Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Nanomedicine ; 10: 1131-43, 2015.
Article in English | MEDLINE | ID: mdl-25709437

ABSTRACT

PURPOSE: In the present study, silica nanoparticles (sNP) coupled with insulin-like growth factor 1 (IGF-1) were loaded on a collagen-based scaffold intended for cartilage repair, and the influence on the viability, proliferation, and differentiation potential of human primary articular chondrocytes was examined. METHODS: Human chondrocytes were isolated from the hyaline cartilage of patients (n=4, female, mean age: 73±5.1 years) undergoing primary total knee joint replacement. Cells were dedifferentiated and then cultivated on a bioresorbable collagen matrix supplemented with fluorescent sNP coupled with IGF-1 (sNP-IGF-1). After 3, 7, and 14 days of cultivation, cell viability and integrity into the collagen scaffold as well as metabolic cell activity and synthesis rate of matrix proteins (collagen type I and II) were analyzed. RESULTS: The number of vital cells increased over 14 days of cultivation, and the cells were able to infiltrate the collagen matrix (up to 120 µm by day 7). Chondrocytes cultured on the collagen scaffold supplemented with sNP-IGF-1 showed an increase in metabolic activity (5.98-fold), and reduced collagen type I (1.58-fold), but significantly increased collagen type II expression levels (1.53-fold; P=0.02) after 7 days of cultivation compared to 3 days. In contrast, chondrocytes grown in a monolayer on plastic supplemented with sNP-IGF-1 had significantly lower metabolic activity (1.32-fold; P=0.007), a consistent amount of collagen type I, and significantly reduced collagen type II protein expression (1.86-fold; P=0.001) after 7 days compared to 3 days. CONCLUSION: Collagen-based scaffolds enriched with growth factors, such as IGF-1 coupled to nanoparticles, represent an improved therapeutic intervention for the targeted and controlled treatment of articular cartilage lesions.


Subject(s)
Cell Differentiation/drug effects , Chondrocytes/drug effects , Collagen/chemistry , Insulin-Like Growth Factor I/pharmacology , Nanoparticles/chemistry , Tissue Scaffolds/chemistry , Aged , Cells, Cultured , Chondrocytes/cytology , Female , Humans , Male
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.
Int J Mol Sci ; 15(2): 2454-64, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24521883

ABSTRACT

By means of plasma polymerization, positively charged, nanometre-thin coatings can be applied to implant surfaces. The aim of the present study was to quantify the adhesion of human bone cells in vitro and to evaluate the bone ongrowth in vivo, on titanium surfaces modified by plasma polymer coatings. Different implant surface configurations were examined: titanium alloy (Ti6Al4V) coated with plasma-polymerized allylamine (PPAAm) and plasma-polymerized ethylenediamine (PPEDA) versus uncoated. Shear stress on human osteoblast-like MG-63 cells was investigated in vitro using a spinning disc device. Furthermore, bone-to-implant contact (BIC) was evaluated in vivo. Custom-made conical titanium implants were inserted at the medial tibia of female Sprague-Dawley rats. After a follow-up of six weeks, the BIC was determined by means of histomorphometry. The quantification of cell adhesion showed a significantly higher shear stress for MG-63 cells on PPAAm and PPEDA compared to uncoated Ti6Al4V. Uncoated titanium alloyed implants showed the lowest BIC (40.4%). Implants with PPAAm coating revealed a clear but not significant increase of the BIC (58.5%) and implants with PPEDA a significantly increased BIC (63.7%). In conclusion, plasma polymer coatings demonstrate enhanced cell adhesion and bone ongrowth compared to uncoated titanium surfaces.


Subject(s)
Osseointegration , Polymerization , Prostheses and Implants , Titanium , Alloys , Animals , Cell Adhesion , Cell Line , Coated Materials, Biocompatible , Female , Humans , Osteoblasts/metabolism , Rats
4.
Int J Exp Pathol ; 90(4): 387-99, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19659897

ABSTRACT

The cytosolic cysteine protease calpain is implicated in a multitude of cellular functions but also plays a role in cell damage. Our previous results suggest that an activation of calpain accompanied by a decrease in its endogenous inhibitor calpastatin may contribute to pancreatic damage during cerulein-induced acute pancreatitis. The present study aimed at the time course of secretagogue-induced calpain activation and cellular substrates of the protease. Isolated rat pancreatic acini were incubated with a supramaximal concentration of cholecystokinin (0.1 microM CCK) for 30 min in the presence or absence of the calpain inhibitor Z-Val-Phe methyl ester (100 microM ZVP). The activation of calpain and the expression of calpastatin and the actin cytoskeleton-associated proteins alphaII-spectrin, E-cadherin and vinculin were studied by immunoblotting. The cell damage was assessed by lactate dehydrogenase release and ultrastructural analysis including fluorescence-labelled actin filaments. Immediately after administration, CCK led to activation of both calpain isoforms, mu- and m-calpain. The protease activation was accompanied by a decrease in the E-cadherin level and formation of calpain-specific breakdown products of alphaII-spectrin. A calpain-specific cleavage product of vinculin appeared concomitantly with changes in the actin filament organization. No effect of CCK on calpastatin was found. Inhibition of calpain by ZVP reduced CCK-induced damage of the actin-associated proteins and the cellular ultrastructure including the actin cytoskeleton. The results suggest that CCK-induced acinar cell damage requires activation of calpain and that the actin cytoskeleton belongs to the cellular targets of the protease.


Subject(s)
Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Cytoskeletal Proteins/metabolism , Dipeptides/pharmacology , Pancreas/enzymology , Pancreatitis/enzymology , Actins/analysis , Actins/metabolism , Acute Disease , Animals , Blotting, Western/methods , Cadherins/analysis , Cadherins/metabolism , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calpain/genetics , Calpain/metabolism , Ceruletide , Cholecystokinin/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/enzymology , Cytoskeleton/ultrastructure , Enzyme Activation , Female , Gene Expression , Microscopy, Confocal , Microscopy, Electron , Models, Animal , Organ Culture Techniques , Pancreas/drug effects , Pancreas/ultrastructure , Pancreatitis/pathology , RNA, Messenger/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods , Spectrin/metabolism , Stimulation, Chemical , Time Factors , Vinculin/metabolism
5.
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
6.
Acta Biomater ; 4(6): 1985-95, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18595788

ABSTRACT

Bioinert titanium (Ti) materials are generally encapsulated by fibrous tissue after implantation into the living body. To improve the bone-bonding ability of Ti implants, we activated commercially pure titanium (cpTi) by a simple chemical pre-treatment in HCl and NaOH. Subsequently, we exposed the treated samples to simulated body fluid (SBF) for 2 (TiCT) and 14 days (TiHCA), respectively, to mimic the early stages of bone bonding and to investigate the in vitro response of osteoblasts on thus altered biomimetic surfaces. Sample surfaces were characterized by scanning electron microscopy, energy-dispersive X-ray analysis, cross-sectional transmission electron microscopy analyses, Fourier transform infrared and Raman spectroscopy. It was shown that the efflorescence consisting of sodium titanate that is present on pre-treated cpTi surfaces transformed to calcium titanate after 2 days in SBF. After 14 days in SBF a homogeneous biomimetic apatite layer precipitated. Human osteoblasts (MG-63) revealed a well spread morphology on both functionalized Ti surfaces. On TiCT, the gene expression of the differentiation proteins alkaline phosphatase (ALP) and bone sialo protein was increased after 2 days. On both TiCT and TiHCA, the collagen I and ALP expression on the protein level was enhanced at 7 and 14 days. The TiCT and the TiHCA surfaces reveal the tendency to increase the differentiated cell function of MG-63 osteoblasts. Thus, chemical pre-treatment of titanium seems to be a promising method to generate osteoconductive surfaces.


Subject(s)
Biocompatible Materials/chemistry , Biomimetics , Osteoblasts/metabolism , Titanium/chemistry , Alkaline Phosphatase/metabolism , Bone Substitutes/chemistry , Bone and Bones/metabolism , Cell Line, Tumor , Humans , Microscopy, Confocal , Microscopy, Electron , Oxides/chemistry , Spectroscopy, Fourier Transform Infrared , Spectrum Analysis, Raman/methods , Surface Properties
7.
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
8.
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
9.
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
10.
Biomaterials ; 26(4): 383-92, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15275812

ABSTRACT

The mechanisms of cell adhesion to the extracellular matrix (ECM) which are of fundamental importance for function, survival, and growth of cells involve the formation of focal adhesions to facilitate integrin signaling. Recently, it became evident that focal adhesions are not stable but move to enable cell migration and ECM formation. We examined the number, size, and dynamic behavior of focal adhesions in living MG-63 osteoblastic cells, which were cultured on titanium surfaces with different roughnesses and on stainless steel (SS). As a marker for focal adhesions we used GFP-tagged vinculin, a cytoskeletal protein. Focal adhesions were smaller on titanium and on SS than on collagen-coated glass coverslips. The corundum-blasted rough surface of titanium induced the smallest adhesions. On all the surfaces that we have tested, we observed a mobility of focal adhesions. On collagen-coated coverslips focal adhesions moved with a speed of 60 nm/min. The speed was reduced on titanium and still more restricted on SS. The topography did not affect the mobility of focal adhesions. We conclude that on the material surfaces that we have studied a reduced mobility of focal adhesions may strengthen the linkages between cell and ECM but impair the ability to dynamically organize and remodel the ECM. The results may have a great impact in the functional evaluation of tailored biomaterial surfaces for the application in tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Focal Adhesions/pathology , Focal Adhesions/physiology , Osteoblasts/cytology , Osteoblasts/physiology , Stainless Steel/chemistry , Titanium/chemistry , Cell Adhesion/physiology , Cell Movement/physiology , Extracellular Matrix/physiology , Humans , Materials Testing , Surface Properties
11.
Biomaterials ; 26(15): 2423-40, 2005 May.
Article in English | MEDLINE | ID: mdl-15585246

ABSTRACT

Mechanisms of cell adhesion and extracellular matrix formation are primary processes in the interaction with the material surface of an implant which are controlled by integrin receptors. The aim of our study was to find out whether beta1- and beta3-integrins of osteoblastic cells sense the surface topography of titanium, and if structural alterations of integrin adhesions were involved in the organization of fibronectin. Pure titanium surfaces were modified by polishing (P), machining (NT), blasting with glass spheres (GB), and blasting with corundum particles (CB) resulting in increasing roughness. Confocal microscopic investigations revealed fibrillar adhesions of beta1- and alpha5-integrins on P, NT, and GB, but on CB with its sharp edges these integrin subunits did not form fibrillar adhesions. beta3 generally appeared in focal adhesions. We observed aligned fibrillar structures of fibronectin on NT not only on the basal site but interestingly, also on the apical cell surface. In contrast, on CB, fibronectin appeared apically clustered. We suggest that this alignment of fibronectin fibrils depends on the directed actin cytoskeleton and in particular, on the capability of the beta1-integrins to form fibrillar adhesions, which is affected by the surface roughness of titanium.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Adhesion/physiology , Fibronectins/metabolism , Integrin beta1/metabolism , Integrin beta3/metabolism , Osteoblasts/cytology , Osteoblasts/physiology , Titanium/chemistry , Adsorption , Binding Sites , Biocompatible Materials/chemistry , Cell Movement/physiology , Cell Size , Cells, Cultured , Cytoskeletal Proteins/metabolism , Hardness , Humans , Materials Testing , Protein Binding , Surface Properties , Titanium/analysis
12.
J Mater Sci Mater Med ; 15(4): 437-40, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15332613

ABSTRACT

Electrochemically deposited calcium phosphate (CaP) coatings are fast resorbable and existent only during the first period of osseointegration. In the present study, composite coatings with varying solubility (hydroxyapatite (HA), brushite with less HA and monetite (M) with less HA) were prepared and the influence of the degradation and the reprecipitation of CaP on osteoblastic cells were investigated. On the brushite composite coating a new precipitated, finely structured CaP phase was observed during immersion in cell culture medium with or without osteoblastic cells. The surface morphology of monetite and HA coatings were entirely unmodified under the same conditions. So it could be assumed that electrochemically deposited brushite with less HA acts as a precursor for new precipitated CaP. On this surface osteoblastic cells revealed a well-spread morphology with pronounced actin cytoskeleton and demonstrated good proliferation behaviour. Thus we suggest that brushite seems to be especially suitable for coating of implants as a matrix for nucleation and growth of new bone.


Subject(s)
Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Calcium Phosphates/chemistry , Coated Materials, Biocompatible/chemistry , Electrochemistry/methods , Osteoblasts/cytology , Osteoblasts/metabolism , Cell Division , Cell Line , Humans , Manufactured Materials/analysis , Materials Testing , Surface Properties , Titanium/chemistry
13.
Pancreatology ; 4(1): 12-21, 2004.
Article in English | MEDLINE | ID: mdl-14988654

ABSTRACT

BACKGROUND: One of the early events leading to alcoholic pancreatitis seems to be the effect of ethanol on stimulus-secretion coupling. This study examines ethanol-induced modifications of filamentous actin (F-actin) content and localization in acini, the resulting alpha-amylase secretion and the role of protein kinase C (PKC) activity in these processes. METHODS: Freshly isolated acini were treated with different concentrations of ethanol or cholecystokinin octapeptide (CCK-8) for different periods. F-actin was localized by confocal laser scanning microscopy; its quantity was determined fluorometrically, and the alpha-amylase secretion was measured. RESULTS: Ethanol caused F-actin reorganization resembling the effects of supramaximal CCK-8 stimulation and of direct PKC activation by phorbol-12-myristate-13-acetate. The polyphasic time course of the F-actin content also resembled that under supramaximal CCK-8 stimulation and was counteracted by inhibition of PKC. The PKC inhibitor bisindolylmaleimide I did not increase the ethanol- induced alpha-amylase secretion, but the suboptimally CCK-8-stimulated secretion via high-affinity receptors. CONCLUSION: Ethanol, like supramaximal CCK-8 concentrations, inhibits acinar secretion by reorganization of the actin cytoskeleton via PKC activation. This effect is suggested to be mediated by low-affinity CCK-A receptors. Together with the ethanol-induced stimulation of early steps of stimulus-secretion coupling, this may be a pancreas-damaging mechanism resembling that in experimental hyperstimulation pancreatitis.


Subject(s)
Actins/analysis , Cytoskeleton/chemistry , Cytoskeleton/drug effects , Ethanol/pharmacology , Pancreas/ultrastructure , Sincalide/pharmacology , Animals , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Female , L-Lactate Dehydrogenase/metabolism , Pancreas/enzymology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Rats, Inbred Lew , Tetradecanoylphorbol Acetate/pharmacology , alpha-Amylases/metabolism
14.
J Bone Miner Res ; 17(4): 603-11, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11918217

ABSTRACT

Following the idea that integrin receptors function as mechanotransducers, we applied defined physical forces to integrins in osteoblastic cells using a magnetic drag force device to show how cells sense different modes of physical forces. Application of mechanical stress to the beta1-integrin subunit revealed that cyclic forces of 1 Hz were more effective to stimulate the cellular calcium response than continuous load. Cyclic forces also induced an enhanced cytoskeletal anchorage of tyrosine-phosphorylated proteins and increased activation of the focal adhesion kinase (FAK) and mitogen activated protein (MAP) kinase. These events were dependent on an intact cytoskeleton and the presence of intracellular calcium. Analyses of the intracellular spatial organization of the calcium responses revealed that calcium signals originate in a restricted region in the vicinity of the stressed receptors, which indicates that cells are able to sense locally applied stress on the cell surface via integrins. The calcium signals can spread throughout the cell including the nucleus, which shows that calcium also is a candidate to transmit mechanically induced information into different cellular compartments.


Subject(s)
Calcium/metabolism , Integrin beta1/metabolism , Osteoblasts/physiology , Signal Transduction , Cells, Cultured , Cytoskeleton/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Stress, Mechanical , Tyrosine/metabolism
15.
Pancreas ; 24(1): 63-74, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11741184

ABSTRACT

INTRODUCTION: Calpains, cytosolic Ca(2+)-dependent cysteine proteases, are expressed in a variety of mammalian cells and have been found to participate in stimulus-secretion coupling in platelets and alveolar cells. AIMS: In pancreatic acinar cells, expression of calpains and their role in the secretory process have not yet been elucidated. Both subjects, therefore, were examined in the current study. METHODOLOGY: mu-calpain and m-calpain were detected immunochemically. Calpain activation was measured by fluorescence spectrophotometry and single-cell fluorometry using Suc-Leu-Leu-Val-Tyr-AMC as substrate. Amylase secretion and cell damage, characterized by lactate dehydrogenase release, were measured by colorimetric assays. RESULTS: Immunochemistry revealed cytoplasmic localization of both calpain isoforms. Immediately after increasing the cytosolic Ca(2+) concentration with ionomycin, a marked dose-dependent protease activation and cellular damage were observed. Inhibition of ionomycin-mediated enzyme activation through preincubation of cells with Ca(2+)-free medium, BAPTA-AM, or Z-Leu-Leu-Tyr-CHN(2) significantly reduced cell injury. Cholecystokinin (100 pM) also induced proteolytic activity, preceding cholecystokinin-stimulated amylase secretion. Protease activity and amylase release were significantly inhibited by Z-Leu-Leu-Tyr-CHN(2 ) retreatment. CONCLUSION: Calpains are expressed in pancreatic acinar cells and may participate in stimulus-secretion coupling. In addition, our study indicates that pathologic calpain activation may contribute to Ca(2+)-mediated acinar cell damage.


Subject(s)
Calpain/biosynthesis , Pancreas/metabolism , Amylases/analysis , Amylases/metabolism , Animals , Calcium/metabolism , Calcium/pharmacology , Calpain/analysis , Calpain/antagonists & inhibitors , Cells, Cultured , Cholecystokinin , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Immunohistochemistry , Ionomycin , Isoenzymes/biosynthesis , Oligopeptides/pharmacology , Pancreas/enzymology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...