Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Neurosci ; 59(2): 308-315, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38086536

ABSTRACT

Multiple system atrophy (MSA) is a rare and rapidly progressive atypical parkinsonian disorder characterized by oligodendroglial cytoplasmic inclusions containing α-synuclein (α-syn), demyelination, inflammation and neuronal loss. To date, no disease-modifying therapy is available. Targeting α-syn-driven oligodendroglial dysfunction and demyelination presents a potential therapeutic approach for restricting axonal dysfunction, neuronal loss and disease progression. The present study investigated the promyelinogenic potential of sobetirome, a blood-brain barrier permeable and central nervous system selective thyromimetic in the context of an in vitro MSA model. Oligodendrocyte precursor cells (OPCs) were obtained from transgenic mice overexpressing human α-syn specifically in oligodendrocytes (MBP29 mouse line), a well-described MSA model, and non-transgenic littermates. mRNA and protein expression analyses revealed a substantial rescue effect of sobetirome on myelin-specific proteins in control and α-syn overexpressing oligodendrocytes. Furthermore, myelination analysis using nanofibres confirmed that sobetirome increases both the length and number of myelinated segments per oligodendrocyte in primary murine α-syn overexpressing oligodendrocytes and their respective control. These results suggest that sobetirome may be a promising thyromimetic compound targeting an important neuropathological hallmark of MSA.


Subject(s)
Demyelinating Diseases , Multiple System Atrophy , Phenols , Mice , Humans , Animals , Multiple System Atrophy/drug therapy , Multiple System Atrophy/genetics , Multiple System Atrophy/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Acetates/metabolism , Mice, Transgenic , Oligodendroglia/metabolism , Demyelinating Diseases/metabolism , Disease Models, Animal
2.
BMC Mol Cell Biol ; 24(1): 18, 2023 May 15.
Article in English | MEDLINE | ID: mdl-37189080

ABSTRACT

BACKGROUND: For the purpose of skeletal muscle engineering, primary myoblasts (Mb) and adipogenic mesenchymal stem cells (ADSC) can be co-cultured and myogenically differentiated. Electrospun composite nanofiber scaffolds represent suitable matrices for tissue engineering of skeletal muscle, combining both biocompatibility and stability Although growth differentiation factor 11 (GDF11) has been proposed as a rejuvenating circulating factor, restoring skeletal muscle function in aging mice, some studies have also described a harming effect of GDF11. Therefore, the aim of the study was to analyze the effect of GDF11 on co-cultures of Mb and ADSC on poly-ε-caprolactone (PCL)-collagen I-polyethylene oxide (PEO)-nanofibers. RESULTS: Human Mb were co-cultured with ADSC two-dimensionally (2D) as monolayers or three-dimensionally (3D) on aligned PCL-collagen I-PEO-nanofibers. Differentiation media were either serum-free with or without GDF11, or serum containing as in a conventional differentiation medium. Cell viability was higher after conventional myogenic differentiation compared to serum-free and serum-free + GDF11 differentiation as was creatine kinase activity. Immunofluorescence staining showed myosine heavy chain expression in all groups after 28 days of differentiation without any clear evidence of more or less pronounced expression in either group. Gene expression of myosine heavy chain (MYH2) increased after serum-free + GDF11 stimulation compared to serum-free stimulation alone. CONCLUSIONS: This is the first study analyzing the effect of GDF11 on myogenic differentiation of Mb and ADSC co-cultures under serum-free conditions. The results of this study show that PCL-collagen I-PEO-nanofibers represent a suitable matrix for 3D myogenic differentiation of Mb and ADSC. In this context, GDF11 seems to promote myogenic differentiation of Mb and ADSC co-cultures compared to serum-free differentiation without any evidence of a harming effect.


Subject(s)
Mesenchymal Stem Cells , Nanofibers , Humans , Mice , Animals , Tissue Scaffolds , Polyethylene/metabolism , Polyethylene/pharmacology , Polyesters/metabolism , Polyesters/pharmacology , Mesenchymal Stem Cells/metabolism , Myoblasts/metabolism , Cell Differentiation , Polyethylene Glycols/metabolism , Polyethylene Glycols/pharmacology , Collagen/metabolism , Collagen/pharmacology , Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factors/metabolism
3.
Cells ; 11(23)2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36497034

ABSTRACT

Electrospun nanofibers represent an ideal matrix for the purpose of skeletal muscle tissue engineering due to their highly aligned structure in the nanoscale, mimicking the extracellular matrix of skeletal muscle. However, they often consist of high-density packed fibers, which might impair vascularization. The integration of polyethylene oxide (PEO) sacrificial fibers, which dissolve in water, enables the creation of less dense structures. This study examines potential benefits of poly-ε-caprolactone-collagen I-PEO-nanoscaffolds (PCP) in terms of neovascularization and distribution of newly formed vessels compared to poly-ε-caprolactone -collagen I-nanoscaffolds (PC) in a modified arteriovenous loop model in the rat. For this purpose, the superficial inferior epigastric artery and vein as well as a motor nerve branch were integrated into a multilayer three-dimensional nanofiber scaffold construct, which was enclosed by an isolation chamber. Numbers and spatial distribution of sprouting vessels as well as macrophages were analyzed via immunohistochemistry after two and four weeks of implantation. After four weeks, aligned PC showed a higher number of newly formed vessels, regardless of the compartments formed in PCP by the removal of sacrificial fibers. Both groups showed cell influx and no difference in macrophage invasion. In this study, a model of combined axial vascularization and neurotization of a PCL-collagen I-nanofiber construct could be established for the first time. These results provide a foundation for the in vivo implantation of cells, taking a major step towards the generation of functional skeletal muscle tissue.


Subject(s)
Nanofibers , Tissue Scaffolds , Rats , Animals , Tissue Scaffolds/chemistry , Polyesters/chemistry , Nanofibers/chemistry , Collagen/chemistry , Collagen Type I , Polyethylene Glycols/chemistry
4.
J Vis Exp ; (188)2022 10 06.
Article in English | MEDLINE | ID: mdl-36282700

ABSTRACT

With the evolution of suture materials, there has been a change in paradigms in primary and secondary tendon repair. Improved mechanical properties allow more aggressive rehabilitation and earlier recovery. However, for the repair to hold against higher mechanical demands, more advanced suturing and knotting techniques must be assessed in combination with those materials. In this protocol, the use of polytetrafluoroethylene (PTFE) as a suture material in combination with different repair techniques was investigated. In the first part of the protocol, both linear tension strength and elongation of knotted against not-knotted strands of three different materials used in flexor tendon repair were evaluated. The three different materials are polypropylene (PPL), ultra-high molecular weight polyethylene with a braided jacket of polyester (UHMWPE), and polytetrafluoroethylene (PTFE). In the next part (ex vivo experiments with cadaveric flexor tendons), the behavior of PTFE using different suture techniques was assessed and compared with PPL and UHMWPE. This experiment is comprised of four steps: harvesting of the flexor tendons from fresh cadaveric hands, transection of the tendons in a standardized manner, tendon repair by four different techniques, mounting, and measurement of the tendon repairs on a standard linear dynamometer. The UHMWPE and PTFE showed comparable mechanical properties and were significantly superior to PPL in terms of linear traction strength. Repairs with four- and six-strand techniques proved stronger than two-strand techniques. Handling and knotting of PTFE are a challenge due to very low surface friction but fastening of the four- or six-strand repair is comparatively easy to achieve. Surgeons routinely use PTFE suture material in cardiovascular surgery and breast surgery. The PTFE strands are suitable for use in tendon surgery, providing a robust tendon repair so that early active motion regimens for rehabilitation can be applied.


Subject(s)
Polytetrafluoroethylene , Tendon Injuries , Humans , Tendon Injuries/surgery , Polypropylenes , Tensile Strength , Sutures , Suture Techniques , Tendons , Polyesters , Cadaver , Biomechanical Phenomena
5.
Cells ; 11(9)2022 04 24.
Article in English | MEDLINE | ID: mdl-35563742

ABSTRACT

For the purpose of skeletal muscle tissue engineering, different cell types have been investigated regarding their myogenic differentiation potential, including co-cultured myoblasts and adipogenic mesenchymal stromal cells (Mb/ADSC). As neural cells enhance synaptic junction formation, the aim of this study was to co-culture Schwann cells (SCs) with Mb/ADSC on biocompatible electrospun aligned poly-ε-polycaprolacton (PCL)-collagen I-nanofibers. It was hypothesized that SCs, as part of the peripheral nervous system, promote the myogenic differentiation of Mb/ADSC co-cultures. Mb/ADSC were compared to Mb/ADSC/SC regarding their capacity for myogenic differentiation via immunofluorescent staining and gene expression of myogenic markers. Mb/ADSC/SC showed more myotubes after 28 days of differentiation (p ≤ 0.05). After 28 days of differentiation on electrospun aligned PCL-collagen I-nanofibers, gene expression of myosin heavy chains (MYH2) and myogenin (MYOG) was upregulated in Mb/ADSC/SC compared to Mb/ADSC (p ≤ 0.01 and p ≤ 0.05, respectively). Immunofluorescent staining for MHC showed highly aligned multinucleated cells as possible myotube formation in Mb/ADSC/SC. In conclusion, SCs promote myogenic differentiation of Mb/ADSC. The co-culture of primary Mb/ADSC/SC on PCL-collagen I-nanofibers serves as a physiological model for skeletal muscle tissue engineering, applicable to future clinical applications.


Subject(s)
Mesenchymal Stem Cells , Nanofibers , Caproates , Collagen/metabolism , Collagen Type I/metabolism , Lactones , Mesenchymal Stem Cells/metabolism , Myoblasts/metabolism , Schwann Cells
6.
Int J Mol Sci ; 23(5)2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35269908

ABSTRACT

Due to its ability to reduce scarring and inflammation, human amniotic membrane is a widely used graft for wound dressings after corneal surgery. To overcome donor dependency and biological variances in the donor tissue, artificial nanofibrous grafts acting as drug carrier systems are promising substitutes. Electrospun nanofibrous scaffolds seem to be an appropriate approach as they offer the properties of permeable scaffolds with a high specific surface, the latter one depending on the fiber diameter. Electrospun scaffolds with fiber diameter of 35 nm, 113 nm, 167 nm and 549 nm were manufactured and coated by the layer-by-layer (LbL) technology with either hyaluronic acid or heparin for enhanced regeneration of corneal tissue after surgery. Studies on drug loading capacity and release kinetics defined a lower limit for nanofibrous scaffolds for effective drug loading. Additionally, scaffold characteristics and resulting mechanical properties from the application-oriented characterization of suture pullout from suture retention tests were examined. Finally, scaffolds consisting of nanofibers with a mean fiber diameter of 113 nm were identified as the best-performing scaffolds, concerning drug loading efficiency and resistance against suture pullout.


Subject(s)
Hyaluronic Acid , Nanofibers , Bandages , Drug Carriers , Heparin/pharmacology , Humans , Hyaluronic Acid/pharmacology , Nanofibers/therapeutic use , Polyesters , Tissue Engineering , Tissue Scaffolds
7.
Nanomaterials (Basel) ; 11(12)2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34947541

ABSTRACT

The transparency of nanofibrous scaffolds is of highest interest for potential applications like corneal wound dressings in corneal tissue engineering. In this study, we provide a detailed analysis of light transmission through electrospun polycaprolactone (PCL) scaffolds. PCL scaffolds were produced via electrospinning, with fiber diameters in the range from (35 ± 13) nm to (167 ± 35) nm. Light transmission measurements were conducted using UV-vis spectroscopy in the range of visible light and analyzed with respect to the influence of scaffold thickness, fiber diameter, and surrounding medium. Contour plots were compiled for a straightforward access to light transmission values for arbitrary scaffold thicknesses. Depending on the fiber diameter, transmission values between 15% and 75% were observed for scaffold thicknesses of 10 µm. With a decreasing fiber diameter, light transmission could be improved, as well as with matching refractive indices of fiber material and medium. For corneal tissue engineering, scaffolds should be designed as thin as possible and fabricated from polymers with a matching refractive index to that of the human cornea. Concerning fiber diameter, smaller fiber diameters should be favored for maximizing graft transparency. Finally, a novel, semi-empirical formulation of light transmission through nanofibrous scaffolds is presented.

8.
Sci Rep ; 11(1): 18858, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34552187

ABSTRACT

Posterior lamellar transplantation of the eye' s cornea (DSAEK, DMEK) currently is the gold standard for treating patients with corneal endothelial cell and back surface pathologies resulting in functional impairment. An artificial biomimetic graft carrying human corneal endothelium could minimize the dependency on human donor corneas giving access to this vision-restoring surgery to large numbers of patients, thus reducing current long waiting lists. In this study, four groups of electrospun nanofibrous scaffolds were compared: polycaprolactone (PCL), PCL/collagen, PCL/gelatin and PCL/chitosan. Each of the scaffolds were tissue-engineered with human corneal endothelial cells (HCEC-B4G12) and analyzed with regard to their potential application as artificial posterior lamellar grafts. Staining with ZO-1 and Na+/K+-ATPase antibodies revealed intact cell functionalities. It could be shown, that blending leads to decreasing contact angle, whereby a heterogeneous blend morphology could be revealed. Scaffold cytocompatibility could be confirmed for all groups via live/dead staining, whereby a significant higher cell viability could be observed for the collagen and gelatine blended matrices with 97 ± 3% and 98 ± 2% living cells respectively. TEM images show the superficial anchoring of the HCECs onto the scaffolds. This work emphasizes the benefit of blended PCL nanofibrous scaffolds for corneal endothelial keratoplasty.


Subject(s)
Endothelium, Corneal/cytology , Nanofibers/chemistry , Polyesters/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Biocompatible Materials , Cell Line , Chitosan/chemistry , Collagen/chemistry , Gelatin/chemistry , Humans , Nanofibers/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...