Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
2.
Angiogenesis ; 27(3): 523-542, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38771392

ABSTRACT

Induced pluripotent stem cell (iPSC) derived endothelial cells (iECs) have emerged as a promising tool for studying vascular biology and providing a platform for modelling various vascular diseases, including those with genetic origins. Currently, primary ECs are the main source for disease modelling in this field. However, they are difficult to edit and have a limited lifespan. To study the effects of targeted mutations on an endogenous level, we generated and characterized an iPSC derived model for venous malformations (VMs). CRISPR-Cas9 technology was used to generate a novel human iPSC line with an amino acid substitution L914F in the TIE2 receptor, known to cause VMs. This enabled us to study the differential effects of VM causative mutations in iECs in multiple in vitro models and assess their ability to form vessels in vivo. The analysis of TIE2 expression levels in TIE2L914F iECs showed a significantly lower expression of TIE2 on mRNA and protein level, which has not been observed before due to a lack of models with endogenous edited TIE2L914F and sparse patient data. Interestingly, the TIE2 pathway was still significantly upregulated and TIE2 showed high levels of phosphorylation. TIE2L914F iECs exhibited dysregulated angiogenesis markers and upregulated migration capability, while proliferation was not affected. Under shear stress TIE2L914F iECs showed reduced alignment in the flow direction and a larger cell area than TIE2WT iECs. In summary, we developed a novel TIE2L914F iPSC-derived iEC model and characterized it in multiple in vitro models. The model can be used in future work for drug screening for novel treatments for VMs.


Subject(s)
Endothelial Cells , Gene Knock-In Techniques , Induced Pluripotent Stem Cells , Receptor, TIE-2 , Humans , Induced Pluripotent Stem Cells/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Endothelial Cells/metabolism , Mutation/genetics , CRISPR-Cas Systems/genetics , Vascular Malformations/genetics , Vascular Malformations/pathology , Vascular Malformations/metabolism
3.
NPJ Parkinsons Dis ; 9(1): 24, 2023 Feb 11.
Article in English | MEDLINE | ID: mdl-36774388

ABSTRACT

The pathogenesis and clinical heterogeneity of Parkinson's disease (PD) have been evaluated from molecular, pathophysiological, and clinical perspectives. High-throughput proteomic analysis of cerebrospinal fluid (CSF) opened new opportunities for scrutinizing this heterogeneity. To date, this is the most comprehensive CSF-based proteomics profiling study in PD with 569 patients (350 idiopathic patients, 65 GBA + mutation carriers and 154 LRRK2 + mutation carriers), 534 controls, and 4135 proteins analyzed. Combining CSF aptamer-based proteomics with genetics we determined protein quantitative trait loci (pQTLs). Analyses of pQTLs together with summary statistics from the largest PD genome wide association study (GWAS) identified 68 potential causal proteins by Mendelian randomization. The top causal protein, GPNMB, was previously reported to be upregulated in the substantia nigra of PD patients. We also compared the CSF proteomes of patients and controls. Proteome differences between GBA + patients and unaffected GBA + controls suggest degeneration of dopaminergic neurons, altered dopamine metabolism and increased brain inflammation. In the LRRK2 + subcohort we found dysregulated lysosomal degradation, altered alpha-synuclein processing, and neurotransmission. Proteome differences between idiopathic patients and controls suggest increased neuroinflammation, mitochondrial dysfunction/oxidative stress, altered iron metabolism and potential neuroprotection mediated by vasoactive substances. Finally, we used proteomic data to stratify idiopathic patients into "endotypes". The identified endotypes show differences in cognitive and motor disease progression based on previously reported protein-based risk scores.Our findings not only contribute to the identification of new therapeutic targets but also to shape personalized medicine in CNS neurodegeneration.

4.
Sci Adv ; 8(46): eadd5430, 2022 11 18.
Article in English | MEDLINE | ID: mdl-36383675

ABSTRACT

We performed collapsing analyses on 454,796 UK Biobank (UKB) exomes to detect gene-level associations with diabetes. Recessive carriers of nonsynonymous variants in MAP3K15 were 30% less likely to develop diabetes (P = 5.7 × 10-10) and had lower glycosylated hemoglobin (ß = -0.14 SD units, P = 1.1 × 10-24). These associations were independent of body mass index, suggesting protection against insulin resistance even in the setting of obesity. We replicated these findings in 96,811 Admixed Americans in the Mexico City Prospective Study (P < 0.05)Moreover, the protective effect of MAP3K15 variants was stronger in individuals who did not carry the Latino-enriched SLC16A11 risk haplotype (P = 6.0 × 10-4). Separately, we identified a Finnish-enriched MAP3K15 protein-truncating variant associated with decreased odds of both type 1 and type 2 diabetes (P < 0.05) in FinnGen. No adverse phenotypes were associated with protein-truncating MAP3K15 variants in the UKB, supporting this gene as a therapeutic target for diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , MAP Kinase Kinase Kinases , Humans , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Monocarboxylic Acid Transporters/genetics , Obesity/genetics , Prospective Studies , MAP Kinase Kinase Kinases/genetics
5.
Genome Biol ; 20(1): 142, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31315641

ABSTRACT

We develop CellSIUS (Cell Subtype Identification from Upregulated gene Sets) to fill a methodology gap for rare cell population identification for scRNA-seq data. CellSIUS outperforms existing algorithms for specificity and selectivity for rare cell types and their transcriptomic signature identification in synthetic and complex biological data. Characterization of a human pluripotent cell differentiation protocol recapitulating deep-layer corticogenesis using CellSIUS reveals unrecognized complexity in human stem cell-derived cellular populations. CellSIUS enables identification of novel rare cell populations and their signature genes providing the means to study those populations in vitro in light of their role in health and disease.


Subject(s)
Single-Cell Analysis/methods , Transcriptome , Algorithms , Cell Line , Humans , Neurons/cytology
6.
Cell Stem Cell ; 19(6): 703-708, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27912091

ABSTRACT

Zika virus (ZIKV) can cross the placental barrier, resulting in infection of the fetal brain and neurological defects including microcephaly. The cellular tropism of ZIKV and the identity of attachment factors used by the virus to gain access to key cell types involved in pathogenesis are under intense investigation. Initial studies suggested that ZIKV preferentially targets neural progenitor cells (NPCs), providing an explanation for the developmental phenotypes observed in some pregnancies. The AXL protein has been nominated as a key attachment factor for ZIKV in several cell types including NPCs. However, here we show that genetic ablation of AXL has no effect on ZIKV entry or ZIKV-mediated cell death in human induced pluripotent stem cell (iPSC)-derived NPCs or cerebral organoids. These findings call into question the utility of AXL inhibitors for preventing birth defects after infection and suggest that further studies of viral attachment factors in NPCs are needed.


Subject(s)
Cerebrum/pathology , Gene Deletion , Neural Stem Cells/metabolism , Neural Stem Cells/virology , Neuroprotection , Organoids/virology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Zika Virus Infection/prevention & control , Cell Death , Gene Knockout Techniques , Humans , Neural Stem Cells/pathology , Organoids/metabolism , Organoids/pathology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Zika Virus Infection/pathology , Axl Receptor Tyrosine Kinase
7.
Elife ; 52016 10 01.
Article in English | MEDLINE | ID: mdl-27692071

ABSTRACT

Evolutionary differences in gene regulation between humans and lower mammalian experimental systems are incompletely understood, a potential translational obstacle that is challenging to surmount in neurons, where primary tissue availability is poor. Rodent-based studies show that activity-dependent transcriptional programs mediate myriad functions in neuronal development, but the extent of their conservation in human neurons is unknown. We compared activity-dependent transcriptional responses in developing human stem cell-derived cortical neurons with those induced in developing primary- or stem cell-derived mouse cortical neurons. While activity-dependent gene-responsiveness showed little dependence on developmental stage or origin (primary tissue vs. stem cell), notable species-dependent differences were observed. Moreover, differential species-specific gene ortholog regulation was recapitulated in aneuploid mouse neurons carrying human chromosome-21, implicating promoter/enhancer sequence divergence as a factor, including human-specific activity-responsive AP-1 sites. These findings support the use of human neuronal systems for probing transcriptional responses to physiological stimuli or indeed pharmaceutical agents.


Subject(s)
Biological Evolution , Gene Expression Regulation, Developmental , Neural Stem Cells/physiology , Neurons/physiology , Transcription, Genetic , Animals , Cells, Cultured , Humans , Mice
8.
Nat Commun ; 6: 7066, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25967870

ABSTRACT

Forebrain neurons have weak intrinsic antioxidant defences compared with astrocytes, but the molecular basis and purpose of this is poorly understood. We show that early in mouse cortical neuronal development in vitro and in vivo, expression of the master-regulator of antioxidant genes, transcription factor NF-E2-related-factor-2 (Nrf2), is repressed by epigenetic inactivation of its promoter. Consequently, in contrast to astrocytes or young neurons, maturing neurons possess negligible Nrf2-dependent antioxidant defences, and exhibit no transcriptional responses to Nrf2 activators, or to ablation of Nrf2's inhibitor Keap1. Neuronal Nrf2 inactivation seems to be required for proper development: in maturing neurons, ectopic Nrf2 expression inhibits neurite outgrowth and aborization, and electrophysiological maturation, including synaptogenesis. These defects arise because Nrf2 activity buffers neuronal redox status, inhibiting maturation processes dependent on redox-sensitive JNK and Wnt pathways. Thus, developmental epigenetic Nrf2 repression weakens neuronal antioxidant defences but is necessary to create an environment that supports neuronal development.


Subject(s)
Antioxidants/metabolism , Cerebral Cortex/cytology , Epigenesis, Genetic/physiology , Gene Expression Regulation, Developmental/physiology , NF-E2-Related Factor 2/metabolism , Neurons/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cerebral Cortex/embryology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Electrophysiological Phenomena , Kelch-Like ECH-Associated Protein 1 , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics
9.
J Physiol ; 592(19): 4353-63, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25172951

ABSTRACT

We have assessed, using whole-cell patch-clamp recording and RNA-sequencing (RNA-seq), the properties and composition of GABAA receptors (GABAARs) and strychnine-sensitive glycine receptors (GlyRs) expressed by excitatory cortical neurons derived from human embryonic stem cells (hECNs). The agonists GABA and muscimol gave EC50 values of 278 µm and 182 µm, respectively, and the presence of a GABAAR population displaying low agonist potencies is supported by strong RNA-seq signals for α2 and α3 subunits. GABAAR-mediated currents, evoked by EC50 concentrations of GABA, were blocked by bicuculline and picrotoxin with IC50 values of 2.7 and 5.1 µm, respectively. hECN GABAARs are predominantly γ subunit-containing as assessed by the sensitivity of GABA-evoked currents to diazepam and insensitivity to Zn(2+), together with the weak direct agonist action of gaboxadol; RNA-seq indicated a predominant expression of the γ2 subunit. Potentiation of GABA-evoked currents by propofol and etomidate and the lack of inhibition of currents by salicylidine salycylhydrazide (SCS) indicate expression of the ß2 or ß3 subunit, with RNA-seq analysis indicating strong expression of ß3 in hECN GABAARs. Taken together our data support the notion that hECN GABAARs have an α2/3ß3γ2 subunit composition - a composition that also predominates in immature rodent cortex. GlyRs expressed by hECNs were activated by glycine with an EC50 of 167 µm. Glycine-evoked (500 µm) currents were blocked by strychnine (IC50 = 630 nm) and picrotoxin (IC50 = 197 µm), where the latter is suggestive of a population of heteromeric receptors. RNA-seq indicates GlyRs are likely to be composed of α2 and ß subunits.


Subject(s)
Cerebral Cortex/metabolism , Neurons/metabolism , Pluripotent Stem Cells/metabolism , Protein Subunits/metabolism , Receptors, GABA-A/metabolism , Receptors, Glycine/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Humans , Muscimol/pharmacology , Neurons/cytology , Neurons/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , gamma-Aminobutyric Acid/pharmacology
10.
Nat Chem Biol ; 10(8): 677-85, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24974230

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) have distinct clinical features but a common pathology--cytoplasmic inclusions rich in transactive response element DNA-binding protein of 43 kDa (TDP43). Rare TDP43 mutations cause ALS or FTD, but abnormal TDP43 levels and localization may cause disease even if TDP43 lacks a mutation. Here we show that individual neurons vary in their ability to clear TDP43 and are exquisitely sensitive to TDP43 levels. To measure TDP43 clearance, we developed and validated a single-cell optical method that overcomes the confounding effects of aggregation and toxicity and discovered that pathogenic mutations shorten TDP43 half-life. New compounds that stimulate autophagy improved TDP43 clearance and localization and enhanced survival in primary murine neurons and in human stem cell-derived neurons and astrocytes harboring mutant TDP43. These findings indicate that the levels and localization of TDP43 critically determine neurotoxicity and show that autophagy induction mitigates neurodegeneration by acting directly on TDP43 clearance.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Autophagy , DNA-Binding Proteins/metabolism , Neurons/metabolism , Amino Acid Sequence , Amyotrophic Lateral Sclerosis/pathology , Animals , Astrocytes/metabolism , Autophagy/drug effects , Cell Survival , Cells, Cultured , DNA-Binding Proteins/genetics , Fluphenazine/pharmacology , Half-Life , Humans , Induced Pluripotent Stem Cells/physiology , Methotrimeprazine/pharmacology , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Mutation , Rats , Reproducibility of Results , Single-Cell Analysis/methods , Small Molecule Libraries/pharmacology , Stem Cells/metabolism
11.
J Neurosci ; 34(11): 4070-5, 2014 Mar 12.
Article in English | MEDLINE | ID: mdl-24623784

ABSTRACT

Rodent-based studies have shown that neurons undergo major developmental changes to ion channel expression and ionic gradients that determine their excitation-inhibition balance. Neurons derived from human pluripotent stem cells theoretically offer the potential to study classical developmental processes in a human-relevant system, although this is currently not well explored. Here, we show that excitatory cortical-patterned neurons derived from multiple human pluripotent stem cell lines exhibit native-like maturation changes in AMPAR composition such that there is an increase in the expression of GluA2(R) subunits. Moreover, we observe a dynamic shift in intracellular Cl- levels, which determines the reversal potential of GABAAR-mediated currents and is influenced by neurotrophic factors. The shift is concomitant with changes in KCC2 and NKCC1 expression. Because some human diseases are thought to involve perturbations to AMPAR GluA2 content and others in the chloride reversal potential, human stem-cell-derived neurons represent a valuable tool for studying these fundamental properties.


Subject(s)
Cerebral Cortex/cytology , Neurons/cytology , Neurons/physiology , Pluripotent Stem Cells/cytology , Receptors, AMPA/physiology , Receptors, GABA-A/physiology , Cell Differentiation/physiology , Cell Line , Embryonic Stem Cells/cytology , Excitatory Postsynaptic Potentials/physiology , Female , Humans , Male , Patch-Clamp Techniques , Receptors, AMPA/genetics , Receptors, GABA-A/genetics , Solute Carrier Family 12, Member 2/genetics , Solute Carrier Family 12, Member 2/physiology , Symporters/genetics , Symporters/physiology , K Cl- Cotransporters
12.
PLoS One ; 9(3): e91269, 2014.
Article in English | MEDLINE | ID: mdl-24651281

ABSTRACT

TDP-43 is found in cytoplasmic inclusions in 95% of amyotrophic lateral sclerosis (ALS) and 60% of frontotemporal lobar degeneration (FTLD). Approximately 4% of familial ALS is caused by mutations in TDP-43. The majority of these mutations are found in the glycine-rich domain, including the variant M337V, which is one of the most common mutations in TDP-43. In order to investigate the use of allele-specific RNA interference (RNAi) as a potential therapeutic tool, we designed and screened a set of siRNAs that specifically target TDP-43(M337V) mutation. Two siRNA specifically silenced the M337V mutation in HEK293T cells transfected with GFP-TDP-43(wt) or GFP-TDP-43(M337V) or TDP-43 C-terminal fragments counterparts. C-terminal TDP-43 transfected cells show an increase of cytosolic inclusions, which are decreased after allele-specific siRNA in M337V cells. We then investigated the effects of one of these allele-specific siRNAs in induced pluripotent stem cells (iPSCs) derived from an ALS patient carrying the M337V mutation. These lines showed a two-fold increase in cytosolic TDP-43 compared to the control. Following transfection with the allele-specific siRNA, cytosolic TDP-43 was reduced by 30% compared to cells transfected with a scrambled siRNA. We conclude that RNA interference can be used to selectively target the TDP-43(M337V) allele in mammalian and patient cells, thus demonstrating the potential for using RNA interference as a therapeutic tool for ALS.


Subject(s)
Alleles , Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Induced Pluripotent Stem Cells/pathology , Mutation/genetics , Neural Stem Cells/metabolism , Amino Acid Substitution/genetics , Base Sequence , HEK293 Cells , Humans , Inclusion Bodies/metabolism , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism
13.
Neuron ; 81(3): 536-543, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24507191

ABSTRACT

The RNA-binding protein TDP-43 regulates RNA metabolism at multiple levels, including transcription, RNA splicing, and mRNA stability. TDP-43 is a major component of the cytoplasmic inclusions characteristic of amyotrophic lateral sclerosis and some types of frontotemporal lobar degeneration. The importance of TDP-43 in disease is underscored by the fact that dominant missense mutations are sufficient to cause disease, although the role of TDP-43 in pathogenesis is unknown. Here we show that TDP-43 forms cytoplasmic mRNP granules that undergo bidirectional, microtubule-dependent transport in neurons in vitro and in vivo and facilitate delivery of target mRNA to distal neuronal compartments. TDP-43 mutations impair this mRNA transport function in vivo and in vitro, including in stem cell-derived motor neurons from ALS patients bearing any one of three different TDP-43 ALS-causing mutations. Thus, TDP-43 mutations that cause ALS lead to partial loss of a novel cytoplasmic function of TDP-43.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Axonal Transport/genetics , DNA-Binding Proteins/genetics , Motor Neurons/metabolism , Mutation/genetics , RNA, Messenger/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Animals, Genetically Modified , Cells, Cultured , Cerebral Cortex/cytology , Drosophila , Drosophila Proteins/genetics , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Luminescent Proteins/genetics , Mice , Mitochondria/metabolism , Motor Neurons/ultrastructure , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
14.
PLoS One ; 9(1): e85932, 2014.
Article in English | MEDLINE | ID: mdl-24465796

ABSTRACT

Widespread use of human pluripotent stem cells (hPSCs) to study neuronal physiology and function is hindered by the ongoing need for specialist expertise in converting hPSCs to neural precursor cells (NPCs). Here, we describe a new methodology to generate cryo-preservable hPSC-derived NPCs that retain an anterior identity and are propagatable long-term prior to terminal differentiation, thus abrogating regular de novo neuralization. Key to achieving passagable NPCs without loss of identity is the combination of both absence of EGF and propagation in physiological levels (3%) of O2. NPCs generated in this way display a stable long-term anterior forebrain identity and importantly retain developmental competence to patterning signals. Moreover, compared to NPCs maintained at ambient O2 (21%), they exhibit enhanced uniformity and speed of functional maturation, yielding both deep and upper layer cortical excitatory neurons. These neurons display multiple attributes including the capability to form functional synapses and undergo activity-dependent gene regulation. The platform described achieves long-term maintenance of anterior neural precursors that can give rise to forebrain neurones in abundance, enabling standardised functional studies of neural stem cell maintenance, lineage choice and neuronal functional maturation for neurodevelopmental research and disease-modelling.


Subject(s)
Epidermal Growth Factor/metabolism , Neural Stem Cells/cytology , Oxygen/pharmacology , Pluripotent Stem Cells/cytology , Cell Differentiation/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Fibroblast Growth Factor 2/metabolism , Humans , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Signal Transduction/drug effects , Synapses/drug effects , Synapses/metabolism , Time Factors
15.
Stem Cell Reports ; 1(5): 437-50, 2013.
Article in English | MEDLINE | ID: mdl-24286031

ABSTRACT

We have established and efficient system to specify NG2/PDGF-Rα/OLIG2+ oligodendrocyte precursor cells (OPCs) from human embryonic stem cells (hESCs) at low, physiological (3%) oxygen levels. This was achieved via both forebrain and spinal cord origins, with up to 98% of cells expressing NG2. Developmental insights reveal a critical role for fibroblast growth factor 2 (FGF-2) in OLIG2 induction via ventral forebrain pathways. The OPCs mature in vitro to express O4 (46%) and subsequently become galactocerebroside (GALC), O1, and myelin basic protein-positive (MBP+) multibranching oligodendrocytes. These were cultured alongside hESC-derived neurons. The electrophysiological properties of human OPCs are similar to those of rat OPCs, with large voltage-gated sodium currents and the ability to fire action potentials. Exposure to a selective retinoid X receptor agonist increased the proportion of O4+ oligodendrocytes that express MBP from 5% to 30%. Thus, we have established a developmentally engineered system to investigate the biological properties of human OPCs and test the effects of putative remyelinating agents prior to clinical application.


Subject(s)
Cell Lineage , Embryonic Stem Cells/cytology , Neural Stem Cells/cytology , Neurogenesis , Oligodendroglia/cytology , Oxygen/pharmacology , Action Potentials , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fibroblast Growth Factor 2/pharmacology , Galactosylceramides/metabolism , Humans , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/physiology , Prosencephalon/cytology , Retinoid X Receptors/antagonists & inhibitors , Sodium/metabolism , Spinal Cord/cytology
17.
Sci Transl Med ; 5(188): 188le2, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23740897

ABSTRACT

Egawa et al. recently showed the value of patient-specific induced pluripotent stem cells (iPSCs) for modeling amyotrophic lateral sclerosis in vitro. Their study and our work highlight the need for complementary assays to detect small, but potentially important, phenotypic differences between control iPSC lines and those carrying disease mutations.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Drug Evaluation, Preclinical/methods , Induced Pluripotent Stem Cells/cytology , Motor Neurons/cytology , Humans
18.
Stem Cells Transl Med ; 2(6): 464-72, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23677643

ABSTRACT

Traditionally, in vitro stem cell systems have used oxygen tensions that are far removed from the in vivo situation. This is particularly true for the central nervous system, where oxygen (O2) levels range from 8% at the pia to 0.5% in the midbrain, whereas cells are usually cultured in a 20% O2 environment. Cell transplantation strategies therefore typically introduce a stress challenge at the time of transplantation as the cells are switched from 20% to 3% O2 (the average in adult organs). We have modeled the oxygen stress that occurs during transplantation, demonstrating that in vitro transfer of neonatal rat cortical neural precursor cells (NPCs) from a 20% to a 3% O2 environment results in significant cell death, whereas maintenance at 3% O2 is protective. This survival benefit translates to the in vivo environment, where culture of NPCs at 3% rather than 20% O2 approximately doubles survival in the immediate post-transplantation phase. Furthermore, NPC fate is affected by culture at low, physiological O2 tensions (3%), with particularly marked effects on the oligodendrocyte lineage, both in vitro and in vivo. We propose that careful consideration of physiological oxygen environments, and particularly changes in oxygen tension, has relevance for the practical approaches to cellular therapies.


Subject(s)
Hyperoxia/pathology , Neural Stem Cells/drug effects , Oligodendroglia/drug effects , Oxygen/pharmacology , Stem Cell Transplantation/methods , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Survival/drug effects , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Gene Expression/drug effects , Graft Survival/drug effects , Hippocampus , Hyperoxia/physiopathology , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Oxidative Stress , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Tubulin/genetics , Tubulin/metabolism
19.
Proc Natl Acad Sci U S A ; 110(12): 4697-702, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23401527

ABSTRACT

Glial proliferation and activation are associated with disease progression in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar dementia. In this study, we describe a unique platform to address the question of cell autonomy in transactive response DNA-binding protein (TDP-43) proteinopathies. We generated functional astroglia from human induced pluripotent stem cells carrying an ALS-causing TDP-43 mutation and show that mutant astrocytes exhibit increased levels of TDP-43, subcellular mislocalization of TDP-43, and decreased cell survival. We then performed coculture experiments to evaluate the effects of M337V astrocytes on the survival of wild-type and M337V TDP-43 motor neurons, showing that mutant TDP-43 astrocytes do not adversely affect survival of cocultured neurons. These observations reveal a significant and previously unrecognized glial cell-autonomous pathological phenotype associated with a pathogenic mutation in TDP-43 and show that TDP-43 proteinopathies do not display an astrocyte non-cell-autonomous component in cell culture, as previously described for SOD1 ALS. This study highlights the utility of induced pluripotent stem cell-based in vitro disease models to investigate mechanisms of disease in ALS and other TDP-43 proteinopathies.


Subject(s)
Amyotrophic Lateral Sclerosis , Astrocytes , Induced Pluripotent Stem Cells , Motor Neurons , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Astrocytes/metabolism , Astrocytes/pathology , Cell Line , Cell Proliferation , Cell Survival , Coculture Techniques , DNA-Binding Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Male , Middle Aged , Motor Neurons/metabolism , Motor Neurons/pathology , Mutation
20.
Proc Natl Acad Sci U S A ; 109(15): 5803-8, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-22451909

ABSTRACT

Transactive response DNA-binding (TDP-43) protein is the dominant disease protein in amyotrophic lateral sclerosis (ALS) and a subgroup of frontotemporal lobar degeneration (FTLD-TDP). Identification of mutations in the gene encoding TDP-43 (TARDBP) in familial ALS confirms a mechanistic link between misaccumulation of TDP-43 and neurodegeneration and provides an opportunity to study TDP-43 proteinopathies in human neurons generated from patient fibroblasts by using induced pluripotent stem cells (iPSCs). Here, we report the generation of iPSCs that carry the TDP-43 M337V mutation and their differentiation into neurons and functional motor neurons. Mutant neurons had elevated levels of soluble and detergent-resistant TDP-43 protein, decreased survival in longitudinal studies, and increased vulnerability to antagonism of the PI3K pathway. We conclude that expression of physiological levels of TDP-43 in human neurons is sufficient to reveal a mutation-specific cell-autonomous phenotype and strongly supports this approach for the study of disease mechanisms and for drug screening.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Motor Neurons/pathology , Mutation/genetics , TDP-43 Proteinopathies/genetics , Adult , Cell Differentiation/drug effects , DNA-Binding Proteins/metabolism , Detergents/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Induced Pluripotent Stem Cells/drug effects , Male , Middle Aged , Motor Neurons/drug effects , Motor Neurons/metabolism , Organ Specificity/drug effects , Solubility/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL