Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Mol Cancer ; 23(1): 180, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39217332

ABSTRACT

BACKGROUND: Neuroblastoma (NB) is a heterogeneous embryonal malignancy and the deadliest tumor of infancy. It is a complex disease that can result in diverse clinical outcomes. In some children, tumors regress spontaneously. Others respond well to existing treatments. But for the high-risk group, which constitutes approximately 40% of all patients, the prognosis remains dire despite collaborative efforts in basic and clinical research. While its exact cellular origin is still under debate, NB is assumed to arise from the neural crest cell lineage including multipotent Schwann cell precursors (SCPs), which differentiate into sympatho-adrenal cell states eventually producing chromaffin cells and sympathoblasts. METHODS: To investigate clonal development of neuroblastoma cell states, we performed haplotype-specific analysis of human tumor samples using single-cell multi-omics, including joint DNA/RNA sequencing of sorted single cells (DNTR-seq). Samples were also assessed using immunofluorescence stainings and fluorescence in-situ hybridization (FISH). RESULTS: Beyond adrenergic tumor cells, we identify subpopulations of aneuploid SCP-like cells, characterized by clonal expansion, whole-chromosome 17 gains, as well as expression programs of proliferation, apoptosis, and a non-immunomodulatory phenotype. CONCLUSION: Aneuploid pre-malignant SCP-like cells represent a novel feature of NB. Genetic evidence and tumor phylogeny suggest that these clones and malignant adrenergic populations originate from aneuploidy-prone cells of migrating neural crest or SCP origin, before lineage commitment to sympatho-adrenal cell states. Our findings expand the phenotypic spectrum of NB cell states. Considering the multipotency of SCPs in development, we suggest that the transformation of fetal SCPs may represent one possible mechanism of tumor initiation in NB with chromosome 17 aberrations as a characteristic element.


Subject(s)
Gene Expression Profiling , Neuroblastoma , Schwann Cells , Single-Cell Analysis , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/metabolism , Schwann Cells/metabolism , Schwann Cells/pathology , Transcriptome , Gene Expression Regulation, Neoplastic , In Situ Hybridization, Fluorescence
2.
Nat Commun ; 15(1): 4778, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862479

ABSTRACT

Impairment of the central nervous system (CNS) poses a significant health risk for astronauts during long-duration space missions. In this study, we employed an innovative approach by integrating single-cell multiomics (transcriptomics and chromatin accessibility) with spatial transcriptomics to elucidate the impact of spaceflight on the mouse brain in female mice. Our comparative analysis between ground control and spaceflight-exposed animals revealed significant alterations in essential brain processes including neurogenesis, synaptogenesis and synaptic transmission, particularly affecting the cortex, hippocampus, striatum and neuroendocrine structures. Additionally, we observed astrocyte activation and signs of immune dysfunction. At the pathway level, some spaceflight-induced changes in the brain exhibit similarities with neurodegenerative disorders, marked by oxidative stress and protein misfolding. Our integrated spatial multiomics approach serves as a stepping stone towards understanding spaceflight-induced CNS impairments at the level of individual brain regions and cell types, and provides a basis for comparison in future spaceflight studies. For broader scientific impact, all datasets from this study are available through an interactive data portal, as well as the National Aeronautics and Space Administration (NASA) Open Science Data Repository (OSDR).


Subject(s)
Brain , Neurons , Space Flight , Animals , Mice , Female , Brain/metabolism , Brain/pathology , Neurons/metabolism , Transcriptome , Neurogenesis , Single-Cell Analysis , Mice, Inbred C57BL , Synaptic Transmission , Weightlessness/adverse effects , Astrocytes/metabolism , Oxidative Stress , Gene Expression Profiling , Multiomics
3.
Dev Cell ; 58(20): 2140-2162.e5, 2023 10 23.
Article in English | MEDLINE | ID: mdl-37591247

ABSTRACT

A wealth of specialized cell populations within the skin facilitates its hair-producing, protective, sensory, and thermoregulatory functions. How the vast cell-type diversity and tissue architecture develops is largely unexplored. Here, with single-cell transcriptomics, spatial cell-type assignment, and cell-lineage tracing, we deconstruct early embryonic mouse skin during the key transitions from seemingly uniform developmental precursor states to a multilayered, multilineage epithelium, and complex dermal identity. We identify the spatiotemporal emergence of hair-follicle-inducing, muscle-supportive, and fascia-forming fibroblasts. We also demonstrate the formation of the panniculus carnosus muscle (PCM), sprouting blood vessels without pericyte coverage, and the earliest residence of mast and dendritic immune cells in skin. Finally, we identify an unexpected epithelial heterogeneity within the early single-layered epidermis and a signaling-rich periderm layer. Overall, this cellular and molecular blueprint of early skin development-which can be explored at https://kasperlab.org/tools-establishes histological landmarks and highlights unprecedented dynamic interactions among skin cells.


Subject(s)
Epidermis , Skin , Mice , Animals , Hair Follicle/pathology , Hair , Epithelium
4.
Clin Cancer Res ; 29(20): 4139-4152, 2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37540566

ABSTRACT

PURPOSE: Although CD19 chimeric antigen receptor T cells (CAR-T) therapy has shown remarkable success in B-cell malignancies, a substantial fraction of patients do not obtain a long-term clinical response. This could be influenced by the quality of the individual CAR-T infusion product. To shed some light on this, clinical outcome was correlated to characteristics of CAR-T infusion products. PATIENTS AND METHODS: In this phase II study, patients with B-cell lymphoma (n = 23) or leukemia (n = 1) received one or two infusions of third-generation CD19-directed CAR-Ts (2 × 108/m2). The clinical trial was registered at clinicaltrials.gov: NCT03068416. We investigated the transcriptional profile of individual CD19 CAR-T infusion products using targeted single-cell RNA sequencing and multicolor flow cytometry. RESULTS: Two CAR-T infusions were not better than one in the settings used in this study. As for the CAR-T infusion products, we found that effector-like CD8+CAR-Ts with a high polyfunctionality, high cytotoxic and cytokine production profile, and low dysfunctional signature were associated with clinical response. An extended ex vivo expansion time during CAR-T manufacturing negatively influenced the proportion of effector CD8+CAR-Ts in the infusion product. CONCLUSIONS: We identified cell-intrinsic characteristics of effector CD8+CAR-Ts correlating with response that could be used as an indicator for clinical outcome. The results in the study also serve as a guide to CAR-T manufacturing practices.

5.
Dev Cell ; 58(6): 450-460.e6, 2023 03 27.
Article in English | MEDLINE | ID: mdl-36893754

ABSTRACT

Building a blastema from the stump is a key step of salamander limb regeneration. Stump-derived cells temporarily suspend their identity as they contribute to the blastema by a process generally referred to as dedifferentiation. Here, we provide evidence for a mechanism that involves an active inhibition of protein synthesis during blastema formation and growth. Relieving this inhibition results in a higher number of cycling cells and enhances the pace of limb regeneration. By small RNA profiling and fate mapping of skeletal muscle progeny as a cellular model for dedifferentiation, we find that the downregulation of miR-10b-5p is critical for rebooting the translation machinery. miR-10b-5p targets ribosomal mRNAs, and its artificial upregulation causes decreased blastema cell proliferation, reduction in transcripts that encode ribosomal subunits, diminished nascent protein synthesis, and retardation of limb regeneration. Taken together, our data identify a link between miRNA regulation, ribosome biogenesis, and protein synthesis during newt limb regeneration.


Subject(s)
MicroRNAs , RNA, Small Untranslated , Animals , Urodela/genetics , RNA, Small Untranslated/metabolism , Muscle, Skeletal/metabolism , Ribosomes/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Extremities/physiology
6.
iScience ; 26(1): 105857, 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36624836

ABSTRACT

Cardiomyocytes play key roles during cardiogenesis, but have poorly understood features, especially in prenatal stages. Here, we characterized human prenatal cardiomyocytes, 6.5-7 weeks post-conception, by integrating single-cell RNA sequencing, spatial transcriptomics, and ligand-receptor interaction information. Using a computational workflow developed to dissect cell type heterogeneity, localize cell types, and explore their molecular interactions, we identified eight types of developing cardiomyocyte, more than double compared to the ones identified in the Human Developmental Cell Atlas. These have high variability in cell cycle activity, mitochondrial content, and connexin gene expression, and are differentially distributed in the ventricles, including outflow tract, and atria, including sinoatrial node. Moreover, cardiomyocyte ligand-receptor crosstalk is mainly with non-cardiomyocyte cell types, encompassing cardiogenesis-related pathways. Thus, early prenatal human cardiomyocytes are highly heterogeneous and develop unique location-dependent properties, with complex ligand-receptor crosstalk. Further elucidation of their developmental dynamics may give rise to new therapies.

7.
Genome Res ; 2022 Aug 10.
Article in English | MEDLINE | ID: mdl-35948369

ABSTRACT

The preconceptual, intrauterine, and early life environments can have a profound and long-lasting impact on the developmental trajectories and health outcomes of the offspring. Given the relatively low success rates of assisted reproductive technologies (ART; ∼25%), additives and adjuvants, such as glucocorticoids, are used to improve the success rate. Considering the dynamic developmental events that occur during this window, these exposures may alter blastocyst formation at a molecular level, and as such, affect not only the viability of the embryo and the ability of the blastocyst to implant, but also the developmental trajectory of the first three cell lineages, ultimately influencing the physiology of the embryo. In this study, we present a comprehensive single-cell transcriptome, methylome, and small RNA atlas in the day 7 human embryo. We show that, despite no change in morphology and developmental features, preimplantation glucocorticoid exposure reprograms the molecular profile of the TE lineage, and these changes are associated with an altered metabolic and inflammatory response. Our data also suggest that glucocorticoids can precociously mature the TE sublineages, supported by the presence of extravillous trophoblast markers in the polar sublineage and presence of X Chromosome dosage compensation. Further, we have elucidated that epigenetic regulation-DNA methylation and microRNAs (miRNAs)-likely underlies the transcriptional changes observed. This study suggests that exposures to exogenous compounds during preimplantation may unintentionally reprogram the human embryo, possibly leading to suboptimal development and longer-term health outcomes.

8.
Immunity ; 54(9): 2005-2023.e10, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525339

ABSTRACT

Cell fate decisions during early B cell activation determine the outcome of responses to pathogens and vaccines. We examined the early B cell response to T-dependent antigen in mice by single-cell RNA sequencing. Early after immunization, a homogeneous population of activated precursors (APs) gave rise to a transient wave of plasmablasts (PBs), followed a day later by the emergence of germinal center B cells (GCBCs). Most APs rapidly exited the cell cycle, giving rise to non-GC-derived early memory B cells (eMBCs) that retained an AP-like transcriptional profile. Rapid decline of antigen availability controlled these events; provision of excess antigen precluded cell cycle exit and induced a new wave of PBs. Fate mapping revealed a prominent contribution of eMBCs to the MBC pool. Quiescent cells with an MBC phenotype dominated the early response to immunization in primates. A reservoir of APs/eMBCs may enable rapid readjustment of the immune response when failure to contain a threat is manifested by increased antigen availability.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Immunity, Humoral/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Animals , Antigen Presentation/immunology , Cell Differentiation/immunology , Mice , Plasma Cells/immunology , Precursor Cells, B-Lymphoid/immunology
9.
Sci Rep ; 11(1): 4076, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33603054

ABSTRACT

Transcriptome analysis has mainly relied on analyzing RNA sequencing data from whole cells, overlooking the impact of subcellular RNA localization and its influence on our understanding of gene function, and interpretation of gene expression signatures in cells. Here, we separated cytosolic and nuclear RNA from human fetal and adult brain samples and performed a comprehensive analysis of cytosolic and nuclear transcriptomes. There are significant differences in RNA expression for protein-coding and lncRNA genes between cytosol and nucleus. We show that transcripts encoding the nuclear-encoded mitochondrial proteins are significantly enriched in the cytosol compared to the rest of protein-coding genes. Differential expression analysis between fetal and adult frontal cortex show that results obtained from the cytosolic RNA differ from results using nuclear RNA both at the level of transcript types and the number of differentially expressed genes. Our data provide a resource for the subcellular localization of thousands of RNA transcripts in the human brain and highlight differences in using the cytosolic or the nuclear transcriptomes for expression analysis.


Subject(s)
Brain/metabolism , Cell Nucleus/metabolism , Cytosol/metabolism , Transcriptome , Cell Nucleus/genetics , Gene Expression Profiling , Humans , RNA/genetics , RNA/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Nuclear/genetics , RNA, Nuclear/metabolism , Subcellular Fractions/metabolism , Transcription, Genetic , Transcriptome/genetics
10.
Cell Res ; 31(5): 554-568, 2021 05.
Article in English | MEDLINE | ID: mdl-33420427

ABSTRACT

The impact of the microenvironment on innate lymphoid cell (ILC)-mediated immunity in humans remains largely unknown. Here we used full-length Smart-seq2 single-cell RNA-sequencing to unravel tissue-specific transcriptional profiles and heterogeneity of CD127+ ILCs across four human tissues. Correlation analysis identified gene modules characterizing the migratory properties of tonsil and blood ILCs, and signatures of tissue-residency, activation and modified metabolism in colon and lung ILCs. Trajectory analysis revealed potential differentiation pathways from circulating and tissue-resident naïve ILCs to a spectrum of mature ILC subsets. In the lung we identified both CRTH2+ and CRTH2- ILC2 with lung-specific signatures, which could be recapitulated by alarmin-exposure of circulating ILC2. Finally, we describe unique TCR-V(D)J-rearrangement patterns of blood ILC1-like cells, revealing a subset of potentially immature ILCs with TCR-δ rearrangement. Our study provides a useful resource for in-depth understanding of ILC-mediated immunity in humans, with implications for disease.


Subject(s)
Immunity, Innate , Lymphocytes , Cell Differentiation , Humans , Immunity, Innate/genetics , RNA
11.
Commun Biol ; 3(1): 338, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32620779

ABSTRACT

The subthalamic nucleus (STN) is crucial for normal motor, limbic and associative function. STN dysregulation is correlated with several brain disorders, including Parkinson's disease and obsessive compulsive disorder (OCD), for which high-frequency stimulation of the STN is increasing as therapy. However, clinical progress is hampered by poor knowledge of the anatomical-functional organization of the STN. Today, experimental mouse genetics provides outstanding capacity for functional decoding, provided selective promoters are available. Here, we implemented single-nuclei RNA sequencing (snRNASeq) of the mouse STN followed through with histological analysis of 16 candidate genes of interest. Our results demonstrate that the mouse STN is composed of at least four spatio-molecularly defined domains, each distinguished by defined sets of promoter activities. Further, molecular profiles dissociate the STN from the adjoining para-STN (PSTN) and neighboring structures of the hypothalamus, mammillary nuclei and zona incerta. Enhanced knowledge of STN´s internal organization should prove useful towards genetics-based functional decoding of this clinically relevant brain structure.


Subject(s)
Brain/anatomy & histology , Brain/metabolism , Cell Nucleus/metabolism , Glutamic Acid/metabolism , Receptors, GABA/metabolism , Subthalamic Nucleus/metabolism , Transcriptome , Animals , Female , Male , Mice , Single-Cell Analysis , Spatial Analysis
13.
Proc Natl Acad Sci U S A ; 117(30): 17854-17863, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32647059

ABSTRACT

Pacemaker neurons exert control over neuronal circuit function by their intrinsic ability to generate rhythmic bursts of action potential. Recent work has identified rhythmic gut contractions in human, mice, and hydra to be dependent on both neurons and the resident microbiota. However, little is known about the evolutionary origin of these neurons and their interaction with microbes. In this study, we identified and functionally characterized prototypical ANO/SCN/TRPM ion channel-expressing pacemaker cells in the basal metazoan Hydra by using a combination of single-cell transcriptomics, immunochemistry, and functional experiments. Unexpectedly, these prototypical pacemaker neurons express a rich set of immune-related genes mediating their interaction with the microbial environment. Furthermore, functional experiments gave a strong support to a model of the evolutionary emergence of pacemaker cells as neurons using components of innate immunity to interact with the microbial environment and ion channels to generate rhythmic contractions.


Subject(s)
Biological Clocks , Hydra/physiology , Microbiota , Neurons/physiology , Action Potentials , Animals , Biological Evolution , Cluster Analysis , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation , Genome-Wide Association Study , Humans , Mice
14.
Nat Commun ; 11(1): 2434, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32415072

ABSTRACT

Cell replacement is a long-standing and realistic goal for the treatment of Parkinson's disease (PD). Cells for transplantation can be obtained from fetal brain tissue or from stem cells. However, after transplantation, dopamine (DA) neurons are seen to be a minor component of grafts, and it has remained difficult to determine the identity of other cell types. Here, we report analysis by single-cell RNA sequencing (scRNA-seq) combined with comprehensive histological analyses to characterize intracerebral grafts from human embryonic stem cells (hESCs) and fetal tissue after functional maturation in a pre-clinical rat PD model. We show that neurons and astrocytes are major components in both fetal and stem cell-derived grafts. Additionally, we identify a cell type closely resembling a class of recently identified perivascular-like cells in stem cell-derived grafts. Thus, this study uncovers previously unknown cellular diversity in a clinically relevant cell replacement PD model.


Subject(s)
Dopaminergic Neurons/cytology , Parkinson Disease/therapy , Stem Cell Transplantation , Stem Cells/cytology , Animals , Brain/metabolism , Cell Differentiation , Corpus Striatum , Disease Models, Animal , Dopamine/metabolism , Embryonic Stem Cells/cytology , Female , Graft Survival , Humans , Multigene Family , RNA-Seq , Rats , Rats, Nude , Regeneration , Single-Cell Analysis , Transcriptome
15.
Genome Biol ; 20(1): 68, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30935387

ABSTRACT

Accurate variant calling and genotyping represent major limiting factors for downstream applications of single-cell genomics. Here, we report Conbase for the identification of somatic mutations in single-cell DNA sequencing data. Conbase leverages phased read data from multiple samples in a dataset to achieve increased confidence in somatic variant calls and genotype predictions. Comparing the performance of Conbase to three other methods, we find that Conbase performs best in terms of false discovery rate and specificity and provides superior robustness on simulated data, in vitro expanded fibroblasts and clonal lymphocyte populations isolated directly from a healthy human donor.


Subject(s)
Mutation , Single-Cell Analysis , Software , CD8-Positive T-Lymphocytes , Fibroblasts , Humans , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
16.
Nat Commun ; 10(1): 581, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30718509

ABSTRACT

Midbrain dopamine (mDA) neurons constitute a heterogenous group of cells that have been intensely studied, not least because their degeneration causes major symptoms in Parkinson's disease. Understanding the diversity of mDA neurons - previously well characterized anatomically - requires a systematic molecular classification at the genome-wide gene expression level. Here, we use single cell RNA sequencing of isolated mouse neurons expressing the transcription factor Pitx3, a marker for mDA neurons. Analyses include cells isolated during development up until adulthood and the results are validated by histological characterization of newly identified markers. This identifies seven neuron subgroups divided in two major branches of developing Pitx3-expressing neurons. Five of them express dopaminergic markers, while two express glutamatergic and GABAergic markers, respectively. Analysis also indicate evolutionary conservation of diversity in humans. This comprehensive molecular characterization will provide a valuable resource for elucidating mDA neuron subgroup development and function in the mammalian brain.


Subject(s)
Brain/cytology , Dopaminergic Neurons/metabolism , Sequence Analysis, RNA/methods , Animals , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mice , Transcription Factors/metabolism
17.
Nat Commun ; 9(1): 5150, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30514914

ABSTRACT

Cancer-associated fibroblasts (CAFs) are a major constituent of the tumor microenvironment, although their origin and roles in shaping disease initiation, progression and treatment response remain unclear due to significant heterogeneity. Here, following a negative selection strategy combined with single-cell RNA sequencing of 768 transcriptomes of mesenchymal cells from a genetically engineered mouse model of breast cancer, we define three distinct subpopulations of CAFs. Validation at the transcriptional and protein level in several experimental models of cancer and human tumors reveal spatial separation of the CAF subclasses attributable to different origins, including the peri-vascular niche, the mammary fat pad and the transformed epithelium. Gene profiles for each CAF subtype correlate to distinctive functional programs and hold independent prognostic capability in clinical cohorts by association to metastatic disease. In conclusion, the improved resolution of the widely defined CAF population opens the possibility for biomarker-driven development of drugs for precision targeting of CAFs.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts , Sequence Analysis, RNA , Transcriptome , Adipose Tissue/metabolism , Animals , Biomarkers, Tumor/genetics , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/classification , Cell Cycle/genetics , Cell Line, Tumor , Cluster Analysis , Disease Progression , Epithelium/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Intercellular Junctions/genetics , Logistic Models , Mice , Mice, Transgenic , Prognosis , Transcription Factors/genetics , Transcriptome/genetics
18.
Nat Commun ; 9(1): 4639, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389943

ABSTRACT

In the originally published version of this Article, financial support was not fully acknowledged. The PDF and HTML versions of the Article have now been corrected to include support to Thomas Perlmann provided by Knut and Alice Wallenberg Foundation (grant 2013.0075) and Swedish Research Council (VR; grant 2016-02506).

19.
EMBO Rep ; 19(11)2018 11.
Article in English | MEDLINE | ID: mdl-30166336

ABSTRACT

SOX transcription factors have important roles during astrocyte and oligodendrocyte development, but how glial genes are specified and activated in a sub-lineage-specific fashion remains unknown. Here, we define glial-specific gene expression in the developing spinal cord using single-cell RNA-sequencing. Moreover, by ChIP-seq analyses we show that these glial gene sets are extensively preselected already in multipotent neural precursor cells through prebinding by SOX3. In the subsequent lineage-restricted glial precursor cells, astrocyte genes become additionally targeted by SOX9 at DNA regions strongly enriched for Nfi binding motifs. Oligodendrocyte genes instead are prebound by SOX9 only, at sites which during oligodendrocyte maturation are targeted by SOX10. Interestingly, reporter gene assays and functional studies in the spinal cord reveal that SOX3 binding represses the synergistic activation of astrocyte genes by SOX9 and NFIA, whereas oligodendrocyte genes are activated in a combinatorial manner by SOX9 and SOX10. These genome-wide studies demonstrate how sequentially expressed SOX proteins act on lineage-specific regulatory DNA elements to coordinate glial gene expression both in a temporal and in a sub-lineage-specific fashion.


Subject(s)
Astrocytes/physiology , Oligodendroglia/physiology , SOX9 Transcription Factor/genetics , SOXB1 Transcription Factors/genetics , Spinal Cord/cytology , Animals , Cell Differentiation/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Mice , Neural Stem Cells , Neuroglia/cytology , Neuroglia/physiology , SOX9 Transcription Factor/metabolism , SOXB1 Transcription Factors/metabolism , Spinal Cord/growth & development
20.
Dev Cell ; 46(4): 504-517.e7, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30078729

ABSTRACT

Pdgfra+ oligodendrocyte precursor cells (OPCs) arise in distinct specification waves during embryogenesis in the central nervous system (CNS). It is unclear whether there is a correlation between these waves and different oligodendrocyte (OL) states at adult stages. Here, we present bulk and single-cell transcriptomics resources providing insights on how transitions between these states occur. We found that post-natal OPCs from brain and spinal cord present similar transcriptional signatures. Moreover, post-natal OPC progeny of E13.5 Pdgfra+ cells present electrophysiological and transcriptional profiles similar to OPCs derived from subsequent specification waves, indicating that Pdgfra+ pre-OPCs rewire their transcriptional network during development. Single-cell RNA-seq and lineage tracing indicates that a subset of E13.5 Pdgfra+ cells originates cells of the pericyte lineage. Thus, our results indicate that embryonic Pdgfra+ cells in the CNS give rise to distinct post-natal cell lineages, including OPCs with convergent transcriptional profiles in different CNS regions.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Cell Proliferation/physiology , Oligodendroglia/cytology , Animals , Cells, Cultured , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Spinal Cord/metabolism , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL