Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 40
1.
Exp Hematol ; 76: 38-48.e2, 2019 08.
Article En | MEDLINE | ID: mdl-31295506

A better understanding of the development and progression of acute myelogenous leukemia (AML) is necessary to improve patient outcome. Here we define roles for the transcription factor Oct1/Pou2f1 in AML and normal hematopoiesis. Inappropriate reactivation of the CDX2 gene is widely observed in leukemia patients and in leukemia mouse models. We show that Oct1 associates with the CDX2 promoter in both normal and AML primary patient samples, but recruits the histone demethylase Jmjd1a/Kdm3a to remove the repressive H3K9me2 mark only in malignant specimens. The CpG DNA immediately adjacent to the Oct1 binding site within the CDX2 promoter exhibits variable DNA methylation in healthy control blood and bone marrow samples, but complete demethylation in AML samples. In MLL-AF9-driven mouse models, partial loss of Oct1 protects from myeloid leukemia. Complete Oct1 loss completely suppresses leukemia but results in lethality from bone marrow failure. Loss of Oct1 in normal hematopoietic transplants results in superficially normal long-term reconstitution; however, animals become acutely sensitive to 5-fluorouracil, indicating that Oct1 is dispensable for normal hematopoiesis but protects blood progenitor cells against external chemotoxic stress. These findings elucidate a novel and important role for Oct1 in AML.


Leukemia, Myeloid, Acute/genetics , Neoplasm Proteins/physiology , Octamer Transcription Factor-1/physiology , Animals , Bone Marrow/pathology , Bone Marrow Failure Disorders/etiology , Bone Marrow Failure Disorders/genetics , CDX2 Transcription Factor/biosynthesis , CDX2 Transcription Factor/genetics , Cell Transformation, Neoplastic/genetics , CpG Islands , DNA Methylation , Disease Progression , Fluorouracil/toxicity , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/drug effects , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Leukemia, Experimental/genetics , Leukemia, Experimental/prevention & control , Leukemia, Myeloid, Acute/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice, Inbred C57BL , Octamer Transcription Factor-1/deficiency , Oncogene Proteins, Fusion/physiology , Promoter Regions, Genetic , Radiation Chimera
2.
Nature ; 549(7673): 476-481, 2017 09 28.
Article En | MEDLINE | ID: mdl-28825709

Stem-cell fate can be influenced by metabolite levels in culture, but it is not known whether physiological variations in metabolite levels in normal tissues regulate stem-cell function in vivo. Here we describe a metabolomics method for the analysis of rare cell populations isolated directly from tissues and use it to compare mouse haematopoietic stem cells (HSCs) to restricted haematopoietic progenitors. Each haematopoietic cell type had a distinct metabolic signature. Human and mouse HSCs had unusually high levels of ascorbate, which decreased with differentiation. Systemic ascorbate depletion in mice increased HSC frequency and function, in part by reducing the function of Tet2, a dioxygenase tumour suppressor. Ascorbate depletion cooperated with Flt3 internal tandem duplication (Flt3ITD) leukaemic mutations to accelerate leukaemogenesis, through cell-autonomous and possibly non-cell-autonomous mechanisms, in a manner that was reversed by dietary ascorbate. Ascorbate acted cell-autonomously to negatively regulate HSC function and myelopoiesis through Tet2-dependent and Tet2-independent mechanisms. Ascorbate therefore accumulates within HSCs to promote Tet activity in vivo, limiting HSC frequency and suppressing leukaemogenesis.


Ascorbic Acid/metabolism , Carcinogenesis/metabolism , Hematopoietic Stem Cells/cytology , Leukemia/pathology , Animals , Ascorbic Acid/analysis , Ascorbic Acid Deficiency/genetics , Ascorbic Acid Deficiency/metabolism , Carcinogenesis/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Female , Hematopoietic Stem Cells/metabolism , Humans , Leukemia/genetics , Male , Metabolomics , Mice , Myelopoiesis/genetics , Proto-Oncogene Proteins/metabolism , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
3.
Stem Cells ; 34(1): 67-82, 2016 Jan.
Article En | MEDLINE | ID: mdl-26439305

Splenomegaly is a major manifestation of primary myelofibrosis (PMF) contributing to clinical symptoms and hematologic abnormalities. The spleen from PMF patients contains increased numbers of hematopoietic stem cells (HSC) and megakaryocytes (MK). These MK express high levels of P-selectin (P-sel) that, by triggering neutrophil emperipolesis, may cause TGF-ß release and disease progression. This hypothesis was tested by deleting the P-sel gene in the myelofibrosis mouse model carrying the hypomorphic Gata1(low) mutation that induces megakaryocyte abnormalities that recapitulate those observed in PMF. P-sel(null) Gata1(low) mice survived splenectomy and lived 3 months longer than P-sel(WT) Gata1(low) littermates and expressed limited fibrosis and osteosclerosis in the marrow or splenomegaly. Furthermore, deletion of P-sel disrupted megakaryocyte/neutrophil interactions in spleen, reduced TGF-ß content, and corrected the HSC distribution that in Gata1(low) mice, as in PMF patients, is abnormally expanded in spleen. Conversely, pharmacological inhibition of TGF-ß reduced P-sel expression in MK and corrected HSC distribution. Spleens, but not marrow, of Gata1(low) mice contained numerous cKIT(pos) activated fibrocytes, probably of dendritic cell origin, whose membrane protrusions interacted with MK establishing niches hosting immature cKIT(pos) hematopoietic cells. These activated fibrocytes were not detected in spleens from P-sel(null) Gata1(low) or TGF-ß-inhibited Gata1(low) littermates and were observed in spleen, but not in marrow, from PMF patients. Therefore, in Gata1(low) mice, and possibly in PMF, abnormal P-sel expression in MK may mediate the pathological cell interactions that increase TGF-ß content in MK and favor establishment of a microenvironment that supports myelofibrosis-related HSC in spleen.


GATA1 Transcription Factor/metabolism , Hematopoiesis, Extramedullary , P-Selectin/metabolism , Primary Myelofibrosis/metabolism , Animals , Cell Differentiation , Disease Models, Animal , Emperipolesis , Female , Humans , Male , Megakaryocytes/pathology , Megakaryocytes/ultrastructure , Mice , Neutrophils/metabolism , Phenotype , Primary Myelofibrosis/pathology , Spleen/pathology , Spleen/ultrastructure , Transforming Growth Factor beta/metabolism
4.
Hum Antibodies ; 22(1-2): 21-9, 2013.
Article En | MEDLINE | ID: mdl-24284306

BACKGROUND: c3orf75 is a conserved open reading frame within the human genome and has recently been identified as the Elongator subunit, ELP6 [1]. The Elongator enzyme complex has diverse roles, including translational control, neuronal development, cell migration and tumorigenicity [2]. OBJECTIVE: To identify genes expressed early in human eosinophil development. METHODS: Eosinophilopoiesis was investigated by gene profiling of IL-5 stimulated CD34+ cells; ELP6 mRNA is upregulated. A monoclonal antibody was raised to the recombinant protein predicted by the open reading frame. RESULTS: ELP6 transcripts are upregulated in a human tissue culture model of eosinophil development during gene profiling experiments. Transcripts are expressed in most tissue types, as shown by reverse-transcriptase PCR. Western blot experiments show that human ELP6 is a 30 kDa protein expressed in the bone marrow, as well as in many other tissues. Flow cytometry experiments of human bone marrow mononuclear cells show that ELP6 is expressed intracellularly, in developing and mature human neutrophils, eosinophils and monocytes. CONCLUSIONS: ELP6 is expressed intracellularly in developing and mature granulocytes and monocytes but not in lymphocytes and erythrocytes.


Eosinophils/metabolism , Monocytes/metabolism , Neutrophils/metabolism , Proteins/genetics , RNA, Messenger/genetics , Antigens, CD34/genetics , Antigens, CD34/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Eosinophils/cytology , Eosinophils/drug effects , Fetal Blood/cytology , Fetal Blood/drug effects , Fetal Blood/metabolism , Gene Expression Profiling , Gene Expression Regulation , Histone Acetyltransferases , Humans , Immunophenotyping , Interleukin-5/pharmacology , Monocytes/cytology , Monocytes/drug effects , Neutrophils/cytology , Neutrophils/drug effects , Proteins/metabolism , RNA, Messenger/metabolism
5.
PLoS One ; 8(3): e57481, 2013.
Article En | MEDLINE | ID: mdl-23469197

How inflammatory responses are mechanistically modulated by nicotinic acetylcholine receptors (nAChR), especially by receptors composed of alpha7 (α7) subunits, is poorly defined. This includes a precise definition of cells that express α7 and how these impact on innate inflammatory responses. To this aim we used mice generated through homologous recombination that express an Ires-Cre-recombinase bi-cistronic extension of the endogenous α7 gene that when crossed with a reporter mouse expressing Rosa26-LoxP (yellow fluorescent protein (YFP)) marks in the offspring those cells of the α7 cell lineage (α7(lin+)). In the adult, on average 20-25 percent of the total CD45(+) myeloid and lymphoid cells of the bone marrow (BM), blood, spleen, lymph nodes, and Peyers patches are α7(lin+), although variability between litter mates in this value is observed. This hematopoietic α7(lin+) subpopulation is also found in Sca1(+)cKit(+) BM cells suggesting the α7 lineage is established early during hematopoiesis and the ratio remains stable in the individual thereafter as measured for at least 18 months. Both α7(lin+) and α7(lin-) BM cells can reconstitute the immune system of naïve irradiated recipient mice and the α7(lin+):α7(lin-) beginning ratio is stable in the recipient after reconstitution. Functionally the α7(lin+):α7(lin-) lineages differ in response to LPS challenge. Most notable is the response to LPS as demonstrated by an enhanced production of IL-12/23(p40) by the α7(lin+) cells. These studies demonstrate that α7(lin+) identifies a novel subpopulation of bone marrow cells that include hematopoietic progenitor cells that can re-populate an animal's inflammatory/immune system. These findings suggest that α7 exhibits a pleiotropic role in the hematopoietic system that includes both the direct modulation of pro-inflammatory cell composition and later in the adult the role of modulating pro-inflammatory responses that would impact upon an individual's lifelong response to inflammation and infection.


Gene Expression Regulation, Developmental/immunology , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Receptors, Nicotinic/immunology , Adoptive Transfer , Animals , Biomarkers/metabolism , Cell Lineage/drug effects , Cell Lineage/immunology , Crosses, Genetic , Female , Gene Expression Regulation, Developmental/drug effects , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Inflammation/genetics , Inflammation/immunology , Interleukin-12/biosynthesis , Interleukin-12/immunology , Interleukin-23/biosynthesis , Interleukin-23/immunology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Lipopolysaccharides/pharmacology , Lymphocytes/cytology , Lymphocytes/immunology , Male , Mice , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/immunology , Receptors, Nicotinic/genetics , Whole-Body Irradiation , alpha7 Nicotinic Acetylcholine Receptor
6.
Antioxid Redox Signal ; 19(17): 2054-67, 2013 Dec 10.
Article En | MEDLINE | ID: mdl-23249337

AIMS: Vitamin C (ascorbic acid) is thought to enhance immune function, but the mechanisms involved are obscure. We utilized an in vitro model of T-cell maturation to evaluate the role of ascorbic acid in lymphocyte development. RESULTS: Ascorbic acid was essential for the developmental progression of mouse bone marrow-derived progenitor cells to functional T-lymphocytes in vitro and also played a role in vivo. Ascorbate-mediated enhancement of T-cell development was lymphoid cell-intrinsic and independent of T-cell receptor (TCR) rearrangement. Analysis of TCR rearrangements demonstrated that ascorbic acid enhanced the selection of functional TCRαß after the stage of ß-selection. Genes encoding the coreceptor CD8 as well as the kinase ZAP70 were upregulated by ascorbic acid. Pharmacologic inhibition of methylation marks on DNA and histones enhanced ascorbate-mediated differentiation, suggesting an epigenetic mechanism of Cd8 gene regulation via active demethylation by ascorbate-dependent Fe(2+) and 2-oxoglutarate-dependent dioxygenases. INNOVATION: We speculate that one aspect of gene regulation mediated by ascorbate occurs at the level of chromatin demethylation, mediated by Jumonji C (JmjC) domain enzymes that are known to be reliant upon ascorbate as a cofactor. JmjC domain enzymes are also known to regulate transcription factor activity. These two mechanisms are likely to play key roles in the modulation of immune development and function by ascorbic acid. CONCLUSION: Our results provide strong experimental evidence supporting a role for ascorbic acid in T-cell maturation as well as insight into the mechanism of ascorbate-mediated enhancement of immune function.


Ascorbic Acid/pharmacology , Immunologic Factors/pharmacology , T-Lymphocytes/drug effects , Animals , Azepines/pharmacology , Cells, Cultured , Culture Media , Epigenesis, Genetic/drug effects , Gene Expression/drug effects , Gene Rearrangement, T-Lymphocyte/drug effects , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Methylation , Mice , Mice, Inbred C57BL , Phthalimides/pharmacology , Protein Processing, Post-Translational , Quinazolines/pharmacology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
7.
PLoS Genet ; 8(11): e1003048, 2012.
Article En | MEDLINE | ID: mdl-23144633

Defining master transcription factors governing somatic and cancer stem cell identity is an important goal. Here we show that the Oct4 paralog Oct1, a transcription factor implicated in stress responses, metabolic control, and poised transcription states, regulates normal and pathologic stem cell function. Oct1(HI) cells in the colon and small intestine co-express known stem cell markers. In primary malignant tissue, high Oct1 protein but not mRNA levels strongly correlate with the frequency of CD24(LO)CD44(HI) cancer-initiating cells. Reducing Oct1 expression via RNAi reduces the proportion of ALDH(HI) and dye efflux(HI) cells, and increasing Oct1 increases the proportion of ALDH(HI) cells. Normal ALDH(HI) cells harbor elevated Oct1 protein but not mRNA levels. Functionally, we show that Oct1 promotes tumor engraftment frequency and promotes hematopoietic stem cell engraftment potential in competitive and serial transplants. In addition to previously described Oct1 transcriptional targets, we identify four Oct1 targets associated with the stem cell phenotype. Cumulatively, the data indicate that Oct1 regulates normal and cancer stem cell function.


Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells , Organic Cation Transporter 1 , Stem Cells , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Biomarkers/metabolism , CD24 Antigen/metabolism , Colon/cytology , Colon/metabolism , HeLa Cells , Humans , Hyaluronan Receptors/metabolism , Intestine, Small/cytology , Intestine, Small/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Organic Cation Transporter 1/genetics , Organic Cation Transporter 1/metabolism , Phenotype , RNA, Messenger/metabolism , Stem Cells/cytology , Stem Cells/metabolism
8.
Eur J Immunol ; 42(4): 1038-43, 2012 Apr.
Article En | MEDLINE | ID: mdl-22531927

The altered expression of transcription factors in hematopoietic stem cells and their subsequent lineages can alter the development of lymphoid and myeloid lineages. The role of the transcriptional repressor Snai3 protein in the derivation of cells of the hemato-poietic system was investigated. Snai3 is expressed in terminal T-cell and myeloid lineages, therefore, we chose to determine if expressing Snai3 in the early stages of hematopoietic development would influence cell-lineage determination. Expression of Snai3 by retroviral transduction of hematopoietic stem cells using bone marrow chimera studies demonstrated a block in lymphoid-cell development and enhanced expansion of myeloid-lineage cells. Analysis of Snai3-expressing hematopoietic precursor cells showed normal numbers of immature cells, but a block in the development of cells committed to lymphoid lineages. These data indicate that the overexpression of Snai3 does alter bone marrow cell development and that the identification of genes whose expression is altered by the presence of Snai3 would aid in our understanding of these developmental pathways.


Gene Expression Regulation/immunology , Lymphocytes/immunology , Myeloid Cells/immunology , Repressor Proteins/immunology , Transcription Factors/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Gene Expression Regulation/genetics , Lymphocytes/metabolism , Mice , Mice, Transgenic , Myeloid Cells/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transduction, Genetic
9.
Blood ; 119(24): 5621-31, 2012 Jun 14.
Article En | MEDLINE | ID: mdl-22490804

To detect targeted antileukemia agents we have designed a novel, high-content in vivo screen using genetically engineered, T-cell reporting zebrafish. We exploited the developmental similarities between normal and malignant T lymphoblasts to screen a small molecule library for activity against immature T cells with a simple visual readout in zebrafish larvae. After screening 26 400 molecules, we identified Lenaldekar (LDK), a compound that eliminates immature T cells in developing zebrafish without affecting the cell cycle in other cell types. LDK is well tolerated in vertebrates and induces long-term remission in adult zebrafish with cMYC-induced T-cell acute lymphoblastic leukemia (T-ALL). LDK causes dephosphorylation of members of the PI3 kinase/AKT/mTOR pathway and delays sensitive cells in late mitosis. Among human cancers, LDK selectively affects survival of hematopoietic malignancy lines and primary leukemias, including therapy-refractory B-ALL and chronic myelogenous leukemia samples, and inhibits growth of human T-ALL xenografts. This work demonstrates the utility of our method using zebrafish for antineoplastic candidate drug identification and suggests a new approach for targeted leukemia therapy. Although our efforts focused on leukemia therapy, this screening approach has broad implications as it can be translated to other cancer types involving malignant degeneration of developmentally arrested cells.


Antineoplastic Agents/toxicity , Hydrazones/toxicity , Leukemia/pathology , Quinolines/toxicity , Zebrafish/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Blast Crisis/pathology , Cell Differentiation/drug effects , Disease Models, Animal , Disease Progression , Humans , Hydrazones/chemistry , Hydrazones/pharmacokinetics , Hydrazones/therapeutic use , Leukemia/drug therapy , Mice , Mitosis/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Quinolines/chemistry , Quinolines/pharmacokinetics , Quinolines/therapeutic use , Signal Transduction/drug effects , T-Lymphocytes/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
10.
J Immunol ; 188(3): 1381-93, 2012 Feb 01.
Article En | MEDLINE | ID: mdl-22180617

IL-10 is a nonredundant inflammatory modulator that suppresses arthritis development in Borrelia burgdorferi-infected mice. Infected C57BL/6 (B6) IL-10(-/-) mice were previously found to have a prolonged IFN-inducible response in joint tissue. Infection of B6 IL-10 reporter mice identified macrophages and CD4(+) T cells as the primary sources of IL-10 in the infected joint tissue, suggesting that early local production of IL-10 dampened the proarthritic IFN response. Treatment of B6 IL-10(-/-) mice with anti-IFN-γ reduced the increase in arthritis severity and suppressed IFN-inducible transcripts to wild-type levels, thereby linking dysregulation of IFN-γ to disease in the B6 IL-10(-/-) mouse. Arthritis in B6 IL-10(-/-) mice was associated with elevated numbers of NK cell, NKT cell, α/ß T cell, and macrophage infiltration of the infected joint. FACS lineage sorting revealed NK cells and CD4(+) T cells as sources of IFN-γ in the joint tissue of B6 IL-10(-/-) mice. These findings suggest the presence of a positive-feedback loop in the joint tissue of infected B6 IL-10(-/-) mice, in which production of inflammatory chemokines, infiltration of IFN-γ-producing cells, and additional production of inflammatory cytokines result in arthritis. This mechanism of arthritis is in contrast to that seen in C3H/He mice, in which arthritis development is linked to transient production of type I IFN and develops independently of IFN-γ. Due to the sustained IFN response driven by NK cells and T cells, we propose the B6 IL-10(-/-) mouse as a potential model to study the persistent arthritis observed in some human Lyme disease patients.


Cell Movement/immunology , Inflammation/immunology , Interferon-gamma/pharmacology , Interleukin-10/biosynthesis , Lyme Disease/immunology , Animals , Borrelia burgdorferi , Feedback, Physiological , Humans , Interferon-gamma/immunology , Interleukin-10/deficiency , Killer Cells, Natural/immunology , Mice , Mice, Knockout , T-Lymphocytes/immunology
11.
J Immunol ; 187(10): 5203-10, 2011 Nov 15.
Article En | MEDLINE | ID: mdl-21998453

The details of the bifurcation of the lymphoid and myeloid lineages following commitment by multipotent progenitor cells (MPP) remain a topic of controversy. We report that the surface glycoprotein CD62L can be characterized as a novel marker of this and other stages of early hematopoietic differentiation. Cell isolation and transplant studies demonstrated CD62L(neg/low) long-term hematopoietic stem cells and CD62L(high) MPP within the traditionally defined c-kit(pos)Lin(neg/low)Sca-1(pos) stem/progenitor cell population. Within the MPP population, previously defined as c-kit(pos)Lin(neg/low)Sca-1(pos)-Thy-1.1(neg)Flt3(pos), Sca-1 and CD62L resolved four populations and segregated Sca-1(high)CD62L(neg/low) MPP from Sca-1(high)CD62L(high) leukocyte-biased progenitors. Using a novel transplantation method that allows tracking of erythroid and platelet engraftment as an alternative to the classical method of in vitro colony formation, we characterized Sca-1(high)CD62L(neg/low) cells as MPP, based on transient engraftment of these lineages. These data establish CD62L as a useful tool in the study of early hematopoiesis and emphasize the power of trilineage-engraftment studies in establishing the lineage potential of MPP subsets.


Cell Differentiation/immunology , Hematopoietic Stem Cell Transplantation/methods , L-Selectin/immunology , Multipotent Stem Cells/immunology , Animals , Antigens, Ly/biosynthesis , Antigens, Ly/blood , Biomarkers/blood , Cell Lineage/immunology , L-Selectin/biosynthesis , Membrane Proteins/biosynthesis , Membrane Proteins/blood , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/metabolism , Proto-Oncogene Proteins c-kit/biosynthesis , fms-Like Tyrosine Kinase 3/biosynthesis
12.
Blood ; 117(20): 5494-502, 2011 May 19.
Article En | MEDLINE | ID: mdl-21310927

Mitoferrin1 is 1 of 2 homologous mitochondrial iron transporters and is required for mitochondrial iron delivery in developing erythroid cells. We show that total deletion of Mfrn1 in embryos leads to embryonic lethality. Selective deletion of Mfrn1 in adult hematopoietic tissues leads to severe anemia because of a deficit in erythroblast formation. Deletion of Mfrn1 in hepatocytes has no phenotype or biochemical effect under normal conditions. In the presence of increased porphyrin synthesis, however, deletion of Mfrn1 in hepatocytes results in a decreased ability to convert protoporphyrin IX into heme, leading to protoporphyria, cholestasis, and bridging cirrhosis. Our results show that the activity of mitoferrin1 is required to manage an increase in heme synthesis. The data also show that alterations in heme synthesis within hepatocytes can lead to protoporphyria and hepatotoxicity.


Anemia/etiology , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Protoporphyria, Erythropoietic/etiology , Anemia/genetics , Animals , Base Sequence , DNA Primers/genetics , Embryo Loss/genetics , Female , Gene Targeting , Heme/biosynthesis , Hepatocytes/metabolism , Iron/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Pregnancy , Protoporphyria, Erythropoietic/genetics , Protoporphyrins/metabolism
13.
Cell ; 141(5): 775-85, 2010 May 28.
Article En | MEDLINE | ID: mdl-20510925

Mouse Hoxb8 mutants show unexpected behavior manifested by compulsive grooming and hair removal, similar to behavior in humans with the obsessive-compulsive disorder spectrum disorder trichotillomania. As Hox gene disruption often has pleiotropic effects, the root cause of this behavioral deficit was unclear. Here we report that, in the brain, Hoxb8 cell lineage exclusively labels bone marrow-derived microglia. Furthermore, transplantation of wild-type bone marrow into Hoxb8 mutant mice rescues their pathological phenotype. It has been suggested that the grooming dysfunction results from a nociceptive defect, also exhibited by Hoxb8 mutant mice. However, bone marrow transplant experiments and cell type-specific disruption of Hoxb8 reveal that these two phenotypes are separable, with the grooming phenotype derived from the hematopoietic lineage and the sensory defect derived from the spinal cord cells. Immunological dysfunctions have been associated with neuropsychiatric disorders, but the causative relationships are unclear. In this mouse, a distinct compulsive behavioral disorder is associated with mutant microglia.


Grooming , Homeodomain Proteins/metabolism , Obsessive-Compulsive Disorder/genetics , Obsessive-Compulsive Disorder/physiopathology , Animals , B-Lymphocytes/metabolism , Behavior, Animal , Bone Marrow Transplantation , Brain/cytology , Brain/physiopathology , Homeodomain Proteins/genetics , Humans , Mice , Microglia/metabolism , Spinal Cord/metabolism , T-Lymphocytes/metabolism
14.
Blood Cells Mol Dis ; 44(1): 1-6, 2010 Jan 15.
Article En | MEDLINE | ID: mdl-19836979

Transgenic expression of a gain-of-function truncated mouse erythropoietin receptor gene (EpoR) leads to expansion of the HSC pool in response to human erythropoietin (Epo). We have re-examined this observation using a knock-in mouse model, wherein the mouse EpoR gene was replaced in its proper genetic locus by a single copy of either a wild-type human or a polycythemia-inducing truncated human EPOR gene. Bone marrow cells obtained from knock-in mice were transplanted together with competitor bone marrow cells in a model that allows tracking of erythroid, platelet, and leukocyte contributions by each genotype. Secondary transplants were also performed. Stem/progenitor cells were identified phenotypically and isolated for colony-forming assays to evaluate cytokine responsiveness by cells with the wild-type human or truncated human EPOR gene. Augmented Epo signaling increased erythroid repopulation post-transplant as expected, but had no effect on short-term or long-term leukocyte repopulation. However, the wild-type human EPOR knock-in mouse showed decreases in both erythroid and platelet repopulation compared to marrow cells from the mutant human EPOR knock-in mouse or normal B6 animals. These results provide evidence supporting a role for Epo signaling in megakaryopoiesis in vivo and suggest a role for Epo signaling early in hematopoietic development.


Erythroid Precursor Cells/cytology , Erythropoiesis , Megakaryocyte Progenitor Cells/cytology , Receptors, Erythropoietin/physiology , Signal Transduction/genetics , Thrombopoiesis , Animals , Bone Marrow Transplantation/methods , Cells, Cultured , Cytokines/pharmacology , Erythrocytes/metabolism , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoiesis/drug effects , Gene Knock-In Techniques , Genotype , Graft Survival/genetics , Humans , Megakaryocyte Progenitor Cells/drug effects , Megakaryocyte Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Protein Isoforms/analysis , Protein Isoforms/genetics , Receptors, Erythropoietin/agonists , Receptors, Erythropoietin/genetics , Time Factors , beta-Globins/analysis , beta-Globins/genetics
15.
Cell Transplant ; 18(8): 887-97, 2009.
Article En | MEDLINE | ID: mdl-19520051

Copper chelation has been shown to favor the expansion of human hematopoietic stem/progenitor cells in vitro. To further understand the effects of copper modulation on defined subsets of stem cells versus progenitor cells, we extended the studies in a mouse system. We isolated mouse hematopoietic stem cells (HSCs) or hematopoietic progenitor cells (HPCs) and cultured them with or without the copper chelator tetraethylenepentamine (TEPA) or CuCl(2). Cytokine-stimulated HPC cultures treated with TEPA for 7 days generated about two to three times more total and erythroid colony-forming cells (CFCs) compared to control cultures. In contrast, CuCl(2) treatment decreased the CFC numbers. Similar results were seen with HSC after 14, but not 7, days of culture. Transplant studies showed that HPCs cultured for 7 days in TEPA had about twofold higher short-term erythroid repopulation potential compared to control cultures, while CuCl(2) decreased the erythroid potential of cultured HPCs compared to control cultures. HSCs cultured with TEPA for 7 days did not exhibit significantly higher repopulation potential in either leukocyte or erythrocyte lineages compared to control cultures in short-term or long-term assays. Based on JC-1 staining, the mitochondrial membrane potential of HPCs cultured with TEPA was lower relative to control cultures. Our data suggest that decreasing the cellular copper content with TEPA results in preferential expansion or maintenance of HPC that are biased for erythroid differentiation in vivo, but does not enhance the maintenance of HSC activity in culture.


Cell Differentiation/drug effects , Copper/pharmacology , Hematopoietic Stem Cells/drug effects , Animals , Cell Culture Techniques , Cells, Cultured , Ethylenediamines/pharmacology , Hematopoietic Stem Cells/physiology , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic
16.
J Biomed Mater Res A ; 91(1): 209-20, 2009 Oct.
Article En | MEDLINE | ID: mdl-18814276

Asparaginase (ASNase) is an enzyme drug presently approved for the induction of remission in the treatment of patients with acute lymphoblastic leukemia (ALL). The cytotoxic effect of ASNase is derived from its ability to deplete asparagine, an essential amino acid required by certain types of leukemia cells for protein synthesis and survival. Despite its efficacy in enhancing disease remission rate and prolonging complete remission duration in ALL patients, ASNase therapy is nevertheless confounded by a number of serious toxic effects, particularly to organs associated with high protein production (e.g., liver, pancreas), due to the systemic depletion of asparagine. Presented herein is a modified version of our previously established ATTEMPTS protein delivery system that carries the potential to permit a tumor specific, intracellular delivery of ASNase, thereby allowing for a significant reduction of ASNase-induced systemic toxicity. In a previous paper, we already demonstrated the in vitro feasibility of this heparin/protamine-regulated, TAT-mediated system in delivering ASNase directly into ASNase-sensitive murine lymphoma cells. In this article, we further validated the in vivo applicability of this system in animals harboring ASNase-encapsulated L5178Y lymphoma cells. Preliminary results showed that animals inoculated with L5178Y cells containing TAT-ASNase exhibited an extended survival rate of approximately 13% over those harboring L5178Y cells without the encapsulation of ASNase. Furthermore, the TAT-ASNase-treated mice also displayed a significantly improved hematological and liver histological status than the control groups. These findings bring promise to the use of the modified ATTEMPTS delivery system in achieving enhanced ASNase therapy.


Antineoplastic Agents/administration & dosage , Asparaginase/administration & dosage , Drug Delivery Systems , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Amino Acid Sequence , Animals , Antineoplastic Agents/therapeutic use , Asparaginase/therapeutic use , Cell Line, Tumor , Cell Survival , Female , Gene Products, tat/chemistry , Hematologic Tests , Humans , Lymph Nodes/cytology , Mice , Mice, Inbred DBA , Molecular Sequence Data , Peptides/chemistry
17.
Curr Protoc Immunol ; Chapter 4: 4.6.1-4.6.9, 2008 May.
Article En | MEDLINE | ID: mdl-18491297

Transplantation of marrow between mouse strains congenic for CD45 after lethal irradiation establishes hematopoiesis driven by genetically marked cells in recipient animals. After several weeks, peripheral blood or primary and secondary lymphoid organs of transplant recipients can be evaluated for the presence of donor-derived cells. Two- or three-color flow cytometry can be used to identify the progeny of transplanted cells, to document their cell-surface phenotypes, and to follow development of T, B, and myeloid lineages in vivo.


Bone Marrow Transplantation , Hematopoiesis , Leukocyte Common Antigens/metabolism , Lymphocytes/immunology , Radiation Chimera/immunology , Animals , Cell Lineage , Cell Proliferation , Female , Flow Cytometry , Gene Transfer Techniques , Green Fluorescent Proteins/genetics , Immunophenotyping , Leukocyte Common Antigens/genetics , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Transgenic , Pseudomonas Infections/etiology , Pseudomonas Infections/microbiology , Pseudomonas Infections/veterinary , Pseudomonas aeruginosa/isolation & purification , Radiation Dosage , Whole-Body Irradiation/adverse effects
18.
Blood ; 109(12): 5191-8, 2007 Jun 15.
Article En | MEDLINE | ID: mdl-17351112

Gene expression profiling of early eosinophil development shows increased transcript levels of proinflammatory cytokines, chemokines, transcription factors, and a novel gene, EGO (eosinophil granule ontogeny). EGO is nested within an intron of the inositol triphosphate receptor type 1 (ITPR1) gene and is conserved at the nucleotide level; however, the largest open reading frame (ORF) is 86 amino acids. Sucrose density gradients show that EGO is not associated with ribosomes and therefore is a noncoding RNA (ncRNA). EGO transcript levels rapidly increase following interleukin-5 (IL-5) stimulation of CD34(+) hematopoietic progenitors. EGO RNA also is highly expressed in human bone marrow and in mature eosinophils. RNA silencing of EGO results in decreased major basic protein (MBP) and eosinophil derived neurotoxin (EDN) mRNA expression in developing CD34(+) hematopoietic progenitors in vitro and in a CD34(+) cell line model. Therefore, EGO is a novel ncRNA gene expressed during eosinophil development and is necessary for normal MBP and EDN transcript expression.


Eosinophil Granule Proteins/genetics , Eosinophil Major Basic Protein/genetics , Eosinophil-Derived Neurotoxin/genetics , Gene Expression Regulation/genetics , RNA, Untranslated/physiology , Cells, Cultured , Eosinophils/metabolism , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , RNA, Untranslated/genetics , Transcription, Genetic
19.
Ann N Y Acad Sci ; 1106: 82-8, 2007 Jun.
Article En | MEDLINE | ID: mdl-17395732

Prospective isolation of hematopoietic stem and progenitor cell subsets depends upon the premise that expression of combinations of surface antigens reflects developmental potential. During the process of differentiation, however, the loss of antigens associated with stem cells and the concomitant gain of those associated with progenitor cells often occurs as a continuum rather than by discrete binary steps. Coupled with the fact that assay conditions can profoundly influence the developmental fates of prospectively isolated cells, gradients of antigen expression during differentiation have led to a variety of interpretations of lineage commitment in hematopoiesis.


Antigens/biosynthesis , Gene Expression Regulation, Developmental , Gene Expression Regulation , Hematopoiesis , Animals , Antigens/metabolism , Bone Marrow Cells/cytology , Cell Separation , Erythropoiesis , Flow Cytometry , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Stem Cells/cytology , Time Factors
20.
Blood ; 109(6): 2618-21, 2007 Mar 15.
Article En | MEDLINE | ID: mdl-17148589

Congenital erythropoietic porphyria (CEP), an autosomal recessive disorder, is due to mutations of uroporphyrinogen III synthase (UROS). Deficiency of UROS results in excess uroporphyrin I, which causes photosensitization. We evaluated a 3-year-old boy with CEP. A hypochromic, microcytic anemia was present from birth, and platelet counts averaged 70 x 10(9)/L (70,000/microL). Erythrocyte UROS activity was 21% of controls. Red cell morphology and globin chain labeling studies were compatible with beta-thalassemia. Hb electrophoresis revealed 36.3% A, 2.4% A(2), 59.5% F, and 1.8% of an unidentified peak. No UROS or alpha- and beta-globin mutations were found in the child or the parents. The molecular basis of the phenotype proved to be a mutation of GATA1, an X-linked transcription factor common to globin genes and heme biosynthetic enzymes in erythrocytes. A mutation at codon 216 in the child and on one allele of his mother changed arginine to tryptophan (R216W). This is the first report of a human porphyria due to a mutation in a trans-acting factor and the first association of CEP with thalassemia and thrombocytopenia. The Hb F level of 59.5% suggests a role for GATA-1 in globin switching. A bone marrow allograft corrected both the porphyria and the thalassemia.


GATA1 Transcription Factor/genetics , Mutation/genetics , Porphyria, Erythropoietic/genetics , Transcriptional Activation/genetics , Amino Acid Sequence , Child, Preschool , Female , GATA1 Transcription Factor/chemistry , Humans , Male , Molecular Sequence Data , Pedigree , Porphyria, Erythropoietic/etiology , Porphyria, Erythropoietic/pathology , Protein Binding , Zinc Fingers
...