Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Cell Neurosci ; 16: 917181, 2022.
Article in English | MEDLINE | ID: mdl-35936502

ABSTRACT

Dax-1 (dosage-sensitive sex reversal adrenal hypoplasia congenital region on X-chromosome gene 1) blocks 17ß-estradiol biosynthesis and its knockdown would be expected to increase 17ß-estradiol production. We hypothesized that knockdown of Dax-1 in a conditionally immortalized neural stem cell (NSC) line, MHP36, is a useful approach to increase 17ß-estradiol production. Short hairpin (sh) RNA targeted to Dax-1 in NSCs, namely MHP36-Dax1KD cells, resulted in the degradation of Dax-1 RNA and attenuation of Dax-1 protein expression. In vitro, MHP36-Dax1KD cells exhibited overexpression of aromatase and increased 17ß-estradiol secretion compared to MHP36 cells. As 17ß-estradiol has been shown to promote the efficacy of cell therapy, we interrogated the application of 17ß-estradiol-enriched NSCs in a relevant in vivo disease model. We hypothesized that MHP36-Dax1KD cells will enhance functional recovery after transplantation in a stroke model. C57BL/6 male adult mice underwent ischemia/reperfusion by left middle cerebral artery occlusion for 45 min using an intraluminal thread. Two days later male mice randomly received vehicle, MHP36 cells, MHP36-Dax1KD cells, and MHP36 cells suspended in 17ß-estradiol (100 nm) or 17ß-estradiol alone (100 nm) with serial behavioral testing over 28 days followed by post-mortem histology and blinded analysis. Recovery of sensorimotor function was accelerated and enhanced, and lesion volume was reduced by MHP36-Dax1KD transplants. Regarding mechanisms, immunofluorescence indicated increased synaptic plasticity and neuronal differentiation after MHP36-Dax1KD transplants. In conclusion, knockdown of Dax-1 is a useful target to increase 17ß-estradiol biosynthesis in NSCs and improves functional recovery after stroke in vivo, possibly mediated through neuroprotection and improved synaptic plasticity. Therefore, targeting 17ß-estradiol biosynthesis in stem cells may be a promising therapeutic strategy for enhancing the efficacy of stem cell-based therapies for stroke.

2.
RNA ; 22(10): 1620-30, 2016 10.
Article in English | MEDLINE | ID: mdl-27539784

ABSTRACT

Clonal level random allelic expression imbalance and random monoallelic expression provides cellular heterogeneity within tissues by modulating allelic dosage. Although such expression patterns have been observed in multiple cell types, little is known about when in development these stochastic allelic choices are made. We examine allelic expression patterns in human neural progenitor cells before and after epigenetic reprogramming to induced pluripotency, observing that loci previously characterized by random allelic expression imbalance (0.63% of expressed genes) are generally reset to a biallelic state in induced pluripotent stem cells (iPSCs). We subsequently neuralized the iPSCs and profiled isolated clonal neural stem cells, observing that significant random allelic expression imbalance is reestablished at 0.65% of expressed genes, including novel loci not found to show allelic expression imbalance in the original parental neural progenitor cells. Allelic expression imbalance was associated with altered DNA methylation across promoter regulatory regions, with clones characterized by skewed allelic expression being hypermethylated compared to their biallelic sister clones. Our results suggest that random allelic expression imbalance is established during lineage commitment and is associated with increased DNA methylation at the gene promoter.


Subject(s)
Allelic Imbalance , Cellular Reprogramming , Epigenesis, Genetic , Cells, Cultured , DNA Methylation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism
3.
Cell Signal ; 27(3): 621-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25479589

ABSTRACT

Members of the cyclic-AMP response-element binding protein (CREB) transcription factor family regulate the expression of genes needed for long-term memory formation. Loss of Notch impairs long-term, but not short-term, memory in flies and mammals. We investigated if the Notch-1 (N1) exerts an effect on CREB-dependent gene transcription. We observed that N1 inhibits CREB mediated activation of cyclic-AMP response element (CRE) containing promoters in a γ-secretase-dependent manner. We went on to find that the γ-cleaved N1 intracellular domain (N1ICD) sequesters nuclear CREB1α, inhibits cAMP/PKA-mediated neurite outgrowth and represses the expression of specific CREB regulated genes associated with learning and memory in primary cortical neurons. Similar transcriptional effects were observed with the N2ICD, N3ICD and N4ICDs. Together, these observations indicate that the effects of Notch on learning and memory are, at least in part, via an effect on CREB-regulated gene expression.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Receptor, Notch1/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/pharmacology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Female , HEK293 Cells , Humans , Memory, Long-Term/physiology , Mice , Mice, Inbred C57BL , Neurites/physiology , Neurons/cytology , Neurons/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Rats , Receptor, Notch1/chemistry , Receptor, Notch1/genetics , Transcription, Genetic/drug effects
4.
Psychopharmacology (Berl) ; 231(6): 1079-88, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23839283

ABSTRACT

Until now, models of psychiatric diseases have typically been animal models. Whether they were to be used to further understand the pathophysiology of the disorder, or as drug discovery tools, animal models have been the choice of preference in mimicking psychiatric disorders in an experimental setting. While there have been cellular models, they have generally been lacking in validity. This situation is changing with the advent of patient-specific induced pluripotent stem cells (iPSCs). In this article, we give a methodological evaluation of the current state of the iPS technology with reference to our own work in generating patient-specific iPSCs for the study of autistic spectrum disorder (ASD). In addition, we will give a broader perspective on the validity of this technology and to what extent it can be expected to complement animal models of ASD in the coming years.


Subject(s)
Child Development Disorders, Pervasive , Induced Pluripotent Stem Cells , Models, Biological , Animals , Child Development Disorders, Pervasive/genetics , Child Development Disorders, Pervasive/physiopathology , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/physiology , Stem Cell Research
5.
Cell Stem Cell ; 13(6): 706-19, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24139759

ABSTRACT

During embryogenesis, the Activin/Nodal pathway promotes the mesendodermal lineage and inhibits neural fate. The molecular mechanisms underlying this role of the Activin/Nodal pathway are not clear. In this study, we report a role for protein tyrosine phosphatase 1B (PTP1B) in Activin-mediated early fate decisions during ESC differentiation and show that PTP1B acts as an effector of the Activin pathway to specify mesendodermal or neural fate. We found that the Activin/ALK4 pathway directly recruits PTP1B and stimulates its release from the endoplasmic reticulum through ALK4-mediated cleavage. Subsequently, PTP1B suppresses p-ERK1/2 signaling to inhibit neural specification and promote mesendodermal commitment. These findings suggest that a noncanonical Activin signaling pathway functions in lineage specification of mouse and human embryonic stem cells.


Subject(s)
Activins/metabolism , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Neurons/cytology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Signal Transduction , Activin Receptors, Type I/chemistry , Activin Receptors, Type I/metabolism , Amino Acid Sequence , Animals , Benzamides/pharmacology , Cell Differentiation/drug effects , Cell Lineage/drug effects , Dioxoles/pharmacology , Embryonic Stem Cells/drug effects , Endoderm/cytology , Endoderm/drug effects , Endoderm/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Molecular Sequence Data , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Signal Transduction/drug effects , Smad2 Protein/metabolism
6.
Stem Cells ; 28(11): 1950-60, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20872847

ABSTRACT

Neural induction is the first step in the formation of the vertebrate central nervous system. The emerging consensus of the mechanisms underlying neural induction is the combined influences from inhibiting bone morphogenetic protein (BMP) signaling and activating fibroblast growth factor (FGF)/Erk signaling, which act extrinsically via either autocrine or paracrine fashions. However, do intrinsic forces (cues) exist and do they play decisive roles in neural induction? These questions remain to be answered. Here, we have identified a novel neural initiator, neuronatin (Nnat), which acts as an intrinsic factor to promote neural fate in mammals and Xenopus. ESCs lacking this intrinsic factor fail to undergo neural induction despite the inhibition of the BMP pathway. We show that Nnat initiates neural induction in ESCs through increasing intracellular Ca(2+) ([Ca(2+) ](i)) by antagonizing Ca(2+) -ATPase isoform 2 (sarco/endoplasmic reticulum Ca(2+) -ATPase isoform 2) in the endoplasmic reticulum, which in turn increases the phosphorylation of Erk1/2 and inhibits the BMP4 pathway and leads to neural induction in conjunction with FGF/Erk pathway.


Subject(s)
Calcium Signaling/physiology , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Blotting, Western , Calcium Signaling/genetics , Cell Differentiation/genetics , Flow Cytometry , Immunohistochemistry , Immunoprecipitation , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Phosphorylation , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
7.
Stem Cells Dev ; 19(4): 579-92, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19645571

ABSTRACT

In this study, we have used an in vitro co-culture system to investigate the competency of a conditionally immortalized multipotential neural progenitor cell line (MHP36) to adopt "dorsal" or "striatal" telencephalic fates. We report that MHP36 cells, unlike primary fetal neural progenitors cells, do not express either dorsal or ventral telencephalic positional specification genes; at both the mRNA and protein levels, but that they quickly turn on expression of the appropriate set of proteins when cultured in either a dorsal (cortical) or a ventral (striatal) environment. This control has 2 components: transcriptional activation of positional specification genes, and translational control whereby only the appropriate set of mRNAs appears as immunoreactive protein. We show furthermore that this positional specification gene expression is modulated by the RNA-binding protein Musashi1. We postulate that it is the ability of MHP36 cells to adopt either cortical or striatal positional specification that is key to their functional efficacy in a number of models of neurological disease.


Subject(s)
Basal Ganglia , Cell Lineage , Cerebral Cortex , Fetal Stem Cells , Multipotent Stem Cells , Nerve Tissue Proteins/genetics , Neurons , RNA-Binding Proteins/genetics , Animals , Basal Ganglia/cytology , Basal Ganglia/metabolism , Cell Differentiation , Cell Line , Cell Lineage/genetics , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Coculture Techniques , Fetal Stem Cells/cytology , Fetal Stem Cells/metabolism , Gene Expression Regulation, Developmental , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Organogenesis/genetics , Rats , Transcriptional Activation
8.
J Neurochem ; 99(3): 989-99, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17076661

ABSTRACT

p73, like Notch, has been implicated in neurodevelopment and in the maintenance of the mature central nervous system. In this study, by the use of reporter-gene assays, we demonstrate that C-promoter binding factor-1 (CBF-1)-dependent gene transcription driven by the Notch-1 intracellular domain (N1(ICD)) is potently antagonized by exogenously expressed transactivating (TA) p73 splice variants in SH-SY5Y neuroblastomas and in primary neurones. Time course analysis indicated that the inhibitory effects of TAp73 are direct and are not mediated via the product of a downstream target gene. We found that endogenous TAp73 stabilized by either c-Abl or cisplatin treatment also potently antagonized N1(ICD)/CBF-1-dependent gene transcription. Furthermore, western blotting revealed that exogenous TAp73 suppressed endogenous hairy and enhancer of split-1 (HES-1) protein levels and antagonized the increase in HES-1 protein induced by exogenous N1(ICD) expression. Evidence of a direct physical interaction between N1(ICD) and TAp73alpha was demonstrated by co-immunoprecipitation. Using Notch deletion constructs, we demonstrate that TAp73alpha binds the N1(ICD) in a region C-terminal of aa 2094. Interestingly, DeltaNp73alpha and TAp73alpha(R292H) also co-purified with N1(ICD), but neither inhibited N1(ICD)/CBF-1-dependent transcription. This suggests that an intact transactivation (TA) domain and the ability to bind DNA are necessary for TAp73 to antagonize Notch signalling. Finally we found that TAp73alpha reversed the N1(ICD)-mediated repression of retinoic acid-induced differentiation of SH-SY5Y neuroblastomas, providing functional evidence for an inhibitory effect of TAp73alpha on notch signalling. Collectively, these findings may have ramifications for neurodevelopment, neurodegeneration and oncogenesis.


Subject(s)
Brain Neoplasms/physiopathology , DNA-Binding Proteins/physiology , Neuroblastoma/physiopathology , Neurons/physiology , Nuclear Proteins/physiology , Receptor, Notch1/physiology , Tumor Suppressor Proteins/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Cycle Proteins/genetics , Cell Differentiation/drug effects , Cell Line, Tumor , DNA-Binding Proteins/biosynthesis , Humans , Immunoprecipitation , Luciferases/metabolism , Microscopy, Confocal , Nuclear Proteins/biosynthesis , Repressor Proteins/genetics , Signal Transduction/physiology , Transfection , Tretinoin/antagonists & inhibitors , Tretinoin/pharmacology , Tumor Suppressor Proteins/biosynthesis
9.
Brain Res Dev Brain Res ; 151(1-2): 13-23, 2004 Jul 19.
Article in English | MEDLINE | ID: mdl-15246688

ABSTRACT

Neural stem cells (NSCs) are believed to repair brain damage primarily through cell replacement: i.e., the ability to regenerate lost neurons and glia in a site-specific fashion. The neural stem cell line, MHP36, has been shown to have this capacity, but we have little idea of the molecular mechanisms that control the differentiation of such cells during brain repair. In this study we show that an early event in the differentiation of MHP36 cells, both in vivo and in vitro, is the loss of expression of the intermediate filament protein, nestin. We use a co-culture assay to show that loss of nestin is fast, being detectable after just 1 h and complete in 4 h, and is controlled by proteasome degradation rather than down-regulation of de novo nestin synthesis. We also show that nestin loss is regulated by Notch, and mediated by cell contact.


Subject(s)
Acetylcysteine/analogs & derivatives , Cysteine Endopeptidases/metabolism , Intermediate Filament Proteins/metabolism , Membrane Proteins/physiology , Multienzyme Complexes/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Signal Transduction/physiology , Stem Cells/metabolism , Acetylcysteine/pharmacology , Animals , Cell Count/methods , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques/methods , Cricetinae , Cricetulus , Cysteine Proteinase Inhibitors/pharmacology , Embryo, Mammalian , Fluorescent Dyes , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins , Hippocampus/metabolism , Immunohistochemistry/methods , Intermediate Filament Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Nerve Tissue Proteins/genetics , Nestin , Neuroglia/metabolism , Organic Chemicals , Phosphopyruvate Hydratase/metabolism , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Notch , Reverse Transcriptase Polymerase Chain Reaction/methods , Stem Cell Transplantation/methods , Temperature , Time Factors , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...