Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 121(26): e2315100121, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38889143

ABSTRACT

Synapses containing γ-aminobutyric acid (GABA) constitute the primary centers for inhibitory neurotransmission in our nervous system. It is unclear how these synaptic structures form and align their postsynaptic machineries with presynaptic terminals. Here, we monitored the cellular distribution of several GABAergic postsynaptic proteins in a purely glutamatergic neuronal culture derived from human stem cells, which virtually lacks any vesicular GABA release. We found that several GABAA receptor (GABAAR) subunits, postsynaptic scaffolds, and major cell-adhesion molecules can reliably coaggregate and colocalize at even GABA-deficient subsynaptic domains, but remain physically segregated from glutamatergic counterparts. Genetic deletions of both Gephyrin and a Gephyrin-associated guanosine di- or triphosphate (GDP/GTP) exchange factor Collybistin severely disrupted the coassembly of these postsynaptic compositions and their proper apposition with presynaptic inputs. Gephyrin-GABAAR clusters, developed in the absence of GABA transmission, could be subsequently activated and even potentiated by delayed supply of vesicular GABA. Thus, molecular organization of GABAergic postsynapses can initiate via a GABA-independent but Gephyrin-dependent intrinsic mechanism.


Subject(s)
Carrier Proteins , Membrane Proteins , Presynaptic Terminals , Receptors, GABA-A , Synapses , gamma-Aminobutyric Acid , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , gamma-Aminobutyric Acid/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA-A/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Presynaptic Terminals/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Rho Guanine Nucleotide Exchange Factors/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics
3.
Biomedicines ; 11(11)2023 Oct 31.
Article in English | MEDLINE | ID: mdl-38001943

ABSTRACT

Cofilactin rod pathology, which can initiate synapse loss, has been extensively studied in rodent neurons, hippocampal slices, and in vivo mouse models of human neurodegenerative diseases such as Alzheimer's disease (AD). In these systems, rod formation induced by disease-associated factors, such as soluble oligomers of Amyloid-ß (Aß) in AD, utilizes a pathway requiring cellular prion protein (PrPC), NADPH oxidase (NOX), and cytokine/chemokine receptors (CCR5 and/or CXCR4). However, rod pathways have not been systematically assessed in a human neuronal model. Here, we characterize glutamatergic neurons differentiated from human-induced pluripotent stem cells (iPSCs) for the formation of rods in response to activators of the PrPC-dependent pathway. Optimization of substratum, cell density, and use of glial-conditioned medium yielded a robust system for studying the development of Aß-induced rods in the absence of glia, suggesting a cell-autonomous pathway. Rod induction in younger neurons requires ectopic expression of PrPC, but this dependency disappears by Day 55. The quantification of proteins within the rod-inducing pathway suggests that increased PrPC and CXCR4 expression may be factors in the doubling of the rod response to Aß between Days 35 and 55. FDA-approved antagonists to CXCR4 and CCR5 inhibit the rod response. Rods were predominantly observed in dendrites, although severe cytoskeletal disruptions prevented the assignment of over 40% of the rods to either an axon or dendrite. In the absence of glia, a condition in which rods are more readily observed, neurons mature and fire action potentials but do not form functional synapses. However, PSD95-containing dendritic spines associate with axonal regions of pre-synaptic vesicles containing the glutamate transporter, VGLUT1. Thus, our results identified stem cell-derived neurons as a robust model for studying cofilactin rod formation in a human cellular environment and for developing effective therapeutic strategies for the treatment of dementias arising from multiple proteinopathies with different rod initiators.

4.
J Biol Chem ; 299(12): 105361, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37865312

ABSTRACT

In recent years, elegant glycomic and glycoproteomic approaches have revealed an intricate glycosylation profile of mammalian brain with enormous spatial and temporal diversities. Nevertheless, at a cellular level, it is unclear how these post-translational modifications affect various proteins to influence crucial neuronal properties. Here, we have investigated the impact of N-linked glycosylation on neuroligins (NLGNs), a class of cell-adhesion molecules that play instructive roles in synapse organization. We found that endogenous NLGN proteins are differentially glycosylated across several regions of murine brain in a sex-independent but isoform-dependent manner. In both rodent primary neurons derived from brain sections and human neurons differentiated from stem cells, all NLGN variants were highly enriched with multiple N-glycan subtypes, which cumulatively ensured their efficient trafficking to the cell surface. Removal of these N-glycosylation residues only had a moderate effect on NLGNs' stability or expression levels but particularly enhanced their retention at the endoplasmic reticulum. As a result, the glycosylation-deficient NLGNs exhibited considerable impairments in their dendritic distribution and postsynaptic accumulation, which in turn, virtually eliminated their ability to recruit presynaptic terminals and significantly reduced NLGN overexpression-induced assemblies of both glutamatergic and GABAergic synapse structures. Therefore, our results highlight an essential mechanistic contribution of N-linked glycosylations in facilitating the appropriate secretory transport of a major synaptic cell-adhesion molecule and promoting its cellular function in neurons.


Subject(s)
Neuroligins , Synapses , Animals , Humans , Mice , Glycosylation , Neuroligins/genetics , Neuroligins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Synapses/metabolism , Neurons/metabolism , Cells, Cultured , Polysaccharides/metabolism , Protein Transport/physiology
5.
Proc Natl Acad Sci U S A ; 120(22): e2300773120, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37216537

ABSTRACT

δ-catenin is expressed in excitatory synapses and functions as an anchor for the glutamatergic AMPA receptor (AMPAR) GluA2 subunit in the postsynaptic density. The glycine 34 to serine (G34S) mutation in the δ-catenin gene has been found in autism spectrum disorder (ASD) patients and results in loss of δ-catenin functions at excitatory synapses, which is presumed to underlie ASD pathogenesis in humans. However, how the G34S mutation causes loss of δ-catenin functions to induce ASD remains unclear. Here, using neuroblastoma cells, we identify that the G34S mutation increases glycogen synthase kinase 3ß (GSK3ß)-dependent δ-catenin degradation to reduce δ-catenin levels, which likely contributes to the loss of δ-catenin functions. Synaptic δ-catenin and GluA2 levels in the cortex are significantly decreased in mice harboring the δ-catenin G34S mutation. The G34S mutation increases glutamatergic activity in cortical excitatory neurons while it is decreased in inhibitory interneurons, indicating changes in cellular excitation and inhibition. δ-catenin G34S mutant mice also exhibit social dysfunction, a common feature of ASD. Most importantly, pharmacological inhibition of GSK3ß activity reverses the G34S-induced loss of δ-catenin function effects in cells and mice. Finally, using δ-catenin knockout mice, we confirm that δ-catenin is required for GSK3ß inhibition-induced restoration of normal social behavior in δ-catenin G34S mutant animals. Taken together, we reveal that the loss of δ-catenin functions arising from the ASD-associated G34S mutation induces social dysfunction via alterations in glutamatergic activity and that GSK3ß inhibition can reverse δ-catenin G34S-induced synaptic and behavioral deficits.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Delta Catenin , Animals , Humans , Mice , Autism Spectrum Disorder/metabolism , Autistic Disorder/genetics , Autistic Disorder/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Mice, Knockout , Social Behavior , Synapses/metabolism
6.
Sci Rep ; 12(1): 22425, 2022 12 27.
Article in English | MEDLINE | ID: mdl-36575212

ABSTRACT

The chromodomain helicase DNA-binding protein CHD8 is the most frequently mutated gene in autism spectrum disorder. Despite its prominent disease involvement, little is known about its molecular function in the human brain. CHD8 is a chromatin regulator which binds to the promoters of actively transcribed genes through genomic targeting mechanisms which have yet to be fully defined. By generating a conditional loss-of-function and an endogenously tagged allele in human pluripotent stem cells, we investigated the molecular function and the interaction of CHD8 with chromatin in human neurons. Chromatin accessibility analysis and transcriptional profiling revealed that CHD8 functions as a transcriptional activator at its target genes in human neurons. Furthermore, we found that CHD8 chromatin targeting is cell context-dependent. In human neurons, CHD8 preferentially binds at ETS motif-enriched promoters. This enrichment is particularly prominent on the promoters of genes whose expression significantly changes upon the loss of CHD8. Indeed, among the ETS transcription factors, we identified ELK1 as being most highly correlated with CHD8 expression in primary human fetal and adult cortical neurons and most highly expressed in our stem cell-derived neurons. Remarkably, ELK1 was necessary to recruit CHD8 specifically to ETS motif-containing sites. These findings imply that ELK1 and CHD8 functionally cooperate to regulate gene expression and chromatin states at MAPK/ERK target genes in human neurons. Our results suggest that the MAPK/ERK/ELK1 axis potentially contributes to the pathogenesis caused by CHD8 mutations in human neurodevelopmental disorders.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Humans , Autistic Disorder/genetics , Autistic Disorder/metabolism , Autism Spectrum Disorder/genetics , Chromatin/genetics , Chromatin/metabolism , Neurons/metabolism , Risk Factors , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism
7.
Nat Commun ; 13(1): 3060, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35650274

ABSTRACT

A vital question in neuroscience is how neurons align their postsynaptic structures with presynaptic release sites. Although synaptic adhesion proteins are known to contribute in this process, the role of neurotransmitters remains unclear. Here we inquire whether de novo biosynthesis and vesicular release of a noncanonical transmitter can facilitate the assembly of its corresponding postsynapses. We demonstrate that, in both stem cell-derived human neurons as well as in vivo mouse neurons of purely glutamatergic identity, ectopic expression of GABA-synthesis enzymes and vesicular transporters is sufficient to both produce GABA from ambient glutamate and transmit it from presynaptic terminals. This enables efficient accumulation and consistent activation of postsynaptic GABAA receptors, and generates fully functional GABAergic synapses that operate in parallel but independently of their glutamatergic counterparts. These findings suggest that presynaptic release of a neurotransmitter itself can signal the organization of relevant postsynaptic apparatus, which could be directly modified to reprogram the synapse identity of neurons.


Subject(s)
Synapses , gamma-Aminobutyric Acid , Animals , Glutamic Acid/metabolism , Mice , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Receptors, GABA-A/metabolism , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism
8.
Stem Cell Reports ; 16(7): 1763-1776, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34171286

ABSTRACT

The differentiation of pluripotent stem cells can be accomplished by sequential activation of signaling pathways or through transcription factor programming. Multistep differentiation imitates embryonic development to obtain authentic cell types, but it suffers from asynchronous differentiation with variable efficiency. Transcription factor programming induces synchronous and efficient differentiation with higher reproducibility but may not always yield authentic cell types. We systematically explored the generation of dopaminergic induced neuronal cells from mouse and human pluripotent stem cells. We found that the proneural factor Ascl1 in combination with mesencephalic factors Lmx1a and Nurr1 induce peripheral dopaminergic neurons. Co-delivery of additional midbrain transcription factors En1, FoxA2, and Pitx3 resulted in facile and robust generation of functional dopaminergic neurons of midbrain character. Our results suggest that more complex combinations of transcription factors may be needed for proper regional specification of induced neuronal cells generated by direct lineage induction.


Subject(s)
Cell Culture Techniques , Dopaminergic Neurons/cytology , Mesencephalon/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Dopamine/metabolism , Embryonic Stem Cells/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Mice , Signal Transduction , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/metabolism , Wnt1 Protein/metabolism
9.
J Neurosci ; 41(3): 392-407, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33268543

ABSTRACT

Neuroligins (NLGNs) are a class of postsynaptic cell adhesion molecules that interact with presynaptic neurexins (NRXNs) and regulate synapse function. NLGN4 is a member of the NLGN family and consists of a unique amino acid sequence in humans that is not evolutionarily well conserved in rodents. The human-specific NLGN4 gene has been reported to be mutated in many patients with autism and other neurodevelopmental disorders. However, it remained unclear how these mutations might alter the molecular properties of NLGN4 and affect synaptic transmission in human neurons. Here, we describe a severely autistic male patient carrying a single amino acid substitution (R101Q) in the NLGN4 gene. When expressed in HEK293 cells, the R101Q mutation in NLGN4 did not affect its binding affinity for NRXNs or its capacity to form homodimers. This mutation, however, impaired the maturation of NLGN4 protein by inhibiting N-linked glycosylation at an adjacent residue (N102), which is conserved in all NLGNs. As a result, the R101Q substitution significantly decreased the surface trafficking of NLGN4 and increased its retention in the endoplasmic reticulum and Golgi apparatus. In human neurons derived from male stem cell lines, the R101Q mutation also similarly reduced the synaptic localization of NLGN4, resulting in a loss-of-function phenotype. This mutation-induced trafficking defect substantially diminished the ability of NLGN4 to form excitatory synapses and modulate their functional properties. Viewed together, our findings suggest that the R101Q mutation is pathogenic for NLGN4 and can lead to synaptic dysfunction in autism.


Subject(s)
Autistic Disorder/genetics , Cell Adhesion Molecules, Neuronal/genetics , Excitatory Postsynaptic Potentials/physiology , Mutation/genetics , Synaptic Transmission/physiology , Amino Acid Substitution , Autistic Disorder/psychology , Child , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Male , Mutation, Missense/genetics , Neural Stem Cells , Neuropsychological Tests , Patch-Clamp Techniques , Synapses/metabolism
10.
Nat Cell Biol ; 22(4): 401-411, 2020 04.
Article in English | MEDLINE | ID: mdl-32231311

ABSTRACT

The on-target pioneer factors Ascl1 and Myod1 are sequence-related but induce two developmentally unrelated lineages-that is, neuronal and muscle identities, respectively. It is unclear how these two basic helix-loop-helix (bHLH) factors mediate such fundamentally different outcomes. The chromatin binding of Ascl1 and Myod1 was surprisingly similar in fibroblasts, yet their transcriptional outputs were drastically different. We found that quantitative binding differences explained differential chromatin remodelling and gene activation. Although strong Ascl1 binding was exclusively associated with bHLH motifs, strong Myod1-binding sites were co-enriched with non-bHLH motifs, possibly explaining why Ascl1 is less context dependent. Finally, we observed that promiscuous binding of Myod1 to neuronal targets results in neuronal reprogramming when the muscle program is inhibited by Myt1l. Our findings suggest that chromatin access of on-target pioneer factors is primarily driven by the protein-DNA interaction, unlike ordinary context-dependent transcription factors, and that promiscuous transcription factor binding requires specific silencing mechanisms to ensure lineage fidelity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , MyoD Protein/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Transcription Factors/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , Cell Lineage/genetics , Cellular Reprogramming , Chromatin/chemistry , Chromatin/metabolism , Embryo, Mammalian , Fibroblasts/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , MyoD Protein/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Nucleotide Motifs , Protein Binding , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic
11.
Neuron ; 103(4): 617-626.e6, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31257103

ABSTRACT

The autism-associated synaptic-adhesion gene Neuroligin-4 (NLGN4) is poorly conserved evolutionarily, limiting conclusions from Nlgn4 mouse models for human cells. Here, we show that the cellular and subcellular expression of human and murine Neuroligin-4 differ, with human Neuroligin-4 primarily expressed in cerebral cortex and localized to excitatory synapses. Overexpression of NLGN4 in human embryonic stem cell-derived neurons resulted in an increase in excitatory synapse numbers but a remarkable decrease in synaptic strength. Human neurons carrying the syndromic autism mutation NLGN4-R704C also formed more excitatory synapses but with increased functional synaptic transmission due to a postsynaptic mechanism, while genetic loss of NLGN4 did not significantly affect synapses in the human neurons analyzed. Thus, the NLGN4-R704C mutation represents a change-of-function mutation. Our work reveals contrasting roles of NLGN4 in human and mouse neurons, suggesting that human evolution has impacted even fundamental cell biological processes generally assumed to be highly conserved.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Synaptic Transmission/physiology , Animals , Autistic Disorder/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Cerebral Cortex/physiology , Embryonic Stem Cells/cytology , Excitatory Postsynaptic Potentials/physiology , Genes, Reporter , Glutamic Acid/physiology , Humans , Mice , Miniature Postsynaptic Potentials/physiology , Mutation, Missense , Neurogenesis , Neurons/physiology , Phenotype , Receptors, Glutamate/physiology , Species Specificity , Synapses/chemistry
12.
Cell Stem Cell ; 25(1): 103-119.e6, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31155484

ABSTRACT

Human pluripotent stem cells can be rapidly converted into functional neurons by ectopic expression of proneural transcription factors. Here we show that directly reprogrammed neurons, despite their rapid maturation kinetics, can model teratogenic mechanisms that specifically affect early neurodevelopment. We delineated distinct phases of in vitro maturation during reprogramming of human neurons and assessed the cellular phenotypes of valproic acid (VPA), a teratogenic drug. VPA exposure caused chronic impairment of dendritic morphology and functional properties of developing neurons, but not those of mature neurons. These pathogenic effects were associated with VPA-mediated inhibition of the histone deacetylase (HDAC) and glycogen synthase kinase-3 (GSK-3) pathways, which caused transcriptional downregulation of many genes, including MARCKSL1, an actin-stabilizing protein essential for dendritic morphogenesis and synapse maturation during early neurodevelopment. Our findings identify a developmentally restricted pathogenic mechanism of VPA and establish the use of reprogrammed neurons as an effective platform for modeling teratogenic pathways.


Subject(s)
Calmodulin-Binding Proteins/metabolism , Electrical Synapses/metabolism , Microfilament Proteins/metabolism , Neurons/physiology , Pluripotent Stem Cells/physiology , Teratoma/metabolism , Animals , Calmodulin-Binding Proteins/genetics , Carcinogenesis , Cells, Cultured , Cellular Reprogramming , Glycogen Synthase Kinase 3/metabolism , Histone Deacetylases/metabolism , Humans , Mice , Microfilament Proteins/genetics , Neurogenesis , Signal Transduction , Teratoma/chemically induced , Teratoma/pathology , Valproic Acid/toxicity
13.
Proc Natl Acad Sci U S A ; 115(25): 6470-6475, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29866841

ABSTRACT

Human cell models for disease based on induced pluripotent stem (iPS) cells have proven to be powerful new assets for investigating disease mechanisms. New insights have been obtained studying single mutations using isogenic controls generated by gene targeting. Modeling complex, multigenetic traits using patient-derived iPS cells is much more challenging due to line-to-line variability and technical limitations of scaling to dozens or more patients. Induced neuronal (iN) cells reprogrammed directly from dermal fibroblasts or urinary epithelia could be obtained from many donors, but such donor cells are heterogeneous, show interindividual variability, and must be extensively expanded, which can introduce random mutations. Moreover, derivation of dermal fibroblasts requires invasive biopsies. Here we show that human adult peripheral blood mononuclear cells, as well as defined purified T lymphocytes, can be directly converted into fully functional iN cells, demonstrating that terminally differentiated human cells can be efficiently transdifferentiated into a distantly related lineage. T cell-derived iN cells, generated by nonintegrating gene delivery, showed stereotypical neuronal morphologies and expressed multiple pan-neuronal markers, fired action potentials, and were able to form functional synapses. These cells were stable in the absence of exogenous reprogramming factors. Small molecule addition and optimized culture systems have yielded conversion efficiencies of up to 6.2%, resulting in the generation of >50,000 iN cells from 1 mL of peripheral blood in a single step without the need for initial expansion. Thus, our method allows the generation of sufficient neurons for experimental interrogation from a defined, homogeneous, and readily accessible donor cell population.


Subject(s)
Cell Differentiation/physiology , Cell Transdifferentiation/physiology , Leukocytes, Mononuclear/cytology , Neurons/cytology , T-Lymphocytes/cytology , Adolescent , Adult , Aged , Cellular Reprogramming/physiology , Female , Fibroblasts/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Male , Middle Aged , Young Adult
14.
J Neurosci ; 37(29): 6816-6836, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28607166

ABSTRACT

Neuroligins are evolutionarily conserved postsynaptic cell adhesion molecules that interact with presynaptic neurexins. Neurons express multiple neuroligin isoforms that are targeted to specific synapses, but their synaptic functions and mechanistic redundancy are not completely understood. Overexpression or RNAi-mediated knockdown of neuroligins, respectively, causes a dramatic increase or decrease in synapse density, whereas genetic deletions of neuroligins impair synapse function with only minor effects on synapse numbers, raising fundamental questions about the overall physiological role of neuroligins. Here, we have systematically analyzed the effects of conditional genetic deletions of all major neuroligin isoforms (i.e., NL1, NL2, and NL3), either individually or in combinations, in cultured mouse hippocampal and cortical neurons. We found that conditional genetic deletions of neuroligins caused no change or only a small change in synapses numbers, but strongly impaired synapse function. This impairment was isoform specific, suggesting that neuroligins are not functionally redundant. Sparse neuroligin deletions produced phenotypes comparable to those of global deletions, indicating that neuroligins function in a cell-autonomous manner. Mechanistically, neuroligin deletions decreased the synaptic levels of neurotransmitter receptors and had no effect on presynaptic release probabilities. Overexpression of neuroligin-1 in control or neuroligin-deficient neurons increased synaptic transmission and synapse density but not spine numbers, suggesting that these effects reflect a gain-of-function mechanism; whereas overexpression of neuroligin-3, which, like neuroligin-1 is also targeted to excitatory synapses, had no comparable effect. Our data demonstrate that neuroligins are required for the physiological organization of neurotransmitter receptors in postsynaptic specializations and suggest that they do not play a major role in synapse formation.SIGNIFICANCE STATEMENT Human neuroligin genes have been associated with autism, but the cellular functions of different neuroligins and their molecular mechanisms remain incompletely understood. Here, we performed comparative analyses in cultured mouse neurons of all major neuroligin isoforms, either individually or in combinations, using conditional knockouts. We found that neuroligin deletions did not affect synapse numbers but differentially impaired excitatory or inhibitory synaptic functions in an isoform-specific manner. These impairments were due, at least in part, to a decrease in synaptic distribution of neurotransmitter receptors upon deletion of neuroligins. Conversely, the overexpression of neuroligin-1 increased synapse numbers but not spine numbers. Our results suggest that various neuroligin isoforms perform unique postsynaptic functions in organizing synapses but are not essential for synapse formation or maintenance.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Inhibition/physiology , Neurogenesis/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Animals, Newborn , Cells, Cultured , Excitatory Postsynaptic Potentials/physiology , Female , Gene Expression Regulation, Developmental/physiology , Male , Mice , Mice, Knockout
15.
Nat Methods ; 14(6): 621-628, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28504679

ABSTRACT

Approaches to differentiating pluripotent stem cells (PSCs) into neurons currently face two major challenges-(i) generated cells are immature, with limited functional properties; and (ii) cultures exhibit heterogeneous neuronal subtypes and maturation stages. Using lineage-determining transcription factors, we previously developed a single-step method to generate glutamatergic neurons from human PSCs. Here, we show that transient expression of the transcription factors Ascl1 and Dlx2 (AD) induces the generation of exclusively GABAergic neurons from human PSCs with a high degree of synaptic maturation. These AD-induced neuronal (iN) cells represent largely nonoverlapping populations of GABAergic neurons that express various subtype-specific markers. We further used AD-iN cells to establish that human collybistin, the loss of gene function of which causes severe encephalopathy, is required for inhibitory synaptic function. The generation of defined populations of functionally mature human GABAergic neurons represents an important step toward enabling the study of diseases affecting inhibitory synaptic transmission.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , GABAergic Neurons/cytology , GABAergic Neurons/physiology , Homeodomain Proteins/genetics , Pluripotent Stem Cells/physiology , Transcription Factors/genetics , Animals , Cell Engineering , Cells, Cultured , Humans , Mice , Pluripotent Stem Cells/cytology
16.
Nature ; 544(7649): 245-249, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28379941

ABSTRACT

Normal differentiation and induced reprogramming require the activation of target cell programs and silencing of donor cell programs. In reprogramming, the same factors are often used to reprogram many different donor cell types. As most developmental repressors, such as RE1-silencing transcription factor (REST) and Groucho (also known as TLE), are considered lineage-specific repressors, it remains unclear how identical combinations of transcription factors can silence so many different donor programs. Distinct lineage repressors would have to be induced in different donor cell types. Here, by studying the reprogramming of mouse fibroblasts to neurons, we found that the pan neuron-specific transcription factor Myt1-like (Myt1l) exerts its pro-neuronal function by direct repression of many different somatic lineage programs except the neuronal program. The repressive function of Myt1l is mediated via recruitment of a complex containing Sin3b by binding to a previously uncharacterized N-terminal domain. In agreement with its repressive function, the genomic binding sites of Myt1l are similar in neurons and fibroblasts and are preferentially in an open chromatin configuration. The Notch signalling pathway is repressed by Myt1l through silencing of several members, including Hes1. Acute knockdown of Myt1l in the developing mouse brain mimicked a Notch gain-of-function phenotype, suggesting that Myt1l allows newborn neurons to escape Notch activation during normal development. Depletion of Myt1l in primary postmitotic neurons de-repressed non-neuronal programs and impaired neuronal gene expression and function, indicating that many somatic lineage programs are actively and persistently repressed by Myt1l to maintain neuronal identity. It is now tempting to speculate that similar 'many-but-one' lineage repressors exist for other cell fates; such repressors, in combination with lineage-specific activators, would be prime candidates for use in reprogramming additional cell types.


Subject(s)
Cell Lineage/genetics , Cellular Reprogramming/genetics , Gene Silencing , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Neurons/cytology , Neurons/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Brain/cytology , Brain/embryology , Brain/metabolism , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mice , Nerve Tissue Proteins/deficiency , Organ Specificity/genetics , Protein Domains , Receptors, Notch/deficiency , Repressor Proteins/chemistry , Repressor Proteins/deficiency , Signal Transduction , Transcription Factor HES-1/deficiency , Transcription Factors/deficiency
17.
Cell Stem Cell ; 20(3): 329-344.e7, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28089908

ABSTRACT

Understanding the relative contributions of genetic and epigenetic abnormalities to acute myeloid leukemia (AML) should assist integrated design of targeted therapies. In this study, we generated induced pluripotent stem cells (iPSCs) from AML patient samples harboring MLL rearrangements and found that they retained leukemic mutations but reset leukemic DNA methylation/gene expression patterns. AML-iPSCs lacked leukemic potential, but when differentiated into hematopoietic cells, they reacquired the ability to give rise to leukemia in vivo and reestablished leukemic DNA methylation/gene expression patterns, including an aberrant MLL signature. Epigenetic reprogramming was therefore not sufficient to eliminate leukemic behavior. This approach also allowed us to study the properties of distinct AML subclones, including differential drug susceptibilities of KRAS mutant and wild-type cells, and predict relapse based on increased cytarabine resistance of a KRAS wild-type subclone. Overall, our findings illustrate the value of AML-iPSCs for investigating the mechanistic basis and clonal properties of human AML.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Leukemia, Myeloid, Acute/pathology , Models, Biological , Blast Crisis/pathology , Cell Line, Tumor , Cell Lineage , Cell Shape , Cellular Reprogramming , Chromosome Aberrations , Clone Cells , DNA Methylation/genetics , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Leukemic , HEK293 Cells , Hematopoiesis/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Molecular Targeted Therapy , Mutation/genetics , Neoplasm Invasiveness , Phenotype
18.
Proc Natl Acad Sci U S A ; 113(30): 8514-9, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27402759

ABSTRACT

We and others have shown that embryonic and neonatal fibroblasts can be directly converted into induced neuronal (iN) cells with mature functional properties. Reprogramming of fibroblasts from adult and aged mice, however, has not yet been explored in detail. The ability to generate fully functional iN cells from aged organisms will be particularly important for in vitro modeling of diseases of old age. Here, we demonstrate production of functional iN cells from fibroblasts that were derived from mice close to the end of their lifespan. iN cells from aged mice had apparently normal active and passive neuronal membrane properties and formed abundant synaptic connections. The reprogramming efficiency gradually decreased with fibroblasts derived from embryonic and neonatal mice, but remained similar for fibroblasts from postnatal mice of all ages. Strikingly, overexpression of a transcription factor, forkhead box O3 (FoxO3), which is implicated in aging, blocked iN cell conversion of embryonic fibroblasts, whereas knockout or knockdown of FoxO3 increased the reprogramming efficiency of adult-derived but not of embryonic fibroblasts and also enhanced functional maturation of resulting iN cells. Hence, FoxO3 has a central role in the neuronal reprogramming susceptibility of cells, and the importance of FoxO3 appears to change during development.


Subject(s)
Aging , Cellular Reprogramming/genetics , Forkhead Box Protein O3/genetics , Neurons/metabolism , Animals , Animals, Newborn , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Forkhead Box Protein O3/deficiency , Gene Expression Regulation, Developmental , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology
19.
Stem Cell Reports ; 3(2): 282-96, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25254342

ABSTRACT

Direct conversion of nonneural cells to functional neurons holds great promise for neurological disease modeling and regenerative medicine. We previously reported rapid reprogramming of mouse embryonic fibroblasts (MEFs) into mature induced neuronal (iN) cells by forced expression of three transcription factors: ASCL1, MYT1L, and BRN2. Here, we show that ASCL1 alone is sufficient to generate functional iN cells from mouse and human fibroblasts and embryonic stem cells, indicating that ASCL1 is the key driver of iN cell reprogramming in different cell contexts and that the role of MYT1L and BRN2 is primarily to enhance the neuronal maturation process. ASCL1-induced single-factor neurons (1F-iN) expressed mature neuronal markers, exhibited typical passive and active intrinsic membrane properties, and formed functional pre- and postsynaptic structures. Surprisingly, ASCL1-induced iN cells were predominantly excitatory, demonstrating that ASCL1 is permissive but alone not deterministic for the inhibitory neuronal lineage.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cellular Reprogramming , Neural Stem Cells/cytology , Action Potentials , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Humans , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , POU Domain Factors/genetics , POU Domain Factors/metabolism , Patch-Clamp Techniques , Potassium Channels/metabolism , Sodium Channels/metabolism , Synapses/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Cell ; 155(3): 621-35, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24243019

ABSTRACT

Direct lineage reprogramming is a promising approach for human disease modeling and regenerative medicine, with poorly understood mechanisms. Here, we reveal a hierarchical mechanism in the direct conversion of fibroblasts into induced neuronal (iN) cells mediated by the transcription factors Ascl1, Brn2, and Myt1l. Ascl1 acts as an "on-target" pioneer factor by immediately occupying most cognate genomic sites in fibroblasts. In contrast, Brn2 and Myt1l do not access fibroblast chromatin productively on their own; instead, Ascl1 recruits Brn2 to Ascl1 sites genome wide. A unique trivalent chromatin signature in the host cells predicts the permissiveness for Ascl1 pioneering activity among different cell types. Finally, we identified Zfp238 as a key Ascl1 target gene that can partially substitute for Ascl1 during iN cell reprogramming. Thus, a precise match between pioneer factors and the chromatin context at key target genes is determinative for transdifferentiation to neurons and likely other cell types.


Subject(s)
Cellular Reprogramming , Embryo, Mammalian/cytology , Fibroblasts/cytology , Gene Regulatory Networks , Neurons/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Chromatin/metabolism , Fibroblasts/metabolism , Genome-Wide Association Study , Humans , Mice , Nerve Tissue Proteins/metabolism , Neurons/metabolism , POU Domain Factors/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...