Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Neuroendocrinology ; 113(12): 1248-1261, 2023.
Article in English | MEDLINE | ID: mdl-36257292

ABSTRACT

INTRODUCTION: Hippocampal newborn neurons integrate into functional circuits where they play an important role in learning and memory. We previously showed that perinatal exposure to Aroclor 1254, a commercial mixture of polychlorinated biphenyls (PCBs) associated with alterations of cognitive function in children, disrupted the normal maturation of excitatory synapses in the dentate gyrus. We hypothesized that hippocampal immature neurons underlie some of the cognitive effects of PCBs. METHODS: We used newly generated neurons to examine the effects of PCBs in mice following maternal exposure. Newborn dentate granule cells were tagged with enhanced green fluorescent protein using a transgenic mouse line. The transcriptome of the newly generated granule cells was assessed using RNA sequencing. RESULTS: Gestational and lactational exposure to 6 mg/kg/day of Aroclor 1254 disrupted the mRNA expression of 1,308 genes in newborn granule cells. Genes involved in mitochondrial functions were highly enriched with 154 genes significantly increased in exposed compared to control mice. The upregulation of genes involved in oxidative phosphorylation was accompanied by signs of endoplasmic reticulum stress and an increase in lipid peroxidation, a marker of oxidative stress, in the subgranular zone of the dentate gyrus but not in mature granule cells in the granular zone. Aroclor 1254 exposure also disrupted the expression of synaptic genes. Using laser-captured subgranular and granular zones, this effect was restricted to the subgranular zone, where newborn neurons are located. CONCLUSION: Our data suggest that gene expression in newborn granule cells is disrupted by Aroclor 1254 and provide clues to the effects of endocrine-disrupting chemicals on the brain.


Subject(s)
Polychlorinated Biphenyls , Humans , Female , Pregnancy , Child , Mice , Animals , Polychlorinated Biphenyls/pharmacology , Hippocampus , Neurons/physiology , Mice, Transgenic , Brain , Oxidative Stress , Gene Expression , Dentate Gyrus , Neurogenesis
2.
Front Neural Circuits ; 15: 787436, 2021.
Article in English | MEDLINE | ID: mdl-34975417

ABSTRACT

Dendritic spines, the distinctive postsynaptic feature of central nervous system (CNS) excitatory synapses, have been studied extensively as electrical and chemical compartments, as well as scaffolds for receptor cycling and positioning of signaling molecules. The dynamics of the shape, number, and molecular composition of spines, and how they are regulated by neural activity, are critically important in synaptic efficacy, synaptic plasticity, and ultimately learning and memory. Dendritic spines originate as outward protrusions of the cell membrane, but this aspect of spine formation and stabilization has not been a major focus of investigation compared to studies of membrane protrusions in non-neuronal cells. We review here one family of proteins involved in membrane curvature at synapses, the BAR (Bin-Amphiphysin-Rvs) domain proteins. The subfamily of inverse BAR (I-BAR) proteins sense and introduce outward membrane curvature, and serve as bridges between the cell membrane and the cytoskeleton. We focus on three I-BAR domain proteins that are expressed in the central nervous system: Mtss2, MIM, and IRSp53 that promote negative, concave curvature based on their ability to self-associate. Recent studies suggest that each has distinct functions in synapse formation and synaptic plasticity. The action of I-BARs is also shaped by crosstalk with other signaling components, forming signaling platforms that can function in a circuit-dependent manner. We discuss another potentially important feature-the ability of some BAR domain proteins to impact the function of other family members by heterooligomerization. Understanding the spatiotemporal resolution of synaptic I-BAR protein expression and their interactions should provide insights into the interplay between activity-dependent neural plasticity and network rewiring in the CNS.


Subject(s)
Neuronal Plasticity , Synapses , Cell Membrane , Dendritic Spines , Learning , Signal Transduction
3.
Elife ; 82019 06 24.
Article in English | MEDLINE | ID: mdl-31232686

ABSTRACT

Exercise is a potent enhancer of learning and memory, yet we know little of the underlying mechanisms that likely include alterations in synaptic efficacy in the hippocampus. To address this issue, we exposed mice to a single episode of voluntary exercise, and permanently marked activated mature hippocampal dentate granule cells using conditional Fos-TRAP mice. Exercise-activated neurons (Fos-TRAPed) showed an input-selective increase in dendritic spines and excitatory postsynaptic currents at 3 days post-exercise, indicative of exercise-induced structural plasticity. Laser-capture microdissection and RNASeq of activated neurons revealed that the most highly induced transcript was Mtss1L, a little-studied I-BAR domain-containing gene, which we hypothesized could be involved in membrane curvature and dendritic spine formation. shRNA-mediated Mtss1L knockdown in vivo prevented the exercise-induced increases in spines and excitatory postsynaptic currents. Our results link short-term effects of exercise to activity-dependent expression of Mtss1L, which we propose as a novel effector of activity-dependent rearrangement of synapses.


Subject(s)
Hippocampus/physiology , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Neuronal Plasticity , Neurons/physiology , Physical Conditioning, Animal , Action Potentials , Animals , Base Sequence , Gene Expression Profiling , Mice
4.
J Neurosci ; 38(26): 5843-5853, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29793975

ABSTRACT

Mature dentate granule cells in the hippocampus receive input from the entorhinal cortex via the perforant path in precisely arranged lamina, with medial entorhinal axons innervating the middle molecular layer and lateral entorhinal cortex axons innervating the outer molecular layer. Although vastly outnumbered by mature granule cells, adult-generated newborn granule cells play a unique role in hippocampal function, which has largely been attributed to their enhanced excitability and plasticity (Schmidt-Hieber et al., 2004; Ge et al., 2007). Inputs from the medial and lateral entorhinal cortex carry different informational content. Thus, the distribution of inputs onto newly integrated granule cells will affect their function in the circuit. Using retroviral labeling in combination with selective optogenetic activation of medial or lateral entorhinal inputs, we examined the functional innervation and synaptic maturation of newly generated dentate granule cells in the mouse hippocampus. Our results indicate that lateral entorhinal inputs provide the majority of functional innervation of newly integrated granule cells at 21 d postmitosis. Despite preferential functional targeting, the dendritic spine density of immature granule cells was similar in the outer and middle molecular layers, which we speculate could reflect an unequal distribution of shaft synapses. However, chronic blockade of neurotransmitter release of medial entorhinal axons with tetanus toxin disrupted normal synapse development of both medial and lateral entorhinal inputs. Our results support a role for preferential lateral perforant path input onto newly generated neurons in mediating pattern separation, but also indicate that medial perforant path input is necessary for normal synaptic development.SIGNIFICANCE STATEMENT The formation of episodic memories involves the integration of contextual and spatial information. Newly integrated neurons in the dentate gyrus of the hippocampus play a critical role in this process, despite constituting only a minor fraction of the total number of granule cells. Here we demonstrate that these neurons preferentially receive information thought to convey the context of an experience. Each newly integrated granule cell plays this unique role for ∼1 month before reaching maturity.


Subject(s)
Dentate Gyrus/physiology , Entorhinal Cortex/physiology , Neurons/physiology , Perforant Pathway/physiology , Animals , Dentate Gyrus/cytology , Entorhinal Cortex/cytology , Female , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Perforant Pathway/cytology , Synapses/physiology
5.
eNeuro ; 3(1)2016.
Article in English | MEDLINE | ID: mdl-27011954

ABSTRACT

Despite representing only a small fraction of hippocampal granule cells, adult-generated newborn granule cells have been implicated in learning and memory (Aimone et al., 2011). Newborn granule cells undergo functional maturation and circuit integration over a period of weeks. However, it is difficult to assess the accompanying gene expression profiles in vivo with high spatial and temporal resolution using traditional methods. Here we used a novel method ["thiouracil (TU) tagging"] to map the profiles of nascent mRNAs in mouse immature newborn granule cells compared with mature granule cells. We targeted a nonmammalian uracil salvage enzyme, uracil phosphoribosyltransferase, to newborn neurons and mature granule cells using retroviral and lentiviral constructs, respectively. Subsequent injection of 4-TU tagged nascent RNAs for analysis by RNA sequencing. Several hundred genes were significantly enhanced in the retroviral dataset compared with the lentiviral dataset. We compared a selection of the enriched genes with steady-state levels of mRNAs using quantitative PCR. Ontology analysis revealed distinct patterns of nascent mRNA expression, with newly generated immature neurons showing enhanced expression for genes involved in synaptic function, and neural differentiation and development, as well as genes not previously associated with granule cell maturation. Surprisingly, the nascent mRNAs enriched in mature cells were related to energy homeostasis and metabolism, presumably indicative of the increased energy demands of synaptic transmission and their complex dendritic architecture. The high spatial and temporal resolution of our modified TU-tagging method provides a foundation for comparison with steady-state RNA analyses by traditional transcriptomic approaches in defining the functional roles of newborn neurons.


Subject(s)
Dentate Gyrus/metabolism , Gene Expression Profiling/methods , Neurogenesis , Neurons/metabolism , Thiouracil/metabolism , Animals , Base Sequence , Female , Genetic Vectors/administration & dosage , Lentivirus/genetics , Lentivirus/physiology , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Thiouracil/administration & dosage
6.
Elife ; 4: e08722, 2015 Oct 17.
Article in English | MEDLINE | ID: mdl-26476335

ABSTRACT

The majority of adult hippocampal newborn cells die during early differentiation from intermediate progenitors (IPCs) to immature neurons. Neural stem cells in vivo are located in a relative hypoxic environment, and hypoxia enhances their survival, proliferation and stemness in vitro. Thus, we hypothesized that migration of IPCs away from hypoxic zones within the SGZ might result in oxidative damage, thus triggering cell death. Hypoxic niches were observed along the SGZ, composed of adult NSCs and early IPCs, and oxidative byproducts were present in adjacent late IPCs and neuroblasts. Stabilizing hypoxia inducible factor-1α with dimethyloxallyl glycine increased early survival, but not proliferation or differentiation, in neurospheres in vitro and in newly born SGZ cells in vivo. Rescue experiments in Bax(fl/fl) mutants supported these results. We propose that localized hypoxia within the SGZ contributes to the neurogenic microenvironment and determines the early, activity-independent survival of adult hippocampal newborn cells.


Subject(s)
Hippocampus/physiology , Hypoxia , Neural Stem Cells/physiology , Animals , Animals, Newborn , Cell Differentiation , Cell Proliferation , Cell Survival , Hippocampus/growth & development , Mice, Inbred C57BL , Mice, Transgenic
7.
Dev Dyn ; 242(9): 1056-65, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23765990

ABSTRACT

BACKGROUND: Retinoic acid (RA) signaling controls patterning and neuronal differentiation within the hindbrain, but forebrain RA function remains controversial. RA is produced from metabolism of retinol to retinaldehyde by retinol dehydrogenase (RDH), followed by metabolism of retinaldehyde to RA by retinaldehyde dehydrogenase (RALDH). Previous studies on Raldh2-/- and Raldh3-/- mice demonstrated an RA requirement for γ-aminobutyric acid (GABA)ergic and dopaminergic differentiation in forebrain basal ganglia, but no RA requirement was observed during early forebrain patterning or subsequent forebrain cortical expansion. However, other studies suggested that RA controls forebrain patterning, and analysis of ethylnitrosourea-induced Rdh10 mutants suggested that RA synthesized in the meninges stimulates forebrain cortical expansion. RESULTS: We generated Rdh10-/- mouse embryos that lack RA activity early in the head and later in the meninges. We observed defects in hindbrain patterning and eye RA signaling, but early forebrain patterning was unaffected. Retinaldehyde treatment of Rdh10-/- embryos from E7-E9 rescues a cranial skeletal defect, resulting in E14.5 embryos lacking meningeal RA activity but maintaining normal forebrain shape and cortical expansion. CONCLUSIONS: Rdh10-/- embryos demonstrate that RA controls hindbrain but not early forebrain patterning, while studies on retinaldehyde-rescued Rdh10-/- embryos show that meningeal RA synthesis is unnecessary to stimulate forebrain cortical expansion.


Subject(s)
Alcohol Oxidoreductases/metabolism , Embryo, Mammalian/embryology , Neurogenesis/physiology , Prosencephalon/embryology , Retinaldehyde/metabolism , Tretinoin/metabolism , Alcohol Oxidoreductases/genetics , Animals , Embryo, Mammalian/cytology , Eye/cytology , Eye/embryology , Mice , Mice, Knockout , Prosencephalon/cytology , Retinaldehyde/genetics , Signal Transduction/physiology
8.
PLoS Biol ; 9(4): e1000609, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21532733

ABSTRACT

Although retinoic acid (RA) has been implicated as an extrinsic signal regulating forebrain neurogenesis, the processes regulated by RA signaling remain unclear. Here, analysis of retinaldehyde dehydrogenase mutant mouse embryos lacking RA synthesis demonstrates that RA generated by Raldh3 in the subventricular zone of the basal ganglia is required for GABAergic differentiation, whereas RA generated by Raldh2 in the meninges is unnecessary for development of the adjacent cortex. Neurospheres generated from the lateral ganglionic eminence (LGE), where Raldh3 is highly expressed, produce endogenous RA, which is required for differentiation to GABAergic neurons. In Raldh3⁻/⁻ embryos, LGE progenitors fail to differentiate into either GABAergic striatal projection neurons or GABAergic interneurons migrating to the olfactory bulb and cortex. We describe conditions for RA treatment of human embryonic stem cells that result in efficient differentiation to a heterogeneous population of GABAergic interneurons without the appearance of GABAergic striatal projection neurons, thus providing an in vitro method for generation of GABAergic interneurons for further study. Our observation that endogenous RA is required for generation of LGE-derived GABAergic neurons in the basal ganglia establishes a key role for RA signaling in development of the forebrain.


Subject(s)
Basal Ganglia/metabolism , GABA Agents/metabolism , Gene Expression Regulation, Developmental , Interneurons/metabolism , Tretinoin/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Basal Ganglia/cytology , Basal Ganglia/embryology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Embryo, Mammalian , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Humans , Interneurons/cytology , Interneurons/drug effects , Mice , Mice, Knockout , Prosencephalon/drug effects , Prosencephalon/metabolism , Retinal Dehydrogenase/deficiency , Retinal Dehydrogenase/genetics , Signal Transduction , Tretinoin/pharmacology
9.
Dev Dyn ; 240(5): 1142-50, 2011 May.
Article in English | MEDLINE | ID: mdl-21360789

ABSTRACT

Retinoic acid (RA) is purported to be required for expression of genes controlling proximodistal (Meis2) or anteroposterior (Shh) limb patterning. Embryos lacking RDH10, the primary enzyme synthesizing retinaldehyde during mouse development, survive until E14.5 with stunted forelimbs but apparently normal hindlimbs. Using embryos carrying the RARE-lacZ RA-reporter transgene, we show that endogenous RA activity in Rdh10(trex/trex) mutants is detected in neuroectoderm but not limbs during initiation and patterning. Treatment of Rdh10 mutants with 25 nM RA restores RARE-lacZ activity to limb mesoderm, validating RARE-lacZ and verifying that RA is absent in mutant limbs. In Rdh10 mutants, hindlimbs exhibit normal Meis2/Shh expression and skeletal patterning, and although forelimbs are growth-retarded their Meis2 expression remains normal. Later in development, Rdh10 mutants lack interdigital RA activity and accordingly fail to exhibit normal loss of interdigital mesenchyme. These findings demonstrate that RA is unnecessary for limb patterning but required later for interdigital tissue loss.


Subject(s)
Alcohol Oxidoreductases/metabolism , Body Patterning/physiology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Limb Buds/metabolism , Limb Deformities, Congenital/metabolism , Tretinoin/pharmacology , Alcohol Oxidoreductases/genetics , Animals , Body Patterning/genetics , Female , In Situ Hybridization , In Vitro Techniques , Limb Buds/embryology , Limb Deformities, Congenital/genetics , Mice , Pregnancy
10.
Nat Commun ; 2: 151, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21224842

ABSTRACT

Sex-specific initiation of meiosis in the fetal ovary has been suggested to require retinoic acid (RA) for induction of Stra8, with expression of the RA-degrading enzyme Cyp26b1 in fetal testis delaying meiosis until postnatal development. In this study, we investigate Raldh2(-/-) mice lacking RA synthesis and signalling in mesonephros and adjacent gonad and reveal that Stra8 expression in the fetal ovary does not require RA signalling. In contrast to previous observations, we find that Stra8 is expressed in the absence of physiologically detectable levels of RA. Ketoconazole inhibition of Cyp26b1 in Raldh2(-/-) testis allows RA-independent induction of Stra8, but only when the mesonephros remains attached, pointing to a non-RA signal from the mesonephros that induces Stra8 in the adjacent gonad. These findings demonstrate that Cyp26b1 prevents the onset of meiosis by metabolizing a substrate other than RA that controls Stra8 expression, thus changing the paradigm for how studies on Cyp26 function are conducted.

11.
Development ; 138(1): 139-48, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21138976

ABSTRACT

Epicardial signaling and Rxra are required for expansion of the ventricular myocardial compact zone. Here, we examine Raldh2(-/-) and Rxra(-/-) mouse embryos to investigate the role of retinoic acid (RA) signaling in this developmental process. The heart phenotypes of Raldh2 and Rxra mutants are very similar and are characterized by a prominent defect in ventricular compact zone growth. Although RA activity is completely lost in Raldh2(-/-) epicardium and the adjacent myocardium, RA activity is not lost in Rxra(-/-) hearts, suggesting that RA signaling in the epicardium/myocardium is not required for myocardial compact zone formation. We explored the possibility that RA-mediated target gene transcription in non-cardiac tissues is required for this process. We found that hepatic expression of erythropoietin (EPO), a secreted factor implicated in myocardial expansion, is dependent on both Raldh2 and Rxra. Chromatin immunoprecipitation studies support Epo as a direct target of RA signaling in embryonic liver. Treatment of an epicardial cell line with EPO, but not RA, upregulates Igf2. Furthermore, both Raldh2(-/-) and Rxra(-/-) hearts exhibit downregulation of Igf2 mRNA in the epicardium. EPO treatment of cultured Raldh2(-/-) hearts restores epicardial Igf2 expression and rescues ventricular cardiomyocyte proliferation. We propose a new model for the mechanism of RA-mediated myocardial expansion in which RA directly induces hepatic Epo resulting in activation of epicardial Igf2 that stimulates compact zone growth. This RA-EPO-IGF2 signaling axis coordinates liver hematopoiesis with heart development.


Subject(s)
Erythropoietin/metabolism , Insulin-Like Growth Factor II/metabolism , Myocardium/metabolism , Pericardium/metabolism , Tretinoin/pharmacology , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Animals , Cells, Cultured , Chromatin Immunoprecipitation , Erythropoietin/genetics , Heart/drug effects , Heart/embryology , Immunohistochemistry , In Situ Hybridization , Insulin-Like Growth Factor II/genetics , Keratolytic Agents/pharmacology , Liver/drug effects , Liver/metabolism , Mice , Mice, Transgenic , Organ Culture Techniques , Pericardium/drug effects , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
Neural Dev ; 5: 21, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20735826

ABSTRACT

BACKGROUND: Nolz1 is a zinc finger transcription factor whose expression is enriched in the lateral ganglionic eminence (LGE), although its function is still unknown. RESULTS: Here we analyze the role of Nolz1 during LGE development. We show that Nolz1 expression is high in proliferating neural progenitor cells (NPCs) of the LGE subventricular zone. In addition, low levels of Nolz1 are detected in the mantle zone, as well as in the adult striatum. Similarly, Nolz1 is highly expressed in proliferating LGE-derived NPC cultures, but its levels rapidly decrease upon cell differentiation, pointing to a role of Nolz1 in the control of NPC proliferation and/or differentiation. In agreement with this hypothesis, we find that Nolz1 over-expression promotes cell cycle exit of NPCs in neurosphere cultures and negatively regulates proliferation in telencephalic organotypic cultures. Within LGE primary cultures, Nolz1 over-expression promotes the acquisition of a neuronal phenotype, since it increases the number of ß-III tubulin (Tuj1)- and microtubule-associated protein (MAP)2-positive neurons, and inhibits astrocyte generation and/or differentiation. Retinoic acid (RA) is one of the most important morphogens involved in striatal neurogenesis, and regulates Nolz1 expression in different systems. Here we show that Nolz1 also responds to this morphogen in E12.5 LGE-derived cell cultures. However, Nolz1 expression is not regulated by RA in E14.5 LGE-derived cell cultures, nor is it affected during LGE development in mouse models that present decreased RA levels. Interestingly, we find that Gsx2, which is necessary for normal RA signaling during LGE development, is also required for Nolz1 expression, which is lost in Gsx2 knockout mice. These findings suggest that Nolz1 might act downstream of Gsx2 to regulate RA-induced neurogenesis. Keeping with this hypothesis, we show that Nolz1 induces the selective expression of the RA receptor (RAR)ß without altering RARα or RARγ. In addition, Nozl1 over-expression increases RA signaling since it stimulates the RA response element. This RA signaling is essential for Nolz1-induced neurogenesis, which is impaired in a RA-free environment or in the presence of a RAR inverse agonist. It has been proposed that Drosophila Gsx2 and Nolz1 homologues could cooperate with the transcriptional co-repressors Groucho-TLE to regulate cell proliferation. In agreement with this view, we show that Nolz1 could act in collaboration with TLE-4, as they are expressed at the same time in NPC cultures and during mouse development. CONCLUSIONS: Nolz1 promotes RA signaling in the LGE, contributing to the striatal neurogenesis during development.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Corpus Striatum/cytology , Gene Expression Regulation, Developmental/drug effects , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Nuclear Proteins/metabolism , Signal Transduction/physiology , Tretinoin/pharmacology , Animals , Carrier Proteins/genetics , Cell Count , Cell Proliferation , Cells, Cultured , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/physiology , Geniculate Bodies/embryology , Homeodomain Proteins/genetics , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred CBA , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurogenesis/physiology , Neuroglia/drug effects , Neuroglia/physiology , Neurons/physiology , Nuclear Proteins/genetics , Pregnancy , Retinal Dehydrogenase/deficiency , Signal Transduction/genetics , Tubulin/metabolism
13.
Stem Cells Dev ; 19(7): 951-60, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20064013

ABSTRACT

We have introduced 1 to 2 copies of a deletion mutant (betaDeltaC) of the human retinoic acid receptor beta into mouse embryonic stem (ES) cells. The betaDeltaC-expressing cells were 10 to 100 times less sensitive to RA-induced differentiation in comparison with their parental cells. In the presence of 10(-7) M RA in monolayer culture, they showed no growth arrest or differentiation, but remained pluripotent. Embryoid bodies (EBs) derived from betaDeltaC-expressing cells differentiated into cardiomyocytes rather than neurons after treatment with 10(-6) M RA, and became neurons upon exposure to 10(-5) or 10(-4) M RA. Remarkably, after 10 passages of continuous culture in the presence of 10(-7) M RA, they still were able to form chimeras after injection into blastocysts. These data suggest that appropriate levels of normal retinoid receptors are crucial for lineage-specific differentiation of mouse ES cells in vitro. The betaDeltaC mutant protein may prove to be useful in promoting "stemness" of ES cells in culture.


Subject(s)
Cell Differentiation/physiology , Cell Lineage , Embryo, Mammalian , Embryonic Stem Cells/physiology , Receptors, Retinoic Acid , Sequence Deletion , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Humans , Mice , Neurons/cytology , Neurons/physiology , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Tretinoin/metabolism , Tretinoin/pharmacology
14.
J Chem Biol ; 3(3): 113-25, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-21326630

ABSTRACT

UNLABELLED: Two new large poly-1,3-dodecylpyridinium salts, APS12 and APS12-2 of 12.5- and 14.7-kDa size, respectively, were synthesised and tested for their pore-forming and transfection capabilities in HEK 293 and undifferentiated mouse ES cells using patch-clamp recording, Ca(2+) imaging and flow cytometry. Polymerisation reactions were enhanced by microwaves, and the product sizes were controlled by altering the irradiation time. This method can also be applied to obtain polymers with variable linking chains as shown by the preparation of poly-(1,3-octylpyridinium) salt of 11.9-kDa size. Molecular weights of the final products were determined using ESIMS analysis, which also indicated the products to be amongst the largest macro-cycles ever recorded, up to a 900-membered ring. Anti-bacterial, haemolytic and anti-acetylcholinesterase activities were also reported for the two dodecyl pyridinium polymers. These biological activities are characteristic to the structurally related marine toxin, poly-APS. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s12154-010-0036-4) contains supplementary material, which is available to authorized users.

15.
Exp Neurol ; 217(2): 407-16, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19348800

ABSTRACT

Embryonic stem cells (ESCs) promise an unlimited source of defined cells for cell transplantation therapy, while protocols for derivation of homogeneous populations of desirable cell types are yet to be developed and/or refined. Gamma aminobutyric acid (GABA) is a major inhibitory neurotransmitter in the central nervous system, and disturbed GABAergic signaling is associated with a host of neurological conditions. We developed a simple ES cell differentiation protocol which led to the production of uniform GABAergic neurons in approximately 2 weeks. The differentiation protocol involved treatment of embryoid bodies (EBs) with high concentrations (10(-5)-10(-)(4) M) of all-trans-retinoic acid (RA) for 3 days. After plating these EBs on attached dishes in neural supportive medium, 93-96% of the cells became GABA-positive neurons in 7-11 days. These cells also expressed immature neuronal markers with voltage-gated delayed rectifier potassium currents, suggesting that they were immature GABAergic neurons. The technology may have implications for modeling and treatment of GABAergic signaling-related diseases and injuries.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Interneurons/metabolism , Neurogenesis/physiology , Stem Cell Transplantation/methods , gamma-Aminobutyric Acid/metabolism , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation/drug effects , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Interneurons/cytology , Mice , Neurogenesis/drug effects , Potassium Channels, Voltage-Gated/metabolism , Spheroids, Cellular , Tretinoin/pharmacology
16.
J Neurosci ; 28(43): 10893-904, 2008 Oct 22.
Article in English | MEDLINE | ID: mdl-18945897

ABSTRACT

Disrupted-in-Schizophrenia-1 (DISC1), identified by positional cloning of a balanced translocation (1;11) with the breakpoint in intron 8 of a large Scottish pedigree, is associated with a range of neuropsychiatric disorders including schizophrenia. To model this mutation in mice, we have generated Disc1(tr) transgenic mice expressing 2 copies of truncated Disc1 encoding the first 8 exons using a bacterial artificial chromosome (BAC). With this partial simulation of the human situation, we have discovered a range of phenotypes including a series of novel features not previously reported. Disc1(tr) transgenic mice display enlarged lateral ventricles, reduced cerebral cortex, partial agenesis of the corpus callosum, and thinning of layers II/III with reduced neural proliferation at midneurogenesis. Parvalbumin GABAergic neurons are reduced in the hippocampus and medial prefrontal cortex, and displaced in the dorsolateral frontal cortex. In culture, transgenic neurons grow fewer and shorter neurites. Behaviorally, transgenic mice exhibit increased immobility and reduced vocalization in depression-related tests, and impairment in conditioning of latent inhibition. These abnormalities in Disc1(tr) transgenic mice are consistent with findings in severe schizophrenia.


Subject(s)
Behavior, Animal/physiology , Mutation , Nerve Tissue Proteins/genetics , Neurons/pathology , Phenotype , Schizophrenia/genetics , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Embryo, Mammalian , Gene Expression Regulation/genetics , Green Fluorescent Proteins/biosynthesis , Hindlimb Suspension/methods , Inhibition, Psychological , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurites/drug effects , Neurites/pathology , Neurons/drug effects , Parvalbumins/metabolism , Schizophrenia/pathology , Schizophrenia/physiopathology , Swimming
SELECTION OF CITATIONS
SEARCH DETAIL
...