Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Ther ; 27(7): 1299-1312, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31043343

ABSTRACT

In this study, we found that undifferentiated human pluripotent stem cells (hPSCs; up to 30% of total cells) present in the cultures of neural stem or precursor cells (NPCs) completely disappeared within several days when cultured under neural differentiation culture conditions. Intriguingly, the disappearance of undifferentiated cells was not due to cell death but was instead mediated by neural conversion of hPSCs. Based on these findings, we propose pre-conditioning of donor NPC cultures under terminal differentiation culture conditions as a simple but efficient method of eliminating undifferentiated cells to treat neurologic disorders. In addition, we could establish a new neural differentiation protocol, in which undifferentiated hPSCs co-cultured with NPCs become differentiated neurons or NPCs in an extremely efficient, fast, and reproducible manner across the hESC and human-induced pluripotent stem cell (hiPSC) lines.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Cell Line, Tumor , Coculture Techniques , Fibroblast Growth Factor 2/metabolism , Green Fluorescent Proteins/metabolism , Humans , Nervous System Diseases/therapy , Octamer Transcription Factor-3/metabolism , Phenotype , Stem Cell Transplantation
2.
Stem Cell Reports ; 9(4): 1192-1206, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28943252

ABSTRACT

Cultured neural stem/precursor cells (NSCs) are regarded as a potential systematic cell source to treat Parkinson's disease (PD). However, the therapeutic potential of these cultured NSCs is lost during culturing. Here, we show that treatment of vitamin C (VC) enhances generation of authentic midbrain-type dopamine (mDA) neurons with improved survival and functions from ventral midbrain (VM)-derived NSCs. VC acted by upregulating a series of mDA neuron-specific developmental and phenotype genes via removal of DNA methylation and repressive histone code (H3K9m3, H3K27m3) at associated gene promoter regions. Notably, the epigenetic changes induced by transient VC treatment were sustained long after VC withdrawal. Accordingly, transplantation of VC-treated NSCs resulted in improved behavioral restoration, along with enriched DA neuron engraftment, which faithfully expressed midbrain-specific markers in PD model rats. These results indicate that VC treatment to donor NSCs could be a simple, efficient, and safe therapeutic strategy for PD in the future.


Subject(s)
Ascorbic Acid/pharmacology , Epigenesis, Genetic/drug effects , Gene Expression Regulation/drug effects , Mesencephalon/drug effects , Mesencephalon/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Animals , Behavior, Animal , Biomarkers , Cell Differentiation , Cell Proliferation , Cell- and Tissue-Based Therapy , Cells, Cultured , Disease Models, Animal , Dopaminergic Neurons/cytology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Female , Neural Stem Cells/cytology , Neurogenesis , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/therapy , Phenotype , Presynaptic Terminals/metabolism , Promoter Regions, Genetic , Rats , Stem Cell Transplantation
3.
Brain ; 139(Pt 10): 2722-2739, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27538419

ABSTRACT

The original properties of tissue-specific stem cells, regardless of their tissue origins, are inevitably altered during in vitro culturing, lessening the clinical and research utility of stem cell cultures. Specifically, neural stem cells derived from the ventral midbrain lose their dopamine neurogenic potential, ventral midbrain-specific phenotypes, and repair capacity during in vitro cell expansion, all of which are critical concerns in using the cultured neural stem cells in therapeutic approaches for Parkinson's disease. In this study, we observed that the culture-dependent changes of neural stem cells derived from the ventral midbrain coincided with loss of RNA-binding protein LIN28A expression. When LIN28A expression was forced and sustained during neural stem cell expansion using an inducible expression-vector system, loss of dopamine neurogenic potential and midbrain phenotypes after long-term culturing was blocked. Furthermore, dopamine neurons that differentiated from neural stem cells exhibited remarkable survival and resistance against toxic insults. The observed effects were not due to a direct action of LIN28A on the differentiated dopamine neurons, but rather its action on precursor neural stem cells as exogene expression was switched off in the differentiating/differentiated cultures. Remarkable and reproducible behavioural recovery was shown in all Parkinson's disease rats grafted with neural stem cells expanded with LIN28A expression, along with extensive engraftment of dopamine neurons expressing mature neuronal and midbrain-specific markers. These findings suggest that LIN28A expression during stem cell expansion could be used to prepare therapeutically competent donor cells.

4.
Sci Rep ; 6: 32025, 2016 08 24.
Article in English | MEDLINE | ID: mdl-27554447

ABSTRACT

The intracellular Raf-Erk signaling pathway is activated during neural stem cell (NSC) proliferation, and neuronal and astrocytic differentiation. A key question is how this signal can evoke multiple and even opposing NSC behaviors. We show here, using a constitutively active Raf (ca-Raf), that Raf-Erk activation in NSCs induces neuronal differentiation in a cell-autonomous manner. By contrast, it causes NSC proliferation and the formation of astrocytes in an extrinsic autocrine/paracrine manner. Thus, treatment of NSCs with medium (CM) conditioned in ca-Raf-transduced NSCs (Raf-CM; RCM) became activated to form proliferating astrocytes resembling radial glial cells (RGCs) or adult-type NSCs. Infusion of Raf-CM into injured mouse brains caused expansion of the NSC population in the subventricular zone, followed by the formation of new neurons that migrated to the damaged site. Our study shows an example how molecular mechanisms dissecting NSC behaviors can be utilized to develop regenerative therapies in brain disorders.


Subject(s)
Brain/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Neural Stem Cells/metabolism , Regeneration/physiology , raf Kinases/metabolism , Animals , Astrocytes/cytology , Brain/embryology , Cell Count , Cell Differentiation , Cells, Cultured , Culture Media, Conditioned/pharmacology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/cytology , Neurons/cytology , Neurons/metabolism , Rats, Sprague-Dawley
6.
Stem Cell Res ; 15(3): 608-613, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26513556

ABSTRACT

Culturing human embryonic stem and induced pluripotent stem cells (hESCs/iPSCs) is one of the most costly and labor-intensive tissue cultures, as media containing expensive factors/cytokines should be changed every day to maintain and propagate undifferentiated hESCs/iPSCs in vitro. We recently reported that doxycycline, an anti-bacterial agent, had dramatic effects on hESC/iPSC survival and promoted self-renewal. In this study, we extended the effects of doxycycline to a more practical issue to save cost and labor in hESC/iPSC cultures. Regardless of cultured cell conditions, hESCs/iPSCs in doxycycline-supplemented media were viable and proliferating for at least 3 days without media change, while none or few viable cells were detected in the absence of doxycycline in the same conditions. Thus, hESCs/iPSCs supplemented with doxycycline can be cultured for a long period of time with media changes at 3-day intervals without altering their self-renewal and pluripotent properties, indicating that doxycycline supplementation can reduce the frequency of media changes and the amount of media required by 1/3. These findings strongly encourage the use of doxycycline to save cost and labor in culturing hESCs/iPSCs.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Doxycycline/therapeutic use , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Anti-Bacterial Agents/administration & dosage , Cells, Cultured , Doxycycline/administration & dosage , Embryonic Stem Cells/cytology , Humans , Pluripotent Stem Cells/cytology , Time Factors
7.
EMBO Mol Med ; 7(5): 510-25, 2015 May.
Article in English | MEDLINE | ID: mdl-25759364

ABSTRACT

Use of the physiological mechanisms promoting midbrain DA (mDA) neuron survival seems an appropriate option for developing treatments for Parkinson's disease (PD). mDA neurons are specifically marked by expression of the transcription factors Nurr1 and Foxa2. We show herein that Nurr1 and Foxa2 interact to protect mDA neurons against various toxic insults, but their expression is lost during aging and degenerative processes. In addition to their proposed cell-autonomous actions in mDA neurons, forced expression of these factors in neighboring glia synergistically protects degenerating mDA neurons in a paracrine mode. As a consequence of these bimodal actions, adeno-associated virus (AAV)-mediated gene delivery of Nurr1 and Foxa2 in a PD mouse model markedly protected mDA neurons and motor behaviors associated with nigrostriatal DA neurotransmission. The effects of the combined gene delivery were dramatic, highly reproducible, and sustained for at least 1 year, suggesting that expression of these factors is a promising approach in PD therapy.


Subject(s)
Dopaminergic Neurons/physiology , Genetic Therapy/methods , Hepatocyte Nuclear Factor 3-beta/metabolism , Mesencephalon/pathology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Parkinson Disease/pathology , Parkinson Disease/therapy , Animals , Cells, Cultured , Disease Models, Animal , Hepatocyte Nuclear Factor 3-beta/genetics , Male , Mice, Inbred ICR , Neuroglia/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Treatment Outcome
8.
Stem Cells ; 33(4): 1320-32, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25535150

ABSTRACT

Intracellular Vitamin C (VC) is maintained at high levels in the developing brain by the activity of sodium-dependent VC transporter 2 (Svct2), suggesting specific VC functions in brain development. A role of VC as a cofactor for Fe(II)-2-oxoglutarate-dependent dioxygenases has recently been suggested. We show that VC supplementation in neural stem cell cultures derived from embryonic midbrains greatly enhanced differentiation toward midbrain-type dopamine (mDA) neurons, the neuronal subtype associated with Parkinson's disease. VC induced gain of 5-hydroxymethylcytosine (5hmC) and loss of H3K27m3 in DA phenotype gene promoters, which are catalyzed by Tet1 and Jmjd3, respectively. Consequently, VC enhanced DA phenotype gene transcriptions in the progenitors by Nurr1, a transcription factor critical for mDA neuron development, to be more accessible to the gene promoters. Further mechanism studies including Tet1 and Jmjd3 knockdown/inhibition experiments revealed that both the 5hmC and H3K27m3 changes, specifically in the progenitor cells, are indispensible for the VC-mediated mDA neuron differentiation. We finally show that in Svct2 knockout mouse embryos, mDA neuron formation in the developing midbrain decreased along with the 5hmC/H3k27m3 changes. These findings together indicate an epigenetic role of VC in midbrain DA neuron development.


Subject(s)
Ascorbic Acid/pharmacology , Cell Differentiation/physiology , Dioxygenases/metabolism , Dopaminergic Neurons/metabolism , Epigenesis, Genetic/physiology , Jumonji Domain-Containing Histone Demethylases/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Dopaminergic Neurons/drug effects , Epigenesis, Genetic/drug effects , Mesencephalon/cytology , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/drug effects , Neurogenesis/physiology , Rats , Rats, Sprague-Dawley
9.
Stem Cell Reports ; 3(2): 353-64, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25254347

ABSTRACT

We here report that doxycycline, an antibacterial agent, exerts dramatic effects on human embryonic stem and induced pluripotent stem cells (hESC/iPSCs) survival and self-renewal. The survival-promoting effect was also manifest in cultures of neural stem cells (NSCs) derived from hESC/iPSCs. These doxycycline effects are not associated with its antibacterial action, but mediated by direct activation of a PI3K-AKT intracellular signal. These findings indicate doxycycline as a useful supplement for stem cell cultures, facilitating their growth and maintenance.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Proliferation/drug effects , Doxycycline/pharmacology , Pluripotent Stem Cells/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Embryonic Stem Cells/cytology , Humans , Karyotyping , Neural Stem Cells/cytology , Neurons/cytology , Phosphatidylinositol 3-Kinases/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
10.
Development ; 141(4): 761-72, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24496614

ABSTRACT

Understanding how dopamine (DA) phenotypes are acquired in midbrain DA (mDA) neuron development is important for bioassays and cell replacement therapy for mDA neuron-associated disorders. Here, we demonstrate a feed-forward mechanism of mDA neuron development involving Nurr1 and Foxa2. Nurr1 acts as a transcription factor for DA phenotype gene expression. However, Nurr1-mediated DA gene expression was inactivated by forming a protein complex with CoREST, and then recruiting histone deacetylase 1 (Hdac1), an enzyme catalyzing histone deacetylation, to DA gene promoters. Co-expression of Nurr1 and Foxa2 was established in mDA neuron precursor cells by a positive cross-regulatory loop. In the presence of Foxa2, the Nurr1-CoREST interaction was diminished (by competitive formation of the Nurr1-Foxa2 activator complex), and CoREST-Hdac1 proteins were less enriched in DA gene promoters. Consequently, histone 3 acetylation (H3Ac), which is responsible for open chromatin structures, was strikingly increased at DA phenotype gene promoters. These data establish the interplay of Nurr1 and Foxa2 as the crucial determinant for DA phenotype acquisition during mDA neuron development.


Subject(s)
Dopaminergic Neurons/physiology , Epigenesis, Genetic/physiology , Gene Expression Regulation/physiology , Hepatocyte Nuclear Factor 3-beta/metabolism , Mesencephalon/cytology , Neurogenesis/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Analysis of Variance , Animals , Chromatin Immunoprecipitation , Co-Repressor Proteins , Dopaminergic Neurons/metabolism , Fluorescent Antibody Technique , Genetic Vectors , Histone Deacetylase 1/metabolism , Immunoprecipitation , Mice , Microarray Analysis , Nerve Tissue Proteins/metabolism , Real-Time Polymerase Chain Reaction , Repressor Proteins/metabolism , Retroviridae , Transduction, Genetic
11.
Hum Mol Genet ; 23(3): 657-67, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24087792

ABSTRACT

DNA methylation and hydroxymethylation have been implicated in normal development and differentiation, but our knowledge is limited about the genome-wide distribution of 5-methylcytosine (5 mC) and 5-hydroxymethylcytosine (5 hmC) during cellular differentiation. Using an in vitro model system of gradual differentiation of human embryonic stem (hES) cells into ventral midbrain-type neural precursor cells and terminally into dopamine neurons, we observed dramatic genome-wide changes in 5 mC and 5 hmC patterns during lineage commitment. The 5 hmC pattern was dynamic in promoters, exons and enhancers. DNA hydroxymethylation within the gene body was associated with gene activation. The neurogenesis-related genes NOTCH1, RGMA and AKT1 acquired 5 hmC in the gene body and were up-regulated during differentiation. DNA methylation in the promoter was associated with gene repression. The pluripotency-related genes POU5F1, ZFP42 and HMGA1 acquired 5 mC in their promoters and were down-regulated during differentiation. Promoter methylation also acted as a locking mechanism to maintain gene silencing. The mesoderm development-related genes NKX2-8, TNFSF11 and NFATC1 acquired promoter methylation during neural differentiation even though they were already silenced in hES cells. Our findings will help elucidate the molecular mechanisms underlying lineage-specific differentiation of pluripotent stem cells during human embryonic development.


Subject(s)
Cell Differentiation/physiology , DNA Methylation , Embryonic Stem Cells/physiology , Neurons/physiology , 5-Methylcytosine/metabolism , Cell Lineage/genetics , Cells, Cultured , Cytosine/analogs & derivatives , Cytosine/metabolism , Embryonic Stem Cells/cytology , GPI-Linked Proteins/genetics , Gene Expression Regulation , Gene Silencing , Homeodomain Proteins/genetics , Humans , Mesoderm/physiology , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Neurons/cytology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/genetics , RANK Ligand/genetics , Receptor, Notch1/genetics , Transcription Factors/genetics
12.
Proc Natl Acad Sci U S A ; 110(35): E3281-90, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23918355

ABSTRACT

The future of safe cell-based therapy rests on overcoming teratoma/tumor formation, in particular when using human pluripotent stem cells (hPSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). Because the presence of a few remaining undifferentiated hPSCs can cause undesirable teratomas after transplantation, complete removal of these cells with no/minimal damage to differentiated cells is a prerequisite for clinical application of hPSC-based therapy. Having identified a unique hESC signature of pro- and antiapoptotic gene expression profile, we hypothesized that targeting hPSC-specific antiapoptotic factor(s) (i.e., survivin or Bcl10) represents an efficient strategy to selectively eliminate pluripotent cells with teratoma potential. Here we report the successful identification of small molecules that can effectively inhibit these antiapoptotic factors, leading to selective and efficient removal of pluripotent stem cells through apoptotic cell death. In particular, a single treatment of hESC-derived mixed population with chemical inhibitors of survivin (e.g., quercetin or YM155) induced selective and complete cell death of undifferentiated hPSCs. In contrast, differentiated cell types (e.g., dopamine neurons and smooth-muscle cells) derived from hPSCs survived well and maintained their functionality. We found that quercetin-induced selective cell death is caused by mitochondrial accumulation of p53 and is sufficient to prevent teratoma formation after transplantation of hESC- or hiPSC-derived cells. Taken together, these results provide the "proof of concept" that small-molecule targeting of hPSC-specific antiapoptotic pathway(s) is a viable strategy to prevent tumor formation by selectively eliminating remaining undifferentiated pluripotent cells for safe hPSC-based therapy.


Subject(s)
Pluripotent Stem Cells/cytology , Small Molecule Libraries , Teratoma/pathology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Apoptosis , B-Cell CLL-Lymphoma 10 Protein , Cell Differentiation , Cells, Cultured , Gene Expression Profiling , Humans , Imidazoles/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Mitochondria/metabolism , Naphthoquinones/pharmacology , Pluripotent Stem Cells/metabolism , Stem Cell Transplantation , Survivin , Teratoma/genetics , Tumor Suppressor Protein p53/metabolism
13.
Development ; 139(13): 2447-51, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22627286

ABSTRACT

Neural stem/progenitor cell (NSC/NPC) cultures can be a source of dopamine (DA) neurons for experimental and transplantation purposes. Nurr1, a steroid receptor transcription factor, can overcome the limitations associated with differentiation of cultured NPCs into DA neurons. However, forced Nurr1 expression in NPC cultures generates non-neuronal and/or immature DA cells. We show here that the Nurr1 level and period of expression crucially affect the differentiation and maturation of Nurr1-induced DA neurons. Mature DA neurons were generated by manipulating Nurr1 expression patterns to resemble those in the developing midbrain.


Subject(s)
Dopaminergic Neurons/metabolism , Mesencephalon/growth & development , Neurogenesis , Nuclear Receptor Subfamily 4, Group A, Member 2/biosynthesis , Animals , Cells, Cultured , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Dopamine/biosynthesis , Embryonic Development , Mesencephalon/metabolism , Rats
14.
Stem Cells ; 29(11): 1861-73, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21922608

ABSTRACT

Understanding midbrain dopamine (DA) neuron differentiation is of importance, because of physiological and clinical implications of this neuronal subtype. We show that prolonged membrane depolarization induced by KCl treatment promotes DA neuron differentiation from neural precursor cells (NPCs) derived from embryonic ventral midbrain (VM). Interestingly, the depolarization-induced increase of DA neuron yields was not abolished by L-type calcium channel blockers, along with no depolarization-mediated change of intracellular calcium level in the VM-derived NPCs (VM-NPCs), suggesting that the depolarization effect is due to a calcium-independent mechanism. Experiments with labeled DA neuron progenitors indicate that membrane depolarization acts at the differentiation fate determination stage and promotes the expression of DA phenotype genes (tyrosine hydroxylase [TH] and DA transporter [DAT]). Recruitment of Nurr1, a transcription factor crucial for midbrain DA neuron development, to the promoter of TH gene was enhanced by depolarization, along with increases of histone 3 acetylation (H3Ac) and trimethylation of histone3 on lysine 4 (H3K4m3), and decreases of H3K9m3 and H3K27m3 in the consensus Nurr1 binding regions of TH promoter. Depolarization stimuli on differentiating VM-NPCs also induced dissociation of methyl CpG binding protein 2 and related repressor complex molecules (repressor element-1 silencing transcription factor corepressor and histone deacetylase 1) from the CpG sites of TH and DAT promoters. Based on these findings, we suggest that membrane depolarization promotes DA neuron differentiation by opening chromatin structures surrounding DA phenotype genes and inhibiting the binding of corepressors, thus allowing transcriptional activators such as Nurr1 to access DA neuron differentiation gene promoter regions.


Subject(s)
Cell Differentiation/physiology , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Histones/metabolism , Mesencephalon/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Calcium/metabolism , Cell Differentiation/genetics , Cell Proliferation , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Histones/genetics , Immunohistochemistry , Methyl-CpG-Binding Protein 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Rats , Real-Time Polymerase Chain Reaction
15.
J Clin Invest ; 121(6): 2326-35, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21576821

ABSTRACT

Parkinson disease (PD) involves the selective loss of midbrain dopamine (mDA) neurons and is a possible target disease for stem cell-based therapy. Human induced pluripotent stem cells (hiPSCs) are a potentially unlimited source of patient-specific cells for transplantation. However, it is critical to evaluate the safety of hiPSCs generated by different reprogramming methods. Here, we compared multiple hiPSC lines derived by virus- and protein-based reprogramming to human ES cells (hESCs). Neuronal precursor cells (NPCs) and dopamine (DA) neurons delivered from lentivirus-based hiPSCs exhibited residual expression of exogenous reprogramming genes, but those cells derived from retrovirus- and protein-based hiPSCs did not. Furthermore, NPCs derived from virus-based hiPSCs exhibited early senescence and apoptotic cell death during passaging, which was preceded by abrupt induction of p53. In contrast, NPCs derived from hESCs and protein-based hiPSCs were highly expandable without senescence. DA neurons derived from protein-based hiPSCs exhibited gene expression, physiological, and electrophysiological properties similar to those of mDA neurons. Transplantation of these cells into rats with striatal lesions, a model of PD, significantly rescued motor deficits. These data support the clinical potential of protein-based hiPSCs for personalized cell therapy of PD.


Subject(s)
Cellular Reprogramming , Dopamine/metabolism , Induced Pluripotent Stem Cells/physiology , Kruppel-Like Transcription Factors/physiology , Neurons/cytology , Octamer Transcription Factor-3/physiology , Parkinsonian Disorders/surgery , Proto-Oncogene Proteins c-myc/physiology , SOXB1 Transcription Factors/physiology , Animals , Apoptosis , Arginine , Cell Differentiation , Cell Line/transplantation , Cell Lineage , Cellular Senescence , Gene Expression Regulation, Developmental , Genes, p53 , Genetic Vectors/pharmacology , Humans , Induced Pluripotent Stem Cells/transplantation , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Lentivirus/physiology , Neurons/metabolism , Octamer Transcription Factor-3/genetics , Proto-Oncogene Proteins c-myc/genetics , Rats , Retroviridae/physiology , SOXB1 Transcription Factors/genetics , Tumor Suppressor Protein p53/biosynthesis
16.
Stem Cells ; 28(3): 501-12, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20049900

ABSTRACT

Effective dopamine (DA) neuron differentiation from neural precursor cells (NPCs) is prerequisite for precursor/stem cell-based therapy of Parkinson's disease (PD). Nurr1, an orphan nuclear receptor, has been reported as a transcription factor that can drive DA neuron differentiation from non-dopaminergic NPCs in vitro. However, Nurr1 alone neither induces full neuronal maturation nor expression of proteins found specifically in midbrain DA neurons. In addition, Nurr1 expression is inefficient in inducing DA phenotype expression in NPCs derived from certain species such as mouse and human. We show here that Foxa2, a forkhead transcription factor whose role in midbrain DA neuron development was recently revealed, synergistically cooperates with Nurr1 to induce DA phenotype acquisition, midbrain-specific gene expression, and neuronal maturation. Thus, the combinatorial expression of Nurr1 and Foxa2 in NPCs efficiently yielded fully differentiated nigral (A9)-type midbrain neurons with clearly detectable DA neuronal activities. The effects of Foxa2 in DA neuron generation were observed regardless of the brain regions or species from which NPCs were derived. Furthermore, DA neurons generated by ectopic Foxa2 expression were more resistant to toxins. Importantly, Foxa2 expression resulted in a rapid cell cycle exit and reduced cell proliferation. Consistently, transplantation of NPCs transduced with Nurr1 and Foxa2 generated grafts enriched with midbrain-type DA neurons but reduced number of proliferating cells, and significantly reversed motor deficits in a rat PD model. Our findings can be applied to ongoing attempts to develop an efficient and safe precursor/stem cell-based therapy for PD.


Subject(s)
Cell Differentiation/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Neurons/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Stem Cell Transplantation/methods , Stem Cells/metabolism , Animals , Cell Proliferation , Cell Survival/genetics , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Dopamine/metabolism , Humans , Mice , Neurogenesis/genetics , Neurons/cytology , Neurons/transplantation , Parkinson Disease/surgery , Phenotype , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Substantia Nigra/cytology , Substantia Nigra/metabolism , Transfection/methods , Treatment Outcome
17.
Stem Cells ; 27(9): 2238-46, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19522012

ABSTRACT

Nurr1 is a transcription factor specific for the development and maintenance of the midbrain dopamine (DA) neurons. Exogenous Nurr1 in neural precursor (NP) cells induces the differentiation of DA neurons in vitro that are capable of reversing motor dysfunctions in a rodent model for Parkinson disease. The promise of this therapeutic approach, however, is unclear due to poor cell survival and phenotype loss of DA cells after transplantation. We herein demonstrate that Nurr1 proteins undergo ubiquitin-proteasome-system-mediated degradation in differentiating NP cells. The degradation process is activated by a direct Akt-mediated phosphorylation of Nurr1 proteins and can be prevented by abolishing the Akt-target sequence in Nurr1 (Nurr1(Akt)). Overexpression of Nurr1(Akt) in NP cells yielded DA neurons in which Nurr1 protein levels were maintained for prolonged periods. The sustained Nurr1 expression endowed the Nurr1(Akt)-induced DA neurons with resistance to toxic stimuli, enhanced survival, and sustained DA phenotypes in vitro and in vivo after transplantation.


Subject(s)
Dopamine/metabolism , Neurons/cytology , Neurons/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Blotting, Western , Butadienes/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cell Survival/genetics , Cell Survival/physiology , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Immunoprecipitation , Mesencephalon/cytology , Morpholines/pharmacology , Nitriles/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Protein Stability/drug effects , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...