Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Nat Cancer ; 2(8): 835-852, 2021 08.
Article En | MEDLINE | ID: mdl-34734190

Comparison of intratumor genetic heterogeneity in cancer at diagnosis and relapse suggests that chemotherapy induces bottleneck selection of subclonal genotypes. However, evolutionary events subsequent to chemotherapy could also explain changes in clonal dominance seen at relapse. We, therefore, investigated the mechanisms of selection in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL) during induction chemotherapy where maximal cytoreduction occurs. To distinguish stochastic versus deterministic events, individual leukemias were transplanted into multiple xenografts and chemotherapy administered. Analyses of the immediate post-treatment leukemic residuum at single-cell resolution revealed that chemotherapy has little impact on genetic heterogeneity. Rather, it acts on extensive, previously unappreciated, transcriptional and epigenetic heterogeneity in BCP-ALL, dramatically reducing the spectrum of cell states represented, leaving a genetically polyclonal but phenotypically uniform population with hallmark signatures relating to developmental stage, cell cycle and metabolism. Hence, canalization of cell state accounts for a significant component of bottleneck selection during induction chemotherapy.


Burkitt Lymphoma , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Burkitt Lymphoma/drug therapy , Cell Cycle , Humans , Induction Chemotherapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Recurrence
2.
Cell Stem Cell ; 26(4): 527-541.e8, 2020 04 02.
Article En | MEDLINE | ID: mdl-32197066

Umbilical cord blood (UCB) has had considerable impact in pediatric stem cell transplantation, but its wider use is limited in part by unit size. Long-term ex vivo culture offers one approach to increase engraftment capacity by seeking to expand stem and progenitor cells. Here, we show brief incubation (8 h) of UCB CD34+ cells with the matricellular regulator Nov (CCN3) increases the frequency of serially transplantable hematopoietic stem cells (HSCs) 6-fold. This rapid response suggests recruitment rather than expansion of stem cells; accordingly, in single-cell assays, Nov increases the clonogenicity of phenotypic HSCs without increasing their number through cell division. Recruitment is associated with both metabolic and transcriptional changes, and tracing of cell divisions demonstrates that the increased clonogenic activity resides within the undivided fraction of cells. Harnessing latent stem cell potential through recruitment-based approaches will inform understanding of stem cell state transitions with implications for translation to the clinic.


Fetal Blood , Hematopoietic Stem Cell Transplantation , Antigens, CD34 , Child , Hematopoietic Stem Cells , Humans
3.
Dev Biol ; 424(2): 236-245, 2017 04 15.
Article En | MEDLINE | ID: mdl-28189604

Hematopoietic stem cells (HSCs) emerge during development via an endothelial-to-hematopoietic transition from hemogenic endothelium of the dorsal aorta (DA). Using in situ hybridization and analysis of a knock-in RedStar reporter, we show that the transcriptional regulator Hhex is expressed in endothelium of the dorsal aorta (DA) and in clusters of putative HSCs as they are specified during murine development. We exploited this observation, using the Hhex locus to define cis regulatory elements, enhancers and interacting transcription factors that are both necessary and sufficient to support gene expression in the emerging HSC. We identify an evolutionarily conserved non-coding region (ECR) in the Hhex locus with the capacity to bind the hematopoietic-affiliated transcriptional regulators Gata2, SCL, Fli1, Pu.1 and Ets1/2. This region is sufficient to drive the expression of a transgenic GFP reporter in the DA endothelium and intra-aortic hematopoietic clusters. GFP-positive AGM cells co-expressed HSC-associated markers c-Kit, CD34, VE-Cadherin, and CD45, and were capable of multipotential differentiation and long term engraftment when transplanted into myelo-ablated recipients. The Hhex ECR was also sufficient to drive expression at additional blood sites including the yolk sac blood islands, fetal liver, vitelline and umbilical arteries and the adult bone marrow, suggesting a common mechanism for Hhex regulation throughout ontogenesis of the blood system. To explore the physiological requirement for the Hhex ECR region during hematoendothelial development, we deleted the ECR element from the endogenous locus in the context of a targeted Hhex-RedStar reporter allele. Results indicate a specific requirement for the ECR in blood-associated Hhex expression during development and further demonstrate a requirement for this region in the adult HSC compartment. Taken together, our results identified the ECR region as an enhancer both necessary and sufficient for gene expression in HSC development and homeostasis. The Hhex ECR thus appears to be a core node for the convergence of the transcription factor network that governs the emergence of HSCs.


Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Cell Compartmentation , Cell Lineage/genetics , Colony-Forming Units Assay , Conserved Sequence/genetics , Embryo, Mammalian/metabolism , Genetic Loci , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/genetics
4.
Exp Hematol ; 44(5): 399-409.e5, 2016 May.
Article En | MEDLINE | ID: mdl-26876150

Transforming growth factor ß (TGFß) is a potent inhibitor of hematopoietic stem and progenitor cell proliferation. However, the precise mechanism for this effect is unknown. Here, we have identified the transcription factor Gata2, previously described as an important regulator of hematopoietic stem cell function, as an early and direct target gene for TGFß-induced Smad signaling in hematopoietic progenitor cells. We also report that Gata2 is involved in mediating a significant part of the TGFß response in primitive hematopoietic cells. Interestingly, the cell cycle regulator and TGFß signaling effector molecule p57 was found to be upregulated as a secondary response to TGFß. We observed Gata2 binding upstream of the p57 genomic locus, and importantly, loss of Gata2 abolished TGFß-stimulated induction of p57 as well as the resulting growth arrest of hematopoietic progenitors. Our results connect key molecules involved in hematopoietic stem cell self-renewal and reveal a functionally relevant network, regulating proliferation of primitive hematopoietic cells.


Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p57/genetics , GATA2 Transcription Factor/genetics , Hematopoietic Stem Cells/drug effects , Smad4 Protein/genetics , Transforming Growth Factor beta/pharmacology , Animals , Cell Line , Cell Proliferation/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p57/metabolism , GATA2 Transcription Factor/metabolism , Gene Expression Profiling/methods , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/genetics , HEK293 Cells , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Genetic , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Smad4 Protein/metabolism
5.
Cell Rep ; 11(10): 1503-10, 2015 Jun 16.
Article En | MEDLINE | ID: mdl-26051941

We explore cell heterogeneity during spontaneous and transcription-factor-driven commitment for network inference in hematopoiesis. Since individual genes display discrete OFF states or a distribution of ON levels, we compute and combine pairwise gene associations from binary and continuous components of gene expression in single cells. Ddit3 emerges as a regulatory node with positive linkage to erythroid regulators and negative association with myeloid determinants. Ddit3 loss impairs erythroid colony output from multipotent cells, while forcing Ddit3 in granulo-monocytic progenitors (GMPs) enhances self-renewal and impedes differentiation. Network analysis of Ddit3-transduced GMPs reveals uncoupling of myeloid networks and strengthening of erythroid linkages. RNA sequencing suggests that Ddit3 acts through development or stabilization of a precursor upstream of GMPs with inherent Meg-E potential. The enrichment of Gata2 target genes in Ddit3-dependent transcriptional responses suggests that Ddit3 functions in an erythroid transcriptional network nucleated by Gata2.


Gene Regulatory Networks , Hematopoiesis/genetics , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Animals , Cell Differentiation/genetics , GATA2 Transcription Factor/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Single-Cell Analysis/methods
6.
Trends Cell Biol ; 25(8): 459-67, 2015 Aug.
Article En | MEDLINE | ID: mdl-26004869

Regulation of lineage commitment in multipotential cells is key to maintaining a balanced hematopoietic output throughout life while retaining the capacity to respond to stress and infection. Cell fate decisions are made by individual stem cells, but population-level analysis obscures the mechanics of cell fate choice by averaging the molecular and functional heterogeneity that exists even in the most highly purified stem cell populations. Therefore, single cell analysis of both molecular and cellular phenotypes is crucial to delineate and interrogate the process of lineage commitment. We review recent single cell expression profiling, imaging, and clonal tracking studies that have provided new insights into commitment, focusing on the hematopoietic system, and suggest how new technologies may illuminate our understanding of lineage commitment in the near future.


Cell Differentiation/physiology , Cell Lineage/physiology , Gene Regulatory Networks/physiology , Single-Cell Analysis/methods , Animals , Humans , Single-Cell Analysis/trends
7.
Cell Stem Cell ; 13(6): 754-68, 2013 Dec 05.
Article En | MEDLINE | ID: mdl-24120743

We used the paradigmatic GATA-PU.1 axis to explore, at the systems level, dynamic relationships between transcription factor (TF) binding and global gene expression programs as multipotent cells differentiate. We combined global ChIP-seq of GATA1, GATA2, and PU.1 with expression profiling during differentiation to erythroid and neutrophil lineages. Our analysis reveals (1) differential complexity of sequence motifs bound by GATA1, GATA2, and PU.1; (2) the scope and interplay of GATA1 and GATA2 programs within, and during transitions between, different cell compartments, and the extent of their hard-wiring by DNA motifs; (3) the potential to predict gene expression trajectories based on global associations between TF-binding data and target gene expression; and (4) how dynamic modeling of DNA-binding and gene expression data can be used to infer regulatory logic of TF circuitry. This rubric exemplifies the utility of this cross-platform resource for deconvoluting the complexity of transcriptional programs controlling stem/progenitor cell fate in hematopoiesis.


Cell Lineage/genetics , Gene Expression Regulation , Genome/genetics , Hematopoiesis/genetics , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Chromatin Immunoprecipitation , Erythroid Cells/cytology , Erythroid Cells/metabolism , GATA1 Transcription Factor/metabolism , GATA2 Transcription Factor/metabolism , Humans , Mice , Models, Biological , Molecular Sequence Data , Nucleotide Motifs/genetics , Protein Binding/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism
8.
Blood ; 113(12): 2661-72, 2009 Mar 19.
Article En | MEDLINE | ID: mdl-19168794

Evidence suggests the transcription factor GATA-2 is a critical regulator of murine hematopoietic stem cells. Here, we explore the relation between GATA-2 and cell proliferation and show that inducing GATA-2 increases quiescence (G(0) residency) of murine and human hematopoietic cells. In human cord blood, quiescent fractions (CD34(+)CD38(-)Hoechst(lo)Pyronin Y(lo)) express more GATA-2 than cycling counterparts. Enforcing GATA-2 expression increased quiescence of cord blood cells, reducing proliferation and performance in long-term culture-initiating cell and colony-forming cell (CFC) assays. Gene expression analysis places GATA-2 upstream of the quiescence regulator MEF, but enforcing MEF expression does not prevent GATA-2-conferred quiescence, suggesting additional regulators are involved. Although known quiescence regulators p21(CIP1) and p27(KIP1) do not appear to be responsible, enforcing GATA-2 reduced expression of regulators of cell cycle such as CCND3, CDK4, and CDK6. Enforcing GATA-2 inhibited human hematopoiesis in vivo: cells with highest exogenous expression (GATA-2(hi)) failed to contribute to hematopoiesis in nonobese diabetic-severe combined immunodeficient (NOD-SCID) mice, whereas GATA-2(lo) cells contributed with delayed kinetics and low efficiency, with reduced expression of Ki-67. Thus, GATA-2 activity inhibits cell cycle in vitro and in vivo, highlighting GATA-2 as a molecular entry point into the transcriptional program regulating quiescence in human hematopoietic stem and progenitor cells.


Cell Cycle , GATA2 Transcription Factor/physiology , Hematopoietic Stem Cells/cytology , Animals , Apoptosis , Cells, Cultured/cytology , Cells, Cultured/drug effects , Cord Blood Stem Cell Transplantation , Estradiol/pharmacology , Fetal Blood/cytology , GATA2 Transcription Factor/biosynthesis , GATA2 Transcription Factor/genetics , Gene Expression Regulation/genetics , Genes, Synthetic , Genes, cdc , Humans , Interleukin-3/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/physiology , Resting Phase, Cell Cycle , Tamoxifen/pharmacology , Transcription, Genetic
9.
Blood ; 112(13): 4862-73, 2008 Dec 15.
Article En | MEDLINE | ID: mdl-18840712

The zinc finger transcription factor GATA-2 has been implicated in the regulation of hematopoietic stem cells. Herein, we explored the role of GATA-2 as a candidate regulator of the hematopoietic progenitor cell compartment. We showed that bone marrow from GATA-2 heterozygote (GATA-2(+/-)) mice displayed attenuated granulocyte-macrophage progenitor function in colony-forming cell (CFC) and serial replating CFC assays. This defect was mapped to the Lin(-)CD117(+)Sca-1(-)CD34(+)CD16/32(high) granulocyte-macrophage progenitor (GMP) compartment of GATA-2(+/-) marrow, which was reduced in size and functionally impaired in CFC assays and competitive transplantation. Similar functional impairments were obtained using a RNA interference approach to stably knockdown GATA-2 in wild-type GMP. Although apoptosis and cell-cycle distribution remained unperturbed in GATA-2(+/-) GMP, quiescent cells from GATA-2(+/-) GMP exhibited altered functionality. Gene expression analysis showed attenuated expression of HES-1 mRNA in GATA-2-deficient GMP. Binding of GATA-2 to the HES-1 locus was detected in the myeloid progenitor cell line 32Dcl3, and enforced expression of HES-1 expression in GATA-2(+/-) GMP rectified the functional defect, suggesting that GATA-2 regulates myeloid progenitor function through HES-1. These data collectively point to GATA-2 as a novel, pivotal determinant of GMP cell fate.


GATA2 Transcription Factor/physiology , Granulocyte-Macrophage Progenitor Cells/cytology , Animals , Cell Line , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression Profiling , Genotype , Granulocyte-Macrophage Progenitor Cells/physiology , Mice , Mice, Mutant Strains , Protein Binding , RNA Interference
10.
Cell Stem Cell ; 2(3): 264-73, 2008 Mar 06.
Article En | MEDLINE | ID: mdl-18371451

Regulatory mechanisms of human hematopoiesis remain largely uncharacterized. Through expression profiling of prospectively isolated stem and primitive progenitor cells as well as committed progenitors from cord blood (CB), we identified MLLT3 as a candidate regulator of erythroid/megakaryocytic (E/Meg) lineage decisions. Through the analysis of the hematopoietic potential of primitive cord blood cells in which MLLT3 expression has been knocked down, we identify a requirement for MLLT3 in the elaboration of the erythroid and megakaryocytic lineages. Conversely, forced expression of MLLT3 promotes the output of erythroid and megakaryocytic progenitors, and analysis of MLLT3 mutants suggests that this capacity of MLLT3 depends on its transcriptional regulatory activity. Gene expression and cis-regulatory element analyses reveal crossregulatory interactions between MLLT3 and E/Meg-affiliated transcription factor GATA-1. Taken together, the data identify MLLT3 as a regulator of early erythroid and megakaryocytic cell fate in the human system.


Erythropoiesis/physiology , Fetal Blood/metabolism , Megakaryocytes/metabolism , Nuclear Proteins/metabolism , Thrombopoiesis/physiology , Cells, Cultured , Fetal Blood/cytology , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Silencing , Humans , Megakaryocytes/cytology , Mutation , Nuclear Proteins/genetics , Response Elements/physiology
11.
Dev Biol ; 305(2): 695-713, 2007 May 15.
Article En | MEDLINE | ID: mdl-17412320

Lineage specification of multipotent progenitor cells is governed by a balance of lineage-affiliated transcription factors, such as GATA1 and PU.1, which regulate the choice between erythroid and myelomonocytic fates. But how ratios of lineage-determining transcription factors stabilize progenitor cells and resolve their indeterminacy to commit them to discrete, mutually exclusive fates remains unexplained. We used a simple model and experimental measurements to analyze the dynamics of a binary fate decision governed by a gene-circuit containing auto-stimulation and cross-inhibition, as embodied by the GATA1-PU.1 paradigm. This circuit generates stable attractors corresponding to erythroid and myelomonocytic fates, as well as an uncommitted metastable state characterized by coexpression of both regulators, explaining the phenomenon of "multilineage priming". GATA1 and PU.1 mRNA and transcriptome dynamics of differentiating progenitor cells confirm that commitment occurs in two stages, as suggested by the model: first, the progenitor state is destabilized in an almost symmetrical bifurcation event, resulting in a poised state at the boundary between the two lineage-specific attractors; second, the cell is driven to the respective, now accessible attractors. This minimal model captures fundamental features of binary cell fate decisions, uniting the concepts of stochastic (selective) and deterministic (instructive) regulation, and hence, may apply to a wider range of binary fate decision points.


Cell Differentiation/physiology , Cell Lineage/physiology , Erythroid Precursor Cells/cytology , Granulocyte Precursor Cells/cytology , Models, Biological , Multipotent Stem Cells/cytology , Animals , Cell Line , Erythroid Precursor Cells/metabolism , GATA1 Transcription Factor/physiology , Gene Expression Regulation, Developmental/physiology , Granulocyte Precursor Cells/metabolism , Mice , Multipotent Stem Cells/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Trans-Activators/biosynthesis , Trans-Activators/genetics
12.
Ann N Y Acad Sci ; 1106: 30-40, 2007 Jun.
Article En | MEDLINE | ID: mdl-17442775

Identifying the transcription factor interactions that are responsible for cell-specific gene expression programs is key to understanding the regulation of cell behaviors, such as self-renewal, proliferation, differentiation, and death. The rapidly increasing availability of microarray-derived global gene expression data sets, coupled with genome sequence information from multiple species, has driven the development of computational methods to reverse engineer and dynamically model genetic regulatory networks. An understanding of the architecture and behavior of transcriptional networks should lend insight into how the huge number of potential gene expression programs is constrained and facilitates efforts to direct or redirect cell fate.


Hematopoietic Stem Cells/cytology , Transcription, Genetic , Animals , Cell Differentiation , Cell Lineage , Computational Biology , Computer Simulation , GATA1 Transcription Factor/metabolism , Gene Regulatory Networks , Hematopoietic System , Humans , Models, Biological , Models, Theoretical , Oligonucleotide Array Sequence Analysis , Software
13.
Cell Res ; 14(3): 241-50, 2004 Jun.
Article En | MEDLINE | ID: mdl-15225418

Characterising the mechanisms of cell death following focal cerebral ischaemia has been hampered by a lack of an in vitro assay emulating both the apoptotic and necrotic features observed in vivo. The present study systematically characterised oxygen-glucose-deprivation (OGD) in primary rat cortical neurones to establish a reproducible model with components of both cell-death endpoints. OGD induced a time-dependent reduction in cell viability, with 80% cell death occurring 24 h after 3 h exposure to 0% O2 and 0.5 mM glucose. Indicative of a necrotic component to OGD-induced cell death, N-methyl-D-aspartate (NMDA) receptor inhibition with MK-801 attenuated neuronal loss by 60%. The lack of protection by the caspase inhibitors DEVD-CHO and z-VAD-fmk suggested that under these conditions neurones did not die by an apoptotic mechanism. Moderating the severity of the insult by decreasing OGD exposure to 60 min did not reduce the amount of necrosis, but did induce a small degree of apoptosis (a slight reduction in cell death was observed in the presence of 10 uM DEVD-CHO). In separate experiments purported to enhance the apoptotic component, cells were gradually deprived of O2, exposed to 4% O2 (as opposed to 0%) during the OGD period, or maintained in serum-containing media throughout. While NMDA receptor antagonism significantly reduced cortical cell death under all conditions, a caspase-inhibitor sensitive component of cell death was not uncovered. These studies suggest that OGD of cultured cortical cells models the excitotoxic, but not the apoptotic component of cell death observed in vivo.


Apoptosis/physiology , Brain Ischemia/pathology , Cerebral Cortex/pathology , Neurons/pathology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Brain Ischemia/metabolism , Caspases/drug effects , Caspases/metabolism , Cell Hypoxia/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glucose/metabolism , Glucose/pharmacology , Models, Biological , N-Methylaspartate/pharmacology , Neurons/drug effects , Neurons/metabolism , Oligopeptides/pharmacology , Oxygen/metabolism , Oxygen/pharmacology , Rats , Rats, Sprague-Dawley , Staurosporine/pharmacology , Time Factors
14.
EMBO J ; 21(14): 3770-81, 2002 Jul 15.
Article En | MEDLINE | ID: mdl-12110589

The developmental plasticity of transplanted adult stem cells challenges the notion that tissue-restricted stem cells have stringently limited lineage potential and prompts a re-evaluation of the stability of lineage commitment. Transformed cell systems are inappropriate for such studies, since transformation potentially dysregulates the processes governing lineage commitment. We have therefore assessed the stability of normal lineage commitment in primary adult haematopoietic cells. For these studies we have used prospectively isolated primary bipotent progenitors, which normally display only neutrophil and monocyte differentiation in vitro. In response to ectopic transcription factor expression, these neutrophil/monocyte progenitors were reprogrammed to take on erythroid, eosinophil and basophil-like cell fates, with the resultant colonies resembling the mixed lineage colonies normally generated by multipotential progenitors. Clone-marking and daughter cell experiments identified lineage switching rather than differential cell selection as the mechanism of altered lineage output. These results demonstrate that the cell type-specific programming of apparently committed primary progenitors is not irrevocably fixed, but may be radically re-specified in response to a single transcriptional regulator.


Cell Lineage , DNA-Binding Proteins/physiology , Stem Cells/cytology , Transcription Factors/physiology , Blotting, Northern , Blotting, Western , Cytokines/pharmacology , Erythroid-Specific DNA-Binding Factors , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/drug effects
...