Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Mol Aspects Med ; 91: 101148, 2023 06.
Article in English | MEDLINE | ID: mdl-36257857

ABSTRACT

Advances in genome sequencing have greatly facilitated the identification of genomic variants underlying rare neurodevelopmental and neurodegenerative disorders. Understanding the fundamental causes of rare monogenic disorders has made gene therapy a possible treatment approach for these conditions. RNA interference (RNAi) technologies such as small interfering RNA (siRNA), microRNA (miRNA), and short hairpin RNA (shRNA), and other oligonucleotide-based modalities such as antisense oligonucleotides (ASOs) are being developed as potential therapeutic approaches for manipulating expression of the genes that cause a variety of neurological diseases. Here, we offer a brief review of the mechanism of action of these RNAi approaches; provide deeper discussion of the advantages, challenges, and specific considerations related to the development of RNAi therapeutics for neurological disease; and highlight examples of rare neurological diseases for which RNAi therapeutics hold great promise.


Subject(s)
MicroRNAs , Humans , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , Genetic Therapy
2.
Cell Rep Med ; 2(8): 100377, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34467252

ABSTRACT

New research from Pandya and colleagues1 identifies PEG10 as a UBE3A-regulated protein that may underlie pathophysiology in Angelman syndrome neurons. PEG10 is a secreted protein, and this work suggests that it may be a potential biomarker for Angelman syndrome therapeutics under development.


Subject(s)
Angelman Syndrome , Ubiquitin-Protein Ligases , Angelman Syndrome/genetics , Animals , Biomarkers , Disease Models, Animal , Neurons , Ubiquitin-Protein Ligases/genetics
3.
Hum Mol Genet ; 29(19): 3285-3295, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32977341

ABSTRACT

Prader-Willi syndrome (PWS) is characterized by neonatal hypotonia, developmental delay and hyperphagia/obesity. This disorder is caused by the absence of paternally expressed gene products from chromosome 15q11-q13. We previously demonstrated that knocking out ZNF274, a Kruppel-associated box-A-domain zinc finger protein capable of recruiting epigenetic machinery to deposit the H3K9me3 repressive histone modification, can activate expression from the normally silent maternal allele of SNORD116 in neurons derived from PWS induced pluripotent stem cells (iPSCs). However, ZNF274 has many other targets in the genome in addition to SNORD116. Depleting ZNF274 will surely affect the expression of other important genes and disrupt other pathways. Here, we used CRISPR/Cas9 to delete ZNF274 binding sites at the SNORD116 locus to determine whether activation of the maternal copy of SNORD116 could be achieved without altering ZNF274 protein levels. We obtained similar activation of gene expression from the normally silenced maternal allele in neurons derived from PWS iPSCs, compared with ZNF274 knockout, demonstrating that ZNF274 is directly involved in the repression of SNORD116. These results suggest that interfering with ZNF274 binding at the maternal SNORD116 locus is a potential therapeutic strategy for PWS.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Kruppel-Like Transcription Factors/metabolism , Neurons/pathology , Prader-Willi Syndrome/pathology , RNA, Messenger, Stored/genetics , RNA, Small Nucleolar/genetics , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Transcription Factors/genetics , Neurons/metabolism , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/metabolism
4.
Hum Mol Genet ; 29(18): 3021-3031, 2020 11 04.
Article in English | MEDLINE | ID: mdl-32833011

ABSTRACT

Loss of UBE3A expression, a gene regulated by genomic imprinting, causes Angelman syndrome (AS), a rare neurodevelopmental disorder. The UBE3A gene encodes an E3 ubiquitin ligase with three known protein isoforms in humans. Studies in mouse suggest that the human isoforms may have differences in localization and neuronal function. A recent case study reported mild AS phenotypes in individuals lacking one specific isoform. Here we have used CRISPR/Cas9 to generate isogenic human embryonic stem cells (hESCs) that lack the individual protein isoforms. We demonstrate that isoform 1 accounts for the majority of UBE3A protein in hESCs and neurons. We also show that UBE3A predominantly localizes to the cytoplasm in both wild type and isoform-null cells. Finally, we show that neurons lacking isoform 1 display a less severe electrophysiological AS phenotype.


Subject(s)
Angelman Syndrome/genetics , Genetic Predisposition to Disease , Ubiquitin-Protein Ligases/genetics , Angelman Syndrome/pathology , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Electrophysiological Phenomena/genetics , Genomic Imprinting/genetics , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/pathology , Humans , Mice , Neurons/metabolism , Neurons/pathology , Protein Isoforms/genetics
5.
Adv Neurobiol ; 25: 55-77, 2020.
Article in English | MEDLINE | ID: mdl-32578144

ABSTRACT

The chromosome 15q11-q13 region of the human genome is regulated by genomic imprinting, an epigenetic phenomenon in which genes are expressed exclusively from one parental allele. Several genes within the 15q11-q13 region are expressed exclusively from the paternally inherited chromosome 15. At least one gene UBE3A, shows exclusive expression of the maternal allele, but this allele-specific expression is restricted to neurons. The appropriate regulation of imprinted gene expression across chromosome 15q11-q13 has important implications for human disease. Three different neurodevelopmental disorders result from aberrant expression of imprinted genes in this region: Prader-Willi syndrome (PWS), Angelman syndrome (AS), and 15q duplication syndrome.


Subject(s)
Angelman Syndrome , Prader-Willi Syndrome , Angelman Syndrome/genetics , Chromosomes , Genomic Imprinting/genetics , Humans , Prader-Willi Syndrome/genetics
6.
Proc Natl Acad Sci U S A ; 116(6): 2181-2186, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30674673

ABSTRACT

Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by the loss of function from the maternal allele of UBE3A, a gene encoding an E3 ubiquitin ligase. UBE3A is only expressed from the maternally inherited allele in mature human neurons due to tissue-specific genomic imprinting. Imprinted expression of UBE3A is restricted to neurons by expression of UBE3A antisense transcript (UBE3A-ATS) from the paternally inherited allele, which silences the paternal allele of UBE3A in cis However, the mechanism restricting UBE3A-ATS expression and UBE3A imprinting to neurons is not understood. We used CRISPR/Cas9-mediated genome editing to functionally define a bipartite boundary element critical for neuron-specific expression of UBE3A-ATS in humans. Removal of this element led to up-regulation of UBE3A-ATS without repressing paternal UBE3A However, increasing expression of UBE3A-ATS in the absence of the boundary element resulted in full repression of paternal UBE3A, demonstrating that UBE3A imprinting requires both the loss of function from the boundary element as well as the up-regulation of UBE3A-ATS These results suggest that manipulation of the competition between UBE3A-ATS and UBE3A may provide a potential therapeutic approach for AS.


Subject(s)
Chromatin/genetics , Genomic Imprinting , Neurons/metabolism , Ubiquitin-Protein Ligases/genetics , Angelman Syndrome/genetics , Binding Sites , Chromatin/metabolism , Epistasis, Genetic , Exons , Gene Expression , Gene Expression Regulation , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Protein Binding , RNA, Antisense , RNA, Long Noncoding , Sequence Deletion
7.
Nat Commun ; 8: 15038, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28436452

ABSTRACT

Angelman syndrome (AS) is a neurogenetic disorder caused by deletion of the maternally inherited UBE3A allele and is characterized by developmental delay, intellectual disability, ataxia, seizures and a happy affect. Here, we explored the underlying pathophysiology using induced pluripotent stem cell-derived neurons from AS patients and unaffected controls. AS-derived neurons showed impaired maturation of resting membrane potential and action potential firing, decreased synaptic activity and reduced synaptic plasticity. These patient-specific differences were mimicked by knocking out UBE3A using CRISPR/Cas9 or by knocking down UBE3A using antisense oligonucleotides. Importantly, these phenotypes could be rescued by pharmacologically unsilencing paternal UBE3A expression. Moreover, selective effects of UBE3A disruption at late stages of in vitro development suggest that changes in action potential firing and synaptic activity may be secondary to altered resting membrane potential. Our findings provide a cellular phenotype for investigating pathogenic mechanisms underlying AS and identifying novel therapeutic strategies.


Subject(s)
Action Potentials/physiology , Angelman Syndrome/pathology , Induced Pluripotent Stem Cells/physiology , Neurons/physiology , Action Potentials/genetics , Angelman Syndrome/genetics , Angelman Syndrome/metabolism , Cell Differentiation , Cells, Cultured , Female , Gene Knockout Techniques , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Membrane Potentials/genetics , Membrane Potentials/physiology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Neurons/metabolism , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
8.
Methods Mol Biol ; 1353: 45-64, 2016.
Article in English | MEDLINE | ID: mdl-25520291

ABSTRACT

Induced pluripotent stem cell (iPSC) technology has allowed for the invaluable modeling of many genetic disorders including disorders associated with genomic imprinting. Genomic imprinting involves differential DNA and histone methylation and results in allele-specific gene expression. Most of the epigenetic marks in somatic cells are erased and reestablished during the process of reprogramming into iPSCs. Therefore, in generating models of disorders associated with genomic imprinting, it is important to verify that the imprinting status and allele-specific gene expression patterns of the parental somatic cells are maintained in their derivative iPSCs. Here, we describe three techniques: DNA methylation analysis, allele-specific PCR, and RNA FISH, which we use to analyze genomic imprinting in iPSC models of neurogenetic disorders involving copy number variations of the chromosome 15q11-q13 region.


Subject(s)
Epigenesis, Genetic , Genomic Imprinting , Induced Pluripotent Stem Cells/metabolism , Models, Genetic , Prader-Willi Syndrome/genetics , Alleles , Animals , Cell Differentiation , Cells, Cultured , DNA Copy Number Variations , DNA Methylation , DNA Primers/chemical synthesis , DNA Primers/metabolism , Feeder Cells/cytology , Fibroblasts/cytology , Humans , In Situ Hybridization, Fluorescence/methods , Induced Pluripotent Stem Cells/pathology , Mice , Polymerase Chain Reaction/methods , Prader-Willi Syndrome/diagnosis , Prader-Willi Syndrome/pathology , RNA/genetics , RNA/metabolism , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
9.
Stem Cells Dev ; 24(8): 1022-33, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25472739

ABSTRACT

Embryonic stem cells (ESCs) undergoing neural differentiation form radial arrays of neural stem cells, termed neural rosettes. These structures manifest many of the properties associated with embryonic and adult neurogenesis, including cell polarization, interkinetic nuclear migration (INM), and a gradient of neuronal differentiation. We now identify novel rosette structural features that serve to localize key regulators of neurogenesis. Cells within neural rosettes have specialized basal as well as apical surfaces, based on localization of the extracellular matrix receptor ß1 integrin. Apical processes of cells in mature rosettes terminate at the lumen, where adherens junctions are apparent. Primary cilia are randomly distributed in immature rosettes and tightly associated with the neural stem cell's apical domain as rosettes mature. Components of two signaling pathways known to regulate neurogenesis in vivo and in rosettes, Hedgehog and Notch, are apically localized, with the Hedgehog effector Smoothened (Smo) associated with primary cilia and the Notch pathway γ-secretase subunit Presenilin 2 associated with the adherens junction. Increased neuron production upon treatment with the Notch inhibitor DAPT suggests a major role for Notch signaling in maintaining the neural stem cell state, as previously described. A less robust outcome was observed with manipulation of Hedgehog levels, though consistent with a role in neural stem cell survival or proliferation. Inhibition of both pathways resulted in an additive effect. These data support a model by which cells extending a process to the rosette lumen maintain neural stem cell identity whereas release from this association, either through asymmetric cell division or apical abscission, promotes neuronal differentiation.


Subject(s)
Cell Polarity , Embryonic Stem Cells/cytology , Neurogenesis , Cells, Cultured , Embryonic Stem Cells/metabolism , Hedgehog Proteins/metabolism , Humans , Integrin beta1/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Presenilin-2/metabolism , Receptors, Notch/metabolism
10.
Hum Mol Genet ; 23(17): 4674-85, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24760766

ABSTRACT

Prader-Willi syndrome (PWS), a disorder of genomic imprinting, is characterized by neonatal hypotonia, hypogonadism, small hands and feet, hyperphagia and obesity in adulthood. PWS results from the loss of paternal copies of the cluster of SNORD116 C/D box snoRNAs and their host transcript, 116HG, on human chromosome 15q11-q13. We have investigated the mechanism of repression of the maternal SNORD116 cluster and 116HG. Here, we report that the zinc-finger protein ZNF274, in association with the histone H3 lysine 9 (H3K9) methyltransferase SETDB1, is part of a complex that binds to the silent maternal but not the active paternal alleles. Knockdown of SETDB1 in PWS-specific induced pluripotent cells (iPSCs) causes a decrease in the accumulation of H3K9 trimethylation (H3K9me3) at 116HG and corresponding accumulation of the active chromatin mark histone H3 lysine 4 dimethylation (H3K4me2). We also show that upon knockdown of SETDB1 in PWS-specific iPSCs, expression of maternally silenced 116HG RNA is partially restored. SETDB1 knockdown in PWS iPSCs also disrupts DNA methylation at the PWS-IC where a decrease in 5-methylcytosine is observed in association with a concomitant increase in 5-hydroxymethylcytosine. This observation suggests that the ZNF274/SETDB1 complex bound to the SNORD116 cluster may protect the PWS-IC from DNA demethylation during early development. Our findings reveal novel epigenetic mechanisms that function to repress the maternal 15q11-q13 region.


Subject(s)
Gene Knockdown Techniques , Induced Pluripotent Stem Cells/metabolism , Multigene Family , Prader-Willi Syndrome/genetics , Protein Methyltransferases/genetics , RNA, Small Nucleolar/genetics , Transcriptional Activation , Alleles , Cell Line , DNA Methylation/genetics , Epigenesis, Genetic , Female , Gene Silencing , Histone-Lysine N-Methyltransferase , Humans , Kruppel-Like Transcription Factors/genetics , Prader-Willi Syndrome/pathology
11.
Mol Autism ; 5: 44, 2014.
Article in English | MEDLINE | ID: mdl-25694803

ABSTRACT

BACKGROUND: Duplications of the chromosome 15q11-q13.1 region are associated with an estimated 1 to 3% of all autism cases, making this copy number variation (CNV) one of the most frequent chromosome abnormalities associated with autism spectrum disorder (ASD). Several genes located within the 15q11-q13.1 duplication region including ubiquitin protein ligase E3A (UBE3A), the gene disrupted in Angelman syndrome (AS), are involved in neural function and may play important roles in the neurobehavioral phenotypes associated with chromosome 15q11-q13.1 duplication (Dup15q) syndrome. METHODS: We have generated induced pluripotent stem cell (iPSC) lines from five different individuals containing CNVs of 15q11-q13.1. The iPSC lines were differentiated into mature, functional neurons. Gene expression across the 15q11-q13.1 locus was compared among the five iPSC lines and corresponding iPSC-derived neurons using quantitative reverse transcription PCR (qRT-PCR). Genome-wide gene expression was compared between neurons derived from three iPSC lines using mRNA-Seq. RESULTS: Analysis of 15q11-q13.1 gene expression in neurons derived from Dup15q iPSCs reveals that gene copy number does not consistently predict expression levels in cells with interstitial duplications of 15q11-q13.1. mRNA-Seq experiments show that there is substantial overlap in the genes differentially expressed between 15q11-q13.1 deletion and duplication neurons, Finally, we demonstrate that UBE3A transcripts can be pharmacologically rescued to normal levels in iPSC-derived neurons with a 15q11-q13.1 duplication. CONCLUSIONS: Chromatin structure may influence gene expression across the 15q11-q13.1 region in neurons. Genome-wide analyses suggest that common neuronal pathways may be disrupted in both the Angelman and Dup15q syndromes. These data demonstrate that our disease-specific stem cell models provide a new tool to decipher the underlying cellular and genetic disease mechanisms of ASD and may also offer a pathway to novel therapeutic intervention in Dup15q syndrome.

12.
Stem Cells Dev ; 22(10): 1477-89, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23351095

ABSTRACT

Gamma aminobutyric acid (GABA)-expressing interneurons are the major inhibitory cells of the cerebral cortex and hippocampus. These interneurons originate in the medial ganglionic eminence (MGE) and lateral ganglionic eminence of the ventral forebrain during embryonic development and show reduced survival and function in a variety of neurological disorders, including temporal lobe epilepsy. We and others have proposed that embryonic stem cell (ESC)-derived ventral forebrain progenitors might provide a source of new GABAergic interneurons for cell-based therapies. While human ESCs (hESCs) are readily differentiated in vitro into dorsal telencephalic neural progenitors, standard protocols for generating ventral subtypes of telencephalic progenitors are less effective. We now report efficient derivation of GABAergic progenitors using an established hESC reporter line that expresses green fluorescent protein (GFP) under the control of an endogenous NKX2.1 promoter. GABAergic progenitors were derived from this hESC line by a modified monolayer neural differentiation protocol. Consistent with sonic hedgehog (SHH)-dependent specification of NKX2.1-positive progenitors in the embryonic MGE, we show a dose-dependent increase in the generation of NKX2.1:GFP-positive progenitors after SHH treatment in vitro. Characterization of NKX2.1:GFP-positive cells confirms their identity as MGE-like neural progenitors, based on gene expression profiles and their ability to differentiate into GABAergic interneurons. We are also able to generate highly enriched populations of NKX2.1:GFP-positive progenitors, including cells with telencephalic identity, by fluorescence-activated cell sorting. These hESC-derived ventral forebrain progenitors are suitable candidates for cell-based therapies that aim at replacing dysfunctional or damaged cortical or hippocampal GABAergic interneurons.


Subject(s)
Cell Separation/methods , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Nuclear Proteins/metabolism , Prosencephalon/cytology , Transcription Factors/metabolism , Animals , Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Flow Cytometry , Green Fluorescent Proteins/metabolism , Hedgehog Proteins/pharmacology , Humans , Mice , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Recombinant Proteins/pharmacology , Thyroid Nuclear Factor 1
13.
Cell Transplant ; 21(8): 1603-11, 2012.
Article in English | MEDLINE | ID: mdl-22776287

ABSTRACT

Embryonic stem cells (ESCs) hold great therapeutic potential due to their ability to differentiate into cells of the three primary germ layers, which can be used to repopulate disease-damaged tissues. In fact, two cell therapies using ESC derivatives are currently in phase I clinical trials. A main concern in using ESCs and their derivatives for cell transplantation is the ability of undifferentiated ESCs to generate tumors in the host. Positive selection steps are often included in protocols designed to generate particular cell types from ESCs; however, the transition from ESC to progenitor cell or terminally differentiated cell is not synchronous, and residual undifferentiated cells often remain. In our transplants of ESC-derived neural progenitors (ESNPs) into the adult mouse hippocampus, we have observed the formation of teratocarcinomas. We set out to reduce teratocarcinoma formation by enrichment of ESNPs using fluorescence-activated cell sorting (FACS) and have found that, although enrichment prior to transplant reduces the overall rate of teratocarcinoma formation, the tumorigenicity of cell batches can vary widely, even after FACS enrichment to as much as 95% ESNPs. Our data suggest that this variability may be due to the percentage of residual ESCs remaining in the transplant cell population and to the presence of pluripotent epiblast-like cells, not previously identified in transplant batches. Our data emphasize the need for stringent characterization of transplant cell populations that will be used for cell replacement therapies in order to reduce the risk of tumor formation.


Subject(s)
Embryonic Stem Cells/cytology , Hippocampus/pathology , Neural Stem Cells/transplantation , Teratocarcinoma/pathology , Animals , Cell Differentiation , Cell Separation , Cells, Cultured , Flow Cytometry , Immunohistochemistry , Mice , Neural Stem Cells/cytology
14.
J Cell Biochem ; 111(3): 535-42, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20589755

ABSTRACT

The prospect of using embryonic stem cell (ESC)-derived neural progenitors and neurons to treat neurological disorders has led to great interest in defining the conditions that guide the differentiation of ESCs, and more recently induced pluripotent stem cells (iPSCs), into neural stem cells (NSCs) and a variety of neuronal and glial subtypes. Over the past decade, researchers have looked to the embryo to guide these studies, applying what we know about the signaling events that direct neural specification during development. This has led to the design of a number of protocols that successfully promote ESC neurogenesis, terminating with the production of neurons and glia with diverse regional addresses and functional properties. These protocols demonstrate that ESCs undergo neural specification in two, three, and four dimensions, mimicking the cell-cell interactions, patterning, and timing that characterizes the in vivo process. We therefore propose that these in vitro systems can be used to examine the molecular regulation of neural specification.


Subject(s)
Embryonic Stem Cells/physiology , Neural Stem Cells/cytology , Neurogenesis , Animals , Embryonic Stem Cells/cytology , Humans , Neurons/cytology , Research Design , Tissue Culture Techniques
15.
J Marital Fam Ther ; 28(2): 213-23, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11977381

ABSTRACT

Feminist approaches to therapy with adolescents emphasize an empowering focus on the strengths of adolescents while simultaneously insisting that therapists become aware of their own biases toward today's adolescents. However, a review of the family therapy literature finds little mention of feminist approaches for addressing injustices (e.g., family scapegoating, negative societal views of adolescents, and gender oppression) that arise in family therapy with adolescents. Therefore, this study explores clinical approaches and resources suggested by a surveyed group of self-identified feminist family therapists. In addition, we also recommend several approaches and resources that will aid family therapists in creating a more just climate for family therapy with youth.


Subject(s)
Adolescent Behavior/psychology , Family Therapy/methods , Social Justice , Adolescent , Counseling , Family Therapy/standards , Female , Feminism , Gender Identity , Health Services Research , Humans , Male , Power, Psychological , Professional-Patient Relations , United States
SELECTION OF CITATIONS
SEARCH DETAIL