Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Genomics ; 116(2): 110793, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38220132

ABSTRACT

Single-cell RNA sequencing (scRNA-Seq) has emerged as a powerful tool for understanding cellular heterogeneity and function. However the choice of sample multiplexing reagents can impact data quality and experimental outcomes. In this study, we compared various multiplexing reagents, including MULTI-Seq, Hashtag antibody, and CellPlex, across diverse sample types such as human peripheral blood mononuclear cells (PBMCs), mouse embryonic brain and patient-derived xenografts (PDXs). We found that all multiplexing reagents worked well in cell types robust to ex vivo manipulation but suffered from signal-to-noise issues in more delicate sample types. We compared multiple demultiplexing algorithms which differed in performance depending on data quality. We find that minor improvements to laboratory workflows such as titration and rapid processing are critical to optimal performance. We also compared the performance of fixed scRNA-Seq kits and highlight the advantages of the Parse Biosciences kit for fragile samples. Highly multiplexed scRNA-Seq experiments require more sequencing resources, therefore we evaluated CRISPR-based destruction of non-informative genes to enhance sequencing value. Our comprehensive analysis provides insights into the selection of appropriate sample multiplexing reagents and protocols for scRNA-Seq experiments, facilitating more accurate and cost-effective studies.


Subject(s)
Leukocytes, Mononuclear , Single-Cell Analysis , Humans , Animals , Mice , RNA-Seq , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Algorithms , Gene Expression Profiling/methods
2.
Development ; 148(20)2021 10 15.
Article in English | MEDLINE | ID: mdl-34550360

ABSTRACT

Blood vessel growth and remodelling are essential during embryonic development and disease pathogenesis. The diversity of endothelial cells (ECs) is transcriptionally evident and ECs undergo dynamic changes in gene expression during vessel growth and remodelling. Here, we investigated the role of the histone acetyltransferase HBO1 (KAT7), which is important for activating genes during development and for histone H3 lysine 14 acetylation (H3K14ac). Loss of HBO1 and H3K14ac impaired developmental sprouting angiogenesis and reduced pathological EC overgrowth in the retinal endothelium. Single-cell RNA sequencing of retinal ECs revealed an increased abundance of tip cells in Hbo1-deficient retinas, which led to EC overcrowding in the retinal sprouting front and prevented efficient tip cell migration. We found that H3K14ac was highly abundant in the endothelial genome in both intra- and intergenic regions, suggesting that HBO1 acts as a genome organiser that promotes efficient tip cell behaviour necessary for sprouting angiogenesis. This article has an associated 'The people behind the papers' interview.


Subject(s)
Histone Acetyltransferases/metabolism , Neovascularization, Pathologic/metabolism , Acetylation , Animals , Cell Movement/physiology , Cells, Cultured , Embryonic Development/physiology , Endothelial Cells/metabolism , Female , Histones/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Lysine/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
3.
Immunity ; 37(6): 1009-23, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23219391

ABSTRACT

Cytopenias are key prognostic indicators of life-threatening infection, contributing to immunosuppression and mortality. Here we define a role for Caspase-1-dependent death, known as pyroptosis, in infection-induced cytopenias by studying inflammasome activation in hematopoietic progenitor cells. The NLRP1a inflammasome is expressed in hematopoietic progenitor cells and its activation triggers their pyroptotic death. Active NLRP1a induced a lethal systemic inflammatory disease that was driven by Caspase-1 and IL-1ß but was independent of apoptosis-associated speck-like protein containing a CARD (ASC) and ameliorated by IL-18. Surprisingly, in the absence of IL-1ß-driven inflammation, active NLRP1a triggered pyroptosis of hematopoietic progenitor cells resulting in leukopenia at steady state. During periods of hematopoietic stress induced by chemotherapy or lymphocytic choriomeningitis virus (LCMV) infection, active NLRP1a caused prolonged cytopenia, bone marrow hypoplasia, and immunosuppression. Conversely, NLRP1-deficient mice showed enhanced recovery from chemotherapy and LCMV infection, demonstrating that NLRP1 acts as a cellular sentinel to alert Caspase-1 to hematopoietic and infectious stress.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis , Hematopoietic Stem Cells/metabolism , Inflammasomes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins/genetics , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Dermatitis/immunology , Dermatitis/metabolism , Fluorouracil/pharmacology , Hematopoiesis/drug effects , Hematopoiesis/immunology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/virology , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interferon-gamma/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Knockout , Mutation , Pancytopenia/immunology , Pancytopenia/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
Nucleic Acids Res ; 47(D1): D780-D785, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30395284

ABSTRACT

During haematopoiesis, haematopoietic stem cells differentiate into restricted potential progenitors before maturing into the many lineages required for oxygen transport, wound healing and immune response. We have updated Haemopedia, a database of gene-expression profiles from a broad spectrum of haematopoietic cells, to include RNA-seq gene-expression data from both mice and humans. The Haemopedia RNA-seq data set covers a wide range of lineages and progenitors, with 57 mouse blood cell types (flow sorted populations from healthy mice) and 12 human blood cell types. This data set has been made accessible for exploration and analysis, to researchers and clinicians with limited bioinformatics experience, on our online portal Haemosphere: https://www.haemosphere.org. Haemosphere also includes nine other publicly available high-quality data sets relevant to haematopoiesis. We have added the ability to compare gene expression across data sets and species by curating data sets with shared lineage designations or to view expression gene vs gene, with all plots available for download by the user.


Subject(s)
Databases, Genetic , Gene Expression/genetics , Hematopoiesis/genetics , Transcriptome/genetics , Animals , Computational Biology , Hematopoietic Stem Cells/metabolism , High-Throughput Nucleotide Sequencing/trends , Humans , Mice , RNA-Seq , Software
6.
Immunol Cell Biol ; 96(10): 1083-1094, 2018 11.
Article in English | MEDLINE | ID: mdl-29870118

ABSTRACT

Plasmacytoid dendritic cells (pDCs) play a critical role in bridging the innate and adaptive immune systems. pDCs are specialized type I interferon (IFN) producers, which has implicated them as initiators of autoimmune pathogenesis. However, little is known about the downstream effectors of type I IFN signaling that amplify autoimmune responses. Here, we have used a chemokine reporter mouse to determine the CXCR3 ligand responses in DCs subsets. Following TLR7 stimulation, conventional type 1 and type 2 DCs (cDC1 and cDC2, respectively) uniformly upregulate CXCL10. By contrast, the proportion of chemokine positive pDCs was significantly less, and stable CXCL10+ and CXCL10- populations could be distinguished. CXCL9 expression was induced in all cDC1s, in half of the cDC2 but not by pDCs. The requirement for IFNAR signaling for chemokine reporter expression was interrogated by receptor blocking and deficiency and shown to be critical for CXCR3 ligand expression in Flt3-ligand-derived DCs. Chemokine-producing potential was not concordant with the previously identified markers of pDC heterogeneity. Finally, we show that CXCL10+ and CXCL10- populations are transcriptionally distinct, expressing unique transcriptional regulators, IFN signaling molecules, chemokines, cytokines, and cell surface markers. This work highlights CXCL10 as a downstream effector of type I IFN signaling and suggests a division of labor in pDCs subtypes that likely impacts their function as effectors of viral responses and as drivers of inflammation.


Subject(s)
Chemokine CXCL10/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation , Toll-Like Receptor 7/agonists , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Cells, Cultured , Chemokine CXCL10/metabolism , Cytokines/metabolism , Gene Expression Profiling , Immunophenotyping , Interferon Type I/metabolism , Mice , Receptors, CXCR3/metabolism , Signal Transduction
7.
PLoS Genet ; 11(5): e1005211, 2015 May.
Article in English | MEDLINE | ID: mdl-25973911

ABSTRACT

Down syndrome (DS), with trisomy of chromosome 21 (HSA21), is the commonest human aneuploidy. Pre-leukemic myeloproliferative changes in DS foetal livers precede the acquisition of GATA1 mutations, transient myeloproliferative disorder (DS-TMD) and acute megakaryocytic leukemia (DS-AMKL). Trisomy of the Erg gene is required for myeloproliferation in the Ts(1716)65Dn DS mouse model. We demonstrate here that genetic changes specifically attributable to trisomy of Erg lead to lineage priming of primitive and early multipotential progenitor cells in Ts(1716)65Dn mice, excess megakaryocyte-erythroid progenitors, and malignant myeloproliferation. Gene expression changes dependent on trisomy of Erg in Ts(1716)65Dn multilineage progenitor cells were correlated with those associated with trisomy of HSA21 in human DS hematopoietic stem and primitive progenitor cells. These data suggest a role for ERG as a regulator of hematopoietic lineage potential, and that trisomy of ERG in the context of DS foetal liver hemopoiesis drives the pre-leukemic changes that predispose to subsequent DS-TMD and DS-AMKL.


Subject(s)
Chromosomes, Human, Pair 21/genetics , Down Syndrome/genetics , Oncogene Proteins/genetics , Stem Cells/cytology , Trans-Activators/genetics , Transcription Factors/genetics , Trisomy , ADP-ribosyl Cyclase 1/metabolism , Alleles , Animals , Antigens, CD34/metabolism , Cell Lineage , Cell Proliferation , Disease Models, Animal , Erythroid Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genotype , Hematopoiesis/genetics , Hematopoietic System/cytology , Hematopoietic System/metabolism , Humans , Megakaryocytes/metabolism , Mice , Mice, Knockout , Microarray Analysis , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Sequence Analysis, RNA , Stem Cells/metabolism , Transcriptional Regulator ERG , Transcriptome
8.
Proc Natl Acad Sci U S A ; 111(16): 5884-9, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24711413

ABSTRACT

Thrombopoietin (TPO) acting via its receptor, the cellular homologue of the myeloproliferative leukemia virus oncogene (Mpl), is the major cytokine regulator of platelet number. To precisely define the role of specific hematopoietic cells in TPO-dependent hematopoiesis, we generated mice that express the Mpl receptor normally on stem/progenitor cells but lack expression on megakaryocytes and platelets (Mpl(PF4cre/PF4cre)). Mpl(PF4cre/PF4cre) mice displayed profound megakaryocytosis and thrombocytosis with a remarkable expansion of megakaryocyte-committed and multipotential progenitor cells, the latter displaying biological responses and a gene expression signature indicative of chronic TPO overstimulation as the underlying causative mechanism, despite a normal circulating TPO level. Thus, TPO signaling in megakaryocytes is dispensable for platelet production; its key role in control of platelet number is via generation and stimulation of the bipotential megakaryocyte precursors. Nevertheless, Mpl expression on megakaryocytes and platelets is essential to prevent megakaryocytosis and myeloproliferation by restricting the amount of TPO available to stimulate the production of megakaryocytes from the progenitor cell pool.


Subject(s)
Blood Platelets/metabolism , Megakaryocytes/metabolism , Myeloid Cells/cytology , Receptors, Thrombopoietin/metabolism , Thrombopoiesis , Animals , Antigens, CD34/metabolism , Blood Platelets/cytology , Cell Compartmentation , Cell Proliferation , Clone Cells , Gene Expression Profiling , Gene Expression Regulation , Gene Targeting , Genetic Loci/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Integrases/metabolism , Megakaryocytes/cytology , Mice , Models, Biological , Myeloid Cells/metabolism , Thrombocytosis , Thrombopoietin/genetics , Thrombopoietin/metabolism , Transcription, Genetic
9.
Proc Natl Acad Sci U S A ; 110(22): 9031-5, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23671076

ABSTRACT

Previous studies have shown that mouse bone marrow cells can produce mast cells when stimulated in vitro by stem cell factor (SCF) and interleukin-3 (IL-3). Experiments to define the marrow cells able to generate mast cells showed that the most active subpopulations were the Kit(+) Sca1(-) progenitor cell fraction and the more ancestral Kit(+) Sca1(+) blast colony-forming cell fraction. In clonal cultures, up to 64% of blast colony-forming cells were able to generate mast cells when stimulated by SCF and IL-3, and, of these, the most active were those in the CD34(-) Flt3R(-) long-term repopulating cell fraction. Basophils, identified by the monoclonal antibody mMCP-8 to mouse mast cell serine protease-8, were also produced by 50% of blast colony-forming cells with a strong concordance in the production of both cell types by individual blast colony-forming cells. Enriched populations of marrow-derived basophils were shown to generate variable numbers of mast cells after a further incubation with SCF and IL-3. The data extend the repertoire of lineage-committed cells able to be produced by multipotential hematopoietic blast colony-forming cells and show that basophils and mast cells can have common ancestral cells and that basophils can probably generate mast cells at least under defined in vitro conditions.


Subject(s)
Basophils/cytology , Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Mast Cells/cytology , Animals , Antibodies, Monoclonal , Azure Stains , Flow Cytometry , In Vitro Techniques , Mice , Mice, Inbred C57BL , Stem Cell Factor
10.
J Immunol ; 188(1): 122-34, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22105998

ABSTRACT

The lamin B receptor (LBR) is a highly unusual inner nuclear membrane protein with multiple functions. Reduced levels are associated with decreased neutrophil lobularity, whereas complete absence of LBR results in severe skeletal dysplasia and in utero/perinatal lethality. We describe a mouse pedigree, Lym3, with normal bone marrow and thymic development but profound and progressive lymphopenia particularly within the T cell compartment. This defect arises from a point mutation within the Lbr gene with only trace mutant protein detectable in homozygotes, albeit sufficient for normal development. Reduced T cell homeostatic proliferative potential and life span in vivo were found to contribute to lymphopenia. To investigate the role of LBR in gene silencing in hematopoietic cells, we examined gene expression in wild-type and mutant lymph node CD8 T cells and bone marrow neutrophils. Although LBR deficiency had a very mild impact on gene expression overall, for common genes differentially expressed in both LBR-deficient CD8 T cells and neutrophils, gene upregulation prevailed, supporting a role for LBR in their suppression. In summary, this study demonstrates that LBR deficiency affects not only nuclear architecture but also proliferation, cell viability, and gene expression of hematopoietic cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cellular Senescence/immunology , Hematopoietic Stem Cells/immunology , Lymphopenia/immunology , Point Mutation , Receptors, Cytoplasmic and Nuclear , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Cell Survival/genetics , Cell Survival/immunology , Cellular Senescence/genetics , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Hematopoietic Stem Cells/pathology , Lymphopenia/genetics , Lymphopenia/pathology , Mice , Mice, Knockout , Lamin B Receptor
11.
Mucosal Immunol ; 17(3): 371-386, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38492744

ABSTRACT

Interleukin-(IL) 22 production by intestinal group 3 innate lymphoid cells (ILC3) is critical to maintain gut homeostasis. However, IL-22 needs to be tightly controlled; reduced IL-22 expression is associated with intestinal epithelial barrier defect while its overexpression promotes tumor development. Here, using a single-cell ribonucleic acid sequencing approach, we identified a core set of genes associated with increased IL-22 production by ILC3. Among these genes, programmed cell death 1 (PD-1), extensively studied in the context of cancer and chronic infection, was constitutively expressed on a subset of ILC3. These cells, found in the crypt of the small intestine and colon, displayed superior capacity to produce IL-22. PD-1 expression on ILC3 was dependent on the microbiota and was induced during inflammation in response to IL-23 but, conversely, was reduced in the presence of Notch ligand. PD-1+ ILC3 exhibited distinct metabolic activity with increased glycolytic, lipid, and polyamine synthesis associated with augmented proliferation compared with their PD-1- counterparts. Further, PD-1+ ILC3 showed increased expression of mitochondrial antioxidant proteins which enable the cells to maintain their levels of reactive oxygen species. Loss of PD-1 signaling in ILC3 led to reduced IL-22 production in a cell-intrinsic manner. During inflammation, PD-1 expression was increased on natural cytotoxicity receptor (NCR)- ILC3 while deficiency in PD-1 expression resulted in increased susceptibility to experimental colitis and failure to maintain gut barrier integrity. Collectively, our findings uncover a new function of the PD-1 and highlight the role of PD-1 signaling in the maintenance of gut homeostasis mediated by ILC3 in mice.


Subject(s)
Homeostasis , Immunity, Innate , Interleukin-22 , Interleukins , Lymphocytes , Mice, Knockout , Programmed Cell Death 1 Receptor , Animals , Mice , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Lymphocytes/immunology , Lymphocytes/metabolism , Interleukins/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Signal Transduction , Colitis/immunology , Intestines/immunology , Mice, Inbred C57BL , Humans , Disease Models, Animal
12.
Sci Immunol ; 8(85): eabo4365, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37450574

ABSTRACT

Vγ9Vδ2 T cells are the largest population of γδ T cells in adults and can play important roles in providing effective immunity against cancer and infection. Many studies have suggested that peripheral Vγ9Vδ2 T cells are derived from the fetal liver and thymus and that the postnatal thymus plays little role in the development of these cells. More recent evidence suggested that these cells may also develop postnatally in the thymus. Here, we used high-dimensional flow cytometry, transcriptomic analysis, functional assays, and precursor-product experiments to define the development pathway of Vγ9Vδ2 T cells in the postnatal thymus. We identify three distinct stages of development for Vγ9Vδ2 T cells in the postnatal thymus that are defined by the progressive acquisition of functional potential and major changes in the expression of transcription factors, chemokines, and other surface markers. Furthermore, our analysis of donor-matched thymus and blood revealed that the molecular requirements for the development of functional Vγ9Vδ2 T cells are delivered predominantly by the postnatal thymus and not in the periphery. Tbet and Eomes, which are required for IFN-γ and TNFα expression, are up-regulated as Vγ9Vδ2 T cells mature in the thymus, and mature thymic Vγ9Vδ2 T cells rapidly express high levels of these cytokines after stimulation. Similarly, the postnatal thymus programs Vγ9Vδ2 T cells to express the cytolytic molecules, perforin, granzyme A, and granzyme K. This study provides a greater understanding of how Vγ9Vδ2 T cells develop in humans and may lead to opportunities to manipulate these cells to treat human diseases.


Subject(s)
Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocyte Subsets , Adult , Humans , Thymus Gland , Gene Expression Profiling
13.
Proc Natl Acad Sci U S A ; 106(33): 13814-9, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19666492

ABSTRACT

Fli-1 and Erg are closely related members of the Ets family of transcription factors. Both genes are translocated in human cancers, including Ewing's sarcoma, leukemia, and in the case of Erg, more than half of all prostate cancers. Although evidence from mice and humans suggests that Fli-1 is required for megakaryopoiesis, and that Erg is required for normal adult hematopoietic stem cell (HSC) regulation, their precise physiological roles remain to be defined. To elucidate the relationship between Fli-1 and Erg in hematopoiesis, we conducted an analysis of mice carrying mutations in both genes. Our results demonstrate that there is a profound genetic interaction between Fli-1 and Erg. Double heterozygotes displayed phenotypes more dramatic than single heterozygotes: severe thrombocytopenia, with a significant deficit in megakaryocyte numbers and evidence of megakaryocyte dysmorphogenesis, and loss of HSCs accompanied by a reduction in the number of committed hematopoietic progenitor cells. These results illustrate an indispensable requirement for both Fli-1 and Erg in normal HSC and megakaryocyte homeostasis, and suggest these transcription factors may coregulate common target genes.


Subject(s)
Gene Expression Regulation , Megakaryocytes/cytology , Oncogene Proteins/chemistry , Proto-Oncogene Protein c-fli-1/chemistry , Animals , Blood Platelets/metabolism , Cell Lineage , Crosses, Genetic , Heterozygote , Humans , Mice , Mice, Transgenic , Models, Genetic , Mutation , Thrombopoietin/metabolism , Transcription Factors , Transcriptional Regulator ERG
14.
Nat Cell Biol ; 23(3): 219-231, 2021 03.
Article in English | MEDLINE | ID: mdl-33649477

ABSTRACT

Regulation of haematopoietic stem and progenitor cell (HSPC) fate is crucial during homeostasis and under stress conditions. Here we examine the aetiology of the Flt3 ligand (Flt3L)-mediated increase of type 1 conventional dendritic cells (cDC1s). Using cellular barcoding we demonstrate this occurs through selective clonal expansion of HSPCs that are primed to produce cDC1s and not through activation of cDC1 fate by other HSPCs. In particular, multi/oligo-potent clones selectively amplify their cDC1 output, without compromising the production of other lineages, via a process we term tuning. We then develop Divi-Seq to simultaneously profile the division history, surface phenotype and transcriptome of individual HSPCs. We discover that Flt3L-responsive HSPCs maintain a proliferative 'early progenitor'-like state, leading to the selective expansion of multiple transitional cDC1-primed progenitor stages that are marked by Irf8 expression. These findings define the mechanistic action of Flt3L through clonal tuning, which has important implications for other models of 'emergency' haematopoiesis.


Subject(s)
Cell Proliferation/drug effects , Dendritic Cells/drug effects , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Membrane Proteins/pharmacology , RNA-Seq , Single-Cell Analysis , Transcriptome/drug effects , Animals , Cell Lineage , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
15.
Infect Immun ; 78(6): 2734-44, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20368343

ABSTRACT

Genetic linkage studies of the host response to Leishmania major, the causative agent of cutaneous leishmaniasis, have identified significant genetic complexity in humans and mice. In the mouse model, multiple loci have been implicated in susceptibility to infection, but to date, the genes underlying these loci have not been identified. We now describe the contribution of a novel candidate gene, Fli1, to both L. major resistance and enhanced wound healing. We have previously mapped the L. major response locus, lmr2, to proximal chromosome 9 in a genetic cross between the resistant C57BL/6 strain and the susceptible BALB/c strain. We now show that the presence of the resistant C57BL/6 lmr2 allele in susceptible BALB/c mice confers an enhanced L. major resistance and wound healing phenotype. Fine mapping of the lmr2 locus permitted the localization of the lmr2 quantitative trait locus to a 5-Mb interval comprising 21 genes, of which microarray analysis was able to identify differential expression in 1 gene-Fli1. Analysis of Fli1 expression in wounded and L. major-infected skin and naïve and infected lymph nodes validated the importance of Fli1 in lesion resolution and wound healing and identified 3 polymorphisms in the Fli1 promoter, among which a GA repeat element may be the important contributor.


Subject(s)
Genetic Predisposition to Disease , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Proto-Oncogene Protein c-fli-1/physiology , Wound Healing , Animals , Chromosome Mapping , Crosses, Genetic , Female , Gene Expression Profiling , Genetic Loci , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Polymorphism, Genetic , Promoter Regions, Genetic
16.
Cell Stem Cell ; 25(2): 258-272.e9, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31374198

ABSTRACT

Tumors are composed of phenotypically heterogeneous cancer cells that often resemble various differentiation states of their lineage of origin. Within this hierarchy, it is thought that an immature subpopulation of tumor-propagating cancer stem cells (CSCs) differentiates into non-tumorigenic progeny, providing a rationale for therapeutic strategies that specifically eradicate CSCs or induce their differentiation. The clinical success of these approaches depends on CSC differentiation being unidirectional rather than reversible, yet this question remains unresolved even in prototypically hierarchical malignancies, such as acute myeloid leukemia (AML). Here, we show in murine and human models of AML that, upon perturbation of endogenous expression of the lineage-determining transcription factor PU.1 or withdrawal of established differentiation therapies, some mature leukemia cells can de-differentiate and reacquire clonogenic and leukemogenic properties. Our results reveal plasticity of CSC maturation in AML, highlighting the need to therapeutically eradicate cancer cells across a range of differentiation states.


Subject(s)
Cell Differentiation/physiology , Cell Transdifferentiation/physiology , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/physiology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Carcinogenesis , Cell Plasticity , Cells, Cultured , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Tretinoin/metabolism
17.
J Leukoc Biol ; 104(1): 195-204, 2018 07.
Article in English | MEDLINE | ID: mdl-29758105

ABSTRACT

Eosinophils are important in fighting parasitic infections and are implicated in the pathogenesis of asthma and allergy. IL-5 is a critical regulator of eosinophil development, controlling proliferation, differentiation, and maturation of the lineage. Mice that constitutively express IL-5 have in excess of 10-fold more eosinophils in the hematopoietic organs than their wild type (WT) counterparts. We have identified that much of this expansion is in a population of Siglec-F high eosinophils, which are rare in WT mice. In this study, we assessed transcription in myeloid progenitors, eosinophil precursors, and Siglec-F medium and Siglec-F high eosinophils from IL-5 transgenic mice and in doing so have created a useful resource for eosinophil biologists. We have then utilized these populations to construct an eosinophil trajectory based on gene expression and to identify gene sets that are associated with eosinophil lineage progression. Cell cycle genes were significantly associated with the trajectory, and we experimentally demonstrate an increasing trend toward quiescence along the trajectory. Additionally, we found gene expression changes associated with constitutive IL-5 signaling in eosinophil progenitors, many of which were not observed in eosinophils.


Subject(s)
Eosinophils/immunology , Gene Expression Profiling , Interleukin-5/immunology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Eosinophils/cytology , Eosinophils/metabolism , Interleukin-5/metabolism , Mice , Mice, Transgenic , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/metabolism
18.
J Leukoc Biol ; 104(1): 123-133, 2018 07.
Article in English | MEDLINE | ID: mdl-29645346

ABSTRACT

In recent years multi-parameter flow cytometry has enabled identification of cells at major stages in myeloid development; from pluripotent hematopoietic stem cells, through populations with increasingly limited developmental potential (common myeloid progenitors and granulocyte-macrophage progenitors), to terminally differentiated mature cells. Myeloid progenitors are heterogeneous, and the surface markers that define transition states from progenitors to mature cells are poorly characterized. Siglec-F is a surface glycoprotein frequently used in combination with IL-5 receptor alpha (IL5Rα) for the identification of murine eosinophils. Here, we describe a CD11b+ Siglec-F+ IL5Rα- myeloid population in the bone marrow of C57BL/6 mice. The CD11b+ Siglec-F+ IL5Rα- cells are retained in eosinophil deficient PHIL mice, and are not expanded upon overexpression of IL-5, indicating that they are upstream or independent of the eosinophil lineage. We show these cells to have GMP-like developmental potential in vitro and in vivo, and to be transcriptionally distinct from the classically described GMP population. The CD11b+ Siglec-F+ IL5Rα- population expands in the bone marrow of Myb mutant mice, which is potentially due to negative transcriptional regulation of Siglec-F by Myb. Lastly, we show that the role of Siglec-F may be, at least in part, to regulate GMP viability.


Subject(s)
Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Animals , Cell Differentiation/physiology , Mice , Mice, Inbred C57BL
19.
Stem Cell Reports ; 7(3): 571-582, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27499199

ABSTRACT

Hematopoiesis is a multistage process involving the differentiation of stem and progenitor cells into distinct mature cell lineages. Here we present Haemopedia, an atlas of murine gene-expression data containing 54 hematopoietic cell types, covering all the mature lineages in hematopoiesis. We include rare cell populations such as eosinophils, mast cells, basophils, and megakaryocytes, and a broad collection of progenitor and stem cells. We show that lineage branching and maturation during hematopoiesis can be reconstructed using the expression patterns of small sets of genes. We also have identified genes with enriched expression in each of the mature blood cell lineages, many of which show conserved lineage-enriched expression in human hematopoiesis. We have created an online web portal called Haemosphere to make analyses of Haemopedia and other blood cell transcriptional datasets easier. This resource provides simple tools to interrogate gene-expression-based relationships between hematopoietic cell types and genes of interest.


Subject(s)
Blood Cells/cytology , Blood Cells/metabolism , Computational Biology , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cluster Analysis , Computational Biology/methods , Gene Expression Profiling , Humans , Mice , Web Browser
20.
Infect Immun ; 71(12): 6830-4, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14638769

ABSTRACT

Inbred strains of mice infected with Leishmania major have been classified as genetically resistant or susceptible on the basis of their ability to cure their lesions, the parasite burden in the draining lymph nodes, and their type of T helper cell immune responses to the parasite. Using the intradermal infection at the base of the tail and the ear pinna, we compared for the first time the above-mentioned parameters in six strains of mice infected with metacyclic promastigotes, and we show that the severity of disease depends greatly on the site of infection. Although the well-documented pattern of disease susceptibility of BALB/c and C57BL/6 mice described for the footpad and base-of-the-tail models of leishmaniasis were confirmed, C3H/HeN and DBA/2 mice, which are intermediate and susceptible, respectively, in the tail and other models, were resistant to ear infection. Moreover, in the CBA/H, C3H/HeN, C57BL/6J, and DBA/2 mouse strains, there was little correlation between the pattern of cytokines produced and the disease phenotype observed at the ear and tail sites. We conclude that the definition of susceptibility and the immune mechanisms leading to susceptibility or resistance to infection may differ substantially depending on the route of infection.


Subject(s)
Ear/parasitology , Leishmania major/pathogenicity , Leishmaniasis, Cutaneous/immunology , Severity of Illness Index , Tail/parasitology , Animals , Female , Injections, Intradermal , Leishmania major/immunology , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/physiopathology , Mice , Mice, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL