Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Tissue Eng Part A ; 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37885209

ABSTRACT

The bioextrusion of mesenchymal stromal cells (MSCs) directly seeded in a bioink enables the production of three-dimensional (3D) constructs, promoting their chondrogenic differentiation. Our study aimed to evaluate the effect of different type I collagen concentrations in the bioink on MSCs' chondrogenic differentiation. We printed 3D constructs using an alginate, gelatin, and fibrinogen-based bioink cellularized with MSCs, with four different quantities of type I collagen addition (0.0, 0.5, 1.0, and 5.0 mg per bioink syringe). We assessed the influence of the bioprinting process, the bioink composition, and the growth factor (TGF-ꞵ1) on the MSCs' survival rate. We confirmed the biocompatibility of the process and the bioinks' cytocompatibility. We evaluated the chondrogenic effects of TGF-ꞵ1 and collagen addition on the MSCs' chondrogenic properties through macroscopic observation, shrinking ratio, reverse transcription polymerase chain reaction, glycosaminoglycan synthesis, histology, and type II collagen immunohistochemistry. The bioink containing 0.5 mg of collagen produces the richest hyaline-like extracellular matrix, presenting itself as a promising tool to recreate the superficial layer of hyaline cartilage. The bioink containing 5.0 mg of collagen enhances the synthesis of a calcified matrix, making it a good candidate for mimicking the calcified cartilaginous layer. Type I collagen thus allows the dose-dependent design of specific hyaline cartilage layers.

2.
Int J Pharm ; 624: 122026, 2022 Aug 25.
Article in English | MEDLINE | ID: mdl-35863592

ABSTRACT

Intra-articular (IA) injection of a chondroprotective candidate may delay the osteoarthritis (OA) course, but its rapid absorption into systemic circulation may limit efficacy and produce untoward effects. We compared the pharmacokinetics (PK) of IA rapamycin injected as sustained release in nanoparticles (NPs) versus a free rapamycin suspension in the rat knee compared to an intravenous (IV) free rapamycin shot taken as a reference. Rats received either a single IV injection of free rapamycin (10 µM) or an IA of free or NPs-loaded rapamycin. After sequential exsanguination (15, 30, 60, 180, 360 min, D1, and D7), knee synovial tissue (ST) and cartilage histology were performed. Blood and ST concentrations (LC-MS/MS), PK parameters (area under the curve: AUC; mean residence time: MRT; elimination half-life: T1/2), and IA biocompatibility were assessed. AUCIV was significantly higher for IV than for both IA injections (AUCIA free and AUCIA NPs), with 4248 vs 28 and 74 µg.min.L-1. For ST parameters, we observed a significant difference between AUCIA free and AUCIA NPs with 3735 and 10513 µg.min.L-1 correspondingly. Articular T1/2 and MRT were higher after NPs than after free rapamycin injection: 57.8 and 5.0 h for T1/2 and 80.6 and 5.5 h for MRT, respectively. Histological analysis revealed no chondral injuries and slight transient synovitis only 3 h after the administration of NPs. In the rat knee, rapamycin-loaded NPs delivery via a single IA injection is biocompatible and prolongs synovium joint residency, diminishes blood levels, and reduces detrimental systemic exposure.


Subject(s)
Nanoparticles , Sirolimus , Animals , Chromatography, Liquid , Injections, Intra-Articular , Knee Joint , Rats , Synovial Membrane , Tandem Mass Spectrometry
3.
Int J Pharm ; 609: 121198, 2021 Nov 20.
Article in English | MEDLINE | ID: mdl-34662644

ABSTRACT

Osteoarthritis (OA) is the most common degenerative joint disease. Rapamycin is a potential candidate for OA treatment by increasing the autophagy process implicated in its physiopathology. To optimize Rapamycin profit and avoid systemic side effects, intra-articular (i.a.) administration appeared helpful. However, Rapamycin's highly hydrophobic nature and low bioavailability made it challenging to develop purpose-made drug delivery systems to overcome these limitations. We developed Rapamycin-loaded nanoparticles (NPs) using poly (lactic-co-glycolic acid) by emulsion/evaporation method. We evaluated these NPs' cytocompatibility towards cartilage (chondrocytes) and synovial membrane cells (synoviocytes) for a potential i.a. administration. The in vitro characterization of Rapamycin-loaded NPs had shown a suitable profile for an i.a. administration. In vitro biocompatibility of NPs was highlighted to 10 µM of Rapamycin for both synoviocytes and chondrocytes, but significant toxicity was observed with higher concentrations. Besides, synoviocytes are more sensitive to Rapamycin-loaded NPs than chondrocytes. Finally, we observed in vitro that an adapted formulated Rapamycin-loaded NPs could be safe at suitable i.a. injection concentrations. The toxic effect of Rapamycin encapsulated in these NPs on both articular cells was dose-dependent. After Rapamycin-loaded NPs i.a. administration, local retention, in situ safety, and systemic release should be evaluated with experimental in vivo models.


Subject(s)
Nanoparticles , Sirolimus , Drug Carriers , Glycols , Injections, Intra-Articular , Nanoparticles/toxicity , Polylactic Acid-Polyglycolic Acid Copolymer , Sirolimus/toxicity
4.
Cells ; 10(1)2020 12 22.
Article in English | MEDLINE | ID: mdl-33374921

ABSTRACT

Hyaline cartilage is deficient in self-healing properties. The early treatment of focal cartilage lesions is a public health challenge to prevent long-term degradation and the occurrence of osteoarthritis. Cartilage tissue engineering represents a promising alternative to the current insufficient surgical solutions. 3D printing is a thriving technology and offers new possibilities for personalized regenerative medicine. Extrusion-based processes permit the deposition of cell-seeded bioinks, in a layer-by-layer manner, allowing mimicry of the native zonal organization of hyaline cartilage. Mesenchymal stem cells (MSCs) are a promising cell source for cartilage tissue engineering. Originally isolated from bone marrow, they can now be derived from many different cell sources (e.g., synovium, dental pulp, Wharton's jelly). Their proliferation and differentiation potential are well characterized, and they possess good chondrogenic potential, making them appropriate candidates for cartilage reconstruction. This review summarizes the different sources, origins, and densities of MSCs used in extrusion-based bioprinting (EBB) processes, as alternatives to chondrocytes. The different bioink constituents and their advantages for producing substitutes mimicking healthy hyaline cartilage is also discussed.


Subject(s)
Bioprinting/methods , Mesenchymal Stem Cells/cytology , Osteoarthritis/therapy , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds , Alginates/therapeutic use , Animals , Cartilage, Articular/cytology , Humans , Hyaline Cartilage/cytology , Hydrogels/therapeutic use
5.
Stem Cell Res Ther ; 11(1): 316, 2020 07 25.
Article in English | MEDLINE | ID: mdl-32711576

ABSTRACT

BACKGROUND: MSCs isolated from bone marrow (BM-MSCs) have well-established chondrogenic potential, but MSCs derived from the synovial membrane (SM-MSCs) and synovial fluid (SF-MSCs) are thought to possess superior chondrogenicity. This study aimed to compare the in vitro immunophenotype and trilineage and chondrogenic potential of BM-MSCs to SM-MSCs and SF-MSCs. METHODS: MSCs were isolated from bone marrow (BM-MSCs), synovial membrane (SM-MSCs), and synovial fluid (SF-MSCs) extracted from the hips (BM) and knees (SM and SF) of advanced OA patients undergoing arthroplasty. Flow cytometric analysis was used at P2 to evaluate cell stemness. The trilinear differentiation test was performed at P2. At P3, MSC-seeded collagen sponges were cultured in chondrogenic medium for 28 days. Chondrogenic gene expression was quantified by qRT-PCR. Finally, the implants were stained to assess the deposition of proteoglycans and type II collagen. RESULTS: Despite variability, the immunophenotyping of BM-MSCs, SM-MSCs, and SF-MSCs was quite similar. All cell types were positive for the expression of stem cell markers and negative for exclusion markers. Additionally, chondrogenic differentiation and hypertrophy were more pronounced in BM-MSCs (ACAN, SOX9, COL2B, and COL10A) than in SF-MSCs, with SM-MSCs having intermediate characteristics. Concerning matrix synthesis, the three cell types were equipotent in terms of GAG content, while BM-MSC ECM synthesis of type II collagen was superior. CONCLUSIONS: Chondrogenic MSCs are easily collected from SM and SF in advanced human OA, but in vitro chondrogenesis that is superior to age-matched BM-MSCs should not be expected. However, due to intra-articular priming, SF-MSCs did not overexpress hypertrophic gene.


Subject(s)
Chondrogenesis , Mesenchymal Stem Cells , Bone Marrow , Bone Marrow Cells , Cell Differentiation , Cells, Cultured , Humans , Synovial Fluid , Synovial Membrane
6.
Stem Cells Int ; 2020: 2487072, 2020.
Article in English | MEDLINE | ID: mdl-32399041

ABSTRACT

3D bioprinting offers interesting opportunities for 3D tissue printing by providing living cells with appropriate scaffolds with a dedicated structure. Biological advances in bioinks are currently promising for cell encapsulation, particularly that of mesenchymal stem cells (MSCs). We present herein the development of cartilage implants by 3D bioprinting that deliver MSCs encapsulated in an original bioink at low concentration. 3D-bioprinted constructs (10 × 10 × 4 mm) were printed using alginate/gelatin/fibrinogen bioink mixed with human bone marrow MSCs. The influence of the bioprinting process and chondrogenic differentiation on MSC metabolism, gene profiles, and extracellular matrix (ECM) production at two different MSC concentrations (1 million or 2 million cells/mL) was assessed on day 28 (D28) by using MTT tests, real-time RT-PCR, and histology and immunohistochemistry, respectively. Then, the effect of the environment (growth factors such as TGF-ß1/3 and/or BMP2 and oxygen tension) on chondrogenicity was evaluated at a 1 M cell/mL concentration on D28 and D56 by measuring mitochondrial activity, chondrogenic gene expression, and the quality of cartilaginous matrix synthesis. We confirmed the safety of bioextrusion and gelation at concentrations of 1 million and 2 million MSC/mL in terms of cellular metabolism. The chondrogenic effect of TGF-ß1 was verified within the substitute on D28 by measuring chondrogenic gene expression and ECM synthesis (glycosaminoglycans and type II collagen) on D28. The 1 M concentration represented the best compromise. We then evaluated the influence of various environmental factors on the substitutes on D28 (differentiation) and D56 (synthesis). Chondrogenic gene expression was maximal on D28 under the influence of TGF-ß1 or TGF-ß3 either alone or in combination with BMP-2. Hypoxia suppressed the expression of hypertrophic and osteogenic genes. ECM synthesis was maximal on D56 for both glycosaminoglycans and type II collagen, particularly in the presence of a combination of TGF-ß1 and BMP-2. Continuous hypoxia did not influence matrix synthesis but significantly reduced the appearance of microcalcifications within the extracellular matrix. The described strategy is very promising for 3D bioprinting by the bioextrusion of an original bioink containing a low concentration of MSCs followed by the culture of the substitutes in hypoxic conditions under the combined influence of TGF-ß1 and BMP-2.

7.
Stem Cell Res Ther ; 9(1): 329, 2018 11 28.
Article in English | MEDLINE | ID: mdl-30486903

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) are found in synovial fluid (SF) and can easily be harvested during arthrocentesis or arthroscopy. However, SF-MSC characterization and chondrogenicity in collagen sponges have been poorly documented as well as their hypothetical in vivo chondroprotective properties with intra-articular injections during experimental osteoarthritis (OA). METHODS: SF-MSCs were isolated from human SF aspirates in patients suffering from advanced OA undergoing total knee joint replacements. SF-MSCs at passage 2 (P2) were characterized by flow cytometry for epitope profiling. SF-MSCs at P2 were subsequently cultured in vitro to assess their multilineage potentials. To assess their chondrogenicity, SF-MSCs at P4 were seeded in collagen sponges for 4 weeks under various oxygen tensions and growth factors combinations to estimate their gene profile and matrix production. Also, SF-MSCs were injected into the joints in a nude rat anterior cruciate ligament transection (ACLT) to macroscopically and histologically assess their possible chondroprotective properties,. RESULTS: We characterized the stemness (CD73+, CD90+, CD105+, CD34-, CD45-) and demonstrated the multilineage potency of SF-MSCs in vitro. Furthermore, the chondrogenic induction (TGF-ß1 ± BMP-2) of these SF-MSCs in collagen sponges demonstrated a good capacity of chondrogenic gene induction and extracellular matrix synthesis. Surprisingly, hypoxia did not enhance matrix synthesis, although it boosted chondrogenic gene expression (ACAN, SOX9, COL2A1). Besides, intra-articular injections of xenogenic SF-MSCs did exert neither chondroprotection nor inflammation in ACLT-induced OA in the rat knee. CONCLUSIONS: Advanced OA SF-MSCs seem better candidates for cell-based constructs conceived for cartilage defects rather than intra-articular injections for diffuse OA.


Subject(s)
Cartilage, Articular/pathology , Mesenchymal Stem Cells/cytology , Osteoarthritis, Knee/pathology , Synovial Fluid/cytology , Wound Healing , Animals , Cells, Cultured , Disease Models, Animal , Glycosaminoglycans/metabolism , Humans , Immunophenotyping , Injections, Intra-Articular , Male , Multipotent Stem Cells/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Nude , Tissue Engineering , Tissue Scaffolds/chemistry
8.
J Tissue Eng Regen Med ; 12(3): e1757-e1766, 2018 03.
Article in English | MEDLINE | ID: mdl-28485490

ABSTRACT

Cartilage tissue engineering is making progress, but the competing available strategies still leave room for improvement and consensual overviews regarding the best combinations of scaffolds and cell sources are limited by the capacity to compare them directly. In addition, because most strategies involve autologous cell transfer, once these are optimized, the resulting implants require individual quality control prior to grafting in order to emphasize patient-to-patient differential responsiveness to engineering processes. Here, cartilage substitutes prepared from human mesenchymal stem cells undergoing chondrogenic differentiation within distinct scaffolds were used as pilot samples to investigate the pertinence of a novel method with the aim of characterizing the implants. The limits and advantages of analysing, by label-free liquid chromatography-coupled matrix-assisted laser desorption and ionization (LC-MALDI) mass spectrometry, the secreted proteome released into culture medium by engineered cartilage tissues were investigated and compared with more classically used methods for biomaterial characterization. This method did not require sacrificing the biomaterials and robustly evidenced their chondrogenic statuses. In more detail, the method highlighted differences between batches prepared from distinct donors. It was adapted to distinct scaffolds and allowed a comparison of the influence of individual engineering steps, such as growth factor combinations and oxygen tension. Finally, it evidenced subtle changes between replicate substitutes within a series, thereby distinguishing the least and most accomplished ones. We conclude that relative quantification of secreted proteins through label-free LC-MALDI will be useful, not only to orientate engineering methodologies, but also to ultimately provide non-invasive quality control of engineered tissue substitutes for the repair of cartilage and possibly other connective tissues.


Subject(s)
Bioengineering , Cartilage, Articular/pathology , Chondrogenesis , Proteomics/methods , Regeneration , Tissue Scaffolds/chemistry , Aged , Aged, 80 and over , Biocompatible Materials/pharmacology , Cell Differentiation/drug effects , Cell Hypoxia/drug effects , Cells, Cultured , Cellular Microenvironment/drug effects , Chondrogenesis/drug effects , Humans , Implants, Experimental , Middle Aged , Proteome/metabolism , Quality Control , Regeneration/drug effects , Staining and Labeling , Tissue Donors
9.
Biomed Mater Eng ; 28(s1): S229-S235, 2017.
Article in English | MEDLINE | ID: mdl-28372299

ABSTRACT

In tissue engineering approaches, the quality of substitutes is a key element to determine its ability to treat cartilage defects. However, in clinical practice, the evaluation of tissue-engineered cartilage substitute quality is not possible due to the invasiveness of the standard procedure, which is to date histology. The aim of this work was to validate a new innovative system performed from two-photon excitation laser adapted to an optical macroscope to evaluate at macroscopic scale the collagen network in cartilage tissue-engineered substitutes in confrontation with gold standard histologic techniques or immunohistochemistry to visualize type II collagen. This system permitted to differentiate the quality of collagen network between ITS and TGF-ß1 treatments. Multiscale large field imaging combined to multimodality approaches (SHG-TCSPC) at macroscopical scale represent an innovative and non-invasive technique to monitor the quality of collagen network in cartilage tissue-engineered substitutes before in vivo implantation.


Subject(s)
Cartilage/anatomy & histology , Chondrocytes/cytology , Collagen Type II/analysis , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Cartilage/chemistry , Cartilage/cytology , Cartilage/growth & development , Chondrocytes/metabolism , Chondrogenesis , Humans , Mesenchymal Stem Cells/metabolism , Transforming Growth Factor beta1/administration & dosage , Transforming Growth Factor beta1/metabolism
10.
Tissue Eng Part A ; 23(17-18): 913-922, 2017 09.
Article in English | MEDLINE | ID: mdl-28385113

ABSTRACT

We examined the respective influence of a sequential or a continuous hypoxia during expansion and transforming growth factor beta 1-driven chondrogenic differentiation of human bone marrow mesenchymal stem cells (MSCs). The differentiation was performed within alginate beads, a classical tool for the implantation of MSCs within the joint. The standard normoxic 2D (expansion) and 3D (differentiation) MSCs cultures served as reference. To determine the quality of chondrogenesis, we analyzed typical markers such as type II and X collagens, SOX9, COMP, versican, and aggrecan mRNAs using polymerase chain reaction and we assessed the production of type II collagen and hypoxia-inducible factor (HIF)-1α by histological stainings. We simultaneously assessed the expression of osteogenic mRNAs (Alkaline Phosphatase, RUNX2, and Osteocalcin) and the presence of micro-calcifications by Alizarin red and Raman spectroscopy. Chondrogenic differentiation is clearly improved by hypoxia in 3D. Best results were obtained when the entire process, that is, 2D expansion and 3D differentiation, was performed under continuous 5% hypoxic condition. In addition, no calcification (hydroxyapatite, proved by RAMAN) was observed after 2D hypoxic expansion even in the case of a normoxic differentiation, in contrast with controls. Finally, a better chondrogenic differentiation of human MSCs is achieved when a reduced oxygen tension is applied during both expansion and differentiation times, avoiding in vitro osteogenic commitment of cells and subsequently the calcification deposition.


Subject(s)
Alginates/chemistry , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondrogenesis/drug effects , Mesenchymal Stem Cells/metabolism , Transforming Growth Factor beta/pharmacology , Aged , Cell Hypoxia/drug effects , Cells, Cultured , Female , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Male , Mesenchymal Stem Cells/cytology , Middle Aged
11.
Stem Cell Res Ther ; 6: 260, 2015 Dec 30.
Article in English | MEDLINE | ID: mdl-26718750

ABSTRACT

BACKGROUND: Due to their intrinsic properties, stem cells are promising tools for new developments in tissue engineering and particularly for cartilage tissue regeneration. Although mesenchymal stromal/stem cells from bone marrow (BM-MSC) have long been the most used stem cell source in cartilage tissue engineering, they have certain limits. Thanks to their properties such as low immunogenicity and particularly chondrogenic differentiation potential, mesenchymal stromal/stem cells from Wharton's jelly (WJ-MSC) promise to be an interesting source of MSC for cartilage tissue engineering. METHODS: In this study, we propose to evaluate chondrogenic potential of WJ-MSC embedded in alginate/hyaluronic acid hydrogel over 28 days. Hydrogels were constructed by the original spraying method. Our main objective was to evaluate chondrogenic differentiation of WJ-MSC on three-dimensional scaffolds, without adding growth factors, at transcript and protein levels. We compared the results to those obtained from standard BM-MSC. RESULTS: After 3 days of culture, WJ-MSC seemed to be adapted to their new three-dimensional environment without any detectable damage. From day 14 and up to 28 days, the proportion of WJ-MSC CD73(+), CD90(+), CD105(+) and CD166(+) decreased significantly compared to monolayer marker expression. Moreover, WJ-MSC and BM-MSC showed different phenotype profiles. After 28 days of scaffold culture, our results showed strong upregulation of cartilage-specific transcript expression. WJ-MSC exhibited greater type II collagen synthesis than BM-MSC at both transcript and protein levels. Furthermore, our work highlighted a relevant result showing that WJ-MSC expressed Runx2 and type X collagen at lower levels than BM-MSC. CONCLUSIONS: Once seeded in the hydrogel scaffold, WJ-MSC and BM-MSC have different profiles of chondrogenic differentiation at both the phenotypic level and matrix synthesis. After 4 weeks, WJ-MSC, embedded in a three-dimensional environment, were able to adapt to their environment and express specific cartilage-related genes and matrix proteins. Today, WJ-MSC represent a real alternative source of stem cells for cartilage tissue engineering.


Subject(s)
Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Tissue Engineering , Adult , Alginates/chemistry , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cartilage/physiology , Cell Differentiation , Cell Survival , Cells, Cultured , Chondrogenesis , Collagen Type II/metabolism , Collagen Type X/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Middle Aged , Phenotype , Regeneration , Wharton Jelly/cytology , Wharton Jelly/metabolism
12.
J Exp Orthop ; 2(1): 8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26914876

ABSTRACT

Damage to the articular cartilage is an important, prevalent, and unsolved clinical issue for the orthopaedic surgeon. This review summarizes innovative basic research approaches that may improve the current understanding of cartilage repair processes and lead to novel therapeutic options. In this regard, new aspects of cartilage tissue engineering with a focus on the choice of the best-suited cell source are presented. The importance of non-destructive cartilage imaging is highlighted with the recent availability of adapted experimental tools such as Second Harmonic Generation (SHG) imaging. Novel insights into cartilage pathophysiology based on the involvement of the infrapatellar fat pad in osteoarthritis are also described. Also, recombinant adeno-associated viral vectors are discussed as clinically adapted, efficient tools for potential gene-based medicines in a variety of articular cartilage disorders. Taken as a whole, such advances in basic research in diverse fields of articular cartilage repair may lead to the development of improved therapies in the clinics for an improved, effective treatment of cartilage lesions in a close future.

13.
PLoS One ; 9(5): e98451, 2014.
Article in English | MEDLINE | ID: mdl-24878844

ABSTRACT

AIM: The aim of this work was the development of successful cell therapy techniques for cartilage engineering. This will depend on the ability to monitor non-invasively transplanted cells, especially mesenchymal stem cells (MSCs) that are promising candidates to regenerate damaged tissues. METHODS: MSCs were labeled with superparamagnetic iron oxide particles (SPIO). We examined the effects of long-term labeling, possible toxicological consequences and the possible influence of progressive concentrations of SPIO on chondrogenic differentiation capacity. RESULTS: No influence of various SPIO concentrations was noted on human bone marrow MSC viability or proliferation. We demonstrated long-term (4 weeks) in vitro retention of SPIO by human bone marrow MSCs seeded in collagenic sponges under TGF-ß1 chondrogenic conditions, detectable by Magnetic Resonance Imaging (MRI) and histology. Chondrogenic differentiation was demonstrated by molecular and histological analysis of labeled and unlabeled cells. Chondrogenic gene expression (COL2A2, ACAN, SOX9, COL10, COMP) was significantly altered in a dose-dependent manner in labeled cells, as were GAG and type II collagen staining. As expected, SPIO induced a dramatic decrease of MRI T2 values of sponges at 7T and 3T, even at low concentrations. CONCLUSIONS: This study clearly demonstrates (1) long-term in vitro MSC traceability using SPIO and MRI and (2) a deleterious dose-dependence of SPIO on TGF-ß1 driven chondrogenesis in collagen sponges. Low concentrations (12.5-25 µg Fe/mL) seem the best compromise to optimize both chondrogenesis and MRI labeling.


Subject(s)
Cell Differentiation/drug effects , Chondrogenesis/drug effects , Ferric Compounds/pharmacology , Mesenchymal Stem Cells/drug effects , Staining and Labeling/methods , Bone Marrow/drug effects , Bone Marrow/metabolism , Cartilage/drug effects , Cartilage/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Collagen Type II/metabolism , Gene Expression/drug effects , Humans , In Vitro Techniques/methods , Magnetic Resonance Imaging/methods , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Transforming Growth Factor beta1/metabolism
14.
J Biomed Mater Res A ; 101(11): 3211-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23554327

ABSTRACT

Nacre (or mother of pearl) can facilitate bone cell differentiation and can speed up their mineralization. Here we report on the capability of nacre to induce differentiation of human bone marrow mesenchymal stem cells (hBM-MSCs) and the production of extracellular matrix. hBM-MSCs were encapsulated in an alginate hydrogel containing different concentrations of powdered nacre and cultured in the same environment until Day 28. Analysis of osteogenic gene expression, histochemistry, second harmonic generation (SHG) microscopy, and Raman scattering spectroscopy were used to characterize the synthesis of the extracellular matrix. In the presence of nacre powder, a significant increase in matrix synthesis from D21 in comparison with pure alginate was observed. Histochemistry revealed the formation of a new tissue composed of collagen fibers in the presence of nacre (immunostaining and SHG), and hydroxyapatite crystals (Raman) in the alginate beads. These results suggest that nacre is efficient in hBM-MSCs differentiation, extracellular matrix production and mineralization in alginate 3D biomaterials.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Mesenchymal Stem Cells/cytology , Nacre/pharmacology , Osteogenesis/drug effects , Aged , Alginates/pharmacology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Collagen Type X/genetics , Collagen Type X/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation/drug effects , Glucuronic Acid/pharmacology , Hexuronic Acids/pharmacology , Humans , Microscopy, Fluorescence, Multiphoton , Microspheres , Middle Aged , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/genetics , Osteopontin/genetics , Osteopontin/metabolism , Powders , Spectrum Analysis, Raman
15.
Biomed Mater Eng ; 22(4): 209-18, 2012.
Article in English | MEDLINE | ID: mdl-22785364

ABSTRACT

Mesenchymal stem cells (MSCs) are regarded as a potential autologous source for cartilage repair, because they can differentiate into chondrocytes by transforming growth factor-beta (TGF-ß) treatment under the 3-dimensional (3-D) culture condition. In addition to these molecular and biochemical methods, the mechanical regulation of differentiation and matrix formation by MSCs is only starting to be considered. Recently, mechanical loading has been shown to induce chondrogenesis of MSCs in vitro. In this study, we investigated the effects of a calibrated agitation on the chondrogenesis of human bone MSCs (MSCs) in a 3-D alginate culture (day 28) and on the maintenance of chondrogenic phenotypes. Biomechanical stimulation of MSCs increased: (i) types 1 and 2 collagen formation; (ii) the expression of chondrogenic markers such as COMP and SOX9; and (iii) the capacity to maintain the chondrogenic phenotypes. Notably, these effects were shown without TGF-ß treatment. These results suggest that a mechanical stimulation could be an efficient method to induce chondrogenic differentiation of MSCs in vitro for cartilage tissue engineering in a 3-D environment. Additionally, it appears that MSCs and chondrocyte responses to mechanical stimulation are not identical.


Subject(s)
Alginates/chemistry , Chondrocytes/cytology , Chondrogenesis , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Cell Differentiation , Cell Survival , Cells, Cultured , Chondrocytes/metabolism , Collagen Type I/analysis , Collagen Type II/analysis , Gene Expression Regulation , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Mesenchymal Stem Cells/metabolism , Mitochondria/metabolism , Weight-Bearing
16.
Biomed Mater Eng ; 22(4): 235-42, 2012.
Article in English | MEDLINE | ID: mdl-22785367

ABSTRACT

AIM: To determine whether viscosupplementation with intra-articular (i.a.) low- or high-molecular-weight hyaluronate (HA) injections influenced both chondral and synovial lesions in rats with surgically-induced OA knee. METHODS: On D0, rats underwent anterior cruciate ligament transection (ACLX) and were divided in 4 groups: sham group, ACLX-saline control group, ACLX-hyaluronate group, ACLX-hylan group. IA injections were performed on D7, D14 and D21. Histological grading of chondral and synovial lesions were performed on D28. Concomitant immunostainings of Caspase3a and Hsp70 were also performed. RESULTS: Articular damages were significantly reduced in both HAs-treated knee joints. In contrast, a significant increase of histological score of synovial inflammation was noted in both ACLX + HAs groups. Apoptotic events significantly decreased as anti-apoptotic Hsp70 expression increased significantly in both HAs groups. CONCLUSION: HAs may exert, independently of its molecular weight, ambivalent properties on articular structures, simultaneously exerting chondroprotective properties and promoting long-term subacute synovitis.


Subject(s)
Biocompatible Materials/therapeutic use , Hyaluronic Acid/analogs & derivatives , Hyaluronic Acid/therapeutic use , Osteoarthritis, Knee/drug therapy , Viscosupplements/therapeutic use , Animals , Apoptosis/drug effects , Biocompatible Materials/administration & dosage , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Hyaluronic Acid/administration & dosage , Injections, Intra-Articular , Knee Joint/drug effects , Knee Joint/pathology , Male , Osteoarthritis, Knee/pathology , Rats , Rats, Wistar , Synovial Fluid/drug effects
17.
Arthritis Res Ther ; 14(2): R60, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22414623

ABSTRACT

INTRODUCTION: We have taken advantage of the large screening capacity of a multiplex immunoassay to better define the respective contribution of articular versus systemic cytokines in experimental arthritis. METHODS: We performed a follow up (from 7 hours to 14 days) multiplex analysis of 24 cytokines in synovial fluid and sera of rats developing Antigen-Induced Arthritis (AIA) and confronted their protein level changes with molecular, biochemical, histological and clinical events occurring in the course of the disease. RESULTS: The time-scheduled findings in arthritic joints correlated with time-dependent changes of cytokine amounts in joint effusions but not with their blood levels. From seven hours after sensitization, high levels of chemokines (MCP-1, MIP1α, GRO/KC, RANTES, eotaxin) were found in synovial fluid of arthritic knees whereas perivascular infiltration occurred in the synovium; local release of inflammatory cytokines (IFNγ, IL-1ß, IL-6) preceded the spreading of inflammation and resulted in progressive degradation of cartilage and bone. Finally a local overexpression of several cytokines/adipocytokines poorly described in arthritis (IL-13, IL-18, leptin) was observed. CONCLUSIONS: Distinct panels of cytokines were found in arthritic fluid during AIA, and the expected effect of mediators correlated well with changes occurring in joint tissues. Moreover, multiplex analysis could be helpful to identify new pathogenic mediators and to elucidate the mechanisms supporting the efficacy of putative targeted therapies.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Experimental/physiopathology , Cartilage, Articular/metabolism , Cartilage, Articular/physiopathology , Cytokines/metabolism , Inflammation Mediators/physiology , Synovial Fluid/metabolism , Animals , Arthritis, Experimental/pathology , Biomarkers/blood , Biomarkers/metabolism , Cartilage, Articular/pathology , Cytokines/blood , Joints/metabolism , Joints/pathology , Joints/physiopathology , Male , Rats , Rats, Wistar
18.
Mol Ther ; 19(10): 1887-95, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21811249

ABSTRACT

Tumor necrosis factor-α (TNF-α), a proinflammatory cytokine, plays a key role in the pathogenesis of many inflammatory diseases, including arthritis. Neutralization of this cytokine by anti-TNF-α antibodies has shown its efficacy in rheumatoid arthritis (RA) and is now widely used. Nevertheless, some patients currently treated with anti-TNF-α remain refractory or become nonresponder to these treatments. In this context, there is a need for new or complementary therapeutic strategies. In this study, we investigated in vitro and in vivo anti-inflammatory potentialities of an anti-TNF-α triplex-forming oligonucleotide (TFO), as judged from effects on two rat arthritis models. The inhibitory activity of this TFO on articular cells (synoviocytes and chondrocytes) was verified and compared to that of small interfering RNA (siRNA) in vitro. The use of the anti-TNF-α TFO as a preventive and local treatment in both acute and chronic arthritis models significantly reduced disease development. Furthermore, the TFO efficiently blocked synovitis and cartilage and bone destruction in the joints. The results presented here provide the first evidence that gene targeting by anti-TNF-α TFO modulates arthritis in vivo, thus providing proof-of-concept that it could be used as therapeutic tool for TNF-α-dependent inflammatory disorders.


Subject(s)
Arthritis/drug therapy , Autoantibodies/therapeutic use , Immunotherapy , Tumor Necrosis Factor-alpha/immunology , Animals , Arthritis/immunology , Autoantibodies/immunology , Cells, Cultured , Disease Models, Animal , RNA, Small Interfering/genetics , Rats , Tumor Necrosis Factor-alpha/genetics
19.
Biomed Mater Eng ; 20(3): 175-81, 2010.
Article in English | MEDLINE | ID: mdl-20930325

ABSTRACT

This study investigated the gene expression profile of human mesenchymal stem cells seeded in collagen sponge for 28 days in three different mediums: (1) basal medium as control containing ITS alone, (2) ITS+TGF-ß1 alone or (3) ITS 1% supplemented sequentially by TGF-ß1 (D3-D14) followed by BMP-2 (D15-D28). Differential expression of 84 genes implicated in chondrogenic and osteogenic differentiation of MSCs was analyzed at D28 by real-time RT-PCR array technology. TGF-ß1 alone down-regulated two genes, CD36 and cathepsin K. Sixteen genes were significantly up-regulated, notably type 2 and type 10 collagens, COMP and Sox9. The sequential combination of TGF-ß1 and BMP-2 produced a similar profile with prominent expression of type 2 collagen and the alkaline phosphatase gene. Interestingly, in this in vitro condition, RUNX2 was not up-regulated, suggesting that the sequential combination of TGF-ß1/BMP2 enhances the hypertrophic chondrogenic profile without turning towards the osteoblastic pathway.


Subject(s)
Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis/physiology , Gene Expression Profiling/methods , Intercellular Signaling Peptides and Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Protein Array Analysis/methods , Biotechnology/methods , Cell Differentiation , Cells, Cultured , Humans
20.
Biomed Mater Eng ; 20(3): 189-94, 2010.
Article in English | MEDLINE | ID: mdl-20930327

ABSTRACT

Due to the actual interest for bioengineering in the osteoarthritis (OA) healing context, researchers need accurate qualitative and quantitative methodologies to evaluate in vivo the integration and functionality of their cartilage-like biomaterials. As in clinical diagnostic strategies, advances in Magnetic Resonance Imaging (MRI) seem promising for non-vulnerant assessments of articular cartilage bio-architecture and morphology in small animal models. These experimental models are commonly used to monitor the physiopathology of OA and to evaluate therapeutic responses mediated by chondroprotective drugs or tissue engineering. Nowadays, the application of MR protocols to in vivo small animal cartilage imaging is achievable with the development of high magnetic fields and the adaptation of methodologies to reach the required spatial resolution and contrast. The purpose of this article is to summarize these current MRI strategies used for in vivo small animal articular cartilage assessments.


Subject(s)
Cartilage, Articular/pathology , Cell Tracking/methods , Image Enhancement/methods , Magnetic Resonance Imaging/methods , Magnetic Resonance Imaging/veterinary , Osteoarthritis/pathology , Animals , Mice , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...