Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cytotherapy ; 26(4): 340-350, 2024 04.
Article in English | MEDLINE | ID: mdl-38349309

ABSTRACT

BACKGROUND AIMS: Age-related macular degeneration (AMD) is the most common cause of blindness in elderly patients within developed countries, affecting more than 190 million worldwide. In AMD, the retinal pigment epithelial (RPE) cell layer progressively degenerates, resulting in subsequent loss of photoreceptors and ultimately vision. There is currently no cure for AMD, but therapeutic strategies targeting the complement system are being developed to slow the progression of the disease. METHODS: Replacement therapy with pluripotent stem cell-derived (hPSC) RPEs is an alternative treatment strategy. A cell therapy product must be produced in accordance with Good Manufacturing Practices at a sufficient scale to facilitate extensive pre-clinical and clinical testing. Cryopreservation of the final cell product is therefore highly beneficial, as the manufacturing, pre-clinical and clinical testing can be separated in time and location. RESULTS: We found that mature hPSC-RPE cells do not survive conventional cryopreservation techniques. However, replating the cells 2-5 days before cryopreservation facilitates freezing. The replated and cryopreserved hPSC-RPE cells maintained their identity, purity and functionality as characteristic RPEs, shown by cobblestone morphology, pigmentation, transcriptional profile, RPE markers, transepithelial resistance and pigment epithelium-derived factor secretion. Finally, we showed that the optimal replating time window can be tracked noninvasively by following the change in cobblestone morphology. CONCLUSIONS: The possibility of cryopreserving the hPSC-RPE product has been instrumental in our efforts in manufacturing and performing pre-clinical testing with the aim for clinical translation.


Subject(s)
Macular Degeneration , Pluripotent Stem Cells , Humans , Aged , Cell Differentiation , Macular Degeneration/therapy , Cryopreservation , Epithelial Cells , Retinal Pigments
2.
bioRxiv ; 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38328127

ABSTRACT

Across a range of biological processes, cells undergo coordinated changes in gene expression, resulting in transcriptome dynamics that unfold within a low-dimensional manifold. Single-cell RNA-sequencing (scRNA-seq) only measures temporal snapshots of gene expression. However, information on the underlying low-dimensional dynamics can be extracted using RNA velocity, which models unspliced and spliced RNA abundances to estimate the rate of change of gene expression. Available RNA velocity algorithms can be fragile and rely on heuristics that lack statistical control. Moreover, the estimated vector field is not dynamically consistent with the traversed gene expression manifold. Here, we develop a generative model of RNA velocity and a Bayesian inference approach that solves these problems. Our model couples velocity field and manifold estimation in a reformulated, unified framework, so as to coherently identify the parameters of an autonomous dynamical system. Focusing on the cell cycle, we implemented VeloCycle to study gene regulation dynamics on one-dimensional periodic manifolds and validated using live-imaging its ability to infer actual cell cycle periods. We benchmarked RNA velocity inference with sensitivity analyses and demonstrated one- and multiple-sample testing. We also conducted Markov chain Monte Carlo inference on the model, uncovering key relationships between gene-specific kinetics and our gene-independent velocity estimate. Finally, we applied VeloCycle to in vivo samples and in vitro genome-wide Perturb-seq, revealing regionally-defined proliferation modes in neural progenitors and the effect of gene knockdowns on cell cycle speed. Ultimately, VeloCycle expands the scRNA-seq analysis toolkit with a modular and statistically rigorous RNA velocity inference framework.

3.
Stem Cell Reports ; 17(6): 1458-1475, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35705015

ABSTRACT

Human embryonic stem cell-derived retinal pigment epithelial cells (hESC-RPE) are a promising cell source to treat age-related macular degeneration (AMD). Despite several ongoing clinical studies, a detailed mapping of transient cellular states during in vitro differentiation has not been performed. Here, we conduct single-cell transcriptomic profiling of an hESC-RPE differentiation protocol that has been developed for clinical use. Differentiation progressed through a culture diversification recapitulating early embryonic development, whereby cells rapidly acquired a rostral embryo patterning signature before converging toward the RPE lineage. At intermediate steps, we identified and examined the potency of an NCAM1+ retinal progenitor population and showed the ability of the protocol to suppress non-RPE fates. We demonstrated that the method produces a pure RPE pool capable of maturing further after subretinal transplantation in a large-eyed animal model. Our evaluation of hESC-RPE differentiation supports the development of safe and efficient pluripotent stem cell-based therapies for AMD.


Subject(s)
Human Embryonic Stem Cells , Macular Degeneration , Animals , Cell Differentiation/genetics , Humans , Macular Degeneration/genetics , Macular Degeneration/therapy , Retinal Pigment Epithelium , Retinal Pigments
4.
Science ; 376(6590): eabh1623, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35420948

ABSTRACT

Human cells produce thousands of lipids that change during cell differentiation and can vary across individual cells of the same type. However, we are only starting to characterize the function of these cell-to-cell differences in lipid composition. Here, we measured the lipidomes and transcriptomes of individual human dermal fibroblasts by coupling high-resolution mass spectrometry imaging with single-cell transcriptomics. We found that the cell-to-cell variations of specific lipid metabolic pathways contribute to the establishment of cell states involved in the organization of skin architecture. Sphingolipid composition is shown to define fibroblast subpopulations, with sphingolipid metabolic rewiring driving cell-state transitions. Therefore, cell-to-cell lipid heterogeneity affects the determination of cell states, adding a new regulatory component to the self-organization of multicellular systems.


Subject(s)
Fibroblasts , Skin , Sphingolipids , Fibroblasts/chemistry , Fibroblasts/classification , Fibroblasts/metabolism , Humans , Lipidomics/methods , Metabolic Networks and Pathways , Skin/chemistry , Skin/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Sphingolipids/analysis , Sphingolipids/metabolism , Transcriptome
5.
Nature ; 596(7870): 92-96, 2021 08.
Article in English | MEDLINE | ID: mdl-34321664

ABSTRACT

The mammalian brain develops through a complex interplay of spatial cues generated by diffusible morphogens, cell-cell interactions and intrinsic genetic programs that result in probably more than a thousand distinct cell types. A complete understanding of this process requires a systematic characterization of cell states over the entire spatiotemporal range of brain development. The ability of single-cell RNA sequencing and spatial transcriptomics to reveal the molecular heterogeneity of complex tissues has therefore been particularly powerful in the nervous system. Previous studies have explored development in specific brain regions1-8, the whole adult brain9 and even entire embryos10. Here we report a comprehensive single-cell transcriptomic atlas of the embryonic mouse brain between gastrulation and birth. We identified almost eight hundred cellular states that describe a developmental program for the functional elements of the brain and its enclosing membranes, including the early neuroepithelium, region-specific secondary organizers, and both neurogenic and gliogenic progenitors. We also used in situ mRNA sequencing to map the spatial expression patterns of key developmental genes. Integrating the in situ data with our single-cell clusters revealed the precise spatial organization of neural progenitors during the patterning of the nervous system.


Subject(s)
Brain/cytology , Brain/embryology , Single-Cell Analysis , Transcriptome , Animals , Animals, Newborn/genetics , Brain/anatomy & histology , Female , Gastrulation/genetics , Male , Mice , Neural Tube/anatomy & histology , Neural Tube/cytology , Neural Tube/embryology
6.
Curr Opin Biotechnol ; 63: 70-78, 2020 06.
Article in English | MEDLINE | ID: mdl-31918114

ABSTRACT

Single-cell transcriptomics enables the measurement of gene expression in complex biological systems at the resolution of individual cells. Multivariate analysis of single-cell data helps describe the variation in expression accompanying cellular processes during embryonic development, disease progression and in response to stimuli. Likewise, new methods have extended the possibilities of single-cell analysis by measuring the transcriptome while simultaneously capturing information on lineage or past molecular events. These emerging approaches have the common strategy of querying a static snapshot for information related to different temporal stages. Single-cell temporal-omics methods open new possibilities such as extrapolating the future or correlating past events to present gene expression. We highlight advancements in the single-cell field, describe novel toolkits for investigation, and consider the potential impact of temporal-omics approaches for the study of disease progression.


Subject(s)
Computational Biology , Transcriptome , Single-Cell Analysis
7.
Genetics ; 212(3): 711-728, 2019 07.
Article in English | MEDLINE | ID: mdl-31092540

ABSTRACT

The Polymerase Associated Factor 1 complex (Paf1C) is a multifunctional regulator of eukaryotic gene expression important for the coordination of transcription with chromatin modification and post-transcriptional processes. In this study, we investigated the extent to which the functions of Paf1C combine to regulate the Saccharomyces cerevisiae transcriptome. While previous studies focused on the roles of Paf1C in controlling mRNA levels, here, we took advantage of a genetic background that enriches for unstable transcripts, and demonstrate that deletion of PAF1 affects all classes of Pol II transcripts including multiple classes of noncoding RNAs (ncRNAs). By conducting a de novo differential expression analysis independent of gene annotations, we found that Paf1 positively and negatively regulates antisense transcription at multiple loci. Comparisons with nascent transcript data revealed that many, but not all, changes in RNA levels detected by our analysis are due to changes in transcription instead of post-transcriptional events. To investigate the mechanisms by which Paf1 regulates protein-coding genes, we focused on genes involved in iron and phosphate homeostasis, which were differentially affected by PAF1 deletion. Our results indicate that Paf1 stimulates phosphate gene expression through a mechanism that is independent of any individual Paf1C-dependent histone modification. In contrast, the inhibition of iron gene expression by Paf1 correlates with a defect in H3 K36 trimethylation. Finally, we showed that one iron regulon gene, FET4, is coordinately controlled by Paf1 and transcription of upstream noncoding DNA. Together, these data identify roles for Paf1C in controlling both coding and noncoding regions of the yeast genome.


Subject(s)
Gene Expression Regulation, Fungal , Nuclear Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcriptome , Chromatin/metabolism , Copper Transport Proteins/genetics , Copper Transport Proteins/metabolism , Histones/metabolism , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Protein Processing, Post-Translational , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism
8.
Nat Biotechnol ; 36(6): 512-520, 2018 07.
Article in English | MEDLINE | ID: mdl-29734294

ABSTRACT

Our understanding of how genotype controls phenotype is limited by the scale at which we can precisely alter the genome and assess the phenotypic consequences of each perturbation. Here we describe a CRISPR-Cas9-based method for multiplexed accurate genome editing with short, trackable, integrated cellular barcodes (MAGESTIC) in Saccharomyces cerevisiae. MAGESTIC uses array-synthesized guide-donor oligos for plasmid-based high-throughput editing and features genomic barcode integration to prevent plasmid barcode loss and to enable robust phenotyping. We demonstrate that editing efficiency can be increased more than fivefold by recruiting donor DNA to the site of breaks using the LexA-Fkh1p fusion protein. We performed saturation editing of the essential gene SEC14 and identified amino acids critical for chemical inhibition of lipid signaling. We also constructed thousands of natural genetic variants, characterized guide mismatch tolerance at the genome scale, and ascertained that cryptic Pol III termination elements substantially reduce guide efficacy. MAGESTIC will be broadly useful to uncover the genetic basis of phenotypes in yeast.


Subject(s)
DNA Barcoding, Taxonomic/methods , Gene Editing/methods , Saccharomyces cerevisiae/genetics , Amino Acid Substitution , Biotechnology , CRISPR-Cas Systems , DNA, Fungal/genetics , Genome, Fungal , Homologous Recombination , Phospholipid Transfer Proteins/genetics , Plasmids/genetics , RNA, Fungal/genetics , Saccharomyces cerevisiae Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...