Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Neurobiol Dis ; 132: 104581, 2019 12.
Article in English | MEDLINE | ID: mdl-31445161

ABSTRACT

Mutations in parkin, encoded by the PARK2 gene, causes early-onset familial Parkinson's disease (PD), but dysfunctional parkin has also been implicated in sporadic PD. By combining human isogenic induced pluripotent stem cells (iPSCs) with and without PARK2 knockout (KO) and a novel large-scale mass spectrometry based proteomics and post-translational modification (PTM)-omics approach, we have mapped changes in protein profiles and PTMs caused by parkin deficiency in neurons. Our study identifies changes to several proteins previously shown to be dysregulated in brains of sporadic PD patients. Pathway analysis and subsequent in vitro assays reveal perturbations in migration and neurite outgrowth in the PARK2 KO neurons. We confirm the neurite defects using long-term engraftment of neurons in the striatum of immunosuppressed hemiparkinsonian adult rats. The GTP-binding protein RhoA was identified as a key upstream regulator, and RhoA activity was significantly increased in PARK2 KO neurons. By inhibiting RhoA signalling the migration and neurite outgrowth phenotypes could be rescued. Our study provides new insight into the pathogenesis of PD and demonstrates the broadly applicable potential of proteomics and PTMomics for elucidating the role of disease-causing mutations.


Subject(s)
Cell Movement/physiology , Dopaminergic Neurons/metabolism , Neurogenesis/physiology , Parkinson Disease/metabolism , Ubiquitin-Protein Ligases/genetics , rhoA GTP-Binding Protein/metabolism , Animals , Gene Knockout Techniques , Humans , Induced Pluripotent Stem Cells , Mutation , Parkinson Disease/genetics , Rats , Signal Transduction/physiology , Ubiquitin-Protein Ligases/deficiency
2.
Cell Physiol Biochem ; 48(5): 1854-1869, 2018.
Article in English | MEDLINE | ID: mdl-30092592

ABSTRACT

BACKGROUND/AIMS: The aim of the current study was to identify potential prognostic long non-coding RNA (lncRNA) biomarkers for predicting survival in patients with hepatocellular carcinoma (HCC) using The Cancer Genome Atlas (TCGA) dataset and bioinformatics analysis. METHODS: RNA sequencing and clinical data of HCC patients from TCGA were used for prognostic association assessment by univariate Cox analysis. A prognostic signature was built using stepwise multivariable Cox analysis, and a comprehensive analysis was performed to evaluate its prognostic value. The prognostic signature was further evaluated by functional assessment and bioinformatics analysis. RESULTS: Thirteen differentially expressed lncRNAs (DELs) were identified and used to construct a single prognostic signature. Patients with high risk scores showed a significantly increased risk of death (adjusted P < 0.0001, adjusted hazard ratio = 3.522, 95% confidence interval = 2.307-5.376). In the time-dependent receiver operating characteristic analysis, the prognostic signature performed well for HCC survival prediction with an area under curve of 0.809, 0.782 and 0.79 for 1-, 3- and 5-year survival, respectively. Comprehensive survival analysis of the 13-DEL prognostic signature suggested that it serves as an independent factor in HCC, showing a better performance for prognosis prediction than traditional clinical indicators. Functional assessment and bioinformatics analysis suggested that the prognostic signature was associated with the cell cycle and peroxisome proliferator-activated receptor signaling pathway. CONCLUSIONS: The novel lncRNA expression signature identified in the present study may be a potential biomarker for predicting the prognosis of HCC patients.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , RNA, Long Noncoding/metabolism , Aged , Area Under Curve , Carcinoma, Hepatocellular/mortality , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Liver Neoplasms/mortality , Male , Middle Aged , Neoplasm Staging , Peroxisome Proliferator-Activated Receptors/metabolism , Prognosis , Proportional Hazards Models , RNA, Long Noncoding/genetics , ROC Curve , Risk Factors , Signal Transduction
3.
Development ; 142(23): 4010-25, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26483210

ABSTRACT

Mechanisms of initial cell fate decisions differ among species. To gain insights into lineage allocation in humans, we derived ten human embryonic stem cell lines (designated UCSFB1-10) from single blastomeres of four 8-cell embryos and one 12-cell embryo from a single couple. Compared with numerous conventional lines from blastocysts, they had unique gene expression and DNA methylation patterns that were, in part, indicative of trophoblast competence. At a transcriptional level, UCSFB lines from different embryos were often more closely related than those from the same embryo. As predicted by the transcriptomic data, immunolocalization of EOMES, T brachyury, GDF15 and active ß-catenin revealed differential expression among blastomeres of 8- to 10-cell human embryos. The UCSFB lines formed derivatives of the three germ layers and CDX2-positive progeny, from which we derived the first human trophoblast stem cell line. Our data suggest heterogeneity among early-stage blastomeres and that the UCSFB lines have unique properties, indicative of a more immature state than conventional lines.


Subject(s)
Blastomeres/cytology , Embryo Culture Techniques , Embryonic Stem Cells/cytology , Trophoblasts/cytology , Blastocyst/cytology , Cell Differentiation , Cell Line , Cell Lineage , DNA Methylation , Endoderm/metabolism , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Growth Differentiation Factor 15/metabolism , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neural Stem Cells/cytology , Oligonucleotide Array Sequence Analysis , Transcription, Genetic , Transcriptome , beta Catenin/metabolism
4.
Cytotherapy ; 20(6): 861-872, 2018 06.
Article in English | MEDLINE | ID: mdl-29793831

ABSTRACT

BACKGROUND AIMS: We have previously reported the generation of a current Good Manufacture Practice (cGMP)-compliant induced pluripotent stem cell (iPSC) line for clinical applications. Here we show that multiple cellular products currently being considered for therapy can be generated from a single master cell bank of this or any other clinically compliant iPSC line METHODS: Using a stock at passage 20 prepared from the cGMP-compliant working cell bank (WCB), we tested differentiation into therapeutically relevant cell types of the three germ layers using standardized but generic protocols. Cells that we generated include (i) neural stem cells, dopaminergic neurons and astrocytes; (ii) retinal cells (retinal pigment epithelium and photoreceptors); and (iii) hepatocyte, endothelial and mesenchymal cells. To confirm that these generic protocols can also be used for other iPSC lines, we tested the reproducibility of our methodology with a second clinically compliant line RESULTS: Our results confirmed that well-characterized iPSC lines have broad potency, and, despite allelic variability, the same protocols could be used with minimal modifications with multiple qualified lines. In addition, we introduced a constitutively expressed GFP cassette in Chr13 safe harbor site using a standardized previously described method and observed no significant difference in growth and differentiation between the engineered line and the control line indicating that engineered products can be made using a standardized methodology CONCLUSIONS: We believe that our demonstration that multiple products can be made from the same WCB and that the same protocols can be used with multiple lines offers a path to a cost-effective strategy for developing cellular products from iPSC lines.


Subject(s)
Cell Engineering/methods , Cell Engineering/standards , Cell Lineage , Guideline Adherence , Induced Pluripotent Stem Cells/cytology , Astrocytes/cytology , Astrocytes/physiology , Cell Differentiation , Cell Line , Dopaminergic Neurons/cytology , Dopaminergic Neurons/physiology , Endothelial Cells/cytology , Endothelial Cells/physiology , Guideline Adherence/standards , Hepatocytes/cytology , Hepatocytes/physiology , Humans , Induced Pluripotent Stem Cells/physiology , Mesoderm/cytology , Mesoderm/physiology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Practice Guidelines as Topic/standards , Reference Standards , Reproducibility of Results , Retina/cytology , Tissue Banks/standards
5.
Hum Mol Genet ; 24(20): 5775-88, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26220978

ABSTRACT

Gaucher disease (GD) is caused by mutations in the GBA1 gene, which encodes the lysosomal enzyme glucocerebrosidase (GCase). The severe forms of GD are associated with neurodegeneration with either rapid (Type 2) or slow progression (Type 3). Although the neurodegenerative process in GD has been linked to lysosomal dysfunction, the mechanisms involved are largely unknown. To identify the lysosomal alterations in GD neurons and uncover the mechanisms involved, we used induced pluripotent stem cells (iPSCs) derived from patients with GD. In GD iPSC-derived neuronal cells (iPSC-NCs), GBA1 mutations caused widespread lysosomal depletion, and a block in autophagic flux due to defective lysosomal clearance of autophagosomes. Autophagy induction by rapamycin treatment in GD iPSC-NCs led to cell death. Further analysis showed that in GD iPSC-NCs, expression of the transcription factor EB (TFEB), the master regulator of lysosomal genes, and lysosomal gene expression, were significantly downregulated. There was also reduced stability of the TFEB protein and altered lysosomal protein biosynthesis. Treatment of mutant iPSC-NCs with recombinant GCase (rGCase) reverted the lysosomal depletion and autophagy block. The effect of rGCase on restoring lysosomal numbers in mutant cells was enhanced in the presence of overexpressed TFEB, but TFEB overexpression alone did not reverse the lysosomal depletion phenotype. Our results suggest that GBA1 mutations interfere with TFEB-mediated lysosomal biogenesis, and that the action of GCase in maintaining a functioning pool of lysosomes is exerted in part through TFEB. The lysosomal alterations described here are likely to be a major determinant in GBA1-associated neurodegeneration.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gaucher Disease/physiopathology , Glucosylceramidase/genetics , Lysosomes/physiology , Neurons/physiology , Organelle Biogenesis , Cell Differentiation , Gaucher Disease/metabolism , Induced Pluripotent Stem Cells/physiology , Lysosomes/metabolism , Mutation , Neurons/metabolism
6.
Mol Ther ; 23(1): 119-29, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25138755

ABSTRACT

Intracerebral cell transplantation is being pursued as a treatment for many neurological diseases, and effective cell delivery is critical for clinical success. To facilitate intracerebral cell transplantation at the scale and complexity of the human brain, we developed a platform technology that enables radially branched deployment (RBD) of cells to multiple target locations at variable radial distances and depths along the initial brain penetration tract with real-time interventional magnetic resonance image (iMRI) guidance. iMRI-guided RBD functioned as an "add-on" to standard neurosurgical and imaging workflows, and procedures were performed in a commonly available clinical MRI scanner. Multiple deposits of super paramagnetic iron oxide beads were safely delivered to the striatum of live swine, and distribution to the entire putamen was achieved via a single cannula insertion in human cadaveric heads. Human embryonic stem cell-derived dopaminergic neurons were biocompatible with the iMRI-guided RBD platform and successfully delivered with iMRI guidance into the swine striatum. Thus, iMRI-guided RBD overcomes some of the technical limitations inherent to the use of straight cannulas and standard stereotactic targeting. This platform technology could have a major impact on the clinical translation of a wide range of cell therapeutics for the treatment of many neurological diseases.


Subject(s)
Cell Transplantation , Magnetic Resonance Imaging, Interventional/methods , Stereotaxic Techniques/instrumentation , Animals , Cadaver , Catheterization , Corpus Striatum/surgery , Female , Humans , Magnetic Resonance Imaging, Interventional/instrumentation , Putamen/surgery , Swine
7.
Mol Cell Proteomics ; 13(1): 311-28, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24173317

ABSTRACT

Human embryonic stem cells (hESCs) can differentiate into neural stem cells (NSCs), which can further be differentiated into neurons and glia cells. Therefore, these cells have huge potential as source for treatment of neurological diseases. Membrane-associated proteins are very important in cellular signaling and recognition, and their function and activity are frequently regulated by post-translational modifications such as phosphorylation and glycosylation. To obtain information about membrane-associated proteins and their modified amino acids potentially involved in changes of hESCs and NSCs as well as to investigate potential new markers for these two cell stages, we performed large-scale quantitative membrane-proteomic of hESCs and NSCs. This approach employed membrane purification followed by peptide dimethyl labeling and peptide enrichment to study the membrane subproteome as well as changes in phosphorylation and sialylation between hESCs and NSCs. Combining proteomics and modification specific proteomics we identified a total of 5105 proteins whereof 57% contained transmembrane domains or signal peptides. The enrichment strategy yielded a total of 10,087 phosphorylated peptides in which 78% of phosphopeptides were identified with ≥99% confidence in site assignment and 1810 unique formerly sialylated N-linked glycopeptides. Several proteins were identified as significantly regulated in hESCs and NSC, including proteins involved in the early embryonic and neural development. In the latter group of proteins, we could identify potential NSC markers as Crumbs 2 and several novel proteins. A motif analysis of the altered phosphosites showed a sequence consensus motif (R-X-XpS/T) significantly up-regulated in NSC. This motif is among other kinases recognized by the calmodulin-dependent protein kinase-2, emphasizing a possible importance of this kinase for this cell stage. Collectively, this data represent the most diverse set of post-translational modifications reported for hESCs and NSCs. This study revealed potential markers to distinguish NSCs from hESCs and will contribute to improve our understanding on the differentiation process.


Subject(s)
Cell Differentiation/genetics , Membrane Proteins/biosynthesis , Phosphoproteins/biosynthesis , Proteomics , Embryonic Stem Cells/metabolism , Glycopeptides/metabolism , Humans , Membrane Proteins/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction/genetics
8.
Proteomics ; 15(4): 656-74, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25418965

ABSTRACT

Stem cells are unspecialized cells capable of self-renewal and to differentiate into the large variety of cells in the body. The possibility to differentiate these cells into neural precursors and neural cells in vitro provides the opportunity to study neural development, nerve cell biology, neurological disease as well as contributing to clinical research. The neural differentiation process is associated with changes at protein and their post-translational modifications (PTMs). PTMs are important regulators of proteins physicochemical properties, function, activity, and interaction with other proteins, DNA/RNA, and complexes. Moreover, the interplay between PTMs is essential to regulate a range of cellular processes that abnormalities in PTM signaling are associated with several diseases. Altogether, this makes PTMs very relevant to study in order to uncover disease pathogenesis and increase the understanding of molecular processes in cells. Substantial advances in PTM enrichment methods and mass spectrometry has allowed the characterization of a subset of PTMs in large-scale studies. This review focuses on the current state-of-the-art of proteomic, as well as PTMomic studies related to human neural differentiation from pluripotent stem cells. Moreover, some of the challenges in stem cell biology, differentiation, and proteomics/PTMomics that are not exclusive to neural development will be discussed.


Subject(s)
Neurogenesis , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Protein Processing, Post-Translational , Proteomics/methods , Animals , Biomedical Research , Humans
9.
Physiol Genomics ; 47(6): 232-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25852171

ABSTRACT

Reference genes, often referred to as housekeeping genes (HKGs), are frequently used to normalize gene expression data based on the assumption that they are expressed at a constant level in the cells. However, several studies have shown that there may be a large variability in the gene expression levels of HKGs in various cell types. In a previous study, employing human embryonic stem cells (hESCs) subjected to spontaneous differentiation, we observed that the expression of commonly used HKG varied to a degree that rendered them inappropriate to use as reference genes under those experimental settings. Here we present a substantially extended study of the HKG signature in human pluripotent stem cells (hPSC), including nine global gene expression datasets from both hESC and human induced pluripotent stem cells, obtained during directed differentiation toward endoderm-, mesoderm-, and ectoderm derivatives. Sets of stably expressed genes were compiled, and a handful of genes (e.g., EID2, ZNF324B, CAPN10, and RABEP2) were identified as generally applicable reference genes in hPSCs across all cell lines and experimental conditions. The stability in gene expression profiles was confirmed by reverse transcription quantitative PCR analysis. Taken together, the current results suggest that differentiating hPSCs have a distinct HKG signature, which in some aspects is different from somatic cell types, and underscore the necessity to validate the stability of reference genes under the actual experimental setup used. In addition, the novel putative HKGs identified in this study can preferentially be used for normalization of gene expression data obtained from differentiating hPSCs.


Subject(s)
Cell Differentiation/genetics , Genes , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Databases, Genetic , Gene Expression Profiling , Gene Expression Regulation , Humans , Oligonucleotide Array Sequence Analysis , Reference Standards
10.
Stem Cells ; 31(5): 941-52, 2013 May.
Article in English | MEDLINE | ID: mdl-23341249

ABSTRACT

Astrocytes can be generated from various tissue sources including human pluripotent stem cells (PSC). In this manuscript, we describe a chemically defined xeno-free medium culture system for rapidly generating astrocytes from neural stem cells derived from PSC. We show that astrocyte development in vitro, mimics normal development in vivo, and also passes through a CD44(+) astrocyte precursor stage. Astrocytes generated by our method display similar gene expression patterns, morphological characteristics and functional properties to primary astrocytes, and they survive and integrate after xenotransplantation. Whole genome expression profiling of astrocyte differentiation was performed at several time points of differentiation, and the results indicate the importance of known regulators and identify potential novel regulators and stage-specific lineage markers.


Subject(s)
Astrocytes/cytology , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Animals , Astrocytes/metabolism , Cell Differentiation/physiology , Cells, Cultured , Cryopreservation , Cytological Techniques/methods , Gene Expression , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Transplantation, Heterologous
11.
Cytotherapy ; 16(9): 1305-12, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25065637

ABSTRACT

BACKGROUND AIMS: We have previously reported a Good Manufacturing Practice (GMP)-compatible process for generating authentic dopaminergic neurons in defined media from human pluripotent stem cells and determined the time point at which dopaminergic precursors/neurons (day 14 after neuronal stem cell [NSC] stage) can be frozen, shipped and thawed without compromising their viability and ability to mature in vitro. One important issue we wished to address is whether dopaminergic precursors/neurons manufactured by our GMP-compatible process can be cryopreserved and engrafted in animal Parkinson disease (PD) models. METHODS: In this study, we evaluated the efficacy of freshly prepared and cryopreserved dopaminergic neurons in the 6-hydroxydopamine-lesioned rat PD model. RESULTS: We showed functional recovery up to 6 months post-transplantation in rats transplanted with our cells, whether freshly prepared or cryopreserved. In contrast, no motor improvement was observed in two control groups receiving either medium or cells at a slightly earlier stage (day 10 after NSC stage). Histologic analysis at the end point of the study (6 months post-transplantation) showed robust long-term survival of donor-derived tyrosine hydroxylase (TH)(+) dopaminergic neurons in rats transplanted with day 14 dopaminergic neurons. Moreover, TH(+) fibers emanated from the graft core into the surrounding host striatum. Consistent with the behavioral analysis, no or few TH(+) neurons were detected in animals receiving day 10 cells, although human cells were present in the graft. Importantly, no tumors were detected in any grafted rats, but long-term tumorigenic studies will need to determine the safety of our products. CONCLUSIONS: Dopaminergic neurons manufactured by a GMP-compatible process from human ESC survived and engrafted efficiently in the 6-OHDA PD rat model.


Subject(s)
Cryopreservation/methods , Dopaminergic Neurons/physiology , Neural Stem Cells/physiology , Parkinson Disease/therapy , Stem Cell Transplantation , Animals , Biotechnology , Cell Line , Cell Survival , Corpus Striatum/pathology , Disease Models, Animal , Dopaminergic Neurons/pathology , Female , Humans , Manufacturing Industry , Nerve Fibers/pathology , Neural Stem Cells/pathology , Neurogenesis , Oxidopamine/administration & dosage , Oxidopamine/adverse effects , Parkinson Disease/etiology , Practice Guidelines as Topic , Rats , Rats, Sprague-Dawley , Recovery of Function , Transplantation, Heterologous
12.
Nat Genet ; 37(10): 1099-103, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16142235

ABSTRACT

Cultured human embryonic stem cell (hESC) lines are an invaluable resource because they provide a uniform and stable genetic system for functional analyses and therapeutic applications. Nevertheless, these dividing cells, like other cells, probably undergo spontaneous mutation at a rate of 10(-9) per nucleotide. Because each mutant has only a few progeny, the overall biological properties of the cell culture are not altered unless a mutation provides a survival or growth advantage. Clonal evolution that leads to emergence of a dominant mutant genotype may potentially affect cellular phenotype as well. We assessed the genomic fidelity of paired early- and late-passage hESC lines in the course of tissue culture. Relative to early-passage lines, eight of nine late-passage hESC lines had one or more genomic alterations commonly observed in human cancers, including aberrations in copy number (45%), mitochondrial DNA sequence (22%) and gene promoter methylation (90%), although the latter was essentially restricted to 2 of 14 promoters examined. The observation that hESC lines maintained in vitro develop genetic and epigenetic alterations implies that periodic monitoring of these lines will be required before they are used in in vivo applications and that some late-passage hESC lines may be unusable for therapeutic purposes.


Subject(s)
Embryo, Mammalian/cytology , Genome, Human/genetics , Mutation , Stem Cells/metabolism , Cell Culture Techniques , Cell Line , DNA/genetics , DNA/metabolism , DNA Methylation , DNA, Mitochondrial/chemistry , Humans , Promoter Regions, Genetic
13.
iScience ; 27(3): 109231, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38439966

ABSTRACT

ApoE regulates neurogenesis, although how it influences genetic programs remains elusive. Cortical neurons induced from isogenic control and ApoE-/- human neural stem cells (NSCs) recapitulated key transcriptomic signatures of in vivo counterparts identified from single-cell human midbrain. Surprisingly, ApoE expression in NSC and neural progenitor cells (NPCs) is not required for differentiation. Instead, ApoE prevents the over-proliferation of non-neuronal cells during extended neuronal culture when it is not expressed. Elevated miR-199a-5p level in ApoE-/- cells lowers the EZH1 protein and the repressive H3K27me3 mark, a phenotype rescued by miR-199a-5p steric inhibitor. Reduced H3K27me3 at genes linked to extracellular matrix organization and angiogenesis in ApoE-/- NPC correlates with their aberrant expression and phenotypes in neurons. Interestingly, the ApoE coding sequence, which contains many predicted miR-199a-5p binding sites, can repress miR-199a-5p without translating into protein. This suggests that ApoE maintains neurons integrity through the target-directed miRNA degradation of miR-199a-5p, imparting the H3K27me3-mediated repression of non-neuronal genes during differentiation.

14.
J Cell Sci ; 124(Pt 3): 348-58, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21242311

ABSTRACT

Here, we have investigated mitochondrial biology and energy metabolism in human embryonic stem cells (hESCs) and hESC-derived neural stem cells (NSCs). Although stem cells collectively in vivo might be expected to rely primarily on anaerobic glycolysis for ATP supply, to minimise production of reactive oxygen species, we show that in vitro this is not so: hESCs generate an estimated 77% of their ATP through oxidative phosphorylation. Upon differentiation of hESCs into NSCs, oxidative phosphorylation declines both in absolute rate and in importance relative to glycolysis. A bias towards ATP supply from oxidative phosphorylation in hESCs is consistent with the expression levels of the mitochondrial gene regulators peroxisome-proliferator-activated receptor γ coactivator (PGC)-1α, PGC-1ß and receptor-interacting protein 140 (RIP140) in hESCs when compared with a panel of differentiated cell types. Analysis of the ATP demand showed that the slower ATP turnover in NSCs was associated with a slower rate of most energy-demanding processes but occurred without a reduction in the cellular growth rate. This mismatch is probably explained by a higher rate of macromolecule secretion in hESCs, on the basis of evidence from electron microscopy and an analysis of conditioned media. Taken together, our developmental model provides an understanding of the metabolic transition from hESCs to more quiescent somatic cell types, and supports important roles for mitochondria and secretion in hESC biology.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Differentiation , Embryonic Stem Cells , Mitochondria , Neural Stem Cells , Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Cell Proliferation , Culture Media, Conditioned , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/ultrastructure , Energy Metabolism , Heat-Shock Proteins/metabolism , Humans , Mitochondria/metabolism , Mitochondria/ultrastructure , Neural Stem Cells/metabolism , Neural Stem Cells/ultrastructure , Nuclear Proteins/metabolism , Nuclear Receptor Interacting Protein 1 , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA-Binding Proteins , Transcription Factors/metabolism
15.
Cytotherapy ; 15(8): 999-1010, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23664011

ABSTRACT

BACKGROUND AIMS: We have previously described a xeno-free scalable system to generate transplantable dopaminergic neurons from human pluripotent stem cells. However, several important questions remain to be answered about our cell therapy efforts. These include determining the exact time at which cells should be transplanted and whether cells at this stage can be frozen, shipped, thawed and injected without compromising their ability to mature and survive the transplantation procedure. We also needed to determine whether further optimization of the culture process could shorten the development time and reduce variability and whether a current Good Manufacture Practice (CGMP) facility could manufacture cells with fidelity. METHODS: We developed an optimized protocol that included modulating the sonic hedgehog homolog gradient with bone morphogenetic proteins (BMP2) and addition of activin to the culture medium, which shortened the time to generate Lmx1A and FoxA2 immunoreactive cells by 4-6 days. RESULTS: We showed that cells at this stage could be safely frozen and thawed while retaining an excellent ability to continue to mature in vitro and survive transplant in vivo. Importantly, we successfully adapted this process to a CGMP facility and manufactured two lots of transplant-ready dopaminergic neurons (>250 vials) under CGMP-compatible conditions. In vitro characterization, including viability/recovery on thawing, whole genome expression as well as expression of midbrain/dopaminergic markers, showed that the cells manufactured under GMP-compatible conditions were similar to cells produced at lab scale. CONCLUSIONS: Our results suggest that this optimized protocol can be used to generate dopaminergic neurons for Investigational New Drug enabling studies.


Subject(s)
Cell Culture Techniques , Dopaminergic Neurons/cytology , Dopaminergic Neurons/transplantation , Induced Pluripotent Stem Cells/metabolism , Neurogenesis , Pluripotent Stem Cells/cytology , Activins , Animals , Bone Morphogenetic Protein 2/metabolism , Cell Proliferation , Cell Survival , Cell- and Tissue-Based Therapy , Cells, Cultured , Cryopreservation/methods , Dopamine/analysis , Dopamine/biosynthesis , Drug Discovery/methods , Hedgehog Proteins/metabolism , Hepatocyte Nuclear Factor 3-beta/immunology , Humans , LIM-Homeodomain Proteins/immunology , Male , Mice , Mice, Inbred C57BL , Transcription Factors/immunology
16.
PLoS One ; 18(9): e0263021, 2023.
Article in English | MEDLINE | ID: mdl-37751438

ABSTRACT

Disease is a neurodegenerative disorder characterised by the progressive loss of dopaminergic cells of the substantia nigra pars compacta. Even though successful transplantation of dopamine-producing cells into the striatum exhibits favourable effects in animal models and clinical trials; transplanted cell survival is low. Since every transplant elicits an inflammatory response which can affect cell survival and differentiation, we aimed to study in vivo and in vitro the impact of the pro-inflammatory environment on human dopaminergic precursors. We first observed that transplanted human dopaminergic precursors into the striatum of immunosuppressed rats elicited an early and sustained activation of astroglial and microglial cells after 15 days' post-transplant. This long-lasting response was associated with Tumour necrosis factor alpha expression in microglial cells. In vitro, conditioned media from activated BV2 microglial cells increased cell death, decreased Tyrosine hydroxylase-positive cells and induced morphological alterations on human neural stem cells-derived dopaminergic precursors at two differentiation stages: 19 days and 28 days. Those effects were ameliorated by inhibition of Tumour necrosis factor alpha, a cytokine which was previously detected in vivo and in conditioned media from activated BV-2 cells. Our results suggest that a pro-inflammatory environment is sustained after transplantation under immunosuppression, providing a window of opportunity to modify this response to increase transplant survival and differentiation. In addition, our data show that the microglia-derived pro-inflammatory microenvironment has a negative impact on survival and differentiation of dopaminergic precursors. Finally, Tumour necrosis factor alpha plays a key role in these effects, suggesting that this cytokine could be an interesting target to increase the efficacy of human dopaminergic precursors transplantation in Parkinson's Disease.


Subject(s)
Microglia , Tumor Necrosis Factor-alpha , Humans , Animals , Rats , Tumor Necrosis Factor-alpha/pharmacology , Culture Media, Conditioned/pharmacology , Dopamine , Cell Differentiation , Cytokines
17.
J Cell Biochem ; 113(12): 3610-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22807388

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder. The motor symptoms of PD are caused by the loss of dopaminergic (DA) neurons in the substantia nigra pars compacta of mesencephalon. The causes for death of DA neurons are not well understood, but the strongest risk factor is increasing age. There is no cure currently available for PD, and treatment is limited to management of PD symptoms in patients. Primary DA neurons are virtually unobtainable from living patients and animal studies have proven inadequate for studying the mechanism of PD development. Pluripotent stem cells (PSC) are primary self-renewing cells capable of differentiating into all cell types of an organism, including DA neurons. PSCs represent an abundant source of cells that can be genetically modified or isolated from patients with complex diseases, enabling the production of large quantities of DA neurons for disease modeling, drug screening, and gene function studies. Furthermore, since PD arises as a result of deterioration of DA neurons in a specific brain region, it has been suggested that a relatively small number of cells could restore normal function. PSCs could provide a source of DA neurons for cell replacement therapy. In this Prospects article, we focus on the development and in vitro derivation of DA neurons from PSCs, as well as current applications of the technological advances, with the emphasis on future directions and efforts in the field.


Subject(s)
Dopaminergic Neurons/cytology , Induced Pluripotent Stem Cells/cytology , Neurogenesis , Animals , Cells, Cultured , Coculture Techniques/methods , Humans , Induced Pluripotent Stem Cells/transplantation , Mesencephalon/cytology , Parkinson Disease/pathology , Parkinson Disease/therapy , Regenerative Medicine , Stem Cell Transplantation/methods , Transcription, Genetic
18.
Biochem Biophys Res Commun ; 426(1): 100-5, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22917535

ABSTRACT

Human embryonic stem cells (hESCs) hold promise for the treatment of many human pathologies. For example, hESCs and the neuronal stem cells (NSCs) and neurons derived from them have significant potential as transplantation therapies for a variety of neurodegenerative diseases. Two concerns about the use of hESCs and their differentiated derivatives are their ability to function and their ability to resist neoplastic transformation in response to stresses that inevitably arise during their preparation for transplantation. To begin to understand how these cells handle genotoxic stress, we examined the responses of hESCs and derived NSCs and neurons to ionizing radiation (IR). Undifferentiated hESCs were extremely sensitive to IR, with nearly all the cells undergoing cell death within 5-7 h. NSCs and neurons were substantially more resistant to IR, with neurons showing the most resistant. Of interest, NSCs that survived IR underwent cellular senescence and acquired astrocytic characteristics. Unlike IR-treated astrocytes, however, the NSC-derived astrocytic cells that survived IR did not display the typical pro-inflammatory, pro-carcinogenic senescence-associated secretory phenotype. These findings suggest distinct genotoxic stress-responses of hESCs and derived NSC and neuronal populations, and suggest that damaged NSCs, while failing to function, may not cause local inflammation.


Subject(s)
Cell Differentiation/radiation effects , Cellular Senescence , DNA Damage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/radiation effects , Radiation Tolerance , Astrocytes/cytology , Astrocytes/metabolism , Biomarkers/metabolism , Cell Survival/radiation effects , Cells, Cultured , Humans , Neural Stem Cells/cytology , Neural Stem Cells/radiation effects , Neurons/cytology , Neurons/radiation effects
19.
Stem Cells ; 28(10): 1893-904, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20715183

ABSTRACT

Human induced pluripotent stem cells (iPSCs) reprogrammed from somatic cells represent a promising unlimited cell source for generating patient-specific cells for biomedical research and personalized medicine. As a first step, critical to clinical applications, we attempted to develop defined culture conditions to expand and differentiate human iPSCs into functional progeny such as dopaminergic neurons for treating or modeling Parkinson's disease (PD). We used a completely defined (xeno-free) system that we previously developed for efficient generation of authentic dopaminergic neurons from human embryonic stem cells (hESCs), and applied it to iPSCs. First, we adapted two human iPSC lines derived from different somatic cell types for the defined expansion medium and showed that the iPSCs grew similarly as hESCs in the same medium regarding pluripotency and genomic stability. Second, by using these two independent adapted iPSC lines, we showed that the process of differentiation into committed neural stem cells (NSCs) and subsequently into dopaminergic neurons was also similar to hESCs. Importantly, iPSC-derived dopaminergic neurons were functional as they survived and improved behavioral deficits in 6-hydroxydopamine-leasioned rats after transplantation. In addition, iPSC-derived NSCs and neurons could be efficiently transduced by a baculoviral vector delivering episomal DNA for future gene function study and disease modeling using iPSCs. We also performed genome-wide microarray comparisons between iPSCs and hESCs, and we derived NSC and dopaminergic neurons. Our data revealed overall similarity and visible differences at a molecular level. Efficient generation of functional dopaminergic neurons under defined conditions will facilitate research and applications using PD patient-specific iPSCs.


Subject(s)
Dopamine/metabolism , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Neurons/metabolism , Cell Differentiation , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
20.
Burns ; 47(1): 140-149, 2021 02.
Article in English | MEDLINE | ID: mdl-33279335

ABSTRACT

Hypertrophic scar (HS) is a pathological scar that often occurs in burn patients. Its histology is characterized by the excessive proliferation of fibroblasts (FB) and excessive accumulation of extracellular matrix (ECM). Inhibition of proliferation and activation of FB is essential for the treatment of HS. The crude extracts of traditional Chinese medicines have beneficial therapeutic effects on HS besides possessing fewer side effects and being easily available. Polyphyllin VII (PP7) is an isoprene saponin isolated from Rhizoma paridis. It has a pro-apoptotic effect on cancer cells. In the present study, we demonstrate that PP7 exerts a significant inhibitory effect on hypertrophic scar fibroblasts (HSFs) in vitro. We also demonstrate that PP7 considerably induces the apoptosis of HSFs and inhibits their activity. Our data show that the PP7-induced HSFs cell apoptosis was mainly due to the enhanced expression of apoptotic genes (Bax, Caspase-3, Caspase-9) and decreased expression of Bcl-2. Moreover, PP7 treatment also enhances the expression of JNK, but that of extracellular protein kinases (ERK) was reduced, and induces apoptosis through ERK/JNK pathways. Thus, PP7 can be used as a drug to prevent the formation of HS.


Subject(s)
Apoptosis/drug effects , Saponins/pharmacology , Signal Transduction/drug effects , Animals , Blotting, Western/methods , Burns/pathology , Disease Models, Animal , Fibroblasts/drug effects , Flow Cytometry/methods , Protein Kinases/metabolism , Rabbits/metabolism , Rabbits/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL