Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
IEEE J Transl Eng Health Med ; 10: 1800308, 2022.
Article in English | MEDLINE | ID: mdl-35391755

ABSTRACT

OBJECTIVE: A repeatable and reliable follow-up of knee injuries would be desirable to prevent delayed diagnosis and to monitor the efficacy of the applied treatment over time. Ultrasound (US) techniques are an attractive option to this purpose, since they are safe, low-cost and non-invasive. However, its use in the clinical practice is limited by the high dependency on the operator's experience. Hence, the objective of this study is to provide a standardization of the US image acquisition process for knee osteoarthritis (OA) allowing an extended clinical use of US technologies in this domain. METHODS: Clinical specifications were provided by expert musculoskeletal radiologists thus identifying the subject poses and the US probe positions needed to evaluate the cartilage structure, signs of synovitis and joint effusion. Such considerations were used to derive the technical requirements needed for the development of a wearable brace equipped with specific openings to guide the correct placement of the probe. The feasibility of the developed wearable brace was tested on three healthy volunteers, which were asked to acquire informative US images, similar to the reference images performed by the musculoskeletal radiologist. RESULTS: Thanks to the knee brace, the untrained subjects were able to self-acquire informative B-mode images comparable to the corresponding images acquired by an expert clinician. DISCUSSION/CONCLUSION: The use of a knee brace intended for knee OA US diagnosis demonstrated the possibility to standardize the acquisition protocol and make its application achievable also for untrained subjects, representing a key step toward tele-ultrasonography.


Subject(s)
Osteoarthritis, Knee , Synovitis , Braces , Humans , Knee Joint/diagnostic imaging , Osteoarthritis, Knee/diagnostic imaging , Ultrasonography
2.
Ultrasonics ; 116: 106495, 2021 Jun 15.
Article in English | MEDLINE | ID: mdl-34186322

ABSTRACT

This work aims to describe the development and validation of two low-intensity pulsed ultrasound stimulation systems able to control the dose delivered to the biological target. Transducer characterization was performed in terms of pressure field shape and intensity, for a high-frequency range (500 kHz to 5 MHz) and for a low-frequency value (38 kHz). This allowed defining the distance, on the beam axis, at which biological samples should be placed during stimulation and to exactly know the intensity at the target. Carefully designed retaining systems were developed, for hosting biological samples. Sealing tests proved their impermeability to external contaminants. The assembly/de-assembly time of the systems resulted ~3 min. Time-domain acoustic simulations allowed to precisely estimate the ultrasound beam within the biological sample chamber, thus enabling the possibility to precisely control the pressure to be transmitted to the biological target, by modulating the transducer's input voltage. Biological in vitro tests were also carried out, demonstrating the sterility of the system and the absence of toxic and inflammatory effects on growing cells after multiple immersions in water, over seven days.

3.
Osteoarthritis Cartilage ; 25(7): 1161-1171, 2017 07.
Article in English | MEDLINE | ID: mdl-28153787

ABSTRACT

OBJECTIVE: To define if adipose mesenchymal stromal cell (ASC) treatment mediated switching of the pro-inflammatory profile of M1-like macrophages as a means to develop a tailored in vitro efficacy/potency test. DESIGN: We firstly performed immunohistochemical analysis of CD68, CD80 (M1-like) and CD206 (M2-like) macrophages in osteoarthritic (OA) synovial tissue. ASC were co-cultured in contact and in transwell with activated (GM-CSF + IFNγ)-M1 macrophages. We analyzed IL1ß, TNFα, IL6, MIP1α/CCL3, S100A8, S100A9, IL10, CD163 and CD206 by qRT-PCR or immunoassays. Prostaglandin E2 (PGE2) blocking experiments were performed using PGE2 receptor antagonist. RESULTS: In moderate grade OA synovium we did not always find a higher percentage of CD80 with respect to CD206. M1-like-activated macrophage factors IL1ß, TNFα, IL6, MIP1α/CCL3, S100A8 and S100A9 were down-modulated both in contact and in transwell by ASC. However, in both systems ASC induced the typical M2-like macrophage markers IL10, CD163 and CD206. Activated-M1-like macrophages pre-treated with PGE2 receptor antagonist failed to decrease secretion of TNFα, IL6 and to increase that of IL10, CD163 and CD206 when co-cultured with ASC confirming a PGE2 specific role. CONCLUSIONS: We demonstrated that ASC are responsible for the switching of activated-M1-like inflammatory macrophages to a M2-like phenotype, mainly through PGE2. This evidenced that activated-M1-like macrophages may represent a relevant cell model to test the efficacy/potency of ASC and suggests a specific role of ASC as important determinants in therapeutic dampening of synovial inflammation in OA.


Subject(s)
Adipocytes/drug effects , Dinoprostone/pharmacology , Macrophages/drug effects , Mesenchymal Stem Cells/drug effects , Oxytocics/pharmacology , Adult , Antigens, CD/metabolism , Case-Control Studies , Cell Differentiation/physiology , Cell Movement/drug effects , Cells, Cultured , Female , Humans , Leukocytes, Mononuclear/drug effects , Male , Middle Aged , Osteoarthritis/pathology , Subcutaneous Fat, Abdominal/cytology , Synovial Membrane/cytology , Synovial Membrane/drug effects
4.
J Tissue Eng Regen Med ; 10(5): 374-91, 2016 05.
Article in English | MEDLINE | ID: mdl-23495253

ABSTRACT

Osteochondral lesions require treatment to restore the biology and functionality of the joint. A novel nanostructured biomimetic gradient scaffold was developed to mimic the biochemical and biophysical properties of the different layers of native osteochondral structure. The present results show that the scaffold presents important physicochemical characteristics and can support the growth and differentiation of mesenchymal stromal cells (h-MSCs), which adhere and penetrate into the cartilaginous and bony layers. H-MSCs grown in chondrogenic or osteogenic medium decreased their proliferation during days 14-52 on both scaffold layers and in medium without inducing factors used as controls. Both chondrogenic and osteogenic differentiation of h-MSCs occurred from day 28 and were increased on day 52, but not in the control medium. Safranin O staining and collagen type II and proteoglycans immunostaining confirmed that chondrogenic differentiation was specifically induced only in the cartilaginous layer. Conversely, von Kossa staining, osteocalcin and osteopontin immunostaining confirmed that osteogenic differentiation occurred on both layers. This study shows the specific potential of each layer of the biomimetic scaffold to induce chondrogenic or osteogenic differentiation of h-MSCs. These processes depended mainly on the media used but not the biomaterial itself, suggesting that the local milieu is fundamental for guiding cell differentiation. Copyright © 2013 John Wiley & Sons, Ltd.


Subject(s)
Biomimetic Materials/chemistry , Bone Regeneration , Cell Differentiation , Chondrogenesis , Mesenchymal Stem Cells/metabolism , Nanocomposites/chemistry , Antigens, Differentiation/biosynthesis , Humans , Mesenchymal Stem Cells/cytology
5.
Osteoarthritis Cartilage ; 23(11): 2045-57, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26521751

ABSTRACT

OBJECTIVE: To define whether good manufacturing practice (GMP)-clinical grade adipose stem cell (ASC)-derived conditioned medium (CM) is as effective as GMP-ASC in modulating inflammatory and catabolic factors released by both osteoarthritis (OA) chondrocytes or synoviocytes. METHODS: OA chondrocytes and synoviocytes were treated with ASC-CM or co-cultured with ASC. Inflammatory factors (IL6, CXCL1/GROα,CXCL8/IL8, CCL2/MCP-1, CCL3/MIP-1α and CCL5/RANTES) and proteinases, such as metalloproteinase (MMP13), a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS4, ADAMTS5) and their tissue metalloproteinase inhibitors (TIMP1, TIMP3) were evaluated by qRT-PCR or immunoassays. The involvement of prostaglandin E2 (PGE2) was also analyzed. RESULTS: Most ASC-CM ratios tested did not decrease IL6, CCL2/MCP-1, CCL3/MIP1-α, CCL5/RANTES on basal inflamed chondrocytes or synoviocytes in contrast to what we found using ASC in co-culture. CXCL8/IL8 and CXCL1/GROα were not decreased by ASC-CM on synoviocytes but were only partially reduced on chondrocytes. Moreover, ASC-CM was less efficient both on basal inflamed OA chondrocytes and synoviocytes in reducing proteinases, such as MMP13, ADAMTS4, ADAMTS5 and increasing TIMP1 and TIMP3 compared to ASC in co-culture. The different ratios of ASC-CM contain lower amounts of PGE2 which were not sufficient to reduce inflammatory factors. CONCLUSIONS: These data show that ASC-CM has a limited ability to decrease inflammatory and proteinases factors produced by OA chondrocytes or synoviocytes. ASC-CM is not sufficient to recapitulate the beneficial effect demonstrated using ASC in co-culture with inflamed OA chondrocytes and synoviocytes and shows that their use in clinical trials is fundamental to counteract OA progression.


Subject(s)
Adipocytes/cytology , Chondrocytes/metabolism , Culture Media, Conditioned/pharmacology , Osteoarthritis, Knee/metabolism , Stem Cell Transplantation/methods , Stem Cells/cytology , Synovial Membrane/metabolism , Aged , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes/pathology , Female , Humans , Male , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/therapy , Synovial Membrane/pathology
6.
Osteoarthritis Cartilage ; 23(7): 1226-30, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25797039

ABSTRACT

OBJECTIVE: To contribute to clarify molecular mechanisms supporting senescence and de-differentiation of chondrocytes in chondrocyte pathologies such as osteoarthritis (OA). Specifically, we investigated the relationship between the nuclear lamina protein Lamin B1 and the negative regulator of chondrogenesis Slug transcription factor in osteoarthritic chondrocytes. METHODS: Lamin B1 and Slug proteins were analyzed in cartilage explants from normal subjects and OA patients by immunohistochemical technique. Their expression was confirmed on isolated chondrocytes both at passage 0 and passage 2 (de-differentiated chondrocytes) by immunofluorescence and western blot. Subsequently, we explored the "in vivo" binding of Slug on LMNB1 promoter by chromatin immunoprecipitation assay (ChIP). RESULTS: In this study we demonstrated that nuclear lamina protein Lamin B1 and anti-chondrogenic Slug transcription factor are upregulated in cartilage and OA chondrocytes. Furthermore, we found that Slug is "in vivo" recruited by LMNB1 gene promoter mostly when chondrocytes undergo de-differentiation or OA degeneration. CONCLUSIONS: We described for the first time a potential regulatory role of Slug on the LMNB1 gene expression in OA chondrocytes. These findings may have important implications for the study of premature senescence, and degeneration of cartilage, and may contribute to develop effective therapeutic strategies against signals supporting cartilage damage in different subsets of patients.


Subject(s)
Chondrocytes/metabolism , Laminin/biosynthesis , Osteoarthritis, Knee/metabolism , Transcription Factors/biosynthesis , Aged , Cartilage, Articular/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Female , Humans , Knee Joint/metabolism , Laminin/genetics , Male , Middle Aged , Osteoarthritis, Knee/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Up-Regulation
7.
Leukemia ; 27(2): 451-63, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22781592

ABSTRACT

Multiple myeloma (MM) is characterized by the impaired osteogenic differentiation of human mesenchymal stromal cells (hMSCs). Canonical Wnt signaling is critical for the regulation of bone formation, however, recent evidence suggests that the non-canonical Wnt agonist Wnt5a stimulates human osteoblastogenesis through its co-receptor Ror2. The effects of MM cells on non-canonical Wnt signaling and the effect of the activation of this pathway on MM-induced osteoblast exhaustion are not known and were investigated in this study. We found that the osteogenic differentiation of bone marrow hMSCs toward osteoprogenitor cells (PreOB) significantly increased Ror2 expression, and that MM cells inhibit Ror2 expression by PreOB in co-culture by inhibiting the non-canonical Wnt5a signaling. The activation of the non-canonical Wnt pathway in hMSCs by means of Wnt5a treatment and the overexpression of Wnt5 or Ror2 by lentiviral vectors increased the osteogenic differentiation of hMSCs and blunted the inhibitory effect of MM in co-culture. Consistently, Wnt5a inhibition by specific small interfering RNA reduced the hMSC expression of osteogenic markers. Our findings demonstrate that the Wnt5a/Ror2 pathway is involved in the pathophysiology of MM-induced bone disease and that the activation of the non-canonical Wnt5a/Ror2 pathway in hMSCs increases osteogenic differentiation and may counterbalance the inhibitory effect of MM cells.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation , Multiple Myeloma/pathology , Osteoblasts/cytology , Osteogenesis , Proto-Oncogene Proteins/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Stem Cells/cytology , Wnt Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Bone Marrow Cells/metabolism , Case-Control Studies , Cell Proliferation , Coculture Techniques , Flow Cytometry , Gene Expression Profiling , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Oligonucleotide Array Sequence Analysis , Osteoblasts/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stem Cells/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics , Wnt-5a Protein
8.
J Biol Regul Homeost Agents ; 25(2 Suppl): S3-13, 2011.
Article in English | MEDLINE | ID: mdl-22051166

ABSTRACT

The requirements for a successful regeneration of an osteo-chondral defect could effectively be met by using a bi-layered composite scaffold, able to support proliferation and differentiation of mesenchymal stem cells, while providing a biochemical environment promoting the formations of the two distinct tissues. The novel strategy here presented consists of developing a bio-mimetic scaffolds obtained by the combination of two integrated organic compounds (type I collagen and chitosan) with or without bioactive Mg-doped hydroxyapatite (Mg-HA) nanocrystals, depending on the specific layer, reproducing cartilaginous or subchondral bone tissue. An innovative patented methodology for scaffolds production, called - pH-dependent 3-phasic assembling -, allowed to development of a highly homogenous and chemically stable scaffold, presenting a very good integration among all three components, as confirmed by extensive SEM and thermogravimetric analyses. A preliminary in vitro evaluation was also carried out by seeding bi-layered scaffold with human bone marrow stromal cells (h-MSCs), by giving particular emphasis to cell viability and distribution at day 0, 7 and 14. Cells were viable and uniformly colonized the whole scaffold until day 14, indicating that the scaffold contributed to the maintenance of cell behaviour.


Subject(s)
Biomimetic Materials/chemistry , Bone Marrow Cells/cytology , Bone Regeneration , Cartilage , Materials Testing , Tissue Scaffolds/chemistry , Bone Marrow Cells/metabolism , Bone Substitutes/chemistry , Cells, Cultured , Chitosan/chemistry , Collagen Type I/chemistry , Durapatite/chemistry , Humans , Stromal Cells/cytology , Stromal Cells/metabolism
9.
Leukemia ; 25(3): 527-37, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21183939

ABSTRACT

The deregulation of the homeobox genes as homeoboxB (HOXB)-7 has been previously associated to tumor progression and angiogenesis; here we investigated the potential role of HOXB7 in the pro-angiogenic properties of multiple myeloma (MM) cells. We found that HOXB7 was expressed in 10 out of 22 MM patients analyzed at the diagnosis related to high bone marrow angiogenesis and overexpressed in about 40% of myeloma cell lines compared with normal plasma cells. Enforced HOXB7 expression in MM cells by a lentiviral vector significantly modified their transcriptional and angiogenic profile, checked by combined microarray and angiogenesis PCR analyses, upregulating VEGFA, FGF2, MMP2, WNT5a and PDGFA and downregulating thrombospoindin-2. The pro- and anti-angiogenic HOXB7-related gene signature was also validated in a large independent dataset of MM patients. Accordingly, MM-induced vessel formation was significantly increased by HOXB7 overexpression both in vitro angiogenic and chorioallantoic membrane assays, as well as the HOXB7 silencing by small interfering RNA inhibited the production of angiogenic factors, and the pro-angiogenic properties of MM cells. Finally, in SCID-NOD mice we confirmed that HOXB7 overexpression by MM cells stimulated tumor growth, increased MM-associated angiogenesis and the expression of pro-angiogenic genes by microarray analysis supporting the critical role of HOXB7 in the angiogenic switch in MM.


Subject(s)
Homeodomain Proteins/physiology , Multiple Myeloma/blood supply , Neovascularization, Pathologic/etiology , Aged , Animals , Cell Line, Tumor , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Vascular Endothelial Growth Factor A/biosynthesis
11.
J Biomed Mater Res A ; 73(3): 275-83, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15789422

ABSTRACT

Ligaments are complex structures that maintain the mechanical stability of the joint. Healing of injured ligaments involves the interactions of different cell types, local cellular environment, and the use of devices. To gain new information on the complex interactions between mesenchymal stem cells (MSCs) and a specific hyaluronan-based prototype scaffold (HYAFF, useful for ligament tissue engineering, short time-course experiments were performed to analyze the proliferation, vitality, and phenotype of MSCs grown on the scaffold. MSC proliferation was analyzed using the MTT test, during the early time points (2, 4, 6, days). Viability was assessed using calcein/acetyloxymethylester immunofluorescence dye and confocal microscopy analysis. Hyaluronic acid receptor (CD44), typical matrix ligament proteins (collagen type I, type III, laminin, fibronectin, actin), and chondrogenic/osteogenic markers (collagen type II and bone sialoprotein) were evaluated by immunohistochemistry. Our data demonstrated that MSC growth and viability were cell density-dependent. MSCs completely wrapped the fibers of the scaffold, expressed CD44, collagen type I, type III, laminin, fibronectin, and actin, and were negative to collagen type II and bone sialoprotein. These data demonstrate that MSCs survive well in the hyaluronan-based prototype ligament scaffold, as assessed after 2 days from seeding, and express CD44, a receptor important for scaffold interaction, and proteins responsible for the functional characteristics of the ligaments.


Subject(s)
Cell Proliferation , Culture Techniques , Hyaluronic Acid/analogs & derivatives , Ligaments, Articular , Mesenchymal Stem Cells/physiology , Animals , Biocompatible Materials/metabolism , Cell Shape , Cell Survival , Cells, Cultured , Extracellular Matrix , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Materials Testing , Mesenchymal Stem Cells/cytology , Sheep , Tissue Engineering
12.
Chir Organi Mov ; 88(4): 351-5, 2003.
Article in English, Italian | MEDLINE | ID: mdl-15259550

ABSTRACT

Association of biomaterials with autologous cells can provide a new generation of implantable devices for cartilage and bone repair. Such scaffolds should provide a performed three-dimensional shape, prevent cells from floating out of the defect, have sufficient mechanical strength, facilitate uniform spread of cells, and stimulate the phenotype of transplanted cells. Hyaff-11 is a recently developed hyaluronic-acid based biodegradable polymer, that has been shown to provide successful cell scaffolds for tissue-engineered repair. The aim of this study was to evaluate in vitro the potential of Hyaff-11 to support the growth of human chondrocytes and to maintain their original phenotype. Our data indicate that human chondrocytes seeded on Hyaff-11 express and produce collagen type II and aggrecan and downregulate the production of collagen type I. These results provide an in vitro demonstration of therapeutic potential of Hyaff-11 as a delivery vehicle in tissue-engineered repair of articular cartilage defects.


Subject(s)
Cartilage/cytology , Hyaluronic Acid/analogs & derivatives , Tissue Engineering , Adolescent , Adult , Cells, Cultured , Humans , Tissue Engineering/methods
13.
Chir Organi Mov ; 88(4): 363-7, 2003.
Article in English, Italian | MEDLINE | ID: mdl-15259552

ABSTRACT

Various techniques are widely used to repair bone defects, association of hyaluronan-based biodegradable polymers (Hyaff-11) with bone marrow stromal cells (BMSC) promises to provide successful cell scaffolds for tissue-engineered repair of bone tissue. We evaluate in vitro and in vivo the potential of Hyaff-11 to facilitate mineralization of BMSC. Rat BMSC were seeded on Hyaff-11 and their differentiation were assessed at different time points. Osteogenic differentiation was investigated in vitro analysing the expression of alkaline phosphatase and osteocalcin. Mineralization of bone defects was evaluated also in vivo implanting Hyaff-11 scaffold combined with BMSC in large segmental radius defects. In vitro, we found a decrease expression of alkaline phosphatase and an increase of osteocalcin. In vivo, our data showed that mineralization was induced and basic fibroblast growth factor contributed to this process. These results provide a demonstration to therapeutic potential of Hyaff-11 as appropriate carrier vehicle for differentiation and mineralization of BMSC and for the repair of bone defects.


Subject(s)
Bone Marrow Cells , Calcification, Physiologic , Hyaluronic Acid/analogs & derivatives , Stromal Cells , Animals , Rats , Rats, Inbred F344
14.
Biomaterials ; 23(4): 1043-51, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11791907

ABSTRACT

Osteogenesis of large segmental radius defects in a rat model was studied by implanting a biodegradable non-woven hyaluronic acid-based polymer scaffold (Hyaff 11) alone or in combination with bone marrow stromal cells (BMSCs). These cells had been previously grown in vitro in mineralising medium either supplemented with basic fibroblast growth factor (bFGF) or unsupplemented. The healing of bone defects was evaluated at 40, 80, 160 and 200 days and the repair process investigated by radiographic, histomorphometric (assessment of new bone growth and lamellar bone) and histological analyses (toluidine blue and von Kossa staining). Mineralisation of bone defects occurred in the presence of the Hyaff 11 scaffold alone or when combined with BMSCs grown with or without bFGF, but each process had a different timing. In particular, bFGF significantly induced mineralisation from day 40, whereas 160 days were necessary for direct evidence that a similar process was developing under the other two conditions tested (scaffold alone or with BMSCs). Radiographic score, new bone growth and lamellar bone percentage were highly correlated. The present outcomes were further confirmed by toluidine blue and von Kossa staining. According to these in vivo findings, the Hyaff 11 scaffold is an appropriate carrier vehicle for the repair of bone defects; additionally, it can significantly accelerate bone mineralisation in combination with BMSCs and bFGF.


Subject(s)
Bone Marrow Cells/drug effects , Bone Marrow Transplantation , Fibroblast Growth Factor 2/pharmacology , Hyaluronic Acid/analogs & derivatives , Osteogenesis/physiology , Animals , Biocompatible Materials , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Bone Regeneration , Fracture Healing , In Vitro Techniques , Male , Polymers , Rats , Rats, Inbred F344
15.
Connect Tissue Res ; 42(1): 49-58, 2001.
Article in English | MEDLINE | ID: mdl-11696988

ABSTRACT

Bone marrow stromal cells (BMSCs) for osteoblast differentiation studies can be obtained by gradient isolation techniques or by directly plating a filtered cell suspension. We compared these two procedures to evaluate whether this step is critical in order to obtain a high number of differentiated colonies. Isolated primary rat BMSCs were cultured in vitro with or without insulin-like growth factor II (IGFII), basic fibroblast growth factor (b-FGF), epidermal growth factor (EGF) or transforming growth factor beta 1 (TGF beta 1), and histochemically and biochemically analysed at different time points. The gradient procedure produced a significantly higher number of colonies capable of osteoblastic differentiation. The growth factors had different effects. In particular, b-FGF and EGF significantly increased the number of Alizarin red S positive colonics, while IGFII and TGF beta I exerted inhibitory effects. Nodules obtained on day 21 showed some alkaline phosphatase positive cells and were Von Kossa-positive. These data demonstrate that more differentiated colonies are obtainable from BMSCs isolated by the gradient procedure.


Subject(s)
Bone Marrow Cells/cytology , Cell Separation/methods , Centrifugation, Density Gradient/methods , Osteoblasts/cytology , Stromal Cells/cytology , Alkaline Phosphatase/metabolism , Animals , Bone Marrow Cells/drug effects , Cell Count , Cell Differentiation , Cells, Cultured , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Insulin-Like Growth Factor II/pharmacology , Male , Rats , Rats, Inbred F344 , Stromal Cells/drug effects , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1
16.
Arthritis Rheum ; 44(8): 1800-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11508432

ABSTRACT

OBJECTIVE: To investigate the in vitro effect of therapeutic hyaluronan (HA) of 500-730 kd on anti-Fas-induced apoptosis of chondrocytes from osteoarthritis (OA) patients, and to assess its mechanism of action by analyzing the role of the 2 HA receptors, CD44 and CD54 (intercellular adhesion molecule 1 [ICAM-1]). METHODS: Chondrocytes isolated from human OA knee cartilage were cultured and the effect of HA on both spontaneous and anti-Fas-induced apoptosis was evaluated. Apoptosis was analyzed by JAM test (for quantitative analysis of fragmented DNA), cell death detection immunoassay (for quantitative analysis of oligonucleosome), TUNEL assay, and electron microscopy. Blocking experiments with anti-CD44 and anti-CD54 alone or in combination were performed to investigate the HA mechanism of action. RESULTS: Both quantitative tests demonstrated that anti-Fas significantly induced apoptosis of isolated OA chondrocytes. HA at 1,000 microg/ml significantly reduced the anti-Fas-induced apoptosis of chondrocytes but did not affect spontaneous chondrocyte apoptosis. These data were also confirmed by TUNEL staining and by electron microscopy morphologic evaluation. The antiapoptotic effects of HA on anti-FAS-induced chondrocyte apoptosis were significantly decreased by both anti-CD44 (mean +/- SD 57 +/- 12% inhibition) and anti-ICAM-1 (31 +/- 22% inhibition). The mixture of the 2 antibodies had an additive effect, since the rate of inhibition increased to 87 +/- 13%. CONCLUSION: These data demonstrate that 500-730-kd HA exerts an antiapoptotic effect on anti-FAS-induced chondrocyte apoptosis by binding its specific receptors (CD44 and ICAM-1). Furthermore, this HA fraction may be able to slow down chondrocyte apoptosis in OA by regulating the processes of cartilage matrix degradation.


Subject(s)
Apoptosis/drug effects , Chondrocytes/pathology , Hyaluronan Receptors/physiology , Hyaluronic Acid/pharmacology , Intercellular Adhesion Molecule-1/physiology , Osteoarthritis/pathology , fas Receptor/physiology , Aged , Antibodies/immunology , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/ultrastructure , Chromatin/ultrastructure , Female , Humans , In Situ Nick-End Labeling , Male , fas Receptor/immunology
17.
Biomaterials ; 22(15): 2095-105, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11432589

ABSTRACT

A biodegradable non-woven hyaluronic acid polymer scaffold (Hyaff 11) was analysed in vitro as a carrier vehicle for differentiation and mineralization of rat bone marrow stromal cells (BMSC). BMSC were grown on Hyaff 11 in a mineralizing medium in the presence/absence of basic fibroblast growth factor (bFGF). Osteoblastic differentiation was investigated by light and electron microscopy analysing the expression of osteogenic markers: calcium, alkaline phosphatase (AP), osteopontin (OP), bone sialoprotein (BSP) and collagen type 1. We also measured proliferation, AP activity and mRNA expression of AP and osteocalcin (OC). Electron microscopy and Toluidine-blue staining demonstrated that bFGF accelerated (day 20 vs. day 40) and increased mineralization. With bFGF, calcium, OP and BSP were strongly enhanced at day 40, whereas AP decreased. Our in vitro results demonstrate that Hyaff 11 is a useful vehicle for growth, differentiation and mineralization of rat BMSC, and that it permits bone development.


Subject(s)
Bone Marrow Cells/metabolism , Fibroblast Growth Factor 2/pharmacology , Hyaluronic Acid/chemistry , Polymers/chemistry , Stromal Cells/cytology , Alkaline Phosphatase/metabolism , Animals , Biocompatible Materials/chemistry , Calcium/metabolism , Cell Culture Techniques/methods , Cell Division , Cells, Cultured , Collagen Type I/metabolism , Coloring Agents/pharmacology , Culture Media , Integrin-Binding Sialoprotein , Kinetics , Microscopy, Electron , Osteoblasts/cytology , Osteocalcin/metabolism , Osteopontin , Protein Binding , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Sialoglycoproteins/metabolism , Time Factors , Tolonium Chloride/pharmacology
18.
Arthritis Rheum ; 44(6): 1444-54, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11407707

ABSTRACT

OBJECTIVE: The imbalance between matrix metalloproteinases (MMPs) 1, 3, and 9 and their specific inhibitor, tissue inhibitor of metalloproteinases 1 (TIMP-1), is a critical step in cartilage injury and angiogenesis in arthritis. To explore the therapeutic potential of TIMP-1 gene transfer in erosive arthritis, the effects of an adenoviral vector (Ad-TIMP-1) were assessed in DBA/1 mice with collagen-induced arthritis (CIA). METHODS: DBA/1 mice with CIA received an intravenous injection of replication-deficient adenovirus containing the human TIMP-1 gene or a control LacZ gene on day 28 postimmunization. The efficiency of gene transfer was determined by serum TIMP-1 detection, measurements of paw swelling, as well as radiologic and histologic examination of the paws. RESULTS: A single administration of Ad-TIMP-1 resulted in detectable serum levels of the exogenous protein for at least 13 days. The incidence and onset of arthritis were not statistically modified after human TIMP-1 gene transfer in DBA/1 mice compared with control mice. However, the severity of inflammation was statistically significantly increased in Ad-TIMP-1-treated mice and a similar trend was observed in the histologic and radiologic scores. With regard to the mechanisms of the worsened effect in the Ad-TIMP-1-treated mice, we observed 1) higher serum levels of anti-type II collagen IgG2a, 2) a significant increase in endogenous soluble tumor necrosis factor receptor I (TNFRI) in sera, and 3) increased labeling of mouse tumor necrosis factor alpha and TNFRI within arthritic joints. CONCLUSION: These findings show that overexpression of TIMP-1 does not prevent osteochondral injury in a mouse model of arthritis. Since MMPs have overlapping properties in terms of their roles in extracellular matrix degradation, angiogenesis, and shedding of cell surface adhesion molecules, cytokines, and cytokine receptors, the paradoxical results obtained suggest that TIMP-1 is probably not the main inhibitor to target.


Subject(s)
Arthritis, Experimental/therapy , Gene Transfer Techniques , Genetic Therapy , Tissue Inhibitor of Metalloproteinase-1/genetics , Adenoviridae , Animals , Arthritis, Experimental/blood , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Cells, Cultured , Collagen/immunology , Collagen/pharmacology , Edema/chemically induced , Edema/pathology , Flow Cytometry , Foot/pathology , Genetic Vectors , Hindlimb/drug effects , Hindlimb/pathology , Humans , Male , Mice , Mice, Inbred DBA , Receptors, Tumor Necrosis Factor/blood , Synovial Membrane/cytology , Synovial Membrane/immunology , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/immunology
19.
Osteoarthritis Cartilage ; 9(2): 161-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11237663

ABSTRACT

OBJECTIVE: Many studies have evidenced the clinical efficacy of hyaluronan (HA) in the treatment of osteoarthritis (OA). However, human and animal studies have described proinflammatory effects of HA on cells not involved in OA. We therefore investigated whether different molecular weight HA preparations can affect proinflammatory cytokine (IL1beta and TNFalpha) or chemokine (IL8, MCP-1 and RANTES) expression in human chondrocytes and synoviocytes isolated from OA patients. DESIGN: Human chondrocytes and synoviocytes were cultured in vitro in the presence or absence of three different purified HA pharmaceutical preparations (1x10(6) Kd, 5x10(5) Kd and 6.5x10(4) Kd) and assessed for the production of proinflammatory cytokines and chemokines and their mRNA expression. RESULTS: basal conditions, both chondrocytes and synoviocytes produce only MCP-1 and IL8, along with low quantities of IL1beta and TNFalpha, but not RANTES. IL8 production was generally about 100 times higher in chondrocytes than in synoviocytes, while MCP-1 was roughly twice as high in synoviocytes than in chondrocytes. At the mRNA level, expression of IL1beta, TNFalpha, IL8, MCP-1 and RANTES did not change in the presence of the three HA preparations either in synoviocytes or in chondrocytes with respect to basal condition. None of the three different HA preparations significantly affected production of IL8 or MCP-1. CONCLUSIONS: These data demonstrate that preparations of HA of the same origin but with different MWs do not induce proinflammatory cytokines and chemokines expressed by chondrocytes and synoviocytes that are either directly or indirectly involved in OA progression.


Subject(s)
Adjuvants, Immunologic/pharmacology , Chemokines/metabolism , Chondrocytes/drug effects , Cytokines/metabolism , Hyaluronic Acid/pharmacology , Osteoarthritis/drug therapy , Synovial Membrane/drug effects , Chondrocytes/metabolism , Humans , Middle Aged , Osteoarthritis/metabolism , RNA, Messenger/metabolism , Synovial Membrane/metabolism
20.
Clin Exp Immunol ; 119(2): 346-53, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10632674

ABSTRACT

We investigated both in vitro and ex vivo the role of mature osteoblasts (OB) and bone marrow stromal cells (BMSC) in RA and OA by analysing the expression of the following IL-6-type cytokines: IL-11, leukaemia inhibitory factor (LIF), oncostatin M (OSM) and IL-6. OB and BMSC were isolated from femora of RA, OA and post-traumatic (PT) patients, cultured in vitro in the presence or absence of IL-1beta and tumour necrosis factor-alpha (TNF-alpha), and assessed for the production and mRNA expression of IL-6-type cytokines. Trabecular bone biopsies were obtained from the inner portions of femoral heads and used for cytokine in situ immunostaining. Cultured OB and BMSC from different patients constitutively secreted IL-11 and IL-6 but not OSM. LIF was secreted only by BMSC, at very low levels. Interestingly, IL-11 basal production was significantly higher in BMSC than in OB in all three groups tested. IL-1beta and TNF-alpha strongly stimulated IL-6-type cytokine release (except for OSM) by both OB and BMSC. OSM was expressed only at mRNA levels in all groups studied. Cytokine immunostaining on bone biopsies confirmed the data obtained on cultured cells: IL-11, IL-6 and LIF proteins were detected both in mesenchymal (BMSC and OB) and mononuclear cells; OSM was found only in mononuclear cells. These data demonstrate that IL-6-type cytokines are constitutively expressed in the bone compartment in RA, OA and PT patients and can be secreted by bone cells at different stages of differentiation (BMSC and OB). This suggests that these cytokines may be involved in the mechanisms of bone remodelling in OA and RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Bone Marrow Cells/metabolism , Femur/pathology , Growth Inhibitors/biosynthesis , Interleukin-11/biosynthesis , Lymphokines/biosynthesis , Osteoarthritis/metabolism , Osteoblasts/metabolism , Peptides/metabolism , Arthritis, Rheumatoid/pathology , Biopsy , Bone Marrow Cells/pathology , Cytokines/chemistry , Cytokines/genetics , Cytokines/metabolism , Femur/metabolism , Gene Expression Regulation , Humans , Interleukin-6/biosynthesis , Leukemia Inhibitory Factor , Middle Aged , Oncostatin M , Osteoarthritis/pathology , Osteoblasts/pathology , Stromal Cells/metabolism , Stromal Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...