Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Can J Vet Res ; 88(3): 87-93, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38988333

ABSTRACT

There is a knowledge gap regarding the effect of extracorporeal shockwave treatment (ESWT) on the stress response and immunomodulatory and anti-inflammatory properties of equine umbilical cord blood mesenchymal stromal cells (CB-MSCs). The objective of this study was to investigate the presence of cellular oxidative stress, inflammatory response, and production of growth factors in CB-MSCs after treatment with ESWT. We hypothesized that CB-MSCs treated with ESWT will experience higher levels of cellular stress and increased production of anti-inflammatory cytokines and growth factors compared to untreated CB-MSCs.


Il existe un manque de connaissances concernant l'effet du traitement extracorporel par ondes de choc (ESWT) sur la réponse au stress et les propriétés immunomodulatrices et anti-inflammatoires des cellules stromales mésenchymateuses du sang de cordon ombilical équin (CB-MSCs). L'objectif de cette étude était d'étudier la présence de stress oxydatif cellulaire, de réponse inflammatoire et de production de facteurs de croissance dans les CB-MSCs après un traitement par ESWT. Nous avons émis l'hypothèse que les CB-MSCs traitées par ESWT connaîtront des niveaux plus élevés de stress cellulaire et une production accrue de cytokines anti-inflammatoires et de facteurs de croissance par rapport aux CB-MSCs non traitées.(Traduit par Docteur Serge Messier).


Subject(s)
Fetal Blood , Mesenchymal Stem Cells , Animals , Horses , Fetal Blood/cytology , Extracorporeal Shockwave Therapy/methods , Cytokines/metabolism , Cells, Cultured
2.
Biotechnol Bioeng ; 121(1): 192-205, 2024 01.
Article in English | MEDLINE | ID: mdl-37772415

ABSTRACT

Equine mesenchymal stromal cells (MSCs) have been found to be beneficial for the treatment of many ailments, including orthopedic injuries, due to their superior differentiation potential and immunomodulating properties. Cell therapies require large cell numbers, which are not efficiently generated using conventional static expansion methods. Expansion of equine cord blood-derived MSCs (eCB-MSCs) in bioreactors, using microcarriers as an attachment surface, has the potential to generate large numbers of cells with increased reproducibility and homogeneity compared with static T-flask expansion. This study investigated the development of an expansion process using Vertical-Wheel (VW) bioreactors, a single-use bioreactor technology that incorporates a wheel instead of an impeller. Initially, microcarriers were screened at small scale to assess eCB-MSC attachment and growth and then in bioreactors to assess cell expansion and harvesting. The effect of different donors, serial passaging, and batch versus fed batch were all examined in 0.1 L VW bioreactors. The use of VW bioreactors with an appropriate microcarrier was shown to be able to produce cell densities of up to 1E6 cells/mL, while maintaining cell phenotype and functionality, thus demonstrating great potential for the use of these bioreactors to produce large cell numbers for cell therapies.


Subject(s)
Cell Culture Techniques , Mesenchymal Stem Cells , Animals , Horses , Cell Culture Techniques/methods , Fetal Blood , Reproducibility of Results , Bioreactors , Cell Differentiation , Cell Proliferation
3.
Stem Cells Dev ; 32(21-22): 693-702, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37578107

ABSTRACT

Mesenchymal stromal cells (MSCs) are a promising cell source for cartilage tissue regeneration in animals and humans but with large interdonor variation in their in vitro chondrogenic differentiation potential. Underlying molecular mechanisms responsible for culture-expanded MSC heterogeneity remain poorly understood. In this study, we sought to identify variations in microRNA (miRNA) signatures associated with cultured equine MSC chondrogenic differentiation potential from different donors. Neocartilage tissue generated from equine cord blood-derived MSCs was categorized as having either high or low chondrogenic potential (LCP) based on their histological appearance and quantification of glycosaminoglycan deposition. Using next-generation sequencing, we identified 30 differentially expressed miRNAs among undifferentiated MSC cultures that corresponded with their chondrogenic potential. Of note, MSCs with LCP upregulated miR-146a and miR-487b-3p, which was also observed by quantitative real-time polymerase chain reaction. Our findings suggest that miRNA profiling of equine MSC cultures may have prognostic value in selecting MSC donors with regard to their chondrogenic differentiation potential.


Subject(s)
Mesenchymal Stem Cells , MicroRNAs , Humans , Animals , Horses , MicroRNAs/genetics , Prognosis , Cell Differentiation/genetics , Cartilage , Chondrogenesis/genetics , Cells, Cultured , Chondrocytes
4.
Cartilage ; 12(2): 222-225, 2021 04.
Article in English | MEDLINE | ID: mdl-30841716

ABSTRACT

OBJECTIVES: RNA isolation is necessary for the evaluation of gene expression. Due to the nature of its extracellular matrix, RNA isolation from articular hyaline cartilage is difficult and thus the tissue is commonly enzymatically digested in order to extract RNA from the obtained chondrocytes. We hypothesized that the digestion process affects the expression levels of common cartilage-associated genes. DESIGN: Expression of cartilage-associated genes was compared between intact cartilage and digested chondrocytes from weight bearing and non-weight bearing regions of the equine fetlock joint. RESULTS: The gene expression of SOX9, COL1A2, COL2A1, ACAN, and COLX were analyzed. Digested cartilage showed a significant decrease in the expression of COL1A2, COL2A1, and ACAN compared to intact cartilage in both joint regions, and an increase in COLX expression in non-weight bearing cartilage only. CONCLUSIONS: Enzymatic digestion of cartilage significantly impacts gene expression profile. We conclude that while RNA isolation from intact cartilage is more technically difficult, determination of gene expression should be conducted on intact cartilage if true representation of the in vivo processes is sought.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Extracellular Matrix Proteins/analysis , Gene Expression Profiling/methods , Transcriptome/genetics , Animals , Horses/genetics , Proteolysis , RNA/isolation & purification
5.
J Biol Eng ; 13: 25, 2019.
Article in English | MEDLINE | ID: mdl-30949237

ABSTRACT

Equine mesenchymal stromal cells (MSCs) are increasingly investigated for their clinical therapeutic utility. Such cell-based treatments can require cell numbers in the millions or billions, with conventional expansion methods using static T-flasks typically inefficient in achieving these cell numbers. Equine cord blood-derived MSCs (eCB-MSCs), are promising cell candidates owing to their capacity for chondrogenic differentiation and immunomodulation. Expansion of eCB-MSCs in stirred suspension bioreactors with microcarriers as an attachment surface has the potential to generate clinically relevant numbers of cells while decreasing cost, time and labour requirements and increasing reproducibility and yield when compared to static expansion. As eCB-MSCs have not yet been expanded in stirred suspension bioreactors, a robust protocol was required to expand these cells using this method. This study outlines the development of an expansion bioprocess, detailing the inoculation phase, expansion phase, and harvesting phase, followed by phenotypic and trilineage differentiation characterization of two eCB-MSC donors. The process achieved maximum cell densities up to 75,000 cells/cm2 corresponding to 40 million cells in a 100 mL bioreactor, with a harvesting efficiency of up to 80%, corresponding to a yield of 32 million cells from a 100 mL bioreactor. When compared to cells grown in static T-flasks, bioreactor-expanded eCB-MSC cultures did not change in surface marker expression or trilineage differentiation capacity. This indicates that the bioreactor expansion process yields large quantities of eCB-MSCs with similar characteristics to conventionally grown eCB-MSCs.

6.
Tissue Eng Part B Rev ; 25(2): 114-125, 2019 04.
Article in English | MEDLINE | ID: mdl-30638141

ABSTRACT

IMPACT STATEMENT: In this comprehensive review, we are providing a holistic overview of osteochondral tissue development, disease, pain localization, as well as structural evaluation and current repair strategies. This review is intended to serve as a broad introduction to this multidisciplinary research area. It is a thorough examination of the biological aspects of the osteochondral unit from a tissue engineering perspective, highlighting the importance of the subchondral bone in chondral and osteochondral lesion repair and pain relief.


Subject(s)
Bone and Bones/cytology , Cartilage, Articular/cytology , Chondrocytes/cytology , Knee Injuries/therapy , Tissue Engineering/methods , Animals , Humans
7.
Stem Cells Dev ; 28(3): 227-237, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30484372

ABSTRACT

Mesenchymal stromal cells (MSCs) are the most common cell population studied for therapeutic use in veterinary medicine. MSCs obtained from neonatal sources such as umbilical cord tissue (CT-MSCs) or cord blood (CB-MSCs) are appealing due to the non-invasive nature of procurement and the time allowed for characterization of cells before use. However, it remains unclear as to whether CB- or CT-MSCs have equivalent progenitor and non-progenitor functions. CB-MSCs have been shown to have superior chondrogenic potential to MSCs from other sources, whereas their immunomodulatory capacity does not seem to vary significantly. Using equine CB-MSCs and CT-MSCs from the same donors, we hypothesized that MSCs from both sources would have a similar immunophenotype, that CB-MSCs would be more amenable to differentiation, and that they can equally suppress lymphocyte proliferation. We evaluated cells from both sources for "classic" equine MSC markers CD90, CD105, CD29, and CD44, as well as pericyte markers CD146, NG2, and α-SMA. Contrary to our hypothesis, CB-MSCs showed mid- to high expression of pericyte surface markers CD146 and NG2, whereas expression in CT-MSCs was absent. On trilineage differentiation, CB-MSCs were more osteogenic and chondrogenic based on alkaline phosphatase activity and glycosaminoglycan content, respectively. Finally, using a mononuclear cell (MNC) suppression assay, we determined that both CB-MSCs and CT-MSCs are capable of suppressing stimulated MNC proliferation to a similar degree. We have determined that the choice of MSC tissue source should be made with the intended application in mind. This appears to be particularly relevant if pursuing a progenitor-based treatment strategy.


Subject(s)
Cell Differentiation , Cell Proliferation , Fetal Blood/cytology , Lymphocytes/immunology , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology , Animals , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Female , Horses , Lymphocytes/physiology , Mesenchymal Stem Cells/immunology , Pericytes/cytology , Pericytes/metabolism , Pregnancy
8.
Stem Cells Dev ; 25(1): 80-9, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26414480

ABSTRACT

Multipotent mesenchymal stromal cells (MSCs) are more and more frequently used to treat orthopedic injuries in horses. However, these cells are limited in their expandability and differentiation capacity. Recently, the first equine-induced pluripotent stem cell (iPSC) lines were reported by us [ 1 ]. In vitro differentiation of iPSCs into MSC-like cells is an attractive alternative to using MSCs derived from other sources, as a much larger quantity of patient-specific cells with broad differentiation potential could be generated. However, the differentiation capacity of iPSCs to MSCs and the potential for use in tissue engineering have yet to be explored. In this study, equine iPSCs were induced to differentiate into an MSC-like population. Upon induction, the iPSCs changed morphology toward spindle-shaped cells similar to MSCs. The ensuing iPSC-MSCs exhibited downregulation of pluripotency-associated genes and an upregulation of MSC-associated genes. In addition, the cells expressed the same surface markers as MSCs derived from equine umbilical cord blood. We then assessed the multilineage differentiation potential of iPSC-MSCs. Although chondrogenesis was not achieved after induction with transforming growth factor-beta 3 (TGFß3) and/or bone morphogenic protein 4 (BMP-4) in 3D pellet culture, mineralization characteristic of osteogenesis and lipid droplet accumulation characteristic of adipogenesis were observed after chemical induction. We demonstrate a protocol for the derivation of MSC-like progenitor populations from equine iPS cells.


Subject(s)
Cell Differentiation , Horses , Induced Pluripotent Stem Cells/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Adipogenesis/physiology , Animals , Cell Culture Techniques/veterinary , Cell Separation , Cells, Cultured , Cellular Reprogramming , Chondrogenesis/physiology , Induced Pluripotent Stem Cells/cytology , Mice , Osteogenesis/physiology , Tissue Engineering/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL
...