Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(4)2021 Feb 06.
Article in English | MEDLINE | ID: mdl-33562136

ABSTRACT

Nanoparticle (NP)-assisted procedures including laser tissue soldering (LTS) offer advantages compared to conventional microsuturing, especially in the brain. In this study, effects of polymer-coated silica NPs used in LTS were investigated in human brain endothelial cells (ECs) and blood-brain barrier models. In the co-culture setting with ECs and pericytes, only the cell type directly exposed to NPs displayed a time-dependent internalization. No transfer of NPs between the two cell types was observed. Cell viability was decreased relatively to NP exposure duration and concentration. Protein expression of the nuclear factor ĸ-light-chain-enhancer of activated B cells and various endothelial adhesion molecules indicated no initiation of inflammation or activation of ECs after NP exposure. Differentiation of CD34+ ECs into brain-like ECs co-cultured with pericytes, blood-brain barrier (BBB) characteristics were obtained. The established endothelial layer reduced the passage of integrity tracer molecules. NP exposure did not result in alterations of junctional proteins, BBB formation or its integrity. In a 3-dimensional setup with an endothelial tube formation and tight junctions, barrier formation was not disrupted by the NPs and NPs do not seem to cross the blood-brain barrier. Our findings suggest that these polymer-coated silica NPs do not damage the BBB.


Subject(s)
Blood-Brain Barrier/drug effects , Cerebral Revascularization/methods , Endothelial Cells/metabolism , Nanoparticles/metabolism , Polymers/pharmacology , Silicon Dioxide/pharmacology , Animals , B-Lymphocytes/immunology , Biological Transport/physiology , Blood-Brain Barrier/physiology , Brain/blood supply , Brain/cytology , Brain/metabolism , Cattle , Cell Survival/drug effects , Cells, Cultured , Humans , Laser Therapy/methods , Lymphocyte Activation/immunology , NF-kappa B/metabolism , Pericytes/metabolism
2.
Toxicol In Vitro ; 68: 104963, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32777439

ABSTRACT

The use of body-worn wireless devices with different communication protocols and rapidly changing exposure scenarios is still multiplying and the need to identify possible health effects of radiofrequency electromagnetic field (RF-EMF) exposure with extremely low-frequency (ELF) modulation envelops. In this study, effects of ELF-modulated 935 MHz RF-EMF on apoptosis, autophagy, oxidative stress and electron exchange in N9 microglial and SH-SY5Y neuroblastoma cells were investigated. Cells were exposed at 4 W/kg or sham-exposed for 2 and 24 h. RF-EMF exposure of both cell types did not alter apoptosis, the number of living cells nor the apoptosis-inducing factor (AIF), irrespective of the exposure duration. RF-EMF exposure for 24, but not for 2 h, increased protein levels of the autophagy marker ATG5, whereas LC3B-I and II and pERK were not altered in both cell types and exposure times investigated. A transient increase in glutathione (GSH), but not hydrogen peroxide and cytochrome c oxidase was found only in SH-SY5Y cells, indicating that short-time RF-EMF at SAR levels accepted by today's safety guidelines might cause autophagy and oxidative stress with the effect being dependent on cell type and exposure duration. Further studies are needed to evaluate possible underlying mechanisms involved in pulse-modulated RF-EMF exposure.


Subject(s)
Electromagnetic Fields , Radio Waves , Animals , Apoptosis , Autophagy , Autophagy-Related Protein 5/metabolism , Cell Line , Electron Transport , Electron Transport Complex IV/metabolism , Glutathione/metabolism , Humans , Hydrogen Peroxide/metabolism , Mice , Microglia/metabolism , Neuroblastoma/metabolism , Oxidative Stress
3.
CNS Neurol Disord Drug Targets ; 19(8): 618-629, 2020.
Article in English | MEDLINE | ID: mdl-32640966

ABSTRACT

BACKGROUND: The human neuroblastoma cell line, SH-SY5Y, has been widely used in neuroscience research, especially in studies related to Parkinson's disease. However, differences between clones have been demonstrated, highlighting the importance to characterize the properties of this cell line carefully. OBJECTIVE: The aim of this study was to characterize the phenotype of undifferentiated and differentiated SH-SY5Y cells using various differentiation protocols. METHODS: A morphological and quantitative analysis of markers related to dopaminergic and cholinergic neurons, but also other phenotypes, was performed. RESULTS: Differentiated cells showed the typical neuronal morphology. Undifferentiated cells expressed low levels of Tyrosine Hydroxylase (TH) and higher levels of the high-affinity Choline Transporter (CHT1). Staurosporine (ST)-differentiation resulted in the highest number of THimmunoreactive cells, followed by phorbol ester Phorbol-12-Myristate-13-Acetate (PMA), whereas differentiation with Brain-Derived Neurotrophic Factor (BDNF) did not increase TH-immunoreactive cells. TH, dopamine ß-hydroxylase and vesicular monoamine transporter-2 were also significantly upregulated in ST-differentiated cells compared to both undifferentiated and Retinoic Acid (RA)- differentiated cells. RA induced the highest number of CHT1-immunoreactive cells while ST- and BDNF-differentiation reduced CHT1-immunoreactive cells, indicating a decrease in the cholinergic phenotype. The presynaptic neuronal protein, α-synuclein, was significantly upregulated in RA- and ST-treated cells compared to undifferentiated cells. Ascorbic acid increased the number of CHT1-immunoreactive cells in all differentiation procedures and ST-differentiated TH-positive cells significantly. CONCLUSION: Our findings indicate that a quantitative characterization of the phenotype is crucial when using SH-SY5Y cells to study the pathogenesis or evaluate compounds for treatment of neurodegenerative diseases.


Subject(s)
Biomarkers/metabolism , Cell Differentiation/physiology , Phenotype , Cell Line, Tumor , Cholinergic Neurons/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Humans , Neuroblastoma/pathology , Parkinson Disease/metabolism , Signal Transduction , Tretinoin , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation
4.
Brain Res ; 1720: 146330, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31299185

ABSTRACT

Transplantation of stem and progenitor cells offers a promising tool for brain repair in the context of neuropathological disorders including Parkinson's disease. There is growing proof that the capacity of adult stem and progenitor cells for tissue regeneration relies rather on the release of paracrine factors than on their cell replacement properties. In line with this notion, we have previously reported that conditioned medium (CM) collected from cultured Endothelial Progenitor Cells (EPC) stimulated survival of striatal neurons. In the present study we investigated whether EPC-CM promotes survival of cultured midbrain progenitor cells. For that purpose primary cultures from fetal rat embryonic ventral mesencephalon (VM) were prepared and grown for 7 days in vitro (DIV). EPC-CM was administered from DIV5-7. First, we found that EPC-CM treatment resulted in significantly increased cell densities of TH-ir neurons. Interestingly, this effect was no longer seen after proteolytic digestion of the EPC-CM. EPC-CM also significantly increased densities of beta-III-tubulin positive neurons and lba-1-ir microglial cells. The effect on dopaminergic neurons was not due to higher cell proliferation as no incorporation of EdU was observed in TH-ir cells. Importantly, EPC-CM exerted neuroprotection against MPP+ induced toxicity as in vitro model of Parkinson's disease. Taken together, our findings identified EPC-CM as a powerful tool to promote survival of cultured VM neurons and further support the importance of paracrine factors in the actions of stem and progenitor cells for brain repair.


Subject(s)
Culture Media, Conditioned/pharmacology , Endothelial Progenitor Cells/metabolism , Mesencephalon/metabolism , Animals , Brain/metabolism , Cell Line , Cell Survival/drug effects , Cells, Cultured , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Neuroprotection/drug effects , Neuroprotective Agents/metabolism , Rats , Stem Cells/drug effects , Tyrosine 3-Monooxygenase/metabolism
5.
Beilstein J Nanotechnol ; 10: 941-954, 2019.
Article in English | MEDLINE | ID: mdl-31165021

ABSTRACT

Nanomedicine is a constantly expanding field, facilitating and improving diagnosis and treatment of diseases. As nanomaterials are foreign objects, careful evaluation of their toxicological and functional aspects prior to medical application is imperative. In this study, we aimed to determine the effects of gold and polymer-coated silica nanoparticles used in laser tissue soldering on brain endothelial cells and the blood-brain barrier using rat brain capillary endothelial cells (rBCEC4). All types of nanoparticles were taken up time-dependently by the rBCEC4 cells, albeit to a different extent, causing a time- and concentration-dependent decrease in cell viability. Nanoparticle exposure did not change cell proliferation, differentiation, nor did it induce inflammation. rBCEC4 cells showed blood-brain barrier characteristics including tight junctions. None of the nanoparticles altered the expression of tight junctions or impaired the blood-brain barrier permeability. The findings suggest that effects of these nanoparticles on the metabolic state of cells have to be further characterized before use for medical purposes.

6.
J Nanobiotechnology ; 17(1): 46, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30935413

ABSTRACT

BACKGROUND: Silica-ε-polycaprolactone-nanoparticles (SiPCL-NPs) represent a promising tool for laser-tissue soldering in the brain. After release of the SiPCL-NPs in the brain, neuronal differentiation might be modulated. The present study was performed to determine effects of SiPCL-NP-exposure at different stages of neuronal differentiation in neuron-like SH-SY5Y cells. The resulting phenotypes were analyzed quantitatively and signaling pathways involved in neuronal differentiation and degeneration were studied. SH-SY5Y cells were differentiated with all-trans retinoic acid or staurosporine to obtain predominantly cholinergic or dopaminergic neurons. The resulting phenotype was analyzed at the end of differentiation with and without the SiPCL-NPs given at various times during differentiation. RESULTS: Exposure to SiPCL-NPs before and during differentiation led to a decreased cell viability of SH-SY5Y cells depending on the differentiation protocol used. SiPCL-NPs co-localized with the neuronal marker ß-3-tubulin but did not alter the morphology of these cells. A significant decrease in the number of tyrosine hydroxylase (TH) immunoreactive neurons was found in staurosporine-differentiated cells when SiPCL-NPs were added at the end of the differentiation. TH-protein expression was also significantly downregulated when SiPCL-NPs were applied in the middle of differentiation. Protein expression of the marker for the dopamine active transporter (DAT) was not affected by SiPCL-NPs. SiPCL-NP-exposure predominantly decreased the expression of the high-affinity choline transporter 1 (CHT1) when the NPs were given before the differentiation. Pathways involved in neuronal differentiation, namely Akt, MAP-K, MAP-2 and the neurodegeneration-related markers ß-catenin and GSK-3ß were not altered by NP-exposure. CONCLUSIONS: The decrease in the number of dopaminergic and cholinergic cells may implicate neuronal dysfunction, but the data do not provide evidence that pathways relevant for differentiation and related to neurodegeneration are impaired.


Subject(s)
Cholinergic Neurons/drug effects , Dopaminergic Neurons/drug effects , Nanoparticles/toxicity , Polyesters/toxicity , Silicon Dioxide/toxicity , Cell Differentiation , Cell Line, Tumor , Cell Survival/drug effects , Cholinergic Neurons/cytology , Cholinergic Neurons/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Humans , Nanoparticles/chemistry , Phenotype , Polyesters/chemistry , Signal Transduction , Silicon Dioxide/chemistry , Staurosporine/pharmacology , Tretinoin/pharmacology
7.
Cell Transplant ; 27(4): 648-653, 2018 04.
Article in English | MEDLINE | ID: mdl-29701077

ABSTRACT

Idiopathic Parkinson's disease (PD) is a progressive neurodegenerative disorder, clinically manifested by cardinal motor symptoms including tremor at rest, bradykinesia, and muscle rigidity. Transplantation of dopaminergic (DAergic) neurons is an experimental therapy for PD, however, it is limited by suboptimal integration and low survival of grafts. Pretreatment of donor tissue may offer a strategy to improve properties of transplanted DAergic neurons and thereby clinical outcome. We have previously shown that a combination of neurotrophin-4/5 (NT-4/5) and glial cell line-derived neurotrophic factor (GDNF) demonstrated additive effects on rat ventral mesencephalic (VM) tissue. The present study investigated the effects of NT-4/5 and GDNF as single factors, or in combination on DAergic neurons, in organotypic explant cultures of fetal human ventral mesencephalon. For that purpose, free-floating roller-tube cultures were prepared from VM and the equally sized pieces grown for 1 week in the presence or absence of neurotrophic factors. Both neurotrophic factors increased dopamine content in the culture medium and in the number of tyrosine hydroxylase immunoreactive neurons, most prominently after combined GDNF + NT-4/5 treatment. Culture volumes did not differ between groups while content of lactate dehydrogenase in the culture medium was moderately reduced in all treated groups. In conclusion, we identified that a combination of GDNF and NT-4/5 robustly promoted differentiation and survival of human fetal VM DAergic neurons, an observation with potential promising impact for cell replacement approaches in PD.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Nerve Growth Factors/pharmacology , Neural Stem Cells/cytology , Substantia Nigra/cytology , Cells, Cultured , Humans , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Tyrosine 3-Monooxygenase/metabolism
8.
J Nanobiotechnology ; 15(1): 49, 2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28676089

ABSTRACT

BACKGROUND: Nanomedicine offers a promising tool for therapies of brain diseases, but potential effects on neuronal health and neuronal differentiation need to be investigated to assess potential risks. The aim of this study was to investigate effects of silica-indocyanine green/poly (ε-caprolactone) nanoparticles (PCL-NPs) engineered for laser tissue soldering in the brain before and during differentiation of SH-SY5Y cells. Considering adaptations in mitochondrial homeostasis during neuronal differentiation, metabolic effects of PCL-NP exposure before and during neuronal differentiation were studied. In addition, kinases of the PI3 kinase (PI3-K/Akt) and the MAP kinase (MAP-K/ERK) pathways related to neuronal differentiation and mitochondrial function were investigated. RESULTS: Differentiation resulted in a decrease in the cellular respiration rate and the extracellular acidification rate (ECAR). PCL-NP exposure impaired mitochondrial function depending on the time of exposure. The cellular respiration rate was significantly reduced compared to differentiated controls when PCL-NPs were given before differentiation. The shift in ECAR was less pronounced in PCL-NP exposure during differentiation. Differentiation and PCL-NP exposure had no effect on expression levels and the enzymatic activity of respiratory chain complexes. The activity of the glycolytic enzyme phosphofructokinase was significantly reduced after differentiation with the effect being more pronounced after PCL-NP exposure before differentiation. The increase in mitochondrial membrane potential observed after differentiation was not found in SH-SY5Y cells exposed to PCL-NPs before differentiation. The cellular adenosine triphosphate (ATP) production significantly dropped during differentiation, and this effect was independent of the PCL-NP exposure. Differentiation and nanoparticle exposure had no effect on superoxide levels at the endpoint of the experiments. A slight decrease in the expression of the neuronal differentiation markers was found after PCL-NP exposure, but no morphological variation was observed. CONCLUSIONS: PCL-NP exposure affects mitochondrial function depending on the time of exposure before and during neuronal differentiation. PCL-NP exposure during differentiation was associated with impaired mitochondrial function, which may affect differentiation. Considering the importance of adaptations in cellular respiration for neuronal differentiation and function, further studies are needed to unravel the underlying mechanisms and consequences to assess the possible risks including neurodegeneration.


Subject(s)
Mitochondria/drug effects , Nanoparticles/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Polyesters/metabolism , Silicon Dioxide/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Nanoparticles/toxicity , Neurons/cytology , Neurons/metabolism , Oxidative Phosphorylation/drug effects , Oxygen Consumption/drug effects , Phosphofructokinases/metabolism , Polyesters/toxicity , Silicon Dioxide/toxicity , Superoxides/metabolism
9.
Cell Transplant ; 26(4): 679-691, 2017 04 13.
Article in English | MEDLINE | ID: mdl-27938488

ABSTRACT

Cell replacement therapy is a promising avenue into the investigation and treatment of Parkinson's disease (PD), and in some cases, significant long-term motor improvements have been demonstrated. The main source of donor tissue is the human fetal ventral mesencephalon (FVM), which consists of a mixed neuronal population, and its heterogeneity likely contributes to the inconsistent outcome observed in clinical trials. Therefore, detailed knowledge about the neuronal subpopulations in the VM seems essential for successful cell transplantation. Interestingly, it has been reported that some tyrosine hydroxylase-positive (TH+) neurons in the VM of adult rats and in cultured midbrain-derived neuroblasts coexpress additional neurotransmitters. Thus, the present study investigated, by means of colocalization analyses, the possible expression of GABA or serotonin in TH+ neurons. For that purpose, both fetal rat and human dissociated, organotypic and neurosphere FVM cultures as well as an animal model of PD were investigated. In dissociated rat FVM cultures, approximately 30% of the TH+ neurons coexpressed serotonin, while no colocalization with GABA was observed. Interestingly, coexpression of TH and serotonin was found to be dependent on the time in culture, the plating density, and the exposure to neurotrophic factors, that is, higher cell densities and treatment with brain-derived neurotrophic factor resulted in a significantly reduced coexpression rate. Notably, even though approximately 30% of the dopaminergic neurons in the donor tissue coexpressed serotonin, no colocalization could be detected in grafts 1 month after intrastriatal transplantation into hemiparkinsonian rats. In conclusion, a significant and susceptible subpopulation of dopaminergic neurons in FVM tissues coexpresses serotonin. This might have potential implications for the future selection and handling of cells prior to transplantation in PD.


Subject(s)
Dopaminergic Neurons/metabolism , Mesencephalon/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Serotonin/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cell Count , Cells, Cultured , Disease Models, Animal , Female , Humans , Mesencephalon/drug effects , Rats, Wistar , Tyrosine 3-Monooxygenase/metabolism , gamma-Aminobutyric Acid/metabolism
10.
Nanomedicine ; 13(3): 1195-1204, 2017 04.
Article in English | MEDLINE | ID: mdl-27871963

ABSTRACT

Nanomedicine offers a promising tool for therapies of brain diseases, but they may be associated with potential adverse effects. The aim of this study was to investigate the uptake of silica-nanoparticles engineered for laser-tissue soldering in the brain using SH-SY5Y cells, dissociated and organotypic slice cultures from rat hippocampus. Nanoparticles were predominantly taken up by microglial cells in the hippocampal cultures but nanoparticles were also found in differentiated SH-SY5Y cells. The uptake was time- and concentration-dependent in primary hippocampal cells. Transmission electron microscopy experiments demonstrated nanoparticle aggregates and single particles in the cytoplasm. Nanoparticles were found in the endoplasmic reticulum, but not in other cellular compartments. Nanoparticle exposure did not impair cell viability and neuroinflammation in primary hippocampal cultures at all times investigated. Neurite outgrowth was not significantly altered in SH-SY5Y cells, but the neuronal differentiation markers indicated a reduction in neuronal differentiation induction after nanoparticle exposure.


Subject(s)
Brain/drug effects , Brain/metabolism , Nanoparticles/metabolism , Neurogenesis/drug effects , Silicon Dioxide/pharmacokinetics , Animals , Brain/cytology , Cell Line , Cell Survival/drug effects , Cells, Cultured , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Microglia/drug effects , Microglia/metabolism , Nanoparticles/analysis , Nanoparticles/toxicity , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar , Silicon Dioxide/metabolism , Silicon Dioxide/toxicity
11.
Amino Acids ; 48(8): 1913-27, 2016 08.
Article in English | MEDLINE | ID: mdl-27129463

ABSTRACT

Transplantation of neural progenitor cells (NPCs) is a promising experimental therapy for Huntington's disease (HD). The variables responsible for the success of this approach, including selection of the optimal developmental stage of the grafted cells, are however largely unknown. Supporting cellular energy metabolism by creatine (Cr) supplementation is a clinically translatable method for improving cell transplantation strategies. The present study aims at investigating differences between early (E14) and late (E18) developmental stages of rat striatal NPCs in vitro. NPCs were isolated from E14 and E18 embryos and cultured for 7 days with or without Cr [5 mM]. Chronic treatment significantly increased the percentage of GABA-immunoreactive neurons as compared to untreated controls, both in the E14 (170.4 ± 4.7 %) and the E18 groups (129.3 ± 9.3 %). This effect was greater in E14 cultures (p < 0.05). Similarly, short-term treatment for 24 h resulted in increased induction (p < 0.05) of the GABA-ergic phenotype in E14 (163.0 ± 10.4 %), compared to E18 cultures (133.3 ± 9.5 %). Total neuronal cell numbers and general viability were not affected by Cr (p > 0.05). Protective effects of Cr against a metabolic insult were equal in E14 and E18 NPCs (p > 0.05). Cr exposure promoted morphological differentiation of GABA-ergic neurons, including neurite length in both groups (p < 0.05), but the number of branching points was increased only in the E18 group (p < 0.05). Our results demonstrate that the role of Cr as a GABA-ergic differentiation factor depends on the developmental stage of striatal NPCs, while Cr-mediated neuroprotection is not significantly influenced. These findings have potential implications for optimizing future cell replacement strategies in HD.


Subject(s)
Cell Differentiation/drug effects , Corpus Striatum/embryology , Creatine/pharmacology , Embryo, Mammalian/embryology , GABAergic Neurons/metabolism , Neural Stem Cells/metabolism , Animals , Corpus Striatum/cytology , Embryo, Mammalian/cytology , Female , GABAergic Neurons/cytology , Neural Stem Cells/cytology , Rats , Rats, Wistar
12.
PLoS One ; 10(2): e0116088, 2015.
Article in English | MEDLINE | ID: mdl-25723595

ABSTRACT

Fetal antigen 1/delta-like 1 homologue (FA1/dlk1) belongs to the epidermal growth factor superfamily and is considered to be a non-canonical ligand for the Notch receptor. Interactions between Notch and its ligands are crucial for the development of various tissues. Moreover, FA1/dlk1 has been suggested as a potential supplementary marker of dopaminergic neurons. The present study aimed at investigating the distribution of FA1/dlk1-immunoreactive (-ir) cells in the early postnatal and adult midbrain as well as in the nigrostriatal system of 6-hydroxydopamine (6-OHDA)-lesioned hemiparkinsonian adult rats. FA1/dlk1-ir cells were predominantly distributed in the substantia nigra (SN) pars compacta (SNc) and in the ventral tegmental area. Interestingly, the expression of FA1/dlk1 significantly increased in tyrosine hydroxylase (TH)-ir cells during early postnatal development. Co-localization and tracing studies demonstrated that FA1/dlk1-ir cells in the SNc were nigrostriatal dopaminergic neurons, and unilateral 6-OHDA lesions resulted in loss of both FA1/dlk1-ir and TH-ir cells in the SNc. Surprisingly, increased numbers of FA1/dlk1-ir cells (by 70%) were detected in dopamine-depleted striata as compared to unlesioned controls. The higher number of FA1/dlk1-ir cells was likely not due to neurogenesis as colocalization studies for proliferation markers were negative. This suggests that FA1/dlk1 was up-regulated in intrinsic cells in response to the 6-OHDA-mediated loss of FA1/dlk1-expressing SNc dopaminergic neurons and/or due to the stab wound. Our findings hint to a significant role of FA1/dlk1 in the SNc during early postnatal development. The differential expression of FA1/dlk1 in the SNc and the striatum of dopamine-depleted rats could indicate a potential involvement of FA1/dlk1 in the cellular response to the degenerative processes.


Subject(s)
Gene Expression , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Substantia Nigra/metabolism , Animals , Biomarkers , Female , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mesencephalon/metabolism , Mesencephalon/pathology , Neurons/metabolism , Oxidopamine/adverse effects , Phenotype , Protein Binding , Protein Transport , Rats , Substantia Nigra/drug effects , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
13.
PLoS One ; 8(10): e76592, 2013.
Article in English | MEDLINE | ID: mdl-24116124

ABSTRACT

Trefoil factor 1 (TFF1) belongs to a family of secreted peptides with a characteristic tree-looped trefoil structure. TFFs are mainly expressed in the gastrointestinal tract where they play a critical role in the function of the mucosal barrier. TFF1 has been suggested as a neuropeptide, but not much is known about its expression and function in the central nervous system. We investigated the expression of TFF1 in the developing and adult rat midbrain. In the adult ventral mesencephalon, TFF1-immunoreactive (-ir) cells were predominantly found in the substantia nigra pars compacta (SNc), the ventral tegmental area (VTA) and in periaqueductal areas. While around 90% of the TFF1-ir cells in the SNc co-expressed tyrosine hydroxylase (TH), only a subpopulation of the TH-ir neurons expressed TFF1. Some TFF1-ir cells in the SNc co-expressed the calcium-binding proteins calbindin or calretinin and nearly all were NeuN-ir confirming a neuronal phenotype, which was supported by lack of co-localization with the astroglial marker glial fibrillary acidic protein (GFAP). Interestingly, at postnatal (P) day 7 and P14, a significantly higher proportion of TH-ir neurons in the SNc co-expressed TFF1 as compared to P21. In contrast, the proportion of TFF1-ir cells expressing TH remained unchanged during postnatal development. Furthermore, significantly more TH-ir neurons expressed TFF1 in the SNc, compared to the VTA at all four time-points investigated. Injection of the tracer fluorogold into the striatum of adult rats resulted in retrograde labeling of several TFF1 expressing cells in the SNc showing that a significant fraction of the TFF1-ir cells were projection neurons. This was also reflected by unilateral loss of TFF1-ir cells in SNc of 6-hydroxylase-lesioned hemiparkinsonian rats. In conclusion, we show for the first time that distinct subpopulations of midbrain dopaminergic neurons express TFF1, and that this expression pattern is altered in a rat model of Parkinson's disease.


Subject(s)
Mesencephalon/metabolism , Peptides/metabolism , Animals , Female , Immunohistochemistry , Mesencephalon/cytology , Mesencephalon/growth & development , Microscopy, Fluorescence , Rats , Rats, Wistar , Substantia Nigra/cytology , Substantia Nigra/growth & development , Substantia Nigra/metabolism , Time Factors , Trefoil Factor-1 , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/cytology , Ventral Tegmental Area/growth & development , Ventral Tegmental Area/metabolism
14.
Brain Res ; 1276: 39-49, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19389387

ABSTRACT

Transplantation of fetal dopaminergic (DA) neurons offers an experimental therapy for Parkinson's disease (PD). The low availability and the poor survival and integration of transplanted cells in the host brain are major obstacles in this approach. Glial cell line-derived neurotrophic factor (GDNF) is a potent neurotrophic factor with growth- and survival-promoting capabilities for developing DA neurons. In the present study, we examined whether pretreatment of ventral mesencephalic (VM) free-floating roller tube (FFRT) cultures with GDNF would improve graft survival and function. For that purpose organotypic cultures of E14 rat VM were grown for 2, 4 or 8 days in the absence (control) or presence of GDNF [10 ng/ml] and transplanted into the striatum of 6-hydroxydopamine-lesioned rats. While all groups of rats showed a significant reduction in d-amphetamine-induced rotations at 6 weeks posttransplantation a significantly improved graft function was observed only in the days in vitro (DIV) 4 GDNF pretreated group compared to the control group. In addition, no statistical significant differences between groups were found in the number of surviving tyrosine hydroxylase-immunoreactive (TH-ir) neurons assessed at 9 weeks posttransplantation. However, a tendency for higher TH-ir fiber outgrowth from the transplants in the GDNF pretreated groups as compared to corresponding controls was observed. Furthermore, GDNF pretreatment showed a tendency for a higher number of GIRK2 positive neurons in the grafts. In sum, our findings demonstrate that GDNF pretreatment was not disadvantageous for transplants of embryonic rat VM with the FFRT culture technique but only marginally improved graft survival and function.


Subject(s)
Cell Survival/drug effects , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Mesenchymal Stem Cell Transplantation/methods , Neurons/physiology , Neuroprotective Agents/therapeutic use , Parkinson Disease/therapy , Amphetamine/administration & dosage , Animals , Brain/drug effects , Brain/pathology , Cells, Cultured , Central Nervous System Stimulants/administration & dosage , Disease Models, Animal , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Motor Activity/drug effects , Neurons/pathology , Oxidopamine , Parkinson Disease/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
15.
Brain Res Bull ; 76(4): 329-43, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18502307

ABSTRACT

Creatine kinase catalyses the reversible transphosphorylation of creatine by ATP. In the cell, creatine kinase isoenzymes are specifically localized at strategic sites of ATP consumption to efficiently regenerate ATP in situ via phosphocreatine or at sites of ATP generation to build-up a phosphocreatine pool. Accordingly, the creatine kinase/phosphocreatine system plays a key role in cellular energy buffering and energy transport, particularly in cells with high and fluctuating energy requirements like neurons. Creatine kinases are expressed in the adult and developing human brain and spinal cord, suggesting that the creatine kinase/phosphocreatine system plays a significant role in the central nervous system. Functional impairment of this system leads to a deterioration in energy metabolism, which is phenotypic for many neurodegenerative and age-related diseases. Exogenous creatine supplementation has been shown to reduce neuronal cell loss in experimental paradigms of acute and chronic neurological diseases. In line with these findings, first clinical trials have shown beneficial effects of therapeutic creatine supplementation. Furthermore, creatine was reported to promote differentiation of neuronal precursor cells that might be of importance for improving neuronal cell replacement strategies. Based on these observations there is growing interest on the effects and functions of this compound in the central nervous system. This review gives a short excursion into the basics of the creatine kinase/phosphocreatine system and aims at summarizing findings and concepts on the role of creatine kinase and creatine in the central nervous system with special emphasis on pathological conditions and the positive effects of creatine supplementation.


Subject(s)
Central Nervous System/metabolism , Creatine Kinase/metabolism , Creatine/metabolism , Energy Metabolism , Animals , Brain Diseases, Metabolic/drug therapy , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/physiopathology , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/physiopathology , Creatine/therapeutic use , Humans , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Stroke/drug therapy , Stroke/metabolism , Stroke/physiopathology
16.
Neurosurg Focus ; 24(3-4): E16, 2008.
Article in English | MEDLINE | ID: mdl-18341392

ABSTRACT

Intracerebral hemorrhage (ICH), for which no effective treatment strategy is currently available, constitutes one of the most devastating forms of stroke. As a result, developing therapeutic options for ICH is of great interest to the medical community. The 3 potential therapies that have the most promise are cell replacement therapy, enhancing endogenous repair mechanisms, and utilizing various neuroprotective drugs. Replacement of damaged cells and restoration of function can be accomplished by transplantation of cells derived from different sources, such as embryonic or somatic stem cells, umbilical cord blood, and genetically modified cell lines. Early experimental data showing the benefits of cell transplantation on functional recovery after ICH have been promising. Nevertheless, several studies have focused on another therapeutic avenue, investigating novel ways to activate and direct endogenous repair mechanisms in the central nervous system, through exposure to specific neuronal growth factors or by inactivating inhibitory molecules. Lastly, neuroprotective drugs may offer an additional tool for improving neuronal survival in the perihematomal area. However, a number of scientific issues must be addressed before these experimental techniques can be translated into clinical therapy. In this review, the authors outline the recent advances in the basic science of treatment strategies for ICH.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cerebral Hemorrhage/therapy , Animals , Cell- and Tissue-Based Therapy/trends , Disease Models, Animal , Humans
17.
Swiss Med Wkly ; 138(11-12): 155-72, 2008 Mar 22.
Article in English | MEDLINE | ID: mdl-18363114

ABSTRACT

There is increasing interest in the search for therapeutic options for diseases and injuries of the central nervous system (CNS), for which currently no effective treatment strategies are available. Replacement of damaged cells and restoration of function can be accomplished by transplantation of cells derived from different sources, such as human foetal tissue, genetically modified cell lines, embryonic or somatic stem cells. Preclinical and clinical trials have shown promising results in neurodegenerative disorders, like Parkinson's and Huntington's disease, but also ischaemic stroke, intracerebral haemorrhage, demyelinating disorders, epilepsy and traumatic lesions of the brain and spinal cord. Other studies have focused on finding new ways to activate and direct endogenous repair mechanisms in the CNS, eg, by exposure to specific neuronal growth factors or by inactivating inhibitory molecules. Neuroprotective drugs may offer an additional tool for improving neuronal survival in acute or chronic CNS diseases. Importantly however, a number of scientific issues need to be addressed in order to permit the introduction of these experimental techniques in the wider clinical setting.


Subject(s)
Central Nervous System Diseases/therapy , Nerve Tissue/transplantation , Animals , Humans
18.
J Neurosci Res ; 85(9): 1863-75, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17526013

ABSTRACT

Creatine is a substrate of cytosolic and mitochondrial creatine kinases. Its supplementation augments cellular levels of creatine and phosphocreatine, the rate of ATP resynthesis, and improves the function of the creatine kinase energy shuttle. High cytoplasmatic total creatine levels have been reported to be neuroprotective by inhibiting apoptosis. In addition, creatine has direct antioxidant effects, which may be of importance in amyotrophic lateral sclerosis. In the present study, we investigated the effects of creatine [5 mM] on survival and differentiation of cultured GABA-immunoreactive (-ir) and choline acetyltransferase (ChAT)-ir rat spinal cord neurons. Furthermore, we addressed the neuroprotective potential of creatine supplementation against 3-nitropropionic acid (3-NP) induced toxicity. General cell survival and total neuronal cell density were not altered by chronic creatine treatment. We found, however, after chronic creatine and short-term creatine exposure a significantly higher density of GABA-ir neurons hinting to a differentiation-inducing mechanism of creatine. This notion is further supported by a significant higher content of GAD after creatine exposure. Creatine supplementation also exerted a partial, but significant neuroprotection for GABA-ir neurons against 3-NP induced toxicity. Interestingly, chronic creatine treatment did not alter cell density of ChAT-ir neurons but promoted their morphologic differentiation. Cell soma size and number of primary neurites per neuron were increased significantly after creatine supplementation. Taken together, creatine supplementation promoted the differentiation or the survival of GABAergic neurons and resulted in partial neuroprotection against 3-NP induced toxicity. The data suggest that creatine may play a critical role during development of spinal cord neurons.


Subject(s)
Cell Differentiation/drug effects , Creatine/pharmacology , Neurons/physiology , Spinal Cord/cytology , Stem Cells/physiology , gamma-Aminobutyric Acid/physiology , Animals , Blotting, Western , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Choline O-Acetyltransferase/metabolism , Creatine Kinase/metabolism , Energy Metabolism/drug effects , Female , Immunohistochemistry , Mitogen-Activated Protein Kinase 1/genetics , Nitro Compounds/pharmacology , Pregnancy , Propionates/pharmacology , Rats , Tetrazolium Salts , Thiazoles , Tubulin/genetics
19.
Brain Res ; 1137(1): 50-7, 2007 Mar 16.
Article in English | MEDLINE | ID: mdl-17223088

ABSTRACT

In the present study, we investigated the expression pattern of cytosolic brain specific-BB-CK and ubiquitous mitochondrial-creatine kinases (uMt-CK) in developing human spinal cord. Consequently, we studied the effects of creatine treatment on cultured fetal human spinal cord tissue. We found that both CK isoforms were expressed in fetal spinal cord at all time points investigated (5 to 11.5 weeks post conception) and correspondingly specific CK activity was detected. Chronic creatine exposure resulted in significantly higher densities of GABA-immunoreactive neurons in the cultures, while total neuronal cell density was not altered, suggesting a differentiation inducing mechanism of creatine supplementation. Taken together, our observations favour the view that the creatine phosphocreatine system plays an important role in the developing CNS.


Subject(s)
Creatine/pharmacology , Gene Expression Regulation, Developmental/drug effects , Neurons/drug effects , Spinal Cord/cytology , gamma-Aminobutyric Acid/metabolism , Age Factors , Cell Count/methods , Choline O-Acetyltransferase/metabolism , Creatine Kinase, MM Form/metabolism , Fetus , Humans , Immunohistochemistry/methods , Organ Culture Techniques , Spinal Cord/drug effects
20.
Cell Transplant ; 14(8): 537-50, 2005.
Article in English | MEDLINE | ID: mdl-16355565

ABSTRACT

Cell replacement therapy using mesencephalic precursor cells is an experimental approach for the treatment of Parkinson's disease (PD). A significant problem associated with this procedure is the poor survival of grafted neurons. Impaired energy metabolism is considered to contribute to neuronal cell death after transplantation. Creatine is a substrate for mitochondrial and cytosolic creatine kinases (CK) and buffers cellular ATP resources. Furthermore, elevated cellular creatine levels facilitate metabolic channeling and show antiapoptotic properties. Exogenous creatine supplementation therefore might offer a tool for improvement of dopaminergic neuron survival. The present study aimed at investigating the effects of creatine on cell survival of rat embryonic day 14 (E14) ventral mesencephalic neurons grown as organotypic free-floating roller tube (FFRT) cultures. We found that the brain-specific isoform of CK (BB-CK) and the ubiquitous mitochondrial isoform (uMt-CK) are expressed at high levels in FFRT cultures and colocalize with tyrosine hydroxylase immunoreactive (TH-ir) cells. Exposure of these cultures to creatine induced an increase in the content of the BB-CK isotype. Creatine (5 mM) administration starting at day in vitro (DIV) 7 resulted in a significant increase (+35%) in TH-ir cell density at DIV21. In addition, we observed that creatine treatment provided neuroprotection against 1-methyl-4-phenyl pyridinium ion (MPP+)-induced TH-ir cell loss in the FFRT culture system, resulting in a significantly higher density (+19%) of TH-ir neurons in creatine-treated cultures compared to corresponding controls. The decrease of TH-ir neurons in the MPP+-treated group corresponded with an increase in immunoreactivity for active caspase-3, an effect that was not seen in the group receiving creatine supplementation. In conclusion, our data imply that creatine administration is beneficial for the survival of TH-ir neurons encountering harmful conditions.


Subject(s)
Cell Survival/drug effects , Creatine/pharmacology , Tissue Culture Techniques/methods , 1-Methyl-4-phenylpyridinium , Animals , Cells, Cultured , Creatine/metabolism , Female , Humans , Mesencephalon/drug effects , Mesencephalon/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...