Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters










Publication year range
1.
Int J Cardiol ; 197: 327-32, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26159040

ABSTRACT

BACKGROUND: Myocardial apoptosis has been discussed to play a pivotal role in the development and progression of congestive heart failure (CHF). However, recently there is doubt on the evidence of myocardial apoptosis in heart failure as information on ultrastructural changes by electron microscopy is still scarce. This project therefore aimed to detect direct morphological evidence of myocardial apoptosis in an experimental heart failure model. METHOD: Following IRB approval, an aortocaval fistula (ACF) was induced in male Wistar rats using a 16G needle. 28±2days following ACF rats were examined by hemodynamic measurements, Western blot, immunofluorescence confocal and electron microscopic analysis. RESULTS: Within 28±2days of ACF heart (3.8±0.1 vs. 6.6±0.3mg/g) and lung (3.7±0.2 vs. 6.9±0.5mg/g) weight indices significantly increased in the ACF group accompanied by a restriction in systolic (LVEF: 72±2 vs. 39±3%) and diastolic (dP/dtmin.: -10,435±942 vs. -5982±745mmHg/s) function (p<0.01). Activated caspase-3 was significantly increased in failing hearts concomitant with mitochondrial leakage of cytochrome c into the cytosol. Finally, electron microscopy of the left ventricle (LV) of ACF rats revealed pronounced ultrastructural changes in >70% of examined cardiomyocytes, such as nuclear chromatin condensation, myofibril loss and disarray, contour irregularities and amorphous dense bodies, mitochondriosis and damaged cell-cell-contacts between cardiomyocytes. CONCLUSIONS: Volume overload induced heart failure is associated with activation of the mitochondrial apoptotic pathway. In addition, electron microscopy of the LV revealed direct ultrastructural evidence of extended myocardial apoptosis in ACF rats.


Subject(s)
Apoptosis , Heart Failure/pathology , Myocardium/pathology , Myocardium/ultrastructure , Ventricular Remodeling , Animals , Apoptosis/physiology , Male , Myocytes, Cardiac/ultrastructure , Rats , Rats, Wistar , Ventricular Remodeling/physiology
2.
Res Pharm Sci ; 9(6): 431-43, 2014.
Article in English | MEDLINE | ID: mdl-26339258

ABSTRACT

Amyloid pathology is associated with fibril aggregation of different proteins which results in the progressive damage of affected organs. It is strongly believed that specific small molecules interfere with fibrillation by interacting with the amyloidogenic proteins. We had previously reported the strong and long-term inhibition of fibrillation of hen egg white lysozyme (HEWL) by Cuminum cyminum oil. Herein, it was intended to rationally identify the active anti-amyloidogenic compounds of the oil. After fractionation, the highest inhibitory effect was observed in the toluene-ethyl acetate part of the oil. Gas chromatography-mass spectrometry (GC-MS) analysis of this fraction indicated that eight compounds were predominantly present in the fraction. Unexpectedly, two compounds including terpinolene and limonene, having very similar chemical structures, inhibited and induced fibrillation, respectively. PC12 cells (derived from a transplantable rat pheochromocytoma) were affected by HEWL fibrils, whereas the inhibited forms of fibrils in the presence of terpinolene led to higher levels of viability, as shown by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT), lactate dehydrogenase (LDH) and flow cytometry assays. Molecular local docking analysis suggested a site of interaction for terpinolene in the flexible cleft of the protein. This interaction site is close to tryptophan -62 and -63 and two other hydrophobic residues in the hot spot regions of the protein. Seemingly, these interactions interrupt protein self-assembly and therefore, fibril formation. Despite previously reported small anti-amyloid molecules which have aromatic flat rings, terpinolene ring is not flat. This functionally durable small molecule may aid us toward developing new anti-amyloidogenic compounds with extended activity.

3.
Histol Histopathol ; 28(8): 955-64, 2013 08.
Article in English | MEDLINE | ID: mdl-23463583

ABSTRACT

Tendonitis and tendinitis are terms used to describe an inflamed and painful tendon. Tendinopathy, is a descriptive term for describing clinical conditions arising from tendon injury and overuse both within and around tendons. The aim of this mini-review is to explore the role of pro-inflammatory cytokines, particularly interleukin-1ß (IL-1ß) in tendon disorders. A number of investigators including our group have proposed that pro-inflammatory cytokines such as IL-1ß are initiators of tendinopathies, stimulating inflammation, apoptosis and extracellular matrix (ECM) degradation. This is one of the reasons why IL-1ß is frequently used in culture models of tendon inflammation to study the inflammatory and catabolic responses of tenocytes. However, some researchers oppose this view and suggest that although IL-1ß may play a role in rheumatoid arthritis (RA) and osteoarthritis (OA), the involvement of IL-1ß in the development of tendinopathy is questionable. This mini-review discusses the relevant papers published in this area and summarises the evidence for and against the involvement of pro-inflammatory cytokines such as IL-1ß in tendonitis. Reaching a consensus will be important for the development and refinement of biomimetic models of tendon inflammation and the formulation of new therapeutic strategies for the treatment of tendon injuries.


Subject(s)
Cytokines/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Tendinopathy/metabolism , Achilles Tendon/pathology , Achilles Tendon/ultrastructure , Animals , Apoptosis , Collagen/chemistry , Extracellular Matrix/metabolism , Fluoroquinolones/chemistry , Humans , Inflammation/physiopathology , Rats , Tendons/pathology
4.
Histol Histopathol ; 26(9): 1173-85, 2011 09.
Article in English | MEDLINE | ID: mdl-21751149

ABSTRACT

Tendons have a limited capacity for self-repair due to the low density and mitotic activity of tenocytes. Pro-inflammatory cytokines such as interleukin-1ß (IL-1ß) have been identified as the main initiators of tendinopathies, stimulating inflammation, apoptosis and extracellular matrix (ECM) degradation. The aim of this study was to evaluate the potential of Tendoactive®, a newly developed proprietary nutraceutical formulation that includes mucopolysaccharides, collagen and vitamin C, in an in vitro model of tendon inflammation. The effects of Tendoactive® were studied in primary cultures of human tenocytes treated with IL-1ß for up to 72 h. Expression of collagen type I, integrin ß1, cyclo-oxygenase-2 (COX-2), caspase-3 and matrix metalloproteinase-1 (MMP-1) was monitored by western blotting. The effects of Tendoactive® on the expression, phosphorylation and nuclear translocation of protein components of the NF-κB system were studied by western blotting and immunofluorescence respectively. Treatment of tenocytes with Tendoactive® suppressed IL-1ß-induced NF-κB activation and p65 nuclear translocation. These events correlated with down-regulation of NF-κB targets including COX-2, MMP-1 and activated caspase-3. Tendoactive® also reversed the IL-1ß-induced down-regulation of collagen type I and ß1-integrin receptor expression. These results indicate that Tendoactive® has nutraceutical potential as an anti-inflammatory agent for treating tendinopathy through suppression of NF-κB mediated IL-1ß catabolic signalling pathways in tenocytes.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Dietary Supplements , Metabolism/drug effects , Tendons/cytology , Tendons/drug effects , Adult , Apoptosis/drug effects , Blotting, Western , Cell Nucleus/drug effects , Cell Proliferation/drug effects , Cell Separation , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/drug effects , Culture Media , Fluorescent Antibody Technique , Humans , Interleukin-1beta/physiology , Male , Microscopy, Electron, Transmission , Middle Aged , Mitochondria/drug effects , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Signal Transduction/drug effects , Transcription Factor RelA/metabolism
5.
Histochem Cell Biol ; 135(5): 453-60, 2011 May.
Article in English | MEDLINE | ID: mdl-21476078

ABSTRACT

Little is know about the pathophysiology of acute and degenerative tendon injuries. Although most lesions are uncomplicated, treatment is long and unsatisfactory in a considerable number of cases. Besides the common growth factors that were shown to be relevant for tendon integrity more recently protection against oxidative stress was shown to promote tendon healing. To improve tendon regeneration, many have advocated the use of platelet-rich plasma (PRP), a thrombocyte concentrate that can serve as an autologous source of growth factors. In this study, we investigated the effect of platelet-released growth factors (PRGF) on tenocytes. Tenocytes were isolated from the Achilles tendon of postnatal rats. Tenocyte cell cultures were stimulated with PRGF. We used a CyQuant assay and WST assay to analyse tendon cell growth and viability in different concentrations of PRGF. Migration and proliferation of cells grown in PRGF were assessed by a scratch test. A dual-luciferase assay was used to demonstrate the activation of the anti-oxidant response element (ARE) in tenocytes. A positive effect of PRGF could be shown on tendon cell growth and migratory capacity. PRGF activated the Nrf2-ARE pathway in a dose-dependent manner. Here, we provide evidence of a biological effect of PRGF on tenocytes by the promotion of tenocyte growth and activation of the Nrf2-ARE pathway. This is a novel aspect of the action of platelet concentrates on tendon growth.


Subject(s)
Antioxidants/metabolism , Blood Platelets/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Response Elements/genetics , Tendons/cytology , Tendons/drug effects , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Male , NF-E2-Related Factor 2/metabolism , Phenotype , Rats , Rats, Wistar , Tendons/metabolism
6.
Osteoarthritis Cartilage ; 18(2): 141-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19836480

ABSTRACT

OBJECTIVES: Curcumin (diferuloylmethane) is the principal biochemical component of the spice turmeric and has been shown to possess potent anti-catabolic, anti-inflammatory and antioxidant, properties. This article aims to provide a summary of the actions of curcumin on articular chondrocytes from the available literature with the use of a text-mining tool. We highlight both the potential benefits and drawbacks of using this chemopreventive agent for treating osteoarthritis (OA). We also explore the recent literature on the molecular mechanisms of curcumin mediated alterations in gene expression mediated via activator protein 1 (AP-1)/nuclear factor-kappa B (NF-kappaB) signalling in chondrocytes, osteoblasts and synovial fibroblasts. METHODS: A computer-aided search of the PubMed/Medline database aided by a text-mining tool to interrogate the ResNet Mammalian database 6.0. RESULTS: Recent work has shown that curcumin protects human chondrocytes from the catabolic actions of interleukin-1 beta (IL-1beta) including matrix metalloproteinase (MMP)-3 up-regulation, inhibition of collagen type II and down-regulation of beta1-integrin expression. Curcumin blocks IL-1beta-induced proteoglycan degradation, AP-1/NF-kappaB signalling, chondrocyte apoptosis and activation of caspase-3. CONCLUSIONS: The available data from published in vitro and in vivo studies suggest that curcumin may be a beneficial complementary treatment for OA in humans and companion animals. Nevertheless, before initiating extensive clinical trials, more basic research is required to improve its solubility, absorption and bioavailability and gain additional information about its safety and efficacy in different species. Once these obstacles have been overcome, curcumin and structurally related biochemicals may become safer and more suitable nutraceutical alternatives to the non-steroidal anti-inflammatory drugs that are currently used for the treatment of OA.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/drug effects , Curcumin/pharmacology , Enzyme Inhibitors/pharmacology , Signal Transduction/drug effects , Apoptosis/drug effects , CD18 Antigens/metabolism , Cartilage, Articular/cytology , Caspase 3/metabolism , Cells, Cultured , Chondrocytes/metabolism , Collagen Type II/metabolism , Humans , Inflammation/physiopathology , Interleukin-1beta/metabolism , Matrix Metalloproteinase 3/metabolism , NF-kappa B/physiology , Osteoarthritis/drug therapy , Osteoarthritis/prevention & control , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factor AP-1/physiology
7.
Histol Histopathol ; 24(3): 347-66, 2009 03.
Article in English | MEDLINE | ID: mdl-19130405

ABSTRACT

Defects of load-bearing connective tissues such as articular cartilage, often result from trauma, degenerative or age-related disease. Osteoarthritis (OA) presents a major clinical challenge to clinicians due to the limited inherent repair capacity of articular cartilage. Articular cartilage defects are increasingly common among the elderly population causing pain, reduced joint function and significant disability among affected patients. The poor capacity for self-repair of chondral defects has resulted in the development of a large variety of treatment approaches including Autologous Chondrocyte Transplantation (ACT), microfracture and mosaicplasty methods. In ACT, a cartilage biopsy is taken from the patient and articular chondrocytes are isolated. The cells are then expanded after several passages in vitro and used to fill the cartilage defect. Since its introduction, ACT has become a widely applied surgical method with good to excellent clinical outcomes. More recently, classical ACT has been combined with tissue engineering and implantable scaffolds for improved results. However, there are still major problems associated with the ACT technique which relate mainly to chondrocyte de-differentiation during the expansion phase in monolayer culture and the poor integration of the implants into the surrounding cartilage tissue. Novel approaches using mesenchymal stem cells (MSCs) as an alternative cell source to patient derived chondrocytes are currently on trial. MSCs have shown significant potential for chondrogenesis in animal models. This review article discusses the potential of MSCs in tissue engineering and regenerative medicine and highlights their potential for cartilage repair and cell-based therapies for osteoarthritis and a range of related osteoarticular disorders.


Subject(s)
Cartilage, Articular/pathology , Mesenchymal Stem Cells/cytology , Osteoarthritis/pathology , Regenerative Medicine , Tissue Engineering/methods , Aged , Cartilage, Articular/surgery , Connective Tissue/pathology , Humans , Mesenchymal Stem Cell Transplantation/methods , Models, Biological , Osteoarthritis/surgery
8.
Histochem Cell Biol ; 131(2): 251-66, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18941769

ABSTRACT

Tissue engineering of bone grafts with osteogenic progenitor cells such as adult mesenchymal stem cells (MSC) represents a promising strategy for the treatment of large bone defects. The aim of this experimental study was to evaluate the osteogenic potential of primary osteoblasts on MSCs in co-culture at different ratios. The co-cultures were treated with or without a specific osteogenic induction medium in monolayer and high density cultures. In monolayer co-cultures, MSCs and osteoblasts actively searched for cell-cell contact leading to cell proliferation and only in treated monolayer co-cultures osteogenesis was observed. Ultrastructural evaluation of high density co-cultures using electron microscopy demonstrated osteogenesis with no significant difference between treated or untreated co-cultures. Immunoblotting confirmed expression of collagen type I, beta1-Integrin, the osteogenic-specific transcription factor Cbfa-1 and induction of the MAPKinase pathway (Shc, Erk1/2) in both treated and untreated co-cultures. Although treatment with the induction medium enhanced osteogenesis in the co-cultures compared to untreated co-cultures, the quality of osteogenesis was proportional to the quantity of osteoblasts in the co-cultures. Fifty percent osteoblasts in the co-cultures markedly increased osteogenesis; even the presence of ten percent osteoblasts in the co-culture strongly promoted osteogenesis. This data leads us to conclude that co-culture of MSCs with osteoblasts combined with the three-dimensional environment of the high density culture strongly promotes osteogenesis and stabilizes the osteogenic potential of MSCs. This approach may prove to be of practical benefit in future tissue engineering and regenerative medicine research.


Subject(s)
Cell Communication , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteogenesis , Animals , Cell Differentiation , Cell Proliferation , Coculture Techniques , Collagen Type I/analysis , Core Binding Factor Alpha 1 Subunit/analysis , Dogs , Integrin beta1/analysis , MAP Kinase Signaling System , Tissue Engineering
9.
Ann Anat ; 190(5): 395-412, 2008 Nov 20.
Article in English | MEDLINE | ID: mdl-18842397

ABSTRACT

Osteoarthritis (OA) resulting from trauma, degenerative or age-related disease presents a major clinical challenge due to the limited repair capacity of articular cartilage. This poor self-repair capacity of osteochondral defects has resulted in the development of a wide variety of new treatment approaches. Although the use of chondrocytes in applications of cartilage tissue engineering is still prevalent, concerns associated with donor-site morbidity, cell de-differentiation and the limited lifespan of these cells have brought the use of mesenchymal stem cells (MSCs) to the forefront of such applications. Therefore, in the last two decades MSCs have come into the focus of connective tissue engineering and regenerative medicine and have become increasingly sought after as an alternative cell source for improving well-established methods of osteochondrotic cartilage defect repair such as the Autologous Chondrocyte Transplantation method, but are also being tested as an ideal cell source in combination with newly developed implantable scaffolds or as a target/carrier cell in other new concepts of regenerative medicine. However, up to now, although in animal models MSCs have already shown significant potential for cartilage repair and novel approaches using MSCs as an alternative cell source to patient-derived chondrocytes are being tested, much more research is needed before feasible clinical application of MSCs becomes reality.


Subject(s)
Cartilage/cytology , Mesoderm/cytology , Stem Cells/cytology , Tissue Engineering/methods , Cartilage/physiology , Cell Culture Techniques/methods , Connective Tissue/pathology , Connective Tissue Cells/cytology , Germany , Humans , Regeneration , Tissue Engineering/trends , Wounds and Injuries/therapy
10.
Adv Anat Embryol Cell Biol ; 197: 1-60, 2008.
Article in English | MEDLINE | ID: mdl-18637520

ABSTRACT

Integrins are heterodimeric integral membrane proteins made up of alpha and beta subunits. At least eighteen alpha and eight beta subunit genes have been described in mammals. Integrin family members are plasma membrane receptors involved in cell adhesion and active as intra- and extracellular signalling molecules in a variety of processes including embryogenesis, hemostasis, tissue repair, immune response and metastatic spread of tumour cells. Integrin beta 1 (beta1-integrin), the protein encoded by the ITGB1 gene (also known as CD29 and VLAB), is a multi-functional protein involved in cell-matrix adhesion, cell signalling, cellular defense, cell adhesion, protein binding, protein heterodimerisation and receptor-mediated activity. It is highly expressed in the human body (17.4 times higher than the average gene in the last updated revision of the human genome). The extracellular matrix (ECM) of articular cartilage is a unique environment. Interactions between chondrocytes and the ECM regulate many biological processes important to homeostasis and repair of articular cartilage, including cell attachment, growth, differentiation and survival. The beta1-integrin family of cell surface receptors appears to play a major role in mediating cell-matrix interactions that are important in regulating these fundamental processes. Chondrocyte mechanoreceptors have been proposed to incorporate beta1-integrins and mechanosensitive ion channels which link with key ECM, cytoskeletal and signalling proteins to maintain the chondrocyte phenotype, prevent chondrocyte apoptosis and regulate chondrocyte-specific gene expression. This review focuses on the expression and function of beta1-integrins in articular chondrocytes, its role in the unique biology of these cells and its distribution in cartilage.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Integrins/metabolism , Animals , Cell Culture Techniques , Humans
11.
Orthopade ; 37(5): 440-7, 2008 May.
Article in German | MEDLINE | ID: mdl-18427772

ABSTRACT

BACKGROUND: Autologous chondrocyte transplantation (ACT) has had reasonable success for repairing small articular cartilage defects. A limiting factor for ACT is, however, the in vitro cultivation of chondrocytes because it leads to dedifferentiation. Therefore, the goal of this work was to optimize the monolayer culture of chondrocytes in vitro. MATERIAL AND METHOD: Human articular chondrocytes were plated on either collagen type II or untreated surfaces. The cells were evaluated morphologically and with immunoblotting. RESULTS: On collagen type II surfaces, a stable chondrogenic phenotype, expression of beta1-integrin, and a significant activation of phosphorylated intracellular proteins and the adaptor protein Shc could be observed up to day 20 in culture. Treatment with beta1 integrin antibody led to a loss of cell adhesion (82%). The results indicate that on collagen type II, beta1-integrin receptors are activated. Through the activation of Shc, these stimulate the Ras-MAPK pathway, which stabilizes the chondrogenic phenotype. CONCLUSION: Our results provide a practical and low-cost solution for improved long-term chondrocyte cultivation, thus providing a new perspective for using ACT on larger or arthrotic cartilage defects.


Subject(s)
Cell Culture Techniques/methods , Chondrocytes/cytology , Chondrocytes/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Tissue Engineering/methods , Cell Proliferation , Cell Survival , Cells, Cultured , Chondrocytes/transplantation , Humans
12.
Histochem Cell Biol ; 128(6): 507-20, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17922135

ABSTRACT

Musculoskeletal diseases with osteochondrotic articular cartilage defects, such as osteoarthritis, are an increasing problem for humans and companion animals which necessitates the development of novel and improved therapeutic strategies. Canine mesenchymal stem cells (cMSCs) offer significant promise as a multipotent source for cell-based therapies and could form the basis for the differentiation and cultivation of tissue grafts to replace damaged tissue. However, no comprehensive analysis has been undertaken to characterize the ultrastructure of in vitro differentiated cMSCs. The main goal of this paper was to focus on cMSCs and to analyse their differentiation capacity. To achieve this aim, bone marrow cMSCs from three canine patients were isolated, expanded in monolayer culture and characterized with respect to their ability for osteogenic, adipogenic and chondrogenic differentiation capacities. cMSCs showed proliferative potential and were capable of osteogenic, adipogenic and chondrogenic differentiation. cMSCs treated with the osteogenic induction medium differentiated into osteoblasts, produced typical bone matrix components, beta1-integrins and upregulated the osteogenic specific transcription factor Cbfa-1. cMSCs treated with the adipogenic induction medium showed typical adipocyte morphology, produced adiponectin, collagen type I and beta1-integrins, and upregulated the adipogenic specific transcription factor PPAR-gamma. cMSCs treated with the chondrogenic induction medium exhibited a round to oval shape, produced a cartilage-specific extracellular matrix, beta1-integrins and upregulated the chondrogenic specific transcription factor Sox9. These results demonstrate, at the biochemical, morphological and ultrastructural levels, the multipotency of cMSCs and thus highlight their potential therapeutic value for cell-based tissue engineering.


Subject(s)
Adipocytes/physiology , Chondrogenesis/physiology , Mesenchymal Stem Cells/physiology , Osteogenesis/physiology , Adipocytes/cytology , Adipocytes/ultrastructure , Animals , Cell Culture Techniques/methods , Cell Differentiation , Cell Separation , Dogs , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/ultrastructure , Microscopy, Immunoelectron
13.
Histol Histopathol ; 22(5): 469-82, 2007 05.
Article in English | MEDLINE | ID: mdl-17330802

ABSTRACT

Elevated levels of the pro-inflammatory cytokine, interleukin-18 (IL-18) have recently been demonstrated in osteoarthritic cartilage. However, the effects of IL-18 on chondrocyte signalling and matrix biosynthesis are poorly understood. Therefore, the present study was undertaken to further characterize the impact of IL-18 on human articular chondrocyte in vitro. Human articular chondrocytes were stimulated with various concentrations of recombinant human IL-18 (1, 10, 100 ng/ml) for 0, 4, 8, 12, 24, 48, 72 h in vitro. The effects of IL-18 on the cartilage-specific matrix protein collagen type II, the cytoskeletal protein vinculin, the cell matrix signal transduction receptor beta-integrin, key signalling proteins of the MAPKinase pathway (such as SHC (Sarc Homology Collagen) and activated MAPKinase [ERK-1/-2]), the pro-inflammatory enzyme cyclo-oxygenase-2 (COX-2) and the apoptosis marker activated caspase-3 were evaluated by Western blot analysis and immunofluorescence labelling. Morphological features of IL-18 stimulated chondrocytes were estimated by transmission electron microscopy. IL-18 lead to inhibition of collagen type II-deposition, decreased beta-integrin receptor and vinculin synthesis, SHC and MAPKinase activation, increased COX-2 synthesis and activation of caspase-3 in chondrocytes in a time- and dose-dependent manner. Furthermore, chondrocytes treated with IL-18 exhibited typical morphological features of apoptosis as revealed by transmission electron microscopy. Taken together, the results of the present study underline key catabolic events mediated by IL-18 signalling in chondrocytes such as loss of cartilage-specific matrix and apoptosis. Inhibition of MAPKinase signalling is hypothesized to contribute to these features. Future therapeutics targeting IL-18 signalling pathways may be beneficial in rheumatoid arthritis and osteoarthritis therapy.


Subject(s)
Apoptosis , Chondrocytes/metabolism , Interleukin-18/metabolism , MAP Kinase Signaling System , Aged , Apoptosis/drug effects , Blotting, Western , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Caspase 3/metabolism , Cell Shape/drug effects , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/ultrastructure , Collagen Type II/metabolism , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Fibronectins/metabolism , Humans , Integrin beta Chains/metabolism , Interleukin-18/pharmacology , MAP Kinase Signaling System/drug effects , Membrane Proteins/metabolism , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Time Factors , Vinculin/metabolism
15.
Apoptosis ; 11(3): 347-58, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16538381

ABSTRACT

BACKGROUND: Apoptosis plays a key role in the pathogenesis of cardiac diseases. We examined the influence of the renin-angiotensin system (RAS) on different regulators of apoptosis using an isolated hemoperfused working porcine heart model of acute ischemia (2 h), followed by reperfusion (4 h). METHODS AND RESULTS: 23 porcine hearts were randomized to 5 groups: hemoperfused non-infarcted hearts (C), infarcted hearts (MI: R. circumflexus), infarcted hearts treated with quinaprilat (Q), infarcted hearts treated with angiotensin-I (Ang I), and infarcted hearts treated with angiotensin-I and quinaprilat (QA). Fas, Bax, bcl-2 and p53 proteins were increased in MI hearts and further elevated by Ang I. Quinaprilat reduced Bax and p53. Bcl-2 was elevated in Q and reduced in QA. An early upregulation of caspase-3 gene and protein expression was detected in MI and Ang I hearts compared to C. Q reduced caspase-3 gene expression, but had no effect on caspase-3 and Fas protein. CONCLUSIONS: These data suggest that the RAS plays a pivotal role in cardiac apoptosis which is the early and predominant form of death in myocardial infarction. Ischemia/reperfusion induces programmed cell death via extrinsic and intrinsic pathways. Early treatment with quinaprilat attenuated cardiomyocyte apoptosis.


Subject(s)
Apoptosis/physiology , Heart/physiology , Myocardial Infarction/pathology , Myocardium , Renin-Angiotensin System/physiology , Reperfusion Injury , Angiotensin I/metabolism , Angiotensin II/metabolism , Angiotensin-Converting Enzyme Inhibitors/metabolism , Animals , Caspase 3/genetics , Caspase 3/metabolism , Cell Shape , Female , Myocardial Infarction/metabolism , Myocardium/cytology , Myocardium/metabolism , Myocardium/pathology , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Random Allocation , Swine , Tetrahydroisoquinolines/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism , fas Receptor/metabolism
16.
Apoptosis ; 11(5): 749-64, 2006 May.
Article in English | MEDLINE | ID: mdl-16528471

ABSTRACT

Endothelial cells play a crucial role in the pathogenesis of many diseases and are highly sensitive to low gravity conditions. Using a three-dimensional random positioning machine (clinostat) we investigated effects of simulated weightlessness on the human EA.hy926 cell line (4, 12, 24, 48 and 72 h) and addressed the impact of exposure to VEGF (10 ng/ml). Simulated microgravity resulted in an increase in extracellular matrix proteins (ECMP) and altered cytoskeletal components such as microtubules (alpha-tubulin) and intermediate filaments (cytokeratin). Within the initial 4 h, both simulated microgravity and VEGF, alone, enhanced the expression of ECMP (collagen type I, fibronectin, osteopontin, laminin) and flk-1 protein. Synergistic effects between microgravity and VEGF were not seen. After 12 h, microgravity further enhanced all proteins mentioned above. Moreover, clinorotated endothelial cells showed morphological and biochemical signs of apoptosis after 4 h, which were further increased after 72 h. VEGF significantly attenuated apoptosis as demonstrated by DAPI staining, TUNEL flow cytometry and electron microscopy. Caspase-3, Bax, Fas, and 85-kDa apoptosis-related cleavage fragments were clearly reduced by VEGF. After 72 h, most surviving endothelial cells had assembled to three-dimensional tubular structures. Simulated weightlessness induced apoptosis and increased the amount of ECMP. VEGF develops a cell-protective influence on endothelial cells exposed to simulated microgravity.


Subject(s)
Apoptosis/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Weightlessness Simulation , Cell Line , Collagen Type I/metabolism , Collagen Type I/ultrastructure , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Endothelium, Vascular/metabolism , Endothelium, Vascular/ultrastructure , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Fibronectins/metabolism , Fibronectins/ultrastructure , Humans , Keratins/metabolism , Keratins/ultrastructure , Laminin/metabolism , Laminin/ultrastructure , Osteopontin , Sialoglycoproteins/metabolism , Sialoglycoproteins/ultrastructure , Tubulin/metabolism , Tubulin/ultrastructure , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/ultrastructure , Weightlessness Simulation/instrumentation , Weightlessness Simulation/methods
17.
Histol Histopathol ; 21(5): 467-76, 2006 05.
Article in English | MEDLINE | ID: mdl-16493577

ABSTRACT

Cultivation of phenotypically stable auricular chondrocytes will have applications in autologous chondrocyte transplantation and reconstructive surgery of cartilage. Chondrocytes grown in monolayer culture rapidly dedifferentiate assuming a fibroblast-like morphology and lose their cartilage-specific pattern of gene expression. Three-dimensional high-density culture models mimic more closely the in vivo conditions of cartilage. Therefore, this study was undertaken to test whether the high-density cultures might serve as a suitable model system to acquire phenotypically and functionally differentiated auricular chondrocytes from porcine cartilage. Freshly isolated porcine auricular chondrocytes were cultured for 7 passages in monolayer culture. From each passage (passage 0 and 1-7) cells were introduced to high-density cultures and examined by transmission electron microscopy. Western blotting was used to analyse the expression of cartilage-specific markers, such as collagen type II and cartilage specific proteoglycan, fibronectin, cell adhesion and signal transduction receptor beta1-integrin, matrix metalloproteinases (MMP-9, MMP-13), cyclo-oxygenase (COX)-2 and the apoptosis commitment marker, activated caspase-3. When dedifferentiated auricular chondrocytes from monolayer passages 0-4 were cultured in high-density culture, they recovered their chondrocytic phenotype and formed cartilage nodules surrounded by fibroblast-like cells and synthesised collagen type II, proteoglycans, fibronectin and beta1-integrins. However, chondrocytes from monolayer passages 5-7 did not redifferentiate to chondrocytes even when transferred to high-density culture, and did not synthesize a chondrocyte-specific extracellular matrix. Instead, they produced increasing amounts of MMP-9, MMP-13, COX-2, activated caspase-3 and underwent apoptosis. Three-dimensional high-density cultures may therefore be used to obtain sufficient quantities of fully differentiated auricular chondrocytes for autologous chondrocyte transplantation and reconstructive plastic surgery.


Subject(s)
Cell Culture Techniques/methods , Chondrocytes/cytology , Ear , Plastic Surgery Procedures/methods , Surgery, Plastic/methods , Animals , Blotting, Western , Caspase 3 , Caspases/analysis , Cell Count , Cell Differentiation , Cell Proliferation , Cell Transplantation/methods , Cells, Cultured , Chondrocytes/chemistry , Chondrocytes/transplantation , Chondrocytes/ultrastructure , Collagen Type II/analysis , Collagenases/analysis , Fibronectins/analysis , Integrin beta1/analysis , Matrix Metalloproteinase 13 , Matrix Metalloproteinase 9/analysis , Microscopy, Electron, Transmission , Phenotype , Proteoglycans/analysis , Swine
18.
Mund Kiefer Gesichtschir ; 10(2): 106-17, 2006 Mar.
Article in German | MEDLINE | ID: mdl-16496112

ABSTRACT

BACKGROUND: After presenting two sisters with the rare form of congenital arrhinia, this syndrome is reviewed, an explanation of the pathogenesis is offered and the therapeutic options of the functional and aesthetic reconstruction are discussed. DISCUSSION: In cases of congenital arrhinia different degrees of respiratory distress, cyanotic episodes, and impaired food intake are described. Therefore after birth respiration and food intake need to be monitored to alleviate the situation through intubation or tracheotomy. The following conclusions could be made based on the literature overview. Little is known about the pathophysiology and a great variety of therapeutic interventions and reconstruction solutions with a wide spectrum of complications are described. Due to the numerous forms of complications, which need to be compared with the reconstructive results, indications for surgical reconstruction of the airway and plastic reconstruction of the nose during childhood must be defined very stringently. CONCLUSION: One method to achieve a satisfactory plastic result is with an osseointegrated prosthesis. This facial prosthesis can be inserted without complications and can guarantee an adequate result, whereas no impairment of maxillofacial development was noted.


Subject(s)
Craniofacial Abnormalities/genetics , Nose/abnormalities , Adolescent , Anophthalmos/diagnosis , Anophthalmos/genetics , Anophthalmos/surgery , Child , Child, Preschool , Craniofacial Abnormalities/diagnosis , Craniofacial Abnormalities/surgery , Female , Follow-Up Studies , Genes, Dominant , Humans , Pedigree , Prosthesis Design , Prosthesis Fitting , Prosthesis Implantation , Surgical Flaps , Tomography, X-Ray Computed
19.
Histol Histopathol ; 20(4): 1327-38, 2005 10.
Article in English | MEDLINE | ID: mdl-16136514

ABSTRACT

Articular cartilage is an avascular connective tissue in which the availability of oxygen and glucose is significantly lower than synovial fluid and plasma. Glucose is an important metabolic fuel and structural precursor that plays a key role in the synthesis of extracellular matrix macromolecules in articular cartilage. However, glucose concentrations in cartilage can fluctuate depending on age, physical activity and endocrine status. Chondrocytes are glycolytic cells and must be able to sense the quantities of oxygen and glucose available to them in the extracellular matrix and respond appropriately by adjusting cellular metabolism. Consequently chondrocytes must have the capacity to survive in an extracellular matrix with limited nutrients and low oxygen tensions. The molecular mechanisms responsible for allowing chondrocytes to adapt to these harsh environmental conditions are poorly understood. In this article we present a novel "dual" model of oxygen and glucose sensing in chondrocytes based on recent experimental data. This model incorporates the hypoxia-inducible factor alpha (HIF-1alpha) as an oxygen sensor and the hypoxia responsive facilitative glucose transporters, GLUT1 and GLUT3 as putative components of the glucose sensing apparatus in chondrocytes. Recent studies have shown that GLUT1 and GLUT3 are both expressed in chondrocytes and their HIF-1alpha-mediated transcription may be dually stimulated in response to hypoxia and low glucose conditions which in turn promote anaerobic glycolysis in favor of oxidative phosphorylation. This working model provides, for the first time, a unifying hypothesis to explain how chondrocytes might sense and respond to low oxygen tensions and alterations in extracellular glucose.


Subject(s)
Cartilage, Articular/cytology , Cartilage, Articular/physiology , Chondrocytes/physiology , Glucose Transporter Type 1/physiology , Glucose Transporter Type 3/physiology , Glucose/metabolism , Hypoxia-Inducible Factor 1/physiology , Oxygen/metabolism , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 3/genetics , Humans , Hypoxia-Inducible Factor 1/genetics , Models, Chemical
20.
Cell Biol Int ; 29(4): 249-60, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15943951

ABSTRACT

Glucose transport across the chondrocyte membrane is essential for chondrogenesis and the development of the skeletal system. We have previously used RT-PCR to show that fully developed human articular chondrocytes express transcripts for the GLUT1 and GLUT9 glucose transporters. In this study we report on the expression and immunohistochemical localization of the GLUT1 and GLUT9 proteins in embryonic and mature ovine cartilage. We also provide Western blot evidence for GLUT1 and GLUT9 expression in mature ovine chondrocytes. Ovine embryos (developmental stages E32 to E36 and E42 to E45) were obtained from pregnant ewes humanely killed by injection with sodium pentobarbitone. Embryos were fixed and processed for immunohistochemistry. Polyclonal antibodies to GLUT1 and GLUT9 revealed that both transporters are expressed in developing chondrocytes in ovine embryos and in the superficial, middle and deep layers of ovine cartilage from mature animals. GLUT1 expression was observed in erythrocytes and organs including heart, liver, and kidney. GLUT9 was also found in heart, kidney and liver. Western blotting confirmed the presence of the GLUT1 protein which migrated between the 50 and 64 kDa markers and two specific GLUT9 bands migrating under the 50 and 60 kDa markers, respectively. The presence of GLUT1 and GLUT9 in developing joints of ovine embryos suggests that these proteins may be important in glucose delivery to developing chondroblasts. Expression of these GLUT isoforms may be an important bioenergetic adaptation for chondrocytes in the extracellular matrix of developing cartilage.


Subject(s)
Chondrocytes/metabolism , Monosaccharide Transport Proteins/biosynthesis , Animals , Blotting, Western , Cartilage, Articular/cytology , Cartilage, Articular/embryology , Cartilage, Articular/metabolism , Female , Gene Expression Regulation, Developmental , Glucose Transporter Type 1 , Immunoenzyme Techniques , Sheep/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...