Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Osteoarthritis Cartilage ; 30(6): 875-885, 2022 06.
Article in English | MEDLINE | ID: mdl-35307533

ABSTRACT

OBJECTIVE: In light of the role of immune cells in OA pathogenesis, the development of sophisticated animal models closely mimicking the immune dysregulation during the disease development and progression could be instrumental for the preclinical evaluation of novel treatments. Among these models, immunologically humanized mice may represent a relevant system, particularly for testing immune-interacting DMOADs or cell therapies before their transfer to the clinic. Our objective, therefore, was to develop an experimental model of OA by destabilization of the medial meniscus (DMM) in humanized mice. METHOD: Irradiated 5-week-old NOD/LtSz-scid IL2Rγnull (NSG) mice were humanized by intravenous injection of CD34+ human hematopoietic stem cells. The engraftment efficiency was evaluated by flow cytometry 17 weeks after the humanization procedure. Humanized and non-humanized NSG mice underwent DMM or sham surgery and OA development was assessed 1, 6, and 12 weeks after the surgery. RESULTS: 120 days after the humanization, human T and B lymphocytes, macrophages and NK cells, were present in the blood and spleen of the humanized NSG mice. The DMM surgery induced articular cartilage and meniscal alterations associated with an increase in OA and the meniscal score. Moreover, the surgery triggered an inflammatory response that was sustained at a low grade in the DMM group. CONCLUSIONS: Our study shows for the first time the feasibility of inducing OA by DMM in humanized mice. This novel OA model could constitute a useful tool to bridge the gap between the preclinical and clinical evaluation of immune interacting DMOADs and cell-based therapies.


Subject(s)
Cartilage, Articular , Osteoarthritis , Animals , Cartilage, Articular/pathology , Disease Models, Animal , Menisci, Tibial/pathology , Menisci, Tibial/surgery , Mice , Mice, Inbred NOD , Osteoarthritis/pathology
2.
Osteoarthritis Cartilage ; 28(10): 1373-1384, 2020 10.
Article in English | MEDLINE | ID: mdl-32659345

ABSTRACT

OBJECTIVE: TGFß is a key player in cartilage homeostasis and OA pathology. However, few data are available on the role of TGFß signalling in the different OA phenotypes. Here, we analysed the TGFß pathway by transcriptomic analysis in six mouse models of OA. METHOD: We have brought together seven expert laboratories in OA pathophysiology and, used inter-laboratories standard operating procedures and quality controls to increase experimental reproducibility and decrease bias. As none of the available OA models covers the complexity and heterogeneity of the human disease, we used six different murine models of knee OA: from post-traumatic/mechanical models (meniscectomy (MNX), MNX and hypergravity (HG-MNX), MNX and high fat diet (HF-MNX), MNX and seipin knock-out (SP-MNX)) to aging-related OA and inflammatory OA (collagenase-induced OA (CIOA)). Four controls (MNX-sham, young, SP-sham, CIOA-sham) were added. OsteoArthritis Research Society International (OARSI)-based scoring of femoral condyles and ribonucleic acid (RNA) extraction from tibial plateau samples were done by single operators as well as the transcriptomic analysis of the TGFß family pathway by Custom TaqMan® Array Microfluidic Cards. RESULTS: The transcriptomic analysis revealed specific gene signatures in each of the six models; however, no gene was deregulated in all six OA models. Of interest, we found that the combinatorial Gdf5-Cd36-Ltbp4 signature might discriminate distinct subgroups of OA: Cd36 upregulation is a hallmark of MNX-related OA while Gdf5 and Ltbp4 upregulation is related to MNX-induced OA and CIOA. CONCLUSION: These findings stress the OA animal model heterogeneity and the need of caution when extrapolating results from one model to another.


Subject(s)
CD36 Antigens/genetics , Disease Models, Animal , Growth Differentiation Factor 5/genetics , Latent TGF-beta Binding Proteins/genetics , Mice , Osteoarthritis/genetics , Transforming Growth Factor beta/genetics , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Arthritis, Experimental/physiopathology , Collagenases , Diet, High-Fat , GTP-Binding Protein gamma Subunits/genetics , Gene Expression Profiling , Hypergravity , Meniscectomy , Metabolic Syndrome , Mice, Knockout , Obesity , Osteoarthritis/metabolism , Osteoarthritis/physiopathology , Transcriptome , Transforming Growth Factor beta/metabolism
3.
Haemophilia ; 24(5): 703-710, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29989307

ABSTRACT

Haemophilia is characterized by a congenital deficiency of clotting factor VIII or IX. One of the consequences of haemophilia is joint bleedings. Repetitive haemathroses induce cartilage damage and chronic synovitis leading to joint deterioration, and to definitive haemophilic arthropathy which is source of walking disability. Three-dimension gait analysis (3DGA) appears particularly relevant in the case of haemophilia because it allows an evaluation of several joints in weight-bearing situations. The purpose of this study was to review the interest and the contribution of 3DGA in the management of patients with haemophilia. The greatest interest of gait analysis would be to detect early walking changes with a non-invasive and well-tolerated examination, especially in paediatric population. In adulthood, this technic may be also useful to help detect walking worsening in patients known to have already arthropathy. However, it takes time to realize and needs expensive equipment, which limits its possibility of routine use. Although generalizations of these results remain difficult, especially to compare patients with haemophilia to normal population. Indeed, in the studies, patient groups are small and usually heterogeneous in terms of age and target joints. It certainly results of the rarity of the disease. So, it could be interesting to perform a study with a larger cohort in order to allow subgroup analysis, helping to define clearly the place of 3DGA in the strategy of haemophilia evaluation.


Subject(s)
Gait Analysis/methods , Hemophilia A/complications , Adolescent , Adult , Child , Female , Hemophilia A/pathology , Humans , Male , Young Adult
4.
J Tissue Eng Regen Med ; 11(4): 1152-1164, 2017 04.
Article in English | MEDLINE | ID: mdl-25824373

ABSTRACT

The development of biologically and mechanically competent hydrogels is a prerequisite in cartilage engineering. We recently demonstrated that a marine exopolysaccharide, GY785, stimulates the in vitro chondrogenesis of adipose stromal cells. In the present study, we thus hypothesized that enriching our silated hydroxypropyl methylcellulose hydrogel (Si-HPMC) with GY785 might offer new prospects in the development of scaffolds for cartilage regeneration. The interaction properties of GY785 with growth factors was tested by surface plasmon resonance (SPR). The biocompatibility of Si-HPMC/GY785 towards rabbit articular chondrocytes (RACs) and its ability to maintain and recover a chondrocytic phenotype were then evaluated in vitro by MTS assay, cell counting and qRT-PCR. Finally, we evaluated the potential of Si-HPMC/GY785 associated with RACs to form cartilaginous tissue in vivo by transplantation into the subcutis of nude mice for 3 weeks. Our SPR data indicated that GY785 was able to physically interact with BMP-2 and TGFß. Our analyses also showed that three-dimensionally (3D)-cultured RACs into Si-HPMC/GY785 strongly expressed type II collagen (COL2) and aggrecan transcripts when compared to Si-HPMC alone. In addition, RACs also produced large amounts of extracellular matrix (ECM) containing glycosaminoglycans (GAG) and COL2. When dedifferentiated RACs were replaced in 3D in Si-HPMC/GY785, the expressions of COL2 and aggrecan transcripts were recovered and that of type I collagen decreased. Immunohistological analyses of Si-HPMC/GY785 constructs transplanted into nude mice revealed the production of a cartilage-like extracellular matrix (ECM) containing high amounts of GAG and COL2. These results indicate that GY785-enriched Si-HPMC appears to be a promising hydrogel for cartilage tissue engineering. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Biocompatible Materials/pharmacology , Cartilage, Articular/cytology , Cellulose/pharmacology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Polysaccharides/pharmacology , Tissue Engineering/methods , Animals , Cartilage, Articular/drug effects , Cell Death/drug effects , Cell Dedifferentiation/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Phenotype , Rabbits , Rheology
5.
Ann Phys Rehabil Med ; 59(3): 139-144, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27079583

ABSTRACT

Articular cartilage is a non-vascularized and poorly cellularized connective tissue that is frequently damaged as a result of trauma and degenerative joint diseases such as osteoarthrtis. Because of the absence of vascularization, articular cartilage has low capacity for spontaneous repair. Today, and despite a large number of preclinical data, no therapy capable of restoring the healthy structure and function of damaged articular cartilage is clinically available. Tissue-engineering strategies involving the combination of cells, scaffolding biomaterials and bioactive agents have been of interest notably for the repair of damaged articular cartilage. During the last 30 years, cartilage tissue engineering has evolved from the treatment of focal lesions of articular cartilage to the development of strategies targeting the osteoarthritis process. In this review, we focus on the different aspects of tissue engineering applied to cartilage engineering. We first discuss cells, biomaterials and biological or environmental factors instrumental to the development of cartilage tissue engineering, then review the potential development of cartilage engineering strategies targeting new emerging pathogenic mechanisms of osteoarthritis.


Subject(s)
Cartilage, Articular/surgery , Osteoarthritis/surgery , Tissue Engineering/methods , Biocompatible Materials/therapeutic use , Humans , Stem Cells
6.
Eur J Nucl Med Mol Imaging ; 39(7): 1169-72, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22398956

ABSTRACT

PURPOSE: A rabbit model of osteochondral defects (OD) and spontaneous healing was longitudinally followed over 12 weeks, by in vivo joint scintigraphy using (99m)Tc-NTP 15-5, and histology. METHODS: We used two models, one with one OD (OD1 group) in the femoral condyle of one knee and the other with two ODs (OD2 group) in the femoral condyle of one knee, with the contralateral knees serving as the reference. A serial longitudinal imaging study was performed with the scintigraphic ratio (SR, operated knee uptake/contralateral knee uptake) determined at each time-point. RESULTS: ODs were imaged as radioactive defects. The SR was decreased with respective to controls, with values of 0.73 ± 0.08 and 0.65 ± 0.07 in the OD1 and OD2 groups, respectively, at 4 weeks after surgery. Histology of both OD groups revealed the presence of repair tissue characterized by a small amount of sulphated glycosaminoglycans and collagen. CONCLUSION: (99m)Tc-NTP 15-5 imaging provided quantitative criteria useful for in vivo evaluation of cartilage trauma and healing.


Subject(s)
Cartilage/diagnostic imaging , Cartilage/surgery , Heterocyclic Compounds, 1-Ring , Knee Injuries/diagnostic imaging , Knee Injuries/surgery , Quaternary Ammonium Compounds , Radionuclide Imaging/methods , Technetium Compounds , Wound Healing/physiology , Animals , Collagen/metabolism , Disease Models, Animal , Glycosaminoglycans/metabolism , Knee Joint/diagnostic imaging , Knee Joint/surgery , Longitudinal Studies , Rabbits , Radiopharmaceuticals
7.
Biomed Mater Eng ; 20(3): 159-66, 2010.
Article in English | MEDLINE | ID: mdl-20930323

ABSTRACT

Articular cartilage does not repair itself spontaneously. To promote its repair, the transfer of stem cells from adipose tissue (ATSC) using an injectable self-setting cellulosic-hydrogel (Si-HPMC) appears promising. In this context, the objective of this work was to investigate the influence of in vitro chondrogenic differentiation of ATSC on the in vivo cartilage formation when combined with Si-HPMC. In a first set of experiments, we characterized ATSC for their ability to proliferate, self renew and express typical mesenchymal stem cell surface markers. Then, the potential of ATSC to differentiate towards the chondrogenic lineage and the optimal culture conditions to drive this differentiation were evaluated. Real-time RT-PCR and histological analysis for sulphated glycosaminoglycans and type II collagen revealed that 3-dimensional culture and hypoxic condition favored ATSC chondrogenesis regarding mRNA expression level and the corresponding proteins production. In order to assess the phenotypic stability of chondrogenically-differentiated ATSC, real-time RT-PCR for specific terminal chondrogenic markers and alkaline phosphatase activity assay were performed. In addition to promote chondrogenesis, our culture conditions seem to prevent the terminal differentiation of ATSC. Histological examination of ATSC/Si-HPMC implants suggested that the in vitro chondrogenic pre-commitment of ATSC in monolayer is sufficient to obtain cartilaginous tissue in vivo.


Subject(s)
Cartilage, Articular/cytology , Cartilage, Articular/growth & development , Cellulose/chemistry , Chondrocytes/cytology , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Animals , Biocompatible Materials/chemistry , Cell Culture Techniques/methods , Cell Differentiation , Cells, Cultured , Chondrocytes/physiology , Humans , Materials Testing , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Mice , Mice, Nude
8.
Biotechnol Bioeng ; 102(4): 1259-67, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-18949749

ABSTRACT

Articular cartilage has a low capacity for spontaneous repair. To promote the repair of this tissue, the transfer of autologous chondrocytes using a three-dimensional matrix appears promising. In this context, the aim of the present work was to investigate the potential use of autologous rabbit nasal chondrocytes (RNC) associated with an injectable self-setting cellulose-based hydrogel (Si-HPMC). Firstly, the influence of Si-HPMC on chondrocytic phenotype was investigated by real-time PCR for specific chondrocyte markers (type II collagen and aggrecan) and type I collagen. Thereafter, autologous RNC were amplified in vitro for 4 weeks before transplantation with Si-HPMC into a rabbit articular cartilage defect followed by analysis 6 weeks later. Implants were histologically characterized for the presence of sulfated GAG and type II collagen. Transcripts analysis indicated that dedifferentiated RNC recovered expression of the main chondrocytic markers after in vitro three-dimensional culture within Si-HPMC. Histological analysis of autologous RNC transplanted in an articular cartilage defect revealed the formation of repair tissue with a histological organization similar to that of healthy articular cartilage. In addition, immunohistological analysis of type II collagen suggested that the repair tissue was a hyaline-like cartilage. Si-HPMC hydrogel associated with nasal chondrocytes therefore appears a promising injectable tissue engineering device for the repair of articular cartilage.


Subject(s)
Cartilage, Articular/injuries , Chondrocytes/transplantation , Hydrogel, Polyethylene Glycol Dimethacrylate/therapeutic use , Tissue Engineering/methods , Transplantation, Autologous/methods , Animals , Cells, Cultured , Chondrocytes/physiology , Collagen Type II/metabolism , Gene Expression Profiling , Glycosaminoglycans/metabolism , Injections , Rabbits
9.
J Biomed Mater Res A ; 89(1): 176-85, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18431767

ABSTRACT

Hybrid constructs associating a biodegradable matrix and autologous chondrocytes hold promise for the treatment of articular cartilage defects. In this context, our objective was to investigate the potential use of nasal chondrocytes associated with a fibrin sealant for the treatment of articular cartilage defects. The phenotype of primary nasal chondrocytes (NC) from human (HNC) and rabbit (RNC) origin were characterized by RT-PCR. The ability of constructs associating fibrin sealant and NC to form a cartilaginous tissue in vivo was investigated, firstly in a subcutaneous site in nude mice and secondly in an articular cartilage defect in rabbit. HNC express type II collagen and aggrecan, the two major hallmarks of a chondrocytic phenotype. Furthermore, when injected subcutaneously into nude mice within a fibrin sealant, these chondrocytes were able to form a cartilage-like tissue. Our data indicate that RNC also express type II collagen and aggrecan and maintained their phenotype in three-dimensional culture within a fibrin sealant. Moreover, treatment of rabbit articular cartilage defects with autologous RNC embedded in a fibrin sealant led to the formation of a hyalin-like repair tissue. The use of fibrin sealant containing hybrid autologous NC therefore appears as a promising approach for cell-based therapy of articular cartilage.


Subject(s)
Chondrocytes/physiology , Fibrin Tissue Adhesive/metabolism , Nasal Septum/cytology , Tissue Engineering/methods , Aggrecans/genetics , Aggrecans/metabolism , Animals , Biocompatible Materials/metabolism , Cartilage, Articular/cytology , Cartilage, Articular/pathology , Cell Culture Techniques , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/transplantation , Collagen Type II/genetics , Collagen Type II/metabolism , Guided Tissue Regeneration/methods , Humans , Implants, Experimental , Mice , Mice, Nude , Phenotype , Rabbits , Transplantation, Autologous
10.
J Biomed Mater Res A ; 80(1): 66-74, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16958048

ABSTRACT

Tissue engineering strategies, based on developing three-dimensional scaffolds capable of transferring autologous chondrogenic cells, holds promise for the restoration of damaged cartilage. In this study, the authors aimed at determining whether a recently developed silanized hydroxypropyl methylcellulose (Si-HPMC) hydrogel can be a suitable scaffold for human nasal chondrocytes (HNC)-based cartilage engineering. Methyltetrazolium salt assay and cell counting experiments first revealed that Si-HPMC enabled the proliferation of HNC. Cell tracker green staining further demonstrated that HNC were able to form nodular structures in this three-dimensional scaffold. HNC phenotype was then assessed by RT-PCR analysis of type II collagen and aggrecan expression as well as alcian blue staining of extracellular matrix. Our data indicated that Si-HPMC allowed the maintenance and the recovery of a chondrocytic phenotype. The ability of constructs HNC/Si-HPMC to form a cartilaginous tissue in vivo was finally investigated after 3 weeks of implantation in subcutaneous pockets of nude mice. Histological examination of the engineered constructs revealed the formation of a cartilage-like tissue with an extracellular matrix containing glycosaminoglycans and type II collagen. The whole of these results demonstrate that Si-HPMC hydrogel associated to HNC is a convenient approach for cartilage tissue engineering.


Subject(s)
Cartilage/metabolism , Chondrocytes/metabolism , Hydrogels , Methylcellulose/analogs & derivatives , Tissue Engineering , Aggrecans/biosynthesis , Cartilage/cytology , Cartilage/injuries , Cell Culture Techniques , Cells, Cultured , Chondrocytes/cytology , Collagen Type II/biosynthesis , Extracellular Matrix/metabolism , Gene Expression Regulation , Humans , Hydrogels/chemistry , Hypromellose Derivatives , Methylcellulose/chemistry , Nasal Mucosa/metabolism , Nose/cytology
11.
Biomed Mater Eng ; 16(4 Suppl): S107-13, 2006.
Article in English | MEDLINE | ID: mdl-16823101

ABSTRACT

Tissue engineering is an emerging field of regenerative medicine which holds promise for the restoration of tissues and organs affected by chronic diseases, age-linked degeneration, congenital deformity and trauma. During the past decade, tissue engineering has evolved from the use of naked biomaterials, which may just replace small area of damaged tissue, to the use of controlled three-dimensional scaffolds in which cells can be seeded before implantation. These cellularized constructs aims at being functionally equal to the unaffected tissue and could make possible the regeneration of large tissue defects. Among the recently developed scaffolds for tissue engineering, polymeric hydrogels have proven satisfactory in cartilage and bone repair. Major technological progress and advances in basic knowledge (physiology and developmental biology) are today necessary to bring this proof of concept to clinical reality. The present review focuses on the recent advances in hydrogel-based tissue engineered constructs potentially utilizable in bone and cartilage regenerative medicine.


Subject(s)
Biocompatible Materials/chemistry , Cartilage/chemistry , Hydrogels/chemistry , Regenerative Medicine/methods , Tissue Engineering/methods , Animals , Biomedical Engineering/methods , Bone Regeneration , Bone Substitutes , Bone and Bones/pathology , Cartilage/metabolism , Connective Tissue/metabolism , Humans
12.
Bioessays ; 27(7): 708-16, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15954094

ABSTRACT

Vascular calcifications are the consequence of several pathological conditions such as atherosclerosis, diabetes, hypercholesterolemia and chronic renal insufficiency. They are associated with risks of amputation, ischemic heart disease, stroke and increased mortality. A growing body of evidence indicates that vascular smooth muscle cells (VSMCs) undergo chondrogenic commitment eventually leading to vascular calcification, by mechanisms similar to those governing ossification in the cartilage growth plate. Our knowledge of the formation of cartilage growth plate can therefore help us to understand why and how arteries calcify and, consequently, develop new therapeutic strategies. Reciprocally, thorough consideration of the events leading to ectopic chondrocyte differentiation appears crucial to further increase our understanding of growth plate formation. In this context, we will review the effects of known or suspected factors that promote chondrogenic differentiation in growth plate and arteries.


Subject(s)
Arteries/cytology , Cartilage/physiology , Growth Plate/cytology , Muscle, Smooth, Vascular/cytology , Animals , Apoptosis , Arteries/pathology , Arteriosclerosis , Cartilage/metabolism , Cell Death , Cell Differentiation , Chondrocytes/metabolism , Humans , Mesoderm/metabolism , Models, Biological , Myocytes, Smooth Muscle/cytology
13.
Biomaterials ; 26(33): 6643-51, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15950277

ABSTRACT

Articular cartilage has limited intrinsic repair capacity. In order to promote cartilage repair, the amplification and transfer of autologous chondrocytes using three-dimensional scaffolds have been proposed. We have developed an injectable and self-setting hydrogel consisting of hydroxypropyl methylcellulose grafted with silanol groups (Si-HPMC). The aim of the present work is to assess both the in vitro cytocompatibility of this hydrogel and its ability to maintain a chondrocyte-specific phenotype. Primary chondrocytes isolated from rabbit articular cartilage (RAC) and two human chondrocytic cell lines (SW1353 and C28/I2) were cultured into the hydrogel. Methyl tetrazolium salt (MTS) assay and cell counting indicated that Si-HPMC hydrogel did not affect respectively chondrocyte viability and proliferation. Fluorescent microscopic observations of RAC and C28/I2 chondrocytes double-labeled with cell tracker green and ethidium homodimer-1 revealed that chondrocytes proliferated within Si-HPMC. Phenotypic analysis (RT-PCR and Alcian blue staining) indicates that chondrocytes, when three-dimensionnally cultured within Si-HPMC, expressed transcripts encoding type II collagen and aggrecan and produced sulfated glycosaminoglycans. These results show that Si-HPMC allows the growth of differentiated chondrocytes. Si-HPMC therefore appears as a potential scaffold for three-dimensional amplification and transfer of chondrocytes in cartilage tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Hydrogels/chemistry , Methylcellulose/analogs & derivatives , Silanes/chemistry , Animals , Cartilage/metabolism , Cartilage, Articular/cytology , Cell Proliferation , Cell Survival , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Dimerization , Glycosaminoglycans/chemistry , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Hypromellose Derivatives , Methylcellulose/chemistry , Microscopy, Fluorescence , Phenotype , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tissue Engineering
SELECTION OF CITATIONS
SEARCH DETAIL
...