Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters











Publication year range
1.
Horm Metab Res ; 48(2): 77-82, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26783739

ABSTRACT

The endocrine system involves communication among different tissues in distinct organs, including the pancreas and components of the Hypothalamic-Pituitary-Adrenal Axis. The molecular mechanisms underlying these complex interactions are a subject of intense study as they may hold clues for the progression and treatment of a variety of metabolic and degenerative diseases. A plethora of signaling pathways, activated by hormones and other endocrine factors have been implicated in this communication. Recent advances in the stem cell field introduce a new level of complexity: adult progenitor cells appear to utilize distinct signaling pathways than the more mature cells in the tissue they co-reside. It is therefore important to elucidate the signal transduction requirements of adult progenitor cells in addition to those of mature cells. Recent evidence suggests that a common non-canonical signaling pathway regulates adult progenitors in several different tissues, rendering it as a potentially valuable starting point to explore their biology. The STAT3-Ser/Hes3 Signaling Axis was first identified as a major regulator of neural stem cells and, subsequently, cancer stem cells. In the endocrine/neuroendocrine system, this pathway operates on several levels, regulating other types of plastic cells: (a) it regulates pancreatic islet cell function and insulin release; (b) insulin in turn activates the pathway in broadly distributed neural progenitors and possibly also hypothalamic tanycytes, cells with important roles in the control of the adrenal gland; (c) adrenal progenitors themselves operate this pathway. The STAT3-Ser/Hes3 Signaling Axis therefore deserves additional research in the context of endocrinology.


Subject(s)
DNA-Binding Proteins/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription Factors/metabolism , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Animals , Cell Differentiation , DNA-Binding Proteins/genetics , Humans , Hypothalamo-Hypophyseal System/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Pituitary-Adrenal System/pathology , Repressor Proteins , STAT3 Transcription Factor/genetics , Transcription Factors/genetics
2.
Horm Metab Res ; 45(2): 159-68, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22990992

ABSTRACT

Neural stem cells (NSCs) are pluripotent precursors with the ability to proliferate and differentiate into 3 neural cell lineages, neurons, astrocytes and oligodendrocytes. Elucidation of the mechanisms underlying these biologic processes is essential for understanding both physiologic and pathologic neural development and regeneration after injury. Nuclear hormone receptors (NRs) and their transcriptional coregulators also play crucial roles in neural development, functions and fate. To identify key NRs and their transcriptional regulators in NSC differentiation, we examined mRNA expression of 49 NRs and many of their coregulators during differentiation (0-5 days) of mouse embryonic NSCs induced by withdrawal of fibroblast growth factor-2 (FGF2). 37 out of 49 NRs were expressed in NSCs before induction of differentiation, while receptors known to play major roles in neural development, such as THRα, RXRs, RORs, TRs, and COUP-TFs, were highly expressed. CAR, which plays important roles in xenobiotic metabolism, was also highly expressed. FGF2 withdrawal induced mRNA expression of RORγ, RXRγ, and MR by over 20-fold. Most of the transcriptional coregulators examined were expressed basally and throughout differentiation without major changes, while FGF2 withdrawal strongly induced mRNA expression of several histone deacetylases (HDACs), including HDAC11. Dexamethasone and aldosterone, respectively a synthetic glucocorticoid and natural mineralocorticoid, increased NSC numbers and induced differentiation into neurons and astrocytes. These results indicate that the NRs and their coregulators are present and/or change their expression during NSC differentiation, suggesting that they may influence development of the central nervous system in the absence or presence of their ligands.


Subject(s)
Cell Differentiation , Cell Nucleus/metabolism , Gene Expression Regulation, Developmental , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Nuclear Proteins/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cell Differentiation/drug effects , Cell Nucleus/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Embryo, Mammalian/cytology , Gene Expression Profiling , Glucocorticoids/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Immunohistochemistry , Mice , Mineralocorticoids/pharmacology , Neural Stem Cells/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
3.
Cell Death Dis ; 2: e200, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21881602

ABSTRACT

Malignant gliomas contain a population of self-renewing tumorigenic stem-like cells; however, it remains unclear how these glioma stem cells (GSCs) self-renew or generate cellular diversity at the single-cell level. Asymmetric cell division is a proposed mechanism to maintain cancer stem cells, yet the modes of cell division that GSCs utilize remain undetermined. Here, we used single-cell analyses to evaluate the cell division behavior of GSCs. Lineage-tracing analysis revealed that the majority of GSCs were generated through expansive symmetric cell division and not through asymmetric cell division. The majority of differentiated progeny was generated through symmetric pro-commitment divisions under expansion conditions and in the absence of growth factors, occurred mainly through asymmetric cell divisions. Mitotic pair analysis detected asymmetric CD133 segregation and not any other GSC marker in a fraction of mitoses, some of which were associated with Numb asymmetry. Under growth factor withdrawal conditions, the proportion of asymmetric CD133 divisions increased, congruent with the increase in asymmetric cell divisions observed in the lineage-tracing studies. Using single-cell-based observation, we provide definitive evidence that GSCs are capable of different modes of cell division and that the generation of cellular diversity occurs mainly through symmetric cell division, not through asymmetric cell division.


Subject(s)
Antigens, CD/metabolism , Glioma/pathology , Glycoproteins/metabolism , Neoplastic Stem Cells/metabolism , Peptides/metabolism , AC133 Antigen , Antigens, CD/analysis , Cell Division , Cell Lineage , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Glioma/metabolism , Glycoproteins/analysis , Humans , Laminin/metabolism , Mitosis , Neoplastic Stem Cells/pathology , Peptides/analysis
4.
Oncogene ; 28(45): 3949-59, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19718046

ABSTRACT

Hypoxia contributes to the progression of a variety of cancers by activating adaptive transcriptional programs that promote cell survival, motility and tumor angiogenesis. Although the importance of hypoxia and subsequent hypoxia-inducible factor-1alpha (HIF-1alpha) activation in tumor angiogenesis is well known, their role in the regulation of glioma-derived stem cells is unclear. In this study, we show that hypoxia (1% oxygen) promotes the self-renewal capacity of CD133-positive human glioma-derived cancer stem cells (CSCs). Propagation of the glioma-derived CSCs in a hypoxic environment also led to the expansion of cells bearing CXCR4 (CD184), CD44(low) and A2B5 surface markers. The enhanced self-renewal activity of the CD133-positive CSCs in hypoxia was preceded by upregulation of HIF-1alpha. Knockdown of HIF-1alpha abrogated the hypoxia-mediated CD133-positive CSC expansion. Inhibition of the phosphatidylinositol 3-kinase(PI3K)-Akt or ERK1/2 pathway reduced the hypoxia-driven CD133 expansion, suggesting that these signaling cascades may modulate the hypoxic response. Finally, CSCs propagated at hypoxia robustly retained the undifferentiated phenotype, whereas CSCs cultured at normoxia did not. These results suggest that response to hypoxia by CSCs involves the activation of HIF-1alpha to enhance the self-renewal activity of CD133-positive cells and to inhibit the induction of CSC differentiation. This study illustrates the importance of the tumor microenvironment in determining cellular behavior.


Subject(s)
Antigens, CD/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , Glycoproteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplastic Stem Cells/metabolism , Peptides/metabolism , AC133 Antigen , Brain Neoplasms/pathology , Cell Growth Processes/physiology , Cell Hypoxia/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioma/pathology , Humans , Hyaluronan Receptors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Neoplastic Stem Cells/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR4/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Up-Regulation , Vascular Endothelial Growth Factor A/biosynthesis
5.
Article in English | MEDLINE | ID: mdl-19022746

ABSTRACT

The identification and characterization of multipotent neural precursors open the possibility of transplant therapies, but this approach is complicated by the widespread pathology of many degenerative diseases. Activation of endogenous precursors that support regenerative mechanisms is a possible alternative. We have previously shown that Notch ligands promote stem cell survival in vitro. Here, we show that there is an intimate interaction between insulin and Notch receptor signaling. Notch ligands also expand stem cell numbers in vivo with correlated benefits in brain ischemia. We now show that insulin promotes recovery of injured dopamine neurons in the adult brain. This response suggests that activating survival mechanisms in neural stem cells will promote recovery from progressive degenerative disease.


Subject(s)
Brain Diseases/metabolism , Neurons/metabolism , Stem Cells/metabolism , Adult , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Diseases/drug therapy , Brain Diseases/pathology , Cell Survival/drug effects , Dopamine/metabolism , Humans , Insulin/metabolism , Insulin/pharmacology , Intracellular Signaling Peptides and Proteins , Ligands , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/pathology , Oxidopamine/toxicity , Rabbits , Rats , Receptors, Notch/metabolism , Repressor Proteins , Signal Transduction , Stem Cells/drug effects , Stem Cells/pathology
6.
Neuroscience ; 149(1): 87-98, 2007 Oct 12.
Article in English | MEDLINE | ID: mdl-17826919

ABSTRACT

Matrix metalloproteinase-7 (MMP-7) belongs to a family of zinc dependent endopeptidases that are expressed in a variety of tissues including the brain. MMPs are known to be potent mediators of pericellular proteolysis and likely mediators of dynamic remodelling of neuronal connections. While an association between proteases and the neuronal synapse is emerging, a full understanding of this relationship is lacking. Here, we show that MMP-7 alters the structure and function of presynaptic terminals without affecting neuronal survival. Bath application of recombinant MMP-7 to cultured rat neurons induced long-lasting inhibition of vesicular recycling as measured by synaptotagmin 1 antibody uptake assays and FM4-64 optical imaging. MMP-7 application resulted in reduced abundance of vesicular and active zone proteins locally within synaptic terminals although their general levels remained unaltered. Finally, chronic application of the protease resulted in synaptic atrophy, including smaller terminals and fewer synaptic vesicles, as determined by electron microscopy. Together these results suggest that MMP-7 is a potent modulator of synaptic vesicle recycling and synaptic ultrastructure and that elevated levels of the enzyme, as may occur with brain inflammation, may adversely influence neurotransmission.


Subject(s)
Matrix Metalloproteinase 7/pharmacology , Neurons/drug effects , Synapses/drug effects , Synapses/pathology , Synaptic Vesicles/drug effects , Animals , Atrophy , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation/drug effects , Hippocampus/cytology , Humans , Matrix Metalloproteinase 1/pharmacology , Microscopy, Immunoelectron/methods , Protein Transport/drug effects , Pyridinium Compounds , Quaternary Ammonium Compounds , Rats , Rats, Sprague-Dawley , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , Synaptophysin/metabolism , Synaptotagmin I/metabolism , Vesicle-Associated Membrane Protein 2/metabolism
7.
Philos Trans R Soc Lond B Biol Sci ; 359(1445): 851-6, 2004 May 29.
Article in English | MEDLINE | ID: mdl-15293812

ABSTRACT

The fundamental basis of our work is that organs are generated by multipotent stem cells, whose properties we must understand to control tissue assembly or repair. Central nervous system (CNS) stem cells are now recognized as a well-defined population of precursors that differentiate into cells that are indisputably neurons and glial cells. Work from our group played an important role in defining stem cells of the CNS. Embryonic stem (ES) cells also differentiate to specific neuron and glial types through defined intermediates that are similar to the cellular precursors that normally occur in brain development. There is convincing evidence that the differentiated progeny of ES cells and CNS stem cells show expected functions of neurons and glia. Recent progress has been made on three fundamental developmental processes: (i) cell cycle control; (ii) the control of cell fate; and (iii) early steps in neural differentiation. In addition, our work on CNS stem cells has developed to a stage where there are clinical implications for Parkinson's and other degenerative disorders. These advances establish that stem cell biology contributes to our understanding of brain development and has great clinical promise.


Subject(s)
Central Nervous System/cytology , Neurodegenerative Diseases/therapy , Neuroglia/physiology , Neurons/physiology , Stem Cells/physiology , Bone Morphogenetic Protein Receptors, Type I , Cell Cycle/physiology , Cell Differentiation/physiology , Central Nervous System/physiology , Dopamine/metabolism , Dopamine/therapeutic use , Gene Expression Regulation/physiology , Humans , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Growth Factor/metabolism , Synapses/physiology , Vasoactive Intestinal Peptide/metabolism
8.
Curr Neurovasc Res ; 1(5): 421-7, 2004 Dec.
Article in English | MEDLINE | ID: mdl-16181090

ABSTRACT

The use of cell-based therapy may be a valid therapeutic approach to ischemic brain injury. Stem cells have been proposed as a new form of cell based therapy in a variety of disorders, including acute and degenerative brain diseases. Up to date most efforts have concentrated on transplantation of embryonic stem cells (ESC) or neural stem cells (NSCs) obtained from immortalized cell lines into the diseased brain. These procedures require harvesting the appropriate stem cell, expansion in vitro and transplantation. Endogenous NSCs have been identified in the central nervous system where they reside largely in the subventricular zone and in the subgranular zone of the hippocampus. Endogenous NSCs may be capable of self-renewal and differentiation into functional glia and neurons. Manipulation of endogenous NSCs may bypass the need to use ESC as a form of therapy thus avoiding the complex ethical and biological issues involved with ES cells or immortalized cell lines. This review summarizes the evidence recently gathered in support of a therapeutic role for endogenous NSCs in acute experimental stroke.


Subject(s)
Brain Ischemia/therapy , Neurons/physiology , Recovery of Function/physiology , Stem Cells/physiology , Animals , Cell Survival , Humans , Models, Neurological , Stem Cell Transplantation/methods
9.
Dev Biol ; 262(1): 119-36, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14512023

ABSTRACT

Embryonic stem (ES) cells have revolutionised our understanding of animal physiology. Analysis of chimaeric mice generated from these cells allows us to study the role of genes in development and function of the nervous system. The NMDA receptor, one of the two major ionotropic glutamate receptors, has been proposed to play fundamental roles in the survival, migration, differentiation, and activity-dependent maturation of neural cells. The NMDA receptor subunit 1 (NR1) gene is indispensable for receptor function, and knock-out mice die at birth, inhibiting the study of glutamate signalling in postnatal neurons. Homozygous NR1-/- ES cells were derived from matings of heterozygous mice under feeder-free conditions. Chimaeras were made by incorporating these ES cells into wild-type blastocysts and by the classical aggregation of morulae between wild-type and NR1-/- embryos. The resulting chimaeras survive and develop normally. NR1-/- neurons, identified by their lacZ label, were analysed and quantified in developing and adult brains with varying knock-out contributions in every single brain region. Specifically, postnatal ontogenesis of cerebellum and hippocampus was normal. Accordingly, in chimaeric mice, NMDA receptor-initiated signals are not required for the migration, differentiation, and survival of most types of neurons in the central nervous system, in a cell-autonomous way.


Subject(s)
Brain/growth & development , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Cell Differentiation , Cell Movement , Cerebellum/growth & development , Chimera , Embryo, Mammalian/cytology , Female , Hippocampus/growth & development , Mice , Stem Cells/physiology
10.
J Neurosci Res ; 65(4): 284-8, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11494363

ABSTRACT

The use of in vitro expanded human CNS precursors has the potential to overcome some of the ethical, logistic and technical problems of fetal tissue transplantation in Parkinson disease. Cultured rat mesencephalic precursors proliferate in response to bFGF and upon mitogen withdrawal, differentiate into functional dopamine neurons that alleviate motor symptoms in Parkinsonian rats (Studer et al. [1998] Nat. Neurosci. 1:290-295). The successful clinical application of CNS precursor technology in Parkinson disease will depend on the efficient in vitro generation of human dopaminergic neurons. We demonstrate that human dopamine neurons can be generated from both midbrain and cortical precursors. Transplantation of midbrain precursor-derived dopamine neurons into Parkinsonian rats resulted in grafts rich in tyrosine hydroxylase positive neurons 6 weeks after transplantation. No surviving tyrosine hydroxylase positive neurons could be detected when dopamine neurons derived from cortical precursors were grafted. Our data demonstrate in vitro derivation of human dopamine neurons from expanded CNS precursors and encourage further studies that systematically address in vivo function and clinical potential.


Subject(s)
Brain Tissue Transplantation , Dopamine/physiology , Fetal Tissue Transplantation , Neurons/transplantation , Parkinsonian Disorders/surgery , Stem Cell Transplantation , Animals , Cell Culture Techniques/methods , Cell Differentiation , Cell Survival , Cells, Cultured , Disease Models, Animal , Female , Fetus/cytology , Humans , Neurons/cytology , Oxidopamine , Parkinsonian Disorders/chemically induced , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Sympathomimetics
11.
Genes Dev ; 15(16): 2094-110, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11511541

ABSTRACT

Bone morphogenetic proteins (BMPs) have diverse and sometimes paradoxical effects during embryonic development. To determine the mechanisms underlying BMP actions, we analyzed the expression and function of two BMP receptors, BMPR-IA and BMPR-IB, in neural precursor cells in vitro and in vivo. Neural precursor cells always express Bmpr-1a, but Bmpr-1b is not expressed until embryonic day 9 and is restricted to the dorsal neural tube surrounding the source of BMP ligands. BMPR-IA activation induces (and Sonic hedgehog prevents) expression of Bmpr-1b along with dorsal identity genes in precursor cells and promotes their proliferation. When BMPR-IB is activated, it limits precursor cell numbers by causing mitotic arrest. This results in apoptosis in early gestation embryos and terminal differentiation in mid-gestation embryos. Thus, BMP actions are first inducing (through BMPR-IA) and then terminating (through BMPR-IB), based on the accumulation of BMPR-IB relative to BMPR-IA. We describe a feed-forward mechanism to explain how the sequential actions of these receptors control the production and fate of dorsal precursor cells from neural stem cells.


Subject(s)
Neurons/physiology , Protein Serine-Threonine Kinases/metabolism , Receptors, Cell Surface/physiology , Receptors, Growth Factor/metabolism , Trans-Activators , Animals , Apoptosis , Bone Morphogenetic Protein Receptors , Bone Morphogenetic Protein Receptors, Type I , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/physiology , Cell Count , Cell Differentiation/physiology , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Epithelial Cells/physiology , Female , Hedgehog Proteins , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proteins/physiology , Receptor Cross-Talk , Receptors, Cell Surface/metabolism , Receptors, Growth Factor/antagonists & inhibitors , Signal Transduction
12.
J Comp Neurol ; 435(4): 406-17, 2001 Jul 09.
Article in English | MEDLINE | ID: mdl-11406822

ABSTRACT

Knowing the rate of addition of new granule cells to the adult dentate gyrus is critical to understanding the function of adult neurogenesis. Despite the large number of studies of neurogenesis in the adult dentate gyrus, basic questions about the magnitude of this phenomenon have never been addressed. The S-phase marker bromodeoxyuridine (BrdU) has been extensively used in recent studies of adult neurogenesis, but it has been carefully tested only in the embryonic brain. Here, we show that a high dose of BrdU (300 mg/kg) is a specific, quantitative, and nontoxic marker of dividing cells in the adult rat dentate gyrus, whereas lower doses label only a fraction of the S-phase cells. By using this high dose of BrdU along with a second S-phase marker, [(3)H]thymidine, we found that young adult rats have 9,400 dividing cells proliferating with a cell cycle time of 25 hours, which would generate 9,000 new cells each day, or more than 250,000 per month. Within 5-12 days of BrdU injection, a substantial pool of immature granule neurons, 50% of all BrdU-labeled cells in the dentate gyrus, could be identified with neuron-specific antibodies TuJ1 and TUC-4. This number of new granule neurons generated each month is 6% of the total size of the granule cell population and 30-60% of the size of the afferent and efferent populations (West et al. [1991] Anat Rec 231:482-497; Mulders et al. [1997] J Comp Neurol 385:83-94). The large number of the adult-generated granule cells supports the idea that these new neurons play an important role in hippocampal function.


Subject(s)
Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Animals , Antimetabolites , Bromodeoxyuridine , Cell Cycle/drug effects , Cell Survival/drug effects , Male , Rats , Tissue Fixation
13.
Eur J Neurosci ; 13(7): 1273-82, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11298787

ABSTRACT

We have recently demonstrated that embryonic E16 hippocampal neurons grown in cultures are unable to form fast synaptic connections unless treated with BDNF or NT-3. This experimental system offers an opportunity to define the roles of neurotrophins in processes leading to formation of functional synaptic connections. We have used ultrastructural and electrophysiological methods to explore the cellular locations underlying neurotrophin action on synaptic maturation. The rate of spontaneous miniature excitatory postsynaptic currents (mEPSCs) evoked by hyperosmotic stimulation was 7-16-fold higher in neurotrophin-treated cells than in controls. In addition, the potent neurotransmitter-releasing drug alpha-latrotoxin was virtually ineffective in the control cells while it stimulated synaptic events in neurotrophin-treated cells. Likewise, the membrane-bound dye FM1-43 was taken up by terminals in neurotrophin-treated cultures five-fold more than in controls. Both the total number and the number of docked synaptic vesicles were increased by neurotrophin treatment. Activation of synaptic responses by neurotrophins occurred even when postsynaptic glutamate receptors and action potential discharges were pharmacologically blocked. These results are consistent with a presynaptic locus of action of neurotrophins to increase synaptic vesicle density which is critical for rapid synaptic transmission. They also suggest that neurotrophins can activate synapses in the absence of pre- and postsynaptic neuronal activity.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Neurons/physiology , Neurotrophin 3/pharmacology , Presynaptic Terminals/drug effects , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cells, Cultured , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , Microscopy, Electron , Neurons/cytology , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Rats , Spider Venoms/pharmacology , Synaptic Vesicles/physiology , Synaptic Vesicles/ultrastructure , Tetrodotoxin/pharmacology
14.
Dev Biol ; 231(1): 103-12, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11180955

ABSTRACT

The NMDA receptor, one of the two major ionotropic glutamate receptors, has been proposed to play fundamental roles in the survival, migration, differentiation, and activity-dependent maturation of neural cells. The NR1 gene encodes the major subunit that is responsible for channel function, and NR1 -/- mice die at birth, inhibiting the study of glutamate signaling in postnatal neurons. The properties of cells lacking the NR1 subunit of NMDA receptors were studied by transplanting dissociated telencephalic, diencephalic, and mesencephalic cells of E14 mouse embryos with a targeted deletion of the NR1 gene into the ventricles of embryonic rats using intrauterine transplantation (Brüstle et al., 1995, Neuron 15, 1275-1285). The transplanted cells took part in the normal development of the host brain where they survived after migration into a large number of brain structures. Morphological and immunohistochemical analysis suggests that NR1 -/- cells can differentiate normally in these sites. The results provide evidence that NMDA-receptor-initiated signals are not required for the postnatal differentiation and survival of many types of neurons in the central nervous system, in a noncell autonomous fashion after transplantation into a wild-type environment.


Subject(s)
Brain/embryology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Cell Differentiation , Cell Movement , Cell Survival , Mice , Mice, Knockout , Neurons/transplantation , Rats , Rats, Sprague-Dawley
15.
J Neurochem ; 76(1): 307-11, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11146004

ABSTRACT

CNS precursors derived from E12 rat mesencephalon proliferate in the presence of basic fibroblast growth factor and differentiate in vitro into functional dopaminergic neurons, which upon transplantation alleviate behavioral symptoms in a rat model of Parkinson's disease. Here we show that the efficiency of dopaminergic differentiation decreases in the mesencephalic precursors that were proliferated or passaged for extended periods in vitro. Ascorbic acid treatment restored dopaminergic differentiation in these precursors and led to a greater than 10-fold increase in dopamine neuron yield compared with untreated cultures. The effect of ascorbic acid was stereospecific and could not be mimicked by any other antioxidants. The expression of sodium-dependent vitamin C transporter, a recently identified stereospecific ascorbic acid transporter, was maintained in mesencephalic precursors for extended in vitro periods. Pre-treatment of in vitro expanded mesencephalic precursors with ascorbic acid might facilitate the large-scale generation of dopaminergic neurons for clinical transplantation.


Subject(s)
Ascorbic Acid/pharmacology , Fibroblast Growth Factor 2/pharmacology , Mesencephalon/drug effects , Neurons/drug effects , Stem Cells/drug effects , Animals , Biological Transport , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Cells, Cultured , Cyclic AMP/agonists , Dopamine/metabolism , Immunohistochemistry , Mesencephalon/cytology , Mesencephalon/embryology , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/metabolism , Tyrosine 3-Monooxygenase/metabolism
16.
J Neurosci ; 20(21): 8012-20, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11050122

ABSTRACT

Astrocytes are present in large numbers in the nervous system, are associated with synapses, and propagate ionic signals. Astrocytes influence neuronal physiology by responding to and releasing neurotransmitters, but the mechanisms that establish the close interaction between these cells are not defined. Here we use hippocampal neurons in culture to demonstrate that vasoactive intestinal polypeptide (VIP) promotes neuronal differentiation through activity-dependent neurotrophic factor (ADNF), a protein secreted by VIP-stimulated astroglia. ADNF is produced by glial cells and acts directly on neurons to promote glutamate responses and morphological development. ADNF causes secretion of neurotrophin 3 (NT-3), and both proteins regulate NMDA receptor subunit 2A (NR2A) and NR2B. These data suggest that the VIP-ADNF-NT-3 neuronal-glial pathway regulates glutamate responses from an early stage in the synaptic development of excitatory neurons and may also contribute to the known effects of VIP on learning and behavior in the adult nervous system.


Subject(s)
Cell Differentiation/physiology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Oligopeptides/metabolism , Vasoactive Intestinal Peptide/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Microtubule-Associated Proteins/metabolism , Neuroglia/cytology , Neurons/drug effects , Neurotrophin 3/metabolism , Oligopeptides/pharmacology , Patch-Clamp Techniques , Peptide Fragments/pharmacology , Physical Stimulation , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/drug effects , Stimulation, Chemical , Vasoactive Intestinal Peptide/pharmacology
17.
Nat Biotechnol ; 18(6): 675-9, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10835609

ABSTRACT

Embryonic stem (ES) cells are clonal cell lines derived from the inner cell mass of the developing blastocyst that can proliferate extensively in vitro and are capable of adopting all the cell fates in a developing embryo. Clinical interest in the use of ES cells has been stimulated by studies showing that isolated human cells with ES properties from the inner cell mass or developing germ cells can provide a source of somatic precursors. Previous studies have defined in vitro conditions for promoting the development of specific somatic fates, specifically, hematopoietic, mesodermal, and neurectodermal. In this study, we present a method for obtaining dopaminergic (DA) and serotonergic neurons in high yield from mouse ES cells in vitro. Furthermore, we demonstrate that the ES cells can be obtained in unlimited numbers and that these neuron types are generated efficiently. We generated CNS progenitor populations from ES cells, expanded these cells and promoted their differentiation into dopaminergic and serotonergic neurons in the presence of mitogen and specific signaling molecules. The differentiation and maturation of neuronal cells was completed after mitogen withdrawal from the growth medium. This experimental system provides a powerful tool for analyzing the molecular mechanisms controlling the functions of these neurons in vitro and in vivo, and potentially for understanding and treating neurodegenerative and psychiatric diseases.


Subject(s)
Embryo, Mammalian , Mesencephalon/cytology , Neurons/cytology , Rhombencephalon/cytology , Stem Cells/cytology , Trans-Activators , Animals , Ascorbic Acid/pharmacology , Cell Culture Techniques/methods , Cell Differentiation , Cell Division , Cell Line , Chromatography, High Pressure Liquid , Cyclic AMP/pharmacology , Dopamine/metabolism , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/pharmacology , Glutamic Acid/pharmacology , Hedgehog Proteins , Immunohistochemistry , Mice , Patch-Clamp Techniques , Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/metabolism , Tetrodotoxin/pharmacology , gamma-Aminobutyric Acid/pharmacology
18.
Eur J Neurosci ; 12(5): 1696-704, 2000 May.
Article in English | MEDLINE | ID: mdl-10792447

ABSTRACT

An understanding of developmental mechanisms and new cell therapies can be achieved by transplantation into the nervous system. Multipotential stem cells have been isolated from the foetal and adult central nervous system (CNS). Immortalized and primary precursor cells integrate into the developing brain generating both neurons and glia as defined by immunological and morphological criteria. Here we show for the first time that in vitro-expanded CNS precursors, upon transplantation into the brains of rats, form electrically active and functionally connected neurons. These neurons exhibit spontaneous and evoked postsynaptic events and respond to focal glutamate application. Donor cells were grafted into the foetal hippocampus, and the amplitude and frequency of spontaneous synaptic events were monitored in the grafted cells in area CA1 for the first month of postnatal life. The formation of synapses onto grafted neurons indicates that grafted CNS stem cells can be used to study synaptic development in vivo and has important implications for clinical cell replacement therapies.


Subject(s)
Brain Tissue Transplantation/physiology , Cerebral Cortex/cytology , Fetal Tissue Transplantation/physiology , Neurons/physiology , Neurons/transplantation , Synapses/physiology , Animals , Cells, Cultured , Cerebral Cortex/embryology , Excitatory Postsynaptic Potentials , Genes, Reporter , Green Fluorescent Proteins , In Vitro Techniques , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Membrane Potentials , Rats , Rats, Sprague-Dawley , Retroviridae , Stem Cells , Transfection
19.
J Neurosci ; 20(10): 3725-35, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10804214

ABSTRACT

Cell fate is determined by intrinsic programs and external cues, such as soluble signals and cell-cell contact. Previous studies have demonstrated the roles of soluble factors in the proliferation and differentiation of cortical stem cells and cell-cell contact in maintaining stem cells in a proliferative state. In the present study, we focused on the effect of cell-cell interaction on cell-fate determination. We found that density could exert a strong influence on the cell-type composition when cortical stem cells differentiate. Multipotent stem cells, which normally gave rise to neurons, astrocytes, and oligodendrocytes under high-density culture condition, differentiated almost exclusively into smooth muscle at low density. Clonal analysis indicated that smooth muscle and astrocytes were derived from a common precursor and that the density effect on cell types used an instructive mechanism on the choice of fate rather than an effect of selective survival and/or proliferation. This instructive mechanism depended on the local and not the average density of the cells. This local signal could be mimicked by membrane extract. These findings demonstrate the importance of membrane-bound signals in specifying lineage and provide the first evidence for a short-range regulatory mechanism in cortical stem cell differentiation.


Subject(s)
Cell Communication/physiology , Cerebral Cortex/cytology , Neurons/cytology , Stem Cells/cytology , Animals , Astrocytes/cytology , Cell Count , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Membrane/physiology , Cells, Cultured , Female , Fetus/cytology , Muscle, Smooth/cytology , Oligodendroglia/cytology , Pregnancy , Rats , Rats, Sprague-Dawley
20.
Eur J Neurosci ; 12(2): 677-88, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10712648

ABSTRACT

Stem cell technology promises new and rapid advances in cell therapy and drug discovery. Clearly, the value of this approach will be limited by the differentiated functions displayed by the progeny of stem cells. The foetal and adult central nervous system (CNS) harbour stem cells that can be expanded in vitro and differentiate into immature neurons and glia. Surprisingly, we do not know if neurons derived from stem cells form synapses, a definitive feature of neuronal function. Neuronal differentiation is a complex process and in this paper we establish conditions that permit extensive maturation of neurons in the presence of neurotrophins. These conditions permit the differentiation of rat hippocampal stem cells into both excitatory (glutamatergic) and inhibitory (GABAergic) neurons. The proportion of excitatory and inhibitory synapses was strongly influenced by specific neurotrophins, and these responses reflect the region of origin of the stem cells in the brain. These data show that stem cells can be used to study mechanisms of excitation and inhibition in the nervous system.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Hippocampus/cytology , Nerve Growth Factor/pharmacology , Nerve Growth Factors/pharmacology , Neurons/cytology , Neurotrophin 3/pharmacology , Stem Cells/cytology , Animals , Astrocytes/cytology , Astrocytes/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/analysis , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Lineage , Cells, Cultured/drug effects , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , Glutamic Acid/pharmacology , Immunoenzyme Techniques , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Neurons/classification , Neurons/drug effects , Phosphoprotein Phosphatases/metabolism , Rats , Receptor, trkB/metabolism , Receptor, trkC/metabolism , Receptors, AMPA/metabolism , Stem Cells/drug effects , Synapses/drug effects , Synapses/physiology , Synapsins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL