Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Nat Cancer ; 5(5): 742-759, 2024 May.
Article in English | MEDLINE | ID: mdl-38429414

ABSTRACT

Successful immunotherapy relies on triggering complex responses involving T cell dynamics in tumors and the periphery. Characterizing these responses remains challenging using static human single-cell atlases or mouse models. To address this, we developed a framework for in vivo tracking of tumor-specific CD8+ T cells over time and at single-cell resolution. Our tools facilitate the modeling of gene program dynamics in the tumor microenvironment (TME) and the tumor-draining lymph node (tdLN). Using this approach, we characterize two modes of anti-programmed cell death protein 1 (PD-1) activity, decoupling induced differentiation of tumor-specific activated precursor cells from conventional type 1 dendritic cell (cDC1)-dependent proliferation and recruitment to the TME. We demonstrate that combining anti-PD-1 therapy with anti-4-1BB agonist enhances the recruitment and proliferation of activated precursors, resulting in tumor control. These data suggest that effective response to anti-PD-1 therapy is dependent on sufficient influx of activated precursor CD8+ cells to the TME and highlight the importance of understanding system-level dynamics in optimizing immunotherapies.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy , Tumor Microenvironment , Animals , Mice , Immunotherapy/methods , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Tumor Microenvironment/immunology , Humans , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Dendritic Cells/immunology , Dendritic Cells/drug effects , Cell Line, Tumor
2.
Nat Aging ; 4(1): 129-144, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38062254

ABSTRACT

To understand human longevity, inherent aging processes must be distinguished from known etiologies leading to age-related chronic diseases. Such deconvolution is difficult to achieve because it requires tracking patients throughout their entire lives. Here, we used machine learning to infer health trajectories over the entire adulthood age range using extrapolation from electronic medical records with partial longitudinal coverage. Using this approach, our model tracked the state of patients who were healthy and free from known chronic disease risk and distinguished individuals with higher or lower longevity potential using a multivariate score. We showed that the model and the markers it uses performed consistently on data from Israeli, British and US populations. For example, mildly low neutrophil counts and alkaline phosphatase levels serve as early indicators of healthy aging that are independent of risk for major chronic diseases. We characterize the heritability and genetic associations of our longevity score and demonstrate at least 1 year of extended lifespan for parents of high-scoring patients compared to matched controls. Longitudinal modeling of healthy individuals is thereby established as a tool for understanding healthy aging and longevity.


Subject(s)
Healthy Aging , Humans , Adult , Healthy Aging/genetics , Longevity/genetics , Chronic Disease , Disease Susceptibility , Machine Learning
3.
Genome Biol ; 24(1): 220, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37798781

ABSTRACT

We describe MCProj-an algorithm for analyzing query scRNA-seq data by projections over reference single-cell atlases. We represent the reference as a manifold of annotated metacell gene expression distributions. We then interpret query metacells as mixtures of atlas distributions while correcting for technology-specific gene biases. This approach distinguishes and tags query cells that are consistent with atlas states from unobserved (novel or artifactual) behaviors. It also identifies expression differences observed in successfully mapped query states. We showcase MCProj functionality by projecting scRNA-seq data on a blood cell atlas, deriving precise, quantitative, and interpretable results across technologies and datasets.


Subject(s)
Gene Expression Profiling , Single-Cell Analysis , Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Algorithms
4.
Nat Commun ; 14(1): 3844, 2023 06 29.
Article in English | MEDLINE | ID: mdl-37386027

ABSTRACT

Embryonic development involves massive proliferation and differentiation of cell lineages. This must be supported by chromosome replication and epigenetic reprogramming, but how proliferation and cell fate acquisition are balanced in this process is not well understood. Here we use single cell Hi-C to map chromosomal conformations in post-gastrulation mouse embryo cells and study their distributions and correlations with matching embryonic transcriptional atlases. We find that embryonic chromosomes show a remarkably strong cell cycle signature. Despite that, replication timing, chromosome compartment structure, topological associated domains (TADs) and promoter-enhancer contacts are shown to be variable between distinct epigenetic states. About 10% of the nuclei are identified as primitive erythrocytes, showing exceptionally compact and organized compartment structure. The remaining cells are broadly associated with ectoderm and mesoderm identities, showing only mild differentiation of TADs and compartment structures, but more specific localized contacts in hundreds of ectoderm and mesoderm promoter-enhancer pairs. The data suggest that while fully committed embryonic lineages can rapidly acquire specific chromosomal conformations, most embryonic cells are showing plastic signatures driven by complex and intermixed enhancer landscapes.


Subject(s)
Gastrulation , Regulatory Sequences, Nucleic Acid , Female , Pregnancy , Animals , Mice , Molecular Conformation , Promoter Regions, Genetic/genetics , Chromosomes
5.
Cell ; 186(12): 2610-2627.e18, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37209682

ABSTRACT

The hourglass model describes the convergence of species within the same phylum to a similar body plan during development; however, the molecular mechanisms underlying this phenomenon in mammals remain poorly described. Here, we compare rabbit and mouse time-resolved differentiation trajectories to revisit this model at single-cell resolution. We modeled gastrulation dynamics using hundreds of embryos sampled between gestation days 6.0 and 8.5 and compared the species using a framework for time-resolved single-cell differentiation-flows analysis. We find convergence toward similar cell-state compositions at E7.5, supported by the quantitatively conserved expression of 76 transcription factors, despite divergence in surrounding trophoblast and hypoblast signaling. However, we observed noticeable changes in specification timing of some lineages and divergence of primordial germ cell programs, which in the rabbit do not activate mesoderm genes. Comparative analysis of temporal differentiation models provides a basis for studying the evolution of gastrulation dynamics across mammals.


Subject(s)
Gastrulation , Mesoderm , Animals , Rabbits , Mice , Gastrulation/genetics , Mesoderm/physiology , Cell Differentiation/physiology , Mammals/genetics , Trophoblasts , Gene Expression Regulation, Developmental
6.
Nat Struct Mol Biol ; 29(12): 1252-1265, 2022 12.
Article in English | MEDLINE | ID: mdl-36510023

ABSTRACT

In mammalian embryos, DNA methylation is initialized to maximum levels in the epiblast by the de novo DNA methyltransferases DNMT3A and DNMT3B before gastrulation diversifies it across regulatory regions. Here we show that DNMT3A and DNMT3B are differentially regulated during endoderm and mesoderm bifurcation and study the implications in vivo and in meso-endoderm embryoid bodies. Loss of both Dnmt3a and Dnmt3b impairs exit from the epiblast state. More subtly, independent loss of Dnmt3a or Dnmt3b leads to small biases in mesoderm-endoderm bifurcation and transcriptional deregulation. Epigenetically, DNMT3A and DNMT3B drive distinct methylation kinetics in the epiblast, as can be predicted from their strand-specific sequence preferences. The enzymes compensate for each other in the epiblast, but can later facilitate lineage-specific methylation kinetics as their expression diverges. Single-cell analysis shows that differential activity of DNMT3A and DNMT3B combines with replication-linked methylation turnover to increase epigenetic plasticity in gastrulation. Together, these findings outline a dynamic model for the use of DNMT3A and DNMT3B sequence specificity during gastrulation.


Subject(s)
DNA Methyltransferase 3A , Gastrulation , Animals , Mice , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Embryo, Mammalian/metabolism , DNA/metabolism , Mammals/genetics
7.
Cell ; 185(17): 3169-3185.e20, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35908548

ABSTRACT

Mice deficient for all ten-eleven translocation (TET) genes exhibit early gastrulation lethality. However, separating cause and effect in such embryonic failure is challenging. To isolate cell-autonomous effects of TET loss, we used temporal single-cell atlases from embryos with partial or complete mutant contributions. Strikingly, when developing within a wild-type embryo, Tet-mutant cells retain near-complete differentiation potential, whereas embryos solely comprising mutant cells are defective in epiblast to ectoderm transition with degenerated mesoderm potential. We map de-repressions of early epiblast factors (e.g., Dppa4 and Gdf3) and failure to activate multiple signaling from nascent mesoderm (Lefty, FGF, and Notch) as likely cell-intrinsic drivers of TET loss phenotypes. We further suggest loss of enhancer demethylation as the underlying mechanism. Collectively, our work demonstrates an unbiased approach for defining intrinsic and extrinsic embryonic gene function based on temporal differentiation atlases and disentangles the intracellular effects of the demethylation machinery from its broader tissue-level ramifications.


Subject(s)
Gastrulation , Mesoderm , Animals , Cell Differentiation/genetics , Embryo, Mammalian/metabolism , Gastrulation/genetics , Gene Expression Regulation, Developmental , Mice , Nuclear Proteins/metabolism , Signal Transduction
8.
Cell Stem Cell ; 29(6): 973-989.e10, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35659879

ABSTRACT

The liver carries a remarkable ability to regenerate rapidly after acute zonal damage. Single-cell approaches are necessary to study this process, given the spatial heterogeneity of liver cell types. Here, we use spatially resolved single-cell RNA sequencing (scRNA-seq) to study the dynamics of mouse liver regeneration after acute acetaminophen (APAP) intoxication. We find that hepatocytes proliferate throughout the liver lobule, creating the mitotic pressure required to repopulate the necrotic pericentral zone rapidly. A subset of hepatocytes located at the regenerating front transiently upregulate fetal-specific genes, including Afp and Cdh17, as they reprogram to a pericentral state. Zonated endothelial, hepatic stellate cell (HSC), and macrophage populations are differentially involved in immune recruitment, proliferation, and matrix remodeling. We observe massive transient infiltration of myeloid cells, yet stability of lymphoid cell abundance, in accordance with a global decline in antigen presentation. Our study provides a resource for understanding the coordinated programs of zonal liver regeneration.


Subject(s)
Chemical and Drug Induced Liver Injury , Liver Regeneration , Acetaminophen/metabolism , Acetaminophen/toxicity , Animals , Chemical and Drug Induced Liver Injury/metabolism , Hepatic Stellate Cells , Hepatocytes/metabolism , Liver/metabolism , Mice
9.
Genome Biol ; 23(1): 100, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440087

ABSTRACT

Scaling scRNA-seq to profile millions of cells is crucial for constructing high-resolution maps of transcriptional manifolds. Current analysis strategies, in particular dimensionality reduction and two-phase clustering, offer only limited scaling and sensitivity to define such manifolds. We introduce Metacell-2, a recursive divide-and-conquer algorithm allowing efficient decomposition of scRNA-seq datasets of any size into small and cohesive groups of cells called metacells. Metacell-2 improves outlier cell detection and rare cell type identification, as shown with human bone marrow cell atlas and mouse embryonic data. Metacell-2 is implemented over the scanpy framework for easy integration in any analysis pipeline.


Subject(s)
Algorithms , Single-Cell Analysis , Animals , Cluster Analysis , Mice , Sequence Analysis, RNA , Exome Sequencing
10.
Nat Commun ; 12(1): 5406, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34518533

ABSTRACT

DNA methylation is aberrant in cancer, but the dynamics, regulatory role and clinical implications of such epigenetic changes are still poorly understood. Here, reduced representation bisulfite sequencing (RRBS) profiles of 1538 breast tumors and 244 normal breast tissues from the METABRIC cohort are reported, facilitating detailed analysis of DNA methylation within a rich context of genomic, transcriptional, and clinical data. Tumor methylation from immune and stromal signatures are deconvoluted leading to the discovery of a tumor replication-linked clock with genome-wide methylation loss in non-CpG island sites. Unexpectedly, methylation in most tumor CpG islands follows two replication-independent processes of gain (MG) or loss (ML) that we term epigenomic instability. Epigenomic instability is correlated with tumor grade and stage, TP53 mutations and poorer prognosis. After controlling for these global trans-acting trends, as well as for X-linked dosage compensation effects, cis-specific methylation and expression correlations are uncovered at hundreds of promoters and over a thousand distal elements. Some of these targeted known tumor suppressors and oncogenes. In conclusion, this study demonstrates that global epigenetic instability can erode cancer methylomes and expose them to localized methylation aberrations in-cis resulting in transcriptional changes seen in tumors.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation , Epigenesis, Genetic , Epigenomics/methods , Gene Expression Regulation, Neoplastic , Cohort Studies , CpG Islands/genetics , DNA Replication/genetics , Female , Genome, Human/genetics , Genomic Instability/genetics , Genomics/methods , Humans , MCF-7 Cells , Mutation , Promoter Regions, Genetic/genetics , Survival Analysis
11.
Nat Med ; 27(9): 1582-1591, 2021 09.
Article in English | MEDLINE | ID: mdl-34426707

ABSTRACT

Standardized lab tests are central for patient evaluation, differential diagnosis and treatment. Interpretation of these data is nevertheless lacking quantitative and personalized metrics. Here we report on the modeling of 2.1 billion lab measurements of 92 different lab tests from 2.8 million adults over a span of 18 years. Following unsupervised filtering of 131 chronic conditions and 5,223 drug-test pairs we performed a virtual survey of lab tests distributions in healthy individuals. Age and sex alone explain less than 10% of the within-normal test variance in 89 out of 92 tests. Personalized models based on patients' history explain 60% of the variance for 17 tests and over 36% for half of the tests. This allows for systematic stratification of the risk for future abnormal test levels and subsequent emerging disease. Multivariate modeling of within-normal lab tests can be readily implemented as a basis for quantitative patient evaluation.


Subject(s)
Clinical Laboratory Techniques/standards , Healthy Volunteers , Precision Medicine , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Proportional Hazards Models , Young Adult
12.
Cell ; 184(11): 2825-2842.e22, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33932341

ABSTRACT

Mouse embryonic development is a canonical model system for studying mammalian cell fate acquisition. Recently, single-cell atlases comprehensively charted embryonic transcriptional landscapes, yet inference of the coordinated dynamics of cells over such atlases remains challenging. Here, we introduce a temporal model for mouse gastrulation, consisting of data from 153 individually sampled embryos spanning 36 h of molecular diversification. Using algorithms and precise timing, we infer differentiation flows and lineage specification dynamics over the embryonic transcriptional manifold. Rapid transcriptional bifurcations characterize the commitment of early specialized node and blood cells. However, for most lineages, we observe combinatorial multi-furcation dynamics rather than hierarchical transcriptional transitions. In the mesoderm, dozens of transcription factors combinatorially regulate multifurcations, as we exemplify using time-matched chimeric embryos of Foxc1/Foxc2 mutants. Our study rejects the notion of differentiation being governed by a series of binary choices, providing an alternative quantitative model for cell fate acquisition.


Subject(s)
Embryonic Development/physiology , Gastrulation/physiology , Animals , Cell Differentiation , Cell Lineage , Embryo, Mammalian/cytology , Embryonic Development/genetics , Female , Gene Expression , Mice/embryology , Mice, Inbred C57BL , Mouse Embryonic Stem Cells , Pregnancy , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
13.
Nat Genet ; 52(7): 709-718, 2020 07.
Article in English | MEDLINE | ID: mdl-32601473

ABSTRACT

Propagation of clonal regulatory programs contributes to cancer development. It is poorly understood how epigenetic mechanisms interact with genetic drivers to shape this process. Here, we combine single-cell analysis of transcription and DNA methylation with a Luria-Delbrück experimental design to demonstrate the existence of clonally stable epigenetic memory in multiple types of cancer cells. Longitudinal transcriptional and genetic analysis of clonal colon cancer cell populations reveals a slowly drifting spectrum of epithelial-to-mesenchymal transcriptional identities that is seemingly independent of genetic variation. DNA methylation landscapes correlate with these identities but also reflect an independent clock-like methylation loss process. Methylation variation can be explained as an effect of global trans-acting factors in most cases. However, for a specific class of promoters-in particular, cancer-testis antigens-de-repression is correlated with and probably driven by loss of methylation in cis. This study indicates how genetic sub-clonal structure in cancer cells can be diversified by epigenetic memory.


Subject(s)
Clonal Evolution , Epigenesis, Genetic , Neoplasms/genetics , Neoplasms/pathology , Transcription, Genetic , Cell Line , Cell Proliferation/genetics , DNA Methylation , DNA, Neoplasm/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , RNA, Neoplasm/metabolism , Single-Cell Analysis
15.
Genome Biol ; 20(1): 206, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31604482

ABSTRACT

scRNA-seq profiles each represent a highly partial sample of mRNA molecules from a unique cell that can never be resampled, and robust analysis must separate the sampling effect from biological variance. We describe a methodology for partitioning scRNA-seq datasets into metacells: disjoint and homogenous groups of profiles that could have been resampled from the same cell. Unlike clustering analysis, our algorithm specializes at obtaining granular as opposed to maximal groups. We show how to use metacells as building blocks for complex quantitative transcriptional maps while avoiding data smoothing. Our algorithms are implemented in the MetaCell R/C++ software package.


Subject(s)
Sequence Analysis, RNA , Single-Cell Analysis , Software , Algorithms , CD8-Positive T-Lymphocytes/metabolism , Genomics/methods
16.
Cell ; 176(4): 775-789.e18, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30595452

ABSTRACT

Tumor immune cell compositions play a major role in response to immunotherapy, but the heterogeneity and dynamics of immune infiltrates in human cancer lesions remain poorly characterized. Here, we identify conserved intratumoral CD4 and CD8 T cell behaviors in scRNA-seq data from 25 melanoma patients. We discover a large population of CD8 T cells showing continuous progression from an early effector "transitional" into a dysfunctional T cell state. CD8 T cells that express a complete cytotoxic gene set are rare, and TCR sharing data suggest their independence from the transitional and dysfunctional cell states. Notably, we demonstrate that dysfunctional T cells are the major intratumoral proliferating immune cell compartment and that the intensity of the dysfunctional signature is associated with tumor reactivity. Our data demonstrate that CD8 T cells previously defined as exhausted are in fact a highly proliferating, clonal, and dynamically differentiating cell population within the human tumor microenvironment.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Melanoma/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/immunology , Tumor Microenvironment/immunology
17.
Int J Cancer ; 144(5): 1061-1072, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30350867

ABSTRACT

Lung adenocarcinoma (ADC) is the most prevalent subtype of lung cancer and characterized by considerable morphological and mutational heterogeneity. However, little is known about the epigenomic intratumor variability between spatially separated histological growth patterns of ADC. In order to reconstruct the clonal evolution of histomorphological patterns, we performed global DNA methylation profiling of 27 primary tumor regions, seven matched normal tissues and six lymph node metastases from seven ADC cases. Additionally, we investigated the methylation data from 369 samples of the TCGA ADC cohort. All regions showed varying degrees of methylation changes between segments of different, but also of the same growth patterns. Similarly, copy number variations were seen between spatially distinct segments of each patient. Hierarchical clustering of promoter methylation revealed extensive heterogeneity within and between the cases. Intratumor DNA methylation heterogeneity demonstrated a branched clonal evolution of ADC regions driven by genomic instability with subclonal copy number changes. Notably, methylation profiles within tumors were not more similar to each other than to those from other individuals. In two cases, different tumor regions of the same individuals were represented in distant clusters of the TCGA cohort, illustrating the extensive epigenomic intratumor heterogeneity of ADCs. We found no evidence for the lymph node metastases to be derived from a common growth pattern. Instead, they had evolved early and separately from a particular pattern in each primary tumor. Our results suggest that extensive variation of epigenomic features contributes to the molecular and phenotypic heterogeneity of primary ADCs and lymph node metastases.


Subject(s)
Adenocarcinoma of Lung/genetics , DNA Methylation/genetics , Lung Neoplasms/genetics , Aged , Aged, 80 and over , Clonal Evolution , DNA Copy Number Variations/genetics , Evolution, Molecular , Female , Genetic Heterogeneity , Humans , Male , Middle Aged , Mutation/genetics , Promoter Regions, Genetic/genetics
18.
Cell ; 173(6): 1520-1534.e20, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29856957

ABSTRACT

The emergence and diversification of cell types is a leading factor in animal evolution. So far, systematic characterization of the gene regulatory programs associated with cell type specificity was limited to few cell types and few species. Here, we perform whole-organism single-cell transcriptomics to map adult and larval cell types in the cnidarian Nematostella vectensis, a non-bilaterian animal with complex tissue-level body-plan organization. We uncover eight broad cell classes in Nematostella, including neurons, cnidocytes, and digestive cells. Each class comprises different subtypes defined by the expression of multiple specific markers. In particular, we characterize a surprisingly diverse repertoire of neurons, which comparative analysis suggests are the result of lineage-specific diversification. By integrating transcription factor expression, chromatin profiling, and sequence motif analysis, we identify the regulatory codes that underlie Nematostella cell-specific expression. Our study reveals cnidarian cell type complexity and provides insights into the evolution of animal cell-specific genomic regulation.


Subject(s)
Gene Expression Regulation, Developmental , Neurons/physiology , RNA , Sea Anemones/physiology , Actins/chemistry , Amino Acid Motifs , Animals , Chromatin/metabolism , Cluster Analysis , Gene Expression Profiling , Genome , Genomics , Phylogeny , Sea Anemones/genetics , Sequence Analysis, RNA , Transcriptome , Tubulin/chemistry
19.
Nature ; 540(7632): 296-300, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27919068

ABSTRACT

Chromosomes are folded into highly compacted structures to accommodate physical constraints within nuclei and to regulate access to genomic information. Recently, global mapping of pairwise contacts showed that loops anchoring topological domains (TADs) are highly conserved between cell types and species. Whether pairwise loops synergize to form higher-order structures is still unclear. Here we develop a conformation capture assay to study higher-order organization using chromosomal walks (C-walks) that link multiple genomic loci together into proximity chains in human and mouse cells. This approach captures chromosomal structure at varying scales. Inter-chromosomal contacts constitute only 7-10% of the pairs and are restricted by interfacing TADs. About half of the C-walks stay within one chromosome, and almost half of those are restricted to intra-TAD spaces. C-walks that couple 2-4 TADs indicate stochastic associations between transcriptionally active, early replicating loci. Targeted analysis of thousands of 3-walks anchored at highly expressed genes support pairwise, rather than hub-like, chromosomal topology at active loci. Polycomb-repressed Hox domains are shown by the same approach to enrich for synergistic hubs. Together, the data indicate that chromosomal territories, TADs, and intra-TAD loops are primarily driven by nested, possibly dynamic, pairwise contacts.


Subject(s)
Chromosome Walking , Chromosomes/chemistry , Chromosomes/genetics , Genetic Loci , Nucleic Acid Conformation , Animals , Chromatin/chemistry , Chromatin/genetics , Gene Expression Regulation , Genes, Homeobox , Genetic Loci/genetics , Humans , Imaging, Three-Dimensional , Mice , Polycomb-Group Proteins/metabolism , Stochastic Processes , Transcription, Genetic
20.
PLoS Genet ; 12(11): e1006330, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27846223

ABSTRACT

The development of niches for tissue-specific stem cells is an important aspect of stem cell biology. Determination of niche size and niche numbers during organogenesis involves precise control of gene expression. How this is achieved in the context of a complex chromatin landscape is largely unknown. Here we show that the nuclear protein Combgap (Cg) supports correct ovarian niche formation in Drosophila by controlling ecdysone-Receptor (EcR)- mediated transcription and long-range chromatin contacts in the broad locus (BR-C). Both cg and BR-C promote ovarian growth and the development of niches for germ line stem cells. BR-C levels were lower when Combgap was either reduced or over-expressed, indicating an intricate regulation of the BR-C locus by Combgap. Polytene chromosome stains showed that Cg co-localizes with EcR, the major regulator of BR-C, at the BR-C locus and that EcR binding to chromatin was sensitive to changes in Cg levels. Proximity ligation assay indicated that the two proteins could reside in the same complex. Finally, chromatin conformation analysis revealed that EcR-bound regions within BR-C, which span ~30 KBs, contacted each other. Significantly, these contacts were stabilized in an ecdysone- and Combgap-dependent manner. Together, these results highlight Combgap as a novel regulator of chromatin structure that promotes transcription of ecdysone target genes and ovarian niche formation.


Subject(s)
Chromatin/genetics , Drosophila Proteins/genetics , Receptors, Steroid/genetics , Transcription Factors/genetics , Transcription, Genetic , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Ecdysone/biosynthesis , Ecdysone/genetics , Female , Gene Expression Regulation, Developmental , Organ Specificity , Ovary/growth & development , Ovary/metabolism , Polytene Chromosomes/genetics , Stem Cell Niche/genetics
SELECTION OF CITATIONS
SEARCH DETAIL