. A better understanding of equine meniscus composition and biomechanics could advance not only veterinary therapies for meniscus degeneration or injuries, but also further substantiate the horse as suitable translational animal model for (human) meniscus tissue engineering. Therefore, the aim of this study was to investigate the composition and structure of the equine knee meniscus in a site- and age-specific manner and their relationship with potential site-specific biomechanical properties. The meniscus architecture was investigated histologically. Biomechanical testing included evaluation of the shore hardness (SH), stiffness and energy loss of the menisci. The SH was found to be subjected to both age and site-specific changes, with an overall higher SH of the tibial meniscus surface and increase in SH with age. Stiffness and energy loss showed neither site nor age related significant differences. The macroscopic and histologic similarities between equine and human menisci described in this study, support continued research in this field.


Horses/anatomy & histology , Meniscus/anatomy & histology , Meniscus/physiology , Aging , Animals , Biomechanical Phenomena , Collagen/analysis , Compressive Strength , Female , Gait , Glycosaminoglycans/analysis , Hardness , Hardness Tests , Male , Meniscus/chemistry , Stifle/anatomy & histology , Stifle/physiology , Structure-Activity Relationship , X-Ray Microtomography
12.
Tissue Eng Part C Methods ; 23(5): 298-310, 2017 05.
Article En | MEDLINE | ID: mdl-28504918

Sheep are one of the most frequently used large animal models in stem cell research. However, minimal invasive or noninvasive sources of mesenchymal stem cells (MSCs) in sheep are scarce. In the light of the principles of the 3Rs (reduce, refine, replace), it would therefore be desirable to identify a minimally invasive or noninvasive ovine MSC source. In humans, the chorionic villi of the placenta, which can be noninvasively harvested as part of the afterbirth, have been identified as a rich source of MSCs. Therefore, in the present study, ovine placenta cotyledons, which have similar function and structure to human chorionic villi, were tested for their potential use as a noninvasive source of ovine MSCs. Through mincing of the placental cotyledons, collagenase digestion, and Ficoll density gradient centrifugation, combined with plastic adherence selection, MSCs were successfully isolated. Their morphological, immunophenotypical, and cellular growth characteristics, as well as their proliferation, differentiation, and migration potential, were evaluated and compared to the currently best-researched MSC source, bone marrow-derived stem cells. Ovine cotyledons were shown to be a reliable, abundant source for the noninvasive, pain- and risk-free harvest of MSCs. The collection procedure does not interfere with partum or the initial bonding phase between ewes and lambs and is therefore exempt from ethical debate. Ovine placenta cotyledon-derived MSCs exhibit multipotential characteristics and can be cryopreserved for later use.


Cell Differentiation , Cell Separation/methods , Mesenchymal Stem Cells/cytology , Placenta/cytology , Tissue Engineering/methods , Adipogenesis/physiology , Animals , Cell Movement , Cell Proliferation , Chondrogenesis/physiology , Female , Mesenchymal Stem Cells/physiology , Osteogenesis/physiology , Placenta/physiology , Pregnancy , Sheep
13.
Tissue Eng Part A ; 23(9-10): 390-402, 2017 05.
Article En | MEDLINE | ID: mdl-28095754

Meniscal injuries are the most frequently encountered soft tissue injuries in the equine stifle joint. Due to the inherent limited repair potential of meniscal tissue, meniscal injuries do not only affect the meniscus itself but also lead to impaired joint homeostasis and secondary osteoarthritis. The presented study compares 3D coculture constructs of primary equine mesenchymal stem cells (MSC) and meniscus cells (MC) seeded on three different scaffolds-a cell-laden collagen type I hydrogel (Col I gel), a tissue-derived small intestinal matrix scaffold (SIS-muc) and a combination thereof-for their qualification to be applied for meniscus tissue engineering. To investigate cell attachment of primary MC and MSC on SIS-muc matrix SEM pictures were performed. For molecular analysis, lyophilized samples of coculture constructs with different cell ratios (100% MC, 100% MSC, and 50% MC and 50% MSC, 20% MC, and 80% MSC) were digested and analyzed for DNA and GAG content. Active matrix remodeling of 3D coculture models was indicated by matrix metalloproteinases detection. For comparison of tissue-engineered constructs with the histologic architecture of natural equine menisci, paired lateral and medial menisci of 15 horses representing different age groups were examined. A meniscus phenotype with promising similarity to native meniscus tissue in its GAG/DNA expression in addition to Col I, Col II, and Aggrecan production was achieved using a scaffold composed of Col I gel on SIS-muc combined with a coculture of MC and MSC. The results encourage further development of this scaffold-cell combination for meniscus tissue engineering.


Collagen Type I/chemistry , Extracellular Matrix/chemistry , Hydrogels/chemistry , Intestine, Small/chemistry , Meniscus/metabolism , Mesenchymal Stem Cells/metabolism , Tissue Engineering , Animals , Cells, Cultured , Coculture Techniques , Horses , Meniscus/injuries , Pilot Projects
15.
PeerJ ; 4: e1773, 2016.
Article En | MEDLINE | ID: mdl-27019778

Background. Mesenchymal stromal cells (MSCs) are increasingly used for clinical applications in equine patients. For MSC isolation and expansion, a laboratory step is mandatory, after which the cells are sent back to the attending veterinarian. Preserving the biological properties of MSCs during this transport is paramount. The goal of the study was to compare transport-related parameters (transport container, media, temperature, time, cell concentration) that potentially influence characteristics of culture expanded equine MSCs. Methods. The study was arranged in three parts comparing (I) five different transport containers (cryotube, two types of plastic syringes, glass syringe, CellSeal), (II) seven different transport media, four temperatures (4 °C vs. room temperature; -20 °C vs. -80 °C), four time frames (24 h vs. 48 h; 48 h vs. 72 h), and (III) three MSC concentrations (5 × 10(6), 10 × 10(6), 20 × 10(6) MSC/ml). Cell viability (Trypan Blue exclusion; percent and total number viable cell), proliferation and trilineage differentiation capacity were assessed for each test condition. Further, the recovered volume of the suspension was determined in part I. Each condition was evaluated using samples of six horses (n = 6) and differentiation protocols were performed in duplicates. Results. In part I of the study, no significant differences in any of the parameters were found when comparing transport containers at room temperature. The glass syringe was selected for all subsequent evaluations (highest recoverable volume of cell suspension and cell viability). In part II, media, temperatures, or time frames had also no significant influence on cell viability, likely due to the large number of comparisons and small sample size. Highest cell viability was observed using autologous bone marrow supernatant as transport medium, and "transport" at 4 °C for 24 h (70.6% vs. control group 75.3%); this was not significant. Contrary, viability was unacceptably low (<40%) for all freezing protocols at -20 °C or -80 °C, particularly with bone marrow supernatant or plasma and DMSO. In part III, various cell concentrations also had no significant influence on any of the evaluated parameters. Chondrogenic differentiation showed a trend towards being decreased for all transport conditions, compared to control cells. Discussion. In this study, transport conditions were not found to impact viability, proliferation or ability for trilineage differentiation of MSCs, most likely due to the small sample size and large number of comparisons. The unusual low viability after all freezing protocols is in contrast to previous equine studies. Potential causes are differences in the freezing, but also in thawing method. Also, the selected container (glass syringe) may have impacted viability. Future research may be warranted into the possibly negative effect of transport on chondrogenic differentiation.

16.
Cell Transplant ; 24(6): 1111-25, 2015.
Article En | MEDLINE | ID: mdl-24330785

The purpose of this study was to evaluate the use of three different superparamagnetic iron oxide (SPIO) particles for labeling of ovine and equine bone marrow (BM)-derived multipotent stromal cells (MSCs) in vitro. MSCs were obtained from five adult sheep and horses, respectively. After three passages (p3), cells were labeled with either 1) Molday ION Rhodamine B, 2) Endorem, 3) Resovist, or 4) remained unlabeled as control. Labeling efficiency, marker retention, and long-term detectability in MRI until p7 were evaluated. Further, proliferation capacity and trilineage differentiation as indicators for potential impact on stromal cell characteristics were assessed. MSCs of both species were successfully labeled with all three SPIO products. A high, exclusively intracellular, iron uptake was achieved by Molday ION Rhodamine B only. Labeling with Resovist led to prominent extracellular iron presence; labeling with Endorem was less efficient. During MRI, all labeled cells showed strong hypointense signals, contrary to unlabeled controls. Resovist induced the largest areas of hypointense signals, followed by Molday ION Rhodamine B and Endorem. MRI signal detectability decreased from p4 to p7. Proliferation, adipogenic, and osteogenic differentiation potential were not reduced by cell labeling compared to unlabeled cells. Chondrogenic differentiation capacity decreased with increasing amount of iron associated with the cells. Among the three products, Resovist and Molday were identified as promising labeling agents. While Resovist achieved superior results in most of the assessed parameters, Molday ION Rhodamine B ensured intracellular iron uptake without extracellular SPIO complexes and consistent hypointense signals on MRI.


Dextrans/metabolism , Magnetic Resonance Imaging/methods , Multipotent Stem Cells/metabolism , Staining and Labeling , Adipogenesis , Animals , Cell Lineage , Cell Proliferation , Chondrogenesis , Horses , Magnetite Nanoparticles , Multipotent Stem Cells/cytology , Osteogenesis , Phantoms, Imaging , Sheep , Time Factors
17.
BMC Vet Res ; 9: 221, 2013 Oct 29.
Article En | MEDLINE | ID: mdl-24168625

BACKGROUND: The treatment of tendon lesions with multipotent mesenchymal stromal cells (MSCs) is widely used in equine medicine. Cell sources of MSCs include bone marrow, as well as solid tissues such as adipose tissue. MSCs can be isolated from these solid tissues either by enzymatic digestion or by explant technique. However, the different preparation techniques may potentially influence the properties of the isolated MSCs. Therefore, the aim of this study was to investigate and compare the effects of these two different methods used to isolate MSCs from solid tissues.Equine adipose tissue, tendon and umbilical cord matrix served as solid tissue sources of MSCs with different stiffness and density. Subsequent to tissue harvest, MSCs were isolated either by enzymatic digestion with collagenase or by explant technique. Cell yield, growth, differentiation potential and tendon marker expression were analysed. RESULTS: At first passage, the MSC yield was significantly higher in enzymatically digested tissue samples than in explanted tissue samples, despite a shorter period of time in primary culture. Further analysis of cell proliferation, migration and differentiation revealed no significant differences between MSCs isolated by enzymatic digestion and MSCs isolated by explant technique. Interestingly, analysis of gene expression of tendon markers revealed a significantly higher expression level of scleraxis in MSCs isolated by enzymatic digestion. CONCLUSIONS: Both isolation techniques are feasible methods for successful isolation of MSCs from solid tissues, with no major effects on cellular proliferation, migration or differentiation characteristics. However, higher MSC yields were achieved in a shorter period of time by collagenase digestion, which is advantageous for the therapeutic use of MSCs. Moreover, based on the higher level of expression of scleraxis in MSCs isolated by enzymatic digestion, these cells might be a better choice when attempting tendon regeneration.


Mesenchymal Stem Cells/physiology , Adipogenesis/physiology , Adipose Tissue/cytology , Animals , Cell Count/veterinary , Cell Movement/physiology , Cell Proliferation , Cells, Cultured , Chondrogenesis/physiology , Collagenases , Cytological Techniques/methods , Cytological Techniques/veterinary , Gene Expression/physiology , Horses , Osteogenesis/physiology , Proteolysis , Tendons/cytology , Umbilical Cord/cytology
18.
Vet J ; 195(1): 98-106, 2013 Jan.
Article En | MEDLINE | ID: mdl-22841420

Multipotent mesenchymal stromal cells (MSCs) are a promising therapeutic tool for the treatment of equine tendon and other musculoskeletal injuries. While bone marrow is considered the 'gold standard' source of these cells, various other tissues contain MSCs with potentially useful features. The aim of this study was to compare clinically relevant characteristics of MSCs derived from bone marrow, umbilical cord blood and tissue and from adipose tissue and tendon. Cell yield, proliferation, migration, tendon marker expression and differentiation into adipocytes, chondrocytes and osteoblasts was assessed, quantified and compared. MSC numbers obtained from adipose, tendon or umbilical cord tissues were 222-fold higher than those obtained from bone marrow or cord blood. Cells derived from tendon and adipose tissues exhibited most rapid proliferation. Osteogenic differentiation was most prominent in MSCs derived from bone marrow, and was weak in MSCs derived from umbilical cord blood and tissue. In contrast, the highest levels of chondrogenic differentiation were observed in MSCs derived from these sources. Collagen 1A2 expression was highest in adipose- and tendon-derived MSCs, while scleraxis expression was highest in cord blood- and in tendon-derived MSCs. The findings indicate that MSCs from different sources display significantly diverse properties that may impact on their therapeutic application.


Bone Marrow Cells/physiology , Cell Differentiation/physiology , Cell Proliferation , Horses/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Animals , Biomarkers , Bone Marrow Cells/cytology , Cell Culture Techniques , Fetal Blood/cytology , Gene Expression Regulation/physiology , Tendons/metabolism , Umbilical Cord/cytology
19.
Can J Vet Res ; 76(1): 52-6, 2012 Jan.
Article En | MEDLINE | ID: mdl-22754095

The objective of this study was to compare bone marrow (BM) aspirates from the sternum and the tuber coxae of middle-aged horses. Bone marrow was obtained from the sternum and both tubera coxae of 12 healthy, 13-year-old geldings. Two different puncture techniques were used for the tuber coxae. The 2 syringes used for sternal sampling were evaluated separately. The mononuclear cell (MNC) fraction of the BM was isolated and the mesenchymal stem cells (MSCs) were culture-expanded. At the sternum, BM aspiration was always possible. Bone marrow aspiration at the tuber coxae required straight and deep needle penetration combined with high negative pressure. With this technique a median sample amount of 11.0 mL with large individual variation was obtained. A median of 3.06 × 10(6) MNC/mL BM (1st syringe) and 2.46 × 10(6) MNC/mL BM (2nd syringe) was isolated from sternal samples. In contrast, the tuber coxae yielded a median of 0.27 × 10(6) MNC/mL BM. The first passage yielded a median of 2.19 × 10(6) MSC (1st syringe) and 1.13 × 10(6) MSC (2nd syringe) from sternal samples, compared to a significantly lower median number of MSC from tuber coxae BM (0.06 × 10(6) MSC). The number of MNC and MSC obtainable from the BM aspirates taken from the tuber coxae is significantly lower than that obtained from the sternal BM aspirates. Autologous BM for the equine athlete is particularly clinically relevant at an advanced age. Based on our findings, the tuber coxae cannot be recommended for BM aspiration in middle-aged horses.


Biopsy, Needle/veterinary , Bone Marrow Cells/cytology , Bone Marrow/surgery , Horses/anatomy & histology , Ilium/surgery , Sternum/surgery , Animals , Biopsy, Needle/methods , Cell Count/veterinary , Centrifugation, Density Gradient/veterinary , Male , Statistics, Nonparametric
20.
Cell Tissue Res ; 347(3): 677-688, 2012 03.
Article En | MEDLINE | ID: mdl-22287044

Regenerative medicine is one of the most intensively researched medical branches, with enormous progress every year. When it comes to translating research from bench to bedside, many of the pioneering innovations are achieved by cooperating teams of human and veterinary medical scientists. The veterinary profession has an important role to play in this new and evolving technology, holding a great scientific potential, because animals serve widely as models for human medicine and results obtained from animals may serve as preclinical results for human medicine. Regenerative veterinary medicine utilizing mesenchymal stromal cells (MSC) for the treatment of acute injuries as well as chronic disorders is gradually turning into clinical routine. As orthopaedic disorders represent a major part of all cases in veterinary clinical practice, it is not surprising that they are currently taking a leading role in MSC therapies. Therefore, the purpose of this paper is to give an overview on past and current achievements as well as future perspectives in stem cell-based tissue engineering in veterinary orthopaedics.

...