Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
2.
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.

3.
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
4.
Nat Commun ; 13(1): 5582, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36151095

ABSTRACT

Stably silenced genes that display a high level of CpG dinucleotide methylation are refractory to the current generation of dCas9-based activation systems. To counter this, we create an improved activation system by coupling the catalytic domain of DNA demethylating enzyme TET1 with transcriptional activators (TETact). We show that TETact demethylation-coupled activation is able to induce transcription of suppressed genes, both individually and simultaneously in cells, and has utility across a number of cell types. Furthermore, we show that TETact can effectively reactivate embryonic haemoglobin genes in non-erythroid cells. We anticipate that TETact will expand the existing CRISPR toolbox and be valuable for functional studies, genetic screens and potential therapeutics.


Subject(s)
CRISPR-Cas Systems , DNA Methylation , Clustered Regularly Interspaced Short Palindromic Repeats , Epigenesis, Genetic , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , Transcriptional Activation
5.
Cancer Discov ; 12(6): 1560-1579, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35311997

ABSTRACT

Pharmacologic inhibition of epigenetic enzymes can have therapeutic benefit against hematologic malignancies. In addition to affecting tumor cell growth and proliferation, these epigenetic agents may induce antitumor immunity. Here, we discovered a novel immunoregulatory mechanism through inhibition of histone deacetylases (HDAC). In models of acute myeloid leukemia (AML), leukemia cell differentiation and therapeutic benefit mediated by the HDAC inhibitor (HDACi) panobinostat required activation of the type I interferon (IFN) pathway. Plasmacytoid dendritic cells (pDC) produced type I IFN after panobinostat treatment, through transcriptional activation of IFN genes concomitant with increased H3K27 acetylation at these loci. Depletion of pDCs abrogated panobinostat-mediated induction of type I IFN signaling in leukemia cells and impaired therapeutic efficacy, whereas combined treatment with panobinostat and IFNα improved outcomes in preclinical models. These discoveries offer a new therapeutic approach for AML and demonstrate that epigenetic rewiring of pDCs enhances antitumor immunity, opening the possibility of exploiting this approach for immunotherapies. SIGNIFICANCE: We demonstrate that HDACis induce terminal differentiation of AML through epigenetic remodeling of pDCs, resulting in production of type I IFN that is important for the therapeutic effects of HDACis. The study demonstrates the important functional interplay between the immune system and leukemias in response to HDAC inhibition. This article is highlighted in the In This Issue feature, p. 1397.


Subject(s)
Leukemia, Myeloid, Acute , Cell Differentiation , Dendritic Cells , Epigenesis, Genetic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/genetics , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Panobinostat/pharmacology
6.
Nat Commun ; 12(1): 6546, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34764270

ABSTRACT

Acute myeloid leukemia (AML) is a malignancy of immature progenitor cells. AML differentiation therapies trigger leukemia maturation and can induce remission, but relapse is prevalent and its cellular origin is unclear. Here we describe high resolution analysis of differentiation therapy response and relapse in a mouse AML model. Triggering leukemia differentiation in this model invariably produces two phenotypically distinct mature myeloid lineages in vivo. Leukemia-derived neutrophils dominate the initial wave of leukemia differentiation but clear rapidly and do not contribute to residual disease. In contrast, a therapy-induced population of mature AML-derived eosinophil-like cells persists during remission, often in extramedullary organs. Using genetic approaches we show that restricting therapy-induced leukemia maturation to the short-lived neutrophil lineage markedly reduces relapse rates and can yield cure. These results indicate that relapse can originate from therapy-resistant mature AML cells, and suggest differentiation therapy combined with targeted eradication of mature leukemia-derived lineages may improve disease outcome.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Neoplasm, Residual/metabolism , Cell Differentiation , Humans , Leukemia, Myeloid, Acute/genetics , Neoplasm, Residual/genetics
7.
Cell Rep ; 37(8): 110058, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34818538

ABSTRACT

Mouse hematopoietic tissues contain abundant tissue-resident macrophages that support immunity, hematopoiesis, and bone homeostasis. A systematic strategy to characterize macrophage subsets in mouse bone marrow (BM), spleen, and lymph node unexpectedly reveals that macrophage surface marker staining emanates from membrane-bound subcellular remnants associated with unrelated cells. Intact macrophages are not present within these cell preparations. The macrophage remnant binding profile reflects interactions between macrophages and other cell types in vivo. Depletion of CD169+ macrophages in vivo eliminates F4/80+ remnant attachment. Remnant-restricted macrophage-specific membrane markers, cytoplasmic fluorescent reporters, and mRNA are all detected in non-macrophage cells including isolated stem and progenitor cells. Analysis of RNA sequencing (RNA-seq) data, including publicly available datasets, indicates that macrophage fragmentation is a general phenomenon that confounds bulk and single-cell analysis of disaggregated hematopoietic tissues. Hematopoietic tissue macrophage fragmentation undermines the accuracy of macrophage ex vivo molecular profiling and creates opportunity for misattribution of macrophage-expressed genes to non-macrophage cells.


Subject(s)
Cell Separation/methods , Macrophages/cytology , Single-Cell Analysis/methods , Animals , Bone Marrow/metabolism , Hematopoiesis , Hematopoietic Stem Cells/cytology , Homeostasis , Mice
8.
Pathology ; 53(3): 339-348, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33674147

ABSTRACT

Myeloproliferative neoplasms are characterised by somatic mutations in pathways that regulate cell proliferation, epigenetic modifications, RNA splicing or DNA repair. Assessment of the mutational profile assists diagnosis and classification, but also aids assessment of prognosis, and may guide the use of emerging targeted therapies. The most practical way to provide information on numerous genetic variants is by using massively parallel sequencing, commonly in the form of disease specific next generation sequencing (NGS) panels. This review summarises the diagnostic and prognostic value of somatic mutation testing in Philadelphia-negative myeloproliferative neoplasms: polycythaemia vera, essential thrombocythaemia, primary myelofibrosis, chronic neutrophilic leukaemia, systemic mastocytosis, and chronic eosinophilic leukaemia. NGS panel testing is increasing in routine practice and promises to improve the accuracy and efficiency of pathological diagnosis and prognosis.


Subject(s)
Hypereosinophilic Syndrome/diagnosis , Leukemia/diagnosis , Mastocytosis, Systemic/diagnosis , Myeloproliferative Disorders/diagnosis , High-Throughput Nucleotide Sequencing , Humans , Hypereosinophilic Syndrome/genetics , Leukemia/genetics , Leukemia, Neutrophilic, Chronic/diagnosis , Leukemia, Neutrophilic, Chronic/genetics , Mastocytosis, Systemic/genetics , Mutation , Myeloproliferative Disorders/genetics , Polycythemia Vera/diagnosis , Polycythemia Vera/genetics , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/genetics , Prognosis , Sequence Analysis, DNA , Thrombocythemia, Essential/diagnosis , Thrombocythemia, Essential/genetics
11.
Nat Commun ; 11(1): 2042, 2020 04 27.
Article in English | MEDLINE | ID: mdl-32341362

ABSTRACT

The endothelial cell adhesion molecule E-selectin is a key component of the bone marrow hematopoietic stem cell (HSC) vascular niche regulating balance between HSC self-renewal and commitment. We now report in contrast, E-selectin directly triggers signaling pathways that promote malignant cell survival and regeneration. Using acute myeloid leukemia (AML) mouse models, we show AML blasts release inflammatory mediators that upregulate endothelial niche E-selectin expression. Alterations in cell-surface glycosylation associated with oncogenesis enhances AML blast binding to E-selectin and enable promotion of pro-survival signaling through AKT/NF-κB pathways. In vivo AML blasts with highest E-selectin binding potential are 12-fold more likely to survive chemotherapy and main contributors to disease relapse. Absence (in Sele-/- hosts) or therapeutic blockade of E-selectin using small molecule mimetic GMI-1271/Uproleselan effectively inhibits this niche-mediated pro-survival signaling, dampens AML blast regeneration, and strongly synergizes with chemotherapy, doubling the duration of mouse survival over chemotherapy alone, whilst protecting endogenous HSC.


Subject(s)
Drug Resistance, Neoplasm , E-Selectin/antagonists & inhibitors , E-Selectin/metabolism , Leukemia, Myeloid, Acute/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Bone Marrow , Coculture Techniques , Disease Models, Animal , Female , Glycolipids/therapeutic use , Glycosylation , Hematopoietic Stem Cells/cytology , Humans , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
12.
Sci Rep ; 9(1): 7851, 2019 May 21.
Article in English | MEDLINE | ID: mdl-31110193

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

13.
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
14.
Transfus Med Rev ; 33(2): 111-117, 2019 04.
Article in English | MEDLINE | ID: mdl-31023581

ABSTRACT

Erythroid-specific Krüppel-like factor 1, or KLF1, is an integral transcriptional activator for erythropoiesis. Genetic variants within KLF1 can result in a range of erythropoietic clinical phenotypes from benign to significant. The In(Lu) phenotype refers to changes in the quantitative expression of blood group-associated red cell surface molecules due to KLF1 variants which are otherwise clinically benign. These clinically benign KLF1 variants are associated with a reduced expression of 1 or more red cell membrane proteins/carbohydrates that carry blood group antigens for the LU (Lutheran), IN (Indian), P1PK, LW (Landsteiner-Wiener), KN (Knops), OK, RAPH, and I blood group systems. This is of significance during routine serologic blood typing when expression falls below the test sensitivity and therefore impacts on the ability to accurately detect the presence of affected blood group antigens. This is of clinical importance because the transfusion requirements for individuals with the In(Lu) phenotype differ from those of individuals that have a true Lunull phenotype. With this review, we summarize the current body of knowledge with regard to the In(Lu) phenotype and associated KLF1 variants. Our review also highlights discordant reports and provides insights for future research and management strategies. Serological heterogeneity in blood group expression of In(Lu) individuals has been shown, but studies are limited by the low prevalence of the phenotype and therefore the small numbers of samples. They are further limited by availability and inconsistent application of serological reagents and varying test algorithms. With the advent of genome sequence-based testing, an increasing list of In(Lu)-associated KLF1 variants is being revealed. The spectrum of effects on blood group expression due to these variants warrants further attention, and a consistent methodological approach of studies in larger cohorts is required. We propose that a recently reported testing framework of standardized serological studies, flow cytometry, and variant analysis be adopted; and that the international databases be curated to document KLF1 variability and the resultant In(Lu) red cell blood group expression. This will provide better classification of KLF1 variants affecting blood group expression and allow for phenotype prediction from genotype, accurate typing of In(Lu) individuals, and better transfusion management of related challenging transfusion scenarios.


Subject(s)
Blood Group Antigens/genetics , Kruppel-Like Transcription Factors/genetics , Lutheran Blood-Group System/genetics , Polymorphism, Genetic , Blood Transfusion , Genotype , Humans , Mutation , Phenotype , Sequence Analysis, DNA
15.
Blood Adv ; 3(3): 406-418, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30733301

ABSTRACT

In normoxia, hypoxia-inducible transcription factors (HIFs) are rapidly degraded within the cytoplasm as a consequence of their prolyl hydroxylation by oxygen-dependent prolyl hydroxylase domain (PHD) enzymes. We have previously shown that hematopoietic stem and progenitor cells (HSPCs) require HIF-1 for effective mobilization in response to granulocyte colony-stimulating factor (G-CSF) and CXCR4 antagonist AMD3100/plerixafor. Conversely, HIF PHD inhibitors that stabilize HIF-1 protein in vivo enhance HSPC mobilization in response to G-CSF or AMD3100 in a cell-intrinsic manner. We now show that extrinsic mechanisms involving vascular endothelial growth factor receptor-2 (VEGFR2), via bone marrow (BM) endothelial cells, are also at play. PTK787/vatalanib, a tyrosine kinase inhibitor selective for VEGFR1 and VEGFR2, and neutralizing anti-VEGFR2 monoclonal antibody DC101 blocked enhancement of HSPC mobilization by FG-4497. VEGFR2 was absent on mesenchymal and hematopoietic cells and was detected only in Sca1+ endothelial cells in the BM. We propose that HIF PHD inhibitor FG-4497 enhances HSPC mobilization by stabilizing HIF-1α in HSPCs as previously demonstrated, as well as by activating VEGFR2 signaling in BM endothelial cells, which facilitates HSPC egress from the BM into the circulation.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Isoquinolines/pharmacology , Prolyl-Hydroxylase Inhibitors/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Phthalazines/pharmacology , Pyridines/pharmacology , Vascular Endothelial Growth Factor A/metabolism
16.
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
17.
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
18.
Sci Rep ; 8(1): 12793, 2018 08 24.
Article in English | MEDLINE | ID: mdl-30143664

ABSTRACT

Anemic Nan mice carry a mutation (E339D) in the second zinc finger of erythroid transcription factor KLF1. Nan-KLF1 fails to bind a subset of normal KLF1 targets and ectopically binds a large set of genes not normally engaged by KLF1, resulting in a corrupted fetal liver transcriptome. Here, we performed RNAseq using flow cytometric-sorted spleen erythroid precursors from adult Nan and WT littermates rendered anemic by phlebotomy to identify global transcriptome changes specific to the Nan Klf1 mutation as opposed to anemia generally. Mutant Nan-KLF1 leads to extensive and progressive transcriptome corruption in adult spleen erythroid precursors such that stress erythropoiesis is severely compromised. Terminal erythroid differentiation is defective in the bone marrow as well. Principle component analysis reveals two major patterns of differential gene expression predicting that defects in basic cellular processes including translation, cell cycle, and DNA repair could contribute to disordered erythropoiesis and anemia in Nan. Significant erythroid precursor stage specific changes were identified in some of these processes in Nan. Remarkably, however, despite expression changes in large numbers of associated genes, most basic cellular processes were intact in Nan indicating that developing red cells display significant physiological resiliency and establish new homeostatic set points in vivo.


Subject(s)
Aging/pathology , Anemia/genetics , Anemia/pathology , Cell Differentiation/genetics , Erythropoiesis/genetics , Kruppel-Like Transcription Factors/genetics , Mutation/genetics , Transcriptome/genetics , Animals , Base Sequence , Cell Cycle/genetics , DNA Damage , Erythroid Cells/metabolism , Female , Gene Expression Regulation, Developmental , Gene Ontology , Kruppel-Like Transcription Factors/metabolism , Liver/embryology , Liver/metabolism , Mice , Mice, Mutant Strains , Mitophagy/genetics , Molecular Sequence Annotation , Principal Component Analysis , Reactive Oxygen Species/metabolism , Spleen/embryology , Spleen/metabolism
19.
Sci Rep ; 8(1): 12457, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30127368

ABSTRACT

A direct interaction between the erythropoietin (EPOR) and the beta-common (ßc) receptors to form an Innate Repair Receptor (IRR) is controversial. On one hand, studies have shown a functional link between EPOR and ßc receptor in tissue protection while others have shown no involvement of the ßc receptor in tissue repair. To date there is no biophysical evidence to confirm a direct association of the two receptors either in vitro or in vivo. We investigated the existence of an interaction between the extracellular regions of EPOR and the ßc receptor in silico and in vitro (either in the presence or absence of EPO or EPO-derived peptide ARA290). Although a possible interaction between EPOR and ßc was suggested by our computational and genomic studies, our in vitro biophysical analysis demonstrates that the extracellular regions of the two receptors do not specifically associate. We also explored the involvement of the ßc receptor gene (Csf2rb) under anaemic stress conditions and found no requirement for the ßc receptor in mice. In light of these studies, we conclude that the extracellular regions of the EPOR and the ßc receptor do not directly interact and that the IRR is not involved in anaemic stress.

20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...