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










Publication year range
1.
Adv Sci (Weinh) ; : e2400545, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773714

ABSTRACT

Standard single-cell (sc) proteomics of disease states inferred from multicellular organs or organoids cannot currently be related to single-cell physiology. Here, a scPatch-Clamp/Proteomics platform is developed on single neurons generated from hiPSCs bearing an Alzheimer's disease (AD) genetic mutation and compares them to isogenic wild-type controls. This approach provides both current and voltage electrophysiological data plus detailed proteomics information on single-cells. With this new method, the authors are able to observe hyperelectrical activity in the AD hiPSC-neurons, similar to that observed in the human AD brain, and correlate it to ≈1400 proteins detected at the single neuron level. Using linear regression and mediation analyses to explore the relationship between the abundance of individual proteins and the neuron's mutational and electrophysiological status, this approach yields new information on therapeutic targets in excitatory neurons not attainable by traditional methods. This combined patch-proteomics technique creates a new proteogenetic-therapeutic strategy to correlate genotypic alterations to physiology with protein expression in single-cells.

3.
Nat Chem Biol ; 19(3): 275-283, 2023 03.
Article in English | MEDLINE | ID: mdl-36175661

ABSTRACT

Prevention of infection and propagation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a high priority in the Coronavirus Disease 2019 (COVID-19) pandemic. Here we describe S-nitrosylation of multiple proteins involved in SARS-CoV-2 infection, including angiotensin-converting enzyme 2 (ACE2), the receptor for viral entry. This reaction prevents binding of ACE2 to the SARS-CoV-2 spike protein, thereby inhibiting viral entry, infectivity and cytotoxicity. Aminoadamantane compounds also inhibit coronavirus ion channels formed by envelope (E) protein. Accordingly, we developed dual-mechanism aminoadamantane nitrate compounds that inhibit viral entry and, thus, the spread of infection by S-nitrosylating ACE2 via targeted delivery of the drug after E protein channel blockade. These non-toxic compounds are active in vitro and in vivo in the Syrian hamster COVID-19 model and, thus, provide a novel avenue to pursue therapy.


Subject(s)
COVID-19 , Humans , SARS-CoV-2/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Protein Binding , Peptidyl-Dipeptidase A/metabolism
4.
bioRxiv ; 2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35411336

ABSTRACT

Prevention of infection and propagation of SARS-CoV-2 is of high priority in the COVID-19 pandemic. Here, we describe S-nitrosylation of multiple proteins involved in SARS-CoV-2 infection, including angiotensin converting enzyme 2 (ACE2), the receptor for viral entry. This reaction prevents binding of ACE2 to the SARS-CoV-2 Spike protein, thereby inhibiting viral entry, infectivity, and cytotoxicity. Aminoadamantane compounds also inhibit coronavirus ion channels formed by envelope (E) protein. Accordingly, we developed dual-mechanism aminoadamantane nitrate compounds that inhibit viral entry and thus spread of infection by S-nitrosylating ACE2 via targeted delivery of the drug after E-protein channel blockade. These non-toxic compounds are active in vitro and in vivo in the Syrian hamster COVID-19 model, and thus provide a novel avenue for therapy.

5.
J Neurosci ; 41(10): 2264-2273, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33483428

ABSTRACT

Synaptic and neuronal loss are major neuropathological characteristics of Parkinson's disease. Misfolded protein aggregates in the form of Lewy bodies, comprised mainly of α-synuclein (αSyn), are associated with disease progression, and have also been linked to other neurodegenerative diseases, including Lewy body dementia, Alzheimer's disease, and frontotemporal dementia. However, the effects of αSyn and its mechanism of synaptic damage remain incompletely understood. Here, we show that αSyn oligomers induce Ca2+-dependent release of glutamate from astrocytes obtained from male and female mice, and that mice overexpressing αSyn manifest increased tonic release of glutamate in vivo In turn, this extracellular glutamate activates glutamate receptors, including extrasynaptic NMDARs (eNMDARs), on neurons both in culture and in hippocampal slices of αSyn-overexpressing mice. Additionally, in patch-clamp recording from outside-out patches, we found that oligomerized αSyn can directly activate eNMDARs. In organotypic slices, oligomeric αSyn induces eNMDAR-mediated synaptic loss, which can be reversed by the drug NitroSynapsin. When we expose human induced pluripotent stem cell-derived cerebrocortical neurons to αSyn, we find similar effects. Importantly, the improved NMDAR antagonist NitroSynapsin, which selectively inhibits extrasynaptic over physiological synaptic NMDAR activity, protects synapses from oligomeric αSyn-induced damage in our model systems, thus meriting further study for its therapeutic potential.SIGNIFICANCE STATEMENT Loss of synaptic function and ensuing neuronal loss are associated with disease progression in Parkinson's disease (PD), Lewy body dementia (LBD), and other neurodegenerative diseases. However, the mechanism of synaptic damage remains incompletely understood. α-Synuclein (αSyn) misfolds in PD/LBD, forming Lewy bodies and contributing to disease pathogenesis. Here, we found that misfolded/oligomeric αSyn releases excessive astrocytic glutamate, in turn activating neuronal extrasynaptic NMDA receptors (eNMDARs), thereby contributing to synaptic damage. Additionally, αSyn oligomers directly activate eNMDARs, further contributing to damage. While the FDA-approved drug memantine has been reported to offer some benefit in PD/LBD (Hershey and Coleman-Jackson, 2019), we find that the improved eNMDAR antagonist NitroSynapsin ameliorates αSyn-induced synaptic spine loss, providing potential disease-modifying intervention in PD/LBD.


Subject(s)
Astrocytes/metabolism , Glutamic Acid/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , alpha-Synuclein/metabolism , Animals , Astrocytes/drug effects , Astrocytes/pathology , Cells, Cultured , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synapses/metabolism , Synapses/pathology , alpha-Synuclein/pharmacology
6.
Annu Rev Pharmacol Toxicol ; 61: 701-721, 2021 01 06.
Article in English | MEDLINE | ID: mdl-32997602

ABSTRACT

Excitatory/inhibitory (E/I) balance, defined as the balance between excitation and inhibition of synaptic activity in a neuronal network, accounts in part for the normal functioning of the brain, controlling, for example, normal spike rate. In many pathological conditions, this fine balance is perturbed, leading to excessive or diminished excitation relative to inhibition, termed E/I imbalance, reflected in network dysfunction. E/I imbalance has emerged as a contributor to neurological disorders that occur particularly at the extremes of life, including autism spectrum disorder and Alzheimer's disease, pointing to the vulnerability of neuronal networks at these critical life stages. Hence, it is important to develop approaches to rebalance neural networks. In this review, we describe emerging therapies that can normalize the E/I ratio or the underlying abnormality that contributes to the imbalance in electrical activity, thus improving neurological function in these maladies.


Subject(s)
Autism Spectrum Disorder , Neurodegenerative Diseases , Brain , Humans , Neurons
7.
Mol Psychiatry ; 26(10): 5751-5765, 2021 10.
Article in English | MEDLINE | ID: mdl-32467645

ABSTRACT

Beginning at early stages, human Alzheimer's disease (AD) brains manifest hyperexcitability, contributing to subsequent extensive synapse loss, which has been linked to cognitive dysfunction. No current therapy for AD is disease-modifying. Part of the problem with AD drug discovery is that transgenic mouse models have been poor predictors of potential human treatment. While it is undoubtedly important to test drugs in these animal models, additional evidence for drug efficacy in a human context might improve our chances of success. Accordingly, in order to test drugs in a human context, we have developed a platform of physiological assays using patch-clamp electrophysiology, calcium imaging, and multielectrode array (MEA) experiments on human (h)iPSC-derived 2D cortical neuronal cultures and 3D cerebral organoids. We compare hiPSCs bearing familial AD mutations vs. their wild-type (WT) isogenic controls in order to characterize the aberrant electrical activity in such a human context. Here, we show that these AD neuronal cultures and organoids manifest increased spontaneous action potentials, slow oscillatory events (~1 Hz), and hypersynchronous network activity. Importantly, the dual-allosteric NMDAR antagonist NitroSynapsin, but not the FDA-approved drug memantine, abrogated this hyperactivity. We propose a novel model of synaptic plasticity in which aberrant neural networks are rebalanced by NitroSynapsin. We propose that hiPSC models may be useful for screening drugs to treat hyperexcitability and related synaptic damage in AD.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Action Potentials , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Animals , Disease Models, Animal , Mice , Neural Networks, Computer , Neurons
8.
Elife ; 82019 11 29.
Article in English | MEDLINE | ID: mdl-31782729

ABSTRACT

Human Alzheimer's disease (AD) brains and transgenic AD mouse models manifest hyperexcitability. This aberrant electrical activity is caused by synaptic dysfunction that represents the major pathophysiological correlate of cognitive decline. However, the underlying mechanism for this excessive excitability remains incompletely understood. To investigate the basis for the hyperactivity, we performed electrophysiological and immunofluorescence studies on hiPSC-derived cerebrocortical neuronal cultures and cerebral organoids bearing AD-related mutations in presenilin-1 or amyloid precursor protein vs. isogenic gene corrected controls. In the AD hiPSC-derived neurons/organoids, we found increased excitatory bursting activity, which could be explained in part by a decrease in neurite length. AD hiPSC-derived neurons also displayed increased sodium current density and increased excitatory and decreased inhibitory synaptic activity. Our findings establish hiPSC-derived AD neuronal cultures and organoids as a relevant model of early AD pathophysiology and provide mechanistic insight into the observed hyperexcitability.


Subject(s)
Action Potentials , Alzheimer Disease/physiopathology , Cerebrum/cytology , Cortical Excitability , Electrophysiological Phenomena , Induced Pluripotent Stem Cells/physiology , Neurons/physiology , Amyloid beta-Protein Precursor/genetics , Animals , Cell Size , Cells, Cultured , Fluorescent Antibody Technique , Humans , Mice , Models, Theoretical , Mutant Proteins/genetics , Organoids , Presenilin-1/genetics
9.
Stem Cell Reports ; 7(3): 527-542, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27569059

ABSTRACT

Controlled differentiation of human embryonic stem cells (hESCs) can be utilized for precise analysis of cell type identities during early development. We established a highly efficient neural induction strategy and an improved analytical platform, and determined proteomic and phosphoproteomic profiles of hESCs and their specified multipotent neural stem cell derivatives (hNSCs). This quantitative dataset (nearly 13,000 proteins and 60,000 phosphorylation sites) provides unique molecular insights into pluripotency and neural lineage entry. Systems-level comparative analysis of proteins (e.g., transcription factors, epigenetic regulators, kinase families), phosphorylation sites, and numerous biological pathways allowed the identification of distinct signatures in pluripotent and multipotent cells. Furthermore, as predicted by the dataset, we functionally validated an autocrine/paracrine mechanism by demonstrating that the secreted protein midkine is a regulator of neural specification. This resource is freely available to the scientific community, including a searchable website, PluriProt.


Subject(s)
Cell Differentiation , Neurons/metabolism , Phosphoproteins/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Proteome , Proteomics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Computational Biology/methods , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Neurons/cytology , Proteomics/methods , Signal Transduction , Transcriptome
11.
Sci Rep ; 5: 14781, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26477507

ABSTRACT

Stroke and vascular dementia are leading causes of morbidity and mortality. Neuroprotective therapies have been proposed but none have proven clinically tolerated and effective. While overstimulation of N-methyl-d-aspartate-type glutamate receptors (NMDARs) is thought to contribute to cerebrovascular insults, the importance of NMDARs in physiological function has made this target, at least in the view of many in 'Big Pharma,' 'undruggable' for this indication. Here, we describe novel NitroMemantine drugs, comprising an adamantane moiety that binds in the NMDAR-associated ion channel that is used to target a nitro group to redox-mediated regulatory sites on the receptor. The NitroMemantines are both well tolerated and effective against cerebral infarction in rodent models via a dual allosteric mechanism of open-channel block and NO/redox modulation of the receptor. Targeted S-nitrosylation of NMDARs by NitroMemantine is potentiated by hypoxia and thereby directed at ischemic neurons. Allosteric approaches to tune NMDAR activity may hold therapeutic potential for cerebrovascular disorders.


Subject(s)
Cerebrovascular Disorders/metabolism , Memantine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Anura , Apoptosis/drug effects , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebrovascular Disorders/drug therapy , Cerebrovascular Disorders/pathology , Long-Term Potentiation/drug effects , Maze Learning/drug effects , Memantine/analogs & derivatives , Memantine/therapeutic use , Membrane Potentials/drug effects , Nitric Oxide/metabolism , Oxidation-Reduction/drug effects , Rats , Synaptic Transmission/drug effects
13.
Cell ; 155(6): 1351-64, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24290359

ABSTRACT

Parkinson's disease (PD) is characterized by loss of A9 dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). An association has been reported between PD and exposure to mitochondrial toxins, including environmental pesticides paraquat, maneb, and rotenone. Here, using a robust, patient-derived stem cell model of PD allowing comparison of A53T α-synuclein (α-syn) mutant cells and isogenic mutation-corrected controls, we identify mitochondrial toxin-induced perturbations in A53T α-syn A9 DA neurons (hNs). We report a pathway whereby basal and toxin-induced nitrosative/oxidative stress results in S-nitrosylation of transcription factor MEF2C in A53T hNs compared to corrected controls. This redox reaction inhibits the MEF2C-PGC1α transcriptional network, contributing to mitochondrial dysfunction and apoptotic cell death. Our data provide mechanistic insight into gene-environmental interaction (GxE) in the pathogenesis of PD. Furthermore, using small-molecule high-throughput screening, we identify the MEF2C-PGC1α pathway as a therapeutic target to combat PD.


Subject(s)
Gene-Environment Interaction , Mitochondria/drug effects , Paraquat/toxicity , Parkinson Disease/genetics , Parkinson Disease/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , MEF2 Transcription Factors , Mutation/drug effects , Neurons/metabolism , Oxidative Stress , Parkinson Disease/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Nitrogen Species/metabolism , Substantia Nigra/metabolism , Transcription Factors/metabolism , Transcription, Genetic , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
14.
Proc Natl Acad Sci U S A ; 110(27): E2518-27, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23776240

ABSTRACT

Synaptic loss is the cardinal feature linking neuropathology to cognitive decline in Alzheimer's disease (AD). However, the mechanism of synaptic damage remains incompletely understood. Here, using FRET-based glutamate sensor imaging, we show that amyloid-ß peptide (Aß) engages α7 nicotinic acetylcholine receptors to induce release of astrocytic glutamate, which in turn activates extrasynaptic NMDA receptors (eNMDARs) on neurons. In hippocampal autapses, this eNMDAR activity is followed by reduction in evoked and miniature excitatory postsynaptic currents (mEPSCs). Decreased mEPSC frequency may reflect early synaptic injury because of concurrent eNMDAR-mediated NO production, tau phosphorylation, and caspase-3 activation, each of which is implicated in spine loss. In hippocampal slices, oligomeric Aß induces eNMDAR-mediated synaptic depression. In AD-transgenic mice compared with wild type, whole-cell recordings revealed excessive tonic eNMDAR activity accompanied by eNMDAR-sensitive loss of mEPSCs. Importantly, the improved NMDAR antagonist NitroMemantine, which selectively inhibits extrasynaptic over physiological synaptic NMDAR activity, protects synapses from Aß-induced damage both in vitro and in vivo.


Subject(s)
Amyloid beta-Peptides/toxicity , Astrocytes/metabolism , Glutamic Acid/metabolism , Neural Inhibition/physiology , Peptide Fragments/toxicity , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes/pathology , Coculture Techniques , Female , Fluorescence Resonance Energy Transfer , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Transgenic , Rats , Receptors, Nicotinic/metabolism , Synapses/metabolism , alpha7 Nicotinic Acetylcholine Receptor
15.
J Neurosci ; 32(45): 15837-42, 2012 Nov 07.
Article in English | MEDLINE | ID: mdl-23136422

ABSTRACT

After transplantation, individual stem cell-derived neurons can functionally integrate into the host CNS; however, evidence that neurons derived from transplanted human embryonic stem cells (hESCs) can drive endogenous neuronal network activity in CNS tissue is still lacking. Here, using multielectrode array recordings, we report activation of high-frequency oscillations in the ß and γ ranges (10-100 Hz) in the host hippocampal network via targeted optogenetic stimulation of transplanted hESC-derived neurons.


Subject(s)
Embryonic Stem Cells/transplantation , Hippocampus/physiology , Neural Stem Cells/transplantation , Neurons/transplantation , Action Potentials/physiology , Animals , Embryonic Stem Cells/cytology , Female , Hippocampus/cytology , Humans , Male , Neural Stem Cells/cytology , Neurogenesis/physiology , Neurons/cytology , Optogenetics , Rats , Rats, Sprague-Dawley
16.
PLoS One ; 6(8): e24027, 2011.
Article in English | MEDLINE | ID: mdl-21901155

ABSTRACT

Human embryonic stem cells (hESCs) can potentially differentiate into any cell type, including dopaminergic neurons to treat Parkinson's disease (PD), but hyperproliferation and tumor formation must be avoided. Accordingly, we use myocyte enhancer factor 2C (MEF2C) as a neurogenic and anti-apoptotic transcription factor to generate neurons from hESC-derived neural stem/progenitor cells (NPCs), thus avoiding hyperproliferation. Here, we report that forced expression of constitutively active MEF2C (MEF2CA) generates significantly greater numbers of neurons with dopaminergic properties in vitro. Conversely, RNAi knockdown of MEF2C in NPCs decreases neuronal differentiation and dendritic length. When we inject MEF2CA-programmed NPCs into 6-hydroxydopamine-lesioned parkinsonian rats in vivo, the transplanted cells survive well, differentiate into tyrosine hydroxylase-positive neurons, and improve behavioral deficits to a significantly greater degree than non-programmed cells. The enriched generation of dopaminergic neuronal lineages from hESCs by forced expression of MEF2CA in the proper context may prove valuable in cell-based therapy for CNS disorders such as PD.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , MADS Domain Proteins/metabolism , Myogenic Regulatory Factors/metabolism , Neurogenesis/physiology , Animals , Cell Differentiation/genetics , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Electrophysiology , Embryonic Stem Cells/transplantation , Humans , Immunohistochemistry , In Situ Nick-End Labeling , MADS Domain Proteins/genetics , MEF2 Transcription Factors , Myogenic Regulatory Factors/genetics , Neurogenesis/genetics , Oxidopamine , Parkinson Disease/therapy , Polymerase Chain Reaction , RNA Interference/physiology , Rats , Reverse Transcriptase Polymerase Chain Reaction
17.
Cell Stem Cell ; 9(2): 113-8, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21802386

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) have been generated by reprogramming a number of different somatic cell types using a variety of approaches. In addition, direct reprogramming of mature cells from one lineage to another has emerged recently as an alternative strategy for generating cell types of interest. Here we show that a combination of a microRNA (miR-124) and two transcription factors (MYT1L and BRN2) is sufficient to directly reprogram postnatal and adult human primary dermal fibroblasts (mesoderm) to functional neurons (ectoderm) under precisely defined conditions. These human induced neurons (hiNs) exhibit typical neuronal morphology and marker gene expression, fire action potentials, and produce functional synapses between each other. Our findings have major implications for cell-replacement strategies in neurodegenerative diseases, disease modeling, and neural developmental studies.


Subject(s)
Cell Culture Techniques/methods , Cellular Reprogramming/genetics , Culture Media/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Neurons/cytology , Neurons/metabolism , Adult , Cell Differentiation/drug effects , Cellular Reprogramming/drug effects , Dermis/cytology , Fibroblasts/drug effects , Humans , Infant, Newborn , Neurons/drug effects , Synapses/drug effects , Synapses/metabolism
18.
Proc Natl Acad Sci U S A ; 108(20): 8299-304, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21525408

ABSTRACT

Human embryonic stem cells (hESCs) hold enormous promise for regenerative medicine. Typically, hESC-based applications would require their in vitro differentiation into a desirable homogenous cell population. A major challenge of the current hESC differentiation paradigm is the inability to effectively capture and, in the long-term, stably expand primitive lineage-specific stem/precursor cells that retain broad differentiation potential and, more importantly, developmental stage-specific differentiation propensity. Here, we report synergistic inhibition of glycogen synthase kinase 3 (GSK3), transforming growth factor ß (TGF-ß), and Notch signaling pathways by small molecules can efficiently convert monolayer cultured hESCs into homogenous primitive neuroepithelium within 1 wk under chemically defined condition. These primitive neuroepithelia can stably self-renew in the presence of leukemia inhibitory factor, GSK3 inhibitor (CHIR99021), and TGF-ß receptor inhibitor (SB431542); retain high neurogenic potential and responsiveness to instructive neural patterning cues toward midbrain and hindbrain neuronal subtypes; and exhibit in vivo integration. Our work uniformly captures and maintains primitive neural stem cells from hESCs.


Subject(s)
Embryonic Stem Cells/cytology , Neural Stem Cells/cytology , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Embryonic Stem Cells/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Receptors, Notch/antagonists & inhibitors , Transforming Growth Factor beta/antagonists & inhibitors
19.
Proc Natl Acad Sci U S A ; 108(19): 7838-43, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21521790

ABSTRACT

The simple yet powerful technique of induced pluripotency may eventually supply a wide range of differentiated cells for cell therapy and drug development. However, making the appropriate cells via induced pluripotent stem cells (iPSCs) requires reprogramming of somatic cells and subsequent redifferentiation. Given how arduous and lengthy this process can be, we sought to determine whether it might be possible to convert somatic cells into lineage-specific stem/progenitor cells of another germ layer in one step, bypassing the intermediate pluripotent stage. Here we show that transient induction of the four reprogramming factors (Oct4, Sox2, Klf4, and c-Myc) can efficiently transdifferentiate fibroblasts into functional neural stem/progenitor cells (NPCs) with appropriate signaling inputs. Compared with induced neurons (or iN cells, which are directly converted from fibroblasts), transdifferentiated NPCs have the distinct advantage of being expandable in vitro and retaining the ability to give rise to multiple neuronal subtypes and glial cells. Our results provide a unique paradigm for iPSC-factor-based reprogramming by demonstrating that it can be readily modified to serve as a general platform for transdifferentiation.


Subject(s)
Cell Transdifferentiation , Fibroblasts/cytology , Fibroblasts/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Cell Lineage , Cell Proliferation , Culture Media , Cytological Techniques , Gene Expression , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , Mice , Models, Biological , Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/biosynthesis , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
20.
J Neurosci ; 30(34): 11501-5, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20739572

ABSTRACT

NMDA receptors are typically excited by a combination of glutamate and glycine. Here we describe excitatory responses in CNS myelin that are gated by a glycine agonist alone and mediated by NR1/NR3 "NMDA" receptor subunits. Response properties include activation by d-serine, inhibition by the glycine-site antagonist CNQX, and insensitivity to the glutamate-site antagonist d-APV. d-Serine responses were abrogated in NR3A-deficient mice. Our results suggest the presence of functional NR1/NR3 receptors in CNS myelin.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Glycine/physiology , Myelin Sheath/physiology , Protein Subunits/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Cell Line , Central Nervous System/physiology , Humans , Mice , Mice, Knockout , Protein Subunits/agonists , Protein Subunits/genetics , Rats , Rats, Long-Evans , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Proteins/agonists , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...