Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Br J Haematol ; 203(3): 355-368, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37583269

ABSTRACT

Congenital fibrinogen disorders or CFDs are heterogenous, both in clinical manifestation and array of culprit molecular lesions. Correlations between phenotype and genotype remain poorly defined. This review examines the genetic landscape discovered to date for this rare condition. The question of a possible oligogenic model of inheritance influencing phenotypic heterogeneity is raised, with discussion of the benefits and challenges of sequencing technology used to enhance discovery in this space. Considerable work lies ahead in order to achieve diagnostic and prognostic precision and subsequently provide targeted management to this complex cohort of patients.

2.
Biochem Biophys Res Commun ; 635: 244-251, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36283337

ABSTRACT

Neural tube closure is a dynamic morphogenic event in early embryonic development. Perturbations of this process through either environmental or genetic factors induce the severe congenital malformations known collectively as neural tube defects (NTDs). Deficiencies in maternal folate intake have long been associated with NTDs, as have mutations in critical neurulation genes that include the Grainyhead-like 3 (Grhl3) gene. Mice lacking this gene exhibit fully penetrant thoraco-lumbo-sacral spina bifida and a low incidence of exencephaly. Previous studies have shown that exposure of pregnant mice carrying hypomorphic Grhl3 alleles to exogenous retinoic acid (RA) increases the incidence and severity of NTDs in their offspring. Here, we demonstrate that inhibition of RA signaling using a high affinity pan-RA receptor antagonist administered to pregnant mice at E7.5 induces fully penetrant exencephaly and more severe spina bifida in Grhl3-null mice. Later administration, although prior to neural tube closure has no effect. Similarly, blockade of RA in the context of reduced expression of Grhl2, a related gene known to induce NTDs, has no effect. Taken together, these findings provide new insights into the complexities of the interplay between RA signaling and Grhl3-induced neurulation.


Subject(s)
Neural Tube Defects , Spinal Dysraphism , Pregnancy , Female , Mice , Animals , Transcription Factors/metabolism , Neurulation/genetics , Neural Tube/metabolism , Tretinoin/pharmacology , Tretinoin/metabolism , Neural Tube Defects/metabolism , Mice, Knockout , Spine/metabolism , DNA-Binding Proteins/metabolism
3.
Development ; 144(3): 430-440, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28143845

ABSTRACT

Transcription factor control of cell-specific downstream targets can be significantly altered when the controlling factor is mutated. We show that the semi-dominant neonatal anemia (Nan) mutation in the EKLF/KLF1 transcription factor leads to ectopic expression of proteins that are not normally expressed in the red blood cell, leading to systemic effects that exacerbate the intrinsic anemia in the adult and alter correct development in the early embryo. Even when expressed as a heterozygote, the Nan-EKLF protein accomplishes this by direct binding and aberrant activation of genes encoding secreted factors that exert a negative effect on erythropoiesis and iron use. Our data form the basis for a novel mechanism of physiological deficiency that is relevant to human dyserythropoietic anemia and likely other disease states.


Subject(s)
Anemia, Neonatal/genetics , Kruppel-Like Transcription Factors/genetics , Mutation , Amino Acid Substitution , Anemia, Neonatal/blood , Anemia, Neonatal/embryology , Animals , Animals, Newborn , Cytokines/blood , DNA/genetics , DNA/metabolism , Disease Models, Animal , Erythrocytes/metabolism , Erythropoiesis/genetics , Gene Expression Regulation, Developmental , Heterozygote , Humans , Kruppel-Like Transcription Factors/blood , Kruppel-Like Transcription Factors/deficiency , Mice , Mice, Knockout , Mice, Mutant Strains , Models, Biological , Muscle Proteins/blood , Mutant Proteins/blood , Mutant Proteins/genetics
4.
Blood ; 132(7): 735-749, 2018 08 16.
Article in English | MEDLINE | ID: mdl-29945953

ABSTRACT

Distinct subsets of resident tissue macrophages are important in hematopoietic stem cell niche homeostasis and erythropoiesis. We used a myeloid reporter gene (Csf1r-eGFP) to dissect the persistence of bone marrow and splenic macrophage subsets following lethal irradiation and autologous hematopoietic stem cell transplantation in a mouse model. Multiple recipient bone marrow and splenic macrophage subsets survived after autologous hematopoietic stem cell transplantation with organ-specific persistence kinetics. Short-term persistence (5 weeks) of recipient resident macrophages in spleen paralleled the duration of extramedullary hematopoiesis. In bone marrow, radiation-resistant recipient CD169+ resident macrophages and erythroid-island macrophages self-repopulated long-term after transplantation via autonomous cell division. Posttransplant peak expansion of recipient CD169+ resident macrophage number in bone marrow aligned with the persistent engraftment of phenotypic long-term reconstituting hematopoietic stem cells within bone marrow. Selective depletion of recipient CD169+ macrophages significantly compromised the engraftment of phenotypic long-term reconstituting hematopoietic stem cells and consequently impaired hematopoietic reconstitution. Recipient bone marrow resident macrophages are essential for optimal hematopoietic stem cell transplantation outcomes and could be an important consideration in the development of pretransplant conditioning therapies and/or chemoresistance approaches.


Subject(s)
Bone Marrow/metabolism , Graft Survival , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Macrophages/metabolism , Radiation Injuries, Experimental/metabolism , Animals , Autografts , Bone Marrow/pathology , Cell Survival , Hematopoietic Stem Cells/pathology , Macrophages/pathology , Mice , Mice, Transgenic , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/therapy
5.
BMC Genomics ; 20(1): 417, 2019 May 24.
Article in English | MEDLINE | ID: mdl-31126231

ABSTRACT

BACKGROUND: Mutations in the transcription factor, KLF1, are common within certain populations of the world. Heterozygous missense mutations in KLF1 mostly lead to benign phenotypes, but a heterozygous mutation in a DNA-binding residue (E325K in human) results in severe Congenital Dyserythropoietic Anemia type IV (CDA IV); i.e. an autosomal-dominant disorder characterized by neonatal hemolysis. RESULTS: To investigate the biochemical and genetic mechanism of CDA IV, we generated murine erythroid cell lines that harbor tamoxifen-inducible (ER™) versions of wild type and mutant KLF1 on a Klf1-/- genetic background. Nuclear translocation of wild type KLF1 results in terminal erythroid differentiation, whereas mutant KLF1 results in hemolysis without differentiation. The E to K variant binds poorly to the canonical 9 bp recognition motif (NGG-GYG-KGG) genome-wide but binds at high affinity to a corrupted motif (NGG-GRG-KGG). We confirmed altered DNA-binding specificity by quantitative in vitro binding assays of recombinant zinc-finger domains. Our results are consistent with previously reported structural data of KLF-DNA interactions. We employed 4sU-RNA-seq to show that a corrupted transcriptome is a direct consequence of aberrant DNA binding. CONCLUSIONS: Since all KLF/SP family proteins bind DNA in an identical fashion, these results are likely to be generally applicable to mutations in all family members. Importantly, they explain how certain mutations in the DNA-binding domain of transcription factors can generate neomorphic functions that result in autosomal dominant disease.


Subject(s)
Anemia, Dyserythropoietic, Congenital/genetics , DNA/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Point Mutation , Animals , Cell Line , DNA/chemistry , Gene Expression Regulation , Mice , Nucleotide Motifs , Protein Binding , Transcription, Genetic
7.
Intern Med J ; 49(8): 948-954, 2019 08.
Article in English | MEDLINE | ID: mdl-30411442

ABSTRACT

The classical myeloproliferative neoplasms (MPN) are uncommon clonal haemopoietic malignancies characterised by excessive production of mature blood cells. Clinically, they are associated with thrombosis, haemorrhage, varying degrees of constitutional disturbance and a risk of progression to myelofibrosis or acute myeloid leukaemia. Many of the disease manifestations may be ameliorated by treatment with interferon-α (IFN), but its use in Australian MPN patients has been limited due to the inconvenience of frequent injections and side-effects. The pegylated form of IFN is a long-acting preparation, which is better tolerated, and its Pharmaceutical Benefits Scheme listing is likely to lead to increased usage. We review the literature on risks and benefits of IFN treatment for MPN, suggest criteria for patient selection in each of these diseases and discuss strategies to manage the side-effects of pegylated IFN.


Subject(s)
Hematologic Neoplasms/drug therapy , Interferon-alpha/therapeutic use , Myeloproliferative Disorders/drug therapy , Australia , Disease Progression , Female , Humans , Interferon-alpha/adverse effects , Polyethylene Glycols , Pregnancy , Treatment Outcome
8.
Nucleic Acids Res ; 45(11): 6572-6588, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28541545

ABSTRACT

Krüppel-like factors (KLFs) are a family of 17 transcription factors characterized by a conserved DNA-binding domain of three zinc fingers and a variable N-terminal domain responsible for recruiting cofactors. KLFs have diverse functions in stem cell biology, embryo patterning, and tissue homoeostasis. KLF1 and related family members function as transcriptional activators via recruitment of co-activators such as EP300, whereas KLF3 and related members act as transcriptional repressors via recruitment of C-terminal Binding Proteins. KLF1 directly activates the Klf3 gene via an erythroid-specific promoter. Herein, we show KLF1 and KLF3 bind common as well as unique sites within the erythroid cell genome by ChIP-seq. We show KLF3 can displace KLF1 from key erythroid gene promoters and enhancers in vivo. Using 4sU RNA labelling and RNA-seq, we show this competition results in reciprocal transcriptional outputs for >50 important genes. Furthermore, Klf3-/- mice displayed exaggerated recovery from anemic stress and persistent cell cycling consistent with a role for KLF3 in dampening KLF1-driven proliferation. We suggest this study provides a paradigm for how KLFs work in incoherent feed-forward loops or networks to fine-tune transcription and thereby control diverse biological processes such as cell proliferation.


Subject(s)
Enhancer Elements, Genetic , Kruppel-Like Transcription Factors/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Animals , Cell Line , Coculture Techniques , Erythroid Cells/metabolism , Erythropoiesis , Mice , Transcription, Genetic
9.
Nucleic Acids Res ; 45(3): 1130-1143, 2017 02 17.
Article in English | MEDLINE | ID: mdl-28180284

ABSTRACT

The rules of engagement between zinc finger transcription factors and DNA have been partly defined by in vitro DNA-binding and structural studies, but less is known about how these rules apply in vivo. Here, we demonstrate how a missense mutation in the second zinc finger of Krüppel-like factor-1 (KLF1) leads to degenerate DNA-binding specificity in vivo, resulting in ectopic transcription and anemia in the Nan mouse model. We employed ChIP-seq and 4sU-RNA-seq to identify aberrant DNA-binding events genome wide and ectopic transcriptional consequences of this binding. We confirmed novel sequence specificity of the mutant recombinant zinc finger domain by performing biophysical measurements of in vitro DNA-binding affinity. Together, these results shed new light on the mechanisms by which missense mutations in DNA-binding domains of transcription factors can lead to autosomal dominant diseases.


Subject(s)
DNA/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Transcriptome/genetics , Zinc Fingers/genetics , Animals , Cell Line , Cell Survival/genetics , Erythroid Cells/metabolism , Erythropoiesis/genetics , Humans , Kruppel-Like Transcription Factors/chemistry , Mice , Models, Genetic , Models, Molecular , Mutant Proteins/chemistry , Mutation, Missense , Protein Binding
10.
Transfusion ; 58(10): 2414-2420, 2018 10.
Article in English | MEDLINE | ID: mdl-30222867

ABSTRACT

INTRODUCTION: KLF1 is an essential transcriptional activator that drives erythropoiesis. KLF1 variants can result in the Inhibitor of Lutheran, or In(Lu), phenotype where red blood cells (RBCs) have reduced BCAM (LU) and CD44 (IN). Other RBC surface molecules also have changed expression; however, there is controversy in the literature regarding which are truly impacted. We aimed to investigate KLF1 variants in the Australian population. STUDY DESIGN AND METHODS: In(Lu) samples were sourced through screening and through the RBC reference laboratory. Blood donor samples (8036) were screened to identify weakened/absent Lub antigen. Samples were genotyped by massively parallel sequencing, while surface carbohydrates and blood group molecules were assessed by flow cytometry. Hemoglobin (Hb) types were analyzed by high-performance liquid chromatography. RESULTS: Four of 8036 donors were identified to be In(Lu), and two previously identified In(Lu) samples were provided from the RBC reference laboratory. Five different KLF1 variants were identified; two were novel: c.954G>C/p.Trp318Cys and c.421C>T/p.Arg141*. BCAM and CD44 were reduced in all samples, consistent with previous reports. As a group, In(Lu) RBCs had reduced CD35 (KN), ICAM4 (LW), and CD147 (OK), and demonstrated increased binding of lectins ECA and SNAI. One In(Lu) sample had elevated HbF and another elevated HbA2. CONCLUSION: Different KLF1 variants may potentially produce variable phenotypes. A framework for investigating KLF1 variants and their phenotypic impact has been provided. In the future, given available international databases, further testing algorithms (as advocated here) will allow for correlation of phenotype with genotype and therefore accurately document this variability between KLF1 variants.


Subject(s)
Blood Group Antigens/blood , Erythrocytes/immunology , Genetic Variation , Kruppel-Like Transcription Factors/genetics , Lutheran Blood-Group System/chemistry , Australia , Chromatography, High Pressure Liquid , Flow Cytometry , Genetic Association Studies , Humans , Phenotype
11.
Blood ; 125(15): 2405-17, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25724378

ABSTRACT

We describe a case of severe neonatal anemia with kernicterus caused by compound heterozygosity for null mutations in KLF1, each inherited from asymptomatic parents. One of the mutations is novel. This is the first described case of a KLF1-null human. The phenotype of severe nonspherocytic hemolytic anemia, jaundice, hepatosplenomegaly, and marked erythroblastosis is more severe than that present in congenital dyserythropoietic anemia type IV as a result of dominant mutations in the second zinc-finger of KLF1. There was a very high level of HbF expression into childhood (>70%), consistent with a key role for KLF1 in human hemoglobin switching. We performed RNA-seq on circulating erythroblasts and found that human KLF1 acts like mouse Klf1 to coordinate expression of many genes required to build a red cell including those encoding globins, cytoskeletal components, AHSP, heme synthesis enzymes, cell-cycle regulators, and blood group antigens. We identify novel KLF1 target genes including KIF23 and KIF11 which are required for proper cytokinesis. We also identify new roles for KLF1 in autophagy, global transcriptional control, and RNA splicing. We suggest loss of KLF1 should be considered in otherwise unexplained cases of severe neonatal NSHA or hydrops fetalis.


Subject(s)
Anemia, Neonatal/genetics , Anemia, Neonatal/pathology , Gene Deletion , Hydrops Fetalis/genetics , Hydrops Fetalis/pathology , Kruppel-Like Transcription Factors/genetics , Transcriptome , Anemia, Neonatal/blood , Anemia, Neonatal/complications , Autophagy , Erythroblasts/metabolism , Erythroblasts/pathology , Erythropoiesis , Female , Gene Expression Regulation , Humans , Hydrops Fetalis/blood , Infant, Newborn , Kruppel-Like Transcription Factors/metabolism , Male
12.
Intern Med J ; 47(3): 262-268, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28260257

ABSTRACT

Ruxolitinib is a dual janus kinase 1 (JAK1)/JAK2 inhibitor used to treat splenomegaly and symptoms associated with myelofibrosis (MF). Current therapeutic options for symptomatic MF include supportive care, myelosuppressive therapy (such as hydroxycarbamide) and janus kinase (JAK) inhibitors (in particular ruxolitinib). Allogeneic stem cell transplantation remains the only potentially curative treatment for MF, and younger transplant-eligible patients should still be considered for allogeneic stem cell transplantation; however, this is applicable only to a small proportion of patients. There is now increasing and extensive experience of the efficacy and safety of ruxolitinib in MF, both in clinical trials and in 'real-world' practice. The drug has been shown to be of benefit in intermediate-1 risk patients with symptomatic splenomegaly or other MF-related symptoms, and higher risk disease. Optimal use of the drug is required to maximise clinical benefit, requiring an understanding of the balance between dose-dependent responses and dose-limiting toxicities. There is also increasing experience in the use of ruxolitinib in the pre-transplantation setting. This paper aims to utilise several 'real-life' cases to illustrate several strategies that may help to optimise clinical practice.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Primary Myelofibrosis/drug therapy , Pyrazoles/therapeutic use , Splenomegaly/drug therapy , Adult , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged , Nitriles , Practice Guidelines as Topic , Primary Myelofibrosis/physiopathology , Pyrimidines , Splenomegaly/etiology , Transplantation, Autologous , Treatment Outcome
13.
Growth Factors ; 34(5-6): 210-216, 2016 12.
Article in English | MEDLINE | ID: mdl-28209092

ABSTRACT

Fibroblast growth factor-1 (FGF-1) promotes differentiation of human preadipocytes into mature adipocytes via modulation of a BMP and Activin Membrane-Bound Inhibitor (BAMBI)/Peroxisome proliferator-activated receptor (PPARγ)-dependent network. Here, we combined transcriptomic and functional investigations to identify novel downstream effectors aligned with complementary analyses of gene expression in human adipose tissue to explore relationships with insulin sensitivity. RNA-Seq and qRT-PCR analysis revealed significant down-regulation of carboxypeptidase A4 (CPA4) following FGF-1 treatment or induction of differentiation of human preadipocytes in a BAMBI/PPARγ-independent manner. siRNA-mediated knockdown of CPA4 resulted in enhanced differentiation of human preadipocytes. Furthermore, expression of CPA4 in subcutaneous adipose tissue correlated negatively with indices of local and systemic (liver and muscle) insulin sensitivity. These results identify CPA4 as a negative regulator of adipogenesis that is down-regulated by FGF-1 and a putative deleterious modulator of local and systemic insulin sensitivity. Further investigations are required to define the molecular mechanism(s) involved and potential therapeutic opportunities.


Subject(s)
Adipocytes/metabolism , Adipogenesis , Carboxypeptidases A/metabolism , Fibroblast Growth Factor 1/pharmacology , Insulin Resistance , Adipocytes/cytology , Adipocytes/drug effects , Adult , Carboxypeptidases A/genetics , Cells, Cultured , Down-Regulation , Humans , Liver/metabolism , Male , Membrane Proteins/metabolism , Middle Aged , Muscle, Skeletal/metabolism , PPAR gamma/metabolism
14.
EMBO J ; 31(15): 3309-22, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22751148

ABSTRACT

F-BAR proteins are multivalent adaptors that link plasma membrane and cytoskeleton and coordinate cellular processes such as membrane protrusion and migration. Yet, little is known about the function of F-BAR proteins in vivo. Here we report, that the F-BAR protein NOSTRIN is necessary for proper vascular development in zebrafish and postnatal retinal angiogenesis in mice. The loss of NOSTRIN impacts on the migration of endothelial tip cells and leads to a reduction of tip cell filopodia number and length. NOSTRIN forms a complex with the GTPase Rac1 and its exchange factor Sos1 and overexpression of NOSTRIN in cells induces Rac1 activation. Furthermore, NOSTRIN is required for fibroblast growth factor 2 dependent activation of Rac1 in primary endothelial cells and the angiogenic response to fibroblast growth factor 2 in the in vivo matrigel plug assay. We propose a novel regulatory circuit, in which NOSTRIN assembles a signalling complex containing FGFR1, Rac1 and Sos1 thereby facilitating the activation of Rac1 in endothelial cells during developmental angiogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Blood Vessels/embryology , DNA-Binding Proteins/physiology , Fibroblast Growth Factors/metabolism , Neovascularization, Physiologic/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Animals, Genetically Modified , Animals, Newborn , Blood Vessels/growth & development , Blood Vessels/physiology , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Embryo, Mammalian , Embryo, Nonmammalian , Fibroblast Growth Factors/physiology , Mice , Mice, Knockout , Models, Biological , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Zebrafish/embryology , Zebrafish/genetics
15.
Blood ; 123(17): 2682-90, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24596419

ABSTRACT

The MYB oncogene is widely expressed in acute leukemias and is important for the continued proliferation of leukemia cells, suggesting that MYB may be a therapeutic target in these diseases. However, realization of this potential requires a significant therapeutic window for MYB inhibition, given its essential role in normal hematopoiesis, and an approach for developing an effective therapeutic. We previously showed that the interaction of c-Myb with the coactivator CBP/p300 is essential for its transforming activity. Here, by using cells from Booreana mice which carry a mutant allele of c-Myb, we show that this interaction is essential for in vitro transformation by the myeloid leukemia oncogenes AML1-ETO, AML1-ETO9a, MLL-ENL, and MLL-AF9. We further show that unlike cells from wild-type mice, Booreana cells transduced with AML1-ETO9a or MLL-AF9 retroviruses fail to generate leukemia upon transplantation into irradiated recipients. Finally, we have begun to explore the molecular mechanisms underlying these observations by gene expression profiling. This identified several genes previously implicated in myeloid leukemogenesis and HSC function as being regulated in a c-Myb-p300-dependent manner. These data highlight the importance of the c-Myb-p300 interaction in myeloid leukemogenesis and suggest disruption of this interaction as a potential therapeutic strategy for acute myeloid leukemia.


Subject(s)
Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/metabolism , Proto-Oncogene Proteins c-myb/metabolism , p300-CBP Transcription Factors/metabolism , Alleles , Animals , Cell Transformation, Neoplastic , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Mice , Mice, Mutant Strains , Mutation , Oncogene Proteins, Fusion/metabolism , Oncogenes , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism
17.
Genomics ; 105(2): 116-22, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25451176

ABSTRACT

Position-effect variegation of transgene expression is sensitive to the chromatin state. We previously reported a forward genetic screen in mice carrying a variegated α-globin GFP transgene to find novel genes encoding epigenetic regulators. We named the phenovariant strains "Mommes" for modifiers of murine metastable epialleles. Here we report positional cloning of mutations in two Momme strains which result in suppression of variegation. Both strains harbour point mutations in the erythroid transcription factor, Klf1. One (D11) generates a stop codon in the zinc finger domain and a homozygous null phenotype. The other (D45) generates an amino acid transversion (H350R) within a conserved linker between zinc fingers two and three. Homozygous MommeD45 mice have chronic microcytic anaemia which models the phenotype in a recently described family. This is the first genetic evidence that the linkers between the zinc fingers of transcription factors have a function beyond that of a simple spacer.


Subject(s)
Chromosomal Position Effects , Kruppel-Like Transcription Factors/genetics , Mutation , alpha-Globins/genetics , Anemia/genetics , Animals , Genetic Testing/methods , Mice , Mice, Transgenic/embryology , Mice, Transgenic/genetics , Splenomegaly/genetics , Zinc Fingers/genetics
18.
Curr Opin Hematol ; 22(3): 212-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25693142

ABSTRACT

PURPOSE OF REVIEW: The nature and function of macrophages at the center of erythroblastic islands is not fully understood. This review discusses novel findings on the phenotypic and molecular characterization of erythroblastic island macrophages, and their role in regulating normal and pathological erythropoiesis. RECENT FINDINGS: The phenotype to prospectively isolate erythroblastic island macrophages from mouse bone marrow has been identified. In-vivo depletion of erythroblastic island macrophages causes blockade of erythroblast maturation and delays erythropoietic recovery following chemical insults. The cytokine granulocyte colony-stimulating factor arrests medullary erythropoiesis by depleting erythroblastic island macrophages from the bone marrow. In-vivo ablation of macrophages improves anemia associated with ß-thalassemia and reduces red blood cell counts in the mouse model of polycythemia vera. The role of cell adhesion molecules regulating interactions between erythroblastic island macrophages and erythroblasts has been clarified, and mechanisms of pyrenocyte engulfment by erythroblastic island macrophages have been demonstrated to involve Mer tyrosine kinase receptor. SUMMARY: Prospective isolation of mouse erythroblastic island macrophages together with new genetic mouse models to specifically target erythroblastic island macrophages will enable molecular studies to better define their role in controlling erythroblast maturation. These studies have revealed the key role of erythroblastic island macrophages in regulating normal erythropoiesis and could be interesting targets to treat ß-thalassemia or polycythemia vera.


Subject(s)
Erythropoiesis/physiology , Macrophages/physiology , Anemia , Animals , Blood Cell Count , Erythroblasts/physiology , Humans , Mice , Prospective Studies
19.
Genome Res ; 22(12): 2385-98, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22835905

ABSTRACT

KLF1 (formerly known as EKLF) regulates the development of erythroid cells from bi-potent progenitor cells via the transcriptional activation of a diverse set of genes. Mice lacking Klf1 die in utero prior to E15 from severe anemia due to the inadequate expression of genes controlling hemoglobin production, cell membrane and cytoskeletal integrity, and the cell cycle. We have recently described the full repertoire of KLF1 binding sites in vivo by performing KLF1 ChIP-seq in primary erythroid tissue (E14.5 fetal liver). Here we describe the KLF1-dependent erythroid transcriptome by comparing mRNA-seq from Klf1(+/+) and Klf1(-/-) erythroid tissue. This has revealed novel target genes not previously obtainable by traditional microarray technology, and provided novel insights into the function of KLF1 as a transcriptional activator. We define a cis-regulatory module bound by KLF1, GATA1, TAL1, and EP300 that coordinates a core set of erythroid genes. We also describe a novel set of erythroid-specific promoters that drive high-level expression of otherwise ubiquitously expressed genes in erythroid cells. Our study has identified two novel lncRNAs that are dynamically expressed during erythroid differentiation, and discovered a role for KLF1 in directing apoptotic gene expression to drive the terminal stages of erythroid maturation.


Subject(s)
Erythropoiesis/genetics , Gene Expression Regulation, Developmental , Kruppel-Like Transcription Factors/genetics , RNA, Messenger/genetics , Transcriptome , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Differentiation , Chromosome Mapping , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Expression Profiling , In Situ Nick-End Labeling , Kruppel-Like Transcription Factors/metabolism , Liver/metabolism , Mice , Mice, Inbred BALB C , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Sequence Analysis, RNA/methods , T-Cell Acute Lymphocytic Leukemia Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL