Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Development ; 145(5)2018 03 12.
Article in English | MEDLINE | ID: mdl-29530939

ABSTRACT

During ontogeny, hematopoietic stem and progenitor cells arise from hemogenic endothelium through an endothelial-to-hematopoietic transition that is strictly dependent on the transcription factor RUNX1. Although it is well established that RUNX1 is essential for the onset of hematopoiesis, little is known about the role of RUNX1 dosage specifically in hemogenic endothelium and during the endothelial-to-hematopoietic transition. Here, we used the mouse embryonic stem cell differentiation system to determine if and how RUNX1 dosage affects hemogenic endothelium differentiation. The use of inducible Runx1 expression combined with alterations in the expression of the RUNX1 co-factor CBFß allowed us to evaluate a wide range of RUNX1 levels. We demonstrate that low RUNX1 levels are sufficient and necessary to initiate an effective endothelial-to-hematopoietic transition. Subsequently, RUNX1 is also required to complete the endothelial-to-hematopoietic transition and to generate functional hematopoietic precursors. In contrast, elevated levels of RUNX1 are able to drive an accelerated endothelial-to-hematopoietic transition, but the resulting cells are unable to generate mature hematopoietic cells. Together, our results suggest that RUNX1 dosage plays a pivotal role in hemogenic endothelium maturation and the establishment of the hematopoietic system.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Endothelium, Vascular/physiology , Gene Dosage/physiology , Hemangioblasts/physiology , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/physiology , Gene Expression Regulation , Mice , Mice, Knockout
2.
Mech Dev ; 146: 31-41, 2017 08.
Article in English | MEDLINE | ID: mdl-28577909

ABSTRACT

The transcriptional program that regulates the differentiation of endothelial precursor cells into a highly organized vascular network is still poorly understood. Here we explore the role of SOX7 during this process, performing a detailed analysis of the vascular defects resulting from either a complete deficiency in Sox7 expression or from the conditional deletion of Sox7 in FLK1-expressing cells. We analysed the consequence of Sox7 deficiency from E7.5 onward to determine from which stage of development the effect of Sox7 deficiency can be observed. We show that while Sox7 is expressed at the onset of endothelial specification from mesoderm, Sox7 deficiency does not impact the emergence of the first endothelial progenitors. However, by E8.5, clear signs of defective vascular development are already observed with the presence of highly unorganised endothelial cords rather than distinct paired dorsal aorta. By E10.5, both Sox7 complete knockout and FLK1-specific deletion of Sox7 lead to widespread vascular defects. In contrast, while SOX7 is expressed in the earliest specified blood progenitors, the VAV-specific deletion of Sox7 does not affect the hematopoietic system. Together, our data reveal the unique role of SOX7 in vasculogenesis and angiogenesis during embryonic development.


Subject(s)
Embryonic Development/genetics , Neovascularization, Physiologic/genetics , SOXF Transcription Factors/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Cell Differentiation/genetics , Female , Gene Deletion , Gene Expression Regulation, Developmental , Mesoderm/growth & development , Mice , Mice, Knockout , Morphogenesis/genetics , Pregnancy
3.
Semin Cell Dev Biol ; 63: 50-57, 2017 03.
Article in English | MEDLINE | ID: mdl-27470491

ABSTRACT

Cardiovascular development during embryogenesis involves complex changes in gene regulatory networks regulated by a variety of transcription factors. In this review we discuss the various reported roles of the SOXF factors: SOX7, SOX17 and SOX18 in cardiac, vascular and lymphatic development. SOXF factors have pleiotropic roles during these processes, and there is significant redundancy and functional compensation between SOXF family members. Despite this, evidence suggests that there is some specificity in the transcriptional programs they regulate which is necessary to control the differentiation and behaviour of endothelial subpopulations. Furthermore, SOXF factors appear to have an indirect role in regulating cardiac mesoderm specification and differentiation. Understanding how SOXF factors are regulated, as well as their downstream transcriptional target genes, will be important for unravelling their roles in cardiovascular development and related diseases.


Subject(s)
Cardiovascular System/embryology , Cardiovascular System/metabolism , SOXF Transcription Factors/metabolism , Amino Acid Sequence , Animals , Hemangioblasts/metabolism , Humans , Lymphatic Vessels/embryology , Lymphatic Vessels/metabolism , Organogenesis/genetics , SOXF Transcription Factors/chemistry
4.
Development ; 143(23): 4341-4351, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27802172

ABSTRACT

Endothelial to hematopoietic transition (EHT) is a dynamic process involving the shutting down of endothelial gene expression and switching on of hematopoietic gene transcription. Although the factors regulating EHT in hemogenic endothelium (HE) of the dorsal aorta have been relatively well studied, the molecular regulation of yolk sac HE remains poorly understood. Here, we show that SOX7 inhibits the expression of RUNX1 target genes in HE, while having no effect on RUNX1 expression itself. We establish that SOX7 directly interacts with RUNX1 and inhibits its transcriptional activity. Through this interaction we demonstrate that SOX7 hinders RUNX1 DNA binding as well as the interaction between RUNX1 and its co-factor CBFß. Finally, we show by single-cell expression profiling and immunofluorescence that SOX7 is broadly expressed across the RUNX1+ yolk sac HE population compared with SOX17. Collectively, these data demonstrate for the first time how direct protein-protein interactions between endothelial and hematopoietic transcription factors regulate contrasting transcriptional programs during HE differentiation and EHT.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Endothelium/cytology , Hemangioblasts/cytology , SOXF Transcription Factors/metabolism , Yolk Sac/cytology , Animals , Cell Differentiation , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/antagonists & inhibitors , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , HMGB Proteins/metabolism , Hematopoietic Stem Cells/cytology , Male , Mice , Mice, Inbred C57BL , SOXF Transcription Factors/genetics , Transcription, Genetic/physiology
5.
Cell Cycle ; 15(16): 2108-2114, 2016 Aug 17.
Article in English | MEDLINE | ID: mdl-27399214

ABSTRACT

The first hematopoietic cells are generated very early in ontogeny to support the growth of the embryo and to provide the foundation to the adult hematopoietic system. There is a considerable therapeutic interest in understanding how these first blood cells are generated in order to try to reproduce this process in vitro. This would allow generating blood products, or hematopoietic cell populations from embryonic stem (ES) cells, induced pluripotent stem cells or through directed reprogramming. Recent studies have clearly established that the first hematopoietic cells originate from a hemogenic endothelium (HE) through an endothelial to hematopoietic transition (EHT). The molecular mechanisms underlining this transition remain largely unknown with the exception that the transcription factor RUNX1 is critical for this process. In this Extra Views report, we discuss our recent studies demonstrating that the transcriptional repressors GFI1 and GFI1B have a critical role in the EHT. We established that these RUNX1 transcriptional targets are actively implicated in the downregulation of the endothelial program and the loss of endothelial identity during the formation of the first blood cells. In addition, our results suggest that GFI1 expression provides an ideal novel marker to identify, isolate and study the HE cell population.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Hemangioblasts/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Models, Biological
6.
Dev Cell ; 36(5): 572-87, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26923725

ABSTRACT

Metazoan development involves the successive activation and silencing of specific gene expression programs and is driven by tissue-specific transcription factors programming the chromatin landscape. To understand how this process executes an entire developmental pathway, we generated global gene expression, chromatin accessibility, histone modification, and transcription factor binding data from purified embryonic stem cell-derived cells representing six sequential stages of hematopoietic specification and differentiation. Our data reveal the nature of regulatory elements driving differential gene expression and inform how transcription factor binding impacts on promoter activity. We present a dynamic core regulatory network model for hematopoietic specification and demonstrate its utility for the design of reprogramming experiments. Functional studies motivated by our genome-wide data uncovered a stage-specific role for TEAD/YAP factors in mammalian hematopoietic specification. Our study presents a powerful resource for studying hematopoiesis and demonstrates how such data advance our understanding of mammalian development.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Animals , Cell Lineage/physiology , Homeodomain Proteins/metabolism , Mice , Protein Binding/genetics , Transcription Factors/metabolism
7.
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
8.
Stem Cells Int ; 2011: 274564, 2011.
Article in English | MEDLINE | ID: mdl-22135682

ABSTRACT

The concept of the haematopoietic stem cell (HSC) niche was formulated by Schofield in the 1970s, as a region within the bone marrow containing functional cell types that can maintain HSC potency throughout life. Since then, ongoing research has identified numerous cell types and a plethora of signals that not only maintain HSCs, but also dictate their behaviour with respect to homeostatic requirements and exogenous stresses. It has been proposed that there are endosteal and vascular niches within the bone marrow, which are thought to regulate different HSC populations. However, recent data depicts a more complicated picture, with functional crosstalk between cells in these two regions. In this review, recent research into the endosteal/vascular cell types and signals regulating HSC behaviour are considered, together with the possibility of a single subcompartmentalised niche.

9.
Cancer Res ; 71(3): 1177-86, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21169412

ABSTRACT

Nm23-H1 plays complex roles in the development of diverse cancers including breast carcinoma, high-grade lymphomas, and acute myeloid leukemia (AML). In the case of AML and lymphomas, serum Nm23-H1 protein is elevated with the highest levels correlating with poorest prognosis. A recent study identified that this association is most likely causal in AML and that Nm23-H1 acts as an AML cell survival factor. In this study, we report heterogeneity in the ability of AML samples to bind and respond to Nm23-H1, and we offer evidence that binding is essential for improved survival. Further, we show that the subset of AMLs that bind Nm23-H1 do not do so through the putative Nm23-H1 receptor MUC1*. Although rNm23-H1 promoted the survival of the most primitive blasts within responding AMLs, it was not these cells that actually bound the protein. Instead, rNm23-H1 bound to more mature CD34(lo)/CD34(-) and CD11b(+) cells, revealing an indirect survival benefit of Nm23-H1 on primitive blasts. In support of this finding, the survival of purified blast cells was enhanced by medium conditioned by more mature cells from the clone that had been stimulated by rNm23-H1. Levels of interleukin 1ß (IL1ß) and IL6 in rNm23-H1 conditioned medium mirrored the potency of the conditioned media to promote blast cell survival. Furthermore, Nm23-H1 expression was significantly associated with IL1ß and IL6 expression in primary uncultured AML samples. These findings have implications for the role of Nm23-H1 in AML and its use as a prognostic marker. Additionally, they offer the first evidence of novel cross-talk between cell populations within the tumor clone.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , NM23 Nucleoside Diphosphate Kinases/metabolism , Antigens, CD34/metabolism , CD11b Antigen/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , K562 Cells , Mucin-1/biosynthesis , NM23 Nucleoside Diphosphate Kinases/biosynthesis , NM23 Nucleoside Diphosphate Kinases/pharmacology , Proto-Oncogene Proteins c-kit/metabolism
10.
Antimicrob Agents Chemother ; 53(9): 3832-6, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19596880

ABSTRACT

Nontyphoidal Salmonella enterica strains with a nonclassical quinolone resistance phenotype were isolated from patients returning from Thailand or Malaysia to Finland. A total of 10 isolates of seven serovars were studied in detail, all of which had reduced susceptibility (MIC > or = 0.125 microg/ml) to ciprofloxacin but were either susceptible or showed only low-level resistance (MIC < or = 32 microg/ml) to nalidixic acid. Phenotypic characterization included susceptibility testing by the agar dilution method and investigation of efflux activity. Genotypic characterization included the screening of mutations in the quinolone resistance-determining regions (QRDR) of gyrA, gyrB, parC, and parE by PCR and denaturing high-pressure liquid chromatography and the amplification of plasmid-mediated quinolone resistance (PMQR) genes qnrA, qnrB, qnrS, qnrD, aac(6')-Ib-cr, and qepA by PCR. PMQR was confirmed by plasmid analysis, Southern hybridization, and plasmid transfer. No mutations in the QRDRs of gyrA, gyrB, parC, or parE were detected with the exception of a Thr57-Ser substitution within ParC seen in all but the S. enterica serovar Typhimurium strains. The qnrA and qnrS genes were the only PMQR determinants detected. Plasmids carrying qnr alleles were transferable in vitro, and the resistance phenotype was reproducible in Escherichia coli DH5alpha transformants. These data demonstrate the emergence of a highly mobile qnr genotype that, in the absence of mutation within topoisomerase genes, confers the nontypical quinolone resistance phenotype in S. enterica isolates. The qnr resistance mechanism enables bacteria to survive elevated quinolone concentrations, and therefore, strains carrying qnr alleles may be able to expand during fluoroquinolone treatment. This is of concern since nonclassical quinolone resistance is plasmid mediated and therefore mobilizable.


Subject(s)
Drug Resistance, Bacterial/physiology , Quinolones/pharmacology , Salmonella enterica/drug effects , Salmonella enterica/genetics , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Drug Resistance, Bacterial/genetics , Electroporation , Genotype , Microbial Sensitivity Tests , Plasmids/genetics , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...