Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Cells ; 10(8)2021 08 20.
Article in English | MEDLINE | ID: mdl-34440921

ABSTRACT

Transplantation of xenogenic porcine chondrocytes could represent a future strategy for the treatment of human articular cartilage defects. Major obstacles are humoral and cellular rejection processes triggered by xenogenic epitopes like α-1,3-Gal and Neu5Gc. Besides knockout (KO) of genes responsible for the biosynthesis of respective epitopes (GGTA1 and CMAH), transgenic expression of human complement inhibitors and anti-apoptotic as well as anti-inflammatory factors (CD46, CD55, CD59, TNFAIP3 and HMOX1) could synergistically prevent hyperacute xenograft rejection. Therefore, chondrocytes from different strains of single- or multi-genetically modified pigs were characterized concerning their protection from xenogeneic complement activation. Articular chondrocytes were isolated from the knee joints of WT, GalTKO, GalT/CMAH-KO, human CD59/CD55//CD46/TNFAIP3/HMOX1-transgenic (TG), GalTKO/TG and GalT/CMAHKO/TG pigs. The tissue-specific effectiveness of the genetic modifications was tested on gene, protein and epitope expression level or by functional assays. After exposure to 20% and 40% normal human serum (NHS), deposition of C3b/iC3b/C3c and formation of the terminal complement complex (TCC, C5b-9) was quantified by specific cell ELISAs, and generation of the anaphylatoxin C5a by ELISA. Chondrocyte lysis was analyzed by Trypan Blue Exclusion Assay. In all respective KO variants, the absence of α -1,3-Gal and Neu5Gc epitope was verified by FACS analysis. In chondrocytes derived from TG animals, expression of CD55 and CD59 could be confirmed on gene and protein level, TNFAIP3 on gene expression level as well as by functional assays and CD46 only on gene expression level whereas transgenic HMOX1 expression was not evident. Complement activation in the presence of NHS indicated mainly effective although incomplete protection against C3b/iC3b/C3c deposition, C5a-generation and C5b-9 formation being lowest in single GalTKO. Chondrocyte viability under exposure to NHS was significantly improved even by single GalTKO and completely preserved by all other variants including TG chondrocytes without KO of xenoepitopes.


Subject(s)
Bone Diseases/therapy , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Transplantation, Heterologous/methods , Animals , Animals, Genetically Modified , Bone Diseases/genetics , CD55 Antigens/genetics , CD55 Antigens/metabolism , CD59 Antigens/genetics , CD59 Antigens/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/cytology , Complement System Proteins/genetics , Complement System Proteins/metabolism , Gene Expression , Gene Knockout Techniques , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/metabolism , Swine , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
2.
Cytotherapy ; 20(2): 218-231, 2018 02.
Article in English | MEDLINE | ID: mdl-29223534

ABSTRACT

BACKGROUND: Effective therapy of Acute Lung Injury (ALI) is still a major scientific and clinical problem. To define novel therapeutic strategies for sequelae of blunt chest trauma (TxT) like ALI/Acute Respiratory Distress Syndrome, we have investigated the immunomodulatory and regenerative effects of a single dose of ex vivo expanded human or rat mesenchymal stromal cells (hMSCs/rMSCs) with or without priming, immediately after the induction of TxT in Wistar rats. METHODS: We analyzed the histological score of lung injury, the cell count of the broncho alveolar lavage fluid (BAL), the change in local and systemic cytokine level and the recovery of the administered cells 24 h and 5 days post trauma. RESULTS: The treatment with hMSCs reduced the injury score 24 h after trauma by at least 50% compared with TxT rats without MSCs. In general, TxT rats treated with hMSCs exhibited a lower level of pro-inflammatory cytokines (interleukin [IL]-1B, IL-6) and chemokines (C-X-C motif chemokine ligand 1 [CXCL1], C-C motif chemokine ligand 2 [CCL2]), but a higher tumor necrosis factor alpha induced protein 6 (TNFAIP6) level in the BAL compared with TxT rats after 24 h. Five days after trauma, cytokine levels and the distribution of inflammatory cells were similar to sham rats. In contrast, the treatment with rMSCs did not reveal such therapeutic effects on the injury score and cytokine levels, except for TNFAIP6 level. CONCLUSION: TxT represents a suitable model to study effects of MSCs as an acute treatment strategy after trauma. However, the source of MSCs has to be carefully considered in the design of future studies.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Thoracic Injuries/therapy , Transplantation, Heterologous , Wounds, Nonpenetrating/therapy , Animals , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Cell Shape , Cytokines/metabolism , Disease Models, Animal , Humans , Lung/pathology , Male , Organic Chemicals/metabolism , Rats , Rats, Wistar , Thoracic Injuries/pathology , Transplantation, Homologous , Wounds, Nonpenetrating/pathology
3.
Am J Sports Med ; 43(11): 2822-32, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26362437

ABSTRACT

BACKGROUND: Sport injuries of the knee often lead to posttraumatic arthritis. In addition to direct damage of the cartilage, trauma-associated intra-articular bleeding may cause hemarthrosis. Both blood exposure and trauma are known to induce cell death and inflammation and to enhance proteoglycan release in cartilage. HYPOTHESIS: Blood exposure increases chondrocyte death as well as inflammatory and degenerative processes in traumatized cartilage. STUDY DESIGN: Controlled laboratory study. METHODS: Human macroscopically intact osteoarthritic (OA) cartilage explants were impacted by a drop-tower system (0.59 J) and cultivated with or without 10% blood. Interactive effects were studied concerning cell survival, gene expression, and the release of mediators over 24 hours and 96 hours. To evaluate the effects of trauma and hemarthrosis in vivo, a newly established blunt cartilage trauma model in the rabbit was used. Treatment of the knee joints of mature New Zealand White rabbits consisted of the following groups: control (C), arthrotomy (A), arthrotomy with cartilage trauma (AT; 1.0 J), and arthrotomy with cartilage trauma and blood injection (ATH). After 1 and 12 weeks, inflammatory mediators in the synovial fluid and histological changes of the cartilage were determined, and immunohistological staining was performed. RESULTS: The in vitro studies revealed a significant additional or synergistic effect of blood exposure on trauma-induced chondrocyte death, interleukin (IL)-1ß and prostaglandin-E2 (PGE2) release, and matrix metalloproteinase (MMP)/pro-MMP level. Singular arthrotomy in vivo induced a temporary inflammation. Histologically, cartilage trauma caused significant OA changes that were not aggravated by an additional hemarthrosis. Trauma led to a persistent deposition of terminal complement complex (TCC), being enhanced by hemarthrosis. However, trauma-induced formation of osteophytes and arthrotomy-induced elevation of tumor necrosis factor-α release were reduced by hemarthrosis. CONCLUSION: While blood exposure clearly aggravated trauma-induced OA processes in the in vitro model, a singular blood injection revealed heterogeneous effects in vivo, enhancing TCC deposition but reducing trauma-induced osteophyte formation while the histological score of traumatized cartilage was not further impaired. CLINICAL RELEVANCE: The results of this study indicate that a singular, limited bleeding event might not exacerbate early trauma-induced cartilage degeneration in joint injuries. An early removal of intra-articular blood may not prevent the final resulting cartilage damage.


Subject(s)
Cartilage, Articular/injuries , Chondrocytes/pathology , Inflammation/pathology , Knee Joint/pathology , Aged , Animals , Cell Death , Cell Survival , Dinoprostone/metabolism , Female , Gene Expression , Hemarthrosis/metabolism , Humans , Interleukin-1beta/metabolism , Male , Matrix Metalloproteinases/metabolism , Middle Aged , Proteoglycans/metabolism , Rabbits , Synovial Fluid/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
PLoS One ; 10(1): e0116772, 2015.
Article in English | MEDLINE | ID: mdl-25562599

ABSTRACT

Multipotent mesenchymal stromal cells (MSC) exert immune-modulatory effects and support tissue regeneration in various local trauma models. In case of a polytrauma, high amounts of danger-associated molecular patterns are released, leading to a systemic increase of inflammatory mediators. The influence of such a complex inflammatory microenvironment on human MSC is mainly unknown so far. Therefore, we investigated the effects of a defined serum-free polytrauma "cocktail" containing IL beta, IL6, IL8 and the anaphylatoxins C3a and C5a, in concentrations corresponding to those measured in the blood of polytrauma patients, on human MSC in vitro. The polytrauma cocktail induced directed migration of MSC with C3a representing its major soluble chemoattractive agent. Furthermore, the polytrauma cocktail and IL1beta upregulated the expression of MMP1 indicating a potential role of IL1beta to enhance MSC migration in the tissue context. COX2, PTGES and TSG6 were also found to be upregulated upon stimulation with the polytrauma cocktail or IL1beta, but not through other single factors of the polytrauma cocktail in pathophysiologically relevant concentrations. An RNA expression array of 84 inflammation-related genes revealed that both the polytrauma cocktail and IL1beta induced C3, CSF1, TLR3 and various chemokines without major qualitative or quantitative differences. These results indicate that IL1beta is a crucial mediator of the polytrauma cocktail in terms of immune-modulation and MMP1 expression. Thus, upon encountering the primary sterile, inflammatory milieu of a polytrauma, endogenous or systemically transfused MSC might be able to migrate to sites of injury, secrete TSG6 and PGE2 and to influence macrophage biology as observed in local trauma models.


Subject(s)
Complement C3a/pharmacology , Inflammation Mediators/pharmacology , Interleukin-1beta/pharmacology , Mesenchymal Stem Cells/drug effects , Bone Marrow Cells/cytology , Cell Adhesion Molecules/analysis , Cell Movement/drug effects , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Cyclooxygenase 2/metabolism , Cytokines/genetics , Cytokines/metabolism , Dinoprostone/analysis , Enzyme-Linked Immunosorbent Assay , Humans , Intramolecular Oxidoreductases/metabolism , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Prostaglandin-E Synthases , Up-Regulation/drug effects
5.
PLoS One ; 9(10): e109411, 2014.
Article in English | MEDLINE | ID: mdl-25329487

ABSTRACT

Designing of implant surfaces using a suitable ligand for cell adhesion to stimulate specific biological responses of stem cells will boost the application of regenerative implants. For example, materials that facilitate rapid and guided migration of stem cells would promote tissue regeneration. When seeded on fibronectin (FN) that was homogeneously immmobilized to NCO-sP(EO-stat-PO), which otherwise prevents protein binding and cell adhesion, human mesenchymal stem cells (MSC) revealed a faster migration, increased spreading and a more rapid organization of different cellular components for cell adhesion on fibronectin than on a glass surface. To further explore, how a structural organization of FN controls the behavior of MSC, adhesive lines of FN with varying width between 10 µm and 80 µm and spacings between 5 µm and 20 µm that did not allow cell adhesion were generated. In dependance on both line width and gaps, cells formed adjacent cell contacts, were individually organized in lines, or bridged the lines. With decreasing sizes of FN lines, speed and directionality of cell migration increased, which correlated with organization of the actin cytoskeleton, size and shape of the nuclei as well as of focal adhesions. Together, defined FN lines and gaps enabled a fine tuning of the structural organization of cellular components and migration. Microstructured adhesive substrates can mimic the extracellular matrix in vivo and stimulate cellular mechanisms which play a role in tissue regeneration.


Subject(s)
Cell Adhesion/drug effects , Fibronectins/pharmacology , Mesenchymal Stem Cells/cytology , Regeneration , Actins/metabolism , Cell Line , Cell Movement/drug effects , Cytoskeleton/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/physiology , Humans , Ligands , Mesenchymal Stem Cells/drug effects
6.
PLoS One ; 9(1): e86151, 2014.
Article in English | MEDLINE | ID: mdl-24465929

ABSTRACT

The aim of the present study was to test the biocompatibility and functionality of orthopaedic bone implants with immobilized oligonucleotides serving as anchor stands for rhBMP-2 and rhVEGF-A conjugated with complementary oligonucleotides in an osteoporotic rat model. Al2O3-blasted acid etched Ti6Al4V implants, carrying oligonucleotide anchor strands and hybridized with rhBMP-2 or rhVEGF-A through complementary 31-mer oligonucleotide stands were inserted into the proximal tibia of ovariectomized rats. At the time of surgery (15 weeks after ovariectomy) microCT analysis showed significantly lower bone mineral density compared to non-ovariectomized animals. Bone-implant contact (BIC) and pullout-force were not negatively affected by non-hybridized anchor strands. Twelve weeks after surgery, a significantly higher pullout force was found for BMP-2 hybridized to the anchor strands compared to non-hybridized anchor strands or native samples, and on histomorphometric analysis BIC was highest in the BMP group. Thus, we could show the biocompatibility and in vivo functionality of this modular, self-organizing system for immobilization and subsequent release of BMP-2 in vivo.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Immobilized Proteins/metabolism , Implants, Experimental , Oligonucleotides/metabolism , Osteoporosis/therapy , Tibia/pathology , Titanium/pharmacology , Transforming Growth Factor beta/metabolism , Alloys , Animals , Biomechanical Phenomena/drug effects , Bone Density/drug effects , Female , Humans , Microscopy, Electron, Scanning , Orthopedics , Osteoporosis/diagnostic imaging , Osteoporosis/physiopathology , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Surface Properties , Tibia/diagnostic imaging , Tibia/drug effects , Tibia/physiopathology , X-Ray Microtomography
7.
Biomaterials ; 34(35): 8851-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23968851

ABSTRACT

Hexagonally arranged Gold nanoparticles with controllable diameters and inter-particle distances were deposited on thick SiO2 layers on top of Si wafers and used as masks during subsequent reactive ion etching. In this way, arrays of nanopillars are obtained with well-defined diameters (10/30 nm), inter-pillar distances (50-120 nm) and heights (20-35 nm), all on the nanoscale. Such nanotopographies served as substrate for multipotent mesenchymal stromal cells (MSC) and human osteoblasts (OB) allowing to study cellular responses to purely topographically patterned interfaces. Focus was put on adhesion, proliferation and differentiation of the cells. It turned out experimentally that adhesion is comparable for both cell types practically independent of topographical details at the substrate surface. Topography induced proliferation enhancement, however, is again independent of geometrical details in case of MSC, but significantly sensitive to pillar height in case of OB with a clear preference towards short nanopillars (20 nm). A high sensitivity to topographic details is also observed for osteogenic differentiation of MSC, in that case with a preference towards higher nanopillars (50 nm). The present experimental data also allow the important conclusion that cell proliferation and differentiation can be optimized simultaneously by fine-tuning nanoscaled topographical parameters.


Subject(s)
Mesenchymal Stem Cells/metabolism , Nanoparticles/chemistry , Osteoblasts/metabolism , Surface Properties , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Mesenchymal Stem Cells/cytology , Microscopy, Electron, Scanning , Nanotechnology/methods , Osteoblasts/cytology , Osteogenesis , Silicon Dioxide/chemistry
8.
J Mater Sci Mater Med ; 24(10): 2417-27, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23801500

ABSTRACT

Hydrogel coatings prepared from reactive star shaped polyethylene oxide based prepolymers (NCO-sP(EO-stat-PO)) minimize unspecific protein adsorption in vitro, while proteins immobilized on NCO-sP(EO-stat-PO) coatings retain their structure and biological function. The aim of the present study was to assess biocompatibility and the effect on early osseointegrative properties of a NCO-sP(EO-stat-PO) coating with additional RGD-peptides and augmentation with bone morphogenetic protein-4 (BMP) used on a medical grade high-density polyethylene (HDPE) base under in vivo circumstances. For testing of biocompatibility dishes with large amounts of bulk NCO-sP(EO-stat-PO) were implanted subcutaneously into 14 Wistar rats. In a second set-up functionalization of implants with ultrathin surface layers by coating ammonia-plasma treated HDPE with NCO-sP(EO-stat-PO), functionalization with linear RGD-peptides, and augmentation with RGD and BMP-4 was analyzed. Therefore, implants were placed subcutaneously in the paravertebral tissue and transcortically in the distal femur of another 14 Wistar rats. Both tests revealed no signs of enhanced inflammation of the surrounding tissue analyzed by CD68, IL-1ß-/TNF-α-antibody staining, nor systemic toxic reactions according to histological analysis of various organs. The mean thickness of the fibrous tissue surrounding the femoral implants was highest in native HDPE-implants and tended to be lower in all NCO-sP(EO-stat-PO) modified implants. Micro-CT analysis revealed a significant increase of peri-implant bone volume in RGD/BMP-4 coated samples. These results demonstrate that even very low amounts of surface bound growth factors do have significant effects when immobilized in an environment that retains their biological function. Hence, NCO-sP(EO-stat-PO)-coatings could offer an attractive platform to improve integration of orthopedic implants.


Subject(s)
Coated Materials, Biocompatible/chemistry , Hydrogels/chemistry , Oligopeptides/chemistry , Polyethylene Glycols/chemistry , Adsorption , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Bone Morphogenetic Protein 4/chemistry , Cell Adhesion , Femur/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-1beta/metabolism , Osseointegration , Prostheses and Implants , Rats , Rats, Wistar , Surface Properties , Time Factors , X-Ray Microtomography
9.
Mol Med ; 19: 36-42, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23508571

ABSTRACT

Bone has a high capacity for self-renewal and repair. Prolonged local secretion of interleukin 1ß (IL-1ß), however, is known to be associated with severe bone loss and delayed fracture healing. Since induction of bone resorption by IL-1ß may not sufficiently explain these pathologic processes, we investigated, in vitro, if and how IL-1ß affects migration of multipotent mesenchymal stromal cells (MSC) or osteoblasts. We found that homogenous exposure to IL-1ß significantly diminished both nondirectional migration and site-directed migration toward the chemotactic factors platelet-derived growth factor (PDGF)-BB and insulin like growth factor 1 (IGF-1) in osteoblasts. Exposure to a concentration gradient of IL-1ß induced an even stronger inhibition of migration and completely abolished the migratory response of osteoblasts toward PDGF-BB, IGF-1, vascular endothelial growth factor A (VEGF-A) and the complement factor C5a. IL-1ß induced extracellular signal-regulated kinases 1 and 2 (ERK1/2) and c-Jun N-terminal kinases (JNK) activation and inhibition of these signaling pathways suggested an involvement in the IL-1ß effects on osteoblast migration. In contrast, basal migration of MSC and their migratory activity toward PDGF-BB was found to be unaffected by IL-1ß. These results indicate that the presence of IL-1ß leads to impaired recruitment of osteoblasts which might influence early stages of fracture healing and could have pathological relevance for bone remodeling in inflammatory bone disease.


Subject(s)
Interleukin-1beta/pharmacology , Osteoblasts/drug effects , Becaplermin , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Humans , Insulin-Like Growth Factor I/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Mitogen-Activated Protein Kinases/metabolism , Osteoblasts/physiology , Proto-Oncogene Proteins c-sis/pharmacology , Receptors, Interleukin-1 Type I/metabolism , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
10.
J Mater Sci Mater Med ; 23(10): 2575-82, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22729594

ABSTRACT

The purpose of this study was to prove the effect of cyclic uniaxial intermittent strain on the mRNA expression of ligament-specific marker genes in human mesenchymal stem cells (MSC) and anterior cruciate ligament-derived fibroblasts (ACL-fibroblasts) seeded onto a novel textured poly(L-lactide) scaffold (PLA scaffold). Cell-seeded scaffolds were mechanically stimulated by cyclic uniaxial stretching. The expression of ligament matrix gene markers: collagen types I and III, fibronectin, tenascin C and decorin, as well as the proteolytic enzymes matrix metalloproteinase MMP-1 and MMP-2 and their tissue specific inhibitors TIMP-1 and TIMP-2 was investigated by analysing the mRNA expression using reverse transcriptase polymerase chain reaction and related to the static control. In ACL-fibroblasts seeded on PLA, mechanical load induced up-regulation of collagen types I and III, fibronectin and tenascin C. No effect of mechanical stimulation on the expression of ligament marker genes was found in undifferentiated MSC seeded on PLA. The results indicated that the new textured PLA scaffold could transfer the mechanical load to the ACL-fibroblasts and improved their ligament phenotype. This scaffold might be suitable as a cell-carrying component of ACL prostheses.


Subject(s)
Ligaments/cytology , Mesenchymal Stem Cells/cytology , Polyesters/chemistry , Stress, Physiological , Tissue Engineering , Tissue Scaffolds , Base Sequence , DNA Primers , Extracellular Matrix Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/enzymology , Fibroblasts/metabolism , Gene Expression , Humans , Ligaments/enzymology , Ligaments/metabolism , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism
11.
Int J Artif Organs ; 34(9): 882-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22094570

ABSTRACT

Implant infection still represents a major clinical problem in orthopedic surgery. We therefore tested the in vitro biocompatibility and antibacterial effects of copper (Cu)- and silver (Ag)-ion implantation. Discs of a commonly used titanium alloy (Ti6AlV4) with an aluminium oxide-blasted surface were treated by Cu- or Ag-ion implantation with different dosage regimen (ranging from 1e15-17 ions cm(-2) at energies of 2-20 keV). The samples were seeded with primary human osteoblasts and cell attachment and proliferation was analyzed by an MTT-assay. In comparison to the reference titanium alloy there was no difference in the number of attached viable cells after two days. After seven days the number of viable cells was increased for Cu with 1e17 ions cm(-2) at 2 and 5 keV, and for Ag with 1e16 ions cm(-2) at 5 keV while it was reduced for the highest amount of Ag deposition (1e17 ions cm(-2) at 20 keV). Antibacterial effects on S.aureus and E.coli were marginal for the studied dosages of Cu but clearly present for Ag with 1e16 ions cm(-2) at 2 and 5 keV and 1e17 ions cm(-2) at 20 keV. These results indicate that Ag-ion implantation may be a promising methodological approach for antibacterial functionalization of titanium implants.


Subject(s)
Aluminum Oxide/chemistry , Anti-Bacterial Agents/pharmacology , Cell Proliferation/drug effects , Coated Materials, Biocompatible , Copper/pharmacology , Joint Prosthesis , Osteoblasts/drug effects , Prosthesis-Related Infections/prevention & control , Silver/pharmacology , Titanium/chemistry , Alloys , Cell Adhesion/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Escherichia coli/drug effects , Escherichia coli/growth & development , Humans , Joint Prosthesis/adverse effects , Prosthesis Design , Prosthesis-Related Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Surface Properties , Time Factors
12.
Int J Mol Med ; 27(1): 139-45, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21069259

ABSTRACT

We have previously reported that star shaped poly(ethylene oxide-stat-propylene oxide) macromers with 80% EO content and isocyanate functional groups at the distal ends [NCO-sP(EO-stat-PO)] can be used to generate coatings that are non-adhesive but easily functionalized for specific cell adhesion. In the present study, we investigated whether the NCO-sP(EO-stat-PO) surfaces maintain peptide configuration-specific cell-surface interactions or if differences between dissimilar binding molecules are concealed by the coating. To this end, we have covalently immobilized both linear-RGD peptides (gRGDsc) and cyclic-RGD (RGDfK) peptides in such coatings. Subsequently, SaOS-2 or human multipotent mesenchymal stromal cells (MSC) were seeded on these substrates. Cell adhesion, spreading and survival was observed for up to 30 days. The time span for cell adherence was not different on linear and cyclic RGD peptides, but was shorter in comparison to the unmodified glass surface. MSC proliferation on cyclic RGDfK modified coatings was 4 times higher than on films functionalized by linear gRGDsc sequences, underlining that the NCO-sP(EO-stat-PO) film preserves the configuration-specific biochemical peptide properties. Under basal conditions, MSC expressed osteogenic marker genes after 14 days on cyclic RGD peptides, but not on linear RGD peptides or the unmodified glass surfaces. Our results indicate specific effects of these adhesion peptides on MSC biology and show that this coating system is useful for selective testing of cellular interactions with adhesive ligands.


Subject(s)
Antineoplastic Agents/metabolism , Coated Materials, Biocompatible/chemistry , Oligopeptides/metabolism , Polyethylenes/chemistry , Polypropylenes/chemistry , Adult , Antineoplastic Agents/chemistry , Biomarkers/metabolism , Cell Adhesion/physiology , Cells, Cultured , Humans , Mesoderm/cytology , Oligopeptides/chemistry , Osteogenesis/physiology , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Stromal Cells/cytology , Stromal Cells/physiology , Surface Properties , Young Adult
13.
Int J Artif Organs ; 33(9): 646-53, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20890881

ABSTRACT

Implant surfaces should ideally be designed to promote the attachment of target tissue cells; at the same time, they should prevent bacterial adhesion, achievable through modification strategies comprising three lines of defense. As the first criterion, selective adhesion can be realized by means of non-adhesive coatings that can be functionalized with small peptides, thereby supporting osteogenic cell attachment for implants in bone contact but not bacterial adhesion. The second line of defense, defined by bacterial survival, quorum sensing and biofilm formation, can be addressed by various antimicrobial substances that can be leaching or non-leaching. The possibility of a third line of defense, the disruption of an established biofilm, is just emerging. Since microorganisms are quite ''ingenious'' at finding ways to overcome a certain line of defense, the most promising solution might be a combination of all these antibacterial strategies. Coating systems that allow such different approaches to be combined are scarce. However, ultrathin multifunctional NCO-sP(EO-stat-PO)-based layers may represent a promising platform for such an integrated approach.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Biofilms/drug effects , Coated Materials, Biocompatible , Prosthesis-Related Infections/prevention & control , Adhesins, Bacterial , Humans , Microbial Viability , Prosthesis Design , Prosthesis-Related Infections/microbiology , Quorum Sensing , Surface Properties
14.
Int J Cancer ; 127(6): 1295-307, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20104521

ABSTRACT

The Ewing family of tumors (EFT) is an important group of pediatric malignancies with a guarded prognosis. Little is known about the heterogeneity of EFT cells, and the cellular origin of EFT is disputed. We now add evidence that EFT are heterogeneous by showing that EFT cells from spheres growing in serum-free medium are markedly more tumorigenic than adherently growing EFT cells. Furthermore, EFT cells strongly expressing CD57 (HNK-1), a surface marker for migrating and proliferating neural crest cells, are more tumorigenic than cells with low expression of CD57, possibly mediated in part by enhanced adhesion and invasion. We contribute to the controversy about the cellular origin of EFT by clonal analysis, showing that EFT cells can differentiate similar to neural crest cells. These data increase our knowledge about the pathogenesis and heterogeneity of EFT.


Subject(s)
CD57 Antigens/physiology , Cell Differentiation , Neural Crest/cytology , Sarcoma, Ewing/pathology , Animals , Culture Media, Serum-Free , Flow Cytometry , Humans , Mice , Mice, Knockout , Sarcoma, Ewing/immunology , Tumor Cells, Cultured
15.
Int J Artif Organs ; 32(9): 655-62, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19882552

ABSTRACT

Poor osseointegration and bacterial infection are major causes of orthopedic implant failure. Both problems arise from passive unspecific protein coating that may not optimally support adhesion of osteoblastic cells and which enable bacterial adhesion that subsequently results in biofilm formation. This review addresses emerging concepts of preventing unspecific protein adsorption and biofilm formation by organic coating systems. We especially focus on recent concepts that additionally allow functionalization for preferential cell adhesion using cell adhesion mediating small peptide sequences that do not induce bacterial adherence. One promising approach that is presented and discussed within this context is the use of NCO-sP(EO-stat-PO).


Subject(s)
Bacteria/pathogenicity , Bacterial Adhesion , Biofilms , Coated Materials, Biocompatible , Orthopedic Procedures/instrumentation , Prosthesis-Related Infections/prevention & control , Bacteria/growth & development , Humans , Orthopedic Procedures/adverse effects , Prosthesis Design , Prosthesis-Related Infections/microbiology , Surface Properties , Virulence
16.
Bone ; 45(2): 367-76, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19414075

ABSTRACT

Mesenchymal stem cells are multipotent cells able to differentiate into different mesenchymal lineages. Studies in the past had suggested that two of these mesenchymal differentiation directions, the chondrogenic and the myogenic differentiation, are negatively regulated by the transcription factor NF-kappaB. Although osteogenic differentiation has been extensively studied, the influence of NF-kappaB on this differentiation lineage was not subject of detailed analyses in the past. We have analyzed the consequences of TNF-alpha treatment and genetic manipulation of the NF-kappaB pathway for osteogenic differentiation of hMSCs. Treatment of hMSCs during differentiation with TNF-alpha activates NF-kappaB and this results in enhanced expression of osteogenetic proteins like bone morphogenetic protein2 (BMP-2) and alkaline phosphatase (ALP). In addition, enhanced matrix mineralization was observed. The direct contribution of the NF-kappaB pathway was confirmed in cells that express a constitutively active version of the NF-kappaB-inducing kinase IKK2 (CA-IKK2). The IKK2/NF-kappaB-induced BMP-2 up-regulation results in the enhancement of RUNX2 and Osterix expression, two critical regulators of the osteogenic differentiation program. Interestingly, a genetic block of the NF-kappaB pathway did not interfere with osteogenic differentiation. We conclude that TNFalpha mediated NF-kappaB activation, although not absolutely required for BMP-2 expression and matrix mineralization nevertheless supports osteogenic differentiation and matrix mineralization by increasing BMP-2 expression. Our results therefore suggest that NF-kappaB activation may function in lineage selection during differentiation of hMSCs by fostering osteogenic differentiation at the expense of other differentiation lineages.


Subject(s)
Cell Differentiation/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , NF-kappa B/metabolism , Osteogenesis/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Alkaline Phosphatase/metabolism , Alleles , Bone Morphogenetic Protein 2/metabolism , Calcification, Physiologic/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , I-kappa B Kinase/metabolism , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/virology , Retroviridae
17.
Biotechnol Appl Biochem ; 49(Pt 3): 185-94, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17640172

ABSTRACT

The creation of tissue-engineered constructs with autologous cells is a central goal in regenerative medicine. With respect to ligament replacement, we have evaluated the influences of matrix and growth factors on hMSCs (human mesenchymal stromal cells). hMSCs were seeded in two different 3D (three-dimensional) systems consisting of either a collagen type I gel or a synthetic PLA [poly-(L-lactic acid)] scaffold. After cultivation for 14 days with rhTGFbeta1 (recombinant human transforming growth factor beta1), rhPDGF-BB (recombinant human platelet-derived growth factor homodimer of B-chain) or rhBMP13 (recombinant human bone morphogenetic protein 13), we assessed the proliferation potential, mRNA expression and protein expression of various matrix-interacting and matrix-degrading molecules by quantitative real-time RT (reverse transcriptase)-PCR, immunohistochemistry and gelatin zymography in comparison with unstimulated cells. Cellular reactions to the type of scaffold or soluble factors could be found in the expression of tenascin-C as well as integrin subunits alpha1, alpha3 and beta1. Collagen type X expression was induced by 3D culture and stimulated by rhTGFbeta1 on PLA. The expression of MMP-1 (matrix metalloproteinase 1) tended to increase, and MMP-13 was induced in the collagen culture system. The activation of MMP-2 was stimulated by the cultivation of MSCs within the collagenous matrix. These results demonstrated that various interactive effects of growth factors and scaffolds influence the cell-biological behaviour of MSCs. It is important to take these complex interactions, which partly differ from differentiated cells, into account in further tissue-engineering approaches.


Subject(s)
Mesenchymal Stem Cells/drug effects , Multipotent Stem Cells/drug effects , Platelet-Derived Growth Factor/pharmacology , Tissue Scaffolds , Transforming Growth Factor beta1/pharmacology , Adult , Becaplermin , Bone Morphogenetic Proteins/pharmacology , Cell Proliferation , Collagen/genetics , Collagen Type I , Female , Gels , Growth Differentiation Factor 6 , Humans , Integrins/genetics , Lactic Acid , Male , Matrix Metalloproteinases, Secreted/genetics , Matrix Metalloproteinases, Secreted/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Polyesters , Polymers , Proto-Oncogene Proteins c-sis , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Tenascin/genetics , Tissue Engineering
18.
BMC Cell Biol ; 8: 45, 2007 Oct 31.
Article in English | MEDLINE | ID: mdl-17973995

ABSTRACT

BACKGROUND: CCN-proteins are known to be involved in development, homeostasis and repair of mesenchymal tissues. Since these processes implicate recruitment of cells with the potential to be committed to various phenotypes, we studied the effect of CYR61/CCN1 and WISP3/CCN6 on migration of human bone marrow derived mesenchymal stroma cells (MSCs) in comparison to in vitro osteogenic differentiated MSCs using a modified Boyden chamber assay. RESULTS: CYR61 and WISP3 were purified as fusion proteins with a C-terminal Fc-tag from baculovirus infected SF21 cells using protein G sepharose columns. CYR61 and WISP3 stimulated cell migration of undifferentiated MSCs in a dose-dependent manner. CYR61 and WISP3 had similar effects on committed osteogenic precursor cells. Checkerboard analysis revealed that CYR61 and WISP3 stimulated true directed cell migration (chemotaxis) of MSCs and committed osteogenic precursors. In MSCs the chemotactic activity of WISP3 but not CYR61 was mediated through integrin alphanuss5. CONCLUSION: Our results indicate that CYR61 and WISP3 can function as soluble ligands transmitting chemotactic signals to human MSCs but differ in the involvement of integrin alphanuss5. This may be relevant for their possible role in connective tissue repair.


Subject(s)
Chemotaxis , Immediate-Early Proteins/metabolism , Insulin-Like Growth Factor Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mesenchymal Stem Cells/physiology , Adult , CCN Intercellular Signaling Proteins , Chemotactic Factors/pharmacology , Chemotaxis/drug effects , Cysteine-Rich Protein 61 , Dose-Response Relationship, Drug , Humans , Immediate-Early Proteins/pharmacology , In Vitro Techniques , Insulin-Like Growth Factor Binding Proteins/pharmacology , Integrin alphaVbeta3/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteoblasts/drug effects , Osteoblasts/physiology , Signal Transduction
19.
J Biomed Mater Res B Appl Biomater ; 81(1): 82-90, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16924614

ABSTRACT

Biocompatibility and cell seeding capability of a new cell scaffold made of textured polylactic acid (PLA) fibers was investigated as a new material for tissue engineering of anterior cruciate ligaments (ACL). Adhesion and proliferation of human mesenchymal progenitor cells (MPC) was investigated after 15 days by scanning electron microscopy and standard histology. Expression of collagen type I and III, fibronectin, tenascin C, decorin, smooth muscle actin, and the matrix metalloproteinases MMP-1 and MMP-2, as well as their tissue inhibitors TIMP-1 and TIMP-2 was analyzed using real-time PCR. Protein expression of collagen I and III, tenascin C, and proliferating nuclear antigen (PCNA) was determined by immunohistology. Apoptosis was analyzed by detection of p53 expression and TUNEL staining. MPC seeded the scaffold homogeneously and showed good cell growth and no increased rate of apoptosis. After 15 days, the matrix forming genes collagen type I, tenascin C, and decorin were upregulated, indicating the formation of a ligament-like matrix. MMP-1 and TIMP-1 were also significantly increased, suggesting initial matrix remodeling. It was concluded that the new porous PLA scaffold allowed homogeneous cell seeding, a fibroblastic phenotype and the production of a ligament-like matrix and, therefore, might be a suitable cell carrier for ACL tissue engineering.


Subject(s)
Biocompatible Materials/pharmacology , Ligaments , Mesoderm/drug effects , Polyesters/chemistry , Tissue Engineering , Apoptosis , Biocompatible Materials/chemistry , Cell Adhesion , Cell Differentiation , Cell Proliferation , Extracellular Matrix Proteins/analysis , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression , Humans , Materials Testing , Matrix Metalloproteinase 1/analysis , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Microscopy, Electron, Scanning , Protein Biosynthesis , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/chemistry , Stem Cells/drug effects , Stem Cells/ultrastructure , Tissue Inhibitor of Metalloproteinase-1/analysis , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/analysis , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism
20.
Cells Tissues Organs ; 182(3-4): 143-54, 2006.
Article in English | MEDLINE | ID: mdl-16914917

ABSTRACT

BACKGROUND/AIMS: The aim of our study was to investigate interactions of mesenchymal progenitor cells (MPCs) with collagen matrices. METHODS: Human bone-marrow-derived MPCs were cultivated in collagen type I gels with and without inhibition of beta(1)-integrin by a specific antibody. Collagen gel contraction, cell morphology, expression of integrin subunits and several genes related to matrix synthesis and turnover as well as MPC differentiation were analyzed over 14 days. RESULTS: Human MPCs markedly contracted free-floating collagen gels. Contraction was nearly completely inhibited by blocking beta(1)-integrin. Cellular morphology was elongated in the absence and mostly round in the presence of the antibody. Expression of integrin alpha(1), alpha(2) and beta(1) subunits showed several changes partly dependent on beta(1)-integrin blocking. Expression of matrix metalloproteinase-1 was elevated irrespective of beta(1)-integrin blocking and tenascin-C was subsequently induced during gel contraction. Spontaneous induction of chondrogenic, osteogenic or adipogenic differentiation was observed neither in the presence nor in the absence of the beta(1)-integrin antibody. CONCLUSION: Our results indicate that the interaction of human MPCs with fibrillar collagen type I involves beta(1)- and alpha-integrin subunits and induces changes in gene expression related to extracellular matrix synthesis and turnover but not differentiation to the chondrogenic, osteogenic or adipogenic phenotype.


Subject(s)
Collagen Type I/metabolism , Integrin alpha Chains/metabolism , Integrin beta Chains/metabolism , Mesenchymal Stem Cells/cytology , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/genetics , Extracellular Matrix/metabolism , Humans , Immunohistochemistry , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Integrin alpha1/genetics , Integrin alpha1/metabolism , Integrin alpha2/genetics , Integrin alpha2/metabolism , Integrin alpha3/genetics , Integrin alpha3/metabolism , Integrin beta Chains/genetics , Integrin beta Chains/immunology , Integrin beta1/genetics , Integrin beta1/immunology , Integrin beta1/metabolism , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Mesenchymal Stem Cells/metabolism , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , Tenascin/genetics , Tenascin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...