Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Mol Cell ; 53(1): 19-31, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24289922

ABSTRACT

Fragile X syndrome (FXS) is caused by a CGG repeat expansion in the FMR1 gene that appears to occur during oogenesis and during early embryogenesis. One model proposes that repeat instability depends on the replication fork direction through the repeats such that (CNG)n hairpin-like structures form, causing DNA polymerase to stall and slip. Examining DNA replication fork progression on single DNA molecules at the endogenous FMR1 locus revealed that replication forks stall at CGG repeats in human cells. Furthermore, replication profiles of FXS human embryonic stem cells (hESCs) compared to nonaffected hESCs showed that fork direction through the repeats is altered at the FMR1 locus in FXS hESCs, such that predominantly the CCG strand serves as the lagging-strand template. This is due to the absence of replication initiation that would typically occur upstream of FMR1, suggesting that altered replication origin usage combined with fork stalling promotes repeat instability during early embryonic development.


Subject(s)
DNA Replication , Embryonic Stem Cells/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/embryology , Genetic Loci , Trinucleotide Repeats , Embryonic Development/genetics , Embryonic Stem Cells/pathology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Humans
2.
Proc Natl Acad Sci U S A ; 114(14): 3696-3701, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28325870

ABSTRACT

Gene editing techniques have been extensively used to attempt to model recurrent genomic rearrangements found in tumor cells. These methods involve the induction of double-strand breaks at endogenous loci followed by the identification of breakpoint junctions within a population, which typically arise by nonhomologous end joining. The low frequency of these events, however, has hindered the cloning of cells with the desired rearrangement before oncogenic transformation. Here we present a strategy combining CRISPR-Cas9 technology and homology-directed repair to allow for the selection of human mesenchymal stem cells harboring the oncogenic translocation EWSR1-WT1 found in the aggressive desmoplastic small round cell tumor. The expression of the fusion transcript is under the control of the endogenous EWSR1 promoter and, importantly, can be conditionally expressed using Cre recombinase. This method is easily adapted to generate any cancer-relevant rearrangement.


Subject(s)
Gene Editing/methods , RNA-Binding Protein EWS/genetics , Translocation, Genetic , WT1 Proteins/genetics , CRISPR-Cas Systems , DNA Breaks, Double-Stranded , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Promoter Regions, Genetic
3.
Genome Res ; 23(7): 1182-93, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23568838

ABSTRACT

Chromosomal translocations are signatures of numerous cancers and lead to expression of fusion genes that act as oncogenes. The wealth of genomic aberrations found in cancer, however, makes it challenging to assign a specific phenotypic change to a specific aberration. In this study, we set out to use genome editing with zinc finger (ZFN) and transcription activator-like effector (TALEN) nucleases to engineer, de novo, translocation-associated oncogenes at cognate endogenous loci in human cells. Using ZFNs and TALENs designed to cut precisely at relevant translocation breakpoints, we induced cancer-relevant t(11;22)(q24;q12) and t(2;5)(p23;q35) translocations found in Ewing sarcoma and anaplastic large cell lymphoma (ALCL), respectively. We recovered both translocations with high efficiency, resulting in the expression of the EWSR1-FLI1 and NPM1-ALK fusions. Breakpoint junctions recovered after ZFN cleavage in human embryonic stem (ES) cell-derived mesenchymal precursor cells fully recapitulated the genomic characteristics found in tumor cells from Ewing sarcoma patients. This approach with tailored nucleases demonstrates that expression of fusion genes found in cancer cells can be induced from the native promoter, allowing interrogation of both the underlying mechanisms and oncogenic consequences of tumor-related translocations in human cells. With an analogous strategy, the ALCL translocation was reverted in a patient cell line to restore the integrity of the two participating chromosomes, further expanding the repertoire of genomic rearrangements that can be engineered by tailored nucleases.


Subject(s)
Endonucleases/metabolism , Neoplasms/enzymology , Neoplasms/genetics , Translocation, Genetic , Zinc Fingers , Cell Line , Chromosome Breakpoints , Humans , Nucleophosmin , Protein-Tyrosine Kinases/genetics , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism
4.
Stem Cells ; 33(2): 367-77, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25336442

ABSTRACT

The predominant view of pluripotency regulation proposes a stable ground state with coordinated expression of key transcription factors (TFs) that prohibit differentiation. Another perspective suggests a more complexly regulated state involving competition between multiple lineage-specifying TFs that define pluripotency. These contrasting views were developed from extensive analyses of TFs in pluripotent cells in vitro. An experimentally validated, genome-wide repertoire of the regulatory interactions that control pluripotency within the in vivo cellular contexts is yet to be developed. To address this limitation, we assembled a TF interactome of adult human male germ cell tumors (GCTs) using the Algorithm for the Accurate Reconstruction of Cellular Pathways (ARACNe) to analyze gene expression profiles of 141 tumors comprising pluripotent and differentiated subsets. The network (GCT(Net)) comprised 1,305 TFs, and its ingenuity pathway analysis identified pluripotency and embryonal development as the top functional pathways. We experimentally validated GCT(Net) by functional (silencing) and biochemical (ChIP-seq) analysis of the core pluripotency regulatory TFs POU5F1, NANOG, and SOX2 in relation to their targets predicted by ARACNe. To define the extent of the in vivo pluripotency network in this system, we ranked all TFs in the GCT(Net) according to sharing of ARACNe-predicted targets with those of POU5F1 and NANOG using an odds-ratio analysis method. To validate this network, we silenced the top 10 TFs in the network in H9 embryonic stem cells. Silencing of each led to downregulation of pluripotency and induction of lineage; 7 of the 10 TFs were identified as pluripotency regulators for the first time.


Subject(s)
Algorithms , Models, Biological , Neoplasm Proteins/metabolism , Neoplasms, Germ Cell and Embryonal/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Adult , Cell Line, Tumor , Humans , Male , Neoplasm Proteins/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/pathology , Pluripotent Stem Cells/pathology , Transcription Factors/genetics
5.
Nature ; 461(7262): 402-6, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19693009

ABSTRACT

The isolation of human induced pluripotent stem cells (iPSCs) offers a new strategy for modelling human disease. Recent studies have reported the derivation and differentiation of disease-specific human iPSCs. However, a key challenge in the field is the demonstration of disease-related phenotypes and the ability to model pathogenesis and treatment of disease in iPSCs. Familial dysautonomia (FD) is a rare but fatal peripheral neuropathy, caused by a point mutation in the IKBKAP gene involved in transcriptional elongation. The disease is characterized by the depletion of autonomic and sensory neurons. The specificity to the peripheral nervous system and the mechanism of neuron loss in FD are poorly understood owing to the lack of an appropriate model system. Here we report the derivation of patient-specific FD-iPSCs and the directed differentiation into cells of all three germ layers including peripheral neurons. Gene expression analysis in purified FD-iPSC-derived lineages demonstrates tissue-specific mis-splicing of IKBKAP in vitro. Patient-specific neural crest precursors express particularly low levels of normal IKBKAP transcript, suggesting a mechanism for disease specificity. FD pathogenesis is further characterized by transcriptome analysis and cell-based assays revealing marked defects in neurogenic differentiation and migration behaviour. Furthermore, we use FD-iPSCs for validating the potency of candidate drugs in reversing aberrant splicing and ameliorating neuronal differentiation and migration. Our study illustrates the promise of iPSC technology for gaining new insights into human disease pathogenesis and treatment.


Subject(s)
Dysautonomia, Familial/pathology , Dysautonomia, Familial/therapy , Models, Biological , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/transplantation , Adolescent , Alternative Splicing/drug effects , Alternative Splicing/genetics , Animals , Carrier Proteins/genetics , Cell Dedifferentiation , Cell Differentiation , Cell Lineage , Cell Movement , Cells, Cultured , Child , Dysautonomia, Familial/drug therapy , Dysautonomia, Familial/genetics , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Kinetin/pharmacology , Kinetin/therapeutic use , Male , Mice , Neural Crest/cytology , Neural Crest/drug effects , Organ Specificity , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Transcriptional Elongation Factors
6.
Proc Natl Acad Sci U S A ; 106(26): 10620-5, 2009 Jun 30.
Article in English | MEDLINE | ID: mdl-19549848

ABSTRACT

The precise genetic manipulation of stem and precursor cells offers extraordinary potential for the analysis, prevention, and treatment of human malignancies. Chromosomal translocations are hallmarks of several tumor types where they are thought to have arisen in stem or precursor cells. Although approaches exist to study factors involved in translocation formation in mouse cells, approaches in human cells have been lacking, especially in relevant cell types. The technology of zinc finger nucleases (ZFNs) allows DNA double-strand breaks (DSBs) to be introduced into specified chromosomal loci. We harnessed this technology to induce chromosomal translocations in human cells by generating concurrent DSBs at 2 endogenous loci, the PPP1R12C/p84 gene on chromosome 19 and the IL2Rgamma gene on the X chromosome. Translocation breakpoint junctions for t(19;X) were detected with nested quantitative PCR in a high throughput 96-well format using denaturation curves and DNA sequencing in a variety of human cell types, including embryonic stem (hES) cells and hES cell-derived mesenchymal precursor cells. Although readily detected, translocations were less frequent than repair of a single DSB by gene targeting or nonhomologous end-joining, neither of which leads to gross chromosomal rearrangements. While previous studies have relied on laborious genetic modification of cells and extensive growth in culture, the approach described in this report is readily applicable to primary human cells, including multipotent and pluripotent cells, to uncover both the underlying mechanisms and phenotypic consequences of targeted translocations and other genomic rearrangements.


Subject(s)
DNA Damage , Stem Cells/metabolism , Translocation, Genetic , Base Sequence , Cell Line , Chromosome Breakage , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 6/genetics , Chromosomes, Human, X/genetics , DNA Breaks, Double-Stranded , DNA Breaks, Single-Stranded , DNA Repair , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Rearrangement , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , In Situ Hybridization, Fluorescence , Interleukin Receptor Common gamma Subunit/genetics , Polymerase Chain Reaction , Stem Cells/cytology
7.
Proc Natl Acad Sci U S A ; 106(31): 12759-64, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19549847

ABSTRACT

Human-induced pluripotent stem cells (hiPSCs) are generated from somatic cells by ectopic expression of the 4 reprogramming factors (RFs) Oct-4, Sox2, Klf4, and c-Myc. To better define the stoichiometric requirements and dynamic expression patterns required for successful hiPSC induction, we generated 4 bicistronic lentiviral vectors encoding the 4 RFs co-expressed with discernable fluorescent proteins. Using this system, we define the optimal stoichiometry of RF expression to be highly sensitive to Oct4 dosage, and we demonstrate the impact that variations in the relative ratios of RF expression exert on the efficiency of hiPSC induction. Monitoring of expression of each individual RF in single cells during the course of reprogramming revealed that vector silencing follows acquisition of pluripotent cell markers. Pronounced lentiviral vector silencing was a characteristic of successfully reprogrammed hiPSC clones, but lack of complete silencing did not hinder hiPSC induction, maintenance, or directed differentiation. The vector system described here presents a powerful tool for mechanistic studies of reprogramming and the optimization of hiPSC generation.


Subject(s)
Genes, myc/physiology , Kruppel-Like Transcription Factors/physiology , Octamer Transcription Factor-3/physiology , Pluripotent Stem Cells/cytology , SOXB1 Transcription Factors/physiology , Cell Differentiation , Epigenesis, Genetic , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Octamer Transcription Factor-3/genetics , SOXB1 Transcription Factors/genetics
8.
J Parkinsons Dis ; 11(s2): S135-S140, 2021.
Article in English | MEDLINE | ID: mdl-34250954

ABSTRACT

After many years of preclinical development, cell and gene therapies have advanced from research tools in the lab to clinical-grade products for patients, and today they constitute more than a quarter of all new Phase I clinical trials for Parkinson's disease. Whereas efficacy has been convincingly proven for many of these products in preclinical models, the field is now entering a new phase where the functionality and safety of these products will need to stand the test in clinical trials. If successful, these new products can have the potential to provide patients with a one-time administered treatment which may alleviate them from daily symptomatic dopaminergic medication.


Subject(s)
Parkinson Disease , Genetic Therapy , Humans , Parkinson Disease/drug therapy , Stem Cell Transplantation
9.
Cell Stem Cell ; 28(2): 343-355.e5, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33545081

ABSTRACT

Human pluripotent stem cells show considerable promise for applications in regenerative medicine, including the development of cell replacement paradigms for the treatment of Parkinson's disease. Protocols have been developed to generate authentic midbrain dopamine (mDA) neurons capable of reversing dopamine-related deficits in animal models of Parkinson's disease. However, the generation of mDA neurons at clinical scale suitable for human application remains an important challenge. Here, we present an mDA neuron derivation protocol based on a two-step WNT signaling activation strategy that improves expression of midbrain markers, such as Engrailed-1 (EN1), while minimizing expression of contaminating posterior (hindbrain) and anterior (diencephalic) lineage markers. The resulting neurons exhibit molecular, biochemical, and electrophysiological properties of mDA neurons. Cryopreserved mDA neuron precursors can be successfully transplanted into 6-hydroxydopamine (6OHDA) lesioned rats to induce recovery of amphetamine-induced rotation behavior. The protocol presented here is the basis for clinical-grade mDA neuron production and preclinical safety and efficacy studies.


Subject(s)
Dopaminergic Neurons , Human Embryonic Stem Cells , Animals , Cell Differentiation , Mesencephalon , Rats , Wnt Signaling Pathway
10.
Cancer Res ; 81(19): 4994-5006, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34341072

ABSTRACT

Ewing sarcoma is characterized by pathognomonic translocations, most frequently fusing EWSR1 with FLI1. An estimated 30% of Ewing sarcoma tumors also display genetic alterations in STAG2, TP53, or CDKN2A (SPC). Numerous attempts to develop relevant Ewing sarcoma models from primary human cells have been unsuccessful in faithfully recapitulating the phenotypic, transcriptomic, and epigenetic features of Ewing sarcoma. In this study, by engineering the t(11;22)(q24;q12) translocation together with a combination of SPC mutations, we generated a wide collection of immortalized cells (EWIma cells) tolerating EWSR1-FLI1 expression from primary mesenchymal stem cells (MSC) derived from a patient with Ewing sarcoma. Within this model, SPC alterations strongly favored Ewing sarcoma oncogenicity. Xenograft experiments with independent EWIma cells induced tumors and metastases in mice, which displayed bona fide features of Ewing sarcoma. EWIma cells presented balanced but also more complex translocation profiles mimicking chromoplexy, which is frequently observed in Ewing sarcoma and other cancers. Collectively, these results demonstrate that bone marrow-derived MSCs are a source of origin for Ewing sarcoma and also provide original experimental models to investigate Ewing sarcomagenesis. SIGNIFICANCE: These findings demonstrate that Ewing sarcoma can originate from human bone-marrow-derived mesenchymal stem cells and that recurrent mutations support EWSR1-FLI1 translocation-mediated transformation.


Subject(s)
Cell Transformation, Neoplastic , Disease Susceptibility , Mesenchymal Stem Cells/metabolism , Sarcoma, Ewing/etiology , Sarcoma, Ewing/metabolism , Animals , Biomarkers , CRISPR-Cas Systems , Cells, Cultured , Computational Biology/methods , Disease Models, Animal , Gene Editing , Gene Expression Profiling , Gene Rearrangement , Gene Targeting , Heterografts , Humans , Immunophenotyping , In Situ Hybridization, Fluorescence , Mesenchymal Stem Cells/pathology , Mice , Mutation , Sarcoma, Ewing/pathology , Translocation, Genetic
11.
Cell Stem Cell ; 28(2): 217-229.e7, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33545080

ABSTRACT

Parkinson's disease is characterized by the loss of dopaminergic neurons in the substantia nigra leading to disabling deficits. Dopamine neuron grafts may provide a significant therapeutic advance over current therapies. We have generated midbrain dopamine neurons from human embryonic stem cells and manufactured large-scale cryopreserved dopamine progenitors for clinical use. After optimizing cell survival and phenotypes in short-term studies, the cell product, MSK-DA01, was subjected to an extensive set of biodistribution, toxicity, and tumorigenicity assessments in mice under GLP conditions. A large-scale efficacy study was also performed in rats with the same lot of cells intended for potential human use and demonstrated survival of the grafted cells and behavioral amelioration in 6-hydroxydopamine lesioned rats. There were no adverse effects attributable to the grafted cells, no obvious distribution outside the brain, and no cell overgrowth or tumor formation, thus paving the way for a future clinical trial.


Subject(s)
Dopamine , Human Embryonic Stem Cells , Animals , Cell Differentiation , Dopaminergic Neurons , Mesencephalon , Mice , Rats , Tissue Distribution
12.
Stem Cells ; 27(3): 521-32, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19074416

ABSTRACT

Human embryonic stem cells (hESCs) have enormous potential for applications in basic biology and regenerative medicine. However, harnessing the potential of hESCs toward generating homogeneous populations of specialized cells remains challenging. Here we describe a novel technology for the genetic identification of defined hESC-derived neural cell types using bacterial artificial chromosome (BAC) transgenesis. We generated hESC lines stably expressing Hes5::GFP, Dll1::GFP, and HB9::GFP BACs that yield green fluorescent protein (GFP)(+) neural stem cells, neuroblasts, and motor neurons, respectively. Faithful reporter expression was confirmed by cell fate analysis and appropriate transgene regulation. Prospective isolation of HB9::GFP(+) cells yielded purified human motor neurons with proper marker expression and electrophysiological activity. Global mRNA and microRNA analyses of Hes5::GFP(+) and HB9::GFP(+) populations revealed highly specific expression signatures, suggesting that BAC transgenesis will be a powerful tool for establishing expression libraries that define the human neural lineage and for accessing defined cell types in applications of human disease.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Electrophysiology , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Motor Neurons/cytology , Motor Neurons/metabolism , Receptors, Notch/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism
13.
Cell Stem Cell ; 27(1): 35-49.e6, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32619517

ABSTRACT

Autism is a clinically heterogeneous neurodevelopmental disorder characterized by impaired social interactions, restricted interests, and repetitive behaviors. Despite significant advances in the genetics of autism, understanding how genetic changes perturb brain development and affect clinical symptoms remains elusive. Here, we present a multiplex human pluripotent stem cell (hPSC) platform, in which 30 isogenic disease lines are pooled in a single dish and differentiated into prefrontal cortex (PFC) lineages to efficiently test early-developmental hypotheses of autism. We define subgroups of autism mutations that perturb PFC neurogenesis and are correlated to abnormal WNT/ßcatenin responses. Class 1 mutations (8 of 27) inhibit while class 2 mutations (5 of 27) enhance PFC neurogenesis. Remarkably, autism patient data reveal that individuals carrying subclass-specific mutations differ clinically in their corresponding language acquisition profiles. Our study provides a framework to disentangle genetic heterogeneity associated with autism and points toward converging molecular and developmental pathways of diverse autism-associated mutations.


Subject(s)
Autistic Disorder , Neurodevelopmental Disorders , Pluripotent Stem Cells , Autistic Disorder/genetics , Cell Differentiation/genetics , Humans , Neurogenesis
15.
Nat Commun ; 9(1): 2681, 2018 07 11.
Article in English | MEDLINE | ID: mdl-29992946

ABSTRACT

GLIS3 mutations are associated with type 1, type 2, and neonatal diabetes, reflecting a key function for this gene in pancreatic ß-cell biology. Previous attempts to recapitulate disease-relevant phenotypes in GLIS3-/- ß-like cells have been unsuccessful. Here, we develop a "minimal component" protocol to generate late-stage pancreatic progenitors (PP2) that differentiate to mono-hormonal glucose-responding ß-like (PP2-ß) cells. Using this differentiation platform, we discover that GLIS3-/- hESCs show impaired differentiation, with significant death of PP2 and PP2-ß cells, without impacting the total endocrine pool. Furthermore, we perform a high-content chemical screen and identify a drug candidate that rescues mutant GLIS3-associated ß-cell death both in vitro and in vivo. Finally, we discovered that loss of GLIS3 causes ß-cell death, by activating the TGFß pathway. This study establishes an optimized directed differentiation protocol for modeling human ß-cell disease and identifies a drug candidate for treating a broad range of GLIS3-associated diabetic patients.


Subject(s)
Diabetes Mellitus/prevention & control , Drug Discovery/methods , Hypoglycemic Agents/pharmacology , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Cell Line , DNA-Binding Proteins , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Gene Expression Profiling , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice, SCID , Mutation , Pyrazoles/pharmacology , Quinolines/pharmacology , Repressor Proteins , Trans-Activators , Transcription Factors/metabolism , Transplantation, Heterologous
16.
Nat Commun ; 9(1): 4345, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341316

ABSTRACT

Environmental and genetic risk factors contribute to Parkinson's Disease (PD) pathogenesis and the associated midbrain dopamine (mDA) neuron loss. Here, we identify early PD pathogenic events by developing methodology that utilizes recent innovations in human pluripotent stem cells (hPSC) and chemical sensors of HSP90-incorporating chaperome networks. We show that events triggered by PD-related genetic or toxic stimuli alter the neuronal proteome, thereby altering the stress-specific chaperome networks, which produce changes detected by chemical sensors. Through this method we identify STAT3 and NF-κB signaling activation as examples of genetic stress, and phospho-tyrosine hydroxylase (TH) activation as an example of toxic stress-induced pathways in PD neurons. Importantly, pharmacological inhibition of the stress chaperome network reversed abnormal phospho-STAT3 signaling and phospho-TH-related dopamine levels and rescued PD neuron viability. The use of chemical sensors of chaperome networks on hPSC-derived lineages may present a general strategy to identify molecular events associated with neurodegenerative diseases.


Subject(s)
Dopaminergic Neurons/metabolism , HSP90 Heat-Shock Proteins/metabolism , Mesencephalon/metabolism , Biosensing Techniques , HSP90 Heat-Shock Proteins/physiology , Mesencephalon/pathology , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Stress, Physiological
17.
Mol Ther Methods Clin Dev ; 4: 72-82, 2017 Mar 17.
Article in English | MEDLINE | ID: mdl-28344993

ABSTRACT

Cell replacement therapy in the nervous system has a rich history, with ∼40 years of research and ∼30 years of clinical experience. There is compelling evidence that appropriate cells can integrate and function in the dysfunctioning human nervous system, but the clinical results are mixed in practice. A number of factors conspire to vary patient outcome: the indication, cell source, patient selection, and team performing transplantation are all variables that can affect efficacy. Most early clinical trials have used fetal cells, a limited cell source that resists scale and standardization. Direct fetal cell transplantation creates significant challenges to commercialization that is the ultimate goal of an effective cell therapy. One approach to help scale and standardize fetal cell preparations is the expansion of neural cells in vitro. Expansion is achieved by transformation or through the application of mitogens before cryopreservation. Recently, neural cells derived from pluripotent stem cells have provided a scalable alternative. Pluripotent stem cells are desirable for manufacturing but present alternative concerns and manufacturing obstacles. All cell sources require robust and reproducible manufacturing to make nervous system cell replacement therapy an option for patients. Here, we discuss the challenges and opportunities for cell replacement in the nervous system. In this review, we give an overview of completed and ongoing neural cell transplantation clinical trials, and we discuss the challenges and opportunities for future cell replacement trials with a particular focus on pluripotent stem cell-derived therapies.

18.
Cell Stem Cell ; 21(2): 274-283.e5, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28736217

ABSTRACT

Zika virus (ZIKV) infects fetal and adult human brain and is associated with serious neurological complications. To date, no therapeutic treatment is available to treat ZIKV-infected patients. We performed a high-content chemical screen using human pluripotent stem cell-derived cortical neural progenitor cells (hNPCs) and found that hippeastrine hydrobromide (HH) and amodiaquine dihydrochloride dihydrate (AQ) can inhibit ZIKV infection in hNPCs. Further validation showed that HH also rescues ZIKV-induced growth and differentiation defects in hNPCs and human fetal-like forebrain organoids. Finally, HH and AQ inhibit ZIKV infection in adult mouse brain in vivo. Strikingly, HH suppresses viral propagation when administered to adult mice with active ZIKV infection, highlighting its therapeutic potential. Our approach highlights the power of stem cell-based screens and validation in human forebrain organoids and mouse models in identifying drug candidates for treating ZIKV infection and related neurological complications in fetal and adult patients.


Subject(s)
Antiviral Agents/therapeutic use , Brain/virology , Drug Evaluation, Preclinical/methods , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Organoids/virology , Zika Virus Infection/drug therapy , Zika Virus/physiology , Adolescent , Amaryllidaceae Alkaloids/pharmacology , Amodiaquine/pharmacology , Animals , Antiviral Agents/pharmacology , Cell Line , Child , Female , Fetus/drug effects , Fetus/virology , Humans , Induced Pluripotent Stem Cells/drug effects , Mice, SCID , Neural Stem Cells/drug effects , Organoids/drug effects , Zika Virus/drug effects , Zika Virus Infection/pathology
19.
Stem Cell Reports ; 9(1): 355-365, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28602613

ABSTRACT

Human pluripotent stem cells (PSCs) provide an unlimited cell source for cell therapies and disease modeling. Despite their enormous power, technical aspects have hampered reproducibility. Here, we describe a modification of PSC workflows that eliminates a major variable for nearly all PSC experiments: the quality and quantity of the PSC starting material. Most labs continually passage PSCs and use small quantities after expansion, but the "just-in-time" nature of these experiments means that quality control rarely happens before use. Lack of quality control could compromise PSC quality, sterility, and genetic integrity, which creates a variable that might affect results. This method, called CryoPause, banks PSCs as single-use, cryopreserved vials that can be thawed and immediately used in experiments. Each CryoPause bank provides a consistent source of PSCs that can be pre-validated before use to reduce the possibility that high levels of spontaneous differentiation, contamination, or genetic integrity will compromise an experiment.


Subject(s)
Cryopreservation/methods , Pluripotent Stem Cells/cytology , Animals , Biological Specimen Banks , Cell Differentiation , Cell Line , Cell- and Tissue-Based Therapy , Gene Editing , Humans , Mice , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/transplantation
20.
Nat Biotechnol ; 35(2): 154-163, 2017 02.
Article in English | MEDLINE | ID: mdl-28112759

ABSTRACT

Considerable progress has been made in converting human pluripotent stem cells (hPSCs) into functional neurons. However, the protracted timing of human neuron specification and functional maturation remains a key challenge that hampers the routine application of hPSC-derived lineages in disease modeling and regenerative medicine. Using a combinatorial small-molecule screen, we previously identified conditions to rapidly differentiate hPSCs into peripheral sensory neurons. Here we generalize the approach to central nervous system (CNS) fates by developing a small-molecule approach for accelerated induction of early-born cortical neurons. Combinatorial application of six pathway inhibitors induces post-mitotic cortical neurons with functional electrophysiological properties by day 16 of differentiation, in the absence of glial cell co-culture. The resulting neurons, transplanted at 8 d of differentiation into the postnatal mouse cortex, are functional and establish long-distance projections, as shown using iDISCO whole-brain imaging. Accelerated differentiation into cortical neuron fates should facilitate hPSC-based strategies for disease modeling and cell therapy in CNS disorders.


Subject(s)
Cell Differentiation/physiology , Central Nervous System Agents/administration & dosage , Neurons/cytology , Neurons/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Batch Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Pluripotent Stem Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL