Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Stem Cells Dev ; 25(8): 575-85, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26943017

ABSTRACT

Dipeptidyl peptidase 4 (DPP4)/CD26 truncates certain proteins, and this posttranslational modification can influence their activity. Truncated (T) colony-stimulating factors (CSFs) are decreased in potency for stimulating proliferation of hematopoietic progenitor cells (HPCs). T-CXCL12, a modified chemokine, is inactive as an HPC chemotactic, survival, and enhancing factor for replating or ex-vivo expansion of HPCs. Moreover, T-CSFs and T-CXCL12 specifically downmodulates the positively acting effects of their own full-length molecule. Other chemokines have DPP4 truncation sites. In the present study, we evaluated effects of DPP4 inhibition (by Diprotin A) or gene deletion of HPC on chemokine inhibition of multicytokine-stimulated HPC, and on chemokine-enhancing effects on single CSF-stimulated HPC proliferation, as well as effects of DPP4 treatment of a number of chemokines. Myelosuppressive effects of chemokines with, but not without, a DPP4 truncation site were greatly enhanced in inhibitory potency by pretreating target bone marrow (BM) cells with Diprotin A, or by assaying their activity on dpp4/cd26(-/-) BM cells. DPP4 treatment of myelosuppressive chemokines containing a DPP4 truncation site produced a nonmyelosuppressive molecule, but one which had the capacity to block suppression by that unmodified chemokine both in vitro and in vivo. Additionally, DPP4 treatment ablated the single cytokine-stimulated HPC-enhancing activity of CCL3/MIP-1α and CCL4/MIP-1ß, and blocked the enhancing activity of each unmodified molecule, in vitro and in vivo. These results highlight the functional posttranslational modulating effects of DPP4 on chemokine activities, and information offering additional biological insight into chemokine regulation of hematopoiesis.


Subject(s)
Chemokine CCL3/physiology , Chemokine CCL4/physiology , Dipeptidyl Peptidase 4/physiology , Animals , Cell Proliferation , Chemokine CCL3/chemistry , Chemokine CCL4/chemistry , Dipeptidyl Peptidase 4/chemistry , Female , Hematopoiesis , Hematopoietic Stem Cells/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Processing, Post-Translational , Proteolysis
2.
Cell ; 161(7): 1553-65, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26073944

ABSTRACT

Hematopoietic stem cells (HSCs) reside in hypoxic niches within bone marrow and cord blood. Yet, essentially all HSC studies have been performed with cells isolated and processed in non-physiologic ambient air. By collecting and manipulating bone marrow and cord blood in native conditions of hypoxia, we demonstrate that brief exposure to ambient oxygen decreases recovery of long-term repopulating HSCs and increases progenitor cells, a phenomenon we term extraphysiologic oxygen shock/stress (EPHOSS). Thus, true numbers of HSCs in the bone marrow and cord blood are routinely underestimated. We linked ROS production and induction of the mitochondrial permeability transition pore (MPTP) via cyclophilin D and p53 as mechanisms of EPHOSS. The MPTP inhibitor cyclosporin A protects mouse bone marrow and human cord blood HSCs from EPHOSS during collection in air, resulting in increased recovery of transplantable HSCs. Mitigating EPHOSS during cell collection and processing by pharmacological means may be clinically advantageous for transplantation.


Subject(s)
Bone Marrow , Fetal Blood/cytology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Female , Hematopoietic Stem Cell Transplantation/instrumentation , Hematopoietic Stem Cells/cytology , Humans , Hypoxia , Mice , Mice, Inbred C57BL , Oxygen/metabolism , Tumor Suppressor Protein p53/metabolism
3.
Stem Cells ; 33(6): 1975-84, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25753525

ABSTRACT

Simple efforts are needed to enhance cord blood (CB) transplantation. We hypothesized that short-term exposure of CD34(+) CB cells to 39.5°C would enhance their response to stromal-derived factor-1 (SDF-1), by increasing lipid raft aggregation and CXCR4 expression, thus leading to enhanced engraftment. Mild hyperthermia (39.5°C) significantly increased the percent of CD34(+) CB that migrated toward SDF-1. This was associated with increased expression of CXCR4 on the cells. Mechanistically, mild heating increased the percent of CD34(+) cells with aggregated lipid rafts and enhanced colocalization of CXCR4 within lipid raft domains. Using methyl-ß-cyclodextrin (MßCD), an agent that blocks lipid raft aggregation, it was determined that this enhancement in chemotaxis was dependent upon lipid raft aggregation. Colocalization of Rac1, a GTPase crucial for cell migration and adhesion, with CXCR4 to the lipid raft was essential for the effects of heat on chemotaxis, as determined with an inhibitor of Rac1 activation, NSC23766. Application-wise, mild heat treatment significantly increased the percent chimerism as well as homing and engraftment of CD34(+) CB cells in sublethally irradiated non-obese diabetic severe combined immunodeficiency IL-2 receptor gamma chain d (NSG) mice. Mild heating may be a simple and inexpensive means to enhance engraftment following CB transplantation in patients.


Subject(s)
Cell Movement , Chemokine CXCL12/metabolism , Fetal Blood/cytology , Animals , Antigens, CD34/immunology , Blood Cells/cytology , Bone Marrow/metabolism , Female , Hematopoietic Stem Cell Transplantation/methods , Hot Temperature , Humans , Receptors, CXCR4/metabolism , Receptors, Interleukin-2/deficiency
4.
Stem Cells Dev ; 23(21): 2661-71, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24914588

ABSTRACT

A hyaluronic-acid-rich node and duct system (HAR-NDS) was found on the surface of internal organs of mice, and inside their blood and lymph vessels. The nodes (HAR-Ns) were filled with immune cells of the innate system and were especially enriched with mast cells and histiocytes. They also contained hematopoietic progenitor cells (HPCs), such as granulocyte-macrophage, erythroid, multipotential progenitors, and mast cell progenitors (MCPs). MCPs were the most abundant among the HPCs in HAR-Ns. Their frequency was fivefold higher than that of the MCPs in bone marrow. In addition, the system contained pluripotent stem cells (PSCs) capable of producing CD45(-)Flk1(+) hemangioblast-like cells, which subsequently generated various types of HPCs and differentiated blood cells. Although HAR-Ns did not appear to harbor enough number of cells capable of long-term reconstitution or short-term radioprotection of lethally irradiated recipients, bone marrow cells were able to engraft in the HAR-NDS and reconstitute hematopoietic potentials of the system. PSCs and HPCs were consistently found in intravenous, intralymphatic, and intestinal HAR-ND. We infer that PSCs and HPCs reside in the HAR-ND and that this novel system may serve as an alternative means to traffic immature and mature blood cells throughout the body.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/metabolism , Hyaluronic Acid/metabolism , Pluripotent Stem Cells/metabolism , Animals , Blood Vessels/cytology , Blood Vessels/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation , Flow Cytometry , Hemangioblasts/cytology , Hemangioblasts/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/ultrastructure , Histiocytes/cytology , Histiocytes/metabolism , Immune System/cytology , Immune System/metabolism , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Mast Cells/cytology , Mast Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/ultrastructure , Spleen/cytology , Spleen/metabolism , Stem Cell Transplantation/methods , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Stem Cells Dev ; 22(11): 1678-92, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23311338

ABSTRACT

Hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) play a vital role in replenishment of blood cells. In addition to growth factors, energy metabolism plays an important role in cellular proliferation. Oxidative phosphorylation that occurs in the mitochondria is the major source of ATP. In this study, we have investigated the role of peroxisome proliferator-activated-γ coactivator-1α (PGC-1α), a major regulator of mitochondrial biogenesis, in hematopoiesis. PGC-1α is expressed in HSC/HPCs. Loss of PGC-1α minimally affects basal hematopoiesis; however, it significantly impairs stress hematopoiesis. Recovery of hematopoiesis poststress involves rapid proliferation of HSC/HPCs. Growth factors stimulate HSC/HPC proliferation in a dose-dependent manner and this response is modulated by oxygen tension. Although severe hypoxic conditions inhibit HSC/HPC proliferation, mild hypoxia enhances the clonogenic potential; however, the mechanism underlying this phenomenon remains largely unknown. Our studies demonstrate that PGC-1α-mediated mitochondrial biogenesis is critical for the increased clonogenic potential of progenitors under mild hypoxia. Metabolic programming and increased glucose uptake can drive rapid progenitor cell proliferation under relatively low oxygen tension only if the HPC has the capacity to increase PGC-1α expression and mitochondrial biogenesis. Loss of PGC-1α also impairs the long-term repopulating potential of HSCs. Our findings may have therapeutic applications for rapid recovery of blood cells following myeloablation.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/physiology , Mitochondria/metabolism , Transcription Factors/metabolism , Animals , Cell Hypoxia , Cell Proliferation , Energy Metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Oxygen Species/metabolism , Transcription Factors/genetics
6.
Nat Med ; 18(12): 1786-96, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23160239

ABSTRACT

Enhancement of hematopoietic recovery after radiation, chemotherapy, or hematopoietic stem cell (HSC) transplantation is clinically relevant. Dipeptidylpeptidase (DPP4) cleaves a wide variety of substrates, including the chemokine stromal cell-derived factor-1 (SDF-1). In the course of experiments showing that inhibition of DPP4 enhances SDF-1-mediated progenitor cell survival, ex vivo cytokine expansion and replating frequency, we unexpectedly found that DPP4 has a more general role in regulating colony-stimulating factor (CSF) activity. DPP4 cleaved within the N-termini of the CSFs granulocyte-macrophage (GM)-CSF, G-CSF, interleukin-3 (IL-3) and erythropoietin and decreased their activity. Dpp4 knockout or DPP4 inhibition enhanced CSF activities both in vitro and in vivo. The reduced activity of DPP4-truncated versus full-length human GM-CSF was mechanistically linked to effects on receptor-binding affinity, induction of GM-CSF receptor oligomerization and signaling capacity. Hematopoiesis in mice after radiation or chemotherapy was enhanced in Dpp4(-/-) mice or mice receiving an orally active DPP4 inhibitor. DPP4 inhibition enhanced engraftment in mice without compromising HSC function, suggesting the potential clinical utility of this approach.


Subject(s)
Chemokine CXCL12/metabolism , Dipeptidyl Peptidase 4/metabolism , Drug-Related Side Effects and Adverse Reactions , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hematopoiesis/physiology , Radiotherapy/adverse effects , Signal Transduction/physiology , Animals , Cell Line , DNA Primers/genetics , Dipeptidyl Peptidase 4/genetics , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Humans , Immunophenotyping , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics
7.
J Immunol ; 189(10): 4759-69, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23053511

ABSTRACT

The transcriptional repressor Bcl6 is a critical arbiter of Th cell fate, promoting the follicular Th lineage while repressing other Th cell lineages. Bcl6-deficient (Bcl6(-/-)) mice develop a spontaneous and severe Th2-type inflammatory disease, thus warranting assessment of Bcl6 in regulatory T cell (Treg) function. Bcl6(-/-) Tregs were competent at suppressing T cell proliferation in vitro and Th1-type colitogenic T cell responses in vivo. In contrast, Bcl6(-/-) Tregs strongly exacerbated lung inflammation in a model of allergic airway disease and promoted higher Th2 responses, including systemic upregulation of microRNA-21. Further, Bcl6(-/-) Tregs were selectively impaired at controlling Th2 responses, but not Th1 and Th17 responses, in mixed chimeras of Bcl6(-/-) bone marrow with Foxp3(-/-) bone marrow. Bcl6(-/-) Tregs displayed increased levels of the Th2 transcription factor Gata3 and other Th2 and Treg genes. Bcl6 potently repressed Gata3 transcriptional transactivation, providing a mechanism for the increased expression of Th2 genes by Bcl6(-/-) Tregs. Gata3 has a critical role in regulating Foxp3 expression and functional fitness of Tregs; however, the signal that regulates Gata3 and restricts its transactivation of Th2 cytokines in Tregs has remained unexplored. Our results identify Bcl6 as an essential transcription factor regulating Gata3 activity in Tregs. Thus, Bcl6 represents a crucial regulatory layer in the Treg functional program that is required for specific suppression of Gata3 and Th2 effector responses by Tregs.


Subject(s)
DNA-Binding Proteins/immunology , GATA3 Transcription Factor/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Transcription, Genetic/immunology , Transcriptional Activation/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/immunology , MicroRNAs/metabolism , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/metabolism , Proto-Oncogene Proteins c-bcl-6 , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/cytology , Th2 Cells/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/genetics
8.
Blood ; 120(13): 2589-99, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22665934

ABSTRACT

Nuclear transcription factor Stat3 is important for proper regulation of hematopoietic stem cell (HSC) and hematopoietic progenitor cell (HPC) proliferation, survival, and cytokine signaling responses. A new, noncanonical role for Stat3 in mitochondrial function has been discovered recently. However, there is little information on the role(s) of mitochondrial Stat3 in HSC/HPC function, especially potential effects of Stat3/mitochondrial dysregulation in human diseases. We investigated hematopoietic cell-targeted deletion of the STAT3 gene in HSCs/HPCs with a focus on mitochondrial function. We found that STAT3(-/-) mice, which have a very shortened lifespan, dysfunctional/dysregulated mitochondrial function and excessive reactive oxygen species production in HSCs/HPCs that coincides with pronounced defects in function. These animals have a blood phenotype with similarities to premature aging and to human diseases of myelodysplastic syndrome and myeloproliferative neoplasms such as erythroid dysplasia, anemia, excessive myeloproliferation, and lymphomyeloid ratio shifts. We show herein that the lifespan of STAT3(-/-) animals is lengthened by treatment with a reactive oxygen species scavenger, which lessened the severity of the blood phenotype. These data suggest a need for more detailed studies of role(s) of Stat3 in HSC/HPC mitochondrial function in human diseases and raise the idea that mitochondrial Stat3 could be used as a potential therapeutic target.


Subject(s)
Aging/pathology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mitochondria/pathology , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/physiology , Acetylcysteine/pharmacology , Anemia , Animals , Bone Marrow/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Erythroid Cells/cytology , Erythroid Cells/drug effects , Female , Free Radical Scavengers/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen Consumption/drug effects , Phenotype , Sequence Deletion
9.
Blood ; 119(24): 5731-41, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22535665

ABSTRACT

In the present study, surface CD1d, which is involved in immune cell interactions, was assessed for effects on hematopoiesis. Mouse BM hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) express CD1d. The numbers and cycling status of HPCs in the BM and spleen of different strains of cd1d(-/-) mice were enhanced significantly, suggesting that CD1d is a negative regulator of HPCs. In support of this, CD1d was required for the SCF and Flt3 ligand synergistic enhancement of CSF induction of HPC colony formation and for HPC response to myelosuppressive chemokines. Colony formation by immature subsets of HPCs was greatly enhanced when normal, but not cd1d(-/-), BM cells were pretreated with CD1d Abs in vitro. These effects required the full CD1d cytoplasmic tail. In contrast, long-term, but not short-term, repopulating HSC engraftment was impaired significantly, an effect that was minimally influenced by the presence of a truncated CD1d cytoplasmic tail. Pretreatment of normal BM cells with CD1d Abs greatly enhanced their engraftment of HSCs. The results of the present study implicate CD1d in a previously unrecognized regulatory role of normal and stressed hematopoiesis.


Subject(s)
Antigens, CD1d/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Antibodies/pharmacology , Antigens, CD1d/chemistry , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Count , Cell Proliferation/drug effects , Chemokines/pharmacology , Colony-Forming Units Assay , Galactosylceramides/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Interferon-gamma/pharmacology , Membrane Proteins/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Phenotype , Protein Structure, Tertiary , Stem Cell Factor/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
10.
Stem Cells Dev ; 21(9): 1449-54, 2012 Jun 10.
Article in English | MEDLINE | ID: mdl-21943234

ABSTRACT

DEK is a biochemically distinct protein that is generally found in the nucleus, where it is vital to global heterochromatin integrity. However, DEK is also secreted by cells (eg, macrophages) and influences other adjacent cells (eg, acts as a chemoattractant for certain mature blood cells). We hypothesized that DEK may modulate functions of hematopoietic stem (HSCs) and progenitor (HPCs) cells. C57Bl/6 mice were used to demonstrate that absolute numbers and cycling status of HPCs (colony forming unit-granulocyte macrophage [CFU-GM], burst forming unit-erythroid [BFU-E], and colony forming unit-granulocyte erythroid macrophage megakaryocyte [CFU-GEMM]) in bone marrow (BM) and spleen were significantly enhanced in DEK -/- as compared with wild-type (WT) control mice. Moreover, purified recombinant DEK protein inhibited colony formation in vitro by CFU-GM, BFU-E, and CFU-GEMM from WT BM cells and human cord blood (CB) cells in a dose-dependent fashion, demonstrating that DEK plays a negative role in HPC proliferation in vitro and in vivo. Suppression was direct acting as determined by inhibition of proliferation of single isolated CD34(+) CB cells in vitro. In contrast, DEK -/- BM cells significantly demonstrated reduced long term competitive and secondary mouse repopulating HSC capacity compared with WT BM cells, demonstrating that DEK positively regulates engrafting capability of self-renewing HSCs. This demonstrates that DEK has potent effects on HSCs, HPCs, and hematopoiesis, information of biological and potential clinical interest.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Graft Survival , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Oncogene Proteins/metabolism , Animals , Cell Proliferation , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Knockout , Oncogene Proteins/genetics , Poly-ADP-Ribose Binding Proteins , Transplantation, Heterologous , Transplantation, Homologous
11.
Blood Cells Mol Dis ; 48(1): 25-9, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21983347

ABSTRACT

Several angiopoietin-like (ANGPTL) molecules have been implicated in enhancement of ex-vivo expansion of murine and human (hu) hematopoietic stem cells, but there are no reports on hematopoietic progenitor cells (HPCs). We assessed purified recombinant endotoxin-free hu ANGPTL-2 Coiled-Coil (CC), -3, -3CC, -3 fibrinogen-like domain (FLD), -4, -4CC, -5CC, -6 and -7 for effects on proliferation and survival of HPCs from hu cord blood (CB). None of the ANGPTL molecules stimulated CB HPC proliferation, or enhanced or inhibited colony formation of CB HPC stimulated by various growth factors. However, ANGPTL-2CC, -3, and -3CC significantly enhanced survival of HPC (CFU-GM, BFU-E, CFU-GEMM) subjected to delayed addition of growth factors. Survival enhancing effects of ANGPTL-3 were neutralized by purified anti-ANGPTL-3, but not by anti-ANGPTL-4, -6, or -7. ANGPTL-2CC, -3, and -3CC, but not -4, -6, or -7 also enhanced replating capacity of single CB CFU-GEMM colonies, an estimate of the self-renewal capabilities of HPCs, by greater than 2 fold. Effects of at least ANGPTL-3CC may in part be mediated through phosphorylation of ERK. The ANGPTL molecules did not influence ex-vivo expansion of hu CB CD34(+) cells, alone, or in combination with SCF, TPO, Flt3-ligand, with or without IL-3. Thus, among ANGPTL family members, ANGPTL-2 and -3 had enhancing activities on human HPC survival and replating activity, effects requiring the CC domain of the ANGPTL molecules. This information is of relevance to hu HPC regulation.


Subject(s)
Angiopoietins/genetics , Fetal Blood/drug effects , Hematopoietic Stem Cells/drug effects , Recombinant Proteins/genetics , Signal Transduction/physiology , Angiopoietin-Like Protein 2 , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins , Angiopoietins/antagonists & inhibitors , Angiopoietins/chemistry , Angiopoietins/pharmacology , Antibodies, Neutralizing/pharmacology , Antigens, CD34/biosynthesis , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Erythropoietin/pharmacology , Fetal Blood/cytology , Fetal Blood/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
12.
Stem Cells Dev ; 21(10): 1597-603, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-21936707

ABSTRACT

Cyclin dependent kinase inhibitors (CDKIs) influence proliferation of hematopoietic progenitor cells (HPCs), but little is known of how they influence proliferative responsiveness of HPCs to colony stimulating factors (CSFs), alone and in combination with other hematopoietically active factors, such as the potent co-stimulating cytokine stem cell factor (SCF), or inhibition by myelosuppressive chemokines. Using mice with deletions in p18(INK4c), p21(CIP1/WAF1), or p27(KIP1) genes, and in mice with double gene deletions for either p18/p21 or p18/p27, we determined effects of absence of these CDKIs and their interactions on functional HPC numbers in vivo, and HPC proliferative responsiveness in vitro. There is a decrease in bone marrow HPC proliferation in p18(-/-) mice commensurate with decreased numbers of HPC, suggesting a positive role for p18 on HPC in vivo, similar to that for p21. These positive effects of p18 dominate negative effects of p27 gene deletion. Moreover, the CDKIs differentially regulate responsiveness of granulocyte macrophage (GM) progenitors to synergistic cell proliferation in response to GM-CSF plus SCF, which is considered important for normal hematopoiesis. Responsiveness of HPCs to inhibition by myelosuppressive chemokines is directly related to the capacity of HPCs to respond to synergistic stimulation, and their cell cycle status. P18(INK4c) gene deletion rescued the loss of chemokine suppression of synergistic proliferation due to deletion of p21(CIP1/WAF1). These findings underscore the complex interplay of cell cycle regulators in HPC, and demonstrate that loss of one can sometimes be compensated by loss of another CDKI in both, a pro- or anti-proliferative context.


Subject(s)
Cyclin-Dependent Kinase Inhibitor Proteins/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Hematopoietic Stem Cells/physiology , Stem Cell Factor/physiology , Animals , Bone Marrow Cells/physiology , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cytokines/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Progenitor Cells/physiology , Intercellular Signaling Peptides and Proteins/physiology , Interleukin-6/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , S Phase , Spleen/cytology , Stem Cell Factor/pharmacology
13.
Blood ; 118(3): 565-75, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21628410

ABSTRACT

Neurexin I α (NRXN1α) and Dystroglycan (DAG1) are membrane receptors which serve as mutual ligands in the neuronal system. Neurexophilins (NXPHs) bind NRXN1α. NRXN1α was expressed in primitive populations in human CB (huCB) and murine BM (muBM). DAG1 is ubiquitously expressed in hematopoietic tissue; however, osteoblasts appear to be sites of very high expression within muBM. High concentrations of NXPH were found in huCB plasma and murine lineage-positive splenocytes. We evaluated effects of these molecules on huCB and muBM hematopoietic progenitor cells (HPCs) and HSCs. At both a single and population cell level in vitro, we found that NXPH1 was a potent inhibitor of HPC proliferation acting through NRXN1α an effect down-modulated by DAG1. Injection of recombinant NXPH1 in vivo resulted in myelo- and lymphosuppression in the BM, with absolute numbers and cycling status of functional and phenotypically defined HPCs dose- and time-dependently decreased. Competitive HSC transplantations showed no change in the long-term repopulating activity of HSCs from mice exposed to recombinant NXPH1. These results demonstrate the presence and function of a regulated signaling axis in hematopoiesis centered on NRXN1α and its modulation by DAG1 and NXPH1.


Subject(s)
Glycoproteins/genetics , Glycoproteins/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Neuropeptides/genetics , Neuropeptides/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Calcium-Binding Proteins , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Division/physiology , Cells, Cultured , Dystroglycans/metabolism , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoiesis/physiology , Humans , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Osteoblasts/cytology , Osteoblasts/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/physiology
14.
Blood ; 117(23): 6198-201, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21505191

ABSTRACT

The contribution of specific cell types to the production of cytokines that regulate hematopoiesis is still not well defined. We have previously identified T cell-dependent regulation of hematopoietic progenitor cell (HPC) numbers and cycling. In this report, we demonstrated that HPC activity is decreased in mice with STAT3-deficient T cells, a phenotype that is not because of decreased expression of IL-17 or RORγt. STAT3 expression in T cells was required for IL-21 production by multiple T helper subsets, and neutralization of IL-21 resulted in decreased HPC activity identical to that in mice with STAT3-deficient T cells. Importantly, injection of IL-21 rescued HPC activity in mice with STAT3-deficient T cells. Thus, STAT3-dependent IL-21 production in T cells is required for HPC homeostasis.


Subject(s)
Gene Expression Regulation/immunology , Hematopoietic Stem Cells/immunology , Homeostasis/immunology , Interleukins/immunology , STAT3 Transcription Factor/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeostasis/genetics , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukins/biosynthesis , Interleukins/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism
15.
Blood Cells Mol Dis ; 46(4): 318-20, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21411351

ABSTRACT

Since cord blood (CB) has become a commonly used source of transplantable hematopoietic stem (HSC) and hematopoietic progenitor cells (HPC), there has been a need to overcome the limited HSC and HPC numbers available to transplant from a single CB, especially for adult recipients. Our laboratory previously demonstrated that Rheb2 overexpression significantly impaired the repopulating ability of HSC. Since overexpression of Rheb2 leads to increased signaling through mTOR, we examined the effect of the mTOR inhibitor rapamycin ex vivo on cytokine expanded CD34(+) CB cells for the engraftment of these cells in non-obese diabetic, severe combined immunodeficient, IL-2 receptor γ chain null (NSG) mice. We observed significant enhancement in engraftment of the CB treated ex vivo with cytokines in the presence of rapamycin prior to transplant, effects seen in primary as well as secondary transplants. These pre-clinical results suggest a positive role for rapamycin during ex vivo culture of CB SCID repopulating cells/HSC.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Fetal Blood/drug effects , Sirolimus/pharmacology , Animals , Antigens, CD34 , Graft Survival/drug effects , Humans , Immunosuppressive Agents , Mice , Mice, SCID , Sirolimus/therapeutic use
16.
Blood ; 117(18): 4773-7, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21393480

ABSTRACT

Cryopreservation of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) is crucial for cord blood (CB) banking and transplantation. We evaluated recovery of functional HPC cryopreserved as mononuclear or unseparated cells for up to 23.5 years compared with prefreeze values of the same CB units. Highly efficient recovery (80%-100%) was apparent for granulocyte-macrophage and multipotential hematopoietic progenitors, although some collections had reproducible low recovery. Proliferative potential, response to multiple cytokines, and replating of HPC colonies was extensive. CD34(+) cells isolated from CB cryopreserved for up to 21 years had long-term (≥ 6 month) engrafting capability in primary and secondary immunodeficient mice reflecting recovery of long-term repopulating, self-renewing HSCs. We recovered functionally responsive CD4(+) and CD8(+) T lymphocytes, generated induced pluripotent stem (iPS) cells with differentiation representing all 3 germ cell lineages in vitro and in vivo, and detected high proliferative endothelial colony forming cells, results of relevance to CB biology and banking.


Subject(s)
Blood Preservation , Cryopreservation , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Colony-Forming Units Assay , Endothelial Cells/cytology , Fetal Blood/transplantation , Hematopoietic Stem Cell Transplantation , Humans , In Vitro Techniques , Induced Pluripotent Stem Cells/transplantation , Infant, Newborn , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Time Factors , Transplantation, Heterologous
17.
Blood ; 117(21): 5643-51, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21447833

ABSTRACT

Intracellular factors are involved in and essential for hematopoiesis. HIV-1 Tat-interacting protein of 110 kDa (TIP110; p110(nrb)/SART3/p110) is an RNA-binding nuclear protein implicated in the regulation of HIV-1 gene and host gene transcription, pre-mRNA splicing, and cancer immunology. In the present study, we demonstrate a role for TIP110 in the regulation of hematopoiesis. TIP110 was expressed in human CD34(+) cells and decreased with differentiation of CD34(+) cells. TIP110 mRNA was also expressed in phenotyped mouse marrow hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). Using TIP110 transgenic (TIP110(TG)) and haploinsufficient (TIP110(+/-)) mice, we found that increased TIP110 expression enhanced HPC numbers, survival, and cell cycling, whereas decreased TIP110 expression had the opposite effects. Moreover, TIP110(+/-) bone marrow HPCs responded more effectively, and TIP110(TG) HPCs less effectively, than those of wild-type control mice to recovery from the cell-cycle-active drug 5-fluorouracil (5-FU). Unexplained sex differences were noted in HSC competitive repopulating ability, but not HPC numbers, in TIP110(TG) mice. Intracellularly, TIP110 regulated CMYC and GATA2 expression at the transcriptional level, and TIP110 and CMYC reciprocally regulated the expression of each other. These results demonstrate a role for TIP110 in the regulation of hematopoiesis, effects that are likely linked to TIP110 regulation of CMYC.


Subject(s)
Antigens, Neoplasm/physiology , Bone Marrow/metabolism , Gene Expression Regulation , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Proto-Oncogene Proteins c-myc/genetics , RNA-Binding Proteins/physiology , Animals , Antimetabolites, Antineoplastic/pharmacology , Blotting, Western , Bone Marrow/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Female , Fetal Blood/metabolism , Fluorouracil/pharmacology , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
Exp Hematol ; 39(4): 415-23, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21216274

ABSTRACT

OBJECTIVE: Cell-cycle checkpoints guarantee movement through the cell cycle. Mitotic arrest deficiency 2 (Mad2), a mitotic checkpoint protein, appears crucial for generating the wait anaphase signal to prevent onset of anaphase. We evaluated effects of Mad2 haploinsufficiency on hematopoietic stem (HSC) and progenitor (HPC) function in response to stress. MATERIALS AND METHODS: We studied effects of Mad2(+/-) on in vivo recovery of bone marrow HPC from cytotoxic effects and also effects of cytostatic agents on HPC growth in vitro using Mad2(+/-) mice. RESULTS: Mad2(+/-) HPCs were protected from cytotoxic effects in vivo of a cell-cycle-specific agent, Ara-C, events consistent with Mad2(+/-) HPCs being in a slow or noncycling state, but not from recovery of functional HPC after treatment with non-cycle-specific cyclophosphamide or sublethal irradiation. There were no differences in phenotyped HSCs in Mad2(+/-) &Mad2(+/+) mice, information confirmed by no changes in short- or long-term repopulating HSC assay. To better understand Mad2(+/-) HPC function, E3330, a cytostatic agent, was used to assess redox function of Ape1/Ref-1; colony growth was examined under 5% and 20% O(2) tension. Mad2(+/-) HPCs were less responsive to E3330 than Mad2(+/+) HPCs, and E3330 was more effective under lowered O(2) tension. Mad2(+/-) HPCs were not enhanced at lowered oxygen, as were Mad2(+/+) HPCs. CONCLUSIONS: Our studies have unexpectedly found that Mad2 haploinsufficiency is protective in the presence of a cycle-specific DNA synthesis agent in vivo, and Ape1/Ref-1 inhibitor in vitro.


Subject(s)
Cell Cycle Proteins/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Haploinsufficiency , Hematopoietic Stem Cells/metabolism , Animals , Benzoquinones/pharmacology , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Colony-Forming Units Assay , Cyclophosphamide/pharmacology , Cytarabine/pharmacology , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Immunosuppressive Agents/pharmacology , Mad2 Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction/drug effects , Oxygen/pharmacology , Propionates/pharmacology
19.
J Clin Invest ; 120(12): 4273-88, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21084750

ABSTRACT

The development of effective cancer immunotherapies has been consistently hampered by several factors, including an inability to instigate long-term effective functional antitumor immunity. This is particularly true for immunotherapies that focus on the adoptive transfer of activated or genetically modified mature CD8+ T cells. In this study, we sought to alter and enhance long-term host immunity by genetically modifying, then transplanting, mouse HSCs. We first cloned a previously identified tumor-reactive HLA-DR4-restricted CD4+ TCR specific for the melanocyte differentiation antigen tyrosinase-related protein 1 (Tyrp1), then constructed both a high-expression lentivirus vector and a TCR-transgenic mouse expressing the genes encoding this TCR. Using these tools, we demonstrated that both mouse and human HSCs established durable, high-efficiency TCR gene transfer following long-term transplantation into lethally irradiated mice transgenic for HLA-DR4. Recipients of genetically modified mouse HSCs developed spontaneous autoimmune vitiligo that was associated with the presence of a Th1-polarized memory effector CD4+ T cell population that expressed the Tyrp1-specific TCR. Most importantly, large numbers of CD4+ T cells expressing the Tyrp1-specific TCR were detected in secondary HLA-DR4-transgenic transplant recipients, and these mice were able to destroy subcutaneously administered melanoma cells without the aid of vaccination, immune modulation, or cytokine administration. These results demonstrate the creation of what we believe to be a novel translational model of durable lentiviral gene transfer that results in long-term effective immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hematopoietic Stem Cell Transplantation , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Receptors, Antigen, T-Cell/genetics , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmunity , Cell Line, Tumor , HLA-DR4 Antigen/metabolism , Humans , Immunotherapy , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Transduction, Genetic , Vitiligo/genetics , Vitiligo/immunology
20.
Blood ; 114(16): 3392-401, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19690340

ABSTRACT

Molecular mechanisms preserving hematopoietic stem cell (HSC) self-renewal by maintaining a balance between proliferation, differentiation, and other processes are not fully understood. Hyperactivation of the mammalian target of rapamycin (mTOR) pathway, causing sustained proliferative signals, can lead to exhaustion of HSC repopulating ability. We examined the role of the novel ras gene Rheb2, an activator of the mTOR kinase, in colony-forming ability, survival, and repopulation of immature mouse hematopoietic cells. In a cell line model of mouse hematopoietic progenitor cells (HPCs), we found enhanced proliferation and mTOR signaling in cells overexpressing Rheb2. In addition, overexpression of Rheb2 enhanced colony-forming ability and survival of primary mouse bone marrow HPCs. Expansion of phenotypic HSCs in vitro was enhanced by Rheb2 overexpression. Consistent with these findings, Rheb2 overexpression transiently expanded phenotypically defined immature hematopoietic cells after in vivo transplantation; however, these Rheb2-transduced cells were significantly impaired in overall repopulation of primary and secondary congenic transplantation recipients. Our findings suggest that HPCs and HSCs behave differently in response to growth-promoting signals stimulated by Rheb2. These results may have value in elucidating mechanisms controlling the balance between proliferation and repopulating ability, a finding of importance in clinical uses of HPCs/HSCs.


Subject(s)
Cell Proliferation , Cell Survival/physiology , Gene Expression , Hematopoietic Stem Cells/metabolism , Monomeric GTP-Binding Proteins/biosynthesis , Neuropeptides/biosynthesis , Signal Transduction , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Hematopoietic Stem Cell Transplantation , Humans , Mice , Models, Biological , Monomeric GTP-Binding Proteins/genetics , Neuropeptides/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Ras Homolog Enriched in Brain Protein , TOR Serine-Threonine Kinases , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...