Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Article in English | MEDLINE | ID: mdl-38494072

ABSTRACT

OBJECTIVES: Optimizing rehabilitation strategies for osteoarthritis necessitates a comprehensive understanding of chondrocytes' mechanoresponse in both health and disease, especially in the context of the interplay between loading and key pathways involved in osteoarthritis (OA) development, like canonical Wnt signaling. This study aims to elucidate the role of Wnt signaling in the mechanoresponsiveness of healthy and osteoarthritic human cartilage. METHODS: We used an ex-vivo model involving short-term physiological mechanical loading of human cartilage explants. First, the loading protocol for subsequent experiments was determined. Next, loading was applied to non-OA-explants with or without Wnt activation with CHIR99021. Molecular read-outs of anabolic, pericellular matrix and matrix remodeling markers were used to assess the effect of Wnt on cartilage mechanoresponse. Finally, the same set-up was used to study the effect of loading in cartilage from patients with established OA. RESULTS: Our results confirm that physiological loading maintains expression of anabolic genes in non-OA cartilage, and indicate a deleterious effect of Wnt activation in the chondrocyte mechanoresponsiveness. This suggests that loading-induced regulation of chondrocyte markers occurs downstream of canonical Wnt signaling. Interestingly, our study highlighted contrasting mechanoresponsiveness in the model of Wnt activation and the established OA samples, with established OA cartilage maintaining its mechanoresponsiveness, and mechanical loading rescuing the chondrogenic phenotype. CONCLUSION: This study provides insights into the mechanoresponsiveness of human cartilage in both non-OA and OA conditions. These findings hold the potential to contribute to the development of strategies that optimize the effect of dynamic compression by correcting OA pathological cell signaling.

2.
Osteoarthritis Cartilage ; 31(6): 741-752, 2023 06.
Article in English | MEDLINE | ID: mdl-36669584

ABSTRACT

OBJECTIVES: Current experimental approaches cannot elucidate the effect of maladaptive changes on the main cartilage constituents during the degeneration process in osteoarthritis (OA). In silico approaches, however, allow creating 'virtual knock-out' cases to elucidate these effects in a constituent-specific manner. We used such an approach to study the main mechanisms of cartilage degeneration in different mechanical loadings associated with the following OA etiologies: (1) physiological loading of degenerated cartilage, (2) injurious loading of healthy intact cartilage and (3) physiological loading of cartilage with a focal defect. METHODS: We used the recently developed Cartilage Adaptive REorientation Degeneration (CARED) framework to simulate cartilage degeneration associated with primary and secondary OA (OA cases (1)-(3)). CARED incorporates numerical description of tissue-level cartilage degeneration mechanisms in OA, namely, collagen degradation, collagen reorientation, fixed charged density loss and tissue hydration increase following mechanical loading. We created 'virtual knock-out' scenarios by deactivating these degenerative processes one at a time in each of the three OA cases. RESULTS: In the injurious loading of intact and physiological loading of degenerated cartilage, collagen degradation drives degenerative changes through fixed charge density loss and tissue hydration rise. In contrast, the two later mechanisms were more prominent in the focal defect cartilage model. CONCLUSION: The virtual knock-out models reveal that injurious loading to intact cartilage and physiological loading to degenerated cartilage induce initial degenerative changes in the collagen network, whereas, in the presence of a focal cartilage defect, mechanical loading initially causes proteoglycans (PG) depletion, before changes in the collagen fibril network occur.


Subject(s)
Cartilage, Articular , Osteoarthritis , Humans , Proteoglycans/metabolism , Cartilage, Articular/metabolism , Osteoarthritis/etiology , Osteoarthritis/metabolism , Collagen/metabolism , Extracellular Matrix/metabolism
3.
Ann Rheum Dis ; 82(5): 594-598, 2023 05.
Article in English | MEDLINE | ID: mdl-36702529

ABSTRACT

To detail the unmet clinical and scientific needs in the field of rheumatology. After a 2-year hiatus due to the SARS-CoV-2 pandemic, the 22nd annual international Advances in Targeted Therapies meeting brought together more than 100 leading basic scientists and clinical researchers in rheumatology, immunology, epidemiology, molecular biology and other specialties. Breakout sessions were convened with experts in five rheumatological disease-specific groups including: rheumatoid arthritis (RA), psoriatic arthritis, axial spondyloarthritis, systemic lupus erythematosus and connective tissue diseases (CTDs). In each group, experts were asked to identify and prioritise current unmet needs in clinical and translational research, as well as highlight recent progress in meeting formerly identified unmet needs. Clinical trial design innovation was emphasised across all disease states. Within RA, developing therapies and trials for refractory disease patients remained among the most important identified unmet needs and within lupus and spondyloarthritis the need to account for disease endotypes was highlighted. The RA group also identified the need to better understand the natural history of RA, pre-RA states and the need ultimately for precision medicine. In CTD generally, experts focused on the need to better identify molecular, cellular and clinical signals of early and undifferentiated disease in order to identify novel drug targets. There remains a strong need to develop therapies and therapeutic strategies for those with treatment-refractory disease. Increasingly it is clear that we need to better understand the natural history of these diseases, including their 'predisease' states, and identify molecular signatures, including at a tissue level, which can facilitate disease diagnosis and treatment. As these unmet needs in the field of rheumatic diseases have been identified based on consensus of expert clinicians and scientists in the field, this document may serve individual researchers, institutions and industry to help prioritise their scientific activities.


Subject(s)
Arthritis, Psoriatic , Arthritis, Rheumatoid , COVID-19 , Rheumatic Diseases , Rheumatology , Humans , SARS-CoV-2 , Rheumatic Diseases/drug therapy , Rheumatic Diseases/diagnosis , Arthritis, Rheumatoid/drug therapy , Arthritis, Psoriatic/drug therapy
4.
Osteoarthritis Cartilage ; 31(4): 507-518, 2023 04.
Article in English | MEDLINE | ID: mdl-36370958

ABSTRACT

OBJECTIVES: ANP32A is a key protector of cartilage health, via preventing oxidative stress and Wnt hyper-activation. We aimed to unravel how ANP32A is regulated in cartilage. METHODS: A bioinformatics pipeline was applied to identify regulators of ANP32A. Pathways of interest were targeted to study their impact on ANP32A in in vitro cultures of the human chondrocyte C28/I2 cell-line and primary human articular chondrocytes (hACs) from up to five different donors, using Wnt-activator CHIR99021, hypoxia-mimetic IOX2 and a hypoxia chamber. ANP32A was evaluated using real-time quantitative polymerase chain reaction (RT-qPCR) and Western blot. In vivo, the effect of hypoxia was examined by immunohistochemistry in mice injected intra-articularly with IOX2 after destabilization of the medial meniscus. Effects of Wnt hyper-activation were investigated using Frzb-knockout mice and wild-type mice treated intra-articularly with CHIR99021. Wnt inhibition effects were assessed upon intra-articular injection of XAV939. RESULTS: The hypoxia and Wnt signaling pathways were identified as networks controlling ANP32A expression. In vitro and in vivo experiments demonstrated increases in ANP32A upon hypoxic conditions (1.3-fold in hypoxia in C28/I2 cells with 95% confidence interval (CI) [1.11-1.54] and 1.90-fold in hACs [95% CI: 1.56-2] and 1.67-fold in ANP32A protein levels after DMM surgery with IOX2 injections [95% CI: 1.33-2.08]). Wnt hyper-activation decreased ANP32A in chondrocytes in vitro (1.23-fold decrease [95% CI: 1.02-1.49]) and in mice (1.45-fold decrease after CHIR99021 injection [95% CI: 1.22-1.72] and 1.41-fold decrease in Frzb-knockout mice [95% CI: 1.00-1.96]). Hypoxia and Wnt modulated ataxia-telangiectasia mutated serine/threonine kinase (ATM), an ANP32A target gene, in hACs (1.89-fold increase [95% CI: 1.38-2.60] and 1.41-fold decrease [95% CI: 1.02-1.96]). CONCLUSIONS: Maintaining hypoxia and limiting Wnt activation sustain ANP32A and protect against osteoarthritis.


Subject(s)
Cartilage, Articular , Mice , Humans , Animals , Cartilage, Articular/metabolism , Wnt Signaling Pathway/genetics , Chondrocytes/metabolism , Mice, Knockout , Hypoxia , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/pharmacology
5.
Osteoarthritis Cartilage ; 30(5): 724-734, 2022 05.
Article in English | MEDLINE | ID: mdl-35227892

ABSTRACT

OBJECTIVES: To investigate how ANP32A, previously linked to the antioxidant response, regulates Wnt signaling as unraveled by transcriptome analysis of Anp32a-deficient mouse articular cartilage, and its implications for osteoarthritis (OA) and diseases beyond the joint. METHODS: Anp32a knockdown chondrogenic ATDC5 cells were cultured in micromasses. Wnt target genes, differentiation markers and matrix deposition were quantified. Wnt target genes were determined in articular cartilage from Anp32a-deficient mice and primary human articular chondrocytes upon ANP32A silencing, using qPCR, luciferase assays and immunohistochemistry. Co-immunoprecipitation, immunofluorescence and chromatin-immunoprecipitation quantitative PCR probed the molecular mechanism via which ANP32A regulates Wnt signaling. Anp32a-deficient mice were subjected to the destabilization of the medial meniscus (DMM) OA model and treated with a Wnt inhibitor and an antioxidant. Severity of OA was assessed by cartilage damage and osteophyte formation. Human Protein Atlas data analysis identified additional organs where ANP32A may regulate Wnt signaling. Wnt target genes were determined in heart and hippocampus from Anp32a-deficient mice, and cardiac hypertrophy and fibrosis quantified. RESULTS: Anp32a loss triggered Wnt signaling hyper-activation in articular cartilage. Mechanistically, ANP32A inhibited target gene expression via histone acetylation masking. Wnt antagonist treatment reduced OA severity in Anp32a-deficient mice by preventing osteophyte formation but not cartilage degradation, contrasting with antioxidant treatment. Dual therapy ameliorated more OA features than individual treatments. Anp32a-deficient mice also showed Wnt hyper-activation in the heart, potentially explaining the cardiac hypertrophy phenotype found. CONCLUSIONS: ANP32A is a novel translationally relevant repressor of Wnt signaling impacting osteoarthritis and cardiac disease.


Subject(s)
Cartilage, Articular , Heart Diseases , Osteoarthritis , Osteophyte , Animals , Antioxidants/metabolism , Cardiomegaly/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Heart Diseases/metabolism , Mice , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteophyte/metabolism , Wnt Signaling Pathway/physiology
6.
Semin Immunol ; 58: 101521, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34763975

ABSTRACT

The spondyloarthritides are a cluster of inflammatory rheumatic diseases characterized by different diagnostic entities with heterogeneous phenotypes. The current classification system groups spondyloarthritis patients in two main categories, axial and peripheral spondyloarthritis, providing a framework wherein the clinical picture guides the treatment. However, the heterogeneity of the clinical manifestations of the pathologies, even when residing in the same group, highlights the importance of analyzing the smallest features of each entity to understand how different cellular subsets evolve, what the underlying mechanisms are and what biological markers can be identified and validated to evaluate the stage of disease and the corresponding efficacy of treatments. In this review, we will focus mostly on axial spondyloarthritis, report current knowledge concerning the cellular populations involved in its pathophysiology, and their molecular diversity. We will discuss the implications of such a diversity, and their meaning in terms of patients' stratification.


Subject(s)
Spondylarthritis , Spondylitis, Ankylosing , Humans , Spondylitis, Ankylosing/drug therapy , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/pathology , Spondylarthritis/genetics , Spondylarthritis/diagnosis
8.
Osteoarthritis Cartilage ; 28(5): 675-684, 2020 05.
Article in English | MEDLINE | ID: mdl-31634584

ABSTRACT

OBJECTIVE: Inflammation and innate immune responses may contribute to development and progression of Osteoarthritis (OA). Chondrocytes are the sole cell type of the articular cartilage and produce extracellular-matrix molecules. How inflammatory mediators reach chondrocytes is incompletely understood. Previous studies have shown that chondrocytes express mRNA encoding complement proteins such as C1q, suggesting local protein production, which has not been demonstrated conclusively. The aim of this study is to explore C1q production at the protein level by chondrocytes. DESIGN: We analysed protein expression of C1q in freshly isolated and cultured human articular chondrocytes using Western blot, ELISA and flow cytometry. We examined changes in mRNA expression of collagen, MMP-1 and various complement genes upon stimulation with pro-inflammatory cytokines or C1q. mRNA expression of C1 genes was determined in articular mouse chondrocytes. RESULTS: Primary human articular chondrocytes express genes encoding C1q, C1QA, C1QB, C1QC, and secrete C1q to the extracellular medium. Stimulation of chondrocytes with pro-inflammatory cytokines upregulated C1QA, C1QB, C1QC mRNA expression, although this was not confirmed at the protein level. Extracellular C1q bound to the chondrocyte surface dose dependently. In a pilot study, binding of C1q to chondrocytes resulted in changes in the expression of collagens with a decrease in collagen type 2 and an increase in type 10. Mouse articular chondrocytes also expressed C1QA, C1QB, C1QC, C1R and C1S at the mRNA level. CONCLUSIONS: C1q protein can be expressed and secreted by human articular chondrocytes and is able to bind to chondrocytes influencing the relative collagen expression.


Subject(s)
Chondrocytes/metabolism , Complement C1q/genetics , Complement C1r/genetics , Complement C1s/genetics , Osteoarthritis, Knee/genetics , RNA, Messenger/metabolism , Animals , Cartilage, Articular/cytology , Collagen Type II/genetics , Collagen Type X/genetics , Gene Expression Regulation , Humans , Mice , Osteoarthritis, Knee/metabolism , Pilot Projects
9.
Osteoarthritis Cartilage ; 27(11): 1702-1710, 2019 11.
Article in English | MEDLINE | ID: mdl-31330188

ABSTRACT

OBJECTIVE: Exostosin-1 (Ext1) encodes a glycosyltransferase required for heparan sulfate (HS) chain elongation in HS-proteoglycan biosynthesis. HS chains serve as binding partners for signaling proteins, affecting their distribution and activity. The Wnt/ß-catenin pathway emerged as critical regulator of chondrogenesis. Yet, how EXT1 and HS affect Wnt/ß-catenin signaling during chondrogenesis remains unexplored. METHOD: Ext1 was stably knocked-down or overexpressed in ATDC5 chondrogenic cells cultured as micromasses. HS content was determined using ELISA. Chondrogenic markers Sox9, Col2a1, Aggrecan, and Wnt direct target gene Axin2 were measured by RT-qPCR. Proteoglycan content was evaluated by Alcian blue and DMMB assay, canonical Wnt signaling activation by ß-catenin Western blot and TOP/FOP assay. ATDC5 cells and human articular chondrocytes were treated with Wnt activators CHIR99021 and recombinant WNT3A. RESULTS: Ext1 knock-down reduced HS, and increased chondrogenic markers and proteoglycan accumulation. Ext1 knock-down reduced active Wnt/ß-catenin signaling. Conversely, Ext1 overexpressing cells, with higher HS content, showed decreased chondrogenic differentiation and enhanced Wnt/ß-catenin signaling. Wnt/ß-catenin signaling activation led to a down-regulation of Ext1 expression in ATDC5 cells and in human articular chondrocytes. CONCLUSIONS: EXT1 affects chondrogenic differentiation of precursor cells, in part via changes in the activity of Wnt/ß-catenin signaling. Wnt/ß-catenin signaling controls Ext1 expression, suggesting a regulatory loop between EXT1 and Wnt/ß-catenin signaling during chondrogenesis.


Subject(s)
Chondrocytes/metabolism , Chondrogenesis/genetics , Gene Expression Regulation , N-Acetylglucosaminyltransferases/genetics , RNA/genetics , Wnt Signaling Pathway/genetics , Blotting, Western , Cell Differentiation , Cells, Cultured , Chondrocytes/pathology , Enzyme-Linked Immunosorbent Assay , Humans , N-Acetylglucosaminyltransferases/biosynthesis , Wnt Proteins/biosynthesis , Wnt Proteins/genetics
10.
Osteoarthritis Cartilage ; 27(3): 513-525, 2019 03.
Article in English | MEDLINE | ID: mdl-30513362

ABSTRACT

OBJECTIVE: We earlier identified that the histone methyltransferase Disruptor of telomeric silencing 1-like (DOT1L) is as a master protector of cartilage health via limiting excessive activation of the Wnt pathway. However, cartilage-specific homozygous Dot1l knockout mice exhibited a severe growth phenotype and perinatal death, which hampered their use in induced or ageing models of osteoarthritis (OA). The aim of this study was to generate and examine haploinsufficient and inducible conditional Dot1l-deficient mouse models to evaluate the importance of DOT1L during post-traumatic or ageing-associated OA onset and progression. METHOD: We used cartilage-specific heterozygous and postnatal tamoxifen-inducible Dot1l knockout mice and performed destabilization of the medial meniscus (DMM) and ageing as OA models. Mice were examined histologically using X-rays and micro-computed tomography (µCT), and cartilage damage and osteophyte formation were assessed based on OARSI guidelines. Immunohistochemistry of DOT1L, H3K79me2, TCF1 and COLX was performed. RESULTS: Both Dot1l-deficient strains exhibit a phenotype characterized by joint remodeling with extensive osteophyte formation and ectopic ossification upon ageing, indicating accelerated development of spontaneous osteoarthritis. In the DMM-induced OA mouse model, absence of Dot1l resulted in increased cartilage damage. Wnt signalling hyper-activation and ectopic chondrocyte hypertrophy were observed in the articular cartilage of both Dot1l-deficient mice. CONCLUSIONS: This study demonstrated the functional relevance of DOT1L in vivo during the development of OA using genetically modified mice. Thus, maintaining or enhancing DOT1L activity during ageing or after trauma might prevent OA onset and progression.


Subject(s)
Histone-Lysine N-Methyltransferase/deficiency , Joints/injuries , Osteoarthritis/etiology , Animals , Joints/diagnostic imaging , Joints/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoarthritis/diagnostic imaging , Radiography , X-Ray Microtomography
11.
Ann Rheum Dis ; 75(3): 499-510, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26644232

ABSTRACT

BACKGROUND: Since the publication of the European League Against Rheumatism recommendations for the pharmacological treatment of psoriatic arthritis (PsA) in 2012, new evidence and new therapeutic agents have emerged. The objective was to update these recommendations. METHODS: A systematic literature review was performed regarding pharmacological treatment in PsA. Subsequently, recommendations were formulated based on the evidence and the expert opinion of the 34 Task Force members. Levels of evidence and strengths of recommendations were allocated. RESULTS: The updated recommendations comprise 5 overarching principles and 10 recommendations, covering pharmacological therapies for PsA from non-steroidal anti-inflammatory drugs (NSAIDs), to conventional synthetic (csDMARD) and biological (bDMARD) disease-modifying antirheumatic drugs, whatever their mode of action, taking articular and extra-articular manifestations of PsA into account, but focusing on musculoskeletal involvement. The overarching principles address the need for shared decision-making and treatment objectives. The recommendations address csDMARDs as an initial therapy after failure of NSAIDs and local therapy for active disease, followed, if necessary, by a bDMARD or a targeted synthetic DMARD (tsDMARD). The first bDMARD would usually be a tumour necrosis factor (TNF) inhibitor. bDMARDs targeting interleukin (IL)12/23 (ustekinumab) or IL-17 pathways (secukinumab) may be used in patients for whom TNF inhibitors are inappropriate and a tsDMARD such as a phosphodiesterase 4-inhibitor (apremilast) if bDMARDs are inappropriate. If the first bDMARD strategy fails, any other bDMARD or tsDMARD may be used. CONCLUSIONS: These recommendations provide stakeholders with an updated consensus on the pharmacological treatment of PsA and strategies to reach optimal outcomes in PsA, based on a combination of evidence and expert opinion.


Subject(s)
Algorithms , Antirheumatic Agents/therapeutic use , Arthritis, Psoriatic/drug therapy , Glucocorticoids/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Disease Management , Europe , Humans , Rheumatology , Societies, Medical
12.
Osteoarthritis Cartilage ; 23(2): 275-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25450854

ABSTRACT

OBJECTIVE: To investigate the specific role of Frizzled-related protein (FRZB) and Secreted frizzled-related protein 1 (SFRP1) in the onset and progression of Osteoarthritis (OA) using Frzb(-/-) and Sfrp1(-/-) mice in the destabilization of medial meniscus model (DMM), a slowly progressing model of OA. Secreted frizzled-related proteins (SFRPs) were identified as secreted Wingless-type (Wnt) antagonists. The Wnt signaling cascade is a major regulator in cartilage development, homeostasis and degeneration. METHODS: The DMM model was surgically induced in eight-week-old male C57/Bl6 Frzb(-/-), Sfrp1(-/-) or wild-type mice by transection of the medial meniscotibial ligament. Cartilage damage in the femoral and tibial articular surfaces was calculated following the Osteoarthritis Research Society International (OARSI) histopathology initiative guidelines. Histomorphometry was used to evaluate the subchondral bone plate thickness. RESULTS: OA severity scores were significantly higher in the tibia of Frzb(-/-) mice as compared to littermates, whereas no interaction was seen between genotype and intervention in Sfrp1(-/-) mice. Moreover, the DMM model resulted in significantly greater subchondral bone changes compared to sham but was not different between Frzb(-/-) mice and littermates. In contrast, the subchondral bone properties in Sfrp1(-/-) mice were significantly different from littermates. CONCLUSION: Using the DMM model, we demonstrated that FRZB and SFRP1 differentially modulate joint homeostasis in two distinct compartments of the joint. These data highlight the fine-tuning of Wnt signaling in joint homeostasis and disease, show differential regulation of the cascade in cartilage and subchondral bone, and provide further evidence for a role of endogenous Wnt modulators as key players in OA.


Subject(s)
Glycoproteins/physiology , Homeostasis , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Osteoarthritis/etiology , Animals , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL
14.
Ann Rheum Dis ; 73(2): 336-48, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24285494

ABSTRACT

Osteoarthritis (OA) is the most common form of arthritic disease, and a major cause of disability and impaired quality of life in the elderly. OA is a complex disease of the entire joint, affecting bone, cartilage and synovium that thereby presents multiple targets for treatment. This manuscript will summarise emerging observations from cell biology, preclinical and preliminary clinical trials that elucidate interactions between the bone and cartilage components in particular. Bone and cartilage health are tightly associated. Ample evidence has been found for bone changes during progression of OA including, but not limited to, increased turnover in the subchondral bone, undermineralisation of the trabecular structure, osteophyte formation, bone marrow lesions and sclerosis of the subchondral plate. Meanwhile, a range of investigations has shown positive effects on cartilage health when bone resorption is suppressed, or deterioration of the cartilage when resorption is increased. Known bone therapies, namely oestrogens, selective oestrogen receptor modifiers (SERMs), bisphosphonates, strontium ranelate, calcitonin and parathyroid hormone, might prove useful for treating two critical tissue components of the OA joint, the bone and the cartilage. An optimal treatment for OA likely targets at least these two tissue components. The patient subgroups for whom these therapies are most appropriate have yet to be fully defined but would likely include, at a minimum, those with high bone turnover.


Subject(s)
Anabolic Agents/therapeutic use , Bone Density Conservation Agents/therapeutic use , Bone Remodeling/drug effects , Cartilage, Articular/metabolism , Osteoarthritis/drug therapy , Anabolic Agents/pharmacology , Bone Density Conservation Agents/pharmacology , Bone Remodeling/physiology , Cartilage, Articular/drug effects , Humans , Osteoarthritis/pathology , Osteoarthritis/physiopathology , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/pathology
15.
Fetal Diagn Ther ; 34(4): 225-35, 2013.
Article in English | MEDLINE | ID: mdl-24134897

ABSTRACT

BACKGROUND: Amniotic fluid (AF) cell populations will be applied in perinatology. We aimed to test the feasibility of large-scale cell expansion. STUDY METHODS: We determined the best out of three published expansion protocols for mesenchymal progenitors (AF samples, n = 4) in terms of self-renewal ability. Characterization was performed based on morphology, surface marker analysis, cytogenetic stability, and differentiation potential. The conditions for the best self-renewal ability were further determined in a consecutive series (n = 159). RESULTS: The medium containing fetal bovine serum (FBS), epidermal growth factor, insulin, transferrin, and tri-iodothyronine, combined with seeding on gelatin-coated wells, best stimulated the growth of cells with mesenchymal features, as demonstrated by flow cytometry; however, only osteogenic differentiation was possible. Large-scale testing (n = 44) failed to confirm a robust self-renewal ability. Better results were obtained (n = 88) using optimized FBS or an increased initial cell density. Eventually over 81% of cultures continued growing after the initial medium change and had mesenchymal features but failed differentiation assays. DISCUSSION: Routine in vitro expansion of AF-derived mesenchymal cells remains problematic. Despite an increase in successful cell cultures from 40 up to 80% using optimized serum and an increased cell density, eventually cells failed to demonstrate differentiation abilities. Routine isolation and expansion from unselected AF samples remains a challenge.


Subject(s)
Amniotic Fluid/cytology , Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Animals , Cattle , Cell Count , Cell Differentiation , Cell Proliferation , Culture Media , Female , Humans , Karyotyping , Pregnancy
16.
Arthritis Rheum ; 63(5): 1246-54, 2011 May.
Article in English | MEDLINE | ID: mdl-21337318

ABSTRACT

OBJECTIVE: Rituximab displays therapeutic benefits in the treatment of patients with rheumatoid arthritis (RA) resistant to tumor necrosis factor (TNF) blockade. However, the precise role of B cells in the pathogenesis of RA is still unknown. We undertook this study to investigate the global molecular effects of rituximab in synovial biopsy samples obtained from anti-TNF-resistant RA patients before and after administration of the drug. METHODS: Paired synovial biopsy samples were obtained from the affected knee of anti-TNF-resistant RA patients before (time 0) and 12 weeks after (time 12) initiation of rituximab therapy. Total RNA was extracted, labeled according to standard Affymetrix procedures, and hybridized on GeneChip HGU133 Plus 2.0 slides. Immunohistochemistry and quantitative real-time reverse transcriptase-polymerase chain reaction experiments were performed to confirm the differential expression of selected transcripts. RESULTS: According to Student's paired t-tests, 549 of 54,675 investigated probe sets were differentially expressed between time 0 and time 12. Pathway analysis revealed that genes down-regulated between time 0 and time 12 were significantly enriched in immunoglobulin genes and genes involved in chemotaxis, leukocyte activation, and immune responses (Gene Ontology annotations). In contrast, genes up-regulated between time 0 and time 12 were significantly enriched in transcripts involved in cell development (Gene Ontology annotation) and wound healing (Gene Set Enrichment Analysis). At baseline, higher synovial expression of immunoglobulin genes was associated with response to therapy. CONCLUSION: Rituximab displays unique effects on global gene expression profiles in the synovial tissue of RA patients. These observations open new perspectives in the understanding of the biologic effects of the drug and in the selection of patients likely to benefit from this therapy.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/genetics , Gene Expression/drug effects , Synovial Membrane/drug effects , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Female , Gene Expression/immunology , Humans , Immunohistochemistry , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Rituximab , Synovial Membrane/immunology
17.
Clin Exp Rheumatol ; 27(4 Suppl 55): S10-4, 2009.
Article in English | MEDLINE | ID: mdl-19822039

ABSTRACT

Rheumatoid arthritis and ankylosing spondylitis are common and severe chronic inflammatory skeletal diseases. Recognizing the differences rather than emphasizing similarities is important for a better understanding of the disease processes, the identification of specific therapeutic targets and in the long-term better treatment options for the individual patients. We discuss a number of pathophysiological differences between rheumatoid arthritis and ankylosing spondylitis by looking at the anatomical characteristics, differences and similarities in the autoimmune and autoinflammatory reactions, association with other immune mediated inflammatory diseases, structural outcome, and their potential significance for further therapeutic developments. Further research into the differences between these diseases should focus on the specific nature of the immune/inflammatory components, the role of resident cells in the joint and joint-associated tissues, the types and mechanisms of tissue remodeling and the characteristics of the articular cartilage. Better insights into their individual characteristics may lead to better therapeutic strategies, specific targets and useful biomarkers.


Subject(s)
Arthritis, Rheumatoid/physiopathology , Spondylitis, Ankylosing/physiopathology , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/immunology , Humans , Joints/pathology , Joints/physiopathology , Spondylitis, Ankylosing/diagnosis , Spondylitis, Ankylosing/immunology , Synovitis/immunology , Synovitis/pathology , Synovitis/physiopathology
18.
Clin Exp Rheumatol ; 27(3): 495-8, 2009.
Article in English | MEDLINE | ID: mdl-19604444

ABSTRACT

OBJECTIVES: Growth and differentiation factor 5 (GDF5), member of TGFBeta superfamily, has been implicated in limb development, and is known to play an important role in joint formation. Its absence leads to brachypodism in mice and a number of skeletal malformation syndromes in humans. Recently, an association was shown between osteo-arthritis and a 5' UTR polymorphism in GDF5 gene. In addition, the role of GDF5 may reach beyond the musculoskeletal system. GDF5 appears present in a lipopolysaccharide (LPS) receptor cluster. Absence of GDF5 may limit the response to LPS. This may have consequences for immune responses and macrophage function in general, and for arthritis in particular. Here we compared the sensitivity of Gdf5(Bp-J/Bp-J) mice and wild type (WT) mice to LPS. METHODS: Peritoneal macrophages from Gdf5(Bp-J/Bp-J) mice and WT mice were stimulated for 18h with LPS (0, 10 or 100 ng/ml). The supernatant was collected and TNF release was measured by ELISA and by an indirect luciferase assay using LNF-luc C3 cells. Gdf5(Bp-J/Bp-J) mice and WT mice were injected with LPS i.p. (30 mg/kg) and LPS induced lethality was checked every 3 hours for 36 hours. RESULTS: Gdf5(Bp-J/Bp-J) macrophages showed no difference in TNF expression upon LPS stimulation measured by ELISA and by indirect luciferase assay. Gdf5(Bp-J/Bp-J) mice died upon a lethal dose of LPS, as is seen in WT controls. CONCLUSION: Absence of Gdf5 appears not to affect the LPS response. Mice with a reduced expression of Gdf5 can be used in disease models which are dependent on LPS boost.


Subject(s)
Arthritis/metabolism , Arthritis/physiopathology , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/metabolism , Lipopolysaccharide Receptors/metabolism , Signal Transduction/physiology , Toll-Like Receptors/metabolism , Animals , Arthritis/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Immune System/metabolism , Immune System/physiopathology , Lipopolysaccharide Receptors/drug effects , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Mutant Strains , Mutation/genetics , Toll-Like Receptors/drug effects , Tumor Necrosis Factor-alpha/metabolism
20.
Osteoarthritis Cartilage ; 17(3): 390-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18819822

ABSTRACT

OBJECTIVE: To study the effect of frizzled-related protein (Frzb) deletion in mice on voluntary running wheel exercise performance and osteoarthritis. METHODS: At the age of 7 weeks, Frzb(-/-) and wild-type mice were grouped and a running wheel was introduced into the cage. At week 8, all mice were caged solitarily with a running wheel available. Mice were allowed free exercise for 6-12 months and distances run were recorded daily. Non-running mice were used as additional control group. X-rays of knees and hips were taken at different time points. At the end of the experiment, mice were sacrificed and joints were processed for histological evaluation. Cartilage damage, synovitis and osteophyte formation were scored. Muscle fiber composition of the soleus and extensor digitorum longus was studied by immunofluorescence. RESULTS: At the age of 6 months, both female and male wild-type mice showed a significantly greater exercise performance than the Frzb(-/-) mice (P<0.05). At 1 year, the difference was still significant for male mice, but not for females. Running exercise did not significantly affect severity of osteoarthritis. No statistical differences in osteoarthritis severity were seen between Frzb(-/-) mice and wild-type mice. No differences were seen in muscle composition between Frzb(-/-) mice and wild-type mice. CONCLUSION: Absence of Frzb in mice reduced voluntary exercise performance in running wheels. These experiments demonstrate that the effects of genes in mice can also be evaluated using functional outcomes such as running wheel exercise performance, similar to evolving practice in human clinical trials.


Subject(s)
Glycoproteins/genetics , Osteoarthritis/genetics , Physical Conditioning, Animal/statistics & numerical data , Animals , Cartilage, Articular/pathology , Case-Control Studies , Disease Models, Animal , Female , Gene Deletion , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Knockout , Muscle Fibers, Skeletal/ultrastructure , Osteoarthritis/pathology , Physical Exertion/physiology , Sex Factors , Statistics as Topic , Stifle/pathology , Wnt Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...