Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Neuron ; 98(6): 1155-1169.e6, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29887339

ABSTRACT

Parkinson's disease patients report disturbed sleep patterns long before motor dysfunction. Here, in parkin and pink1 models, we identify circadian rhythm and sleep pattern defects and map these to specific neuropeptidergic neurons in fly models and in hypothalamic neurons differentiated from patient induced pluripotent stem cells (iPSCs). Parkin and Pink1 control the clearance of mitochondria by protein ubiquitination. Although we do not observe major defects in mitochondria of mutant neuropeptidergic neurons, we do find an excess of endoplasmic reticulum-mitochondrial contacts. These excessive contact sites cause abnormal lipid trafficking that depletes phosphatidylserine from the endoplasmic reticulum (ER) and disrupts the production of neuropeptide-containing vesicles. Feeding mutant animals phosphatidylserine rescues neuropeptidergic vesicle production and acutely restores normal sleep patterns in mutant animals. Hence, sleep patterns and circadian disturbances in Parkinson's disease models are explained by excessive ER-mitochondrial contacts, and blocking their formation or increasing phosphatidylserine levels rescues the defects in vivo.


Subject(s)
Endoplasmic Reticulum/metabolism , Hypothalamus/metabolism , Lipid Metabolism , Neurons/metabolism , Parkinson Disease/physiopathology , Phosphatidylserines/metabolism , Sleep Disorders, Circadian Rhythm/physiopathology , Sleep , Animals , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster , Endoplasmic Reticulum/drug effects , Humans , Induced Pluripotent Stem Cells , Mitochondria/metabolism , Neuropeptides/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Phosphatidylserines/pharmacology , Protein Serine-Threonine Kinases/genetics , Sleep/drug effects , Sleep Disorders, Circadian Rhythm/genetics , Sleep Disorders, Circadian Rhythm/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
2.
J Nucl Med ; 58(10): 1659-1665, 2017 10.
Article in English | MEDLINE | ID: mdl-28596158

ABSTRACT

Molecular imaging is indispensable for determining the fate and persistence of engrafted stem cells. Standard strategies for transgene induction involve the use of viral vectors prone to silencing and insertional mutagenesis or the use of nonhuman genes. Methods: We used zinc finger nucleases to induce stable expression of human imaging reporter genes into the safe-harbor locus adeno-associated virus integration site 1 in human embryonic stem cells. Plasmids were generated carrying reporter genes for fluorescence, bioluminescence imaging, and human PET reporter genes. Results: In vitro assays confirmed their functionality, and embryonic stem cells retained differentiation capacity. Teratoma formation assays were performed, and tumors were imaged over time with PET and bioluminescence imaging. Conclusion: This study demonstrates the application of genome editing for targeted integration of human imaging reporter genes in human embryonic stem cells for long-term molecular imaging.


Subject(s)
Embryonic Stem Cells/metabolism , Endoribonucleases/metabolism , Gene Editing , Genes, Reporter/genetics , Genome, Human/genetics , Positron-Emission Tomography , Zinc Fingers , Animals , Cell Differentiation , Cell Line , Endoribonucleases/chemistry , Female , Gene Expression , Humans , Liver/cytology , Mice
3.
J Vis Exp ; (117)2016 11 20.
Article in English | MEDLINE | ID: mdl-27911376

ABSTRACT

Even with the revolution of gene-targeting technologies led by CRISPR-Cas9, genetic modification of human pluripotent stem cells (hPSCs) is still time consuming. Comparative studies that use recombinant lines with transgenes integrated into safe harbor loci could benefit from approaches that use site-specific targeted recombinases, like Cre or FLPe, which are more rapid and less prone to off-target effects. Such methods have been described, although they do not significantly outperform gene targeting in most aspects. Using Zinc-finger nucleases, we previously created a master cell line in the AAVS1 locus of hPSCs that contains a GFP-Hygromycin-tk expressing cassette, flanked by heterotypic FRT sequences. Here, we describe the procedures to perform FLPe recombinase-mediated cassette exchange (RMCE) using this line. The master cell line is transfected with a RMCE donor vector, which contains a promoterless Puromycin resistance, and with FLPe recombinase. Application of both a positive (Puromycin) and negative (FIAU) selection program leads to the selection of RMCE without random integrations. RMCE generates fully characterized pluripotent polyclonal transgenic lines in 15 d with 100% efficiency. Despite the recently described limitations of the AAVS1 locus, the ease of the system paves the way for hPSC transgenesis in isogenic settings, is necessary for comparative studies, and enables semi-high-throughput genetic screens for gain/loss of function analysis that would otherwise be highly time consuming.


Subject(s)
Pluripotent Stem Cells , Recombination, Genetic , Cell Line , Gene Targeting , Humans , Recombinases , Transgenes
4.
Stem Cell Reports ; 4(1): 16-24, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25556567

ABSTRACT

To understand how haploinsufficiency of progranulin (PGRN) causes frontotemporal dementia (FTD), we created induced pluripotent stem cells (iPSCs) from patients carrying the GRN(IVS1+5G > C) mutation (FTD-iPSCs). FTD-iPSCs were fated to cortical neurons, the cells most affected in FTD. Although generation of neuroprogenitors was unaffected, their further differentiation into CTIP2-, FOXP2-, or TBR1-TUJ1 double-positive cortical neurons, but not motorneurons, was significantly decreased in FTD-neural progeny. Zinc finger nuclease-mediated introduction of GRN cDNA into the AAVS1 locus corrected defects in cortical neurogenesis, demonstrating that PGRN haploinsufficiency causes inefficient cortical neuron generation. RNA sequencing analysis confirmed reversal of the altered gene expression profile following genetic correction. We identified the Wnt signaling pathway as one of the top defective pathways in FTD-iPSC-derived neurons, which was reversed following genetic correction. Differentiation of FTD-iPSCs in the presence of a WNT inhibitor mitigated defective corticogenesis. Therefore, we demonstrate that PGRN haploinsufficiency hampers corticogenesis in vitro.


Subject(s)
Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Gene Expression , Induced Pluripotent Stem Cells/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Neurogenesis/genetics , Neurons/metabolism , Biomarkers , Cell Differentiation , Cell Line , Frontotemporal Dementia/therapy , Gene Expression Profiling , Haploinsufficiency , Humans , Induced Pluripotent Stem Cells/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Mutation , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Phenotype , Progranulins , Time Factors , Transcription, Genetic , Transcriptome , Wnt Signaling Pathway
5.
Stem Cell Reports ; 5(5): 918-931, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26455413

ABSTRACT

Tools for rapid and efficient transgenesis in "safe harbor" loci in an isogenic context remain important to exploit the possibilities of human pluripotent stem cells (hPSCs). We created hPSC master cell lines suitable for FLPe recombinase-mediated cassette exchange (RMCE) in the AAVS1 locus that allow generation of transgenic lines within 15 days with 100% efficiency and without random integrations. Using RMCE, we successfully incorporated several transgenes useful for lineage identification, cell toxicity studies, and gene overexpression to study the hepatocyte lineage. However, we observed unexpected and variable transgene expression inhibition in vitro, due to DNA methylation and other unknown mechanisms, both in undifferentiated hESC and differentiating hepatocytes. Therefore, the AAVS1 locus cannot be considered a universally safe harbor locus for reliable transgene expression in vitro, and using it for transgenesis in hPSC will require careful assessment of the function of individual transgenes.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Targeting/methods , Hepatocytes/cytology , Induced Pluripotent Stem Cells/metabolism , Recombinases/metabolism , Transgenes , Cells, Cultured , DNA Methylation , Dependovirus/genetics , Embryonic Stem Cells/cytology , Gene Silencing , Genetic Loci , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Recombinases/genetics
6.
Stem Cells Transl Med ; 3(4): 489-99, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24493854

ABSTRACT

Pancreatic endocrine progenitors obtained from human embryonic stem cells (hESCs) represent a promising source to develop cell-based therapies for diabetes. Although endocrine pancreas progenitor cells have been isolated from mouse pancreata on the basis of Ngn3 expression, human endocrine progenitors have not been isolated yet. As substantial differences exist between human and murine pancreas biology, we investigated whether it is possible to isolate pancreatic endocrine progenitors from differentiating hESC cultures by lineage tracing of NGN3. We targeted the 3' end of NGN3 using zinc finger nuclease-mediated homologous recombination to allow selection of NGN3eGFP(+) cells without disrupting the coding sequence of the gene. Isolated NGN3eGFP(+) cells express PDX1, NKX6.1, and chromogranin A and differentiate in vivo toward insulin, glucagon, and somatostatin single hormone-expressing cells but not to ductal or exocrine pancreatic cells or other endodermal, mesodermal, or ectodermal lineages. This confirms that NGN3(+) cells represent pancreatic endocrine progenitors in humans. In addition, this hESC reporter line constitutes a unique tool that may aid in gaining insight into the developmental mechanisms underlying fate choices in human pancreas and in developing cell-based therapies.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Gene Expression Regulation/physiology , Islets of Langerhans/metabolism , Nerve Tissue Proteins/biosynthesis , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Cell- and Tissue-Based Therapy , Chromogranin A/biosynthesis , Chromogranin A/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Heterografts , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Islets of Langerhans/cytology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Trans-Activators/biosynthesis , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL