Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Front Immunol ; 13: 1095453, 2022.
Article in English | MEDLINE | ID: mdl-36703974

ABSTRACT

Introduction: The granulocyte colony-stimulating factor receptor (G-CSFR), encoded by the CSF3R gene, is involved in the production and function of neutrophilic granulocytes. Somatic mutations in CSF3R leading to truncated G-CSFR forms are observed in acute myeloid leukemia (AML), particularly those subsequent to severe chronic neutropenia (SCN), as well as in a subset of patients with other leukemias. Methods: This investigation introduced equivalent mutations into the zebrafish csf3r gene via genome editing and used a range of molecular and cellular techniques to understand the impact of these mutations on immune cells across the lifespan. Results: Zebrafish harboring truncated G-CSFRs showed significantly enhanced neutrophil production throughout successive waves of embryonic hematopoiesis and a neutrophil maturation defect in adults, with the mutations acting in a partially dominant manner. Discussion: This study has elucidated new insights into the impact of G-CSFR truncations throughout the life-course and created a bone fide zebrafish model for further investigation.


Subject(s)
Hematopoiesis , Receptors, Granulocyte Colony-Stimulating Factor , Animals , Hematopoiesis/genetics , Leukemia, Myeloid, Acute/genetics , Leukopoiesis/genetics , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Zebrafish
2.
Nat Commun ; 12(1): 6843, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824268

ABSTRACT

Integration of external signals and B-lymphoid transcription factor activities organise B cell lineage commitment through alternating cycles of proliferation and differentiation, producing a diverse repertoire of mature B cells. We use single-cell transcriptomics/proteomics to identify differentially expressed gene networks across B cell development and correlate these networks with subtypes of B cell leukemia. Here we show unique transcriptional signatures that refine the pre-B cell expansion stages into pre-BCR-dependent and pre-BCR-independent proliferative phases. These changes correlate with reciprocal changes in expression of the transcription factor EBF1 and the RNA binding protein YBX3, that are defining features of the pre-BCR-dependent stage. Using pseudotime analysis, we further characterize the expression kinetics of different biological modalities across B cell development, including transcription factors, cytokines, chemokines, and their associated receptors. Our findings demonstrate the underlying heterogeneity of developing B cells and characterise developmental nodes linked to B cell transformation.


Subject(s)
B-Lymphocytes/cytology , Gene Regulatory Networks , Leukopoiesis/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Proliferation/genetics , Gene Expression Profiling , Gene Expression Regulation , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Prognosis , Proteomics , Single-Cell Analysis , Trans-Activators/genetics , Trans-Activators/metabolism
3.
Genes (Basel) ; 11(5)2020 05 04.
Article in English | MEDLINE | ID: mdl-32375333

ABSTRACT

The majority of genetic variants affecting complex traits map to regulatory regions of genes, and typically lie in credible intervals of 100 or more SNPs. Fine mapping of the causal variant(s) at a locus depends on assays that are able to discriminate the effects of polymorphisms or mutations on gene expression. Here, we evaluated a moderate-throughput CRISPR-Cas9 mutagenesis approach, based on replicated measurement of transcript abundance in single-cell clones, by deleting candidate regulatory SNPs, affecting four genes known to be affected by large-effect expression Quantitative Trait Loci (eQTL) in leukocytes, and using Fluidigm qRT-PCR to monitor gene expression in HL60 pro-myeloid human cells. We concluded that there were multiple constraints that rendered the approach generally infeasible for fine mapping. These included the non-targetability of many regulatory SNPs, clonal variability of single-cell derivatives, and expense. Power calculations based on the measured variance attributable to major sources of experimental error indicated that typical eQTL explaining 10% of the variation in expression of a gene would usually require at least eight biological replicates of each clone. Scanning across credible intervals with this approach is not recommended.


Subject(s)
CRISPR-Cas Systems , Chromosome Mapping/methods , Genome-Wide Association Study/methods , Multifactorial Inheritance/genetics , Mutagenesis , Polymorphism, Single Nucleotide , Quantitative Trait Loci/genetics , Scientific Experimental Error , Single-Cell Analysis/methods , Causality , Cell Lineage , Clone Cells , Gene Deletion , HL-60 Cells , Humans , Leukopoiesis/genetics , Neutrophils/cytology , Quantitative Trait, Heritable , RNA-Seq , Reproducibility of Results , Sequence Deletion
4.
DNA Cell Biol ; 39(4): 548-554, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32155344

ABSTRACT

The Qing-Tibet Plateau is characterized by low oxygen pressure, which is an important biomedical and ecological stressor. However, the variation in gene expression during periods of stay on the plateau has not been well studied. We recruited eight volunteers to stay on the plateau for 3, 7, and 30 days. Human Clariom D arrays were used to measure transcriptome changes in the mRNA expression profiles in these volunteers' blood. Analysis of variance (ANOVA) indicated that 699 genes were significantly differentially expressed in response to entering the plateau during hypoxic exposure. The genes with changes in transcript abundance were involved in the terms phosphoprotein, acetylation, protein binding, and protein transport. Furthermore, numerous genes involved in hematopoietic functions, including erythropoiesis and immunoregulation, were differentially expressed in response to hypoxia. This phenomenon may be one of reasons why the majority of people entering the plateau do not have excessive erythrocyte proliferation and are susceptible to infection.


Subject(s)
Acclimatization/genetics , Altitude Sickness/genetics , Altitude Sickness/physiopathology , Hypoxia/physiopathology , Leukocytes/physiology , Altitude , Erythrocytes/cytology , Erythropoiesis/genetics , Gene Expression Profiling , Gene Expression Regulation/genetics , Humans , Leukopoiesis/genetics , Male , Oxygen , Tibet
5.
PLoS One ; 14(12): e0226435, 2019.
Article in English | MEDLINE | ID: mdl-31869378

ABSTRACT

Acute myeloid leukemia (AML) is characterized by recurrent mutations that affect normal hematopoiesis. The analysis of human AMLs has mostly been performed using end-point materials, such as cell lines and patient derived AMLs that also carry additional contributing mutations. The molecular effects of a single oncogenic hit, such as expression of the AML associated oncoprotein AML1-ETO on hematopoietic development and transformation into a (pre-) leukemic state still needs further investigation. Here we describe the development and characterization of an induced pluripotent stem cell (iPSC) system that allows in vitro differentiation towards different mature myeloid cell types such as monocytes and granulocytes. During in vitro differentiation we expressed the AML1-ETO fusion protein and examined the effects of the oncoprotein on differentiation and the underlying alterations in the gene program at 8 different time points. Our analysis revealed that AML1-ETO as a single oncogenic hit in a non-mutated background blocks granulocytic differentiation, deregulates the gene program via altering the acetylome of the differentiating granulocytic cells, and induces t(8;21) AML associated leukemic characteristics. Together, these results reveal that inducible oncogene expression during in vitro differentiation of iPS cells provides a valuable platform for analysis of aberrant regulation in disease.


Subject(s)
Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 2 Subunit/physiology , Granulocytes/physiology , Induced Pluripotent Stem Cells/physiology , Oncogene Proteins, Fusion/physiology , RUNX1 Translocation Partner 1 Protein/physiology , Transcriptome , Cell Proliferation/genetics , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , Granulocytes/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukopoiesis/genetics , Monocytes/physiology , Myelopoiesis/genetics , Oncogene Proteins, Fusion/genetics , Oncogenes/physiology , RUNX1 Translocation Partner 1 Protein/genetics , Transcriptome/genetics , Transfection
6.
Stem Cell Reports ; 13(6): 1099-1110, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31708474

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) can serve as a versatile and scalable source of neutrophils for biomedical research and transfusion therapies. Here we describe a rapid efficient serum- and xenogen-free protocol for neutrophil generation, which is based on direct hematoendothelial programming of hiPSCs using ETV2-modified mRNA. Culture of ETV2-induced hematoendothelial progenitors in the presence of GM-CSF, FGF2, and UM171 led to continuous production of generous amounts of CD34+CD33+ myeloid progenitors which could be harvested every 8-10 days for up to 30 days of culture. Subsequently, myeloid progenitors were differentiated into neutrophils in the presence of G-CSF and the retinoic acid agonist Am580. Neutrophils obtained in these conditions displayed a typical somatic neutrophil morphology, produced reactive oxygen species, formed neutrophil extracellular traps and possessed phagocytic and chemotactic activities. Overall, this technology offers an opportunity to generate a significant number of neutrophils as soon as 14 days after initiation of differentiation.


Subject(s)
Cell Differentiation/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Neutrophils/immunology , Neutrophils/metabolism , RNA, Messenger , Transcription Factors/genetics , Biomarkers , Cells, Cultured , Extracellular Traps/genetics , Extracellular Traps/metabolism , Gene Expression Regulation, Developmental , Hematopoiesis , Humans , Immunophenotyping , Leukopoiesis/genetics , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Neutrophils/cytology
7.
Front Immunol ; 10: 1912, 2019.
Article in English | MEDLINE | ID: mdl-31474989

ABSTRACT

Neutrophils are implicated in almost every stage of oncogenesis and paradoxically display anti- and pro-tumor properties. Accumulating evidence indicates that neutrophils display diversity in their phenotype resulting from functional plasticity and/or changes to granulopoiesis. In cancer, neutrophils at a range of maturation stages can be identified in the blood and tissues (i.e., outside of their developmental niche). The functional capacity of neutrophils at different states of maturation is poorly understood resulting from challenges in their isolation, identification, and investigation. Thus, the impact of neutrophil maturity on cancer progression and therapy remains enigmatic. In this review, we discuss the identification, prevalence, and function of immature and mature neutrophils in cancer and the potential impact of this on tumor progression and cancer therapy.


Subject(s)
Cell Differentiation/immunology , Hematopoietic Stem Cells/immunology , Leukopoiesis/immunology , Neoplasms/immunology , Neutrophils/immunology , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/immunology , CCAAT-Binding Factor/metabolism , Cell Differentiation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Leukopoiesis/genetics , Neoplasms/genetics , Neoplasms/therapy , Neutrophils/cytology , Neutrophils/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/immunology , Trans-Activators/metabolism
8.
Sci Rep ; 9(1): 8451, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31186478

ABSTRACT

Nanosecond pulsed electric fields (nsPEFs) have gained attention as a novel physical stimulus for life sciences. Although cancer therapy is currently their promising application, nsPEFs have further potential owing to their ability to elicit various cellular responses. This study aimed to explore stimulatory actions of nsPEFs, and we used HL-60 cells that were differentiated into neutrophils under cultured conditions. Exposure of neutrophil-differentiated HL-60 cells to nsPEFs led to the extracellular release of chromosomal DNA, which appears to be equivalent to neutrophil extracellular traps (NETs) that serve as a host defense mechanism against pathogens. Fluorometric measurement of extracellular DNA showed that DNA extrusion was rapidly induced after nsPEF exposure and increased over time. Western blot analysis demonstrated that nsPEFs induced histone citrullination that is the hydrolytic conversion of arginine to citrulline on histones and facilitates chromatin decondensation. DNA extrusion and histone citrullination by nsPEFs were cell type-specific and Ca2+-dependent events. Taken together, these observations suggest that nsPEFs drive the mechanism for neutrophil-specific immune response without infection, highlighting a novel aspect of nsPEFs as a physical stimulus.


Subject(s)
Apoptosis/radiation effects , Cell Differentiation/radiation effects , Electric Stimulation , Neutrophils/radiation effects , Apoptosis/genetics , Chromatin/genetics , Chromatin/radiation effects , Citrullination/genetics , Citrullination/radiation effects , DNA/genetics , DNA/radiation effects , Extracellular Traps/genetics , Extracellular Traps/radiation effects , HL-60 Cells , HeLa Cells , Histones/genetics , Histones/radiation effects , Humans , Leukopoiesis/genetics , Leukopoiesis/radiation effects
9.
Am J Physiol Regul Integr Comp Physiol ; 315(2): R218-R229, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29641231

ABSTRACT

The interaction between C-X-C chemokine receptor type 4 (CXCR4) and its cognate ligand C-X-C motif chemokine ligand 12 (CXCL12) plays a critical role in regulating hematopoietic stem cell activation and subsequent cellular mobilization. Extensive studies of these genes have been conducted in mammals, but much less is known about the expression and function of CXCR4 and CXCL12 in non-mammalian vertebrates. In the present study, we identify simultaneous expression of CXCR4 and CXCL12 orthologs in the epigonal organ (the primary hematopoietic tissue) of the little skate, Leucoraja erinacea. Genetic and phylogenetic analyses were functionally supported by significant mobilization of leukocytes following administration of Plerixafor, a CXCR4 antagonist and clinically important drug. Our results provide evidence that, as in humans, Plerixafor disrupts CXCR4/CXCL12 binding in the little skate, facilitating release of leukocytes into the bloodstream. Our study illustrates the value of the little skate as a model organism, particularly in studies of hematopoiesis and potentially for preclinical research on hematological and vascular disorders.


Subject(s)
Chemokine CXCL12/metabolism , Fish Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Leukocytes/metabolism , Leukopoiesis , Receptors, CXCR4/metabolism , Skates, Fish/metabolism , Animals , Benzylamines , Chemokine CXCL12/genetics , Cyclams , Fish Proteins/genetics , Gene Expression Regulation , Hematopoietic Stem Cells/drug effects , Heterocyclic Compounds/pharmacology , Leukocytes/drug effects , Leukopoiesis/drug effects , Leukopoiesis/genetics , Phylogeny , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Signal Transduction , Skates, Fish/genetics , Transcriptome
10.
Semin Cell Dev Biol ; 75: 23-39, 2018 03.
Article in English | MEDLINE | ID: mdl-28866328

ABSTRACT

Splicing is a ubiquitous process in eukaryotic cells, long recognised as contributing to diversity of the transcriptome. More specifically, splicing fine-tunes the transcriptome output for highly individual outcomes at different stages of cell development, in specific timeframes, which when perturbed result in significant human diseases. Granulopoiesis provides a particularly well studied example of how splicing can be a highly flexible but tightly regulated process. Focusing on the specific case of granulopoiesis, this review surveys the contribution of cis-splicing variations in individual genes and the trans-regulation of global splicing outcomes during the normal development of neutrophils. Further, the contribution of splicing dysfunction to the pathogenesis of diseases of neutrophil number, function and maturation including hereditary neutropenia, myelodysplasia, and acute myeloid leukaemia is explored.


Subject(s)
Alternative Splicing , Leukopoiesis/genetics , Neutropenia/genetics , Neutrophils/metabolism , Animals , Gene Expression Regulation , Genetic Predisposition to Disease/genetics , Humans , Models, Genetic , Mutation
11.
PLoS Genet ; 13(10): e1007027, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28976973

ABSTRACT

MicroRNAs are universal post-transcriptional regulators in genomes. They have the ability of buffering gene expressional programs, contributing to robustness of biological systems and playing important roles in development, physiology and diseases. Here, we identified a microRNA, miR-125a, as a positive regulator of granulopoiesis. MiR125a knockout mice show reduced infiltration of neutrophils in the lung and alleviated tissue destruction after endotoxin challenge as a consequence of decreased neutrophil numbers. Furthermore, we demonstrated that this significant reduction of neutrophils was due to impaired development of granulocyte precursors to mature neutrophils in an intrinsic manner. We showed that Socs3, a critical repressor for granulopoiesis, was a target of miR-125a. Overall, our study revealed a new microRNA regulating granulocyte development and supported a model in which miR-125a acted as a fine-tuner of granulopoiesis.


Subject(s)
Leukopoiesis/genetics , Leukopoiesis/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , Neutrophils/cytology , Neutrophils/metabolism , 3' Untranslated Regions , Animals , Binding Sites/genetics , Cell Death , Cell Differentiation , Cell Proliferation , Granulocyte Colony-Stimulating Factor/metabolism , Granulocytes/cytology , Granulocytes/metabolism , Mice , Mice, Knockout , Models, Biological , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Shock, Septic/genetics , Shock, Septic/metabolism , Shock, Septic/pathology , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism
12.
Nat Commun ; 8(1): 46, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28663557

ABSTRACT

Transcription factor C/EBPα is a master regulator of myelopoiesis and its inactivation is associated with acute myeloid leukemia. Deregulation of C/EBPα by microRNAs during granulopoiesis or acute myeloid leukemia development has not been studied. Here we show that oncogenic miR-182 is a strong regulator of C/EBPα. Moreover, we identify a regulatory loop between C/EBPα and miR-182. While C/EBPα blocks miR-182 expression by direct promoter binding during myeloid differentiation, enforced expression of miR-182 reduces C/EBPα protein level and impairs granulopoiesis in vitro and in vivo. In addition, miR-182 expression is highly elevated particularly in acute myeloid leukemia patients with C-terminal CEBPA mutations, thereby depicting a mechanism by which C/EBPα blocks miR-182 expression. Furthermore, we present miR-182 expression as a prognostic marker in cytogenetically high-risk acute myeloid leukemia patients. Our data demonstrate the importance of a controlled balance between C/EBPα and miR-182 for the maintenance of healthy granulopoiesis.C/EBPα is a critical transcription factor involved in myelopoiesis and its inactivation is associated with acute myeloid leukemia (AML). Here the authors show a negative feedback loop between C/EBPα and miR-182 and identify this miRNA as a marker of high-risk AML.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Granulocytes , Leukemia, Myeloid, Acute/genetics , Leukopoiesis/genetics , MicroRNAs/genetics , Animals , Blotting, Western , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/genetics , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Mice , Mice, Knockout , MicroRNAs/metabolism , Prognosis , Real-Time Polymerase Chain Reaction
13.
Nat Commun ; 8: 14911, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28382966

ABSTRACT

In response to infection and injury, the neutrophil population rapidly expands and then quickly re-establishes the basal state when inflammation resolves. The exact pathways governing neutrophil/macrophage lineage outputs from a common granulocyte-macrophage progenitor are still not completely understood. From a forward genetic screen in zebrafish, we identify the transcriptional repressor, ZBTB11, as critical for basal and emergency granulopoiesis. ZBTB11 sits in a pathway directly downstream of master myeloid regulators including PU.1, and TP53 is one direct ZBTB11 transcriptional target. TP53 repression is dependent on ZBTB11 cys116, which is a functionally critical, metal ion-coordinating residue within a novel viral integrase-like zinc finger domain. To our knowledge, this is the first description of a function for this domain in a cellular protein. We demonstrate that the PU.1-ZBTB11-TP53 pathway is conserved from fish to mammals. Finally, Zbtb11 mutant rescue experiments point to a ZBTB11-regulated TP53 requirement in development of other organs.


Subject(s)
Leukopoiesis/genetics , Neutrophils , Proto-Oncogene Proteins/metabolism , Repressor Proteins/genetics , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Databases, Protein , Signal Transduction , Zebrafish , Zinc Fingers
14.
J Immunol ; 197(5): 1989-99, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27481851

ABSTRACT

Emergency granulopoiesis refers to the increased production of neutrophils in bone marrow and their release into circulation induced by severe infection. Several studies point to a critical role for G-CSF as the main mediator of emergency granulopoiesis. However, the consequences of G-CSF stimulation on the transcriptome of neutrophils and their precursors have not yet been investigated in humans. In this work, we examine the changes in mRNA expression induced by administration of G-CSF in vivo, as a model of emergency granulopoiesis in humans. Blood samples were collected from healthy individuals after 5 d of G-CSF administration. Neutrophil precursors were sorted into discrete stages of maturation by flow cytometry, and RNA was subjected to microarray analysis. mRNA levels were compared with previously published expression levels in corresponding populations of neutrophil precursors isolated from bone marrow of untreated, healthy individuals. One thousand one hundred and ten mRNAs were differentially expressed >2-fold throughout terminal granulopoiesis. Major changes were seen in pathways involved in apoptosis, cytokine signaling, and TLR pathways. In addition, G-CSF treatment reduced the levels of four of five measured granule proteins in mature neutrophils, including the proantibacterial protein hCAP-18, which was completely deficient in neutrophils from G-CSF-treated donors. These results indicate that multiple biological processes are altered to satisfy the increased demand for neutrophils during G-CSF-induced emergency granulopoiesis in humans.


Subject(s)
Gene Expression , Granulocyte Colony-Stimulating Factor/pharmacology , Leukopoiesis/genetics , Neutrophils/physiology , Antimicrobial Cationic Peptides/deficiency , Antimicrobial Cationic Peptides/genetics , Apoptosis/immunology , Cell Movement , Cytokines/immunology , Cytokines/metabolism , Healthy Volunteers , Humans , Microarray Analysis , Neutrophils/drug effects , Recombinant Proteins/immunology , Cathelicidins
15.
Free Radic Biol Med ; 89: 1203-17, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26524400

ABSTRACT

OBJECTIVE: Cell injury releases nucleic acids supporting inflammation and stem cell activation. Here, the impact of extracellular ribonucleic acid, especially transfer RNA (ex-tRNA), on vasculogenesis and leukopoiesis of mouse embryonic stem (ES) cells was investigated. APPROACH AND RESULTS: ex-tRNA, whole cell RNA and ribosomal RNA (ex-rRNA) but not DNA increased CD31-positive vascular structures in embryoid bodies. Ex-tRNA and ex-rRNA increased numbers of VEGFR2(+), CD31(+) and VE-cadherin(+) vascular cells as well as CD18(+), CD45(+) and CD68(+) cells, indicating leukocyte/macrophage differentiation. This was paralleled by mRNA and protein expression of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor-165 (VEGF165) and neuropilin 1 (NRP1), phosphorylation of phosphatidyl inositol 3-kinase (PI3K) and VEGF receptor 2 (VEGFR2) as well as mRNA expression of α-smooth muscle actin (α-SMA). ex-tRNA was taken up by endosomes, increased expression of the pro-angiogenic semaphorin B4 receptor plexin B1 as well as the ephrin-type B receptor 4 (EphB4) and ephrinB2 ligand and enhanced cell migration, which was inhibited by the VEGFR2 antagonist SU5614 and the PI3K inhibitor LY294002. This likewise abolished the effects of ex-tRNA on vasculogenesis and leukopoiesis of ES cells. Ex-tRNA increased NOX1, NOX2, NOX4 and DUOX2 mRNA and boosted the generation of superoxide and hydrogen peroxide which was inhibited by radical scavengers, the NADPH oxidase inhibitors apocynin, VAS2870, ML171, and plumbagin as well as shRNA silencing of NOX1 and NOX4. CONCLUSIONS: Our findings indicate that ex-tRNA treatment induces vasculogenesis and leukopoiesis of ES cells via superoxide/hydrogen peroxide generated by NADPH oxidase and activation of VEGFR2 and PI3K.


Subject(s)
Embryonic Stem Cells/pathology , Leukopoiesis/genetics , Neovascularization, Physiologic/genetics , RNA, Ribosomal/genetics , RNA, Transfer/genetics , Animals , Blotting, Western , Cell Differentiation , Cell Movement , Cells, Cultured , Embryonic Stem Cells/metabolism , Hydrogen Peroxide/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoenzyme Techniques , Mice , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Superoxides/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
Blood ; 124(10): 1610-21, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25030064

ABSTRACT

Cyclooxygenases (COXs) and their prostanoid products play important roles in a diverse range of physiological processes, including in the immune system. Here, we provide evidence that COX-1 is an essential regulator in early stages of B-cell development. COX-1-deficient mice displayed systematic reduction in total B cells, which was attributed to the arrest of early B-cell development from pro-B to pre-B stage. We further demonstrated that this defect was mediated through downregulation of the Janus kinase/signal transducer and activator of transcription 5 (JAK/STAT5) signaling and its target genes, including Pax5, in COX-1(-/-) mice. Mechanistic studies revealed that COX-1-derived thromboxane A2 (TxA2) could regulate JAK3/STAT5 signaling through the cyclic adenosine monophosphate-protein kinase A pathway, via binding with its receptor thromboxane A2 receptor (TP). Administration of the TP agonist could rescue the defective B-cell development and JAK/STAT5 signaling activity in COX-1-deficient mice. Moreover, administration of low-dose aspirin caused a significant reduction in total B cells in peripheral blood of healthy human volunteers, coincidentally with reduced TxA2 production and downregulation of JAK/STAT5 signaling. Taken together, our results demonstrate that COX-1-derived TxA2 plays a critical role in the stage transition of early B-cell development through regulation of JAK/STAT5 signaling and indicate a potential immune-suppressive effect of low-dose aspirin in humans.


Subject(s)
B-Lymphocytes/physiology , Cell Differentiation , Cyclooxygenase 1/metabolism , Membrane Proteins/metabolism , Thromboxane A2/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Cyclooxygenase 1/genetics , Humans , Janus Kinases/metabolism , Leukopoiesis/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT5 Transcription Factor/metabolism , Signal Transduction/genetics , Thromboxane A2/metabolism
17.
Leuk Res ; 38(9): 1117-25, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25043809

ABSTRACT

CIAPIN1 (cytokine-induced antiapoptotic inhibitor 1) was recently identified as an essential downstream effector of the Ras signaling pathway. However, its potential role in regulating myeloid differentiation remains unclear. In this study, we found depletion of CIAPIN1 by shRNAs led to granulocytic differentiation of K562 cells. Meanwhile, the decrease of NHE1 and up-regulation of phosphorylated ERK1/2 were observed after CIAPIN1 depletion. Interestingly, targeted inhibition of NHE1 further promoted the differentiation of K562 cells with CIAPIN1 silencing. Accordingly, ectopic expression of NHE1 reversed this phenotype. Furthermore, ERK1/2 inhibition with the chemical inhibitor, PD98059, abolished CIAPIN1 silencing-induced differentiation of K562 cells after NHE1 inhibition. Thus, our results revealed important mechanism that CIAPIN1 targeted NHE1 to mediate differentiation of K562 cells via ERK1/2 pathway. Our findings implied CIAPIN1 and NHE1 could be new targets in developing therapeutic strategies against leukemia.


Subject(s)
Cation Transport Proteins/genetics , Cell Differentiation/genetics , Intracellular Signaling Peptides and Proteins/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , MAP Kinase Signaling System/physiology , Sodium-Hydrogen Exchangers/genetics , Cation Transport Proteins/antagonists & inhibitors , Cell Differentiation/drug effects , Gene Expression Regulation, Leukemic/drug effects , Gene Knockdown Techniques , Granulocytes/drug effects , Granulocytes/physiology , Guanidines/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukopoiesis/drug effects , Leukopoiesis/genetics , MAP Kinase Signaling System/drug effects , RNA, Small Interfering/pharmacology , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sulfones/pharmacology
18.
Blood ; 123(7): 1079-89, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24398327

ABSTRACT

CCAAT/enhancer binding protein-ε (C/EBP-ε) is considered a master transcription factor regulating terminal neutrophil maturation. It is essential for expression of secondary granule proteins, but it also regulates proliferation, cell cycle, and maturation during granulopoiesis. Cebpe(-/-) mice have incomplete granulocytic differentiation and increased sensitivity toward bacterial infections. The amount of C/EBP-ε messenger RNA (mRNA) increases with maturation from myeloblasts with peak level in myelocytes (MC)/metamyelocytes (MM), when the cells stop proliferating followed by a decline in more mature cells. In contrast, C/EBP-ε protein is virtually detectable only in the MC/MM population, indicating that expression in more immature cells could be inhibited by microRNAs (miRNAs). We found that miRNA-130a (miR-130a) regulates C/EBP-ε protein expression in both murine and human granulocytic precursors. Overexpression of miR-130a in a murine cell line downregulated C/EBP-ε protein and lactoferrin (Ltf), cathelicidin antimicrobial protein (Camp), and lipocalin-2 (Lcn2) mRNA expression giving rise to cells with a more immature phenotype, as seen in the Cebpe(-/-) mouse. Introduction of a C/EBP-ε mRNA without target site for miR-130a restored both C/EBP-ε production, expression of Camp and Lcn2, and resulted in the cells having a more mature phenotype. We conclude that miR-130a is important for the regulation of the timed expression of C/EBP-ε during granulopoiesis.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Granulocytes/physiology , Leukopoiesis/genetics , MicroRNAs/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Gene Expression Regulation , Granulocyte Precursor Cells/physiology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells
20.
Curr Opin Hematol ; 18(6): 421-6, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21912254

ABSTRACT

PURPOSE OF REVIEW: The molecular genetics of the blood group I system and the regulation mechanism for I antigen expression in postnatal red blood cells are intriguing. This review summarizes their elucidation and recent findings. RECENT FINDINGS: Accumulating data from the molecular analysis of individuals with the adult i phenotype supports the proposed molecular genetic mechanism for the partial association of the adult i phenotype with congenital cataracts. Recent investigations have shown that the regulation of I antigen formation during erythropoiesis is determined by transcription factor CCAAT/enhancer binding protein-α (C/EBPα) and the phosphorylation status of C/EBPα Ser-21 residue. SUMMARY: The human I locus is organized such that it has an uncommon genetic architecture and expresses three different I transcript forms. The results obtained from molecular analysis of two adult i groups, with and without congenital cataracts, demonstrate that the molecular background accounts for the partial association between these two traits and suggest that an I gene defect may lead directly to the development of congenital cataracts. Analysis of the regulation for I antigen expression shows that the regulation during erythropoiesis and granulopoiesis share a common mechanism, with dephosphorylation of the Ser-21 residue on C/EBPα playing the critical role.


Subject(s)
Erythropoiesis/physiology , Glycosphingolipids/metabolism , I Blood-Group System/genetics , I Blood-Group System/immunology , Leukopoiesis/physiology , Cataract/congenital , Cataract/immunology , Erythrocytes/metabolism , Erythropoiesis/genetics , Humans , Leukopoiesis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL