Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Sensors (Basel) ; 21(4)2021 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-33670238

RESUMEN

(1) Background: Postural sway is frequently used to quantify human postural control, balance, injury, and neurological deficits. However, there is considerably less research investigating the value of the metric in horses. Much of the existing equine postural sway research uses force or pressure plates to examine the centre of pressure, inferring change at the centre of mass (COM). This study looks at the inverse, using an inertial measurement unit (IMU) on the withers to investigate change at the COM, exploring the potential of postural sway evaluation in the applied domain. (2) Methods: The lipopolysaccharide model was used to induce transient bilateral lameness in seven equines. Horses were monitored intermittently by a withers fixed IMU over seven days. (3) Results: There was a significant effect of time on total protein, carpal circumference, and white blood cell count in the horses, indicating the presence of, and recovery from, inflammation. There was a greater amplitude of displacement in the craniocaudal (CC) versus the mediolateral (ML) direction. A significant difference was observed in the amplitude of displacement in the ML direction between 4-12 h and 168 h. (4) Conclusions: The significant reduction in ML displacement during the acute inflammation period alongside greater overall CC displacement may be a compensatory behaviour for bilateral lameness.


Asunto(s)
Caballos , Cojera Animal/diagnóstico , Equilibrio Postural , Animales , Estudios de Factibilidad , Cojera Animal/inducido químicamente , Presión , Torso
2.
Connect Tissue Res ; 60(2): 95-106, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-29560747

RESUMEN

AIM: The horse joint, due to its similarity with the human joint, is the ultimate model for translational articular cartilage repair studies. This study was designed to determine the critical size of cartilage defects in the equine carpus and serve as a benchmark for the evaluation of new cartilage treatment options. MATERIAL AND METHODS: Circular full-thickness cartilage defects with a diameter of 2, 4, and 8 mm were created in the left middle carpal joint and similar osteochondral (3.5 mm in depth) defects in the right middle carpal joint of 5 horses. Spontaneously formed repair tissue was examined macroscopically, with MR and µCT imaging, polarized light microscopy, standard histology, and immunohistochemistry at 12 months. RESULTS: Filling of 2 mm chondral defects was good (77.8 ± 8.5%), but proteoglycan depletion was evident in Safranin-O staining and gadolinium-enhanced MRI (T1Gd). Larger chondral defects showed poor filling (50.6 ± 2.7% in 4 mm and 31.9 ± 7.3% in 8 mm defects). Lesion filling in 2, 4, and 8 mm osteochondral defects was 82.3 ± 3.0%, 68.0 ± 4.6% and 70.8 ± 15.4%, respectively. Type II collagen staining was seen in 9/15 osteochondral defects but only in 1/15 chondral defects. Subchondral bone pathologies were evident in 14/15 osteochondral samples but only in 5/15 chondral samples. Although osteochondral lesions showed better neotissue quality than chondral lesions, the overall repair was deemed unsatisfactory because of the subchondral bone pathologies. CONCLUSION: We recommend classifying 4 mm as critical osteochondral lesion size and 2 mm as critical chondral lesion size for cartilage repair research in the equine carpal joint model.


Asunto(s)
Articulaciones del Carpo/patología , Cartílago Articular/patología , Caballos/anatomía & histología , Animales , Articulaciones del Carpo/diagnóstico por imagen , Cartílago Articular/diagnóstico por imagen , Imagen por Resonancia Magnética , Microscopía de Polarización , Factores de Tiempo , Cicatrización de Heridas , Microtomografía por Rayos X
3.
Cytotherapy ; 18(8): 957-969, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27288309

RESUMEN

BACKGROUND AIMS: In regenerative medicine, the use of allogeneic cells could enable the development of "off the shelf" therapies for patients with critical size bone defects, reducing limitations observed with the use of autologous cells, such as cost and time to treat the patient. The idea of the use of allogeneic bone marrow mesenchymal stromal cells (BMSCs) has been of interest in tissue engineering studies. However, little is known about the properties of these cells upon differentiation. Chondrogenically differentiated BMSCs have already been shown to form endochondral bone in immunodeficient and immunocompetent animals. The success of this bone formation is dependent on the host's endogenous cells. This study investigates the interactions between allogeneic chondrogenically differentiated human bone marrow mesenchymal stromal cell (hBMSC) pellets and T lymphocytes in vitro. METHODS: Non-chondrogenic (-transforming growth factor (TGF)ß3) and chondrogenic hBMSC (+TGFß3) pellets were directly co-cultured with unstimulated and CD3/CD28-stimulated peripheral blood mononuclear cells (PBMCs) for 7 days. hBMSC pellets from the co-culture were either fixed for histological analysis or quantitative real time polymerase chain reaction (qRT-PCR). PBMCs were harvested for flow cytometry. RESULTS: Flow cytometic analysis revealed that chondrogenically differentiated hBMSC pellets did not alter the number or proliferation of CD4+, CD8+ T cells or FoxP3+ T regulatory cells (CD4+CD25+CD127-). Chondrogenic hBMSC pellets did not induce immunogenic responses in unstimulated PBMCs. Infiltrating CD3 T cells were found in the matrix of hBMSC pellets. Furthermore, qRT-PCR demonstrated low levels of T-cell activation genes (CD25, CD69, PRF1 and GZMB) in addition to low gene expression levels of the pro-inflammatory gene tumor necrosis factor alpha (TNFα) in chondrogenically differentiated hBMSC pellets cultured with unstimulated PBMCs in comparison with non-chondrogenic hBMSC pellets. CONCLUSIONS: Collectively the results of this study demonstrate that allogeneic chondrogenically differentiated hBMSC pellets are non-immunogenic and do not induce the activation of destructive T-cell responses in vitro.


Asunto(s)
Diferenciación Celular , Condrocitos/fisiología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/fisiología , Linfocitos T/inmunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/fisiología , Células Cultivadas , Condrogénesis/fisiología , Técnicas de Cocultivo , Humanos , Inmunomodulación/fisiología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/fisiología , Activación de Linfocitos , Masculino , Células Madre Mesenquimatosas/citología , Osteogénesis , Trasplante Homólogo
4.
Vet Surg ; 44(6): 723-30, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25998193

RESUMEN

OBJECTIVES: To evaluate knot security for 3 knot types created in 3 commonly used 5 metric suture materials incubated in physiological and pathological fluids. STUDY DESIGN: In vitro mechanical study. SAMPLE POPULATION: Knotted suture loops (n = 5/group). METHODS: Loops of 3 different suture materials (glycolide/lactide copolymer; polyglactin 910; polydioxanone) were created around a 20 mm rod using 3 knot types (square [SQ], surgeon's [SK], and triple knot [TK]) and were tested to failure in distraction (6 mm/min) after tying (day 0) and after being incubated for 14 and 28 days in phosphate buffered saline (PBS) or inflamed peritoneal fluid. Failure load (N) and mode were recorded and compared. RESULTS: For polydioxanone, significant differences in force to knot failure were found between SQ and SK/TK but not between SK and TK. The force required to break all constructs increased after incubation in phosphate buffered saline (PBS). With glycolide/lactide copolymer no differences in force to knot failure were observed. With polyglactin 910, a significant difference between SQ and TK was observed, which was not seen between the other knot types. Incubation in inflamed peritoneal fluid caused a larger and more rapid decrease in force required to cause knot failure than incubation in PBS. CONCLUSIONS: Mechanical properties of suture materials have significant effects on knot security. For polydioxanone, SQ is insufficient to create a secure knot. Additional wraps above a SK confer extra stability in some materials, but this increase may not be clinically relevant or justifiable. Glycolide/lactide copolymer had excellent knot security.


Asunto(s)
Análisis de Falla de Equipo/normas , Polidioxanona/uso terapéutico , Poliglactina 910/uso terapéutico , Técnicas de Sutura/veterinaria , Animales , Líquido Ascítico/patología , Dioxanos/uso terapéutico , Análisis de Falla de Equipo/métodos , Caballos , Fosfatos , Cloruro de Sodio , Resistencia a la Tracción , Factores de Tiempo
5.
Biomaterials ; 306: 122471, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38377846

RESUMEN

Allogeneic stem-cell based regenerative medicine is a promising approach for bone defect repair. The use of chondrogenically differentiated human marrow stromal cells (MSCs) has been shown to lead to bone formation by endochondral ossification in immunodeficient pre-clinical models. However, an insight into the interactions between the allogeneic immune system and the human MSC-derived bone grafts has not been fully achieved yet. The choice of a potent source of MSCs isolated from pediatric donors with consistent differentiation and high proliferation abilities, as well as low immunogenicity, could increase the chance of success for bone allografts. In this study, we employed an immunodeficient animal model humanised with allogeneic immune cells to study the immune responses towards chondrogenically differentiated human pediatric MSCs (ch-pMSCs). We show that ch-differentiated pMSCs remained non-immunogenic to allogeneic CD4 and CD8 T cells in an in vitro co-culture model. After subcutaneous implantation in mice, ch-pMSC-derived grafts were able to initiate bone mineralisation in the presence of an allogeneic immune system for 3 weeks without the onset of immune responses. Re-exposing the splenocytes of the humanised animals to pMSCs did not trigger further T cell proliferation, suggesting an absence of secondary immune responses. Moreover, ch-pMSCs generated mature bone after 8 weeks of implantation that persisted for up to 6 more weeks in the presence of an allogeneic immune system. These data collectively show that human allogeneic chondrogenically differentiated pediatric MSCs might be a safe and potent option for bone defect repair in the tissue engineering and regenerative medicine setting.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Mesenquimatosas , Humanos , Ratones , Animales , Niño , Osteogénesis , Médula Ósea , Células del Estroma , Diferenciación Celular , Células de la Médula Ósea , Células Cultivadas
6.
Mater Today Bio ; 25: 100959, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38327976

RESUMEN

Osteochondral defect repair with a collagen/collagen-magnesium-hydroxyapatite (Col/Col-Mg-HAp) scaffold has demonstrated good clinical results. However, subchondral bone repair remained suboptimal, potentially leading to damage to the regenerated overlying neocartilage. This study aimed to improve the bone repair potential of this scaffold by incorporating newly developed strontium (Sr) ion enriched amorphous calcium phosphate (Sr-ACP) granules (100-150 µm). Sr concentration of Sr-ACP was determined with ICP-MS at 2.49 ± 0.04 wt%. Then 30 wt% ACP or Sr-ACP granules were integrated into the scaffold prototypes. The ACP or Sr-ACP granules were well embedded and distributed in the collagen matrix demonstrated by micro-CT and scanning electron microscopy/energy dispersive x-ray spectrometry. Good cytocompatibility of ACP/Sr-ACP granules and ACP/Sr-ACP enriched scaffolds was confirmed with in vitro cytotoxicity assays. An overall promising early tissue response and good biocompatibility of ACP and Sr-ACP enriched scaffolds were demonstrated in a subcutaneous mouse model. In a goat osteochondral defect model, significantly more bone was observed at 6 months with the treatment of Sr-ACP enriched scaffolds compared to scaffold-only, in particular in the weight-bearing femoral condyle subchondral bone defect. Overall, the incorporation of osteogenic Sr-ACP granules in Col/Col-Mg-HAp scaffolds showed to be a feasible and promising strategy to improve subchondral bone repair.

7.
Radiology ; 269(1): 113-21, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23674789

RESUMEN

PURPOSE: To evaluate the status of articular cartilage and bone in an equine model of spontaneous repair by using the sweep imaging with Fourier transform (SWIFT) magnetic resonance (MR) imaging technique. MATERIALS AND METHODS: Experiments were approved by the Utrecht University Animal Ethics Committee. Six-millimeter-diameter chondral (n = 5) and osteochondral (n = 5, 3-4 mm deep into subchondral bone) defects were created in the intercarpal joints of seven 2-year-old horses and examined with SWIFT at 9.4 T after spontaneous healing for 12 months. Conventional T2 maps and gradient-echo images were obtained for comparison, and histologic assessment of cartilage and micro-computed tomography (CT) of bone were performed for reference. Signal-to-noise ratio (SNR) analysis was performed, and a radiologist evaluated the MR images. Structural bone parameters were derived from SWIFT and micro-CT datasets. Significance of differences was investigated with the Wilcoxon signed rank test and Pearson correlation analysis. RESULTS: SWIFT was able to depict the different outcomes of spontaneous healing of focal chondral versus osteochondral defects. SWIFT produced constant signal intensity throughout cartilage, whereas T2 mapping showed elevated T2 values (P = .06) in repair tissue (mean T2 in superficial region of interest in an osteochondral lesion = 50.0 msec ± 10.2) in comparison to adjacent intact cartilage (mean T2 = 32.7 msec ± 4.2). The relative SNR in the subchondral plate with SWIFT (0.91) was more than four times higher than that with conventional fast spin-echo (0.12) and gradient-echo (0.19) MR imaging. The correlation between bone volume-to-tissue volume fractions determined with SWIFT and micro-CT was significant (r = 0.83, P < .01). CONCLUSION: SWIFT enabled assessment of spontaneous osteochondral repair in an equine model.


Asunto(s)
Curación de Fractura , Fracturas Óseas/patología , Fracturas del Cartílago/patología , Interpretación de Imagen Asistida por Computador/métodos , Articulaciones/lesiones , Articulaciones/patología , Imagen por Resonancia Magnética/métodos , Algoritmos , Animales , Análisis de Fourier , Caballos , Aumento de la Imagen/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
8.
JOR Spine ; 6(3): e1279, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37780829

RESUMEN

Background: A significant hurdle for potential cell-based therapies is the subsequent survival and regenerative capacity of implanted cells. While many exciting developments have demonstrated promise preclinically, cell-based therapies for intervertebral disc (IVD) degeneration fail to translate equivalent clinical efficacy. Aims: This work aims to ascertain the clinical relevance of both a small and large animal model by experimentally investigating and comparing these animal models to human from the perspective of anatomical scale and their cellular metabolic and regenerative potential. Materials and Methods: First, this work experimentally investigated species-specific geometrical scale, native cell density, nutrient metabolism, and matrix synthesis rates for rat, goat, and human disc cells in a 3D microspheroid configuration. Second, these parameters were employed in silico to elucidate species-specific nutrient microenvironments and predict differences in temporal regeneration between animal models. Results: This work presents in silico models which correlate favorably to preclinical literature in terms of the capabilities of animal regeneration and predict that compromised nutrition is not a significant challenge in small animal discs. On the contrary, it highlights a very fine clinical balance between an adequate cell dose for sufficient repair, through de novo matrix deposition, without exacerbating the human microenvironmental niche. Discussion: Overall, this work aims to provide a path towards understanding the effect of cell injection number on the nutrient microenvironment and the "time to regeneration" between preclinical animal models and the large human IVD. While these findings help to explain failed translation of promising preclinical data and the limited results emerging from clinical trials at present, they also enable the research field and clinicians to manage expectations on cell-based regeneration. Conclusion: Ultimately, this work provides a platform to inform the design of clinical trials, and as computing power and software capabilities increase in the future, it is conceivable that generation of patient-specific models could be used for patient assessment, as well as pre- and intraoperative planning.

9.
Animals (Basel) ; 13(20)2023 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-37893914

RESUMEN

This study investigates repeated low-dose lipopolysaccharide (LPS) injections in equine joints as a model for recurrent joint inflammation and its impact on animal welfare. Joint inflammation was induced in eight horses by injecting 0.25 ng of LPS three times at two-week intervals. Welfare scores and clinical parameters were recorded at baseline and over 168 h post-injection. Serial synoviocentesis was performed for the analysis of a panel of synovial fluid biomarkers of inflammation and cartilage turnover. Clinical parameters and a final synoviocentesis were also performed eight weeks after the last sampling point to assess the recovery of normal joint homeostasis. Statistical methods were used to compare the magnitude of response to each of the 3 LPS inductions and to compare the baseline and final measurements. Each LPS injection produced consistent clinical and biomarker responses, with minimal changes in welfare scores. General matrix metalloproteinase (MMP) activity and joint circumference showed greater response to the second LPS induction, but response to the third was comparable to the first. Gylcosaminoglycans (GAG) levels showed a significantly decreased response with each induction, while collagen-cleavage neoepitope of type II collagen (C2C) and carboxypropetide of type II collagen epitope (CPII) showed quicker responses to the second and third inductions. All parameters were comparable to baseline values at the final timepoint. In conclusion, a consistent, reliable intra-articular inflammatory response can be achieved with repeated injections of 0.25 ng LPS, with minimal impact on animal welfare, suggesting potential as a refined translational model of recurrent joint inflammation.

10.
J Funct Biomater ; 14(2)2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36826910

RESUMEN

Despite promising clinical results in osteochondral defect repair, a recently developed bi-layered collagen/collagen-magnesium-hydroxyapatite scaffold has demonstrated less optimal subchondral bone repair. This study aimed to improve the bone repair potential of this scaffold by adsorbing bone morphogenetic protein 2 (BMP-2) and/or platelet-derived growth factor-BB (PDGF-BB) onto said scaffold. The in vitro release kinetics of BMP-2/PDGF-BB demonstrated that PDGF-BB was burst released from the collagen-only layer, whereas BMP-2 was largely retained in both layers. Cell ingrowth was enhanced by BMP-2/PDFG-BB in a bovine osteochondral defect ex vivo model. In an in vivo semi-orthotopic athymic mouse model, adding BMP-2 or PDGF-BB increased tissue repair after four weeks. After eight weeks, most defects were filled with bone tissue. To further investigate the promising effect of BMP-2, a caprine bilateral stifle osteochondral defect model was used where defects were created in weight-bearing femoral condyle and non-weight-bearing trochlear groove locations. After six months, the adsorption of BMP-2 resulted in significantly less bone repair compared with scaffold-only in the femoral condyle defects and a trend to more bone repair in the trochlear groove. Overall, the adsorption of BMP-2 onto a Col/Col-Mg-HAp scaffold reduced bone formation in weight-bearing osteochondral defects, but not in non-weight-bearing osteochondral defects.

11.
Mater Today Bio ; 16: 100343, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35865410

RESUMEN

Articular cartilage defects fail to heal spontaneously, typically progressing to osteoarthritis. Bone marrow stimulation techniques such as microfracture (MFX) are the current surgical standard of care; however MFX typically produces an inferior fibro-cartilaginous tissue which provides only temporary symptomatic relief. Here we implanted solubilised articular cartilage extracellular matrix (ECM) derived scaffolds into critically sized chondral defects in goats, securely anchoring these implants to the joint surface using a 3D-printed fixation device that overcame the need for sutures or glues. In vitro these ECM scaffolds were found to be inherently chondro-inductive, while in vivo they promoted superior articular cartilage regeneration compared to microfracture. In an attempt to further improve the quality of repair, we loaded these scaffolds with a known chemotactic factor, transforming growth factor (TGF)-ß3. In vivo such TGF-ß3 loaded scaffolds promoted superior articular cartilage regeneration. This study demonstrates that ECM derived biomaterials, either alone and particularly when combined with exogenous growth factors, can successfully treat articular cartilage defects in a clinically relevant large animal model.

12.
Acta Biomater ; 143: 266-281, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35278686

RESUMEN

While some clinical advances in cartilage repair have occurred, osteochondral (OC) defect repair remains a significant challenge, with current scaffold-based approaches failing to recapitulate the complex, hierarchical structure of native articular cartilage (AC). To address this need, we fabricated bilayered extracellular matrix (ECM)-derived scaffolds with aligned pore architectures. By modifying the freeze-drying kinetics and controlling the direction of heat transfer during freezing, it was possible to produce anisotropic scaffolds with larger pores which supported homogenous cellular infiltration and improved sulfated glycosaminoglycan deposition. Neo-tissue organization in vitro could also be controlled by altering scaffold pore architecture, with collagen fibres aligning parallel to the long-axis of the pores within scaffolds containing aligned pore networks. Furthermore, we used in vitro and in vivo assays to demonstrate that AC and bone ECM derived scaffolds could preferentially direct the differentiation of mesenchymal stromal cells (MSCs) towards either a chondrogenic or osteogenic lineage respectively, enabling the development of bilayered ECM scaffolds capable of spatially supporting unique tissue phenotypes. Finally, we implanted these scaffolds into a large animal model of OC defect repair. After 6 months in vivo, scaffold implantation was found to improve cartilage matrix deposition, with collagen fibres preferentially aligning parallel to the long axis of the scaffold pores, resulting in a repair tissue that structurally and compositionally was more hyaline-like in nature. These results demonstrate how scaffold architecture and composition can be spatially modulated to direct the regeneration of complex interfaces such as the osteochondral unit, enabling their use as cell-free, off-the-shelf implants for joint regeneration. STATEMENT OF SIGNIFICANCE: The architecture of the extracellular matrix, while integral to tissue function, is often neglected in the design and evaluation of regenerative biomaterials. In this study we developed a bilayered scaffold for osteochondral defect repair consisting of tissue-specific extracellular matrix (ECM)-derived biomaterials to spatially direct stem/progenitor cell differentiation, with a tailored pore microarchitecture to promote the development of a repair tissue that recapitulates the hierarchical structure of native AC. The use of this bilayered scaffold resulted in improved tissue repair outcomes in a large animal model, specifically the ability to guide neo-tissue organization and therefore recapitulate key aspects of the zonal structure of native articular cartilage. These bilayer scaffolds have the potential to become a new therapeutic option for osteochondral defect repair.


Asunto(s)
Cartílago Articular , Andamios del Tejido , Animales , Materiales Biocompatibles/química , Condrogénesis , Colágeno , Matriz Extracelular , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
13.
Front Vet Sci ; 9: 907616, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35812845

RESUMEN

Background: Allogenic mesenchymal stem cell (MSC) secretome is a novel intra-articular therapeutic that has shown promise in in vitro and small animal models and warrants further investigation. Objectives: To investigate if intra-articular allogenic MSC-secretome has anti-inflammatory effects using an equine model of joint inflammation. Study Design: Randomized positively and negatively controlled experimental study. Method: In phase 1, joint inflammation was induced bilaterally in radiocarpal joints of eight horses by injecting 0.25 ng lipopolysaccharide (LPS). After 2 h, the secretome of INFy and TNFα stimulated allogeneic equine MSCs was injected in one randomly assigned joint, while the contralateral joint was injected with medium (negative control). Clinical parameters (composite welfare scores, joint effusion, joint circumference) were recorded, and synovial fluid samples were analyzed for biomarkers (total protein, WBCC; eicosanoid mediators, CCL2; TNFα; MMP; GAGs; C2C; CPII) at fixed post-injection hours (PIH 0, 8, 24, 72, and 168 h). The effects of time and treatment on clinical and synovial fluid parameters and the presence of time-treatment interactions were evaluated. For phase 2, allogeneic MSC-secretome vs. allogeneic equine MSCs (positive control) was tested using a similar methodology. Results: In phase 1, the joint circumference was significantly (p < 0.05) lower in the MSC-secretome treated group compared to the medium control group at PIH 24, and significantly higher peak synovial GAG values were noted at PIH 24 (p < 0.001). In phase 2, no significant differences were noted between the treatment effects of MSC-secretome and MSCs. Main Limitations: This study is a controlled experimental study and therefore cannot fully reflect natural joint disease. In phase 2, two therapeutics are directly compared and there is no negative control. Conclusions: In this model of joint inflammation, intra-articular MSC-secretome injection had some clinical anti-inflammatory effects. An effect on cartilage metabolism, evident as a rise in GAG levels was also noted, although it is unclear whether this could be considered a beneficial or detrimental effect. When directly comparing MSC-secretome to MSCs in this model results were comparable, indicating that MSC-secretome could be a viable off-the-shelf alternative to MSC treatment.

14.
Connect Tissue Res ; 52(5): 380-92, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21405978

RESUMEN

We investigated the effects of lifelong voluntary exercise on articular cartilage of mice. At the age of 4 weeks C57BL mice (n = 152) were divided into two groups, with one group serving as a sedentary control whereas the other was allowed free access to a running wheel from the age of 1 month onward. Mice were euthanized at four different time points (1, 2, 6, and 18 months of age). Articular cartilage samples were gathered from the load-bearing area of the tibial medial plateaus, and osteoarthritis was graded. Additionally, the proteoglycan content distribution was assessed using digital densitometry, collagen fibril orientation, and parallelism with polarized light microscopy, and collagen content using Fourier transform infrared imaging spectroscopy. The incidence of osteoarthritis increased with aging, but exercise had no effect on this trend. Furthermore, the structure and composition revealed significant growth, maturation, and age-dependent properties. Exercise exerted a minor effect on collagen fibril orientation in the superficial zone. Fibril orientation at 2 months of age was more perpendicular to surface (p < 0.05) in controls compared with runners, whereas the situation was reversed at the age of 18 months (p < 0.05). The collagen content of the superficial zone was higher (p < 0.01) at the age of 18 months in controls compared with runners but the proteoglycan content did not display any exercise-dependent changes. In conclusion, growth, maturation, and aging exerted a clear effect on integrity, structure, and composition of medial tibial plateau articular cartilage in mice, whereas lifelong voluntary exercise had only a minor effect on collagen architecture and content.


Asunto(s)
Envejecimiento/fisiología , Cartílago Articular/crecimiento & desarrollo , Actividad Motora/fisiología , Osteoartritis/etiología , Tibia/crecimiento & desarrollo , Animales , Colágeno/ultraestructura , Articulaciones/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoartritis/patología , Carrera , Soporte de Peso
15.
Front Immunol ; 12: 715267, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34659205

RESUMEN

Tissue engineering approaches using progenitor cells such as mesenchymal stromal cells (MSCs) represent a promising strategy to regenerate bone. Previous work has demonstrated the potential of chondrogenically primed human MSCs to recapitulate the process of endochondral ossification and form mature bone in vivo, using immunodeficient xenogeneic models. To further the translation of such MSC-based approaches, additional investigation is required to understand the impact of interactions between human MSC constructs and host immune cells upon the success of MSC-mediated bone formation. Although human MSCs are considered hypoimmunogenic, the potential of chondrogenically primed human MSCs to induce immunogenic responses in vivo, as well as the efficacy of MSC-mediated ectopic bone formation in the presence of fully competent immune system, requires further elucidation. Therefore, the aim of this study was to investigate the capacity of chondrogenically primed human MSC constructs to persist and undergo the process of endochondral ossification in an immune competent xenogeneic model. Chondrogenically differentiated human MSC pellets were subcutaneously implanted to wild-type BALB/c mice and retrieved at 2 and 12 weeks post-implantation. The percentages of CD4+ and CD8+ T cells, B cells, and classical/non-classical monocyte subsets were not altered in the peripheral blood of mice that received chondrogenic MSC constructs compared to sham-operated controls at 2 weeks post-surgery. However, MSC-implanted mice had significantly higher levels of serum total IgG compared to sham-operated mice at this timepoint. Flow cytometric analysis of retrieved MSC constructs identified the presence of T cells and macrophages at 2 and 12 weeks post-implantation, with low levels of immune cell infiltration to implanted MSC constructs detected by CD45 and CD3 immunohistochemical staining. Despite the presence of immune cells in the tissue, MSC constructs persisted in vivo and were not degraded/resorbed. Furthermore, constructs became mineralised, with longitudinal micro-computed tomography imaging revealing an increase in mineralised tissue volume from 4 weeks post-implantation until the experimental endpoint at 12 weeks. These findings indicate that chondrogenically differentiated human MSC pellets can persist and undergo early stages of endochondral ossification following subcutaneous implantation in an immunocompetent xenogeneic model. This scaffold-free model may be further extrapolated to provide mechanistic insight to osteoimmunological processes regulating bone regeneration and homeostasis.


Asunto(s)
Calcificación Fisiológica , Diferenciación Celular , Condrogénesis , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Animales , Biomarcadores , Regeneración Ósea , Diferenciación Celular/genética , Células Cultivadas , Condrogénesis/genética , Humanos , Inmunidad , Ratones , Modelos Animales , Monocitos/inmunología , Monocitos/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Ingeniería de Tejidos , Microtomografía por Rayos X
16.
Equine Vet J ; 53(6): 1277-1286, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33280164

RESUMEN

BACKGROUND: Intra-articular triamcinolone acetonide is a widely used treatment for joint inflammation despite limited scientific evidence of its efficacy. OBJECTIVES: To investigate if intra-articular triamcinolone acetonide has sustained anti-inflammatory effects using an equine model of repeated joint inflammation. STUDY DESIGN: Randomised controlled experimental study. METHOD: For three consecutive cycles 2 weeks apart, inflammation was induced in both middle carpal joints of eight horses by injecting 0.25 ng lipopolysaccharide (LPS). After the first LPS injection only, treatment with 12 mg triamcinolone acetonide (TA) followed in one randomly assigned joint, while the contralateral joint was treated with sterile saline (control). Clinical parameters (composite welfare scores, joint effusion, joint circumference) were recorded and synovial fluid samples were analysed for various biomarkers (total protein, WBCC; PGE2 ; CCL2; TNFα; MMP; GAGs; C2C; CPII) at fixed timepoints (post injection hours 0, 8, 24, 72 and 168). The effects of time and treatment on clinical and synovial fluid parameters and the presence of time-treatment interactions were tested using a linear mixed model for repeated measures with horse as a random effect, and time and treatment as fixed effects. RESULTS: The TA treated joints showed significantly higher peak synovial GAG concentrations (Difference in means 283.1875 µg/mL, 95% CI 179.8, 386.6, P < 0.000), and PGE2 levels (Difference in means 77.8025 pg/mL, 95% CI 21.2, 134.4, P < 0.007) after the first inflammation induction. Significantly lower TP levels were seen with TA treatment after the second induction (Difference in means -7.5 g/L, 95% CI -14.8, -0.20, P < 0.04) . Significantly lower WBCC levels were noted with TA treatment after the first (Difference in means -23.7125 × 109  cells/L, 95% CI -46.7, -0.7, P < 0.04) and second (Difference in means -35.95 × 109  cells/L, 95% CI -59.0, -12.9, P < 0.002) inflammation inductions. Significantly lower general MMP activity was also seen with TA treatment after the second inflammation inductions (Difference in means -51.65 RFU/s, 95% CI -92.4, -10.9, P < 0.01). MAIN LIMITATIONS: This experimental study cannot fully reflect natural joint disease. CONCLUSIONS: In this model, intra-articular TA seems to have some anti-inflammatory activity (demonstrated by reductions in TP, WBCC and general MMP activity) up to 2 weeks post treatment but not at 4 weeks. This anti-inflammatory effect appeared to outlast a shorter-lived, potentially detrimental effect illustrated by increased synovial GAG and PGE2 levels after the first induction.


Asunto(s)
Enfermedades de los Caballos , Triamcinolona Acetonida , Animales , Enfermedades de los Caballos/tratamiento farmacológico , Caballos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/veterinaria , Inyecciones Intraarticulares/veterinaria , Líquido Sinovial , Triamcinolona Acetonida/uso terapéutico
17.
J Anat ; 217(3): 262-74, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20646109

RESUMEN

Articular cartilage composition and structure are maintained and remodeled by chondrocytes under the influence of loading. Exercise-induced changes in the composition, structure, mechanical properties and tissue integrity of growing and aging hamster articular cartilage were investigated. Articular cartilage samples (n = 191) were harvested from the proximal tibiae of hamsters aged 1, 3, 6, 12 and 15 months. The hamsters were divided into runners and controls. The runners had free access to a running wheel between 1 and 3 months (runner groups 3-, 12- and 15-month-old hamsters) or 1 and 6 months (runner group 6-month-old hamsters) of age. Control animals were subjected to a sedentary lifestyle. Mechanical indentation tests and depth-wise compositional and structural analyses were performed for the cartilage samples. Furthermore, the integrity of articular cartilage was assessed using histological osteoarthritis grading. Exercise affected the collagen network organization after a 5-month exercise period, especially in the middle and deep zones. However, no effect on the mechanical properties was detected after exercise. Before the age of 12 months, the runners showed less osteoarthritis than the controls, whereas at 15 months of age the situation was reversed. It is concluded that, in hamsters, physical exercise at a young age enhances cartilage maturation and alters the depth-wise cartilage structure and composition. This may be considered beneficial. However, exercise at a young age demonstrated adverse effects on cartilage at a later age with a significant increase in the incidence of osteoarthritis.


Asunto(s)
Artritis Experimental/prevención & control , Cartílago Articular/fisiología , Actividad Motora/fisiología , Osteoartritis/prevención & control , Envejecimiento/fisiología , Animales , Artritis Experimental/etiología , Artritis Experimental/fisiopatología , Fenómenos Biomecánicos , Cartílago Articular/anatomía & histología , Cartílago Articular/crecimiento & desarrollo , Cartílago Articular/metabolismo , Colágeno/metabolismo , Cricetinae , Femenino , Mesocricetus , Osteoartritis/etiología , Osteoartritis/fisiopatología , Proteoglicanos/metabolismo , Aumento de Peso/fisiología
18.
Acta Biomater ; 113: 130-143, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32505800

RESUMEN

Successful osteochondral defect repair requires regenerating the subchondral bone whilst simultaneously promoting the development of an overlying layer of articular cartilage that is resistant to vascularization and endochondral ossification. During skeletal development articular cartilage also functions as a surface growth plate, which postnatally is replaced by a more spatially complex bone-cartilage interface. Motivated by this developmental process, the hypothesis of this study is that bi-phasic, fibre-reinforced cartilaginous templates can regenerate both the articular cartilage and subchondral bone within osteochondral defects created in caprine joints. To engineer mechanically competent implants, we first compared a range of 3D printed fibre networks (PCL, PLA and PLGA) for their capacity to mechanically reinforce alginate hydrogels whilst simultaneously supporting mesenchymal stem cell (MSC) chondrogenesis in vitro. These mechanically reinforced, MSC-laden alginate hydrogels were then used to engineer the endochondral bone forming phase of bi-phasic osteochondral constructs, with the overlying chondral phase consisting of cartilage tissue engineered using a co-culture of infrapatellar fat pad derived stem/stromal cells (FPSCs) and chondrocytes. Following chondrogenic priming and subcutaneous implantation in nude mice, these bi-phasic cartilaginous constructs were found to support the development of vascularised endochondral bone overlaid by phenotypically stable cartilage. These fibre-reinforced, bi-phasic cartilaginous templates were then evaluated in clinically relevant, large animal (caprine) model of osteochondral defect repair. Although the quality of repair was variable from animal-to-animal, in general more hyaline-like cartilage repair was observed after 6 months in animals treated with bi-phasic constructs compared to animals treated with commercial control scaffolds. This variability in the quality of repair points to the need for further improvements in the design of 3D bioprinted implants for joint regeneration. STATEMENT OF SIGNIFICANCE: Successful osteochondral defect repair requires regenerating the subchondral bone whilst simultaneously promoting the development of an overlying layer of articular cartilage. In this study, we hypothesised that bi-phasic, fibre-reinforced cartilaginous templates could be leveraged to regenerate both the articular cartilage and subchondral bone within osteochondral defects. To this end we used 3D printed fibre networks to mechanically reinforce engineered transient cartilage, which also contained an overlying layer of phenotypically stable cartilage engineered using a co-culture of chondrocytes and stem cells. When chondrogenically primed and implanted into caprine osteochondral defects, these fibre-reinforced bi-phasic cartilaginous grafts were shown to spatially direct tissue development during joint repair. Such developmentally inspired tissue engineering strategies, enabled by advances in biofabrication and 3D printing, could form the basis of new classes of regenerative implants in orthopaedic medicine.


Asunto(s)
Cartílago Articular , Cabras , Impresión Tridimensional , Animales , Regeneración Ósea , Condrogénesis , Ratones , Ratones Desnudos , Ingeniería de Tejidos , Andamios del Tejido
19.
Tissue Eng Part A ; 26(9-10): 490-502, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31797740

RESUMEN

Implantation of chondrogenically differentiated mesenchymal stromal cells (MSCs) leads to bone formation in vivo through the process of endochondral ossification. The use of allogeneic MSCs for this purpose may be a promising new approach to replace the current gold standard of bone regeneration. However, the success of using allogeneic cells depends on the interaction between the implanted cells and the host's endogenous immune cells. Th17 T cells and other CD4 helper T cell subtypes have been shown to negatively impact chondrogenesis, however, it is unclear how the interaction between these cells affects bone regeneration mediated by these cells. The aim of the current work was to assess the effect of chondrogenic MSC pellets on Th1, Th2, Th17, and regulatory T cells in vitro. Human MSCs were nonchondrogenic (-TGFß3) and chondrogenically (+TGFß3) differentiated for 7 or 21 days. Memory T cells (sorted from the CD4 population of peripheral blood mononuclear cells [PBMCs]), as well as total PBMCs were cocultured with allogeneic nonchondrogenic and chondrogenic MSC pellets for 3 days. Seven-day differentiated allogeneic nonchondrogenic and chondrogenic MSC pellets that were cocultured with memory T cells resulted in a significant increase in Th2 and a decrease in Th1 T cells. Furthermore, the co-culture of 21-day differentiated nonchondrogenic and chondrogenic MSC pellets with memory T cells resulted in a significant increase in Th2 and Th17 T cells, as well as a decrease in Th1 and regulatory T cells. Interleukin (IL)-6 was identified as a predominant cytokine involved in this interaction between allogeneic chondrogenically differentiated MSC pellets and memory CD4 T cells, with high levels of IL-6 being secreted in the supernatants of this cocultured condition. The findings of this study highlight the potential of chondrogenically differentiated MSC pellets to alter the ratio of Th1 and Th2 as well as Th17 and regulatory T cell subsets. Additional analysis investigating bone formation by chondrogenically differentiated MSCs in an allogeneic setting may identify a novel role of these T cell subsets in bone regeneration processes mediated by chondrogenically differentiated MSCs. Impact statement Allogeneic mesenchymal stromal cells (MSCs) have the potential to be an off-the-shelf treatment for bone repair. However, the lack of knowledge of the immune cells involved in this process has hampered the progression to the clinic. The current study has shown that allogeneic chondrogenic MSCs have the potential to skew the ratio of specific helper CD4 T cell subsets in vitro. This has now provided insight for future in vivo experiments to investigate the role of these T cell subsets in the early stages of bone regeneration mediated by allogeneic chondrogenic MSCs.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Condrogénesis/fisiología , Células Madre Mesenquimatosas/citología , Linfocitos T Colaboradores-Inductores/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Condrogénesis/genética , Técnicas de Cocultivo , Humanos , Interleucina-6/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo
20.
J Anat ; 215(5): 584-91, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19732210

RESUMEN

The aim of this study was to record growth-related changes in collagen network organization and proteoglycan distribution in intermittently peak-loaded and continuously lower-level-loaded articular cartilage. Cartilage from the proximal phalangeal bone of the equine metacarpophalangeal joint at birth, at 5, 11 and 18 months, and at 6-10 years of age was collected from two sites. Site 1, at the joint margin, is unloaded at slow gaits but is subjected to high-intensity loading during athletic activity; site 2 is a continuously but less intensively loaded site in the centre of the joint. The degree of collagen parallelism was determined with quantitative polarized light microscopy and the parallelism index for collagen fibrils was computed from the cartilage surface to the osteochondral junction. Concurrent changes in the proteoglycan distribution were quantified with digital densitometry. We found that the parallelism index increased significantly with age (up to 90%). At birth, site 2 exhibited a more organized collagen network than site 1. In adult horses this situation was reversed. The superficial and intermediate zones exhibited the greatest reorganization of collagen. Site 1 had a higher proteoglycan content than site 2 at birth but here too the situation was reversed in adult horses. We conclude that large changes in joint loading during growth and maturation in the period from birth to adulthood profoundly affect the architecture of the collagen network in equine cartilage. In addition, the distribution and content of proteoglycans are modified significantly by altered joint use. Intermittent peak-loading with shear seems to induce higher collagen parallelism and a lower proteoglycan content in cartilage than more constant weight-bearing. Therefore, we hypothesize that the formation of mature articular cartilage with a highly parallel collagen network and relatively low proteoglycan content in the peak-loaded area of a joint is needed to withstand intermittent stress and shear, whereas a constantly weight-bearing joint area benefits from lower collagen parallelism and a higher proteoglycan content.


Asunto(s)
Cartílago Articular/crecimiento & desarrollo , Caballos/crecimiento & desarrollo , Proteoglicanos/metabolismo , Envejecimiento/metabolismo , Animales , Cartílago Articular/anatomía & histología , Cartílago Articular/metabolismo , Colágenos Fibrilares , Caballos/anatomía & histología , Caballos/metabolismo , Articulación Metacarpofalángica/anatomía & histología , Articulación Metacarpofalángica/crecimiento & desarrollo , Articulación Metacarpofalángica/metabolismo , Microscopía de Polarización/métodos , Soporte de Peso/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA