Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(5): 114173, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38700984

ABSTRACT

Mutations in the phosphatase and tensin homolog (PTEN) gene are associated with severe neurodevelopmental disorders. Loss of PTEN leads to hyperactivation of the mechanistic target of rapamycin (mTOR), which functions in two distinct protein complexes, mTORC1 and mTORC2. The downstream signaling mechanisms that contribute to PTEN mutant phenotypes are not well delineated. Here, we show that pluripotent stem cell-derived PTEN mutant human neurons, neural precursors, and cortical organoids recapitulate disease-relevant phenotypes, including hypertrophy, electrical hyperactivity, enhanced proliferation, and structural overgrowth. PTEN loss leads to simultaneous hyperactivation of mTORC1 and mTORC2. We dissect the contribution of mTORC1 and mTORC2 by generating double mutants of PTEN and RPTOR or RICTOR, respectively. Our results reveal that the synergistic hyperactivation of both mTORC1 and mTORC2 is essential for the PTEN mutant human neural phenotypes. Together, our findings provide insights into the molecular mechanisms that underlie PTEN-related neural disorders and highlight novel therapeutic targets.


Subject(s)
Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Neurons , Organoids , PTEN Phosphohydrolase , Humans , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Organoids/metabolism , Neurons/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Mutation/genetics , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Signal Transduction , Cell Proliferation , Regulatory-Associated Protein of mTOR/metabolism , Regulatory-Associated Protein of mTOR/genetics , Phenotype
2.
Nat Nanotechnol ; 19(1): 58-69, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37500778

ABSTRACT

Structural DNA nanotechnology enables the fabrication of user-defined DNA origami nanostructures (DNs) for biological applications. However, the role of DN design during cellular interactions and subsequent biodistribution remain poorly understood. Current methods for tracking DN fates in situ, including fluorescent-dye labelling, suffer from low sensitivity and dye-induced artifacts. Here we present origamiFISH, a label-free and universal method for the single-molecule fluorescence detection of DNA origami nanostructures in cells and tissues. origamiFISH targets pan-DN scaffold sequences with hybridization chain reaction probes to achieve 1,000-fold signal amplification. We identify cell-type- and DN shape-specific spatiotemporal distribution patterns within a minute of uptake and at picomolar DN concentrations, 10,000× lower than field standards. We additionally optimize compatibility with immunofluorescence and tissue clearing to visualize DN distribution within tissue cryo-/vibratome sections, slice cultures and whole-mount organoids. Together, origamiFISH enables the accurate mapping of DN distribution across subcellular and tissue barriers for guiding the development of DN-based therapeutics.


Subject(s)
Nanostructures , Nanotechnology , Tissue Distribution , DNA/chemistry , Nanostructures/chemistry , Nucleic Acid Hybridization , Nucleic Acid Conformation
3.
Front Cell Dev Biol ; 11: 1158373, 2023.
Article in English | MEDLINE | ID: mdl-37101616

ABSTRACT

The brain is arguably the most complex part of the human body in form and function. Much remains unclear about the molecular mechanisms that regulate its normal and pathological physiology. This lack of knowledge largely stems from the inaccessible nature of the human brain, and the limitation of animal models. As a result, brain disorders are difficult to understand and even more difficult to treat. Recent advances in generating human pluripotent stem cells (hPSCs)-derived 2-dimensional (2D) and 3-dimensional (3D) neural cultures have provided an accessible system to model the human brain. Breakthroughs in gene editing technologies such as CRISPR/Cas9 further elevate the hPSCs into a genetically tractable experimental system. Powerful genetic screens, previously reserved for model organisms and transformed cell lines, can now be performed in human neural cells. Combined with the rapidly expanding single-cell genomics toolkit, these technological advances culminate to create an unprecedented opportunity to study the human brain using functional genomics. This review will summarize the current progress of applying CRISPR-based genetic screens in hPSCs-derived 2D neural cultures and 3D brain organoids. We will also evaluate the key technologies involved and discuss their related experimental considerations and future applications.

4.
J Mol Biol ; 434(3): 167386, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34883115

ABSTRACT

Developmental brain diseases encompass a group of conditions resulting from genetic or environmental perturbations during early development. Despite the increased research attention in recent years following recognition of the prevalence of these diseases, there is still a significant lack of knowledge of their etiology and treatment options. The genetic and clinical heterogeneity of these diseases, in addition to the limitations of experimental animal models, contribute to this difficulty. In this regard, the advent of brain organoid technology has provided a new means to study the cause and progression of developmental brain diseases in vitro. Derived from human pluripotent stem cells, brain organoids have been shown to recapitulate key developmental milestones of the early human brain. Combined with technological advancements in genome editing, tissue engineering, electrophysiology, and multi-omics analysis, brain organoids have expanded the frontiers of human neurobiology, providing valuable insight into the cellular and molecular mechanisms of normal and pathological brain development. This review will summarize the current progress of applying brain organoids to model human developmental brain diseases and discuss the challenges that need to be overcome to further advance their utility.


Subject(s)
Brain Diseases , Brain , Organoids , Pluripotent Stem Cells , Brain/abnormalities , Brain Diseases/embryology , Cell Culture Techniques , Humans , Organoids/abnormalities
5.
Mol Brain ; 14(1): 131, 2021 08 30.
Article in English | MEDLINE | ID: mdl-34461955

ABSTRACT

The phosphatase and tensin homolog (PTEN) protein, encoded by the PTEN gene on chromosome 10, is a negative regulator of the phosphoinositide 3-kinase (PI3K) signaling pathway. Loss of PTEN has been linked to an array of human diseases, including neurodevelopmental disorders such as macrocephaly and autism. However, it remains unknown whether increased dosage of PTEN can lead to human disease. A recent human genetics study identifies chromosome 10 microduplication encompassing PTEN in patients with microcephaly. Here we generated a human brain organoid model of increased PTEN dosage. We showed that mild PTEN overexpression led to reduced neural precursor proliferation, premature neuronal differentiation, and the formation of significantly smaller brain organoids. PTEN overexpression resulted in decreased AKT activation, and treatment of wild-type organoids with an AKT inhibitor recapitulated the reduced brain organoid growth phenotypes. Together, our findings provide functional evidence that PTEN is a dosage-sensitive gene that regulates human neurodevelopment, and that increased PTEN dosage in brain organoids results in microcephaly-like phenotypes.


Subject(s)
Microcephaly/genetics , Organoids/metabolism , PTEN Phosphohydrolase/biosynthesis , Cell Line , Embryoid Bodies/drug effects , Gene Dosage , Gene Duplication , Genes, Reporter , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction
6.
Sci Adv ; 7(5)2021 01.
Article in English | MEDLINE | ID: mdl-33514545

ABSTRACT

Slow progress in the fight against neurodegenerative diseases (NDs) motivates an urgent need for highly controlled in vitro systems to investigate organ-organ- and organ-immune-specific interactions relevant for disease pathophysiology. Of particular interest is the gut/microbiome-liver-brain axis for parsing out how genetic and environmental factors contribute to NDs. We have developed a mesofluidic platform technology to study gut-liver-cerebral interactions in the context of Parkinson's disease (PD). It connects microphysiological systems (MPSs) of the primary human gut and liver with a human induced pluripotent stem cell-derived cerebral MPS in a systemically circulated common culture medium containing CD4+ regulatory T and T helper 17 cells. We demonstrate this approach using a patient-derived cerebral MPS carrying the PD-causing A53T mutation, gaining two important findings: (i) that systemic interaction enhances features of in vivo-like behavior of cerebral MPSs, and (ii) that microbiome-associated short-chain fatty acids increase expression of pathology-associated pathways in PD.


Subject(s)
Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Parkinson Disease , Brain/metabolism , Humans , Liver/metabolism , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism
7.
J Neurosci ; 40(6): 1186-1193, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32024767

ABSTRACT

In vitro differentiation of pluripotent stem cells provides a systematic platform to study development and disease. Recent advances in brain organoid technology have created new opportunities to investigate the formation and function of the human brain, under physiological and pathological conditions. Brain organoids can be generated to model the cellular and structural development of the human brain, and allow the investigation of the intricate interactions between resident neural and glial cell types. Combined with new advances in gene editing, imaging, and genomic analysis, brain organoid technology can be applied to address questions pertinent to human brain development, disease, and evolution. However, the current iterations of brain organoids also have limitations in faithfully recapitulating the in vivo processes. In this perspective, we evaluate the recent progress in brain organoid technology, and discuss the experimental considerations for its utilization.Dual Perspectives Companion Paper: Integrating CRISPR Engineering and hiPSC-Derived 2D Disease Modeling Systems, by Kristina Rehbach, Michael B. Fernando, and Kristen J. Brennand.


Subject(s)
Brain/physiology , Organoids , Brain/embryology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology
8.
Glia ; 68(9): 1692-1728, 2020 09.
Article in English | MEDLINE | ID: mdl-31958188

ABSTRACT

Development, physiological functions, and pathologies of the brain depend on tight interactions between neurons and different types of glial cells, such as astrocytes, microglia, oligodendrocytes, and oligodendrocyte precursor cells. Assessing the relative contribution of different glial cell types is required for the full understanding of brain function and dysfunction. Over the recent years, several technological breakthroughs were achieved, allowing "glio-scientists" to address new challenging biological questions. These technical developments make it possible to study the roles of specific cell types with medium or high-content workflows and perform fine analysis of their mutual interactions in a preserved environment. This review illustrates the potency of several cutting-edge experimental approaches (advanced cell cultures, induced pluripotent stem cell (iPSC)-derived human glial cells, viral vectors, in situ glia imaging, opto- and chemogenetic approaches, and high-content molecular analysis) to unravel the role of glial cells in specific brain functions or diseases. It also illustrates the translation of some techniques to the clinics, to monitor glial cells in patients, through specific brain imaging methods. The advantages, pitfalls, and future developments are discussed for each technique, and selected examples are provided to illustrate how specific "gliobiological" questions can now be tackled.


Subject(s)
Astrocytes , Neuroglia , Humans , Microglia , Neurons , Oligodendroglia
9.
Proc Natl Acad Sci U S A ; 116(19): 9527-9532, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31019072

ABSTRACT

Zika virus (ZIKV) is a neurotropic and neurovirulent arbovirus that has severe detrimental impact on the developing human fetal brain. To date, little is known about the factors required for ZIKV infection of human neural cells. We identified ZIKV host genes in human pluripotent stem cell (hPSC)-derived neural progenitors (NPs) using a genome-wide CRISPR-Cas9 knockout screen. Mutations of host factors involved in heparan sulfation, endocytosis, endoplasmic reticulum processing, Golgi function, and interferon activity conferred resistance to infection with the Uganda strain of ZIKV and a more recent North American isolate. Host genes essential for ZIKV replication identified in human NPs also provided a low level of protection against ZIKV in isogenic human astrocytes. Our findings provide insights into host-dependent mechanisms for ZIKV infection in the highly vulnerable human NP cells and identify molecular targets for potential therapeutic intervention.


Subject(s)
CRISPR-Cas Systems , Disease Resistance/genetics , Neural Stem Cells/virology , Virus Replication/genetics , Zika Virus Infection/genetics , Zika Virus/physiology , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/virology , Cell Line , Female , Genome-Wide Association Study , Humans , Male , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Zika Virus Infection/metabolism , Zika Virus Infection/pathology
10.
Cell Rep ; 25(2): 368-382.e5, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30304678

ABSTRACT

Most genes mutated in microcephaly patients are expressed ubiquitously, and yet the brain is the only major organ compromised in most patients. Why the phenotype remains brain specific is poorly understood. In this study, we used in vitro differentiation of human embryonic stem cells to monitor the effect of a point mutation in kinetochore null protein 1 (KNL1; CASC5), identified in microcephaly patients, during in vitro brain development. We found that neural progenitors bearing a patient mutation showed reduced KNL1 levels, aneuploidy, and an abrogated spindle assembly checkpoint. By contrast, no reduction of KNL1 levels or abnormalities was observed in fibroblasts and neural crest cells. We established that the KNL1 patient mutation generates an exonic splicing silencer site, which mainly affects neural progenitors because of their higher levels of splicing proteins. Our results provide insight into the brain-specific phenomenon, consistent with microcephaly being the only major phenotype of patients bearing KNL1 mutation.


Subject(s)
Brain/pathology , Kinetochores/pathology , Microcephaly/genetics , Microcephaly/pathology , Microtubule-Associated Proteins/genetics , Mutation , RNA Splicing , Brain/metabolism , Cells, Cultured , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Humans , Kinetochores/metabolism , M Phase Cell Cycle Checkpoints , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Phenotype
11.
Proc Natl Acad Sci U S A ; 115(27): 7117-7122, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29915057

ABSTRACT

Maternal Zika virus (ZIKV) infection during pregnancy is recognized as the cause of an epidemic of microcephaly and other neurological anomalies in human fetuses. It remains unclear how ZIKV accesses the highly vulnerable population of neural progenitors of the fetal central nervous system (CNS), and which cell types of the CNS may be viral reservoirs. In contrast, the related dengue virus (DENV) does not elicit teratogenicity. To model viral interaction with cells of the fetal CNS in vitro, we investigated the tropism of ZIKV and DENV for different induced pluripotent stem cell-derived human cells, with a particular focus on microglia-like cells. We show that ZIKV infected isogenic neural progenitors, astrocytes, and microglia-like cells (pMGLs), but was only cytotoxic to neural progenitors. Infected glial cells propagated ZIKV and maintained ZIKV load over time, leading to viral spread to susceptible cells. DENV triggered stronger immune responses and could be cleared by neural and glial cells more efficiently. pMGLs, when cocultured with neural spheroids, invaded the tissue and, when infected with ZIKV, initiated neural infection. Since microglia derive from primitive macrophages originating in proximity to the maternal vasculature, they may act as a viral reservoir for ZIKV and establish infection of the fetal brain. Infection of immature neural stem cells by invading microglia may occur in the early stages of pregnancy, before angiogenesis in the brain rudiments. Our data are also consistent with ZIKV and DENV affecting the integrity of the blood-brain barrier, thus allowing infection of the brain later in life.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Neuroglia/metabolism , Pregnancy Complications, Infectious/metabolism , Zika Virus Infection/metabolism , Zika Virus/metabolism , Female , Humans , Induced Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/virology , Neural Stem Cells/pathology , Neural Stem Cells/virology , Neuroglia/pathology , Neuroglia/virology , Pregnancy , Pregnancy Complications, Infectious/pathology , Zika Virus Infection/pathology
12.
Cell ; 172(5): 979-992.e6, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29456084

ABSTRACT

Fragile X syndrome (FXS), the most common genetic form of intellectual disability in males, is caused by silencing of the FMR1 gene associated with hypermethylation of the CGG expansion mutation in the 5' UTR of FMR1 in FXS patients. Here, we applied recently developed DNA methylation editing tools to reverse this hypermethylation event. Targeted demethylation of the CGG expansion by dCas9-Tet1/single guide RNA (sgRNA) switched the heterochromatin status of the upstream FMR1 promoter to an active chromatin state, restoring a persistent expression of FMR1 in FXS iPSCs. Neurons derived from methylation-edited FXS iPSCs rescued the electrophysiological abnormalities and restored a wild-type phenotype upon the mutant neurons. FMR1 expression in edited neurons was maintained in vivo after engrafting into the mouse brain. Finally, demethylation of the CGG repeats in post-mitotic FXS neurons also reactivated FMR1. Our data establish that demethylation of the CGG expansion is sufficient for FMR1 reactivation, suggesting potential therapeutic strategies for FXS.


Subject(s)
DNA Methylation/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Gene Editing , Neurons/pathology , Animals , CRISPR-Associated Protein 9/metabolism , Epigenesis, Genetic , HEK293 Cells , Heterochromatin/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Kinetics , Male , Mice , Neurons/metabolism , Phenotype , Promoter Regions, Genetic , RNA, Guide, Kinetoplastida/metabolism , Trinucleotide Repeat Expansion/genetics
13.
Cell Stem Cell ; 20(3): 385-396.e3, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28041895

ABSTRACT

An expansion of the cerebral neocortex is thought to be the foundation for the unique intellectual abilities of humans. It has been suggested that an increase in the proliferative potential of neural progenitors (NPs) underlies the expansion of the cortex and its convoluted appearance. Here we show that increasing NP proliferation induces expansion and folding in an in vitro model of human corticogenesis. Deletion of PTEN stimulates proliferation and generates significantly larger and substantially folded cerebral organoids. This genetic modification allows sustained cell cycle re-entry, expansion of the progenitor population, and delayed neuronal differentiation, all key features of the developing human cortex. In contrast, Pten deletion in mouse organoids does not lead to folding. Finally, we utilized the expanded cerebral organoids to show that infection with Zika virus impairs cortical growth and folding. Our study provides new insights into the mechanisms regulating the structure and organization of the human cortex.


Subject(s)
Cerebrum/cytology , Organoids/cytology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Gene Deletion , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Organoids/drug effects , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Zika Virus/drug effects , Zika Virus/physiology , Zika Virus Infection/pathology , Zika Virus Infection/virology
14.
Nat Med ; 22(11): 1358-1367, 2016 11.
Article in English | MEDLINE | ID: mdl-27668937

ABSTRACT

Microglia, the only lifelong resident immune cells of the central nervous system (CNS), are highly specialized macrophages that have been recognized to have a crucial role in neurodegenerative diseases such as Alzheimer's, Parkinson's and adrenoleukodystrophy (ALD). However, in contrast to other cell types of the human CNS, bona fide microglia have not yet been derived from cultured human pluripotent stem cells. Here we establish a robust and efficient protocol for the rapid production of microglia-like cells from human (h) embryonic stem (ES) and induced pluripotent stem (iPS) cells that uses defined serum-free culture conditions. These in vitro pluripotent stem cell-derived microglia-like cells (termed pMGLs) faithfully recapitulate the expected ontogeny and characteristics of their in vivo counterparts, and they resemble primary fetal human and mouse microglia. We generated these cells from multiple disease-specific cell lines and find that pMGLs derived from an hES model of Rett syndrome are smaller than their isogenic controls. We further describe a platform to study the integration and live behavior of pMGLs in organotypic 3D cultures. This modular differentiation system allows for the study of microglia in highly defined conditions as they mature in response to developmentally relevant cues, and it provides a framework in which to study the long-term interactions of microglia residing in a tissue-like environment.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Microglia/cytology , Humans , In Vitro Techniques , Microglia/immunology , Organ Culture Techniques , Rett Syndrome/immunology
15.
Curr Opin Cell Biol ; 43: 96-103, 2016 12.
Article in English | MEDLINE | ID: mdl-27768957

ABSTRACT

In vitro differentiation of human pluripotent stem cells provides a systematic platform to investigate the physiological development and function of the human nervous system, as well as the etiology and consequence when these processes go awry. Recent development in three-dimensional (3D) organotypic culture systems allows modeling of the complex structure formation of the human CNS, and the intricate interactions between various resident neuronal and glial cell types. Combined with an ever-expanding genome editing and regulation toolkit such as CRISPR/Cas9, it is now a possibility to study human neurological disease in the relevant molecular, cellular and anatomical context. In this article, we review recent progress in 3D neural culture and the implications for disease modeling.


Subject(s)
Central Nervous System Diseases/pathology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Models, Biological , Pluripotent Stem Cells/cytology , Animals , Central Nervous System Diseases/metabolism , Genetic Engineering , Humans , Organoids/pathology
16.
Nature ; 515(7526): 274-8, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25307057

ABSTRACT

Alzheimer's disease is the most common form of dementia, characterized by two pathological hallmarks: amyloid-ß plaques and neurofibrillary tangles. The amyloid hypothesis of Alzheimer's disease posits that the excessive accumulation of amyloid-ß peptide leads to neurofibrillary tangles composed of aggregated hyperphosphorylated tau. However, to date, no single disease model has serially linked these two pathological events using human neuronal cells. Mouse models with familial Alzheimer's disease (FAD) mutations exhibit amyloid-ß-induced synaptic and memory deficits but they do not fully recapitulate other key pathological events of Alzheimer's disease, including distinct neurofibrillary tangle pathology. Human neurons derived from Alzheimer's disease patients have shown elevated levels of toxic amyloid-ß species and phosphorylated tau but did not demonstrate amyloid-ß plaques or neurofibrillary tangles. Here we report that FAD mutations in ß-amyloid precursor protein and presenilin 1 are able to induce robust extracellular deposition of amyloid-ß, including amyloid-ß plaques, in a human neural stem-cell-derived three-dimensional (3D) culture system. More importantly, the 3D-differentiated neuronal cells expressing FAD mutations exhibited high levels of detergent-resistant, silver-positive aggregates of phosphorylated tau in the soma and neurites, as well as filamentous tau, as detected by immunoelectron microscopy. Inhibition of amyloid-ß generation with ß- or γ-secretase inhibitors not only decreased amyloid-ß pathology, but also attenuated tauopathy. We also found that glycogen synthase kinase 3 (GSK3) regulated amyloid-ß-mediated tau phosphorylation. We have successfully recapitulated amyloid-ß and tau pathology in a single 3D human neural cell culture system. Our unique strategy for recapitulating Alzheimer's disease pathology in a 3D neural cell culture model should also serve to facilitate the development of more precise human neural cell models of other neurodegenerative disorders.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Cell Culture Techniques/methods , Models, Biological , Neural Stem Cells/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Cell Differentiation , Drug Evaluation, Preclinical/methods , Extracellular Space/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/pathology , Neurites/metabolism , Phosphorylation , Presenilin-1/metabolism , Protein Aggregation, Pathological , Reproducibility of Results , tau Proteins/chemistry , tau Proteins/metabolism
17.
Cell Rep ; 6(2): 313-24, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24440720

ABSTRACT

Glioblastoma (GBM) harbors subpopulations of therapy-resistant tumor-initiating cells (TICs) that are self-renewing and multipotent. To understand the regulation of the TIC state, we performed an image-based screen for genes regulating GBM TIC maintenance and identified ZFHX4, a 397 kDa transcription factor. ZFHX4 is required to maintain TIC-associated and normal human neural precursor cell phenotypes in vitro, suggesting that ZFHX4 regulates differentiation, and its suppression increases glioma-free survival in intracranial xenografts. ZFHX4 interacts with CHD4, a core member of the nucleosome remodeling and deacetylase (NuRD) complex. ZFHX4 and CHD4 bind to overlapping sets of genomic loci and control similar gene expression programs. Using expression data derived from GBM patients, we found that ZFHX4 significantly affects CHD4-mediated gene expression perturbations, which defines ZFHX4 as a master regulator of CHD4. These observations define ZFHX4 as a regulatory factor that links the chromatin-remodeling NuRD complex and the GBM TIC state.


Subject(s)
Autoantigens/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Homeodomain Proteins/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Transcription Factors/metabolism , Animals , Autoantigens/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line, Tumor , Glioblastoma/genetics , Homeodomain Proteins/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mice, Inbred NOD , Protein Binding , Transcription Factors/genetics , Transcription, Genetic
18.
Cell Stem Cell ; 13(4): 446-58, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24094325

ABSTRACT

Rett syndrome (RTT) is caused by mutations of MECP2, a methyl CpG binding protein thought to act as a global transcriptional repressor. Here we show, using an isogenic human embryonic stem cell model of RTT, that MECP2 mutant neurons display key molecular and cellular features of this disorder. Unbiased global gene expression analyses demonstrate that MECP2 functions as a global activator in neurons but not in neural precursors. Decreased transcription in neurons was coupled with a significant reduction in nascent protein synthesis and lack of MECP2 was manifested as a severe defect in the activity of the AKT/mTOR pathway. Lack of MECP2 also leads to impaired mitochondrial function in mutant neurons. Activation of AKT/mTOR signaling by exogenous growth factors or by depletion of PTEN boosted protein synthesis and ameliorated disease phenotypes in mutant neurons. Our findings indicate a vital function for MECP2 in maintaining active gene transcription in human neuronal cells.


Subject(s)
Embryonic Stem Cells/pathology , Methyl-CpG-Binding Protein 2/metabolism , Neurons/pathology , Protein Biosynthesis/genetics , Rett Syndrome/genetics , Rett Syndrome/pathology , Transcription, Genetic/genetics , Cells, Cultured , Embryonic Stem Cells/metabolism , Humans , Mutation , Neurons/metabolism
19.
Science ; 342(6161): 983-7, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24158904

ABSTRACT

The induced pluripotent stem (iPS) cell field holds promise for in vitro disease modeling. However, identifying innate cellular pathologies, particularly for age-related neurodegenerative diseases, has been challenging. Here, we exploited mutation correction of iPS cells and conserved proteotoxic mechanisms from yeast to humans to discover and reverse phenotypic responses to α-synuclein (αsyn), a key protein involved in Parkinson's disease (PD). We generated cortical neurons from iPS cells of patients harboring αsyn mutations, who are at high risk of developing PD dementia. Genetic modifiers from unbiased screens in a yeast model of αsyn toxicity led to identification of early pathogenic phenotypes in patient neurons. These included nitrosative stress, accumulation of endoplasmic reticulum (ER)-associated degradation substrates, and ER stress. A small molecule identified in a yeast screen (NAB2), and the ubiquitin ligase Nedd4 it affects, reversed pathologic phenotypes in these neurons.


Subject(s)
Benzimidazoles/pharmacology , Neurons/drug effects , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Benzimidazoles/chemistry , Endoplasmic Reticulum Stress/drug effects , Female , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mutation , Neurogenesis , Neurons/metabolism , Neurons/pathology , Parkinson Disease/genetics , Rats , alpha-Synuclein/genetics
20.
Proc Natl Acad Sci U S A ; 108(46): 18714-9, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22065768

ABSTRACT

The current gold standard for the culture of human pluripotent stem cells requires the use of a feeder layer of cells. Here, we develop a spatially defined culture system based on UV/ozone radiation modification of typical cell culture plastics to define a favorable surface environment for human pluripotent stem cell culture. Chemical and geometrical optimization of the surfaces enables control of early cell aggregation from fully dissociated cells, as predicted from a numerical model of cell migration, and results in significant increases in cell growth of undifferentiated cells. These chemically defined xeno-free substrates generate more than three times the number of cells than feeder-containing substrates per surface area. Further, reprogramming and typical gene-targeting protocols can be readily performed on these engineered surfaces. These substrates provide an attractive cell culture platform for the production of clinically relevant factor-free reprogrammed cells from patient tissue samples and facilitate the definition of standardized scale-up friendly methods for disease modeling and cell therapeutic applications.


Subject(s)
Cell Culture Techniques , Pluripotent Stem Cells/cytology , Tissue Engineering/methods , Biocompatible Materials/chemistry , Cells, Cultured , Humans , Materials Testing , Microscopy, Fluorescence/methods , Ozone/chemistry , Polymers/chemistry , Polystyrenes/chemistry , Surface Properties , Transgenes , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...