Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Biomater Appl ; 32(2): 139-149, 2017 08.
Article in English | MEDLINE | ID: mdl-28599578

ABSTRACT

Current strategies in implant technology are directed to generate bioactive implants that are capable to activate the regenerative potential of the surrounding tissue. On the other hand, implant-related infections are a common problem in orthopaedic trauma patients. To meet both challenges, i.e. to generate a bone implant with regenerative and antimicrobial characteristics, we tested the use of copper coated nails for surgical fixation in a rabbit model. Copper acetate was galvanically deposited with a copper load of 1 µg/mm2 onto a porous oxide layer of Ti6Al4V nails, which were used for the fixation of a tibia fracture, inoculated with bacteria. After implantation of the nail the concentration of copper ions did not increase in blood which indicates that copper released from the implant was locally restricted to the fracture site. After four weeks, analyses of the extracted implants revealed a distinct antimicrobial effect of copper, because copper completely prevented both a weak adhesion and firm attachment of biofilm-forming bacteria on the titanium implant. To evaluate fracture healing, radiographic examination demonstrated an increased callus index in animals with copper coated nails. This result indicates a stimulated bone formation by releasing copper ions. We conclude that the use of implants with a defined load of copper ions enables both prevention of bacterial infection and the stimulation of regenerative processes.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bone Nails , Coated Materials, Biocompatible/therapeutic use , Copper/therapeutic use , Osteogenesis/drug effects , Tibial Fractures/surgery , Titanium/therapeutic use , Alloys , Animals , Anti-Bacterial Agents/chemistry , Bone Nails/microbiology , Coated Materials, Biocompatible/chemistry , Copper/chemistry , Female , Fracture Healing/drug effects , Rabbits , Staphylococcal Infections/complications , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Tibial Fractures/complications , Tibial Fractures/drug therapy , Tibial Fractures/microbiology , Titanium/chemistry
2.
Biomaterials ; 44: 36-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25617124

ABSTRACT

The supply of titanium implants which are widely used in orthopaedics with both regenerative and anti-microbial properties will achieve a great progress in bone regeneration. We asked, whether by appropriate concentrations of copper ions it will be possible both to inhibit growth of bacteria and stimulate biological responses in mesenchymal stem cells (MSC). Using titanium material which released galvanically deposited copper at concentrations from 0.3 to 1.75 mM, growth of planktonic Staphylococcus aureus was blocked and more importantly adherent bacteria were cleared from the material surface within 24 h. To test biological responses of human bone marrow derived MSC due to copper ions, we found that copper stimulated the proliferation of MSC in a narrow concentration range around 0.1 mM. Similar copper concentrations enhanced osteogenic differentiation of MSC when cells were cultured in osteogenic differentiation medium. We observed increased activity of alkaline phosphatase (ALP), higher expression of collagen I, osteoprotegerin, osteopontin and finally mineralization of the cells. We conclude that titanium implants that release copper ions can be effective against bacterial infections at higher concentrations of copper near the implant surface and can promote bone regeneration when its concentration becomes lower due to diffusion.


Subject(s)
Copper/pharmacology , Prostheses and Implants , Prosthesis Design , Regenerative Medicine , Anti-Infective Agents/pharmacology , Biomarkers/metabolism , Calcium/metabolism , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Osteogenesis/drug effects , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Surface Properties , Titanium/pharmacology
3.
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
4.
Int J Biochem Cell Biol ; 54: 149-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25066315

ABSTRACT

The pro-inflammatory cytokine tumor necrosis factor (TNF) is well known to induce differentiation of bone matrix-resorbing osteoclasts from hematopoietic stem cells. However, the impact of TNF on differentiation of bone matrix-forming osteoblasts from mesenchymal stem cells (MSC) was only fragmentarily studied so far. Therefore, we investigated what impact long-term TNF treatment has on osteoblastic differentiation of MSC isolated from the adipose tissue (ASC) in vitro. In summary, we found continuous TNF exposure to induce the nuclear factor of kappa B pathway in ASC as well as secretion of the pro-inflammatory chemokine interleukin 8, but not the mitogen-activated protein kinase and the apoptosis pathway in ASC. Moreover, TNF neither induced nor inhibited osteoblastic differentiation of ASC, but strongly increased their proliferation rate. In that manner, pro-inflammatory conditions in vivo may generate significantly increased numbers of progenitor cells, and ASC especially, in conjunction with external stimuli, may contribute to the events of ectopic ossification observed in chronic inflammatory diseases. The substantiation of the translation of our in vitro findings to the disease context encourages further in vivo studies.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation , Mesenchymal Stem Cells/cytology , NF-kappa B/metabolism , Osteoblasts/cytology , Tumor Necrosis Factor-alpha/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Apoptosis/drug effects , Blotting, Western , Cells, Cultured , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , In Vitro Techniques , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mitogen-Activated Protein Kinases/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/drug effects
5.
J Tissue Eng Regen Med ; 8(10): 831-40, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23038605

ABSTRACT

The interaction of mesenchymal stem cells (MSCs) with endothelium in vivo is significant for regenerative processes in organisms. To design concepts for tissue engineering for bone regeneration based on this interaction, the osteogenic differentiation of human bone marrow-derived MSCs in a co-culture with human dermal microvascular endothelial cells (HDMECs) was studied. The experiments were focussed on the regulation of MSCs in a co-culture with HDMECs on different calcium phosphate scaffolds. Alkaline phosphatase (ALP) activity and mRNA expression of various osteogenic markers increased significantly when cells were co-cultured on materials with calcium phosphate scaffolds compared to tissue culture polystyrene or when MSCs were cultured alone. In addition, it was observed that the expression of osteopontin and osteocalcin was highly sensitive to the substrate for cell adhesion. Whereas these late osteogenic markers were down-regulated in co-cultures on polystyrene, they were up-regulated on calcium phosphate and moreover, were differentially expressed on the three calcium phosphate scaffolds tested. To enhance the osteogenic differentiation of MSCs in a co-culture, direct cell-cell interactions were required. Concerning molecular mechanisms in the interactions between both cell types, it was found that connexin 43 was expressed in contact sites and more apparently, endothelial cells grew over the MSCs, which facilitated direct cellular interactions mediated by various adhesion receptors. This study revealed significant findings for the design of implant materials suitable for regeneration of bone by stimulating the functional interaction of MSCs with endothelial cells.


Subject(s)
Calcium Phosphates/pharmacology , Cell Communication/drug effects , Cell Differentiation/drug effects , Endothelial Cells , Mesenchymal Stem Cells , Osteogenesis/drug effects , Tissue Scaffolds , Antigens, Differentiation/metabolism , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism
6.
Materials (Basel) ; 7(2): 1342-1359, 2014 Feb 19.
Article in English | MEDLINE | ID: mdl-28788517

ABSTRACT

Due to the weak regeneration potential of cartilage, there is a high clinical incidence of articular joint disease, leading to a strong demand for cartilaginous tissue surrogates. The aim of this study was to evaluate a gelatin-based hydrogel for its suitability to support chondrogenic differentiation of human mesenchymal stem cells. Gelatin-based hydrogels are biodegradable, show high biocompatibility, and offer possibilities to introduce functional groups and/or ligands. In order to prove their chondrogenesis-supporting potential, a hydrogel film was developed and compared with standard cell culture polystyrene regarding the differentiation behavior of human mesenchymal stem cells. Cellular basis for this study were human adipose tissue-derived mesenchymal stem cells, which exhibit differentiation potential along the adipogenic, osteogenic and chondrogenic lineage. The results obtained show a promotive effect of gelatin-based hydrogels on chondrogenic differentiation of mesenchymal stem cells in vitro and therefore encourage subsequent in vivo studies.

7.
Exp Cell Res ; 319(18): 2856-65, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24055981

ABSTRACT

Cartilaginous matrix-degenerative diseases like osteoarthritis (OA) are characterized by gradual cartilage erosion, and also by increased presence of cells with mesenchymal stem cell (MSC) character within the affected tissues. Moreover, primary chondrocytes long since are known to de-differentiate in vitro and to be chondrogenically re-differentiable. Since both findings appear to conflict with each other, we quantitatively assessed the mesenchymal differentiation potential of OA patient cartilage-derived cells (CDC) towards the osteogenic and adipogenic lineage in vitro and compared it to that of MSC isolated from adipose tissue (adMSC) of healthy donors. We analyzed expression of MSC markers CD29, CD44, CD105, and CD166, and, following osteogenic and adipogenic induction in vitro, quantified their expression of osteogenic and adipogenic differentiation markers. Furthermore, CDC phenotype and proliferation were monitored. We found that CDC exhibit an MSC CD marker expression pattern similar to adMSC and a similar increase in proliferation rate during osteogenic differentiation. In contrast, the marked reduction of proliferation observed during adipogenic differentiation of adMSC was absent in CDC. Quantification of differentiation markers revealed a strong osteogenic differentiation potential for CDC, however almost no capacity for adipogenic differentiation. Since in the pathogenesis of OA, cartilage degeneration coincides with high bone turnover rates, the high osteogenic differentiation potential of OA patient-derived CDC may affect clinical therapeutic regimens aiming at autologous cartilage regeneration in these patients.


Subject(s)
Cartilage, Articular/cytology , Cell Differentiation , Chondrocytes/cytology , Mesenchymal Stem Cells/cytology , Osteogenesis , Antigens, CD/genetics , Biomarkers/metabolism , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Humans , Microscopy, Fluorescence
8.
PLoS One ; 8(7): e71283, 2013.
Article in English | MEDLINE | ID: mdl-23923061

ABSTRACT

Mechanical interactions of mesenchymal stem cells (MSC) with the environment play a significant role in controlling the diverse biological functions of these cells. Mechanical forces are transduced by integrins to the actin cytoskeleton that functions as a scaffold to switch mechanical signals into biochemical pathways. To explore the significance of cytoskeletal mechanisms in human MSC we modulated the actin cytoskeleton using the depolymerising drugs cytochalasin D (CytD) and latrunculin A (LatA), as well as the stabilizing drug jasplakinolide (Jasp) and examined the activation of the signalling molecules ERK and AKT during mechanical loading. All three drugs provoked significant changes in cell morphology and organisation of the cytoskeleton. Application of mechanical forces to ß1-integrin receptors using magnetic beads without deformation of the cell shape induced a phosphorylation of ERK and AKT. Of the two drugs that inhibited the cytoskeletal polymerization, LatA completely blocked the activation of ERK and AKT due to mechanical forces, whereas CytD inhibited the activation of AKT but not of ERK. Activation of both signalling molecules by integrin loading was not affected due to cell treatment with the cytoskeleton stabilizing drug Jasp. To correlate the effects of the drugs on mechanically induced activation of AKT and ERK with parameters of MSC differentiation, we studied ALP activity as a marker for osteogenic differentiation and examined the uptake of fat droplets as marker for adipogenic differentiation in the presence of the drugs. All three drugs inhibited ALP activity of MSC in osteogenic differentiation medium. Adipogenic differentiation was enhanced by CytD and Jasp, but not by LatA. The results indicate that modulation of the cytoskeleton using perturbing drugs can differentially modify both mechanically induced signal transduction and MSC differentiation. In addition to activation of the signalling molecules ERK and AKT, other cytoskeletal mechanisms are involved in MSC differentiation.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Differentiation , Mechanotransduction, Cellular , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Adipogenesis , Cell Differentiation/drug effects , Cytochalasin D/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Proto-Oncogene Proteins c-akt/metabolism
9.
J Appl Biomater Funct Mater ; 11(1): e1-8, 2013 Jun 24.
Article in English | MEDLINE | ID: mdl-22798245

ABSTRACT

AIM: Design optimization and surface modifications of orthopedic implants are focused on adhesive properties depending on specific applications. To obtain an in-vitro understanding of the adhesion interaction of bone cells on implant surfaces the time-dependent adhesion behavior of osteoblastic cells was studied. MATERIALS AND METHODS: MG-63 osteoblastic cells were seeded on discs of polished titanium alloy (Ti6Al4V) and allowed to adhere for various time periods (1 to 48 h). Using a spinning disc device and a confocal laser scanning microscope (LSM) the shear stress required to detach the bone cells from the substrate was determined. An approximation of the adhesion force was calculated from measurements of cell height and contact radius. RESULTS: Shear stress ranged from 40.4 N/m2 to 82.4 N/m2 showing an increase in cell adhesion reaching a maximum after 6 h before decreasing significantly. Using the cell height and contact radii, measured for the various time periods, the lowest adhesion force of 232 nN was approximated after 1 h cell adhesion and analogous to the adhesion strength measurements, the highest of 664 nN after 6 h. Generally, cell adhesion decreased at incubation times longer than 6 h before an increase after 48 h was observed once again. CONCLUSIONS: Differences in adhesion behavior over time indicate dynamic cell-substrate interactions because of cell migration and proliferation processes. The study stresses the importance of calculating the adhesion force rather than shear stress to gain more expressive data regarding cell adhesion.


Subject(s)
Osteoblasts/physiology , Prostheses and Implants , Titanium , Alloys , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Adhesion/drug effects , Humans , Materials Testing , Microscopy, Electron, Scanning/instrumentation , Osteoblasts/drug effects , Osteoblasts/metabolism , Shear Strength/drug effects , Surface Properties , Time Factors , Titanium/chemistry , Titanium/pharmacology , Tumor Cells, Cultured
10.
Biointerphases ; 7(1-4): 51, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22893235

ABSTRACT

To successfully apply implant materials for regenerative processes in the body, understanding the mechanisms at the interface between cells or tissues and the artificial material is of critical importance. This topic is becoming increasing relevant for clinical applications. For the fourth time, around 200 scientists met in Rostock, Germany for the international symposium "Interface Biology of Implants". The aim of the symposium is to promote interdisciplinary dialogue between scientists from different disciplines. The symposium also emphasizes the need of this applied scientific field for permanent input from basic sciences.


Subject(s)
Prostheses and Implants , Surface Properties , Biomedical Research/trends , Germany , Humans
11.
Nephrol Dial Transplant ; 27(6): 2377-83, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22086973

ABSTRACT

BACKGROUND: Albumin is an important transport protein for non-water-soluble protein-bound drugs and uraemic toxins. Its transport capacity is reduced in patients with advanced chronic kidney disease (CKD) and unbound fractions of uraemic toxins are related to complications of CKD. We investigated whether this reduction could be quantified and how it correlated with the stages of CKD. Albumin-binding capacity (ABiC) is a dye-based method that quantifies the remaining binding capacity of one major binding site (site II) of the albumin molecule. METHODS: Blood samples from 104 CKD patients were incubated with a binding site-specific fluorescent marker and the amount of unbound marker was determined by means of fluorescence detection after filtration. Measurements in a pooled human plasma were used for reference. Glomerular filtration rate and serum indoxyl sulphate (IS) levels were also determined. RESULTS: Impairment of renal function was associated with a reduction in ABiC (mean ± SD: 118 ± 12; 111 ± 11; 99 ± 8 and 79 ± 9% for Stages 1/2, 3, 4 and 5, respectively; P < 0.001) and an increase in IS (3.9 ± 1.1; 6.2 ± 3.2; 16.3 ± 14.9 and 56.1 ± 28.6 µmol/L for Stages 1/2, 3, 4 and 5, respectively; P < 0.001). In dialysis patients, ABiC was lower in those with urine outputs <500 mL/day than in those with preserved urine output (73.7 ± 6.0 versus 83.8 ± 8.5%; P < 0.001). CONCLUSION: Impaired albumin function in CKD patients can be quantified, is related to severity of kidney disease and is associated with an accumulation of uraemic albumin-bound retention solutes.


Subject(s)
Fluorescent Dyes/metabolism , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/pathology , Serum Albumin/metabolism , Toxins, Biological/metabolism , Uremia/metabolism , Aged , Cross-Sectional Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis
12.
J Cell Biochem ; 111(6): 1586-97, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21053275

ABSTRACT

The control of mesenchymal stem cells (MSC) by physical cues is of great interest in regenerative medicine. Because integrin receptors function as mechanotransducers, we applied drag forces to ß1 integrins on the apical surface of adherent human MSC. In addition to mechanical forces, the technique we used involved also the exposure of the cells to an inhomogeneous magnetic field. In order to assess the influence of the substrate on cell adhesion, cells were cultured on plain tissue culture polystyrene (TCP) or on coated well plates, which allowed only adhesion to embedded fibronectin or RGD peptides. We found that the expression of collagen I, which is involved in osteogenesis, and VEGF, a factor which stimulates angiogenesis, increased as a result of short-term mechanical integrin stress. Whereas, collagen I expression was stimulated by mechanical forces when the cells were cultured on fibronectin and RGD peptides but not on TCP, VEGF expression was enhanced by physical stimulation on TCP. The study further revealed that magnetic forces enhanced Sox 9 expression, a marker of chondrogenesis, and reduced the expression of ALP. Concerning the intracellular mechanisms involved, we found that the expression of VEGF induced by physical forces depended on Akt activation. Together, the results implicate that biological functions of MSC can be stimulated by integrin-mediated mechanical forces and a magnetic field. However, the responses of cells depend strongly on the substrate to which they adhere and on the cross-talk between integrin-mediated signals and soluble factors.


Subject(s)
Integrins/metabolism , Magnetics , Mesenchymal Stem Cells/metabolism , Stress, Mechanical , Aged , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Blotting, Western , Cell Adhesion/drug effects , Cells, Cultured , Chromones/pharmacology , Female , Flow Cytometry , Humans , Integrins/immunology , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/ultrastructure , Microscopy, Electron, Scanning , Middle Aged , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Reverse Transcriptase Polymerase Chain Reaction
13.
Cell Physiol Biochem ; 24(5-6): 483-92, 2009.
Article in English | MEDLINE | ID: mdl-19910688

ABSTRACT

BACKGROUND: In this study the influence of cultivation and proliferation on energy metabolic characteristics of human umbilical vein endothelial cells (HUVEC) has been examined. The energy metabolic capacities of human endothelial cells freshly isolated from the umbilical vein were compared with those after cultivation for three passages and as subconfluent and confluent cultures. METHODS: Expression of cell type-specific differentiation markers and proliferative activity were studied in dependency on cultivation characteristics. Furthermore, the energy metabolic characteristics of HUVEC were analyzed by measurement of the maximum catalytic activities of marker enzymes of various metabolic pathways. RESULTS: Examination of a typical marker of proliferation, Ki67, confirmed that HUVEC changed in culture from a non-proliferative to a proliferative state. Compared to other cell types, the enzyme pattern of HUVEC showed a high glycolytic and a high NADPH regenerating capacity. These capacities increased by cultivation nearly to the same degree as marker enzymes of other metabolic pathways (e.g. citric acid cycle). CONCLUSION: Our data support the theory that metabolism of EC is primarily by "aerobic glycolysis", i.e. the conversion of glucose to lactate in the presence of oxygen. These characteristics were independent of whether the cells are freshly isolated/non-proliferating or cell culture-adapted/proliferating.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Energy Metabolism/physiology , Glycolysis/physiology , Cell Proliferation , Cells, Cultured , Citric Acid Cycle , Endothelial Cells/cytology , Endothelial Cells/enzymology , Endothelium, Vascular/cytology , Glucose/metabolism , Humans , Ki-67 Antigen/metabolism , Lactic Acid/metabolism , NADP/metabolism , Oxidoreductases/metabolism
14.
Cell Adh Migr ; 3(4): 390-4, 2009.
Article in English | MEDLINE | ID: mdl-19690468

ABSTRACT

Implants are widely used in various clinical disciplines to replace or stabilize organs. The challenge for the future is to apply implant materials to specifically control the biology of the surrounding tissue for repair and regeneration. This field of research is highly interdisciplinary and combines scientists from technical and life sciences disciplines. To successfully apply materials for regenerative processes in the body, the understanding of the mechanisms at the interface between cells or tissues and the artificial material is of critical importance. The research focuses on stem cells, design of material surfaces, and mechanisms of cell adhesion. For the third time around 200 scientists met in Rostock, Germany for the international symposium "Interface Biology of Implants." The aim of the symposium is to promote the interdisciplinary dialogue between the scientists from the different disciplines to develop smart implants for medical use. In addition, researchers from basic sciences, notably cell biology presented new findings concerning mechanisms of cell adhesion to stimulate research in the applied field of implant technology.


Subject(s)
Biocompatible Materials , Cell Physiological Phenomena/physiology , Prostheses and Implants , Regenerative Medicine , Animals , Biomechanical Phenomena , Cell Adhesion/physiology , Extracellular Matrix/physiology , Humans , Polymers/chemistry , Stem Cells/cytology , Stem Cells/physiology , Surface Properties , Tissue Scaffolds
15.
Acta Biomater ; 5(2): 776-84, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18951072

ABSTRACT

Calcium phosphate (CaP) preparations are established coatings for titanium-based medical implants used for bone reconstruction. However, biodegradation of the coating can result in microparticles that subsequently cause inflammatory reactions. The present study was therefore aimed at investigating the inflammatory response to two series of CaP-coated titanium plates: Ti-brushite (Ti-B) and Ti-hydroxyapatite (Ti-H) implants. Fifteen male LEW.1A rats received one plate of each series and a pellet (5 x 2 mm) of sol-gel derived silica/CaP (SCP implants) implanted into the back musculature. After 7, 14 and 28 days, five rats were killed and the implants were removed with the surrounding tissue. Quantitative immunohistochemistry was performed on frozen sections. Total monocytes/macrophages, tissue macrophages, T-cells, MHC-class-II-positive cells and proliferating cells were counted. For the Ti-B implants, the number of monocytes/macrophages remained constant while the other cell populations increased. In contrast, for the Ti-H implants the number of monocytes/macrophages decreased while the other cell populations remained constant. The SCP implants demonstrated degradation and scattering into smaller particles with an increase for all cell populations except the proliferating cells. Human mesenchymal stem cells demonstrated adherence and a flat morphology on Ti-B and Ti-H implants and no remarkable difference between both implants. Taken together, the in vivo data demonstrate that the short-term inflammatory response against a hydroxyapatite coating is lower in comparison to a brushite coating, and that the morphology of cells growing in vitro is similar on both layers.


Subject(s)
Calcium Phosphates/adverse effects , Inflammation/chemically induced , Titanium/adverse effects , Animals , Cell Adhesion , Humans , Immunohistochemistry , Inflammation/immunology , Inflammation/metabolism , Macrophages/immunology , Male , Mesenchymal Stem Cells/cytology , Monocytes/immunology , Prostheses and Implants , Rats , Rats, Inbred Lew , T-Lymphocytes/immunology
16.
J Cell Mol Med ; 12(1): 281-91, 2008.
Article in English | MEDLINE | ID: mdl-18366455

ABSTRACT

Although studies in vivo revealed promising results in bone regeneration after implantation of scaffolds together with osteogenic progenitor cells, basic questions remain how material surfaces control the biology of mesenchymal stem cells (MSC). We used human MSC derived from bone marrow and studied the osteogenic differentiation on calcium phosphate surfaces. In osteogenic differentiation medium MSC differentiated to osteoblasts on hydroxyapatite and BONITmatrix, a degradable xerogel composite, within 14 days. Cells revealed a higher alkaline phosphatase (ALP) activity and increased RNA expression of collagen I and osteocalcin using real-time RTPCR compared with cells on tissue culture plastic. To test whether material surface characteristics alone are able to stimulate osteogenic differentiation, MSC were cultured on the materials in expansion medium without soluble additives for osteogenic differentiation. Indeed, cells on calcium phosphate without osteogenic differentiation additives developed to osteoblasts as shown by increased ALP activity and expression of osteogenic genes, which was not the case on tissue culture plastic. Because we reasoned that the stimulating effect on osteogenesis by calcium phosphate surfaces depends on an altered cell-extracellular matrix interaction we studied the dynamic behaviour of focal adhesions using cells transfected with GFP labelled vinculin. On BONITmatrix, an increased mobility of focal adhesions was observed compared with cells on tissue culture plastic. In conclusion, calcium phosphate surfaces are able to drive MSC to osteoblasts in the absence of osteogenic differentiation supplements in the medium. An altered dynamic behaviour of focal adhesions on calcium phosphate surfaces might be involved in the molecular mechanisms which promote osteogenic differentiation.


Subject(s)
Calcium Phosphates/chemistry , Cell Differentiation , Durapatite/chemistry , Mesenchymal Stem Cells/cytology , Osteogenesis/physiology , Silicon Dioxide/chemistry , Alkaline Phosphatase/metabolism , Bone Marrow Cells/metabolism , Cell Adhesion/physiology , Cell Movement/physiology , Cells, Cultured , Green Fluorescent Proteins/metabolism , Humans , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/metabolism , Vinculin/metabolism
17.
Biomol Eng ; 24(5): 531-6, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17884722

ABSTRACT

Divalent cations like Mn(2+) are known to strongly influence the integrin affinity to ligands and - in consequence - cell adhesion to extracellular matrix proteins. Therefore, divalent cation supplementation of biomaterials could be a promising approach to improve the ingrowth and the integration of implants. We were interested, whether manganese ions affect cellular functions like spreading, proliferation as well as gene expression in human osteoblasts. MG-63 osteoblastic cells were cultured in DMEM with 10% FCS. MnCl(2) was added at a concentration range of 0.01-0.5mM for 24h and 48 h. Spreading (cell area in microm(2)) of PKH26-stained cells (cell membrane dye) was analyzed using confocal microscopy. Cell proliferation was measured by flow cytometry. Quantification of the phosphorylation status of signaling proteins was estimated using the Bio-Plex 200 system. Gene expression of osteogenic markers at the mRNA and protein level was analyzed by quantitative real time RT-PCR and Western blot, respectively. The results demonstrated that at higher concentrations of Mn(2+) cells revealed a spindle shaped morphology. Further analyses indicated a reduced spreading, proliferation as well as phosphorylation of signaling proteins due to the influence of Mn(2+) in a concentration-dependent manner. Although expression of bone sialo protein (BSP) at the mRNA level increased both after 24h and 48 h in the presence of manganese, no increased expression of BSP was detected at the protein level. The expression of alkaline phosphatase (ALP) and collagen 1 (Col 1) mRNA decreased at >0.1mM MnCl(2). We speculate that the effect of manganese cations on cell functions is strongly concentration-dependent and the release of manganese when incorporated in a biomaterial surface has to be thoroughly adjusted.


Subject(s)
Manganese/pharmacology , Osteoblasts/drug effects , Biomarkers, Tumor/genetics , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression Profiling , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/metabolism , Ions/chemistry , Ions/pharmacology , Manganese/chemistry , Osteoblasts/metabolism , Phosphorylation , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
18.
Biomol Eng ; 24(5): 447-54, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17825608

ABSTRACT

Adhesion and spreading of cells on biomaterials are integrin-mediated processes. But recent findings indicate a key role of the cell membrane associated matrix substance hyaluronan (HA) in interface interactions. Because HA is a negatively charged molecule we assume that a biomaterial surface with an opposed charge could boost the first contact of the cell to the surface. Polished cp titanium (R(a)=0.19 microm) was coated with an amino-group containing plasma polymer (Ti PPA). For this purpose, a microwave excited, pulsed, low-pressure plasma was used. Additionally, collagen was immobilized on Ti PPA with polyethylene glycol diacid (PEG-DA), catalyzed by carbodiimide (CDI). The physico-chemical surface analytical techniques like XPS, FT-IR, water contact angle and zeta-potential verified the retention of the allylamine precursor structure. Human osteoblasts were cultured in serum-free Dulbecco's modified Eagle medium (DMEM). Adhesion and cell cycle phases were calculated by flow cytometry. Spreading and actin cytoskeleton were visualized by confocal microscopy. Gene expression of osteogenic markers was detected by real-time RT-PCR. Ti PPA is significantly advantageous concerning initial adhesion and spreading during the first hours of the cell contact to the surface. The proliferation of osteoblasts is positively influenced. Gene expression of the differentiation marker bone sialoprotein was upregulated after 24h. Our results demonstrate that functionalization of titanium with positively charged amino-groups is sufficiently enough to significantly improve initial steps of the cellular contact to the material surface.


Subject(s)
Allylamine/chemistry , Osteoblasts/physiology , Polymers/chemistry , Titanium/chemistry , Actins/chemistry , Alkaline Phosphatase/genetics , Carbodiimides/chemistry , Catalysis , Cell Adhesion/physiology , Cell Cycle , Collagen Type I/chemistry , Collagen Type I/genetics , Collagen Type I/radiation effects , Cytoskeleton/chemistry , Flow Cytometry , Gene Expression Profiling , Humans , Microwaves , Polyethylene Glycols/chemistry , Polyethylene Glycols/radiation effects , Polymers/radiation effects , Procollagen/chemistry , Procollagen/genetics , Procollagen/radiation effects , Reverse Transcriptase Polymerase Chain Reaction , Surface Properties , Titanium/radiation effects , Tumor Cells, Cultured
19.
Biomaterials ; 28(27): 3965-76, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17582491

ABSTRACT

The survival and functioning of a bone biomaterial requires a rapid and stable vascularization after implantation. However, the mechanisms involved in the context of the complex healing microenvironment are poorly understood. To evaluate the vascularization potential of bone biomaterials, angiogenic stimuli were added to human dermal microvascular endothelial cells (HDMEC) growing on three-dimensional (3-D) bone biomaterials consisting of porous hydroxyapatite, porous calcium phosphate, porous nickel-titanium, successfully being used in humans, and also silk fibroin nets. HDMEC did not migrate to form microcapillary-like structures as they did on cell culture plastic. In cocultures of HDMEC and primary human osteoblast cells (HOS) or the human osteoblast-like cell line MG-63 on these biomaterials, a tissue-like self-assembly of cells occurred with time, with endothelial cells forming microcapillary-like structures containing a lumen and giving a strong PECAM-1 expression at cell interfaces. These microcapillary-like structures were intertwined between cell layers of osteoblasts and did not form when exogenous angiogenic stimuli were added to these cocultures. The life span of HDMEC was also significantly enhanced by coculture; with HDMEC being present for up to at least 42 days, compared to the monoculture where cells began to die rapidly after 1 week without passage. This coculture system may be applicable to a prevascularization strategy for biomaterials prior to implantation. Irrespective of this, the coculture model holds promise for studies to deepen our understanding of bone regeneration on 3-D substrates. Most importantly, these data raise important questions concerning the exact nature of pro-angiogenic drug- or gene-delivery systems to be incorporated into scaffolds. Our results underline the necessity to take into account the in situ production of growth factors by invading mesenchymal cells in the regenerative niche.


Subject(s)
Biocompatible Materials/chemistry , Bone Substitutes/chemistry , Capillaries/cytology , Capillaries/physiology , Endothelial Cells/cytology , Neovascularization, Physiologic/physiology , Osteoblasts/cytology , Cells, Cultured , Coculture Techniques , Endothelial Cells/physiology , Humans , Materials Testing , Osteoblasts/physiology , Osteogenesis/physiology , Porosity , Tissue Engineering
20.
Curr Pharm Des ; 12(29): 3799-806, 2006.
Article in English | MEDLINE | ID: mdl-17073678

ABSTRACT

The interaction of leukocytes with the vessel endothelium to facilitate the extravasation into the tissue represents a key process of the body's defense mechanisms. Excessive recruitment of leukocytes into the inflamed tissue in chronic diseases like autoimmune disorders could be prevented by interfering with the mechanisms of leukocyte extravasation. Significant progress in elucidating the molecular basis of the trafficking of leukocytes from the blood stream to the extravascular tissue has been achieved that enables new strategies for therapeutic approaches. The multistep process of leukocyte rolling, firm adhesion and transmigration through the endothelial wall is facilitated by a dynamic interplay of adhesion receptors on both leukocytes and endothelial cells as well as chemokines. In preclinical studies using various animal models, promising results have been received demonstrating that blocking of adhesion receptors of the selectin and integrin families improved the inflammation process in models of ulcerative colitis, autoimmune encephalomyelitis or contact hypersensitivity. In addition to the targeting of adhesion receptors by antibodies, small molecules that mimic epitopes of adhesion receptor ligands have been developed and successfully applied in animal models. Clinical studies revealed a limited response using antibodies to selectins or LFA-1 integrins compared with animal models. However, using humanized antibodies to the alpha4- integrin subunit significant efficacy has been demonstrated in autoimmune diseases like psoriasis, multiple sclerosis and inflammatory bowel disease.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Autoimmune Diseases/drug therapy , Endothelial Cells/metabolism , Leukocyte Rolling , Leukocytes/metabolism , Animals , Autoimmune Diseases/metabolism , Cell Adhesion/drug effects , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Humans , Leukocyte Rolling/drug effects , Leukocytes/drug effects , Randomized Controlled Trials as Topic
SELECTION OF CITATIONS
SEARCH DETAIL