Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 15(1): e16218, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36507558

ABSTRACT

We showed that the chemokine receptor C-X-C Motif Chemokine Receptor 2 (CXCR2) is essential for cartilage homeostasis. Here, we reveal that the CXCR2 ligand granulocyte chemotactic protein 2 (GCP-2) was expressed, during embryonic development, within the prospective permanent articular cartilage, but not in the epiphyseal cartilage destined to be replaced by bone. GCP-2 expression was retained in adult articular cartilage. GCP-2 loss-of-function inhibited extracellular matrix production. GCP-2 treatment promoted chondrogenesis in vitro and in human cartilage organoids implanted in nude mice in vivo. To exploit the chondrogenic activity of GCP-2, we disrupted its chemotactic activity, by mutagenizing a glycosaminoglycan binding sequence, which we hypothesized to be required for the formation of a GCP-2 haptotactic gradient on endothelia. This mutated version (GCP-2-T) had reduced capacity to induce transendothelial migration in vitro and in vivo, without affecting downstream receptor signaling through AKT, and chondrogenic activity. Intra-articular adenoviral overexpression of GCP-2-T, but not wild-type GCP-2, reduced pain and cartilage loss in instability-induced osteoarthritis in mice. We suggest that GCP-2-T may be used for disease modification in osteoarthritis.


Subject(s)
Chemokine CXCL6 , Osteoarthritis , Humans , Animals , Mice , Chemokines, CXC/metabolism , Chemokines, CXC/pharmacology , Mice, Nude , Prospective Studies , Receptors, Chemokine , Chondrogenesis
2.
Methods Mol Biol ; 2598: 337-344, 2023.
Article in English | MEDLINE | ID: mdl-36355303

ABSTRACT

Chondral defects are common and disabling. The development of pharmacological approaches for cartilage repair requires the availability of in vivo models which are amenable for gain and loss of function and ideally to genetic modification. In this chapter, we describe a method to induce full-thickness cartilage defects which, in young DBA/1 mice, heal spontaneously, but fail to heal in C57BL/6 mice of the same age or in aged DBA/1 mice. This model (or variants) has been used for genetic screenings to identify genes associated to repair capacity, to study stem cells involved in cartilage repair, and to study the function of molecules involved in repair mechanisms.


Subject(s)
Cartilage, Articular , Mice , Animals , Mice, Inbred DBA , Mice, Inbred C57BL , Disease Models, Animal
3.
Lab Anim (NY) ; 51(4): 103-120, 2022 04.
Article in English | MEDLINE | ID: mdl-35361989

ABSTRACT

Cartilage regeneration is a priority in medicine for the treatment of osteoarthritis and isolated cartilage defects. Several molecules with potential for cartilage regeneration are under investigation. Unfortunately, in vitro chondrogenesis assays do not always predict the stability of the newly formed cartilage in vivo. Therefore, there is a need for a stringent, quantifiable assay to assess in vivo the capacity of molecules to promote the stable formation of cartilage that is resistant to calcification and endochondral bone formation. We developed an ectopic cartilage formation assay (ECFA) that enables one to assess the capacity of bioactive molecules to support cartilage formation in vivo using cartilage organoids. The ECFA predicted good clinical outcomes when used as a quality control for efficacy of chondrocyte preparations before implantation in patients with cartilage defects. In this assay, articular chondrocytes from human donors or animals are injected either intramuscularly or subcutaneously in nude mice. As early as 2 weeks later, cartilage organoids can be retrieved. The size of the implants and their degree of differentiation can be assessed by histomorphometry, immunostainings of molecular markers and real-time PCR. Mineralization can be assessed by micro-computed tomography or by staining. The effects of molecules on cartilage formation can be tested following the systemic administration of the molecule in mice previously injected with chondrocytes, or after co-injection of chondrocytes with cell lines overexpressing and secreting the protein of interest. Here we describe the ECFA procedure, including steps for harvesting human and bovine articular cartilage, isolating primary chondrocytes, preparing overexpression cell lines, injecting the cells intramuscularly and retrieving the implants. This assay can be performed by technicians and researchers with appropriate animal training within 3 weeks.


Subject(s)
Cartilage, Articular , Chondrogenesis , Animals , Cartilage, Articular/metabolism , Cattle , Chondrocytes/metabolism , Humans , Mice , Mice, Nude , X-Ray Microtomography
4.
J Extracell Vesicles ; 10(7): e12088, 2021 05.
Article in English | MEDLINE | ID: mdl-34025953

ABSTRACT

Cartilage defects repair poorly. Recent genetic studies suggest that WNT3a may contribute to cartilage regeneration, however the dense, avascular cartilage extracellular matrix limits its penetration and signalling to chondrocytes. Extracellular vesicles actively penetrate intact cartilage. This study investigates the effect of delivering WNT3a into large cartilage defects in vivo using exosomes as a delivery vehicle. Exosomes were purified by ultracentrifugation from conditioned medium of either L-cells overexpressing WNT3a or control un-transduced L-cells, and characterized by electron microscopy, nanoparticle tracking analysis and marker profiling. WNT3a loaded on exosomes was quantified by western blotting and functionally characterized in vitro using the SUPER8TOPFlash reporter assay and other established readouts including proliferation and proteoglycan content. In vivo pathway activation was assessed using TCF/Lef:H2B-GFP reporter mice. Wnt3a loaded exosomes were injected into the knees of mice, in which large osteochondral defects were surgically generated. The degree of repair was histologically scored after 8 weeks. WNT3a was successfully loaded on exosomes and resulted in activation of WNT signalling in vitro. In vivo, recombinant WNT3a failed to activate WNT signalling in cartilage, whereas a single administration of WNT3a loaded exosomes activated canonical WNT signalling for at least one week, and eight weeks later, improved the repair of osteochondral defects. WNT3a assembled on exosomes, is efficiently delivered into cartilage and contributes to the healing of osteochondral defects.


Subject(s)
Cartilage/metabolism , Exosomes/metabolism , Wnt3A Protein/metabolism , Animals , Cartilage/injuries , Cartilage, Articular/metabolism , Cell Differentiation , Cell Line , Chondrocytes/cytology , Culture Media, Conditioned/pharmacology , Drug Delivery Systems/methods , Exosomes/physiology , Extracellular Matrix/metabolism , Extracellular Vesicles/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Wnt Signaling Pathway , Wnt3A Protein/genetics
5.
Sci Rep ; 11(1): 5682, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33707504

ABSTRACT

WNT ligands can activate several signalling cascades of pivotal importance during development and regenerative processes. Their de-regulation has been associated with the onset of different diseases. Here we investigated the role of the WNT/Calcium Calmodulin Kinase II (CaMKII) pathway in osteoarthritis. We identified Heme Oxygenase I (HMOX1) and Sox-9 as specific markers of the WNT/CaMKII signalling in articular chondrocytes through a microarray analysis. We showed that the expression of the activated form of CaMKII, phospho-CaMKII, was increased in human and murine osteoarthritis and the expression of HMOX1 was accordingly reduced, demonstrating the activation of the pathway during disease progression. To elucidate its function, we administered the CaMKII inhibitor KN93 to mice in which osteoarthritis was induced by resection of the anterior horn of the medial meniscus and of the medial collateral ligament in the knee joint. Pharmacological blockade of CaMKII exacerbated cartilage damage and bone remodelling. Finally, we showed that CaMKII inhibition in articular chondrocytes upregulated the expression of matrix remodelling enzymes alone and in combination with Interleukin 1. These results suggest an important homeostatic role of the WNT/CaMKII signalling in osteoarthritis which could be exploited in the future for therapeutic purposes.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cartilage, Articular/enzymology , Cartilage, Articular/pathology , Homeostasis , Osteoarthritis/enzymology , Osteoarthritis/pathology , Aged , Animals , Bone Remodeling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cattle , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Models, Animal , Female , Gene Expression Regulation, Enzymologic , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Interleukin-1beta/metabolism , Male , Mice, Inbred C57BL , Middle Aged , Models, Biological , Osteoarthritis/genetics , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcriptome/genetics , Up-Regulation , Wnt3 Protein/metabolism
6.
Dev Dyn ; 250(3): 360-376, 2021 03.
Article in English | MEDLINE | ID: mdl-32738003

ABSTRACT

More than 250 years ago, William Hunter stated that when cartilage is destroyed it never recovers. In the last 20 years, the understanding of the mechanisms that lead to joint formation and the knowledge that some of these mechanisms are reactivated in the homeostatic responses of cartilage to injury has offered an unprecedented therapeutic opportunity to achieve cartilage regeneration. Very large investments in ambitious clinical trials are finally revealing that, although we do not have perfect medicines yet, disease modification is a feasible possibility for human osteoarthritis.


Subject(s)
Cartilage, Articular/physiology , Chondrocytes/metabolism , Chondrogenesis , Osteoarthritis , Regeneration , Animals , Humans , Osteoarthritis/metabolism , Osteoarthritis/physiopathology
7.
Sci Transl Med ; 12(561)2020 09 16.
Article in English | MEDLINE | ID: mdl-32938794

ABSTRACT

Osteoarthritis is characterized by the loss of the articular cartilage, bone remodeling, pain, and disability. No pharmacological intervention can currently halt progression of osteoarthritis. Here, we show that blocking receptor tyrosine kinase-like orphan receptor 2 (ROR2) improves cartilage integrity and pain in osteoarthritis models by inhibiting yes-associated protein (YAP) signaling. ROR2 was up-regulated in the cartilage in response to inflammatory cytokines and mechanical stress. The main ligand for ROR2, WNT5A, and the targets YAP and connective tissue growth factor were up-regulated in osteoarthritis in humans. In vitro, ROR2 overexpression inhibited chondrocytic differentiation. Conversely, ROR2 blockade triggered chondrogenic differentiation of C3H10T1/2 cells and suppressed the expression of the cartilage-degrading enzymes a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4 and ADAMTS-5. The chondrogenic effect of ROR2 blockade in the cartilage was independent of WNT signaling and was mediated by down-regulation of YAP signaling. ROR2 signaling induced G protein and Rho-dependent nuclear accumulation of YAP, and YAP inhibition was required but not sufficient for ROR2 blockade-induced chondrogenesis. ROR2 silencing protected mice from instability-induced osteoarthritis with improved structural outcomes, sustained pain relief, and without apparent side effects or organ toxicity. Last, ROR2 silencing in human articular chondrocytes transplanted in nude mice led to the formation of cartilage organoids with more and better differentiated extracellular matrix, suggesting that the anabolic effect of ROR2 blockade is conserved in humans. Thus, ROR2 blockade is efficacious and well tolerated in preclinical animal models of osteoarthritis.


Subject(s)
Chondrogenesis , Osteoarthritis , Animals , Cell Differentiation , Chondrocytes , Mice , Mice, Nude , Osteoarthritis/drug therapy , Receptor Tyrosine Kinase-like Orphan Receptors
8.
Sci Transl Med ; 12(559)2020 09 02.
Article in English | MEDLINE | ID: mdl-32878982

ABSTRACT

Cartilage loss leads to osteoarthritis, the most common cause of disability for which there is no cure. Cartilage regeneration, therefore, is a priority in medicine. We report that agrin is a potent chondrogenic factor and that a single intraarticular administration of agrin induced long-lasting regeneration of critical-size osteochondral defects in mice, with restoration of tissue architecture and bone-cartilage interface. Agrin attracted joint resident progenitor cells to the site of injury and, through simultaneous activation of CREB and suppression of canonical WNT signaling downstream of ß-catenin, induced expression of the chondrogenic stem cell marker GDF5 and differentiation into stable articular chondrocytes, forming stable articular cartilage. In sheep, an agrin-containing collagen gel resulted in long-lasting regeneration of bone and cartilage, which promoted increased ambulatory activity. Our findings support the therapeutic use of agrin for joint surface regeneration.


Subject(s)
Agrin , Cartilage, Articular , Animals , Cell Differentiation , Chondrocytes , Chondrogenesis , Mice , Sheep , Tissue Scaffolds
9.
J Exp Orthop ; 5(1): 21, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29904905

ABSTRACT

BACKGROUND: To explore potential biomarkers in a meniscectomy-induced knee osteoarthritis model, at forty years after meniscectomy. METHODS: We carried out a forty-year study of 53 patients who, as adolescents, underwent open total meniscectomy and assessed two potential synovial and serum biomarkers, namely glycosaminoglycan (GAG) and matrix metalloproteinase-3 (MMP-3). Of the 30 patients available for review, 8 had contralateral knee operations and were excluded. Of the remaining 22 patients, 17 had successful operated knee synovial fluid aspirations and 8 also had successful contralateral control knee aspirations. GAG and MMP3 levels in the synovial fluid and peripheral serum was measured using Alcian blue precipitation and ELISA quantification, respectively. Patients also had their knee radiographs assessed and their radiographic osteoarthritis classified as per the Kellgren-Lawrence and Ahlbӓck systems. RESULTS: At forty years after meniscectomy, synovial MMP-3 levels remain increased (p = 0.0132) while GAG levels were reduced (p = 0.0487) when compared to controls and these two levels correlate inversely. Furthermore, levels of synovial MMP-3 significantly correlated (p = 0.0032, r = 0.7734; p = 0.0256, r = 0.5552) and GAG levels significantly inversely correlated (p = 0.0308, r = - 0.6220; p = 0.0135, r = - 0.6024), respectively, with both radiological scoring systems. Interestingly, we found that the levels of serum MMP-3 correlated only with the synovial fluid levels of MMP-3 in the operated knee and not with the non-operated joint (p = 0.0252, r = 0.7706 vs. p = 0.0764, r = 0.6576). Multiple regression analysis for patient's quality of life based on these biomarkers revealed an almost perfect result with an R2 of 0.9998 and a p value = 0.0087. CONCLUSION: Our results suggest that serum levels of MMP3 could be used as a potential biomarker for knee osteoarthritis, using a simple blood test. Larger cohorts are desirable in order to prove or disprove this finding.

10.
Ann Rheum Dis ; 76(1): 218-226, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27147711

ABSTRACT

OBJECTIVE: Both excessive and insufficient activation of WNT signalling results in cartilage breakdown and osteoarthritis. WNT16 is upregulated in the articular cartilage following injury and in osteoarthritis. Here, we investigate the function of WNT16 in osteoarthritis and the downstream molecular mechanisms. METHODS: Osteoarthritis was induced by destabilisation of the medial meniscus in wild-type and WNT16-deficient mice. Molecular mechanisms and downstream effects were studied in vitro and in vivo in primary cartilage progenitor cells and primary chondrocytes. The pathway downstream of WNT16 was studied in primary chondrocytes and using the axis duplication assay in Xenopus. RESULTS: WNT16-deficient mice developed more severe osteoarthritis with reduced expression of lubricin and increased chondrocyte apoptosis. WNT16 supported the phenotype of cartilage superficial-zone progenitor cells and lubricin expression. Increased osteoarthritis in WNT16-deficient mice was associated with excessive activation of canonical WNT signalling. In vitro, high doses of WNT16 weakly activated canonical WNT signalling, but, in co-stimulation experiments, WNT16 reduced the capacity of WNT3a to activate the canonical WNT pathway. In vivo, WNT16 rescued the WNT8-induced primary axis duplication in Xenopus embryos. CONCLUSIONS: In osteoarthritis, WNT16 maintains a balanced canonical WNT signalling and prevents detrimental excessive activation, thereby supporting the homeostasis of progenitor cells.


Subject(s)
Arthritis, Experimental/metabolism , Cartilage, Articular/metabolism , Osteoarthritis/metabolism , Wnt Proteins/physiology , Wnt Signaling Pathway/physiology , Animals , Apoptosis/physiology , Arthritis, Experimental/etiology , Arthritis, Experimental/pathology , Cartilage, Articular/pathology , Male , Mice, Knockout , Osteoarthritis/etiology , Osteoarthritis/pathology , Proteoglycans/biosynthesis , Proteoglycans/genetics , RNA, Messenger/genetics , Up-Regulation/physiology , Wnt Proteins/biosynthesis , Wnt Proteins/deficiency , Wnt Proteins/genetics
11.
Ann Rheum Dis ; 75(6): 1228-35, 2016 06.
Article in English | MEDLINE | ID: mdl-26290588

ABSTRACT

OBJECTIVES: Osteoarthritis (OA) is a leading cause of disability for which there is no cure. The identification of molecules supporting cartilage homeostasis and regeneration is therefore a major pursuit in musculoskeletal medicine. Agrin is a heparan sulfate proteoglycan which, through binding to low-density lipoprotein receptor-related protein 4 (LRP4), is required for neuromuscular synapse formation. In other tissues, it connects the cytoskeleton to the basement membrane through binding to α-dystroglycan. Prompted by an unexpected expression pattern, we investigated the role and receptor usage of agrin in cartilage. METHODS: Agrin expression pattern was investigated in human osteoarthritic cartilage and following destabilisation of the medial meniscus in mice. Extracellular matrix (ECM) formation and chondrocyte differentiation was studied in gain and loss of function experiments in vitro in three-dimensional cultures and gain of function in vivo, using an ectopic cartilage formation assay in nude mice. Receptor usage was investigated by disrupting LRP4 and α-dystroglycan by siRNA and blocking antibodies respectively. RESULTS: Agrin was detected in normal cartilage but was progressively lost in OA. In vitro, agrin knockdown resulted in reduced glycosaminoglycan content, downregulation of the cartilage transcription factor SOX9 and other cartilage-specific ECM molecules. Conversely, exogenous agrin supported cartilage differentiation in vitro and ectopic cartilage formation in vivo. In the context of cartilage differentiation, agrin used an unusual receptor repertoire requiring both LRP4 and α-dystroglycan. CONCLUSIONS: We have discovered that agrin strongly promotes chondrocyte differentiation and cartilage formation in vivo. Our results identify agrin as a novel potent anabolic growth factor with strong therapeutic potential in cartilage regeneration.


Subject(s)
Agrin/physiology , Arthritis, Experimental/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Dystroglycans/physiology , Osteoarthritis/metabolism , Receptors, LDL/physiology , Agrin/biosynthesis , Agrin/genetics , Agrin/pharmacology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Cartilage, Articular/pathology , Cells, Cultured , Chondrogenesis/drug effects , Down-Regulation/physiology , Gene Knockdown Techniques , Homeostasis/physiology , Humans , LDL-Receptor Related Proteins/physiology , Male , Mice, Inbred DBA , Mice, Knockout , Osteoarthritis/genetics , Osteoarthritis/pathology , Osteogenesis/physiology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , SOX9 Transcription Factor/biosynthesis , SOX9 Transcription Factor/genetics , Up-Regulation/physiology
12.
Drug Discov Today ; 19(8): 1172-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24880104

ABSTRACT

Cartilage breakdown is the disabling outcome of rheumatic diseases, whether prevalently inflammatory such as rheumatoid arthritis or prevalently mechanical such as osteoarthritis (OA). Despite the differences between immune-mediated arthritides and OA, common mechanisms drive cartilage breakdown. Inflammation, chondrocyte phenotype and homeostatic mechanisms have recently been the focus of research and will be summarised in this review.


Subject(s)
Cartilage/pathology , Cartilage/physiopathology , Wound Healing/physiology , Animals , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/physiopathology , Chondrocytes/pathology , Chondrocytes/physiology , Humans , Inflammation/pathology , Inflammation/physiopathology , Osteoarthritis/pathology , Osteoarthritis/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...