Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.291
1.
Development ; 151(9)2024 May 01.
Article En | MEDLINE | ID: mdl-38722217

Animal evolution is influenced by the emergence of new cell types, yet our understanding of this process remains elusive. This prompts the need for a broader exploration across diverse research organisms, facilitated by recent breakthroughs, such as gene editing tools and single-cell genomics. Essential to our understanding of cell type evolution is the accurate identification of homologous cells. We delve into the significance of considering developmental ontogeny and potential pitfalls when drawing conclusions about cell type homology. Additionally, we highlight recent discoveries in the study of cell type evolution through the application of single-cell transcriptomics and pinpoint areas ripe for further exploration.


Biological Evolution , Single-Cell Analysis , Animals , Single-Cell Analysis/methods , Humans , Cell Lineage/genetics , Transcriptome/genetics , Genomics , Gene Editing
2.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article En | MEDLINE | ID: mdl-38731966

Leukemias are among the most prevalent types of cancer worldwide. Bone marrow mesenchymal stem cells (MSCs) participate in the development of a suitable niche for hematopoietic stem cells, and are involved in the development of diseases such as leukemias, to a yet unknown extent. Here we described the effect of secretome of bone marrow MSCs obtained from healthy donors and from patients with acute myeloid leukemia (AML) on leukemic cell lineages, sensitive (K562) or resistant (K562-Lucena) to chemotherapy drugs. Cell proliferation, viability and death were evaluated, together with cell cycle, cytokine production and gene expression of ABC transporters and cyclins. The secretome of healthy MSCs decreased proliferation and viability of both K562 and K562-Lucena cells; moreover, an increase in apoptosis and necrosis rates was observed, together with the activation of caspase 3/7, cell cycle arrest in G0/G1 phase and changes in expression of several ABC proteins and cyclins D1 and D2. These effects were not observed using the secretome of MSCs derived from AML patients. In conclusion, the secretome of healthy MSCs have the capacity to inhibit the development of leukemia cells, at least in the studied conditions. However, MSCs from AML patients seem to have lost this capacity, and could therefore contribute to the development of leukemia.


Cell Proliferation , Leukemia, Myeloid, Acute , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/genetics , K562 Cells , Apoptosis , Secretome/metabolism , Middle Aged , Female , Male , Bone Marrow Cells/metabolism , Cell Lineage/genetics , Cell Survival , Adult
3.
Nat Commun ; 15(1): 4200, 2024 May 17.
Article En | MEDLINE | ID: mdl-38760342

The developmental fate of cells is regulated by intrinsic factors and the extracellular environment. The extracellular matrix (matrisome) delivers chemical and mechanical cues that can modify cellular development. However, comprehensive understanding of how matrisome factors control cells in vivo is lacking. Here we show that specific matrisome factors act individually and collectively to control germ cell development. Surveying development of undifferentiated germline stem cells through to mature oocytes in the Caenorhabditis elegans germ line enabled holistic functional analysis of 443 conserved matrisome-coding genes. Using high-content imaging, 3D reconstruction, and cell behavior analysis, we identify 321 matrisome genes that impact germ cell development, the majority of which (>80%) are undescribed. Our analysis identifies key matrisome networks acting autonomously and non-autonomously to coordinate germ cell behavior. Further, our results demonstrate that germ cell development requires continual remodeling of the matrisome landscape. Together, this study provides a comprehensive platform for deciphering how extracellular signaling controls cellular development and anticipate this will establish new opportunities for manipulating cell fates.


Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cell Differentiation , Extracellular Matrix , Germ Cells , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Extracellular Matrix/metabolism , Germ Cells/metabolism , Germ Cells/cytology , Cell Differentiation/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Gene Expression Regulation, Developmental , Signal Transduction , Cell Lineage/genetics , Oocytes/metabolism , Oocytes/cytology
4.
BMC Genomics ; 25(1): 464, 2024 May 13.
Article En | MEDLINE | ID: mdl-38741085

Gonad development includes sex determination and divergent maturation of the testes and ovaries. Recent advances in measuring gene expression in single cells are providing new insights into this complex process. However, the underlying epigenetic regulatory mechanisms remain unclear. Here, we profiled chromatin accessibility in mouse gonadal cells of both sexes from embryonic day 11.5 to 14.5 using single-cell assay for transposase accessible chromatin by sequencing (scATAC-seq). Our results showed that individual cell types can be inferred by the chromatin landscape, and that cells can be temporally ordered along developmental trajectories. Integrative analysis of transcriptomic and chromatin-accessibility maps identified multiple putative regulatory elements proximal to key gonadal genes Nr5a1, Sox9 and Wt1. We also uncover cell type-specific regulatory factors underlying cell type specification. Overall, our results provide a better understanding of the epigenetic landscape associated with the progressive restriction of cell fates in the gonad.


Cell Lineage , Chromatin , Gonads , SOX9 Transcription Factor , Single-Cell Analysis , Animals , Chromatin/metabolism , Chromatin/genetics , Mice , Cell Lineage/genetics , Female , Male , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Gonads/metabolism , Gonads/cytology , Gonads/embryology , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism , Testis/metabolism , Testis/cytology , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Ovary/metabolism , Ovary/cytology
5.
Mol Cell ; 84(10): 1855-1869.e5, 2024 May 16.
Article En | MEDLINE | ID: mdl-38593804

RNA transcribed from enhancers, i.e., eRNA, has been suggested to directly activate transcription by recruiting transcription factors and co-activators. Although there have been specific examples of eRNA functioning in this way, it is not clear how general this may be. We find that the AT-hook of SWI/SNF preferentially binds RNA and, as part of the esBAF complex, associates with eRNA transcribed from intronic and intergenic regions. Our data suggest that SWI/SNF is globally recruited in cis by eRNA to cell-type-specific enhancers, representative of two distinct stages that mimic early mammalian development, and not at enhancers that are shared between the two stages. In this manner, SWI/SNF facilitates recruitment and/or activation of MLL3/4, p300/CBP, and Mediator to stage-specific enhancers and super-enhancers that regulate the transcription of metabolic and cell lineage priming-related genes. These findings highlight a connection between ATP-dependent chromatin remodeling and eRNA in cell identity and typical- and super-enhancer activation.


Cell Lineage , DNA Helicases , Enhancer Elements, Genetic , Nuclear Proteins , Transcription Factors , Transcription Factors/metabolism , Transcription Factors/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Cell Lineage/genetics , Animals , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Humans , Mice , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics
6.
Nat Commun ; 15(1): 3432, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38653778

Temporal regulation of super-enhancer (SE) driven transcription factors (TFs) underlies normal developmental programs. Neuroblastoma (NB) arises from an inability of sympathoadrenal progenitors to exit a self-renewal program and terminally differentiate. To identify SEs driving TF regulators, we use all-trans retinoic acid (ATRA) to induce NB growth arrest and differentiation. Time-course H3K27ac ChIP-seq and RNA-seq reveal ATRA coordinated SE waves. SEs that decrease with ATRA link to stem cell development (MYCN, GATA3, SOX11). CRISPR-Cas9 and siRNA verify SOX11 dependency, in vitro and in vivo. Silencing the SOX11 SE using dCAS9-KRAB decreases SOX11 mRNA and inhibits cell growth. Other TFs activate in sequential waves at 2, 4 and 8 days of ATRA treatment that regulate neural development (GATA2 and SOX4). Silencing the gained SOX4 SE using dCAS9-KRAB decreases SOX4 expression and attenuates ATRA-induced differentiation genes. Our study identifies oncogenic lineage drivers of NB self-renewal and TFs critical for implementing a differentiation program.


Cell Differentiation , Gene Expression Regulation, Neoplastic , Neuroblastoma , SOXC Transcription Factors , Tretinoin , Neuroblastoma/metabolism , Neuroblastoma/genetics , Neuroblastoma/pathology , Tretinoin/pharmacology , Tretinoin/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , SOXC Transcription Factors/metabolism , SOXC Transcription Factors/genetics , Humans , Animals , Cell Line, Tumor , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Self Renewal/drug effects , Cell Self Renewal/genetics , GATA3 Transcription Factor/metabolism , GATA3 Transcription Factor/genetics , Cell Lineage/genetics , GATA2 Transcription Factor/metabolism , GATA2 Transcription Factor/genetics , CRISPR-Cas Systems , N-Myc Proto-Oncogene Protein/metabolism , N-Myc Proto-Oncogene Protein/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics
7.
Genome Med ; 16(1): 60, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658971

BACKGROUND: Pituitary neuroendocrine tumors (PitNETs) are common gland neoplasms demonstrating distinctive transcription factors. Although the role of immune cells in PitNETs has been widely recognized, the precise immunological environment and its control over tumor cells are poorly understood. METHODS: The heterogeneity, spatial distribution, and clinical significance of macrophages in PitNETs were analyzed using single-cell RNA sequencing (scRNA-seq), bulk RNA-seq, spatial transcriptomics, immunohistochemistry, and multiplexed quantitative immunofluorescence (QIF). Cell viability, cell apoptosis assays, and in vivo subcutaneous xenograft experiments have confirmed that INHBA-ACVR1B influences the process of tumor cell apoptosis. RESULTS: The present study evaluated scRNA-seq data from 23 PitNET samples categorized into 3 primary lineages. The objective was to explore the diversity of tumors and the composition of immune cells across these lineages. Analyzed data from scRNA-seq and 365 bulk RNA sequencing samples conducted in-house revealed the presence of three unique subtypes of tumor immune microenvironment (TIME) in PitNETs. These subtypes were characterized by varying levels of immune infiltration, ranging from low to intermediate to high. In addition, the NR5A1 lineage is primarily associated with the subtype characterized by limited infiltration of immune cells. Tumor-associated macrophages (TAMs) expressing CX3CR1+, C1Q+, and GPNMB+ showed enhanced contact with tumor cells expressing NR5A1 + , TBX19+, and POU1F1+, respectively. This emphasizes the distinct interaction axes between TAMs and tumor cells based on their lineage. Moreover, the connection between CX3CR1+ macrophages and tumor cells via INHBA-ACVR1B regulates tumor cell apoptosis. CONCLUSIONS: In summary, the different subtypes of TIME and the interaction between TAM and tumor cells offer valuable insights into the control of TIME that affects the development of PitNET. These findings can be utilized as prospective targets for therapeutic interventions.


Macrophages , Neuroendocrine Tumors , Pituitary Neoplasms , Single-Cell Analysis , Transcriptome , Tumor Microenvironment , Humans , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/immunology , Neuroendocrine Tumors/metabolism , Pituitary Neoplasms/genetics , Pituitary Neoplasms/immunology , Pituitary Neoplasms/pathology , Pituitary Neoplasms/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Animals , Mice , Macrophages/metabolism , Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Gene Expression Regulation, Neoplastic , Gene Expression Profiling , Phenotype , Apoptosis/genetics , Cell Lineage/genetics
8.
Life Sci Alliance ; 7(7)2024 Jul.
Article En | MEDLINE | ID: mdl-38664022

Neural stem cells (NSCs) reside in discrete regions of the adult mammalian brain where they can differentiate into neurons, astrocytes, and oligodendrocytes. Several studies suggest that mitochondria have a major role in regulating NSC fate. Here, we evaluated mitochondrial properties throughout NSC differentiation and in lineage-specific cells. For this, we used the neurosphere assay model to isolate, expand, and differentiate mouse subventricular zone postnatal NSCs. We found that the levels of proteins involved in mitochondrial fusion (Mitofusin [Mfn] 1 and Mfn 2) increased, whereas proteins involved in fission (dynamin-related protein 1 [DRP1]) decreased along differentiation. Importantly, changes in mitochondrial dynamics correlated with distinct patterns of mitochondrial morphology in each lineage. Particularly, we found that the number of branched and unbranched mitochondria increased during astroglial and neuronal differentiation, whereas the area occupied by mitochondrial structures significantly reduced with oligodendrocyte maturation. In addition, comparing the three lineages, neurons revealed to be the most energetically flexible, whereas astrocytes presented the highest ATP content. Our work identified putative mitochondrial targets to enhance lineage-directed differentiation of mouse subventricular zone-derived NSCs.


Astrocytes , Cell Differentiation , Cell Lineage , Dynamins , Mitochondria , Mitochondrial Dynamics , Neural Stem Cells , Neurons , Oligodendroglia , Animals , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Mitochondria/metabolism , Mice , Cell Differentiation/genetics , Cell Lineage/genetics , Astrocytes/metabolism , Astrocytes/cytology , Oligodendroglia/metabolism , Oligodendroglia/cytology , Neurons/metabolism , Neurons/cytology , Cells, Cultured , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Neurogenesis , Lateral Ventricles/cytology , Lateral Ventricles/metabolism
9.
Cells ; 13(8)2024 Apr 19.
Article En | MEDLINE | ID: mdl-38667322

Although lineage reprogramming from one cell type to another is becoming a breakthrough technology for cell-based therapy, several limitations remain to be overcome, including the low conversion efficiency and subtype specificity. To address these, many studies have been conducted using genetics, chemistry, physics, and cell biology to control transcriptional networks, signaling cascades, and epigenetic modifications during reprogramming. Here, we summarize recent advances in cellular reprogramming and discuss future directions.


Cell Lineage , Cellular Reprogramming , Cellular Reprogramming/genetics , Humans , Animals , Cell Lineage/genetics , Neurons/metabolism , Neurons/cytology , Epigenesis, Genetic , Cell Differentiation/genetics , Signal Transduction
10.
Genome Res ; 34(3): 484-497, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38580401

Transcriptional regulation controls cellular functions through interactions between transcription factors (TFs) and their chromosomal targets. However, understanding the fate conversion potential of multiple TFs in an inducible manner remains limited. Here, we introduce iTF-seq as a method for identifying individual TFs that can alter cell fate toward specific lineages at a single-cell level. iTF-seq enables time course monitoring of transcriptome changes, and with biotinylated individual TFs, it provides a multi-omics approach to understanding the mechanisms behind TF-mediated cell fate changes. Our iTF-seq study in mouse embryonic stem cells identified multiple TFs that trigger rapid transcriptome changes indicative of differentiation within a day of induction. Moreover, cells expressing these potent TFs often show a slower cell cycle and increased cell death. Further analysis using bioChIP-seq revealed that GCM1 and OTX2 act as pioneer factors and activators by increasing gene accessibility and activating the expression of lineage specification genes during cell fate conversion. iTF-seq has utility in both mapping cell fate conversion and understanding cell fate conversion mechanisms.


Cell Differentiation , Transcription Factors , Animals , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Differentiation/genetics , Single-Cell Analysis/methods , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology , Cell Lineage/genetics , Transcriptome , Sequence Analysis, RNA/methods , RNA-Seq/methods , Gene Expression Profiling/methods , RNA, Small Cytoplasmic/genetics , RNA, Small Cytoplasmic/metabolism , Multiomics , Single-Cell Gene Expression Analysis
11.
Development ; 151(10)2024 May 15.
Article En | MEDLINE | ID: mdl-38682291

The planar polarized organization of hair cells in the vestibular maculae is unique because these sensory organs contain two groups of cells with oppositely oriented stereociliary bundles that meet at a line of polarity reversal (LPR). EMX2 is a transcription factor expressed by one hair cell group that reverses the orientation of their bundles, thereby forming the LPR. We generated Emx2-CreERt2 transgenic mice for genetic lineage tracing and demonstrate Emx2 expression before hair cell specification when the nascent utricle and saccule constitute a continuous prosensory domain. Precursors labeled by Emx2-CreERt2 at this stage give rise to hair cells located along one side of the LPR in the mature utricle or saccule, indicating that this boundary is first established in the prosensory domain. Consistent with this, Emx2-CreERt2 lineage tracing in Dreher mutants, where the utricle and saccule fail to segregate, labels a continuous field of cells along one side of a fused utriculo-saccular-cochlear organ. These observations reveal that LPR positioning is pre-determined in the developing prosensory domain, and that EMX2 expression defines lineages of hair cells with oppositely oriented stereociliary bundles.


Cell Lineage , Cell Polarity , Ear, Inner , Homeodomain Proteins , Mice, Transgenic , Transcription Factors , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mice , Cell Lineage/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Ear, Inner/metabolism , Ear, Inner/embryology , Ear, Inner/cytology , Cell Polarity/genetics , Saccule and Utricle/cytology , Saccule and Utricle/metabolism , Saccule and Utricle/embryology , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology
12.
Stem Cell Reports ; 19(5): 744-757, 2024 May 14.
Article En | MEDLINE | ID: mdl-38579711

Precise insertion of fluorescent proteins into lineage-specific genes in human pluripotent stem cells (hPSCs) presents challenges due to low knockin efficiency and difficulties in isolating targeted cells. To overcome these hurdles, we present the modified mRNA (ModRNA)-based Activation for Gene Insertion and Knockin (MAGIK) method. MAGIK operates in two steps: first, it uses a Cas9-2A-p53DD modRNA with a mini-donor plasmid (without a drug selection cassette) to significantly enhance efficiency. Second, a deactivated Cas9 activator modRNA and a 'dead' guide RNA are used to temporarily activate the targeted gene, allowing for live cell sorting of targeted cells. Consequently, MAGIK eliminates the need for drug selection cassettes or labor-intensive single-cell colony screening, expediting precise gene editing. We showed MAGIK can be utilized to insert fluorescent proteins into various genes, including SOX17, NKX6.1, NKX2.5, and PDX1, across multiple hPSC lines. This underscores its robust performance and offers a promising solution for achieving knockin in hPSCs within a significantly shortened time frame.


Cell Lineage , Gene Knock-In Techniques , Pluripotent Stem Cells , Humans , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Cell Lineage/genetics , Gene Knock-In Techniques/methods , Genes, Reporter , CRISPR-Cas Systems , RNA, Guide, CRISPR-Cas Systems/genetics , Cell Line , Gene Editing/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Stem Cell Reports ; 19(5): 618-628, 2024 May 14.
Article En | MEDLINE | ID: mdl-38579708

SOX2 is a transcription factor involved in the regulatory network maintaining the pluripotency of embryonic stem cells in culture as well as in early embryos. In addition, SOX2 plays a pivotal role in neural stem cell formation and neurogenesis. How SOX2 can serve both processes has remained elusive. Here, we identified a set of SOX2-dependent neural-associated enhancers required for neural lineage priming. They form a distinct subgroup (1,898) among 8,531 OCT4/SOX2/NANOG-bound enhancers characterized by enhanced SOX2 binding and chromatin accessibility. Activation of these enhancers is triggered by neural induction of wild-type cells or by default in Smad4-ablated cells resistant to mesoderm induction and is antagonized by mesodermal transcription factors via Sox2 repression. Our data provide mechanistic insight into the transition from the pluripotency state to the early neural fate and into the regulation of early neural versus mesodermal specification in embryonic stem cells and embryos.


Enhancer Elements, Genetic , Mesoderm , Neural Stem Cells , SOXB1 Transcription Factors , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Animals , Mice , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Mesoderm/cytology , Mesoderm/metabolism , Neurogenesis , Gene Expression Regulation, Developmental , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/genetics , Cell Differentiation/genetics , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , Cell Lineage/genetics , Smad4 Protein/metabolism , Smad4 Protein/genetics , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology , Chromatin/metabolism , Protein Binding
14.
Cell Rep Methods ; 4(5): 100763, 2024 May 20.
Article En | MEDLINE | ID: mdl-38670101

Cellular barcoding is a lineage-tracing methodology that couples heritable synthetic barcodes to high-throughput sequencing, enabling the accurate tracing of cell lineages across a range of biological contexts. Recent studies have extended these methods by incorporating lineage information into single-cell or spatial transcriptomics readouts. Leveraging the rich biological information within these datasets requires dedicated computational tools for dataset pre-processing and analysis. Here, we present BARtab, a portable and scalable Nextflow pipeline, and bartools, an open-source R package, designed to provide an integrated end-to-end cellular barcoding analysis toolkit. BARtab and bartools contain methods to simplify the extraction, quality control, analysis, and visualization of lineage barcodes from population-level, single-cell, and spatial transcriptomics experiments. We showcase the utility of our integrated BARtab and bartools workflow via the analysis of exemplar bulk, single-cell, and spatial transcriptomics experiments containing cellular barcoding information.


High-Throughput Nucleotide Sequencing , Single-Cell Analysis , Transcriptome , Single-Cell Analysis/methods , Humans , Software , DNA Barcoding, Taxonomic/methods , Genome/genetics , Cell Lineage/genetics , Gene Expression Profiling/methods , Computational Biology/methods , Animals
15.
Nature ; 629(8011): 384-392, 2024 May.
Article En | MEDLINE | ID: mdl-38600385

Debate remains around the anatomical origins of specific brain cell subtypes and lineage relationships within the human forebrain1-7. Thus, direct observation in the mature human brain is critical for a complete understanding of its structural organization and cellular origins. Here we utilize brain mosaic variation within specific cell types as distinct indicators for clonal dynamics, denoted as cell-type-specific mosaic variant barcode analysis. From four hemispheres and two different human neurotypical donors, we identified 287 and 780 mosaic variants, respectively, that were used to deconvolve clonal dynamics. Clonal spread and allele fractions within the brain reveal that local hippocampal excitatory neurons are more lineage-restricted than resident neocortical excitatory neurons or resident basal ganglia GABAergic inhibitory neurons. Furthermore, simultaneous genome transcriptome analysis at both a cell-type-specific and a single-cell level suggests a dorsal neocortical origin for a subgroup of DLX1+ inhibitory neurons that disperse radially from an origin shared with excitatory neurons. Finally, the distribution of mosaic variants across 17 locations within one parietal lobe reveals that restriction of clonal spread in the anterior-posterior axis precedes restriction in the dorsal-ventral axis for both excitatory and inhibitory neurons. Thus, cell-type-resolved somatic mosaicism can uncover lineage relationships governing the development of the human forebrain.


Cell Lineage , GABAergic Neurons , Homeodomain Proteins , Mosaicism , Prosencephalon , Transcription Factors , Humans , Prosencephalon/cytology , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Cell Lineage/genetics , Male , Transcription Factors/metabolism , Transcription Factors/genetics , Neurons/cytology , Neurons/metabolism , Female , Hippocampus/cytology , Clone Cells/cytology , Clone Cells/metabolism , Single-Cell Analysis , Parietal Lobe/cytology , Alleles , Neocortex/cytology , Transcriptome
16.
Nature ; 629(8011): 458-466, 2024 May.
Article En | MEDLINE | ID: mdl-38658765

Heteroplasmy occurs when wild-type and mutant mitochondrial DNA (mtDNA) molecules co-exist in single cells1. Heteroplasmy levels change dynamically in development, disease and ageing2,3, but it is unclear whether these shifts are caused by selection or drift, and whether they occur at the level of cells or intracellularly. Here we investigate heteroplasmy dynamics in dividing cells by combining precise mtDNA base editing (DdCBE)4 with a new method, SCI-LITE (single-cell combinatorial indexing leveraged to interrogate targeted expression), which tracks single-cell heteroplasmy with ultra-high throughput. We engineered cells to have synonymous or nonsynonymous complex I mtDNA mutations and found that cell populations in standard culture conditions purge nonsynonymous mtDNA variants, whereas synonymous variants are maintained. This suggests that selection dominates over simple drift in shaping population heteroplasmy. We simultaneously tracked single-cell mtDNA heteroplasmy and ancestry, and found that, although the population heteroplasmy shifts, the heteroplasmy of individual cell lineages remains stable, arguing that selection acts at the level of cell fitness in dividing cells. Using these insights, we show that we can force cells to accumulate high levels of truncating complex I mtDNA heteroplasmy by placing them in environments where loss of biochemical complex I activity has been reported to benefit cell fitness. We conclude that in dividing cells, a given nonsynonymous mtDNA heteroplasmy can be harmful, neutral or even beneficial to cell fitness, but that the 'sign' of the effect is wholly dependent on the environment.


Cell Division , Cell Lineage , DNA, Mitochondrial , Genetic Fitness , Heteroplasmy , Selection, Genetic , Single-Cell Analysis , Animals , Female , Humans , Mice , Cell Division/genetics , Cell Line , Cell Lineage/genetics , DNA, Mitochondrial/genetics , Gene Editing , Heteroplasmy/genetics , Mitochondria/genetics , Mutation , Single-Cell Analysis/methods
17.
Nature ; 629(8012): 688-696, 2024 May.
Article En | MEDLINE | ID: mdl-38658752

Although cancer initiation and progression are generally associated with the accumulation of somatic mutations1,2, substantial epigenomic alterations underlie many aspects of tumorigenesis and cancer susceptibility3-6, suggesting that genetic mechanisms might not be the only drivers of malignant transformation7. However, whether purely non-genetic mechanisms are sufficient to initiate tumorigenesis irrespective of mutations has been unknown. Here, we show that a transient perturbation of transcriptional silencing mediated by Polycomb group proteins is sufficient to induce an irreversible switch to a cancer cell fate in Drosophila. This is linked to the irreversible derepression of genes that can drive tumorigenesis, including members of the JAK-STAT signalling pathway and zfh1, the fly homologue of the ZEB1 oncogene, whose aberrant activation is required for Polycomb perturbation-induced tumorigenesis. These data show that a reversible depletion of Polycomb proteins can induce cancer in the absence of driver mutations, suggesting that tumours can emerge through epigenetic dysregulation leading to inheritance of altered cell fates.


Drosophila Proteins , Drosophila melanogaster , Epigenesis, Genetic , Janus Kinases , Neoplasms , Polycomb-Group Proteins , STAT Transcription Factors , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Polycomb-Group Proteins/metabolism , Polycomb-Group Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , STAT Transcription Factors/metabolism , STAT Transcription Factors/genetics , Janus Kinases/metabolism , Janus Kinases/genetics , Female , Carcinogenesis/genetics , Male , Signal Transduction/genetics , Gene Silencing , Cell Transformation, Neoplastic/genetics , Cell Lineage/genetics , Gene Expression Regulation, Neoplastic
18.
Immunol Rev ; 323(1): 107-117, 2024 May.
Article En | MEDLINE | ID: mdl-38563448

Group 1 innate lymphoid cells (ILCs), comprising ILC1s and natural killer cells (NK cells), belong to a large family of developmentally related innate lymphoid cells that lack rearranged antigen-specific receptors. NK cells and ILC1s both require the transcription factor T-bet for lineage commitment but additionally rely on Eomes and Hobit, respectively, for their development and effector maturation programs. Both ILC1s and NK cells are essential for rapid responses against infections and mediate cancer immunity through production of effector cytokines and cytotoxicity mediators. ILC1s are enriched in tissues and hence generally considered tissue resident cells whereas NK cells are often considered circulatory. Despite being deemed different cell types, ILC1s and NK cells share many common features both phenotypically and functionally. Recent studies employing single cell RNA sequencing (scRNA-seq) technology have exposed previously unappreciated heterogeneity in group 1 ILCs and further broaden our understanding of these cells. Findings from these studies imply that ILC1s in different tissues and organs share a common signature but exhibit some unique characteristics, possibly stemming from tissue imprinting. Also, data from recent fate mapping studies employing Hobit, RORγt, and polychromic reporter mice have greatly advanced our understanding of the developmental and effector maturation programs of these cells. In this review, we aim to outline the fundamental traits of mouse group 1 ILCs and explore recent discoveries related to their developmental programs, phenotypic heterogeneity, plasticity, and transcriptional regulation.


Cell Plasticity , Gene Expression Regulation , Immunity, Innate , Killer Cells, Natural , Animals , Humans , Cell Plasticity/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Cell Differentiation , Cell Lineage/genetics , Transcription, Genetic , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism
19.
Sci Adv ; 10(11): eadk7160, 2024 Mar 15.
Article En | MEDLINE | ID: mdl-38489358

During development, cells make switch-like decisions to activate new gene programs specifying cell lineage. The mechanisms underlying these decisive choices remain unclear. Here, we show that the cardiovascular transcriptional coactivator myocardin (MYOCD) activates cell identity genes by concentration-dependent and switch-like formation of transcriptional condensates. MYOCD forms such condensates and activates cell identity genes at critical concentration thresholds achieved during smooth muscle cell and cardiomyocyte differentiation. The carboxyl-terminal disordered region of MYOCD is necessary and sufficient for condensate formation. Disrupting this region's ability to form condensates disrupts gene activation and smooth muscle cell reprogramming. Rescuing condensate formation by replacing this region with disordered regions from functionally unrelated proteins rescues gene activation and smooth muscle cell reprogramming. Our findings demonstrate that MYOCD condensate formation is required for gene activation during cardiovascular differentiation. We propose that the formation of transcriptional condensates at critical concentrations of cell type-specific regulators provides a molecular switch underlying the activation of key cell identity genes during development.


Myocytes, Smooth Muscle , Transcription Factors , Cell Lineage/genetics , Cell Differentiation/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Myocytes, Smooth Muscle/metabolism , Transcriptional Activation
20.
Nat Commun ; 15(1): 2783, 2024 Mar 30.
Article En | MEDLINE | ID: mdl-38555276

Elucidating the expression of microRNAs in developing single cells is critical for functional discovery. Here, we construct scCAMERA (single-cell cartography of microRNA expression based on reporter assay), utilizing promoter-driven fluorescent reporters in conjunction with imaging and lineage tracing. The cartography delineates the transcriptional activity of 54 conserved microRNAs in lineage-resolved single cells throughout C. elegans embryogenesis. The combinatorial expression of microRNAs partitions cells into fine clusters reflecting their function and anatomy. Notably, the expression of individual microRNAs exhibits high cell specificity and divergence among family members. Guided by cellular expression patterns, we identify developmental functions of specific microRNAs, including miR-1 in pharynx development and physiology, miR-232 in excretory canal morphogenesis by repressing NHR-25/NR5A, and a functional synergy between miR-232 and miR-234 in canal development, demonstrating the broad utility of scCAMERA. Furthermore, integrative analysis reveals that tissue-specific fate determinants activate microRNAs to repress protein production from leaky transcripts associated with alternative, especially neuronal, fates, thereby enhancing the fidelity of developmental fate differentiation. Collectively, our study offers rich opportunities for multidimensional expression-informed analysis of microRNA biology in metazoans.


MicroRNAs , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Caenorhabditis elegans/metabolism , Cell Lineage/genetics , Cell Differentiation/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental
...