Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Stem Cell Res ; 79: 103455, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38896969

ABSTRACT

Calcium indicators are sensitive tools to image neural activity. However, their use in human induced pluripotent stem cells (iPSC)-derived neurons is limited by silencing of the transgene. We generated the iPSC line MSE2336A carrying heterozygous insertion in the safe-harbor locus AAVS1 of the ultrasensitive protein calcium sensor (GCaMP6) under the control of CAG promoter and UCOE to maintain robust transgene expression in differentiated cells. The iPSC exhibited normal cell morphology, expression of pluripotency markers, genome integrity, and the ability to differentiate into the three primary germ layers. This line provides a powerful model to study activity in human neurons.

2.
Nat Commun ; 15(1): 2058, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38448474

ABSTRACT

Genetic and experimental evidence suggests that Alzheimer's disease (AD) risk alleles and genes may influence disease susceptibility by altering the transcriptional and cellular responses of macrophages, including microglia, to damage of lipid-rich tissues like the brain. Recently, sc/nRNA sequencing studies identified similar transcriptional activation states in subpopulations of macrophages in aging and degenerating brains and in other diseased lipid-rich tissues. We collectively refer to these subpopulations of microglia and peripheral macrophages as DLAMs. Using macrophage sc/nRNA-seq data from healthy and diseased human and mouse lipid-rich tissues, we reconstructed gene regulatory networks and identified 11 strong candidate transcriptional regulators of the DLAM response across species. Loss or reduction of two of these transcription factors, BHLHE40/41, in iPSC-derived microglia and human THP-1 macrophages as well as loss of Bhlhe40/41 in mouse microglia, resulted in increased expression of DLAM genes involved in cholesterol clearance and lysosomal processing, increased cholesterol efflux and storage, and increased lysosomal mass and degradative capacity. These findings provide targets for therapeutic modulation of macrophage/microglial function in AD and other disorders affecting lipid-rich tissues.


Subject(s)
Alzheimer Disease , Microglia , Humans , Animals , Mice , Alzheimer Disease/genetics , Macrophages , Cholesterol , Lipids , Homeodomain Proteins , Basic Helix-Loop-Helix Transcription Factors
3.
J Virol ; 97(12): e0159523, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38032195

ABSTRACT

IMPORTANCE: Our mouse model is a powerful tool for investigating the genetic mechanisms governing central nervous system (CNS) human immunodeficiency virus type-1 (HIV-1) infection and latency in the CNS at a single-cell level. A major advantage of our model is that it uses induced pluripotent stem cell-derived microglia, which enables human genetics, including gene function and therapeutic gene manipulation, to be explored in vivo, which is more challenging to study with current hematopoietic stem cell-based models for neuroHIV. Our transgenic tracing of xenografted human cells will provide a quantitative medium to develop new molecular and epigenetic strategies for reducing the HIV-1 latent reservoir and to test the impact of therapeutic inflammation-targeting drug interventions on CNS HIV-1 latency.


Subject(s)
HIV Infections , HIV-1 , Induced Pluripotent Stem Cells , Microglia , Animals , Humans , Mice , Central Nervous System , HIV Infections/metabolism , HIV Infections/pathology , HIV-1/physiology , Microglia/virology , Virus Latency , Heterografts
4.
bioRxiv ; 2023 Apr 27.
Article in English | MEDLINE | ID: mdl-37162838

ABSTRACT

The central nervous system (CNS) is a major human immunodeficiency virus type 1 reservoir. Microglia are the primary target cell of HIV-1 infection in the CNS. Current models have not allowed the precise molecular pathways of acute and chronic CNS microglial infection to be tested with in vivo genetic methods. Here, we describe a novel humanized mouse model utilizing human-induced pluripotent stem cell-derived microglia to xenograft into murine hosts. These mice are additionally engrafted with human peripheral blood mononuclear cells that served as a medium to establish a peripheral infection that then spread to the CNS microglia xenograft, modeling a trans-blood-brain barrier route of acute CNS HIV-1 infection with human target cells. The approach is compatible with iPSC genetic engineering, including inserting targeted transgenic reporter cassettes to track the xenografted human cells, enabling the testing of novel treatment and viral tracking strategies in a comparatively simple and cost-effective way vivo model for neuroHIV.

5.
bioRxiv ; 2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36824752

ABSTRACT

Background: Genetic and experimental evidence strongly implicates myeloid cells in the etiology of AD and suggests that AD-associated alleles and genes may modulate disease risk by altering the transcriptional and cellular responses of macrophages (like microglia) to damage of lipid-rich tissues (like the brain). Specifically, recent single-cell/nucleus RNA sequencing (sc/nRNA-seq) studies identified a transcriptionally distinct state of subsets of macrophages in aging or degenerating brains (usually referred to as disease-associated microglia or DAM) and in other diseased lipid-rich tissues (e.g., obese adipose tissue, fatty liver, and atherosclerotic plaques). We collectively refer to these subpopulations as lipid-associated macrophages or LAMs. Importantly, this particular activation state is characterized by increased expression of genes involved in the phagocytic clearance of lipid-rich cellular debris (efferocytosis), including several AD risk genes. Methods: We used sc/nRNA-seq data from human and mouse microglia from healthy and diseased brains and macrophages from other lipid-rich tissues to reconstruct gene regulatory networks and identify transcriptional regulators whose regulons are enriched for LAM response genes (LAM TFs) across species. We then used gene knock-down/knock-out strategies to validate some of these LAM TFs in human THP-1 macrophages and iPSC-derived microglia in vitro, as well as mouse microglia in vivo. Results: We nominate 11 strong candidate LAM TFs shared across human and mouse networks (BHLHE41, HIF1A, ID2, JUNB, MAF, MAFB, MEF2A, MEF2C, NACA, POU2F2 and SPI1). We also demonstrate a strong enrichment of AD risk alleles in the cistrome of BHLHE41 (and its close homolog BHLHE40), thus implicating its regulon in the modulation of disease susceptibility. Loss or reduction of BHLHE40/41 expression in human THP-1 macrophages and iPSC-derived microglia, as well as loss of Bhlhe40/41 in mouse microglia led to increased expression of LAM response genes, specifically those involved in cholesterol clearance and lysosomal processing, with a concomitant increase in cholesterol efflux and storage, as well as lysosomal mass and degradative capacity. Conclusions: Taken together, this study nominates transcriptional regulators of the LAM response, experimentally validates BHLHE40/41 in human and mouse macrophages/microglia, and provides novel targets for therapeutic modulation of macrophage/microglia function in AD and other disorders of lipid-rich tissues.

6.
Mol Cell ; 82(24): 4627-4646.e14, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36417913

ABSTRACT

Cell lineage specification is accomplished by a concerted action of chromatin remodeling and tissue-specific transcription factors. However, the mechanisms that induce and maintain appropriate lineage-specific gene expression remain elusive. Here, we used an unbiased proteomics approach to characterize chromatin regulators that mediate the induction of neuronal cell fate. We found that Tip60 acetyltransferase is essential to establish neuronal cell identity partly via acetylation of the histone variant H2A.Z. Despite its tight correlation with gene expression and active chromatin, loss of H2A.Z acetylation had little effect on chromatin accessibility or transcription. Instead, loss of Tip60 and acetyl-H2A.Z interfered with H3K4me3 deposition and activation of a unique subset of silent, lineage-restricted genes characterized by a bivalent chromatin configuration at their promoters. Altogether, our results illuminate the mechanisms underlying bivalent chromatin activation and reveal that H2A.Z acetylation regulates neuronal fate specification by establishing epigenetic competence for bivalent gene activation and cell lineage transition.


Subject(s)
Chromatin , Histones , Histones/genetics , Histones/metabolism , Acetylation , Transcriptional Activation , Chromatin/genetics , Protein Processing, Post-Translational , Nucleosomes
7.
Cell Rep ; 35(2): 108991, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852833

ABSTRACT

Transcriptional silencing of the FMR1 gene in fragile X syndrome (FXS) leads to the loss of the RNA-binding protein FMRP. In addition to regulating mRNA translation and protein synthesis, emerging evidence suggests that FMRP acts to coordinate proliferation and differentiation during early neural development. However, whether loss of FMRP-mediated translational control is related to impaired cell fate specification in the developing human brain remains unknown. Here, we use human patient induced pluripotent stem cell (iPSC)-derived neural progenitor cells and organoids to model neurogenesis in FXS. We developed a high-throughput, in vitro assay that allows for the simultaneous quantification of protein synthesis and proliferation within defined neural subpopulations. We demonstrate that abnormal protein synthesis in FXS is coupled to altered cellular decisions to favor proliferative over neurogenic cell fates during early development. Furthermore, pharmacologic inhibition of elevated phosphoinositide 3-kinase (PI3K) signaling corrects both excess protein synthesis and cell proliferation in a subset of patient neural cells.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Biological Assay , Cell Differentiation , Cell Lineage/genetics , Cell Proliferation , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Gene Expression Regulation, Developmental , Humans , Imidazoles/pharmacology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Models, Biological , Morpholines/pharmacology , Neural Stem Cells/drug effects , Neural Stem Cells/pathology , Neurogenesis/genetics , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Piperazines/pharmacology , Primary Cell Culture , Protein Biosynthesis , Pyrimidinones/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
8.
Mol Cell ; 78(3): 459-476.e13, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32240602

ABSTRACT

The cyclin-dependent kinase 1 (Cdk1) drives cell division. To uncover additional functions of Cdk1, we generated knockin mice expressing an analog-sensitive version of Cdk1 in place of wild-type Cdk1. In our study, we focused on embryonic stem cells (ESCs), because this cell type displays particularly high Cdk1 activity. We found that in ESCs, a large fraction of Cdk1 substrates is localized on chromatin. Cdk1 phosphorylates many proteins involved in epigenetic regulation, including writers and erasers of all major histone marks. Consistent with these findings, inhibition of Cdk1 altered histone-modification status of ESCs. High levels of Cdk1 in ESCs phosphorylate and partially inactivate Dot1l, the H3K79 methyltransferase responsible for placing activating marks on gene bodies. Decrease of Cdk1 activity during ESC differentiation de-represses Dot1l, thereby allowing coordinated expression of differentiation genes. These analyses indicate that Cdk1 functions to maintain the epigenetic identity of ESCs.


Subject(s)
CDC2 Protein Kinase/metabolism , Embryonic Stem Cells/physiology , Epigenesis, Genetic , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Animals , CDC2 Protein Kinase/genetics , Cell Differentiation , Cells, Cultured , Chromatin Immunoprecipitation/methods , Female , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , MCF-7 Cells , Male , Mice , Mice, Knockout , Phosphorylation , Saccharomyces cerevisiae Proteins/metabolism
9.
Cell Stem Cell ; 25(4): 531-541.e6, 2019 Oct 03.
Article in English | MEDLINE | ID: mdl-31585094

ABSTRACT

Pelizaeus-Merzbacher disease (PMD) is an X-linked leukodystrophy caused by mutations in Proteolipid Protein 1 (PLP1), encoding a major myelin protein, resulting in profound developmental delay and early lethality. Previous work showed involvement of unfolded protein response (UPR) and endoplasmic reticulum (ER) stress pathways, but poor PLP1 genotype-phenotype associations suggest additional pathogenetic mechanisms. Using induced pluripotent stem cell (iPSC) and gene-correction, we show that patient-derived oligodendrocytes can develop to the pre-myelinating stage, but subsequently undergo cell death. Mutant oligodendrocytes demonstrated key hallmarks of ferroptosis including lipid peroxidation, abnormal iron metabolism, and hypersensitivity to free iron. Iron chelation rescued mutant oligodendrocyte apoptosis, survival, and differentiationin vitro, and post-transplantation in vivo. Finally, systemic treatment of Plp1 mutant Jimpy mice with deferiprone, a small molecule iron chelator, reduced oligodendrocyte apoptosis and enabled myelin formation. Thus, oligodendrocyte iron-induced cell death and myelination is rescued by iron chelation in PMD pre-clinical models.


Subject(s)
Deferiprone/therapeutic use , Induced Pluripotent Stem Cells/physiology , Iron Chelating Agents/therapeutic use , Iron/metabolism , Myelin Proteolipid Protein/metabolism , Oligodendroglia/physiology , Pelizaeus-Merzbacher Disease/therapy , Animals , Cell Differentiation , Cells, Cultured , Ferroptosis , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/transplantation , Lipid Peroxidation , Mice , Mice, Mutant Strains , Mutation/genetics , Myelin Proteolipid Protein/genetics , Oligodendroglia/drug effects , Oligodendroglia/transplantation , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/pathology , Stem Cell Transplantation , Targeted Gene Repair
10.
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
11.
Sci Transl Med ; 10(452)2018 08 01.
Article in English | MEDLINE | ID: mdl-30068571

ABSTRACT

Fragile X syndrome (FXS) is an X chromosome-linked disease leading to severe intellectual disabilities. FXS is caused by inactivation of the fragile X mental retardation 1 (FMR1) gene, but how FMR1 inactivation induces FXS remains unclear. Using human neurons generated from control and FXS patient-derived induced pluripotent stem (iPS) cells or from embryonic stem cells carrying conditional FMR1 mutations, we show here that loss of FMR1 function specifically abolished homeostatic synaptic plasticity without affecting basal synaptic transmission. We demonstrated that, in human neurons, homeostatic plasticity induced by synaptic silencing was mediated by retinoic acid, which regulated both excitatory and inhibitory synaptic strength. FMR1 inactivation impaired homeostatic plasticity by blocking retinoic acid-mediated regulation of synaptic strength. Repairing the genetic mutation in the FMR1 gene in an FXS patient cell line restored fragile X mental retardation protein (FMRP) expression and fully rescued synaptic retinoic acid signaling. Thus, our study reveals a robust functional impairment caused by FMR1 mutations that might contribute to neuronal dysfunction in FXS. In addition, our results suggest that FXS patient iPS cell-derived neurons might be useful for studying the mechanisms mediating functional abnormalities in FXS.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Homeostasis , Mutation/genetics , Neuronal Plasticity , Neurons/metabolism , Signal Transduction , Synapses/metabolism , Tretinoin/metabolism , Alleles , Animals , Base Sequence , Cell Differentiation/drug effects , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Excitatory Postsynaptic Potentials/drug effects , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Homeostasis/drug effects , Humans , Mice , Neuronal Plasticity/drug effects , Neurons/drug effects , Signal Transduction/drug effects , Synapses/drug effects , Tretinoin/pharmacology , Trinucleotide Repeats/genetics , Up-Regulation/drug effects
12.
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
13.
Cell Stem Cell ; 16(1): 39-50, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25467916

ABSTRACT

Mutations in the retinoblastoma tumor suppressor gene Rb are involved in many forms of human cancer. In this study, we investigated the early consequences of inactivating Rb in the context of cellular reprogramming. We found that Rb inactivation promotes the reprogramming of differentiated cells to a pluripotent state. Unexpectedly, this effect is cell cycle independent, and instead reflects direct binding of Rb to pluripotency genes, including Sox2 and Oct4, which leads to a repressed chromatin state. More broadly, this regulation of pluripotency networks and Sox2 in particular is critical for the initiation of tumors upon loss of Rb in mice. These studies therefore identify Rb as a global transcriptional repressor of pluripotency networks, providing a molecular basis for previous reports about its involvement in cell fate pliability, and implicate misregulation of pluripotency factors such as Sox2 in tumorigenesis related to loss of Rb function.


Subject(s)
Carcinogenesis/pathology , Cellular Reprogramming , Induced Pluripotent Stem Cells/metabolism , Retinoblastoma Protein/metabolism , Animals , Carcinogenesis/metabolism , Cell Cycle , Chromatin/metabolism , Fibroblasts/metabolism , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Protein Binding , Repressor Proteins/metabolism , Retinoblastoma Protein/deficiency , SOXB1 Transcription Factors/genetics
14.
Methods Mol Biol ; 1150: 237-46, 2014.
Article in English | MEDLINE | ID: mdl-24744003

ABSTRACT

Nuclear reprogramming by defined transcription factors became of broad interest in 2006 with the work of Takahashi and Yamanaka (Cell 126:663-676, 2006), but the first example of cell fate reshaping via ectopic expression of transcription factor was provided back in 1987 when Davis and colleagues induced features of a muscle cell in fibroblast using the muscle transcription factor MyoD (Davis et al., Cell 51:987-1000, 1987). In 2010 our laboratory described how forced expression of the three neuronal transcription factors Ascl1, Brn2, and Myt1l rapidly converts mouse fibroblasts into neuronal cells that exhibit biochemical and electrophysiological properties of neurons. We named these cells induced neuronal cells (iN cells) (Vierbuchen et al., Nature 463:1035-1041, 2010; Vierbuchen and Wernig, Nat Biotechnol 29:892-907, 2011). Interestingly, iN cells can also be derived from defined endodermal cells such as primary hepatocytes, suggesting the existence of a more general reprogramming paradigm (Marro et al., Cell Stem Cell 9:374-382, 2011). In this chapter we describe the detailed methods used to attain the direct conversion.


Subject(s)
Cell Transdifferentiation , Cellular Reprogramming , Fibroblasts/cytology , Hepatocytes/cytology , Neurons/cytology , Animals , Cell Separation , Fibroblasts/metabolism , Gene Silencing , Hepatocytes/metabolism , Lentivirus/genetics , Mice , Neuroglia/cytology , Transduction, Genetic
15.
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
16.
Proc Natl Acad Sci U S A ; 110(41): 16622-7, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24046374

ABSTRACT

Recent studies suggest that induced neuronal (iN) cells that are directly transdifferentiated from nonneuronal cells provide a powerful opportunity to examine neuropsychiatric diseases. However, the validity of using this approach to examine disease-specific changes has not been demonstrated. Here, we analyze the phenotypes of iN cells that were derived from murine embryonic fibroblasts cultured from littermate wild-type and mutant mice carrying the autism-associated R704C substitution in neuroligin-3. We show that neuroligin-3 R704C-mutant iN cells exhibit a large and selective decrease in AMPA-type glutamate receptor-mediated synaptic transmission without changes in NMDA-type glutamate receptor- or in GABAA receptor-mediated synaptic transmission. Thus, the synaptic phenotype observed in R704C-mutant iN cells replicates the previously observed phenotype of R704C-mutant neurons. Our data show that the effect of the R704C mutation is applicable even to neurons transdifferentiated from fibroblasts and constitute a proof-of-concept demonstration that iN cells can be used for cellular disease modeling.


Subject(s)
Autistic Disorder/physiopathology , Cell Adhesion Molecules, Neuronal/genetics , Disease Models, Animal , Fibroblasts/cytology , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Neurons/cytology , Phenotype , Amino Acid Substitution/genetics , Animals , Autistic Disorder/genetics , Cell Transdifferentiation/physiology , Cells, Cultured , Fibroblasts/physiology , Flow Cytometry , Fluorescent Antibody Technique , Mice , Patch-Clamp Techniques , Real-Time Polymerase Chain Reaction , Receptors, AMPA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Synaptic Transmission/genetics , Synaptic Transmission/physiology
17.
Neuron ; 78(5): 785-98, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23764284

ABSTRACT

Available methods for differentiating human embryonic stem cells (ESCs) and induced pluripotent cells (iPSCs) into neurons are often cumbersome, slow, and variable. Alternatively, human fibroblasts can be directly converted into induced neuronal (iN) cells. However, with present techniques conversion is inefficient, synapse formation is limited, and only small amounts of neurons can be generated. Here, we show that human ESCs and iPSCs can be converted into functional iN cells with nearly 100% yield and purity in less than 2 weeks by forced expression of a single transcription factor. The resulting ES-iN or iPS-iN cells exhibit quantitatively reproducible properties independent of the cell line of origin, form mature pre- and postsynaptic specializations, and integrate into existing synaptic networks when transplanted into mouse brain. As illustrated by selected examples, our approach enables large-scale studies of human neurons for questions such as analyses of human diseases, examination of human-specific genes, and drug screening.


Subject(s)
Biophysical Phenomena/physiology , Gene Expression Regulation/physiology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Pluripotent Stem Cells/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Biophysical Phenomena/genetics , Biophysics , Brain/cytology , Calcium/metabolism , Cells, Cultured , Collagen Type VII/genetics , Electric Stimulation , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/pathology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Fibroblasts , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Humans , Mice , Microscopy, Confocal , Munc18 Proteins/genetics , Munc18 Proteins/metabolism , Mutation/genetics , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , RNA, Small Interfering/physiology , Rhodopsin/genetics , Sodium Channel Blockers/pharmacology , Synapses/physiology , Tetrodotoxin/pharmacology , Time Factors , Transfection
18.
Nat Biotechnol ; 31(5): 434-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23584610

ABSTRACT

Transplantation of oligodendrocyte precursor cells (OPCs) is a promising potential therapeutic strategy for diseases affecting myelin. However, the derivation of engraftable OPCs from human pluripotent stem cells has proven difficult and primary OPCs are not readily available. Here we report the generation of induced OPCs (iOPCs) by direct lineage conversion. Forced expression of the three transcription factors Sox10, Olig2 and Zfp536 was sufficient to reprogram mouse and rat fibroblasts into iOPCs with morphologies and gene expression signatures resembling primary OPCs. More importantly, iOPCs gave rise to mature oligodendrocytes that could ensheath multiple host axons when co-cultured with primary dorsal root ganglion cells and formed myelin after transplantation into shiverer mice. We propose direct lineage reprogramming as a viable alternative approach for the generation of OPCs for use in disease modeling and regenerative medicine.


Subject(s)
Fibroblasts/cytology , Myelin Sheath/metabolism , Oligodendroglia/cytology , Oligodendroglia/physiology , Stem Cells/cytology , Stem Cells/physiology , Transcription Factors/genetics , Animals , Cell Differentiation , Fibroblasts/physiology , Genetic Enhancement/methods , Mice , Stem Cell Transplantation/methods
19.
Blood Cells Mol Dis ; 50(1): 25-30, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22921471

ABSTRACT

Ferroportin (FPN), the sole characterized iron exporter, is mainly controlled by the peptide hormone hepcidin in response to iron, erythroid factors, hypoxia, and inflammation. In addition, intracellular iron level controls FPN translation by modulating the binding of Iron Responsive Proteins at the 5'UTR of FPN mRNA. Recently, hypoxia inducible factor (HIF)2α has been shown to regulate FPN expression in intestinal cells. Here we show that, during experimentally-induced acute anemia in mice, FPN is regulated at transcriptional level in a cell-specific manner. FPN mRNA level increases in duodenum and spleen macrophages, whereas it does not change in liver and is strongly down-regulated in erythroid precursors. These results were confirmed in Caco2, Raw264.7 and K562 cells treated with a hypoxic stimulus. Moreover, we found a differential expression of HIF1α and HIF2α in cells and tissues that might account for the specificity of FPN regulation. Thus, hypoxia, by directly controlling hepcidin and its target FPN, orchestrates a complex regulatory network aimed at ensuring rapid iron recovery from the periphery and efficient iron utilization in the erythroid compartment.


Subject(s)
Anemia/genetics , Antimicrobial Cationic Peptides/genetics , Cation Transport Proteins/genetics , Acute Disease , Anemia/metabolism , Animals , Antimicrobial Cationic Peptides/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cation Transport Proteins/metabolism , Cell Hypoxia , Cell Line, Tumor , Duodenum/metabolism , Gene Expression Regulation , Hepcidins , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Iron/metabolism , Liver/metabolism , Macrophages/metabolism , Male , Mice , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spleen/metabolism , Transcription, Genetic
20.
J Clin Invest ; 122(12): 4569-79, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23187127

ABSTRACT

Feline leukemia virus subgroup C receptor 1 (FLVCR1) is a cell membrane heme exporter that maintains the balance between heme levels and globin synthesis in erythroid precursors. It was previously shown that Flvcr1-null mice died in utero due to a failure of erythropoiesis. Here, we identify Flvcr1b, a mitochondrial Flvcr1 isoform that promotes heme efflux into the cytoplasm. Flvcr1b overexpression promoted heme synthesis and in vitro erythroid differentiation, whereas silencing of Flvcr1b caused mitochondrial heme accumulation and termination of erythroid differentiation. Furthermore, mice lacking the plasma membrane isoform (Flvcr1a) but expressing Flvcr1b had normal erythropoiesis, but exhibited hemorrhages, edema, and skeletal abnormalities. Thus, FLVCR1b regulates erythropoiesis by controlling mitochondrial heme efflux, whereas FLVCR1a expression is required to prevent hemorrhages and edema. The aberrant expression of Flvcr1 isoforms may play a role in the pathogenesis of disorders characterized by an imbalance between heme and globin synthesis.


Subject(s)
Cell Differentiation , Erythrocytes/physiology , Membrane Transport Proteins/physiology , Mitochondrial Proteins/physiology , Receptors, Virus/physiology , Abnormalities, Multiple/genetics , Animals , Bone Diseases, Developmental/genetics , Brain/metabolism , Edema/genetics , Erythrocytes/metabolism , Erythropoiesis , Exons , Gene Expression , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Heme/metabolism , Hemorrhage/genetics , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Organ Specificity , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Receptors, Virus/genetics , Receptors, Virus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...