Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell Tissue Res ; 362(1): 9-20, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25920589

ABSTRACT

Lesions of the central nervous system elicit inflammatory responses that counteract the regeneration of neurites. Microglia and infiltrating macrophages that were activated by trauma have been identified as cellular sources of inhibitory factors. We examine cultured macrophage (RAW264.7) and neuronal (PC12) cell lines to ascertain the potential modulators of the inflammatory impact on neurons. By exposing quiescent macrophages to lipopolysaccharide (LPS) and interferon γ (IFN-γ), cells can be transformed into an activated M1 phenotype. Neurite extension was induced in PC12 cells by culturing them in the presence of nerve growth factor. Neurite outgrowth was quantified by analyzing immunofluorescence and phase contrast microscopy images. Activated macrophages significantly reduced neurite extension. Macrophage activation by LPS/IFN-γ induced a 1000-fold increase in tumor necrosis factor alpha (TNF-α) secretion, as quantified by enzyme-linked immunosorbent assays (ELISA). Recombinant TNF-α inhibited neurite formation at concentrations as low as 0.016 ng/ml. In contrast, the masking of TNF-α with specific functional antibodies abrogated neurite growth inhibition by activated macrophages. Taken together, these results indicated that TNF-α is a key component of inhibitory macrophage action. The transfection of PC12 neurons with microRNA-124 (miR-124) counteracted the inhibition of neurites mediated by both recombinant TNF-α and macrophages. miR-124 did not stimulate neurite formation per se, nor was cell viability affected. These data suggest that miR-124 might be a valuable tool for desensitizing neurons to a repulsive inflammatory environment.


Subject(s)
Inflammation/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , Neurites/metabolism , Animals , Inflammation/genetics , Inflammation/pathology , Mice , MicroRNAs/genetics , Neurites/pathology , PC12 Cells , RAW 264.7 Cells , Rats , Transfection , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
2.
Mol Pharm ; 12(9): 3146-57, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26181293

ABSTRACT

Excessive extracellular matrix formation in organs and tissues arises from an imbalance between the synthesis and degradation of matrix proteins, especially collagen. This condition interferes with proper wound healing and regeneration, and to date, no specific treatment is available. In the present study, we propose a targeted drug delivery system consisting of cell-specific immunoliposomes (ILs) loaded with deferoxamine (DFO) as an antifibrotic drug. ILs were functionalized with polyethylene glycol (PEG) to improve the steric stability and prolong their half-life. In addition, a single-chain Fv (scFv) antibody fragment that specifically targets fibroblast activation protein (FAP) was incorporated. An in vitro fibrosis model was employed to test this construct. This model consisted of highly activated pro-fibrotic fibroblasts with 2- to 6-fold induction of selected fibrosis markers: cell/matrix deposited collagen I, total soluble collagen, and α smooth muscle actin. The activation was accompanied by a significant and cell-specific elevation of FAP expression and activity, thereby confirming that FAP is an adequate target for antifibrotic drug delivery. Purified anti-FAP scFv was shown to bind specifically to these cells without influencing the FAP enzymatic activity. DFO was demonstrated to have a dose-dependent antifibrotic activity as quantified by collagen deposition. Specific binding and intracellular uptake of DiI-labeled ILs into the activated fibroblasts were shown by flow cytometry and microscopy. Finally, DFO-loaded ILs targeted to FAP caused a significant reduction in the collagen deposition, whereas no effect was observed using liposomes that lacked the targeting antibody fragment. These results suggest that the FAP-specific scFv-conjugated liposomes have considerable potential for cell-specific targeting applicable as a therapy for excessive collagen deposition during fibrosis. In general, through liposome encapsulation, bioactive molecules, such as DFO, that have broad effects and poor cell penetration can be converted into cell-specific composites for targeted drug delivery.


Subject(s)
Deferoxamine/administration & dosage , Fibroblasts/drug effects , Fibrosarcoma/drug therapy , Gelatinases/antagonists & inhibitors , Liposomes/chemistry , Lung/drug effects , Membrane Proteins/antagonists & inhibitors , Single-Chain Antibodies/administration & dosage , Cells, Cultured , Drug Delivery Systems , Endopeptidases , Fibroblasts/immunology , Fibroblasts/pathology , Fibrosarcoma/immunology , Fibrosarcoma/pathology , Gelatinases/immunology , Half-Life , Humans , Immunoenzyme Techniques , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/immunology , Liposomes/immunology , Lung/immunology , Lung/pathology , Membrane Proteins/immunology , Polyethylene Glycols/chemistry , Serine Endopeptidases/immunology , Siderophores/administration & dosage , Single-Chain Antibodies/immunology
3.
Circulation ; 125(5): 685-96, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22223428

ABSTRACT

BACKGROUND: CXCR4-positive bone marrow cells (BMCs) are critically involved in cardiac repair mechanisms contributing to preserved cardiac function. Stromal cell-derived factor-1 (SDF-1) is the most prominent BMC homing factor known to augment BMC engraftment, which is a limiting step of stem cell-based therapy. After myocardial infarction, SDF-1 expression is rapidly upregulated and promotes myocardial repair. METHODS AND RESULTS: We have established a bifunctional protein consisting of an SDF-1 domain and a glycoprotein VI (GPVI) domain with high binding affinity to the SDF-1 receptor CXCR4 and extracellular matrix proteins that become exposed after tissue injury. SDF1-GPVI triggers chemotaxis of CXCR4-positive cells, preserves cell survival, enhances endothelial differentiation of BMCs in vitro, and reveals proangiogenic effects in ovo. In a mouse model of myocardial infarction, administration of the bifunctional protein leads to enhanced recruitment of BMCs, increases capillary density, reduces infarct size, and preserves cardiac function. CONCLUSIONS: These results indicate that administration of SDF1-GPVI may be a promising strategy to treat myocardial infarction to promote myocardial repair and to preserve cardiac function.


Subject(s)
Cell- and Tissue-Based Therapy , Chemokine CXCL12/pharmacology , Heart/drug effects , Heart/physiopathology , Ischemic Attack, Transient/therapy , Myocardial Infarction/therapy , Platelet Membrane Glycoproteins/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Chemokine CXCL12/metabolism , Chemokine CXCL12/therapeutic use , Collagen/metabolism , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/physiopathology , Mice , Mice, Inbred C57BL , Models, Animal , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Neovascularization, Physiologic/drug effects , Platelet Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/therapeutic use , Protein Binding , Receptors, CXCR4/metabolism , Treatment Outcome
4.
Cell Mol Life Sci ; 67(14): 2451-65, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20352467

ABSTRACT

By counteracting the shortening of chromosome telomeres, telomerase reverse transcriptase (hTERT) prevents senescence and age-related cell death. Embryonic cells display a high telomerase activity that declines rapidly with cell differentiation. Conversely, de-differentiated tumor cells tend to re-express telomerase. In view of the controversial data on the reciprocal correlation between cell proliferation and differentiation, we questioned whether telomerase overexpression and the resulting immortalization would affect the functional phenotype of human endothelial cells. Our comparative analysis addressed (1) distinct cell adhesion to different ECM-proteins analyzed on miniaturized multisubstrate arrays (MSA), (2) protein expression of diverse markers, (3) the uptake of DiI-Ac-LDL, (4) the inflammatory response based on upregulation of ICAM-1, (5) tube formation, and (6) the barrier properties of cell monolayers in transfilter cultures. Our results, based on some 40 data sets, demonstrate that immortalization of primary endothelial cells by hTERT maintains the typical endothelial characteristics without any sign of functional de-differentiation.


Subject(s)
Cell Dedifferentiation , Endothelial Cells/cytology , Endothelial Cells/physiology , Telomerase/metabolism , Carbocyanines/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement , Cell Transformation, Neoplastic , Cells, Cultured , Cloning, Molecular , Humans , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/immunology , Lipoproteins, LDL/metabolism , Permeability , Proteins/metabolism , Telomerase/genetics , Transduction, Genetic , Tumor Necrosis Factor-alpha/immunology , Vascular Endothelial Growth Factor A/metabolism , von Willebrand Factor/metabolism
5.
Nano Lett ; 10(10): 3933-9, 2010 Oct 13.
Article in English | MEDLINE | ID: mdl-20795625

ABSTRACT

Microstructured 20 µm thick polymer filaments used as nerve implants were loaded with chitosan/siRNA nanoparticles to promote nerve regeneration and ensure local delivery of nanotherapeutics. The stable nanoparticles were rapidly internalized by cells and did not affect cell viability. Target mRNA was successfully reduced by 65-75% and neurite outgrowth was enhanced even in an inhibitory environment. This work, thus, supports the application of nanobiofunctionalized implants as a novel approach for spinal cord and nerve repair.


Subject(s)
Chitosan/chemistry , Nanoparticles/chemistry , Neurons/cytology , Prostheses and Implants , RNA, Small Interfering/administration & dosage , Animals , Cell Line , Nanoparticles/ultrastructure , Nerve Regeneration , Neurites/metabolism , Neurons/metabolism , Prostheses and Implants/ultrastructure , RNA, Small Interfering/genetics , Transfection , rhoA GTP-Binding Protein/genetics
6.
Cell Transplant ; 17(5): 543-8, 2008.
Article in English | MEDLINE | ID: mdl-18714673

ABSTRACT

The quantity of therapeutic gene products released from genetically engineered cells can be controlled externally at different levels. The widely used approach of controlling expression, however, generally has the disadvantage that chemical substances must be applied for stimulation. An alternative strategy aims at controlling gene products at posttranslational levels such as secretion. The secretion of a therapeutic agent can be regulated if the agent is targeted to the regulated secretory pathway and stored in the secretory granules until its release. In this article we address the question of whether the release of beta-endorphin, an opioid with a potent analgesic effect, could be induced by electrically stimulating stably transfected Neuro-2a cells. Throughout this study we used the human proopiomelanocortin (POMC) gene, which is the precursor molecule for human beta-endorphin. We analyzed its subcellular localization and found it in the regulated secretory pathway in Neuro-2a cells. Using electrical field stimulation we were able to identify a stimulation pattern that significantly increased the release of beta-endorphin-immunoreactive material, although to a limited extent. This result indicates that electrical stimulation of secretion could be used to manipulate the amount of a therapeutic agent released from transplanted cells.


Subject(s)
Neurons/metabolism , Secretory Pathway/physiology , beta-Endorphin/metabolism , Cell Line , Cell Transplantation/methods , Electric Stimulation , Genetic Engineering , Humans , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Protein Processing, Post-Translational , Tissue Engineering , beta-Endorphin/genetics
7.
Eur J Cell Biol ; 86(4): 207-19, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17307274

ABSTRACT

Schwann cells (SCs) can support the regeneration of lesioned fiber tracts of the peripheral and central nervous system and have been transplanted alone or in combination with synthetic nerve guides. For neuronal tissue engineering purposes, the cells must be isolated from small biopsies and expanded in vitro. In this study we analyze the impact of cell expansion on 9 different cell parameters, comparing short- and long-term cultured rat SCs, which we refer to as 'young' and 'old' or 'aged' cells, respectively. In comparison to young SCs, old SCs doubled the axonal outgrowth from dorsal root ganglion explants and displayed only one-third as much adhesion to the gray and white matter of spinal cord cryosections. In a 3-dimensional extracellular matrix the two cell populations showed very different cellular responses with regard to cell morphology and cell-cell adhesion. Cell proliferation of old SCs was independent of serum components and was not hampered by contact inhibition. In addition, population doubling times were reduced by a factor of almost three compared to those of young SCs. Despite considerable karyotype changes, with an average of 68.7 chromosomes versus 42 in native rat cells, old SCs did not show any increase in telomerase activity and loss of anchorage dependence--characteristics that are typical of tumor cells. The data also provide biological insights into which cell characteristics (proliferation and adhesion, for example) are functionally clustered and either change or remain constant with aging in vitro. Though the data indicate a lack of tumorigenic transformation coupled with increased neurite outgrowth-promoting activity after extensive SC expansion in vitro, thus suggesting better regeneration qualities, we strongly recommend that in vitro aged rat SCs (>11 passages) should not be employed for tissue engineering.


Subject(s)
Cellular Senescence , Nerve Regeneration , Neurons/physiology , Schwann Cells/physiology , Sciatic Nerve/physiology , Tissue Engineering , Animals , Animals, Newborn , Cell Adhesion , Cell Communication , Cell Proliferation , Cell Shape , Cells, Cultured , Chick Embryo , Coculture Techniques , Extracellular Matrix/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , Karyotyping , Neurites/physiology , Neurons/cytology , Rats , Rats, Inbred Lew , Schwann Cells/cytology , Schwann Cells/metabolism , Sciatic Nerve/cytology , Spinal Cord/metabolism , Telomerase/metabolism , Time Factors , Tissue Engineering/methods
8.
Biomaterials ; 28(36): 5536-43, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17889331

ABSTRACT

To overcome limitations on regeneration in the nervous system and other organs caused by insufficient blood supply, we have developed a gelatin sponge material which stimulates blood vessel formation, i.e. angiogenesis. Controlled chemical cross-linking was employed to slow down enzymatic degradation of the gelatin matrix. Four different in vitro assays using L929 fibroblasts and purified endothelial cells indicated that the sponge material did not release toxic components, but provided a permissive substratum for cell attachment, cell migration and pronounced cell proliferation, all of which are crucial for the formation of vasculature. Two in vivo models were employed to directly monitor the pro-angiogenic impact of the sponge material. Implantation of gelatin sponges onto the chorioallantoic membrane of fertilized chicken eggs induced robust attraction of endothelial cells and formation of blood vessels. Angiogenesis inside gelatin implants occurred more than 200 times faster than in a commercial collagen sponge. Similarly, after subcutaneous implantation of tube-like sponges into mice, an increasing immigration of cells and subsequent formation of functional vasculature became evident. Immunocytochemistry revealed no fibronection accumulation and no scarring. In summary, our matrix based on cross-linked gelatin promises to be a valuable component of future implants, improving neuronal and non-neuronal regeneration by concomitant pro-angiogenic stimulation.


Subject(s)
Cross-Linking Reagents/chemistry , Gelatin/chemistry , Gelatin/pharmacology , Neovascularization, Physiologic/drug effects , Animals , Cattle , Cells, Cultured , Chickens , Gelatin/metabolism , Materials Testing , Mice , Models, Animal
9.
Restor Neurol Neurosci ; 25(2): 131-41, 2007.
Article in English | MEDLINE | ID: mdl-17726272

ABSTRACT

PURPOSE: Recently we successfully used a conduit of epsilon-caprolactone-co-trimethylene carbonate filled with Schwann cells (SC) across a 20 mm gap in a rat median nerve. In this study we applied the tubes with SC across a 40 mm gap in order to analyse the regenerative potential of the tubes in long nerve defects. METHODS: To augment the nerve defect a cross-chest procedure was used and the tubes were implanted with injected isogeneic SCs inside (group 3). Both ulnar nerves were used for a 40 mm autograft (group 2). For control group non-operated animals were used (group 1). The grasping test, histology (S-100, PAM), electrophysiology, and the muscle weight were used to assess regeneration. RESULTS: After 12 months, grasping was seen only in three animals of group 3 (3.6 g [95% CI: 0 to 7.6 g]). However, in group 2 all rats had a partial functional regeneration (42.8 g [95% CI: 39.1 to 46.6 g]). The grasping force of the non-operated animals (group 1) was 240.9 g [95% CI: 237.2 to 244.7 g] at the time. Histology from group 3 confirmed an irregular arrangement of fibres in contrast to more organized structures in group 2. Electrophysiology in group 3 displayed potentials only in the three animals with functional regeneration. In group 2 all animals exhibited potentials. A significant decrease of muscle weight was observed in groups 2 and 3, most prominent in the latter. CONCLUSION: Regeneration was not successful across the 40 mm gap using the applied tube in combination with SC. For future experiments further consideration should be taken in optimizing the cellular and material components that are critical for a successful application to overcome very large nerve gaps.


Subject(s)
Bioartificial Organs , Nerve Regeneration/physiology , Neural Pathways , Schwann Cells/physiology , Action Potentials , Animals , Axons/ultrastructure , Electrophysiology , Equipment Design , Female , Foot , Forelimb , Median Nerve/physiology , Median Nerve/surgery , Median Nerve/ultrastructure , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/innervation , Muscle, Skeletal/physiology , Nerve Fibers/ultrastructure , Organ Size , Psychomotor Performance , Rats , Rats, Inbred Lew , Reaction Time , Transplantation, Autologous , Ulnar Nerve/physiology , Ulnar Nerve/transplantation
10.
J Tissue Eng Regen Med ; 11(10): 2699-2709, 2017 10.
Article in English | MEDLINE | ID: mdl-27225726

ABSTRACT

To improve the clinical performance of vascular prostheses, which is inacceptably low for implants with small diameters (< 6 mm), biofunctionalization of synthetic implants by endothelialization has become a major, although still unreached, aim. In order to be able to recruit native endothelial progenitor cells (EPCs) to luminal implant surfaces from the blood stream, we generated monoclonal antibodies against the EPC-specific vascular endothelial growth factor receptor 2 (VEGFR-2). Employing the very efficient genetic immunization strategy, > 10 000 hybridoma clones were generated. Screening with various deletion mutants of VEGFR-2, 49 highly-specific monoclonal antibodies (mAbs) covering all seven Ig domains of VEGFR-2 were selected. mAb 9H10 was characterized in detail. Once immobilized on synthetic surfaces, mAb 9H10 allowed, within min, nearly 100-fold enrichment of VEGFR-2-expressing cells under continuous flow conditions. Cell trapping was cell-type specific and essentially not affected by competing VEGFR-2-negative cells. To exclude that the antibody would adversely modify receptor responses, four different in vitro assays were employed. Cell proliferation, angiogenic tube formation, acetylated low-density lipoprotein uptake and VEGFR-2 phosphorylation remained unaffected, suggesting that the antibody did not interfere with the receptor functioning of human umbilical vascular endothelial cells. The molecular and cellular characteristics make the selected monoclonal antibody a very promising tool for the biofunctionalization of vascular implants. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Blood Vessel Prosthesis , Lymphocytes/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Antibodies/chemistry , Antibodies/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunization , Phosphorylation , Protein Domains , Species Specificity , Sus scrofa , Vascular Endothelial Growth Factor A
11.
Biomaterials ; 27(8): 1425-36, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16169587

ABSTRACT

The aim of neuro tissue engineering is to imitate biological features in order to enhance regeneration. Following lesions of peripheral nerves, Schwann cells (SCs) reorganize to form longitudinal bands of Büngner (boB) which function as guides for regrowing axons. In order to imitate boB in synthetic implants designed to bridge nerve lesions, we developed resorbable, semipermeable nerve guide conduits with microstructured internal polymer filaments. We utilized a novel microcell chip and identified three extracellular matrix components conducive for coating non-permissive polymer surfaces. In order to maximize SC alignment, seven different microtopographies were investigated via the silicon chip technology. Special longitudinal microgrooves directed SC orientation and growing axons of dorsal root ganglia (DRG), thus achieving stereotropism. When these results were applied to microgrooved polymer filaments inside nerve guide conduits, we observed highly oriented axon growth without meandering. Since scar-forming fibroblasts could potentially interfere with axonal regrowth, triple cultures with fibroblasts, SC and DRG were conducted. Fibroblasts positioned on the outer nanopore containing conduit wall, did not hamper neuronal and glial differentiation inside the tube. In summary, for more rapid regrowth, functional boB can be induced by guided microtissue engineering. By considering both the negative and positive effects of cell interactions, a more rational design of nerve implants becomes feasible.


Subject(s)
Biocompatible Materials , Nerve Tissue/physiology , Neuroglia/physiology , Tissue Engineering , Animals , Axons/physiology , Cell Communication/physiology , Cells, Cultured , Chick Embryo , Fibroblasts/physiology , Nerve Regeneration/physiology , Nerve Tissue/cytology , Neurites/physiology , Neuroglia/cytology , Rats , Rats, Inbred Lew , Schwann Cells/physiology , Tropism
12.
Int J Dev Neurosci ; 24(1): 65-72, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16338115

ABSTRACT

During the development of the central nervous system (CNS), the correct wiring of outgrowing neurites is mediated by antagonistic mechanisms. Aberrant growth is prevented by repulsive factors such as semaphorins. Expression of the ligands Sema3A and -3E and the receptors neuropilin Npn-1, -2a and -2b in the chick visual system were analyzed by RT-PCR. Whereas Sema3A and its major receptor Npn-1 were abundant, Sema3E and Npn-2 isoform expression was highly restricted and developmentally regulated. Peak expression occurred during retinal axon innervation of the tectum. Functional in vitro assays with recombinant proteins revealed a topography-specific growth cone collapsing activity of Sema3A for tectal axons. Interestingly, whereas tectal axons collapsed in a topographic-specific manner only in the presence of Sema3A, retinal axons responded only to Sema3E. The collapsing activity was intracellularly mediated by cGMP. For a detailed analysis of neuronal responses to sempahorins, time lapse video recording was performed. When tectal and retinal axons were pre-exposed to brain-derived neurotrophic factor (BDNF), a protective effect was evident only in the case of retinal axons. Our results suggest a molecular mechanism whereby ingrowth of retinal axons into the tectum can be regulated by Sema3E/BDNF modulation without disturbing tectal axon growth out of the tectum mediated by Sema3A.


Subject(s)
Avian Proteins/metabolism , Axons/metabolism , Central Nervous System/cytology , Semaphorin-3A/metabolism , Semaphorins/metabolism , Animals , Avian Proteins/genetics , Brain-Derived Neurotrophic Factor/metabolism , Chick Embryo , Neural Pathways/cytology , Neural Pathways/metabolism , Neuropilin-1/genetics , Neuropilin-1/metabolism , Neuropilin-2/genetics , Neuropilin-2/metabolism , Nucleotides, Cyclic/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Semaphorin-3A/genetics , Semaphorins/genetics , Signal Transduction/physiology , Superior Colliculi/cytology , Superior Colliculi/metabolism
13.
Carbohydr Polym ; 143: 25-34, 2016 Jun 05.
Article in English | MEDLINE | ID: mdl-27083340

ABSTRACT

Polyethylene glycol (PEG) is a widely used modification for drug delivery systems. It reduces undesired interaction with biological components, aggregation of complexes and serves as a hydrophilic linker of ligands for targeted drug delivery. However, PEGylation can also lead to undesired changes in physicochemical characteristics of chitosan/siRNA nanoplexes and hamper gene silencing. To address this conflicting issue, PEG-chitosan copolymers were synthesized with stepwise increasing degrees of PEG substitution (1.5% to 8.0%). Subsequently formed PEG-chitosan/siRNA nanoplexes were characterized physicochemically and biologically. The results showed that small ratios of chitosan PEGylation did not affect nanoplex stability and density. However, higher PEGylation ratios reduced nanoplex size and charge, as well as cell uptake and final siRNA knockdown efficiency. Therefore, we recommend fine-tuning of PEGylation ratios to generate PEG-chitosan/siRNA delivery systems with maximum bioactivity. The degree of PEGylation for chitosan/siRNA nanoplexes should be kept low in order to maintain optimal nanoplex efficiency.


Subject(s)
Chitosan/analogs & derivatives , Polyethylene Glycols/chemistry , RNA, Small Nuclear/administration & dosage , Cell Line, Tumor , Cell Survival , Chitosan/chemical synthesis , Chitosan/chemistry , Drug Carriers , Flow Cytometry , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Humans , Hydrogen-Ion Concentration , Indicators and Reagents , Nanoparticles , Oxazines/chemistry , Particle Size , Polyethylene Glycols/chemical synthesis , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/genetics , Solubility , Xanthenes/chemistry
14.
Invest Ophthalmol Vis Sci ; 46(3): 1047-53, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15728564

ABSTRACT

PURPOSE: To determine efflux systems of the outer blood-retina barrier (oBRB) and compare the oBRB with the blood-brain barrier (BBB). METHODS: Porcine oBRB structure and transport characteristics of freshly dissected intact tissue sheets were investigated with scanning electron microscopy, immunocytochemistry, vital dye labeling, and pharmacological agents, using HPLC/mass spectrometry. To compare drug permeation across the oBRB and the BBB, three different systems were used: (1) oBRB tissue sheets in a two-chamber device in vitro; (2) an in vitro BBB model composed of purified astrocytes and brain capillary endothelial cells on transfilter membranes; and (3) an in vivo model based on the brain-plasma ratio of drugs in mice. RESULTS: Efflux pumps (multidrug resistance protein [P-gp] and multidrug resistance-associated protein [MRP]) were demonstrated by antibody staining. Side-specific application of three P-gp and MRP substrates and selective transport inhibition suggested that both membrane proteins were preferentially located on the choroidal side of the oBRB. Therefore, the efflux was directed toward the blood, as in the BBB. To relate the transport characteristics of the oBRB to the BBB, up to nine different test compounds were used. The ranking of the permeability coefficients (P(e)) and the brain-plasma ratios of test compounds indicated that the oBRB has barrier and carrier features similar to those of the BBB in vitro and in vivo. CONCLUSIONS: Despite the fact that epithelial oBRB and endothelial BBB have developed as separate entities with many site-specific functions, their transport and permeation characteristics display surprising similarities, that include the polarized expression of the two major efflux pumps P-gp and MRP.


Subject(s)
Blood-Brain Barrier/physiology , Blood-Retinal Barrier/physiology , Brain/blood supply , Pigment Epithelium of Eye/physiology , Retinal Vessels/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Biological Transport, Active , Capillary Permeability , Cattle , Cerebrovascular Circulation/physiology , Chromatography, High Pressure Liquid , Coculture Techniques , Endothelium, Vascular/metabolism , Immunohistochemistry , Membrane Proteins/metabolism , Microscopy, Electron, Scanning , Multidrug Resistance-Associated Proteins/metabolism , Phosphoproteins/metabolism , Pigment Epithelium of Eye/ultrastructure , Swine , Zonula Occludens-1 Protein
15.
J Neurosurg ; 103(6): 1067-76, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16381194

ABSTRACT

OBJECT: In a rat model, nerve regeneration was evaluated across a 2-cm defect in the median nerve by using a resorbable artificial nerve conduit. The aim of this study was to develop an artificial, biocompatible nerve guide to induce regeneration in the peripheral nervous system. METHODS: The authors compared a nerve conduit of trimethylenecarbonate-co-epsilon-caprolactone (TMC/CL) filled with autologous Schwann cells with both an empty hollow conduit and an autologous nerve graft. Animals that did not undergo surgery served as the control group. Nerve regeneration was evaluated with the grasping test, histological analysis of the nerve, muscle weight analysis (flexor digitorum superficialis muscle), and electrophysiological examination. After an observation period of 9 months, regeneration occurred only in animals that had received an autologous graft or a Schwann cell containing nerve conduit. No signs of regeneration were found in animals supplied with the empty conduit. CONCLUSIONS: Results of this study reveal the important role of Schwann cells in the regeneration process across a 2-cm defect in the rat median nerve. Furthermore, Schwann cell-filled nerve conduits induced functional recovery, as demonstrated in the grasping test, that was comparable with that of the autologous graft 9 months after implantation.


Subject(s)
Absorbable Implants , Median Nerve/injuries , Median Nerve/physiopathology , Nerve Regeneration , Schwann Cells , Wounds, Penetrating/physiopathology , Animals , Electrophysiology , Female , Forelimb , Median Nerve/pathology , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/innervation , Neural Conduction , Organ Size , Rats , Rats, Inbred Lew , Wounds, Penetrating/pathology
16.
Brain Res ; 963(1-2): 321-6, 2003 Feb 14.
Article in English | MEDLINE | ID: mdl-12560139

ABSTRACT

A micro-structured, biodegradable, semipermeable hollow nerve guide implant was developed to bridge nerve lesions. Quantitative comparison of cell migration and axonal growth using time lapse video recording in vitro revealed that axons grow eight times faster than neuritotrophic Schwann cells migrate. To accelerate regeneration, purified Schwann cells are best injected into nerve guides before implantation. Nerve guides made from resorbable poly-lactide-co-glycolide support Schwann cell attachment, cell survival, and axonal outgrowth in vitro. The therapeutic concept aims at the development of an 'intelligent neuroprosthesis' that first mediates regeneration and then disappears.


Subject(s)
Absorbable Implants , Axons/physiology , Nerve Regeneration/physiology , Schwann Cells/physiology , Animals , Animals, Newborn , Biocompatible Materials , Cell Movement/drug effects , Cell Transplantation , Male , Polyesters , Rats , Rats, Wistar , Tissue Engineering
17.
PLoS One ; 9(1): e86740, 2014.
Article in English | MEDLINE | ID: mdl-24475174

ABSTRACT

In the field of regenerative medicine we aim to develop implant matrices for specific tissue needs. By combining two per se, cell-permissive gel systems with enzymatic crosslinkers (gelatin/transglutaminase and fibrinogen/thrombin) to generate a blend (technical term: quattroGel), an unexpected cell-selectivity evolved. QuattroGels were porous and formed cavities in the cell diameter range, possessed gelation kinetics in the minute range, viscoelastic properties and a mechanical strength appropriate for general cell adhesion, and restricted diffusion. Cell proliferation of endothelial cells, chondrocytes and fibroblasts was essentially unaffected. In contrast, on quattroGels neither endothelial cells formed vascular tubes nor did primary neurons extend neurites in significant amounts. Only chondrocytes differentiated properly as judged by collagen isoform expression. The biophysical quattroGel characteristics appeared to leave distinct cell processes such as mitosis unaffected and favored differentiation of sessile cells, but hampered differentiation of migratory cells. This cell-type selectivity is of interest e.g. during articular cartilage or invertebral disc repair, where pathological innervation and angiogenesis represent adverse events in tissue engineering.


Subject(s)
Cells/metabolism , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Regenerative Medicine/instrumentation , Regenerative Medicine/methods , Tissue Scaffolds/chemistry , Viscoelastic Substances/chemistry , Analysis of Variance , Animals , Biomechanical Phenomena , Bromodeoxyuridine , Cell Culture Techniques/methods , Cells, Cultured , Cross-Linking Reagents/metabolism , DNA Primers/genetics , Diffusion , Gene Expression Profiling , Humans , Kinetics , Mice , Osmolar Concentration , Rats , Regenerative Medicine/trends , Rheology , Tissue Engineering/methods
18.
J Control Release ; 168(3): 289-97, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23562632

ABSTRACT

Binding, stabilizing and promoting cellular uptake of siRNA are all critical efforts in creating matrices for the localized delivery of siRNA molecules to target cells. In this study, we describe the generation of chitosan imidazole/siRNA nanoplexes (NPs) embedded in nano scope polyelectrolyte multilayers (PEMs) composed of hyaluronic acid and chitosan for sustained and localized drug delivery. Regular PEM build-up, successful integration of NPs and controlled release under physiological conditions were shown. Biological efficacy was evaluated in neuronal cell culture concerning cell adhesion, viability, NPs uptake and gene silencing. The additionally shown biological functionalization of neuronal implants possesses potential for future applications in the field of regenerative medicine and treatment of spinal cord injuries.


Subject(s)
Chitosan/chemistry , Hyaluronic Acid/chemistry , Imidazoles/chemistry , Nanostructures/administration & dosage , Prostheses and Implants , RNA, Small Interfering/administration & dosage , Animals , Cell Adhesion/drug effects , Cell Survival/drug effects , Gene Knockdown Techniques , Nanostructures/chemistry , Neurons , PC12 Cells , RNA, Small Interfering/chemistry , Rats , rhoA GTP-Binding Protein/genetics
19.
Exp Biol Med (Maywood) ; 236(6): 692-700, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21558092

ABSTRACT

Telomerase reverse transcriptase (TERT) is fundamental in determining the life span by regulating telomere length of chromosomes. To address the question whether the enhancement of the proliferative potential hampers cell differentiation, we generated TERT-over-expressing endothelial cells (ECs) and analyzed in vitro their (1) barrier function; (2) low-density lipoprotein uptake; (3) expression pattern of six selected marker proteins; (4) angiogenic potential in four assays; and (5) inflammatory responses. In contrast to investigations with focus on other cell parameters, we demonstrate that immortalization of ECs by over-expression of TERT resulted in different angiogenic and inflammatory behavior in comparison to cells with low native telomerase levels.


Subject(s)
Endothelial Cells/enzymology , Endothelial Cells/physiology , Telomerase/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Gene Expression , Gene Expression Profiling , Humans , Inflammation Mediators/metabolism , Lipoproteins, LDL/metabolism , Neovascularization, Physiologic , Telomerase/genetics
20.
Neurosci Lett ; 494(1): 14-8, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21352894

ABSTRACT

The manipulation of gene expression by RNA interference could play a key role in future neurotherapies, for example in the development of biohydrid implants to bridge nerve and spinal cord lesion gaps. Such resorbable biomaterial prostheses could serve as growth substrates together with specific siRNA to foster neuronal regeneration. To the best of our knowledge, we are the first to biofunctionalize neuronal prostheses with siRNA. We analyzed neuronal and Schwann cell responses to scrambled siRNA coated polydioxanone polymer filaments designed to imitate pro-regenerative bands of Büngner for oriented axonal regrowth. With a view to future clinical applications we were especially interested in potentially detrimental side effects. We employed a variety of in vitro methods, including a novel impedance electrode microchamber assay, fluorescence and scanning electron microscopy, metabolic labeling and RT-PCR. We found that the application of chitosan/siRNA nanoparticles (1) did not affect glial cell motility or (2) axonal growth in contrast to other formulations, (3) only slightly reduced proliferation, and (4) did not induce inflammatory responses that might hamper axonal regeneration. The data suggest that chitosan/siRNA nanoparticle-coated polymer filaments are suitable for use in biohybrid implants with no significant side effects on neuronal and glial cells.


Subject(s)
Axons/physiology , Neurons/physiology , RNA, Small Interfering/administration & dosage , Analysis of Variance , Animals , Biocompatible Materials , Chitosan , Immunohistochemistry , Nanoparticles , RNA, Small Interfering/genetics , Rats , Rats, Inbred Lew , Schwann Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL