Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Blood ; 2024 May 28.
Article in English | MEDLINE | ID: mdl-38805639

ABSTRACT

Loss of long-term hematopoietic stem cell (LT-HSC) function ex vivo hampers the success of clinical protocols reliant on culture. However, the kinetics and mechanisms by which this occurs remain incompletely characterized. Here, through time-resolved scRNA-Seq, matched in vivo functional analysis and the use of a reversible in vitro system of early G1 arrest, we define the sequence of transcriptional and functional events occurring during the first ex vivo division of human LT-HSCs. We demonstrate that the sharpest loss of LT-HSC repopulation capacity happens early on, between 6 and 24 hours of culture, before LT-HSCs commit to cell cycle progression. During this time window, LT-HSCs adapt to the culture environment, limiting global variability in gene expression and transiently upregulating gene networks involved in signaling and stress responses. From 24 hours, LT-HSC progression past early G1 contributes to the establishment of differentiation programmes in culture. However, contrary to current assumptions, we demonstrate that loss of HSC function ex vivo is independent of cell cycle progression. Finally, we show that targeting LT-HSC adaptation to culture by inhibiting early activation of JAK/STAT signaling improves HSC long-term repopulating function ex vivo. Collectively, our study demonstrates that controlling early LT-HSC adaptation to ex vivo culture, for example via JAK inhibition, is of critical importance to improve HSC gene therapy and expansion protocols.

2.
Biol Open ; 13(1)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38149716

ABSTRACT

As cells exit the pluripotent state and begin to commit to a specific lineage they must activate genes appropriate for that lineage while silencing genes associated with pluripotency and preventing activation of lineage-inappropriate genes. The Nucleosome Remodelling and Deacetylation (NuRD) complex is essential for pluripotent cells to successfully undergo lineage commitment. NuRD controls nucleosome density at regulatory sequences to facilitate transcriptional responses, and also has been shown to prevent unscheduled transcription (transcriptional noise) in undifferentiated pluripotent cells. How these activities combine to ensure cells engage a gene expression program suitable for successful lineage commitment has not been determined. Here, we show that NuRD is not required to silence all genes. Rather, it restricts expression of genes primed for activation upon exit from the pluripotent state, but maintains them in a transcriptionally permissive state in self-renewing conditions, which facilitates their subsequent activation upon exit from naïve pluripotency. We further show that NuRD coordinates gene expression changes, which acts to maintain a barrier between different stable states. Thus NuRD-mediated chromatin remodelling serves multiple functions, including reducing transcriptional noise, priming genes for activation and coordinating the transcriptional response to facilitate lineage commitment.


Subject(s)
DNA-Binding Proteins , Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Nucleosomes , Cell Differentiation/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics
3.
Dermatologie (Heidelb) ; 73(10): 788-794, 2022 Oct.
Article in German | MEDLINE | ID: mdl-35925212

ABSTRACT

We conducted a retrospective data analysis of 26 patients with chronic spontaneous urticaria (CSU), 12 of whom had been treated with anti-IgE therapy (omalizumab). The subcohort of patients treated with omalizumab displayed more severe and prolonged courses of disease. In addition, they had often undergone various inpatient therapies, frequently presenting with concomitant angioedema. Collecting the Urticaria Activity Scores from the seven daily values for wheals and itching (UAS7) proved an important and suitable instrument for the determination and assessment of the course of therapy in the dermatological office. Elaborate laboratory screenings, however, seem far less indicative of the severity, prognosis and course of the disease. Omalizumab proved to be a viable and well-tolerated treatment option. One third of the patients were completely free of all symptoms, another third showed very good improvement, whereas the last third showed no improvement at all, even when omalizumab and/or concomitant therapies were escalated.


Subject(s)
Anti-Allergic Agents , Urticaria , Anti-Allergic Agents/therapeutic use , Antibodies, Anti-Idiotypic , Chronic Disease , Humans , Immunosuppressive Agents/therapeutic use , Omalizumab/therapeutic use , Retrospective Studies , Treatment Outcome , Urticaria/drug therapy
4.
Stem Cell Reports ; 17(8): 1788-1798, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35905741

ABSTRACT

To generate sufficient numbers of transplantable hematopoietic stem cells (HSCs) in vitro, a detailed understanding of how this process takes place in vivo is essential. The endothelial-to-hematopoietic transition (EHT), which culminates in the production of the first HSCs, is a highly complex process during which key regulators are switched on and off at precise moments, and that is embedded into a myriad of microenvironmental signals from surrounding cells and tissues. We have previously demonstrated an HSC-supportive function for GATA3 within the sympathetic nervous system and the sub-aortic mesenchyme, but show here that it also plays a cell-intrinsic role during the EHT. It is expressed in hemogenic endothelial cells and early HSC precursors, where its expression correlates with a more quiescent state. Importantly, endothelial-specific deletion of Gata3 shows that it is functionally required for these cells to mature into HSCs, placing GATA3 at the core of the EHT regulatory network.


Subject(s)
Hemangioblasts , Hematopoietic Stem Cells , Cell Differentiation/genetics , Endothelium , Gonads , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Mesoderm , Mesonephros
5.
Hautarzt ; 72(12): 1071-1078, 2021 Dec.
Article in German | MEDLINE | ID: mdl-34328514

ABSTRACT

We present the results of a retrospective data analysis of a practice cohort of 44 patients with atopic dermatitis treated with the IL-4/13 receptor antibody dupilumab for up to 3 years. Patients were followed up over a period of 21 months during specialized consultation hours named Immunodermatology, which was established to guarantee comprehensive documentation. The patient's characteristics regarding age and sex distribution, severity and duration of disease were comparable with the patient collectives in large, pivotal studies. The therapeutic efficiency however was high (percentage of patients with EASI50, -75, -90 after 16 weeks: 94, 84, 60%, respectively) and long lasting (86% EASI90 after 52 weeks on therapy) under everyday conditions in the clinical setting. Approximately half of the patients had facial skin or eye involvement either in their history or at the start of treatment. This group of patients proved to need more and intense care because facial dermatitis and periocular dermatitis, which often involved conjunctivitis, took longer to heal, relapses occurred, and an additional topical treatment was often required. We did not observe any severe side effects during the 48 patient-years analyzed in this study. Dupilumab proved to be a safe and efficient treatment for atopic dermatitis in dermatological practice.


Subject(s)
Dermatitis, Atopic , Antibodies, Monoclonal, Humanized , Cohort Studies , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/drug therapy , Humans , Retrospective Studies , Severity of Illness Index , Treatment Outcome
6.
Stem Cell Reports ; 16(6): 1614-1628, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33961793

ABSTRACT

Advances in the isolation and gene expression profiling of single hematopoietic stem cells (HSCs) have permitted in-depth resolution of their molecular program. However, long-term HSCs can only be isolated to near purity from adult mouse bone marrow, thereby precluding studies of their molecular program in different physiological states. Here, we describe a powerful 7-day HSC hibernation culture system that maintains HSCs as single cells in the absence of a physical niche. Single hibernating HSCs retain full functional potential compared with freshly isolated HSCs with respect to colony-forming capacity and transplantation into primary and secondary recipients. Comparison of hibernating HSC molecular profiles to their freshly isolated counterparts showed a striking degree of molecular similarity, further resolving the core molecular machinery of HSC self-renewal while also identifying key factors that are potentially dispensable for HSC function, including members of the AP1 complex (Jun, Fos, and Ncor2), Sult1a1 and Cish. Finally, we provide evidence that hibernating mouse HSCs can be transduced without compromising their self-renewal activity and demonstrate the applicability of hibernation cultures to human HSCs.


Subject(s)
Arylsulfotransferase/metabolism , Cell Culture Techniques/methods , Hematopoietic Stem Cells/physiology , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription Factor AP-1/metabolism , Transcriptome , Animals , Bone Marrow Transplantation/methods , Cell Cycle , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Hibernation , Mice , Mice, Inbred C57BL , Multiprotein Complexes/metabolism , Single-Cell Analysis , Stem Cell Niche
7.
Allergy ; 76(6): 1731-1742, 2021 06.
Article in English | MEDLINE | ID: mdl-33078414

ABSTRACT

BACKGROUND: Basophils and mast cells contribute to the development of allergic reactions. Whereas these mature effector cells are extensively studied, the differentiation trajectories from hematopoietic progenitors to basophils and mast cells are largely uncharted at the single-cell level. METHODS: We performed multicolor flow cytometry, high-coverage single-cell RNA sequencing analyses, and cell fate assays to chart basophil and mast cell differentiation at single-cell resolution in mouse. RESULTS: Analysis of flow cytometry data reconstructed a detailed map of basophil and mast cell differentiation, including a bifurcation of progenitors into two specific trajectories. Molecular profiling and pseudotime ordering of the single cells revealed gene expression changes during differentiation. Cell fate assays showed that multicolor flow cytometry and transcriptional profiling successfully predict the bipotent phenotype of a previously uncharacterized population of peritoneal basophil-mast cell progenitors. CONCLUSIONS: A combination of molecular and functional profiling of bone marrow and peritoneal cells provided a detailed road map of basophil and mast cell development. An interactive web resource was created to enable the wider research community to explore the expression dynamics for any gene of interest.


Subject(s)
Basophils , Mast Cells , Animals , Bone Marrow Cells , Cell Differentiation , Mice , Stem Cells
8.
Exp Hematol ; 76: 1-12.e5, 2019 08.
Article in English | MEDLINE | ID: mdl-31326613

ABSTRACT

Pluripotent stem cell (PSC) differentiation in vitro represents a powerful and tractable model to study mammalian development and an unlimited source of cells for regenerative medicine. Within hematology, in vitro PSC hematopoiesis affords novel insights into blood formation and represents an exciting potential approach to generate hematopoietic and immune cell types for transplantation and transfusion. Most studies to date have focused on in vitro hematopoiesis from mouse PSCs and human PSCs. However, differences in mouse and human PSC culture protocols have complicated the translation of discoveries between these systems. We recently developed a novel chemical media formulation, expanded potential stem cell medium (EPSCM), that maintains mouse PSCs in a unique cellular state and extraembryonic differentiation capacity. Herein, we describe how EPSCM can be directly used to stably maintain human PSCs. We further demonstrate that human PSCs maintained in EPSCM can spontaneously form embryoid bodies and undergo in vitro hematopoiesis using a simple differentiation protocol, similar to mouse PSC differentiation. EPSCM-maintained human PSCs generated at least two hematopoietic cell populations, which displayed distinct transcriptional profiles by RNA-sequencing (RNA-seq) analysis. EPSCM also supports gene targeting using homologous recombination, affording generation of an SPI1 (PU.1) reporter PSC line to study and track in vitro hematopoiesis. EPSCM therefore provides a useful tool not only to study pluripotency but also hematopoietic cell specification and developmental-lineage commitment.


Subject(s)
Culture Media/pharmacology , Hematopoiesis/drug effects , Human Embryonic Stem Cells/drug effects , Pluripotent Stem Cells/drug effects , Animals , Cell Culture Techniques/methods , Cell Cycle , Cell Lineage , Cells, Cultured , Cellular Reprogramming Techniques , Embryoid Bodies/drug effects , Fibroblasts/cytology , Genes, Reporter , Human Embryonic Stem Cells/cytology , Humans , Mice , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/transplantation , Sequence Analysis, RNA , Species Specificity , Stem Cell Transplantation/adverse effects , Teratoma/etiology
9.
Cell Stem Cell ; 24(5): 785-801.e7, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31031137

ABSTRACT

The gene regulatory network (GRN) of naive mouse embryonic stem cells (ESCs) must be reconfigured to enable lineage commitment. TCF3 sanctions rewiring by suppressing components of the ESC transcription factor circuitry. However, TCF3 depletion only delays and does not prevent transition to formative pluripotency. Here, we delineate additional contributions of the ETS-family transcription factor ETV5 and the repressor RBPJ. In response to ERK signaling, ETV5 switches activity from supporting self-renewal and undergoes genome relocation linked to commissioning of enhancers activated in formative epiblast. Independent upregulation of RBPJ prevents re-expression of potent naive factors, TBX3 and NANOG, to secure exit from the naive state. Triple deletion of Etv5, Rbpj, and Tcf3 disables ESCs, such that they remain largely undifferentiated and locked in self-renewal, even in the presence of differentiation stimuli. Thus, genetic elimination of three complementary drivers of network transition stalls developmental progression, emulating environmental insulation by small-molecule inhibitors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Neurons/physiology , Pluripotent Stem Cells/physiology , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cell Line , Cell Lineage , Cell Self Renewal , DNA-Binding Proteins/genetics , Gene Knockout Techniques , Gene Regulatory Networks , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , RNA, Small Interfering/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics
10.
Nat Commun ; 9(1): 4100, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30291229

ABSTRACT

Capturing where and how multipotency is lost is crucial to understand how blood formation is controlled. Blood lineage specification is currently thought to occur downstream of multipotent haematopoietic stem cells (HSC). Here we show that, in human, the first lineage restriction events occur within the CD19-CD34+CD38-CD45RA-CD49f+CD90+ (49f+) HSC compartment to generate myelo-lymphoid committed cells with no erythroid differentiation capacity. At single-cell resolution, we observe a continuous but polarised organisation of the 49f+ compartment, where transcriptional programmes and lineage potential progressively change along a gradient of opposing cell surface expression of CLEC9A and CD34. CLEC9AhiCD34lo cells contain long-term repopulating multipotent HSCs with slow quiescence exit kinetics, whereas CLEC9AloCD34hi cells are restricted to myelo-lymphoid differentiation and display infrequent but durable repopulation capacity. We thus propose that human HSCs gradually transition to a discrete lymphoid-primed state, distinct from lymphoid-primed multipotent progenitors, representing the earliest entry point into lymphoid commitment.


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells/physiology , Cell Lineage , Humans , Multipotent Stem Cells/physiology
11.
Blood ; 131(21): e1-e11, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29588278

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) maintain the adult blood system, and their dysregulation causes a multitude of diseases. However, the differentiation journeys toward specific hematopoietic lineages remain ill defined, and system-wide disease interpretation remains challenging. Here, we have profiled 44 802 mouse bone marrow HSPCs using single-cell RNA sequencing to provide a comprehensive transcriptional landscape with entry points to 8 different blood lineages (lymphoid, megakaryocyte, erythroid, neutrophil, monocyte, eosinophil, mast cell, and basophil progenitors). We identified a common basophil/mast cell bone marrow progenitor and characterized its molecular profile at the single-cell level. Transcriptional profiling of 13 815 HSPCs from the c-Kit mutant (W41/W41) mouse model revealed the absence of a distinct mast cell lineage entry point, together with global shifts in cell type abundance. Proliferative defects were accompanied by reduced Myc expression. Potential compensatory processes included upregulation of the integrated stress response pathway and downregulation of proapoptotic gene expression in erythroid progenitors, thus providing a template of how large-scale single-cell transcriptomic studies can bridge between molecular phenotypes and quantitative population changes.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mutation , Proto-Oncogene Proteins c-kit/deficiency , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cells, Cultured , Gene Expression Profiling , Mice , Mice, Knockout , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction , Single-Cell Analysis , Transcriptome
12.
Blood ; 128(8): e20-31, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27365425

ABSTRACT

Maintenance of the blood system requires balanced cell fate decisions by hematopoietic stem and progenitor cells (HSPCs). Because cell fate choices are executed at the individual cell level, new single-cell profiling technologies offer exciting possibilities for mapping the dynamic molecular changes underlying HSPC differentiation. Here, we have used single-cell RNA sequencing to profile more than 1600 single HSPCs, and deep sequencing has enabled detection of an average of 6558 protein-coding genes per cell. Index sorting, in combination with broad sorting gates, allowed us to retrospectively assign cells to 12 commonly sorted HSPC phenotypes while also capturing intermediate cells typically excluded by conventional gating. We further show that independently generated single-cell data sets can be projected onto the single-cell resolution expression map to directly compare data from multiple groups and to build and refine new hypotheses. Reconstruction of differentiation trajectories reveals dynamic expression changes associated with early lymphoid, erythroid, and granulocyte-macrophage differentiation. The latter two trajectories were characterized by common upregulation of cell cycle and oxidative phosphorylation transcriptional programs. By using external spike-in controls, we estimate absolute messenger RNA (mRNA) levels per cell, showing for the first time that despite a general reduction in total mRNA, a subset of genes shows higher expression levels in immature stem cells consistent with active maintenance of the stem-cell state. Finally, we report the development of an intuitive Web interface as a new community resource to permit visualization of gene expression in HSPCs at single-cell resolution for any gene of choice.


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells/cytology , Single-Cell Analysis/methods , Animals , Biomarkers/metabolism , Cell Cycle/genetics , Cell Differentiation/genetics , Female , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Mice, Inbred C57BL , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic
13.
Development ; 142(12): 2121-35, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26015544

ABSTRACT

The transcription factor brachyury (T, BRA) is one of the first markers of gastrulation and lineage specification in vertebrates. Despite its wide use and importance in stem cell and developmental biology, its functional genomic targets in human cells are largely unknown. Here, we use differentiating human embryonic stem cells to study the role of BRA in activin A-induced endoderm and BMP4-induced mesoderm progenitors. We show that BRA has distinct genome-wide binding landscapes in these two cell populations, and that BRA interacts and collaborates with SMAD1 or SMAD2/3 signalling to regulate the expression of its target genes in a cell-specific manner. Importantly, by manipulating the levels of BRA in cells exposed to different signalling environments, we demonstrate that BRA is essential for mesoderm but not for endoderm formation. Together, our data illuminate the function of BRA in the context of human embryonic development and show that the regulatory role of BRA is context dependent. Our study reinforces the importance of analysing the functions of a transcription factor in different cellular and signalling environments.


Subject(s)
Embryonic Stem Cells/cytology , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental , Neurogenesis/physiology , Smad1 Protein/metabolism , T-Box Domain Proteins/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Cell Line , Embryonic Stem Cells/metabolism , Endoderm/cytology , Gastrulation/physiology , Humans , Mesoderm/cytology , Mice , Mice, Transgenic , Smad2 Protein/metabolism , Smad3 Protein/metabolism
14.
Cell Stem Cell ; 16(6): 712-24, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26004780

ABSTRACT

Heterogeneity within the self-renewal durability of adult hematopoietic stem cells (HSCs) challenges our understanding of the molecular framework underlying HSC function. Gene expression studies have been hampered by the presence of multiple HSC subtypes and contaminating non-HSCs in bulk HSC populations. To gain deeper insight into the gene expression program of murine HSCs, we combined single-cell functional assays with flow cytometric index sorting and single-cell gene expression assays. Through bioinformatic integration of these datasets, we designed an unbiased sorting strategy that separates non-HSCs away from HSCs, and single-cell transplantation experiments using the enriched population were combined with RNA-seq data to identify key molecules that associate with long-term durable self-renewal, producing a single-cell molecular dataset that is linked to functional stem cell activity. Finally, we demonstrated the broader applicability of this approach for linking key molecules with defined cellular functions in another stem cell system.


Subject(s)
Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Single-Cell Analysis/methods , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation , Clone Cells , Gene Expression Profiling , Genome , Hematopoietic Stem Cell Transplantation , Humans , Mice, Inbred C57BL
15.
J Craniomaxillofac Surg ; 43(4): 483-90, 2015 May.
Article in English | MEDLINE | ID: mdl-25753474

ABSTRACT

The rat calvarial defect is an established model to evaluate craniofacial bone regeneration using cell-scaffold biocomplexes. Dental pulp harbors stem cells with significant osteogenic properties. Extracellular matrix (ECM)-like scaffolds simulate the environment that cells observe in vivo. In the present study, we evaluated the osteogenic effect of a biocomplex of human dental pulp cells and a hyaluronic-based hydrogel scaffold in calvarial defects of immunocompetent rats. Dental pulp cells at the 2nd passage were characterized by flow cytometry, osteodifferentiated ex vivo for 4 days and the whole population was encapsulated in the synthetic ECM matrix. Cell vitality was verified 24 h upon encapsulation. 5 mm calvarial defects were created in 30 male rats and filled with the biocomplex, the scaffold alone, or left untreated. Histological evaluation at 8 weeks showed incomplete bone regeneration in all groups. The scaffold was not fully degraded and entrapped cells were detected in it. Histomorphometry showed statistically significant superior new bone formation in the biocomplex-treated group, compared to the two other groups. The present study provides evidence that the whole population of human dental pulp cells can advance bone healing when transplanted in immunocompetent animals and highlights the importance of proper scaffold degradation in cell-driven bioengineering treatments.


Subject(s)
Bone Diseases/therapy , Bone Regeneration/physiology , Dental Pulp/cytology , Hyaluronic Acid/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Mesenchymal Stem Cells/physiology , Skull/pathology , Stem Cell Transplantation/methods , Tissue Scaffolds/chemistry , Adolescent , Animals , Bone Diseases/pathology , Cell Culture Techniques , Cell Differentiation/physiology , Cell Survival/physiology , Cells, Cultured , Female , Humans , Male , Osteogenesis/physiology , Random Allocation , Rats , Rats, Wistar , Tissue Engineering/methods
16.
Nat Biotechnol ; 33(3): 269-276, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25664528

ABSTRACT

Reconstruction of the molecular pathways controlling organ development has been hampered by a lack of methods to resolve embryonic progenitor cells. Here we describe a strategy to address this problem that combines gene expression profiling of large numbers of single cells with data analysis based on diffusion maps for dimensionality reduction and network synthesis from state transition graphs. Applying the approach to hematopoietic development in the mouse embryo, we map the progression of mesoderm toward blood using single-cell gene expression analysis of 3,934 cells with blood-forming potential captured at four time points between E7.0 and E8.5. Transitions between individual cellular states are then used as input to develop a single-cell network synthesis toolkit to generate a computationally executable transcriptional regulatory network model of blood development. Several model predictions concerning the roles of Sox and Hox factors are validated experimentally. Our results demonstrate that single-cell analysis of a developing organ coupled with computational approaches can reveal the transcriptional programs that underpin organogenesis.


Subject(s)
Blood Cells/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Single-Cell Analysis/methods , Animals , Base Sequence , Computer Simulation , Diffusion , Female , Gastrulation , Gene Expression Profiling , Male , Mice, Inbred ICR , Models, Genetic , Molecular Sequence Data , Transcription, Genetic
17.
Nucleic Acids Res ; 43(Database issue): D1117-23, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25270877

ABSTRACT

CODEX (http://codex.stemcells.cam.ac.uk/) is a user-friendly database for the direct access and interrogation of publicly available next-generation sequencing (NGS) data, specifically aimed at experimental biologists. In an era of multi-centre genomic dataset generation, CODEX provides a single database where these samples are collected, uniformly processed and vetted. The main drive of CODEX is to provide the wider scientific community with instant access to high-quality NGS data, which, irrespective of the publishing laboratory, is directly comparable. CODEX allows users to immediately visualize or download processed datasets, or compare user-generated data against the database's cumulative knowledge-base. CODEX contains four types of NGS experiments: transcription factor chromatin immunoprecipitation coupled to high-throughput sequencing (ChIP-Seq), histone modification ChIP-Seq, DNase-Seq and RNA-Seq. These are largely encompassed within two specialized repositories, HAEMCODE and ESCODE, which are focused on haematopoiesis and embryonic stem cell samples, respectively. To date, CODEX contains over 1000 samples, including 221 unique TFs and 93 unique cell types. CODEX therefore provides one of the most complete resources of publicly available NGS data for the direct interrogation of transcriptional programmes that regulate cellular identity and fate in the context of mammalian development, homeostasis and disease.


Subject(s)
Databases, Genetic , Embryonic Stem Cells/metabolism , Hematopoietic Stem Cells/metabolism , High-Throughput Nucleotide Sequencing , Animals , Chromatin Immunoprecipitation , Hematopoiesis/genetics , Histones/metabolism , Humans , Internet , Mice , Sequence Analysis, DNA , Sequence Analysis, RNA , Software
18.
Nucleic Acids Res ; 42(22): 13513-24, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25428352

ABSTRACT

Combinatorial transcription factor (TF) binding is essential for cell-type-specific gene regulation. However, much remains to be learned about the mechanisms of TF interactions, including to what extent constrained spacing and orientation of interacting TFs are critical for regulatory element activity. To examine the relative prevalence of the 'enhanceosome' versus the 'TF collective' model of combinatorial TF binding, a comprehensive analysis of TF binding site sequences in large scale datasets is necessary. We developed a motif-pair discovery pipeline to identify motif co-occurrences with preferential distance(s) between motifs in TF-bound regions. Utilizing a compendium of 289 mouse haematopoietic TF ChIP-seq datasets, we demonstrate that haematopoietic-related motif-pairs commonly occur with highly conserved constrained spacing and orientation between motifs. Furthermore, motif clustering revealed specific associations for both heterotypic and homotypic motif-pairs with particular haematopoietic cell types. We also showed that disrupting the spacing between motif-pairs significantly affects transcriptional activity in a well-known motif-pair-E-box and GATA, and in two previously unknown motif-pairs with constrained spacing-Ets and Homeobox as well as Ets and E-box. In this study, we provide evidence for widespread sequence-specific TF pair interaction with DNA that conforms to the 'enhanceosome' model, and furthermore identify associations between specific haematopoietic cell-types and motif-pairs.


Subject(s)
Hematopoiesis/genetics , Regulatory Elements, Transcriptional , Transcription Factors/metabolism , Transcription, Genetic , Animals , Binding Sites , Blood Cells/metabolism , Chromatin Immunoprecipitation , DNA/chemistry , DNA/metabolism , Mice , Nucleotide Motifs , Sequence Analysis, DNA
19.
EMBO J ; 33(11): 1212-26, 2014 Jun 02.
Article in English | MEDLINE | ID: mdl-24760698

ABSTRACT

Despite major advances in the generation of genome-wide binding maps, the mechanisms by which transcription factors (TFs) regulate cell type identity have remained largely obscure. Through comparative analysis of 10 key haematopoietic TFs in both mast cells and blood progenitors, we demonstrate that the largely cell type-specific binding profiles are not opportunistic, but instead contribute to cell type-specific transcriptional control, because (i) mathematical modelling of differential binding of shared TFs can explain differential gene expression, (ii) consensus binding sites are important for cell type-specific binding and (iii) knock-down of blood stem cell regulators in mast cells reveals mast cell-specific genes as direct targets. Finally, we show that the known mast cell regulators Mitf and c-fos likely contribute to the global reorganisation of TF binding profiles. Taken together therefore, our study elucidates how key regulatory TFs contribute to transcriptional programmes in several distinct mammalian cell types.


Subject(s)
Gene Expression Regulation/genetics , Mast Cells/metabolism , Stem Cells/metabolism , Transcription Factors/genetics , Transcription, Genetic/genetics , Animals , Cell Line , Gene Expression Profiling , Genes, Reporter , Genome-Wide Association Study , Hematopoiesis/genetics , Mice , Models, Statistical , Nucleotide Motifs , Oligonucleotide Array Sequence Analysis , Sequence Analysis, DNA , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL
...