Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 295(12): 3906-3917, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32054685

ABSTRACT

Transferrin receptor 2 (TFR2) is a transmembrane protein expressed mainly in hepatocytes and in developing erythroid cells and is an important focal point in systemic iron regulation. Loss of TFR2 function results in a rare form of the iron-overload disease hereditary hemochromatosis. Although TFR2 in the liver has been shown to be important for regulating iron homeostasis in the body, TFR2's function in erythroid progenitors remains controversial. In this report, we analyzed TFR2-deficient mice in the presence or absence of iron overload to distinguish between the effects caused by a high iron load and those caused by loss of TFR2 function. Analysis of bone marrow from TFR2-deficient mice revealed a reduction in the early burst-forming unit-erythroid and an expansion of late-stage erythroblasts that was independent of iron overload. Spleens of TFR2-deficient mice displayed an increase in colony-forming unit-erythroid progenitors and in all erythroblast populations regardless of iron overload. This expansion of the erythroid compartment coincided with increased erythroferrone (ERFE) expression and serum erythropoietin (EPO) levels. Rescue of hepatic TFR2 expression normalized hepcidin expression and the total cell count of the bone marrow and spleen, but it had no effect on erythroid progenitor frequency. On the basis of these results, we propose a model of TFR2's function in murine erythropoiesis, indicating that deficiency in this receptor is associated with increased erythroid development and expression of EPO and ERFE in extrahepatic tissues independent of TFR's role in the liver.


Subject(s)
Erythropoiesis , Iron Overload/pathology , Receptors, Transferrin/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cytokines/metabolism , Erythropoietin/blood , Hepcidins/metabolism , Iron Overload/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/metabolism , Receptors, Transferrin/deficiency , Spleen/pathology , Stem Cells/cytology , Stem Cells/metabolism
2.
Blood ; 123(20): 3175-84, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24652990

ABSTRACT

The corepressor Rcor1 has been linked biochemically to hematopoiesis, but its function in vivo remains unknown. We show that mice deleted for Rcor1 are profoundly anemic and die in late gestation. Definitive erythroid cells from mutant mice arrest at the transition from proerythroblast to basophilic erythroblast. Remarkably, Rcor1 null erythroid progenitors cultured in vitro form myeloid colonies instead of erythroid colonies. The mutant proerythroblasts also aberrantly express genes of the myeloid lineage as well as genes typical of hematopoietic stem cells (HSCs) and/or progenitor cells. The colony-stimulating factor 2 receptor ß subunit (Csf2rb), which codes for a receptor implicated in myeloid cytokine signaling, is a direct target for both Rcor1 and the transcription repressor Gfi1b in erythroid cells. In the absence of Rcor1, the Csf2rb gene is highly induced, and Rcor1(-/-) progenitors exhibit CSF2-dependent phospho-Stat5 hypersensitivity. Blocking this pathway can partially reduce myeloid colony formation by Rcor1-deficient erythroid progenitors. Thus, Rcor1 promotes erythropoiesis by repressing HSC and/or progenitor genes, as well as the genes and signaling pathways that lead to myeloid cell fate.


Subject(s)
Co-Repressor Proteins/metabolism , Erythropoiesis , Animals , Cells, Cultured , Co-Repressor Proteins/genetics , Cytokine Receptor Common beta Subunit/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Erythroblasts/cytology , Erythroblasts/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/pathology , Gene Deletion , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Myeloid Cells/cytology , Receptors, Interleukin-3/metabolism , Signal Transduction
3.
Stem Cells ; 33(11): 3304-14, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26119982

ABSTRACT

Based on its physical interactions with histone-modifying enzymes, the transcriptional corepressor Rcor1 has been implicated in the epigenetic regulation blood cell development. Previously, we have demonstrated that Rcor1 is essential for the maturation of definitive erythroid cells and fetal survival. To determine the functional role of Rcor1 in steady-state hematopoiesis in the adult, we used a conditional knockout approach. Here, we show that the loss of Rcor1 expression results in the rapid onset of severe anemia due to a complete, cell autonomous block in the maturation of committed erythroid progenitors. By contrast, both the frequency of megakaryocyte progenitors and their capacity to produce platelets were normal. Although the frequency of common lymphoid progenitors and T cells was not altered, B cells were significantly reduced and showed increased apoptosis. However, Rcor1-deficient bone marrow sustained normal levels of B-cells following transplantation, indicating a non-cell autonomous requirement for Rcor1 in B-cell survival. Evaluation of the myelomonocytic lineage revealed an absence of mature neutrophils and a significant increase in the absolute number of monocytic cells. Rcor1-deficient monocytes were less apoptotic and showed ∼100-fold more colony-forming activity than their normal counterparts, but did not give rise to leukemia. Moreover, Rcor1(-/-) monocytes exhibited extensive, cytokine-dependent self-renewal and overexpressed genes associated with hematopoietic stem/progenitor cell expansion including Gata2, Meis1, and Hoxa9. Taken together, these data demonstrate that Rcor1 is essential for the normal differentiation of myeloerythroid progenitors and for appropriately regulating self-renewal activity in the monocyte lineage.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Co-Repressor Proteins/metabolism , Erythroid Precursor Cells/metabolism , Neutrophils/metabolism , Animals , Erythroid Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monocytes/metabolism
4.
Biol Blood Marrow Transplant ; 20(1): 132-5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24161922

ABSTRACT

Human cytomegalovirus (HCMV) infection, including primary infection resulting from transmission from a seropositive donor to a seronegative recipient (D(+)/R(-)), remains a significant problem in the setting of peripheral blood stem cell transplantation (PBSCT). The lack of a suitable animal model for studying HCMV transmission after PBSCT is a major barrier to understanding this process and, consequently, developing novel interventions to prevent HCMV infection. Our previous work demonstrated that human CD34(+) progenitor cell-engrafted NOD-scid IL2Rγc(null) (NSG) mice support latent HCMV infection after direct inoculation and reactivation after treatment with granulocyte colony-stimulating factor. To more accurately recapitulate HCMV infection in the D(+)/R(-) PBSCT setting, granulocyte colony-stimulating factor-mobilized peripheral blood stem cells from seropositive donors were used to engraft NSG mice. All recipient mice demonstrated evidence of HCMV infection in liver, spleen, and bone marrow. These findings validate the NSG mouse model for studying HCMV transmission during PBSCT.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Peripheral Blood Stem Cell Transplantation , Animals , Bone Marrow/immunology , Bone Marrow/pathology , Bone Marrow/virology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Humans , Liver/immunology , Liver/pathology , Liver/virology , Mice , Mice, Transgenic , Spleen/immunology , Spleen/pathology , Spleen/virology , Transplantation, Heterologous , Viral Load , Virus Activation , Virus Replication
5.
PLoS Pathog ; 7(12): e1002444, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22241980

ABSTRACT

Clinical strains of HCMV encode 20 putative ORFs within a region of the genome termed ULb' that are postulated to encode functions related to persistence or immune evasion. We have previously identified ULb'-encoded pUL138 as necessary, but not sufficient, for HCMV latency in CD34+ hematopoietic progenitor cells (HPCs) infected in vitro. pUL138 is encoded on polycistronic transcripts that also encode 3 additional proteins, pUL133, pUL135, and pUL136, collectively comprising the UL133-UL138 locus. This work represents the first characterization of these proteins and identifies a role for this locus in infection. Similar to pUL138, pUL133, pUL135, and pUL136 are integral membrane proteins that partially co-localized with pUL138 in the Golgi during productive infection in fibroblasts. As expected of ULb' sequences, the UL133-UL138 locus was dispensable for replication in cultured fibroblasts. In CD34+ HPCs, this locus suppressed viral replication in HPCs, an activity attributable to both pUL133 and pUL138. Strikingly, the UL133-UL138 locus was required for efficient replication in endothelial cells. The association of this locus with three context-dependent phenotypes suggests an exciting role for the UL133-UL138 locus in modulating the outcome of viral infection in different contexts of infection. Differential profiles of protein expression from the UL133-UL138 locus correlated with the cell-type dependent phenotypes associated with this locus. We extended our in vitro findings to analyze viral replication and dissemination in a NOD-scid IL2Rγ(c) (null)-humanized mouse model. The UL133-UL138(NULL) virus exhibited an increased capacity for replication and/or dissemination following stem cell mobilization relative to the wild-type virus, suggesting an important role in viral persistence and spread in the host. As pUL133, pUL135, pUL136, and pUL138 are conserved in virus strains infecting higher order primates, but not lower order mammals, the functions encoded likely represent host-specific viral adaptations.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/physiology , Genetic Loci , Hematopoietic Stem Cells/virology , Host-Pathogen Interactions/physiology , Viral Tropism/physiology , Virus Replication/physiology , Animals , Cell Line , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/pathology , Disease Models, Animal , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID
6.
Blood ; 116(24): 5140-8, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20826722

ABSTRACT

Progressive bone marrow failure is a major cause of morbidity and mortality in human Fanconi Anemia patients. In an effort to develop a Fanconi Anemia murine model to study bone marrow failure, we found that Fancd2(-/-) mice have readily measurable hematopoietic defects. Fancd2 deficiency was associated with a significant decline in the size of the c-Kit(+)Sca-1(+)Lineage(-) (KSL) pool and reduced stem cell repopulation and spleen colony-forming capacity. Fancd2(-/-) KSL cells showed an abnormal cell cycle status and loss of quiescence. In addition, the supportive function of the marrow microenvironment was compromised in Fancd2(-/-) mice. Treatment with Sirt1-mimetic and the antioxidant drug, resveratrol, maintained Fancd2(-/-) KSL cells in quiescence, improved the marrow microenvironment, partially corrected the abnormal cell cycle status, and significantly improved the spleen colony-forming capacity of Fancd2(-/-) bone marrow cells. We conclude that Fancd2(-/-) mice have readily quantifiable hematopoietic defects, and that this model is well suited for pharmacologic screening studies.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia/drug therapy , Hematopoietic System/drug effects , Stilbenes/pharmacology , Animals , Antioxidants , Bone Marrow/drug effects , Cell Cycle , Cell Lineage , Colony-Forming Units Assay , Fanconi Anemia Complementation Group D2 Protein/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Mice , Mice, Knockout , Mice, Transgenic , Resveratrol , Spleen/cytology , Stilbenes/therapeutic use , Treatment Outcome
7.
Proc Natl Acad Sci U S A ; 106(11): 4390-5, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19251665

ABSTRACT

The expression of ASPP2 (53BP2L), a proapoptotic member of a family of p53-binding proteins, is frequently suppressed in many human cancers. Accumulating evidence suggests that ASPP2 inhibits tumor growth; however, the mechanisms by which ASPP2 suppresses tumor formation remain to be clarified. To study this, we targeted the ASPP2 allele in a mouse by replacing exons 10-17 with a neoR gene. ASPP2(-/-) mice were not viable because of an early embryonic lethal event. Although ASPP2(+/-) mice appeared developmentally normal, they displayed an increased incidence of a variety of spontaneous tumors as they aged. Moreover, gamma-irradiated 6-week-old ASPP2(+/-) mice developed an increased incidence of high-grade T cell lymphomas of thymic origin compared with ASPP2(+/+) mice. Primary thymocytes derived from ASPP2(+/-) mice exhibited an attenuated apoptotic response to gamma-irradiation compared with ASPP2(+/+) thymocytes. Additionally, ASPP2(+/-) primary mouse embryonic fibroblasts demonstrated a defective G(0)/G(1) cell cycle checkpoint after gamma-irradiation. Our results demonstrate that ASPP2 is a haploinsufficient tumor suppressor and, importantly, open new avenues for investigation into the mechanisms by which disruption of ASPP2 pathways could play a role in tumorigenesis and response to therapy.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/radiation effects , Cell Cycle/radiation effects , Gamma Rays , Genetic Predisposition to Disease , Heterozygote , Lymphoma, T-Cell/etiology , Lymphoma, T-Cell/genetics , Mice , Mice, Mutant Strains , Neoplasms/etiology , Neoplasms/genetics , Thymus Gland
8.
Blood ; 114(20): 4393-401, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19759357

ABSTRACT

Bone morphogenetic protein 4 (BMP4) is required for mesoderm commitment to the hematopoietic lineage during early embryogenesis. However, deletion of BMP4 is early embryonically lethal and its functional role in definitive hematopoiesis is unknown. Consequently, we used a BMP4 hypomorph to investigate the role of BMP4 in regulating hematopoietic stem cell (HSC) function and maintaining steady-state hematopoiesis in the adult. Reporter gene expression shows that Bmp4 is expressed in cells associated with the hematopoietic microenvironment including osteoblasts, endothelial cells, and megakaryocytes. Although resting hematopoiesis is normal in a BMP4-deficient background, the number of c-Kit+, Sca-1+, Lineage- cells is significantly reduced. Serial transplantation studies reveal that BMP4-deficient recipients have a microenvironmental defect that reduces the repopulating activity of wild-type HSCs. This defect is even more pronounced in a parabiosis model that demonstrates a profound reduction in wild-type hematopoietic cells within the bone marrow of BMP4-deficient recipients. Furthermore, wild-type HSCs that successfully engraft into the BMP4-deficient bone marrow show a marked decrease in functional stem cell activity when tested in a competitive repopulation assay. Taken together, these findings indicate BMP4 is a critical component of the hematopoietic microenvironment that regulates both HSC number and function.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Stem Cell Niche/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Flow Cytometry , Gene Expression , Mice , Parabiosis
9.
Nat Med ; 10(7): 744-8, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15195088

ABSTRACT

Liver repopulation with bone marrow-derived hepatocytes (BMHs) can cure the genetic liver disease fumarylacetoacetate hydrolase (Fah) deficiency. BMHs emerge from fusion between donor bone marrow-derived cells and host hepatocytes. To use such in vivo cell fusion efficiently for therapy requires knowing the nature of the hematopoietic cells that fuse with hepatocytes. Here we show that the transplantation into Fah(-/-) mice of hematopoietic stem cells (HSCs) from lymphocyte-deficient Rag1(-/-) mice, lineage-committed granulocyte-macrophage progenitors (GMPs) or bone marrow-derived macrophages (BMMs) results in the robust production of BMHs. These results provide direct evidence that committed myelomonocytic cells such as macrophages can produce functional epithelial cells by in vivo fusion. Because stable bone marrow engraftment or HSCs are not required for this process, macrophages or their highly proliferative progenitors provide potential for targeted and well-tolerated cell therapy aimed at organ regeneration.


Subject(s)
Cell Fusion , Hematopoietic Stem Cell Transplantation , Hepatocytes/cytology , Hydrolases/deficiency , Macrophages/cytology , Animals , Female , Liver Regeneration , Mice , Mice, Inbred C57BL
10.
Cell Stem Cell ; 28(1): 33-47.e8, 2021 01 07.
Article in English | MEDLINE | ID: mdl-32997960

ABSTRACT

Bone marrow failure (BMF) in Fanconi anemia (FA) patients results from dysfunctional hematopoietic stem and progenitor cells (HSPCs). To identify determinants of BMF, we performed single-cell transcriptome profiling of primary HSPCs from FA patients. In addition to overexpression of p53 and TGF-ß pathway genes, we identified high levels of MYC expression. We correspondingly observed coexistence of distinct HSPC subpopulations expressing high levels of TP53 or MYC in FA bone marrow (BM). Inhibiting MYC expression with the BET bromodomain inhibitor (+)-JQ1 reduced the clonogenic potential of FA patient HSPCs but rescued physiological and genotoxic stress in HSPCs from FA mice, showing that MYC promotes proliferation while increasing DNA damage. MYC-high HSPCs showed significant downregulation of cell adhesion genes, consistent with enhanced egress of FA HSPCs from bone marrow to peripheral blood. We speculate that MYC overexpression impairs HSPC function in FA patients and contributes to exhaustion in FA bone marrow.


Subject(s)
Fanconi Anemia , Animals , Bone Marrow , DNA Damage , Fanconi Anemia/genetics , Hematopoietic Stem Cells , Humans , Mice , Transforming Growth Factor beta
11.
Oncogene ; 38(8): 1200-1210, 2019 02.
Article in English | MEDLINE | ID: mdl-30242244

ABSTRACT

Certain cancers, including gastrointestinal stromal tumor (GIST) and subsets of melanoma, are caused by somatic KIT mutations that result in KIT receptor tyrosine kinase constitutive activity, which drives proliferation. The treatment of KIT-mutant GIST has been revolutionized with the advent of KIT-directed cancer therapies. KIT tyrosine kinase inhibitors (TKI) are superior to conventional chemotherapy in their ability to control advanced KIT-mutant disease. However, these therapies have a limited duration of activity due to drug-resistant secondary KIT mutations that arise (or that are selected for) during KIT TKI treatment. To overcome the problem of KIT TKI resistance, we sought to identify novel therapeutic targets in KIT-mutant GIST and melanoma cells using a human tyrosine kinome siRNA screen. From this screen, we identified lemur tyrosine kinase 3 (LMTK3) and herein describe its role as a novel KIT regulator in KIT-mutant GIST and melanoma cells. We find that LMTK3 regulated the translation rate of KIT, such that loss of LMTK3 reduced total KIT, and thus KIT downstream signaling in cancer cells. Silencing of LMTK3 decreased cell viability and increased cell death in KIT-dependent, but not KIT-independent GIST and melanoma cell lines. Notably, LMTK3 silencing reduced viability of all KIT-mutant cell lines tested, even those with drug-resistant KIT secondary mutations. Furthermore, targeting of LMTK3 with siRNA delayed KIT-dependent GIST growth in a xenograft model. Our data suggest the potential of LMTK3 as a target for treatment of patients with KIT-mutant cancer, particularly after failure of KIT TKIs.


Subject(s)
Gastrointestinal Stromal Tumors/drug therapy , Melanoma/drug therapy , Membrane Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-kit/genetics , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Humans , Imatinib Mesylate/administration & dosage , Melanoma/genetics , Melanoma/pathology , Mice , Mutation/drug effects , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
12.
PLoS One ; 12(11): e0188910, 2017.
Article in English | MEDLINE | ID: mdl-29190822

ABSTRACT

Protein phosphatase 2A (PP2A) is a ubiquitously expressed Serine-Threonine phosphatase mediating 30-50% of protein phosphatase activity. PP2A functions as a heterotrimeric complex, with the B subunits directing target specificity to regulate the activity of many key pathways that control cellular phenotypes. PP2A-B56α has been shown to play a tumor suppressor role and to negatively control c-MYC stability and activity. Loss of B56α promotes cellular transformation, likely at least in part through its regulation of c-MYC. Here we report generation of a B56α hypomorph mouse with very low B56α expression that we used to study the physiologic activity of the PP2A-B56α phosphatase. The predominant phenotype we observed in mice with B56α deficiency in the whole body was spontaneous skin lesion formation with hyperproliferation of the epidermis, hair follicles and sebaceous glands. Increased levels of c-MYC phosphorylation on Serine62 and c-MYC activity were observed in the skin lesions of the B56αhm/hm mice. B56α deficiency was found to increase the number of skin stem cells, and consistent with this, papilloma initiation was accelerated in a carcinogenesis model. Further analysis of additional tissues revealed increased inflammation in spleen, liver, lung, and intestinal lymph nodes as well as in the skin lesions, resembling elevated extramedullary hematopoiesis phenotypes in the B56αhm/hm mice. We also observed an increase in the clonogenicity of bone marrow stem cells in B56αhm/hm mice. Overall, this model suggests that B56α is important for stem cells to maintain homeostasis and that B56α loss leading to increased activity of important oncogenes, including c-MYC, can result in aberrant cell growth and increased stem cells that can contribute to the initiation of malignancy.


Subject(s)
Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , Protein Phosphatase 2/metabolism , Animals , Mice , Mice, Transgenic , Neoplasms, Experimental/enzymology
13.
Exp Hematol ; 31(11): 987-93, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14585360

ABSTRACT

Classical studies of the developing embryo first suggested the existence of the hemangioblast, a precursor cell with the potential to differentiate into both blood and blood vessels. Several lines of investigation demonstrated that many of the genes activated during early hematopoietic development are also expressed in the vascular endothelium. Gene-targeting studies using embryonic stem cells have identified Flk-1, SCL, and Runx-1 as important regulatory molecules that specify both hematopoietic and vascular outcomes. Although it was anticipated that the hemangioblast would be present only during the earliest stages of vascular development in the yolk sac, accumulating evidence now indicates that hematopoietic cells with hemangioblast activity persist into adulthood. In the adult, bone marrow-derived, circulating endothelial progenitors contribute to postnatal neovascularization and enhance vascular repair following ischemic injury. Highly purified populations of hematopoietic stem cells from humans and mice can differentiate into both blood cells and vascular tissue at the single cell level. These recent findings suggest that bone marrow-derived hematopoietic stem cells or their progeny may contribute to the maintenance and repair of both the hematopoietic and the vascular systems during adult life.


Subject(s)
Hematopoietic Stem Cells/physiology , Animals , Cell Differentiation , Embryo, Mammalian/cytology , Endothelium, Vascular/cytology , Humans , Neovascularization, Physiologic/physiology
14.
Exp Hematol ; 31(11): 1119-25, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14585378

ABSTRACT

OBJECTIVE: Synergy between Flt-3 ligand and G-CSF produces a marked expansion of hematopoietic progenitor cells and mobilizes large numbers of stem cells into the peripheral blood. To determine if the activation of the Flt-3 and G-CSF receptors enhances the regenerative capacity of the hematopoietic compartment, we evaluated whether activation of these receptors augments stem cell recovery following lethal doses of radiation. METHODS: C57BL/6 mice received a single injection of the bi-functional Flt-3 and G-GSF agonist progenipoietin-1, 24 hours prior to exposure to 1100 cGy of gamma radiation. Survival, hematopoietic reconstitution, and competitive repopulation potential were evaluated. RESULTS: All cytokine-treated mice survived for up to 9 months. Radioprotected recipients exhibited stable multilineage hematopoiesis and recovered normal numbers of T cells, B cells, and myelomonocytic cells in the blood, bone marrow, and thymus. Between 2 and 3 weeks following radiation, cytokine-treated mice demonstrated threefold higher serum hemoglobin levels, 10-fold higher nucleated blood cell counts, and 20-fold higher platelet counts compared to controls. Radioprotection of self-renewing hematopoietic stem cells was revealed by multilineage hematopoietic reconstitution following transplantation in a competitive repopulation assay. To further evaluate the extent of cytokine-induced radioprotective activity, a cohort of mice received a second cycle of cytokine treatment and a second exposure to radiation (1100 cGy). Survival of this serially irradiated group was 70% and analysis of the peripheral blood revealed sustained multilineage hematopoiesis. CONCLUSION: These results demonstrate that activation of both the Flt-3 and G-CSF receptors provides a high degree of radioprotection to the hematopoietic progenitor cell and stem cell compartment.


Subject(s)
Colony-Stimulating Factors/pharmacology , Hematopoietic Stem Cells/radiation effects , Proto-Oncogene Proteins/physiology , Radiation-Protective Agents/pharmacology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Granulocyte Colony-Stimulating Factor/physiology , Animals , Mice , Mice, Inbred C57BL , Recombinant Proteins , fms-Like Tyrosine Kinase 3
15.
Exp Hematol ; 30(8): 950-6, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12160847

ABSTRACT

OBJECTIVE: Accumulating evidence indicates a common stem cell may be responsible for both vasculogenesis and blood cell production during early embryologic development, yet little is known about the fate of these cells during ontogeny. We sought to determine whether hematopoietic potential is associated with normal blood vessels in the adult. MATERIALS AND METHODS: Segments of adult thoracic aorta or inferior vena cava were transplanted under the kidney capsule of lethally irradiated recipients (1100 cGy). Radioprotection, colony-forming units (CFUs), and the extent of donor-derived hematopoietic constitution were evaluated using both Ly5 congenic and ROSA26 donor mice. RESULTS: As little as 10 mg of transplanted vascular tissue radioprotected 80% of recipients, gave rise to similar numbers of CFUs as 10(5) bone marrow cells and prevented the development of severe anemia. Bromodeoxyuridine labeling studies revealed cell proliferation within the intima of donor vascular tissue within 48 hours of transplantation. ROSA26 donor-derived vascular cells migrated to the recipient spleen; however, CFUs were of host origin, a finding confirmed using sex-mismatched transplants. Although donor-derived cells were readily detected in the peripheral blood 2 to 3 weeks after transplant, they rapidly declined in frequency to approximately 1.0% by 4 weeks and persisted at these levels for more than 1 year. Bone marrow from rescued primary recipients provided radioprotection after transplantation into secondary recipients; however, only CD3(+) donor-derived cells were detected. CONCLUSION: These findings demonstrate the presence of a population of cells within normal adult vascular tissue that has the capacity to protect host hematopoietic stem cells from radiation-induced death.


Subject(s)
Aorta, Thoracic/transplantation , Graft Survival , Hematopoietic Stem Cells/cytology , Transplantation, Heterotopic , Vena Cava, Inferior/transplantation , Animals , Aorta, Thoracic/cytology , CD3 Complex/analysis , Cell Differentiation , Cell Division , Cell Lineage , Colony-Forming Units Assay , Female , Hematopoiesis , Kidney , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Radiation Chimera , Spleen/cytology , Vena Cava, Inferior/cytology
16.
Radiat Res ; 183(3): 338-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25738896

ABSTRACT

Bone marrow suppression due to exposure to ionizing radiation is a significant clinical problem associated with radiation therapy as well as with nonmedical radiation exposure. Currently, there are no small molecule agents available that can enhance hematopoietic regeneration after radiation exposure. Here, we report on the effective mitigation of acute hematopoietic radiation syndrome in mice by the synthetic triterpenoid, RTA 408. The administration of a brief course of RTA 408 treatment, beginning 24 h after lethal doses of radiation to bone marrow, significantly increased overall survival. Importantly, treatment with RTA 408 led to the full recovery of steady state hematopoiesis with normalization of the frequency of hematopoietic stem and progenitor cells. Moreover, hematopoietic stem cells from RTA 408-mitigated mice showed lineage-balanced, long-term, multilineage potential in serial transplantation assays, indicative of their normal self-renewal activity. The potency of RTA 408 in mitigating radiation-induced bone marrow suppression makes it an attractive candidate for potential clinical use in treating both therapy-related and unanticipated radiation exposure.


Subject(s)
Acute Radiation Syndrome/drug therapy , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Triterpenes/administration & dosage , Acute Radiation Syndrome/pathology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Dose-Response Relationship, Radiation , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/radiation effects , Mice , Radiation, Ionizing , Radiotherapy/adverse effects , Whole-Body Irradiation
17.
Leukemia ; 28(10): 1978-1987, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24637335

ABSTRACT

Vascular endothelial cells are a critical component of the hematopoietic microenvironment that regulates blood cell production. Recent studies suggest the existence of functional cross-talk between hematologic malignancies and vascular endothelium. Here we show that human acute myeloid leukemia (AML) localizes to the vasculature in both patients and in a xenograft model. A significant number of vascular tissue-associated AML cells (V-AML) integrate into vasculature in vivo and can fuse with endothelial cells. V-AML cells acquire several endothelial cell-like characteristics, including the upregulation of CD105, a receptor associated with activated endothelium. Remarkably, endothelial-integrated V-AML shows an almost fourfold reduction in proliferative activity compared with non-vascular-associated AML. Primary AML cells can be induced to downregulate the expression of their hematopoietic markers in vitro and differentiate into phenotypically and functionally defined endothelial-like cells. After transplantation, these leukemia-derived endothelial cells are capable of giving rise to AML. These novel functional interactions between AML cells and normal endothelium along with the reversible endothelial cell potential of AML suggest that vascular endothelium may serve as a previously unrecognized reservoir for AML.


Subject(s)
Endothelium, Vascular/metabolism , Leukemia, Myeloid, Acute/physiopathology , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/metabolism , Cell Differentiation , Cell Line , Cell Survival , Cells, Cultured , Endoglin , Female , Humans , In Situ Hybridization, Fluorescence , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Mice, Inbred NOD , Middle Aged , Neoplasm Transplantation , Phenotype , Receptors, Cell Surface/metabolism , Recurrence , Young Adult
18.
Stem Cell Res ; 11(3): 1013-21, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23939266

ABSTRACT

Endothelial cells (ECs) are an essential component of the hematopoietic microenvironment, which maintains and regulates hematopoietic stem cells (HSCs). Although ECs can support the regeneration of otherwise lethally-irradiated HSCs, the mechanisms are not well understood. To further understand this phenomenon, we studied HSC regeneration from irradiated bone marrow using co-culture with human aortic ECs (HAECs). Co-culture with HAECs induced a 24-fold expansion of long-term HSCs (CD150(+), lineage(lo), Sca-1(+), c-Kit(+); CD150(+)LSK cells) in vitro. These cells gave rise to functional hematopoietic stem and progenitor cells (HSPCs) with colony-forming activity, multilineage reconstitution and serial transplantation potential. Furthermore, HAECs significantly reduced DNA damage in irradiated LSK cells within 24h. Remarkably, we were able to delay the exposure of irradiated bone marrow to the regenerative, HAEC-derived signals for up to 48h and still rescue functional HSCs. G-CSF is the gold standard for promoting hematopoietic regeneration in vivo. However, when compared to HAECs, in vitro G-CSF treatment promoted lineage differentiation and regenerated 5-fold fewer CD150(+)LSK cells. Together, our results show that HAECs are powerful, direct mitigators of HSC injury and DNA damage. Identification of the HAEC-derived factors that rescue HSCs may lead to improved therapies for hematopoietic regeneration after radiation injury.


Subject(s)
DNA Damage/radiation effects , Endothelial Cells/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/radiation effects , Whole-Body Irradiation , Animals , Aorta/cytology , Bone Marrow Cells/cytology , Bone Marrow Cells/radiation effects , Bone Marrow Transplantation , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Movement , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Endothelial Cells/metabolism , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoiesis , Humans , Male , Mice
19.
Cancer Res ; 73(1): 285-96, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23087056

ABSTRACT

Kinases are dysregulated in most cancers, but the frequency of specific kinase mutations is low, indicating a complex etiology in kinase dysregulation. Here, we report a strategy to rapidly identify functionally important kinase targets, irrespective of the etiology of kinase pathway dysregulation, ultimately enabling a correlation of patient genetic profiles to clinically effective kinase inhibitors. Our methodology assessed the sensitivity of primary leukemia patient samples to a panel of 66 small-molecule kinase inhibitors over 3 days. Screening of 151 leukemia patient samples revealed a wide diversity of drug sensitivities, with 70% of the clinical specimens exhibiting hypersensitivity to one or more drugs. From this data set, we developed an algorithm to predict kinase pathway dependence based on analysis of inhibitor sensitivity patterns. Applying this algorithm correctly identified pathway dependence in proof-of-principle specimens with known oncogenes, including a rare FLT3 mutation outside regions covered by standard molecular diagnostic tests. Interrogation of all 151 patient specimens with this algorithm identified a diversity of kinase targets and signaling pathways that could aid prioritization of deep sequencing data sets, permitting a cumulative analysis to understand kinase pathway dependence within leukemia subsets. In a proof-of-principle case, we showed that in vitro drug sensitivity could predict both a clinical response and the development of drug resistance. Taken together, our results suggested that drug target scores derived from a comprehensive kinase inhibitor panel could predict pathway dependence in cancer cells while simultaneously identifying potential therapeutic options.


Subject(s)
Drug Resistance, Neoplasm/genetics , Gene Expression Profiling/methods , Leukemia/enzymology , Leukemia/genetics , Protein-Tyrosine Kinases/genetics , Algorithms , Cluster Analysis , Humans , Signal Transduction/genetics
20.
Hum Pathol ; 43(12): 2167-76, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22658276

ABSTRACT

Classification of acute myeloid leukemia increasingly depends on genetic analysis. However, the number of known mutations in acute myeloid leukemia is expanding rapidly. Therefore, we tested a high-throughput screening method for acute myeloid leukemia mutation analysis using a multiplex mass spectrometry-based approach. To our knowledge, this is the first reported application of this approach to genotype leukemias in a clinical setting. One hundred seven acute myeloid leukemia cases were screened for mutations using a panel that covers 344 point mutations across 31 genes known to be associated with leukemia. The analysis was performed by multiplex polymerase chain reaction for mutations in genes of interest followed by primer extension reactions. Products were analyzed on a Sequenom MassARRAY system (San Diego, CA). The multiplex panel yielded mutations in 58% of acute myeloid leukemia cases with normal cytogenetics and 21% of cases with abnormal cytogenetics. Cytogenetics and routine polymerase chain reaction-based screening of NPM1, CEBPA, FLT3-ITD, and KIT was also performed on a subset of cases. When combined with the results of these standard polymerase chain reaction-based tests, the mutation frequency reached 78% in cases with normal cytogenetics. Of these, 42% harbored multiple mutations primarily involving NPM1 with NRAS, KRAS, CEBPA, PTPN11, IDH1, or FLT3. In contrast, cases with abnormal cytogenetics rarely harbored more than 1 mutation (1.5%), suggesting different underlying biology. This study demonstrates the feasibility and utility of broad-based mutation profiling of acute myeloid leukemia in a clinical setting. This approach will be helpful in defining prognostic subgroups of acute myeloid leukemia and contribute to the selection of patients for enrollment into trials with novel inhibitors.


Subject(s)
Biomarkers, Tumor/genetics , DNA Mutational Analysis/methods , High-Throughput Screening Assays , Leukemia, Myeloid, Acute/genetics , Multiplex Polymerase Chain Reaction/methods , CCAAT-Enhancer-Binding Proteins/genetics , Female , Humans , Male , Middle Aged , Nuclear Proteins/genetics , Nucleophosmin , Prognosis , Proto-Oncogene Proteins c-kit/genetics , fms-Like Tyrosine Kinase 3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL