Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 7: 42991, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28256506

ABSTRACT

Astroglial pathology is seen in various neurodegenerative diseases including frontotemporal dementia (FTD), which can be caused by mutations in the gene encoding the microtubule-associated protein TAU (MAPT). Here, we applied a stem cell model of FTD to examine if FTD astrocytes carry an intrinsic propensity to degeneration and to determine if they can induce non-cell-autonomous effects in neighboring neurons. We utilized CRISPR/Cas9 genome editing in human induced pluripotent stem (iPS) cell-derived neural progenitor cells (NPCs) to repair the FTD-associated N279K MAPT mutation. While astrocytic differentiation was not impaired in FTD NPCs derived from one patient carrying the N279K MAPT mutation, FTD astrocytes appeared larger, expressed increased levels of 4R-TAU isoforms, demonstrated increased vulnerability to oxidative stress and elevated protein ubiquitination and exhibited disease-associated changes in transcriptome profiles when compared to astrocytes derived from one control individual and to the isogenic control. Interestingly, co-culture experiments with FTD astrocytes revealed increased oxidative stress and robust changes in whole genome expression in previously healthy neurons. Our study highlights the utility of iPS cell-derived NPCs to elucidate the role of astrocytes in the pathogenesis of FTD.


Subject(s)
Astrocytes/metabolism , Frontotemporal Dementia/pathology , tau Proteins/genetics , Annexin A2/metabolism , Astrocytes/cytology , Astrocytes/pathology , Cell Differentiation , Coculture Techniques , Frontotemporal Dementia/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Oxidative Stress , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , Transcriptome , Ubiquitination
2.
Proc Natl Acad Sci U S A ; 114(11): E2243-E2252, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28246330

ABSTRACT

Rapid and efficient protocols to generate oligodendrocytes (OL) from human induced pluripotent stem cells (iPSC) are currently lacking, but may be a key technology to understand the biology of myelin diseases and to develop treatments for such disorders. Here, we demonstrate that the induction of three transcription factors (SOX10, OLIG2, NKX6.2) in iPSC-derived neural progenitor cells is sufficient to rapidly generate O4+ OL with an efficiency of up to 70% in 28 d and a global gene-expression profile comparable to primary human OL. We further demonstrate that iPSC-derived OL disperse and myelinate the CNS of Mbpshi/shiRag-/- mice during development and after demyelination, are suitable for in vitro myelination assays, disease modeling, and screening of pharmacological compounds potentially promoting oligodendroglial differentiation. Thus, the strategy presented here to generate OL from iPSC may facilitate the studying of human myelin diseases and the development of high-throughput screening platforms for drug discovery.


Subject(s)
Cell Differentiation/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Transcription Factors/genetics , Animals , Biomarkers , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Cell Death/genetics , Cell Lineage/genetics , Cells, Cultured , Cluster Analysis , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Ectopic Gene Expression , Gene Expression Profiling , Humans , Mice , Mutation , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin Sheath/genetics , Myelin Sheath/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oxidative Stress , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/ultrastructure , Transcription Factors/metabolism , Transcriptome , tau Proteins/genetics , tau Proteins/metabolism
3.
Stem Cell Res ; 16(3): 776-81, 2016 05.
Article in English | MEDLINE | ID: mdl-27153350

ABSTRACT

Reprogramming technology enables the production of neural progenitor cells (NPCs) from somatic cells by direct transdifferentiation. However, little is known on how neural programs in these induced neural stem cells (iNSCs) differ from those of alternative stem cell populations in vitro and in vivo. Here, we performed transcriptome analyses on murine iNSCs in comparison to brain-derived neural stem cells (NSCs) and pluripotent stem cell-derived NPCs, which revealed distinct global, neural, metabolic and cell cycle-associated marks in these populations. iNSCs carried a hindbrain/posterior cell identity, which could be shifted towards caudal, partially to rostral but not towards ventral fates in vitro. iNSCs survived after transplantation into the rodent brain and exhibited in vivo-characteristics, neural and metabolic programs similar to transplanted NSCs. However, iNSCs vastly retained caudal identities demonstrating cell-autonomy of regional programs in vivo. These data could have significant implications for a variety of in vitro- and in vivo-applications using iNSCs.


Subject(s)
Brain/metabolism , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Transcriptome , Animals , Brain/pathology , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Cluster Analysis , Fibroblasts/cytology , Gene Expression Profiling , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Stem Cell Reports ; 5(1): 83-96, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26143746

ABSTRACT

Frontotemporal dementia (FTD) is a frequent form of early-onset dementia and can be caused by mutations in MAPT encoding the microtubule-associated protein TAU. Because of limited availability of neural cells from patients' brains, the underlying mechanisms of neurodegeneration in FTD are poorly understood. Here, we derived induced pluripotent stem cells (iPSCs) from individuals with FTD-associated MAPT mutations and differentiated them into mature neurons. Patient iPSC-derived neurons demonstrated pronounced TAU pathology with increased fragmentation and phospho-TAU immunoreactivity, decreased neurite extension, and increased but reversible oxidative stress response to inhibition of mitochondrial respiration. Furthermore, FTD neurons showed an activation of the unfolded protein response, and a transcriptome analysis demonstrated distinct, disease-associated gene expression profiles. These findings indicate distinct neurodegenerative changes in FTD caused by mutant TAU and highlight the unique opportunity to use neurons differentiated from patient-specific iPSCs to identify potential targets for drug screening purposes and therapeutic intervention.


Subject(s)
Cell Differentiation/genetics , Frontotemporal Dementia/genetics , Induced Pluripotent Stem Cells/pathology , tau Proteins/genetics , Frontotemporal Dementia/pathology , Gene Expression Profiling , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Mutation , Neurites/pathology , Oxidative Stress/genetics , Unfolded Protein Response/genetics , tau Proteins/biosynthesis
5.
Cell Rep ; 8(6): 1697-1703, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25220454

ABSTRACT

The differentiation capability of induced pluripotent stem cells (iPSCs) toward certain cell types for disease modeling and drug screening assays might be influenced by their somatic cell of origin. Here, we have compared the neural induction of human iPSCs generated from fetal neural stem cells (fNSCs), dermal fibroblasts, or cord blood CD34(+) hematopoietic progenitor cells. Neural progenitor cells (NPCs) and neurons could be generated at similar efficiencies from all iPSCs. Transcriptomics analysis of the whole genome and of neural genes revealed a separation of neuroectoderm-derived iPSC-NPCs from mesoderm-derived iPSC-NPCs. Furthermore, we found genes that were similarly expressed in fNSCs and neuroectoderm, but not in mesoderm-derived iPSC-NPCs. Notably, these neural signatures were retained after transplantation into the cortex of mice and paralleled with increased survival of neuroectoderm-derived cells in vivo. These results indicate distinct origin-dependent neural cell identities in differentiated human iPSCs both in vitro and in vivo.


Subject(s)
Brain/metabolism , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Animals , Antigens, CD34/metabolism , Cell Differentiation , Cells, Cultured , Fetal Blood/cytology , Fetal Blood/metabolism , Fetus/cytology , Fibroblasts/cytology , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Mesoderm/cytology , Mice , Mice, Inbred NOD , Microscopy, Confocal , Neural Plate/cytology
6.
Acta Neuropathol ; 127(2): 151-73, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24306942

ABSTRACT

The number of patients with neurodegenerative diseases is increasing significantly worldwide. Thus, intense research is being pursued to uncover mechanisms of disease development in an effort to identify molecular targets for therapeutic intervention. Analysis of postmortem tissue from patients has yielded important histological and biochemical markers of disease progression. However, this approach is inherently limited because it is not possible to study patient neurons prior to degeneration. As such, transgenic and knockout models of neurodegenerative diseases are commonly employed. While these animal models have yielded important insights into some molecular mechanisms of disease development, they do not provide the opportunity to study mechanisms of neurodegeneration in human neurons at risk and thus, it is often difficult or even impossible to replicate human pathogenesis with this approach. The generation of patient-specific induced pluripotent stem (iPS) cells offers a unique opportunity to overcome these obstacles. By expanding and differentiating iPS cells, it is possible to generate large numbers of functional neurons in vitro, which can then be used to study the disease of the donating patient. Here, we provide an overview of human stem cell models of neurodegeneration using iPS cells from patients with Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, frontotemporal dementia, Huntington's disease, spinal muscular atrophy and other neurodegenerative diseases. In addition, we describe how further refinements of reprogramming technology resulted in the generation of patient-specific induced neurons, which have also been used to model neurodegenerative changes in vitro.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Models, Biological , Neurodegenerative Diseases/pathology , Cells, Cultured , Disease Progression , Humans , In Vitro Techniques , Mutation , Neurodegenerative Diseases/genetics , Neurons/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...