Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Cell ; 143(4): 527-39, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-21074045

ABSTRACT

Autism spectrum disorders (ASD) are complex neurodevelopmental diseases in which different combinations of genetic mutations may contribute to the phenotype. Using Rett syndrome (RTT) as an ASD genetic model, we developed a culture system using induced pluripotent stem cells (iPSCs) from RTT patients' fibroblasts. RTT patients' iPSCs are able to undergo X-inactivation and generate functional neurons. Neurons derived from RTT-iPSCs had fewer synapses, reduced spine density, smaller soma size, altered calcium signaling and electrophysiological defects when compared to controls. Our data uncovered early alterations in developing human RTT neurons. Finally, we used RTT neurons to test the effects of drugs in rescuing synaptic defects. Our data provide evidence of an unexplored developmental window, before disease onset, in RTT syndrome where potential therapies could be successfully employed. Our model recapitulates early stages of a human neurodevelopmental disease and represents a promising cellular tool for drug screening, diagnosis and personalized treatment.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neurogenesis , Rett Syndrome/drug therapy , Rett Syndrome/pathology , Cell Proliferation , Female , Fibroblasts/cytology , Humans , Rett Syndrome/genetics , Synapses , X Chromosome Inactivation
2.
Proc Natl Acad Sci U S A ; 113(12): 3185-90, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26944080

ABSTRACT

Probing a wide range of cellular phenotypes in neurodevelopmental disorders using patient-derived neural progenitor cells (NPCs) can be facilitated by 3D assays, as 2D systems cannot entirely recapitulate the arrangement of cells in the brain. Here, we developed a previously unidentified 3D migration and differentiation assay in layered hydrogels to examine how these processes are affected in neurodevelopmental disorders, such as Rett syndrome. Our soft 3D system mimics the brain environment and accelerates maturation of neurons from human induced pluripotent stem cell (iPSC)-derived NPCs, yielding electrophysiologically active neurons within just 3 wk. Using this platform, we revealed a genotype-specific effect of methyl-CpG-binding protein-2 (MeCP2) dysfunction on iPSC-derived neuronal migration and maturation (reduced neurite outgrowth and fewer synapses) in 3D layered hydrogels. Thus, this 3D system expands the range of neural phenotypes that can be studied in vitro to include those influenced by physical and mechanical stimuli or requiring specific arrangements of multiple cell types.


Subject(s)
Cell Movement , Hydrogels , Induced Pluripotent Stem Cells/cytology , Methyl-CpG-Binding Protein 2/physiology , Neurons/metabolism , Humans
3.
EMBO Mol Med ; 13(1): e12523, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33501759

ABSTRACT

Duplication or deficiency of the X-linked MECP2 gene reliably produces profound neurodevelopmental impairment. MECP2 mutations are almost universally responsible for Rett syndrome (RTT), and particular mutations and cellular mosaicism of MECP2 may underlie the spectrum of RTT symptomatic severity. No clinically approved treatments for RTT are currently available, but human pluripotent stem cell technology offers a platform to identify neuropathology and test candidate therapeutics. Using a strategic series of increasingly complex human stem cell-derived technologies, including human neurons, MECP2-mosaic neurospheres to model RTT female brain mosaicism, and cortical organoids, we identified synaptic dysregulation downstream from knockout of MECP2 and screened select pharmacological compounds for their ability to treat this dysfunction. Two lead compounds, Nefiracetam and PHA 543613, specifically reversed MECP2-knockout cytologic neuropathology. The capacity of these compounds to reverse neuropathologic phenotypes and networks in human models supports clinical studies for neurodevelopmental disorders in which MeCP2 deficiency is the predominant etiology.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Neurons/drug effects , Organoids , Pyrrolidinones/pharmacology , Quinuclidines/pharmacology , Rett Syndrome , Female , Gene Knockout Techniques , Humans , Methyl-CpG-Binding Protein 2/genetics , Organoids/drug effects , Phenotype , Rett Syndrome/genetics
4.
PLoS One ; 16(4): e0243683, 2021.
Article in English | MEDLINE | ID: mdl-33909614

ABSTRACT

Identification of genomic mutations by molecular testing plays an important role in diagnosis, prognosis, and treatment of myeloid neoplasms. Next-generation sequencing (NGS) is an efficient method for simultaneous detection of clinically significant genomic mutations with high sensitivity. Various NGS based in-house developed and commercial myeloid neoplasm panels have been integrated into routine clinical practice. However, some genes frequently mutated in myeloid malignancies are particularly difficult to sequence with NGS panels (e.g., CEBPA, CARL, and FLT3). We report development and validation of a 48-gene NGS panel that includes genes that are technically challenging for molecular profiling of myeloid neoplasms including acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), and myeloproliferative neoplasms (MPN). Target regions were captured by hybridization with complementary biotinylated DNA baits, and NGS was performed on an Illumina NextSeq500 instrument. A bioinformatics pipeline that was developed in-house was used to detect single nucleotide variations (SNVs), insertions/deletions (indels), and FLT3 internal tandem duplications (FLT3-ITD). An analytical validation study was performed on 184 unique specimens for variants with allele frequencies ≥5%. Variants identified by the 48-gene panel were compared to those identified by a 35-gene hematologic neoplasms panel using an additional 137 unique specimens. The developed assay was applied to a large cohort (n = 2,053) of patients with suspected myeloid neoplasms. Analytical validation yielded 99.6% sensitivity (95% CI: 98.9-99.9%) and 100% specificity (95% CI: 100%). Concordance of variants detected by the 2 tested panels was 100%. Among patients with suspected myeloid neoplasms (n = 2,053), 54.5% patients harbored at least one clinically significant mutation: 77% in AML patients, 48% in MDS, and 45% in MPN. Together, these findings demonstrate that the assay can identify mutations associated with diagnosis, prognosis, and treatment options of myeloid neoplasms even in technically challenging genes.


Subject(s)
Hematologic Neoplasms , High-Throughput Nucleotide Sequencing , Leukemia, Myeloid, Acute , Mutation , Myelodysplastic Syndromes , Neoplasm Proteins , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
5.
Neurotherapeutics ; 12(3): 534-45, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25851569

ABSTRACT

Autism spectrum disorders (ASDs) are a heterogeneous group of neurodevelopmental disorders sharing a core set of symptoms, including impaired social interaction, language deficits, and repetitive behaviors. While ASDs are highly heritable and demonstrate a clear genetic component, the cellular and molecular mechanisms driving ASD etiology remain undefined. The unavailability of live patient-specific neurons has contributed to the difficulty in studying ASD pathophysiology. The recent advent of induced pluripotent stem cells (iPSCs) has provided the ability to generate patient-specific human neurons from somatic cells. The iPSC field has quickly grown, as researchers have demonstrated the utility of this technology to model several diseases, especially neurologic disorders. Here, we review the current literature around using iPSCs to model ASDs, and discuss the notable findings, and the promise and limitations of this technology. The recent report of a nonsyndromic ASD iPSC model and several previous ASD models demonstrating similar results points to the ability of iPSC to reveal potential novel biomarkers and therapeutics.


Subject(s)
Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/physiopathology , Induced Pluripotent Stem Cells/physiology , Neural Stem Cells/physiology , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Drug Evaluation, Preclinical , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Signal Transduction
6.
Exp Neurol ; 261: 1-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24918341

ABSTRACT

The neurotoxin beta-N-methylamino-l-alanine (BMAA) was first identified as a "toxin of interest" in regard to the amyotrophic lateral sclerosis-Parkinsonism Dementia Complex of Guam (ALS/PDC); studies in recent years highlighting widespread environmental sources of BMAA exposure and providing new clues to toxic mechanisms have suggested possible relevance to sporadic ALS as well. However, despite clear evidence of uptake into tissues and a range of toxic effects in cells and animals, an animal model in which BMAA induces a neurodegenerative picture resembling ALS is lacking, possibly in part reflecting limited understanding of critical factors pertaining to its absorption, biodistribution and metabolism. To bypass some of these issues and ensure delivery to a key site of disease pathology, we examined effects of prolonged (30day) intrathecal infusion in wild type (WT) rats, and rats harboring the familial ALS associated G93A SOD1 mutation, over an age range (80±2 to 110±2days) during which the G93A rats are developing disease pathology yet remain asymptomatic. The BMAA exposures induced changes that in many ways resemble those seen in the G93A rats, with degenerative changes in ventral horn motor neurons (MNs) with relatively little dorsal horn pathology, marked ventral horn astrogliosis and increased 3-nitrotyrosine labeling in and surrounding MNs, a loss of labeling for the astrocytic glutamate transporter, GLT-1, surrounding MNs, and mild accumulation and aggregation of TDP-43 in the cytosol of some injured and degenerating MNs. Thus, prolonged intrathecal infusion of BMAA can reproduce a picture in spinal cord incorporating many of the pathological hallmarks of diverse forms of human ALS, including substantial restriction of overt pathological changes to the ventral horn, consistent with the possibility that environmental BMAA exposure could be a risk factor and/or contributor to some human disease.


Subject(s)
Amino Acids, Diamino/toxicity , Amyotrophic Lateral Sclerosis/chemically induced , Amyotrophic Lateral Sclerosis/pathology , Anterior Horn Cells/drug effects , Excitatory Amino Acid Agonists/toxicity , Gliosis/chemically induced , Amyotrophic Lateral Sclerosis/complications , Amyotrophic Lateral Sclerosis/genetics , Animals , Cell Survival/drug effects , Cell Survival/genetics , Cyanobacteria Toxins , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein , Gliosis/genetics , Glutamate Plasma Membrane Transport Proteins/metabolism , Humans , Male , Rats , Rats, Transgenic , Spinal Cord/cytology , Superoxide Dismutase/genetics , Tyrosine/analogs & derivatives , Tyrosine/metabolism
7.
Curr Opin Neurobiol ; 22(5): 785-90, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22717528

ABSTRACT

The cellular and molecular mechanisms of neurodevelopmental conditions such as autism spectrum disorders have been studied intensively for decades. The unavailability of live patient neurons for research, however, has represented a major obstacle in the elucidation of the disease etiologies. Recently, the development of induced pluripotent stem cell (iPSC) technology allows for the generation of human neurons from somatic cells of patients. We review ongoing studies using iPSCs as an approach to model neurodevelopmental disorders, the promise and caveats of this technique and its potential for drug screening. The reproducible findings of relevant phenotypes in Rett syndrome iPSC-derived neurons suggest that iPSC technology offers a novel and unique opportunity for the understanding of and the development of therapeutics for other autism spectrum disorders.


Subject(s)
Cell Differentiation/physiology , Central Nervous System Diseases , Developmental Disabilities , Induced Pluripotent Stem Cells/physiology , Neurons/physiology , Central Nervous System Diseases/complications , Central Nervous System Diseases/pathology , Central Nervous System Diseases/therapy , Developmental Disabilities/complications , Developmental Disabilities/pathology , Developmental Disabilities/therapy , Humans
8.
Neurosci Lett ; 487(2): 129-33, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-19563863

ABSTRACT

Angelman syndrome (AS) is a severe neurological disorder caused by a deficiency of ubiquitin protein ligase E3A (UBE3A), but the pathophysiology of the disease remains unknown. We now report that in the brains of AS mice in which the maternal UBE3A allele is mutated (m-) and the paternal allele is potentially inactivated by imprinting (p+) (UBE3A m-\p+), the mitochondria are abnormal and exhibit a partial oxidative phosphorylation (OXPHOS) defect. Electron microscopy of the hippocampal region of the UBE3A m-\p+ mice (n=6) reveals small, dense mitochondria with altered cristae, relative to wild-type littermates (n=6) and reduced synaptic vesicle density. The specific activity of OXPHOS complex III is reduced in whole brain mitochondria in UBE3A m-\p+ (n=5) mice versus wild-type littermates (n=5). Therefore, mitochondrial dysfunction may contribute to the pathophysiology of Angelman syndrome.


Subject(s)
Angelman Syndrome/enzymology , CA1 Region, Hippocampal/enzymology , Disease Models, Animal , Mitochondria/enzymology , Neurons/enzymology , Ubiquitin-Protein Ligases/deficiency , Angelman Syndrome/genetics , Angelman Syndrome/pathology , Animals , CA1 Region, Hippocampal/pathology , Female , Genotype , Male , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/genetics , Mitochondria/pathology , Neurons/pathology , Neurons/physiology , Purkinje Cells/enzymology , Purkinje Cells/pathology , Synaptic Vesicles/genetics , Synaptic Vesicles/pathology , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL