Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Acta Neuropathol Commun ; 11(1): 40, 2023 03 11.
Article in English | MEDLINE | ID: mdl-36906636

ABSTRACT

The MAPT gene, encoding the microtubule-associated protein tau on chromosome 17q21.31, is result of an inversion polymorphism, leading to two allelic variants (H1 and H2). Homozygosity for the more common haplotype H1 is associated with an increased risk for several tauopathies, but also for the synucleinopathy Parkinson's disease (PD). In the present study, we aimed to clarify whether the MAPT haplotype influences expression of MAPT and SNCA, encoding the protein α-synuclein (α-syn), on mRNA and protein levels in postmortem brains of PD patients and controls. We also investigated mRNA expression of several other MAPT haplotype-encoded genes. Postmortem tissues from cortex of fusiform gyrus (ctx-fg) and of the cerebellar hemisphere (ctx-cbl) of neuropathologically confirmed PD patients (n = 95) and age- and sex-matched controls (n = 81) were MAPT haplotype genotyped to identify cases homozygous for either H1 or H2. Relative expression of genes was quantified using real-time qPCR; soluble and insoluble protein levels of tau and α-syn were determined by Western blotting. Homozygosity for H1 versus H2 was associated with increased total MAPT mRNA expression in ctx-fg regardless of disease state. Inversely, H2 homozygosity was associated with markedly increased expression of the corresponding antisense MAPT-AS1 in ctx-cbl. PD patients had higher levels of insoluble 0N3R and 1N4R tau isoforms regardless of the MAPT genotype. The increased presence of insoluble α-syn in PD patients in ctx-fg validated the selected postmortem brain tissue. Our findings in this small, but well controlled cohort of PD and controls support a putative biological relevance of tau in PD. However, we did not identify any link between the disease-predisposing H1/H1 associated overexpression of MAPT with PD status. Further studies are required to gain a deeper understanding of the potential regulatory role of MAPT-AS1 and its association to the disease-protective H2/H2 condition in the context of PD.


Subject(s)
Genetic Predisposition to Disease , Parkinson Disease , tau Proteins , Humans , Brain/metabolism , Genotype , Haplotypes , Parkinson Disease/metabolism , Polymorphism, Single Nucleotide , RNA, Messenger/genetics , tau Proteins/genetics
2.
Mov Disord ; 38(4): 589-603, 2023 04.
Article in English | MEDLINE | ID: mdl-36692025

ABSTRACT

BACKGROUND: Because human fetal ventral mesencephalic tissue grafts provide promising results in ameliorating Parkinson's disease-implicated motor dysfunctions, human fetal midbrain-derived dopamine neuronal precursor cells are considered good candidates for cell-based therapy for Parkinson's disease in that large quantities of cells can be supplied through a good manufacturing practice-compliant system. OBJECTIVE: We conducted a prospective, phase I/IIa, dose-escalation, open-label "first-in-human" clinical trial with fetal neural precursor cells to assess their safety and therapeutic efficacy in patients with idiopathic Parkinson's disease. METHODS: Fifteen patients were assigned to receive three different doses of cells (4 × 106 , 12 × 106 , and 40 × 106 cells) and completed a 12-month follow-up. The primary outcome was safety, by measuring the presence of grade 3 or higher cells according to National Cancer Institute guidelines and any contaminated cells. Secondary outcomes assessed motor and neurocognitive function, as well as the level of dopamine transporters, by positron emission tomography-computed tomography. RESULTS: Although a pronation-supination and hand/arm movement performance was remarkably enhanced in all three groups (all P < 0.05), the medium- and high-dose-treated groups exhibited significant improvement in Unified Parkinson's Disease Rating Scale Part III only up to 26.16% and 40%, respectively, at 12 months after transplantation without any serious clinical complications or graft-induced dyskinesia in all patients. However, the motor improvements did not correlate with increase in the dopamine transporter on positron emission tomography images. CONCLUSIONS: Our results primarily demonstrate the safety and plausible dose-dependent efficacy of human fetal midbrain-derived dopamine neuronal precursor cells for idiopathic Parkinson's disease. © 2023 International Parkinson and Movement Disorder Society.


Subject(s)
Neural Stem Cells , Parkinson Disease , Humans , Parkinson Disease/therapy , Parkinson Disease/drug therapy , Dopamine , Prospective Studies , Tomography, X-Ray Computed , Mesencephalon/diagnostic imaging
3.
Front Cell Dev Biol ; 9: 726866, 2021.
Article in English | MEDLINE | ID: mdl-34532319

ABSTRACT

The H1 haplotype of the microtubule-associated protein tau (MAPT) gene is a common genetic risk factor for some neurodegenerative diseases such as progressive supranuclear palsy, corticobasal degeneration, and Parkinson's disease. The molecular mechanism causing the increased risk for the named diseases, however, remains unclear. In this paper, we present a valuable tool of eight small molecule neural precursor cell lines (smNPC) homozygous for the MAPT haplotypes (four H1/H1 and four H2/H2 cell lines), which can be used to identify MAPT-dependent phenotypes. The employed differentiation protocol is fast due to overexpression of NEUROGENIN-2 and therefore suitable for high-throughput approaches. A basic characterization of all human cell lines was performed, and their TAU and α-SYNUCLEIN profiles were compared during a differentiation time of 30 days. We could identify higher levels of conformationally altered TAU in cell lines carrying the H2 haplotype. Additionally, we found increased expression levels of α-SYNUCLEIN in H1/H1 cells. With this resource, we aim to fill a gap in neurodegenerative disease modeling with induced pluripotent stem cells (iPSC) for sporadic tauopathies.

4.
Nat Cell Biol ; 23(6): 652-663, 2021 06.
Article in English | MEDLINE | ID: mdl-34083785

ABSTRACT

Expression of exon-specific isoforms from alternatively spliced mRNA is a fundamental mechanism that substantially expands the proteome of a cell. However, conventional methods to assess alternative splicing are either consumptive and work-intensive or do not quantify isoform expression longitudinally at the protein level. Here, we therefore developed an exon-specific isoform expression reporter system (EXSISERS), which non-invasively reports the translation of exon-containing isoforms of endogenous genes by scarlessly excising reporter proteins from the nascent polypeptide chain through highly efficient, intein-mediated protein splicing. We applied EXSISERS to quantify the inclusion of the disease-associated exon 10 in microtubule-associated protein tau (MAPT) in patient-derived induced pluripotent stem cells and screened Cas13-based RNA-targeting effectors for isoform specificity. We also coupled cell survival to the inclusion of exon 18b of FOXP1, which is involved in maintaining pluripotency of embryonic stem cells, and confirmed that MBNL1 is a dominant factor for exon 18b exclusion. EXSISERS enables non-disruptive and multimodal monitoring of exon-specific isoform expression with high sensitivity and cellular resolution, and empowers high-throughput screening of exon-specific therapeutic interventions.


Subject(s)
Alternative Splicing , Forkhead Transcription Factors/metabolism , High-Throughput Screening Assays , Induced Pluripotent Stem Cells/metabolism , Proteomics , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , tau Proteins/metabolism , CRISPR-Cas Systems , Exons , Forkhead Transcription Factors/genetics , HEK293 Cells , Humans , Protein Isoforms , Proteome , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Single-Cell Analysis , tau Proteins/genetics
5.
Cells ; 10(4)2021 03 31.
Article in English | MEDLINE | ID: mdl-33807497

ABSTRACT

BACKGROUND: Consecutive adult neurogenesis is a well-known phenomenon in the ventricular-subventricular zone of the lateral wall of the lateral ventricles (V-SVZ) and has been controversially discussed in so-called "non-neurogenic" brain areas such as the periventricular regions (PVRs) of the aqueduct and the fourth ventricle. Dopamine is a known modulator of adult neural stem cell (aNSC) proliferation and dopaminergic neurogenesis in the olfactory bulb, though a possible interplay between local dopaminergic neurodegeneration and induction of aNSC proliferation in mid/hindbrain PVRs is currently enigmatic. OBJECTIVE/HYPOTHESIS: To analyze the influence of chronic-progressive dopaminergic neurodegeneration on both consecutive adult neurogenesis in the PVRs of the V-SVZ and mid/hindbrain aNSCs in two mechanistically different transgenic animal models of Parkinson´s disease (PD). METHODS: We used Thy1-m[A30P]h α synuclein mice and Leu9'Ser hypersensitive α4* nAChR mice to assess the influence of midbrain dopaminergic neuronal loss on neurogenic activity in the PVRs of the V-SVZ, the aqueduct and the fourth ventricle. RESULTS: In both animal models, overall proliferative activity in the V-SVZ was not altered, though the proportion of B2/activated B1 cells on all proliferating cells was reduced in the V-SVZ in Leu9'Ser hypersensitive α4* nAChR mice. Putative aNSCs in the mid/hindbrain PVRs are known to be quiescent in vivo in healthy controls, and dopaminergic deficiency did not induce proliferative activity in these regions in both disease models. CONCLUSIONS: Our data do not support an activation of endogenous aNSCs in mid/hindbrain PVRs after local dopaminergic neurodegeneration. Spontaneous endogenous regeneration of dopaminergic cell loss through resident aNSCs is therefore unlikely.


Subject(s)
Dopamine/deficiency , Mesencephalon/physiology , Neurogenesis , Animals , Cell Proliferation , Humans , Lateral Ventricles/physiology , Mice, Inbred C57BL , Receptors, Nicotinic/metabolism , Rhombencephalon/physiology , alpha-Synuclein/metabolism
6.
Front Cell Dev Biol ; 9: 561086, 2021.
Article in English | MEDLINE | ID: mdl-33748099

ABSTRACT

Growing evidence suggests that epigenetic mechanisms like microRNA-mediated transcriptional regulation contribute to the pathogenesis of parkinsonism. In order to study the influence of microRNAs (miRNAs), we analyzed the miRNome 2 days prior to major cell death in α-synuclein-overexpressing Lund human mesencephalic neurons, a well-established cell model of Parkinson's disease (PD), by next-generation sequencing. The expression levels of 23 miRNAs were significantly altered in α-synuclein-overexpressing cells, 11 were down- and 12 upregulated (P < 0.01; non-adjusted). The in silico analysis of known target genes of these miRNAs was complemented by the inclusion of a transcriptome dataset (BeadChip) of the same cellular system, revealing the G0/G1 cell cycle transition to be markedly enriched. Out of 124 KEGG-annotated cell cycle genes, 15 were present in the miRNA target gene dataset and six G0/G1 cell cycle genes were found to be significantly altered upon α-synuclein overexpression, with five genes up- (CCND1, CCND2, and CDK4 at P < 0.01; E2F3, MYC at P < 0.05) and one gene downregulated (CDKN1C at P < 0.001). Additionally, several of these altered genes are targeted by miRNAs hsa-miR-34a-5p and hsa-miR-34c-5p, which also modulate α-synuclein expression levels. Functional intervention by siRNA-mediated knockdown of the cell cycle gene cyclin D1 (CCND1) confirmed that silencing of cell cycle initiation is able to substantially reduce α-synuclein-mediated cytotoxicity. The present findings suggest that α-synuclein accumulation induces microRNA-mediated aberrant cell cycle activation in post-mitotic dopaminergic neurons. Thus, the mitotic cell cycle pathway at the level of miRNAs might offer interesting novel therapeutic targets for PD.

7.
Prog Neurobiol ; 180: 101644, 2019 09.
Article in English | MEDLINE | ID: mdl-31238088

ABSTRACT

Tau is a microtubule-associated protein with versatile functions in the dynamic assembly of the neuronal cytoskeleton. Four-repeat (4R-) tauopathies are a group of neurodegenerative diseases defined by cytoplasmic inclusions predominantly composed of tau protein isoforms with four microtubule-binding domains. Progressive supranuclear palsy, corticobasal degeneration, argyrophilic grain disease or glial globular tauopathy belong to the group of 4R-tauopathies. The present review provides an introduction in the current concept of 4R-tauopathies, including an overview of the neuropathological and clinical spectrum of these diseases. It describes the genetic and environmental etiological factors, as well as the contemporary knowledge about the pathophysiological mechanisms, including post-translational modifications, aggregation and fragmentation of tau, as well as the role of protein degradation mechanisms. Furthermore, current theories about disease propagation are discussed, involving different extracellular tau species and their cellular release and uptake mechanisms. Finally, molecular diagnostic tools for 4R-tauopathies, including tau-PET and fluid biomarkers, and investigational therapeutic strategies are presented. In summary, we report on 4R-tauopathies as overarching disease concept based on a shared pathophysiological concept, and highlight the challenges and opportunities on the way towards a causal therapy.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Neurons/metabolism , Tauopathies/metabolism , tau Proteins/metabolism , Humans , Neuropathology/methods
8.
Nat Commun ; 9(1): 2929, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30050033

ABSTRACT

Genetic, epigenetic, and environmental factors contribute to the multifactorial disorder progressive supranuclear palsy (PSP). Here, we study epigenetic changes by genome-wide analysis of DNA from postmortem tissue of forebrains of patients and controls and detect significant (P < 0.05) methylation differences at 717 CpG sites in PSP vs. controls. Four-hundred fifty-one of these sites are associated with protein-coding genes. While differential methylation only affects a few sites in most genes, DLX1 is hypermethylated at multiple sites. Expression of an antisense transcript of DLX1, DLX1AS, is reduced in PSP brains. The amount of DLX1 protein is increased in gray matter of PSP forebrains. Pathway analysis suggests that DLX1 influences MAPT-encoded Tau protein. In a cell system, overexpression of DLX1 results in downregulation of MAPT while overexpression of DLX1AS causes upregulation of MAPT. Our observations suggest that altered DLX1 methylation and expression contribute to pathogenesis of PSP by influencing MAPT.


Subject(s)
DNA Methylation/genetics , Epigenesis, Genetic/genetics , Homeodomain Proteins/metabolism , Supranuclear Palsy, Progressive/genetics , Supranuclear Palsy, Progressive/pathology , Transcription Factors/metabolism , Aged , Aged, 80 and over , Female , Homeodomain Proteins/genetics , Humans , Male , Transcription Factors/genetics , tau Proteins/genetics , tau Proteins/metabolism
9.
Stem Cells Dev ; 27(14): 976-984, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29779467

ABSTRACT

Voltage-gated sodium and calcium channels as well as transient receptor potential (TRP) channels are expressed during the differentiation of human neural progenitor cells (hNPCs) and are likely to be involved in regulating neurogenesis. However, the molecular composition of these ion channels in proliferating and differentiating hNPCs is largely unknown. In this study, we investigated fetal mesencephalic hNPCs in respect to their sodium, calcium, and TRP channel subunit expression and function. Quantitative real-time polymerase chain reaction indicated a significant upregulation of voltage-gated sodium and calcium channel subunits in hNPCs after differentiation for 3 weeks in vitro. In contrast, the TRP channel expression did not increase significantly during hNPC maturation. Intracellular Ca2+ measurements showed the marked reduction of KCl-induced Ca2+ transients through inhibition of voltage-gated Ca2+ channels by verapamil and mibefradil in differentiated hNPCs. Application of TRP channel agonists induced intracellular Ca2+ peaks already in proliferating hNPCs without affecting their cell division. The coincubation of hNPCs with TRP channel agonists pregnenolone sulfate or RN1747 did not have any significant effect on their proliferation and differentiation. These data indicate that hNPCs derived from fetal midbrain tissue acquire essential voltage-gated sodium and calcium channel properties during neuronal maturation in vitro. An early role of TRP channels in neurogenesis which may be important for regenerative clinical applications or cellular models could not be elucidated using hNPCs.


Subject(s)
Calcium Channels/genetics , Neural Stem Cells/metabolism , Stem Cells/metabolism , Voltage-Gated Sodium Channels/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Fetus , Gene Expression Regulation, Developmental/drug effects , Humans , Mesencephalon/cytology , Mesencephalon/metabolism , Neural Stem Cells/drug effects , Pregnenolone/pharmacology , Stem Cells/drug effects , Sulfonamides/pharmacology , TRPA1 Cation Channel/genetics
10.
Stem Cells Transl Med ; 6(2): 576-588, 2017 02.
Article in English | MEDLINE | ID: mdl-28191758

ABSTRACT

We have developed a good manufacturing practice for long-term cultivation of fetal human midbrain-derived neural progenitor cells. The generation of human dopaminergic neurons may serve as a tool of either restorative cell therapies or cellular models, particularly as a reference for phenotyping region-specific human neural stem cell lines such as human embryonic stem cells and human inducible pluripotent stem cells. We cultivated 3 different midbrain neural progenitor lines at 10, 12, and 14 weeks of gestation for more than a year and characterized them in great detail, as well as in comparison with Lund mesencephalic cells. The whole cultivation process of tissue preparation, cultivation, and cryopreservation was developed using strict serum-free conditions and standardized operating protocols under clean-room conditions. Long-term-cultivated midbrain-derived neural progenitor cells retained stemness, midbrain fate specificity, and floorplate markers. The potential to differentiate into authentic A9-specific dopaminergic neurons was markedly elevated after prolonged expansion, resulting in large quantities of functional dopaminergic neurons without genetic modification. In restorative cell therapeutic approaches, midbrain-derived neural progenitor cells reversed impaired motor function in rodents, survived well, and did not exhibit tumor formation in immunodeficient nude mice in the short or long term (8 and 30 weeks, respectively). We conclude that midbrain-derived neural progenitor cells are a promising source for human dopaminergic neurons and suitable for long-term expansion under good manufacturing practice, thus opening the avenue for restorative clinical applications or robust cellular models such as high-content or high-throughput screening. Stem Cells Translational Medicine 2017;6:576-588.


Subject(s)
Cell Proliferation , Dopaminergic Neurons/physiology , Mesencephalon/embryology , Neural Stem Cells/physiology , Neurogenesis , Parkinsonian Disorders/surgery , Stem Cell Transplantation/methods , Animals , Biomarkers/metabolism , Cell Culture Techniques , Cell Line , Disease Models, Animal , Dopaminergic Neurons/metabolism , Female , Gestational Age , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Motor Activity , Neural Stem Cells/metabolism , Oxidopamine , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Phenotype , Rats, Sprague-Dawley , Recovery of Function , Risk Assessment , Stem Cell Transplantation/adverse effects , Teratoma/etiology , Teratoma/pathology , Time Factors
11.
J Tissue Eng Regen Med ; 11(3): 751-764, 2017 03.
Article in English | MEDLINE | ID: mdl-25641599

ABSTRACT

Neural stem or progenitor cells are considered to be a novel therapeutic strategy for amyotrophic lateral sclerosis (ALS), based on their potential to generate a protective environment rather than to replace degenerating motor neurons. Following local injection to the spinal cord, neural progenitor cells may generate glial cells and release neurotrophic factors. In the present study, human spinal cord-derived neural progenitor cells (hscNPCs) were injected into the lumbar spinal cord of G93A-SOD1 ALS transgenic mice. We evaluated the potential effect of hscNPC treatment by survival analysis and behavioural/phenotypic assessments. Immunohistological and real-time PCR experiments were performed at a defined time point to study the underlying mechanisms. Symptom progression in hscNPC-injected mice was significantly delayed at the late stage of disease. On average, survival was only prolonged for 5 days. Animals treated with hscNPCs performed significantly better in motor function tests between weeks 18 and 19. Increased production of GDNF and IGF-1 mRNA was detectable in spinal cord tissue of hscNPC-treated mice. In summary, treatment with hscNPCs led to increased endogenous production of several growth factors and increased the preservation of innervated motor neurons but had only a small effect on overall survival. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Nerve Growth Factors/metabolism , Neural Stem Cells/transplantation , Spinal Cord/cytology , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Cell Lineage , Disease Models, Animal , Disease Progression , Humans , Injections, Spinal , Mice, Transgenic , Motor Activity , Neural Stem Cells/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cell Transplantation , Survival Analysis
12.
Tissue Eng Part A ; 21(3-4): 654-68, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25257211

ABSTRACT

Advances in mechanistic knowledge of human neurological disorders have been hindered by the lack of adequate human in vitro models. Three-dimensional (3D) cellular models displaying higher biological relevance are gaining momentum; however, their lack of robustness and scarcity of analytical tools adapted to three dimensions hampers their widespread implementation. Herein we show that human midbrain-derived neural progenitor cells, cultured as 3D neurospheres in stirred culture systems, reproducibly differentiate into complex tissue-like structures containing functional dopaminergic neurons, as well as astrocytes and oligodendrocytes. Moreover, an extensive toolbox of analytical methodologies has been adapted to 3D neural cell models, allowing molecular and phenotypic profiling and interrogation. The generated neurons underwent synaptogenesis and elicit spontaneous Ca(2+) transients. Synaptic vesicle trafficking and release of dopamine in response to depolarizing stimuli was also observed. Under whole-cell current-and-voltage clamp, recordings showed polarized neurons (Vm=-70 mV) and voltage-dependent potassium currents, which included A-type-like currents. Glutamate-induced currents sensitive to α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and N-methyl-D-aspartate antagonists revealed the existence of functional glutamate receptors. Molecular and phenotypic profiling showed recapitulation of midbrain patterning events, and remodeling toward increased similarity to human brain features, such as extracellular matrix composition and metabolic signature. We have developed a robust and reproducible human 3D neural cell model, which may be extended to patient-derived induced pluripotent stem cells, broadening the applicability of this model.


Subject(s)
Batch Cell Culture Techniques/methods , Dopaminergic Neurons/cytology , Dopaminergic Neurons/physiology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Printing, Three-Dimensional , Biomimetics/methods , Cell Differentiation/physiology , Cells, Cultured , Dopamine , Humans , Tissue Engineering/methods
13.
Methods ; 56(3): 452-60, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22433395

ABSTRACT

Central nervous system (CNS) disorders remain a formidable challenge for the development of efficient therapies. Cell and gene therapy approaches are promising alternatives that can have a tremendous impact by treating the causes of the disease rather than the symptoms, providing specific targeting and prolonged duration of action. Hampering translation of gene-based therapeutic treatments of neurodegenerative diseases from experimental to clinical gene therapy is the lack of valid and reliable pre-clinical models that can contribute to evaluate feasibility and safety. Herein we describe a robust and reproducible methodology for the generation of 3D in vitro models of the human CNS following a systematic technological approach based on stirred culture systems. We took advantage of human midbrain-derived neural progenitor cells (hmNPCs) capability to differentiate into the various neural phenotypes and of their commitment to the dopaminergic lineage to generate differentiated neurospheres enriched in dopaminergic neurons. Furthermore, we describe a protocol for efficient gene transfer into differentiated neurospheres using CAV-2 viral vectors and stable expression of the transgene for at least 10 days. CAV-2 vectors, derived from canine adenovirus type 2, are promising tools to understand and treat neurodegenerative diseases, in particular Parkinson's disease. CAV-2 vectors preferentially transduce neurons and have an impressive level of axonal retrograde transport in vivo. Our model provides a practical and versatile in vitro approach to study the CNS in a 3D cellular context. With the successful differentiation and subsequent genetic modification of neurospheres we are increasing the collection of tools available for neuroscience research and contributing for the implementation and widespread utilization of 3D cellular CNS models. These can be applied to study neurodegenerative diseases such as Parkinson's disease; to study the interaction of viral vectors of therapeutic potential within human neural cell populations, thus enabling the introduction of specific therapeutic genes for treatment of CNS pathologies; to study the fate and effect of delivered therapeutic genes; to study toxicological effects. Furthermore these methodologies may be extended to other sources of human neural stem cells, such as human pluripotent stem cells, including patient-derived induced pluripotent stem cells.


Subject(s)
Cell Culture Techniques/methods , Dopaminergic Neurons/cytology , Neural Stem Cells/cytology , Cell Differentiation , Humans , Reproducibility of Results , Transduction, Genetic
14.
Stem Cells ; 30(3): 570-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22213586

ABSTRACT

Reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) by retroviral overexpression of the transcription factors Oct4, Sox2, Klf4, and c-Myc holds great promise for the development of personalized cell replacement therapies. In an attempt to minimize the risk for chromosomal disruption and to simplify reprogramming, several studies demonstrated that a reduced set of reprogramming factors is sufficient to generate iPSC, albeit at lower efficiency. To elucidate the influence of factor reduction on subsequent differentiation, we compared the efficiency of neuronal differentiation in iPSC generated from postnatal murine neural stem cells with either one (Oct4; iPSC(1F-NSC) ), two (Oct4, Klf4; iPSC(2F-NSC) ), or all four factors (iPSC(4F-NSC) ) with those of embryonic stem cells (ESCs) and iPSC produced from fibroblasts with all four factors (iPSC(4F-MEF) ). After 2 weeks of coculture with PA6 stromal cells, neuronal differentiation of iPSC(1F-NSC) and iPSC(2F-NSC) was less efficient compared with iPSC(4F-NSC) and ESC, yielding lower proportions of colonies that stained positive for early and late neuronal markers. Electrophysiological analyses after 4 weeks of differentiation identified functional maturity in neurons differentiated from ESC, iPSC(2F-NSC) , iPSC(4F-NSC) , and iPSC(4F-MEF) but not in those from iPSC(1F-NSC) . Similar results were obtained after hematoendothelial differentiation on OP9 bone marrow stromal cells, where factor-reduced iPSC generated lower proportions of colonies with hematoendothelial progenitors than colonies of ESC, iPSC(4F-NSC) , and iPSC(4F-MEF) . We conclude that a reduction of reprogramming factors does not only reduce reprogramming efficiency but may also worsen subsequent differentiation and hinder future application of iPSC in cell replacement therapies.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/physiology , Neural Stem Cells/physiology , Animals , Antigens, Differentiation/metabolism , Cell Culture Techniques , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Intermediate Filament Proteins/metabolism , Kruppel-Like Factor 4 , Membrane Potentials , Mice , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Neural Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rats , Stromal Cells/metabolism , Stromal Cells/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
Transl Res ; 156(3): 155-60, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20801412

ABSTRACT

"Regenerative medicine" hopefully will provide novel therapies for diseases that remain without effective therapy. This development is also true for most neurodegenerative disorders including Alzheimer's disease, Huntington's disease, or Parkinson's disease. Transplantation of new neurons to the brain has been performed in Parkinson's disease and in Huntington's disease. The restoration of dopaminergic neurons in patients with Parkinson's disease via implantation of embryonic midbrain tissue was taken from animal experiments to clinical applications, showing a limited efficacy. Clinical trials in patients with Huntington's disease using fetal striatal tissue currently are underway. Today, it seems possible to generate functional dopaminergic or striatal neurons form a variety of stem cells including embryonic or neural stem cells as well as induced pluripotent stem cells. First clinical trials using neural stem cell or embryonic-stem-cell-derived tissue are approved or already underway. Such cells allow for extensive in vitro and in vivo testing as well as "good manufacturing production," reducing the risks in clinical application.


Subject(s)
Embryonic Stem Cells/transplantation , Nervous System Diseases/surgery , Pluripotent Stem Cells/transplantation , Stem Cell Transplantation/methods , Adult , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Brain/cytology , Brain/embryology , Brain/physiology , Cell Differentiation/physiology , Humans , Mesenchymal Stem Cell Transplantation/methods , Neurons/cytology , Neurons/physiology , Risk Assessment , Stem Cell Transplantation/adverse effects
16.
J Neurochem ; 111(1): 204-16, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19659571

ABSTRACT

Human midbrain-derived neural progenitor cells (NPCs) may serve as a continuous source of dopaminergic neurons for the development of novel regenerative therapies in Parkinson's disease. However, the molecular and functional characteristics of glutamate receptors in human NPCs are largely unknown. Here, we show that differentiated human mesencepahlic NPCs display a distinct pattern of glutamate receptors. In whole-cell patch-clamp recordings, l-glutamate and NMDA elicited currents in 93% of NPCs after 3 weeks of differentiation in vitro. The concentration-response plots of differentiated NPCs yielded an EC(50) of 2.2 microM for glutamate and an EC(50) of 36 microM for NMDA. Glutamate-induced currents were markedly inhibited by memantine in contrast to 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) suggesting a higher density of functional NMDA than alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA)/kainate receptors. NMDA-evoked currents and calcium signals were blocked by the NR2B-subunit specific antagonist ifenprodil indicating functional expression of NMDA receptors containing subunits NR1 and NR2B. In calcium imaging experiments, the blockade of voltage-gated calcium channels by verapamil abolished AMPA-induced calcium responses but only partially reduced NMDA-evoked transients suggesting the expression of calcium-impermeable, GluR2-containing AMPA receptors. Quantitative real-time PCR showed a predominant expression of subunits NR2A and NR2B (NMDA), GluR2 (AMPA), GluR7 (kainate), and mGluR3 (metabotropic glutamate receptor). Treatment of NPCs with 100 microM NMDA in vitro during proliferation (2 weeks) and differentiation (1 week) increased the amount of tyrosine hydroxylase-immunopositive cells significantly, which was reversed by addition of memantine. These data suggest that NMDA receptors in differentiating human mesencephalic NPCs are important regulators of dopaminergic neurogenesis in vitro.


Subject(s)
Dopamine/metabolism , Embryonic Stem Cells/drug effects , Mesencephalon/cytology , N-Methylaspartate/pharmacology , Neurogenesis/drug effects , Receptors, Glutamate/physiology , Calcium/metabolism , Cells, Cultured , Fetus , Glutamic Acid/pharmacology , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mesencephalon/embryology , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques/methods
17.
PLoS One ; 4(7): e6168, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19584922

ABSTRACT

BACKGROUND: Voltage-gated potassium (K(v)) channels are among the earliest ion channels to appear during brain development, suggesting a functional requirement for progenitor cell proliferation and/or differentiation. We tested this hypothesis, using human neural progenitor cells (hNPCs) as a model system. METHODOLOGY/PRINCIPAL FINDINGS: In proliferating hNPCs a broad spectrum of K(v) channel subtypes was identified using quantitative real-time PCR with a predominant expression of the A-type channel K(v)4.2. In whole-cell patch-clamp recordings K(v) currents were separated into a large transient component characteristic for fast-inactivating A-type potassium channels (I(A)) and a small, sustained component produced by delayed-rectifying channels (I(K)). During differentiation the expression of I(A) as well as A-type channel transcripts dramatically decreased, while I(K) producing delayed-rectifiers were upregulated. Both K(v) currents were differentially inhibited by selective neurotoxins like phrixotoxin-1 and alpha-dendrotoxin as well as by antagonists like 4-aminopyridine, ammoniumchloride, tetraethylammonium chloride and quinidine. In viability and proliferation assays chronic inhibition of the A-type currents severely disturbed the cell cycle and precluded proper hNPC proliferation, while the blockade of delayed-rectifiers by alpha-dendrotoxin increased proliferation. CONCLUSIONS/SIGNIFICANCE: These findings suggest that A-type potassium currents are essential for proper proliferation of immature multipotent hNPCs.


Subject(s)
Ion Channel Gating , Neurons/metabolism , Potassium Channels/metabolism , Stem Cells/metabolism , Cell Proliferation , Cells, Cultured , Humans , Ion Channel Gating/drug effects , Neurons/drug effects , Patch-Clamp Techniques , Polymerase Chain Reaction , Potassium Channel Blockers/pharmacology , Stem Cells/drug effects
18.
Mol Neurodegener ; 4: 25, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19523245

ABSTRACT

Despite a comprehensive mapping of the Parkinson's disease (PD)-related mRNA and protein leucine-rich repeat kinase 2 (LRRK2) in the mammalian brain, its physiological function in healthy individuals remains enigmatic. Based on its structural features and kinase properties, LRRK2 may interact with other proteins involved in signalling pathways. Here, we show a widespread LRRK2 mRNA and/or protein expression in expanded or differentiated human mesencephalic neural progenitor cells (hmNPCs) and in post-mortem substantia nigra PD patients. Using small interfering RNA duplexes targeting LRRK2 in hmNPCs following their differentiation into glia and neurons, we observed a reduced number of dopaminergic neurons due to apoptosis in LRRK2 knockdown samples. LRRK2-deficient hmNPCs exhibited elevated cell cycle- and cell death-related markers. In conclusion, a reduction of LRRK2 expression in hmNPCs severely impaired dopaminergic differentiation and/or survival of dopaminergic neurons most likely via preserving or reactivating the cell cycle.

19.
Biomaterials ; 30(28): 5049-60, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19560816

ABSTRACT

Biofunctional matrices for in vivo tissue engineering strategies must be modifiable in both biomolecular composition and mechanical characteristics. To address this challenge, we present a modular system of biohybrid hydrogels based on covalently cross-linked heparin and star-shaped poly(ethylene glycols) (star-PEG) in which network characteristics can be gradually varied while heparin contents remain constant. Mesh size, swelling and elastic moduli were shown to correlate well with the degree of gel component cross-linking. Additionally, secondary conversion of heparin within the biohybrid gels allowed the covalent attachment of cell adhesion mediating RGD peptides and the non-covalent binding of soluble mitogens such as FGF-2. We applied the biohybrid gels to demonstrate the impact of mechanical and biomolecular cues on primary nerve cells and neural stem cells. The results demonstrate the cell type-specific interplay of synergistic signaling events and the potential of biohybrid materials to selectively stimulate cell fate decisions. These findings suggest important future uses for this material in cell replacement based-therapies for neurodegenerative diseases.


Subject(s)
Heparin/therapeutic use , Hydrogel, Polyethylene Glycol Dimethacrylate/therapeutic use , Neurodegenerative Diseases/therapy , Polyethylene Glycols/therapeutic use , Animals , Cell Survival , Cells, Cultured , Elastic Modulus , Embryonic Stem Cells/cytology , Female , Fibroblast Growth Factor 2/chemistry , Heparin/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Mesencephalon/cytology , Mice , Mice, Inbred C57BL , Neurons/cytology , Oligopeptides/chemistry , Polyethylene Glycols/chemistry , Pregnancy , Prostheses and Implants , Rats , Rats, Wistar
20.
Cell Calcium ; 45(5): 485-98, 2009 May.
Article in English | MEDLINE | ID: mdl-19386359

ABSTRACT

Nucleotides play an important role in brain development and may exert their action via ligand-gated cationic channels or G protein-coupled receptors. Patch-clamp measurements indicated that in contrast to AMPA, ATP did not induce membrane currents in human midbrain derived neuronal progenitor cells (hmNPCs). Various nucleotide agonists concentration-dependently increased [Ca(2+)](i) as measured by the Fura-2 method, with the rank order of potency ATP>ADP>UTP>UDP. A Ca(2+)-free external medium moderately decreased, whereas a depletion of the intracellular Ca(2+) storage sites by cyclopiazonic acid markedly depressed the [Ca(2+)](i) transients induced by either ATP or UTP. Further, the P2Y(1) receptor antagonistic PPADS and MRS 2179, as well as the nucleotide catalyzing enzyme apyrase, allmost abolished the effects of these two nucleotides. However, the P2Y(1,2,12) antagonistic suramin only slightly blocked the action of ATP, but strongly inhibited that of UTP. In agreement with this finding, UTP evoked the release of ATP from hmNPCs in a suramin-, but not PPADS-sensitive manner. Immunocytochemistry indicated the co-localization of P2Y(1,2,4)-immunoreactivities (IR) with nestin-IR at these cells. In conclusion, UTP may induce the release of ATP from hmNPCs via P2Y(2) receptor-activation and thereby causes [Ca(2+)](i) transients by stimulating a P2Y(1)-like receptor.


Subject(s)
Adenine Nucleotides/metabolism , Calcium/metabolism , Mesencephalon/cytology , Neurons/physiology , Stem Cells/physiology , Uracil Nucleotides/metabolism , Animals , Cell Proliferation , Cells, Cultured , Humans , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Neurogenesis/physiology , Neurons/cytology , Patch-Clamp Techniques , Purinergic P2 Receptor Agonists , Receptors, Purinergic P2/metabolism , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...