Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Am Soc Nephrol ; 24(8): 1263-73, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23744888

ABSTRACT

The renin-angiotensin-aldosterone system (RAAS) regulates BP and salt-volume homeostasis. Juxtaglomerular (JG) cells synthesize and release renin, which is the first and rate-limiting step in the RAAS. Intense pathologic stresses cause a dramatic increase in the number of renin-producing cells in the kidney, termed JG cell recruitment, but how this occurs is not fully understood. Here, we isolated renal CD44(+) mesenchymal stem cell (MSC)-like cells and found that they differentiated into JG-like renin-expressing cells both in vitro and in vivo. Sodium depletion and captopril led to activation and differentiation of these cells into renin-expressing cells in the adult kidney. In summary, CD44(+) MSC-like cells exist in the adult kidney and can differentiate into JG-like renin-producing cells under conditions that promote JG cell recruitment.


Subject(s)
Adult Stem Cells/metabolism , Captopril/pharmacology , Cell Differentiation/physiology , Juxtaglomerular Apparatus/cytology , Kidney/cytology , Mesenchymal Stem Cells/metabolism , Renin-Angiotensin System/physiology , Renin/metabolism , Animals , Cell Differentiation/drug effects , Juxtaglomerular Apparatus/metabolism , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Renin-Angiotensin System/drug effects
2.
PLoS One ; 5(6): e11056, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20585403

ABSTRACT

Medications that can mitigate against radiation injury are limited. In this study, we investigated the ability of recombinant human growth hormone (rhGH) to mitigate against radiation injury in mice and nonhuman primates. BALB/c mice were irradiated with 7.5 Gy and treated post-irradiation with rhGH intravenously at a once daily dose of 20 microg/dose for 35 days. rhGH protected 17 out of 28 mice (60.7%) from lethal irradiation while only 3 out of 28 mice (10.7%) survived in the saline control group. A shorter course of 5 days of rhGH post-irradiation produced similar results. Compared with the saline control group, treatment with rhGH on irradiated BALB/c mice significantly accelerated overall hematopoietic recovery. Specifically, the recovery of total white cells, CD4 and CD8 T cell subsets, B cells, NK cells and especially platelets post radiation exposure were significantly accelerated in the rhGH-treated mice. Moreover, treatment with rhGH increased the frequency of hematopoietic stem/progenitor cells as measured by flow cytometry and colony forming unit assays in bone marrow harvested at day 14 after irradiation, suggesting the effects of rhGH are at the hematopoietic stem/progenitor level. rhGH mediated the hematopoietic effects primarily through their niches. Similar data with rhGH were also observed following 2 Gy sublethal irradiation of nonhuman primates. Our data demonstrate that rhGH promotes hematopoietic engraftment and immune recovery post the exposure of ionizing radiation and mitigates against the mortality from lethal irradiation even when administered after exposure.


Subject(s)
Growth Hormone/administration & dosage , Radiation Injuries, Experimental/prevention & control , Animals , Apoptosis/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Primates , Recombinant Proteins/administration & dosage , T-Lymphocyte Subsets
3.
Nat Med ; 16(4): 475-82, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20305662

ABSTRACT

Hematopoietic stem cell (HSC) self-renewal is regulated by both intrinsic and extrinsic signals. Although some of the pathways that regulate HSC self-renewal have been uncovered, it remains largely unknown whether these pathways can be triggered by deliverable growth factors to induce HSC growth or regeneration. Here we show that pleiotrophin, a neurite outgrowth factor with no known function in hematopoiesis, efficiently promotes HSC expansion in vitro and HSC regeneration in vivo. Treatment of mouse bone marrow HSCs with pleiotrophin caused a marked increase in long-term repopulating HSC numbers in culture, as measured in competitive repopulating assays. Treatment of human cord blood CD34(+)CDCD38(-)Lin(-) cells with pleiotrophin also substantially increased severe combined immunodeficient (SCID)-repopulating cell counts in culture, compared to input and cytokine-treated cultures. Systemic administration of pleiotrophin to irradiated mice caused a pronounced expansion of bone marrow stem and progenitor cells in vivo, indicating that pleiotrophin is a regenerative growth factor for HSCs. Mechanistically, pleiotrophin activated phosphoinositide 3-kinase (PI3K) signaling in HSCs; antagonism of PI3K or Notch signaling inhibited pleiotrophin-mediated expansion of HSCs in culture. We identify the secreted growth factor pleiotrophin as a new regulator of both HSC expansion and regeneration.


Subject(s)
Carrier Proteins/physiology , Cytokines/physiology , Hematopoietic Stem Cells/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Carrier Proteins/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/pharmacology , Dose-Response Relationship, Drug , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, SCID , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Receptors, Notch/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cell Factor/pharmacology , Thrombopoietin/pharmacology
4.
Stem Cells ; 28(3): 523-34, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20054864

ABSTRACT

Hematopoietic stem cells (HSCs) are enriched for aldehyde dehydrogenase (ALDH) activity and ALDH is a selectable marker for human HSCs. However, the function of ALDH in HSC biology is not well understood. We sought to determine the function of ALDH in regulating HSC fate. Pharmacologic inhibition of ALDH with diethylaminobenzaldehyde (DEAB) impeded the differentiation of murine CD34(-)c-kit(+)Sca-1(+)lineage(-) (34(-)KSL) HSCs in culture and facilitated a ninefold expansion of cells capable of radioprotecting lethally irradiated mice compared to input 34(-)KSL cells. Treatment of bone marrow (BM) 34(-)KSL cells with DEAB caused a fourfold increase in 4-week competitive repopulating units, verifying the amplification of short-term HSCs (ST-HSCs) in response to ALDH inhibition. Targeted siRNA of ALDH1a1 in BM HSCs caused a comparable expansion of radioprotective progenitor cells in culture compared to DEAB treatment, confirming that ALDH1a1 was the target of DEAB inhibition. The addition of all trans retinoic acid blocked DEAB-mediated expansion of ST-HSCs in culture, suggesting that ALDH1a1 regulates HSC differentiation via augmentation of retinoid signaling. Pharmacologic inhibition of ALDH has therapeutic potential as a means to amplify ST-HSCs for transplantation purposes.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Cell Proliferation/drug effects , Cytoprotection/physiology , Enzyme Inhibitors/pharmacology , Hematopoietic Stem Cells/enzymology , Stem Cell Transplantation/methods , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Cytoprotection/drug effects , Enzyme Inhibitors/therapeutic use , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Humans , Mice , Mice, Congenic , Mice, Inbred C57BL , RNA, Small Interfering/genetics , Radiation, Ionizing , Retinal Dehydrogenase , Signal Transduction/drug effects , Signal Transduction/physiology , Tretinoin/metabolism , Tretinoin/pharmacology , p-Aminoazobenzene/analogs & derivatives , p-Aminoazobenzene/pharmacology , p-Aminoazobenzene/therapeutic use
5.
J Nanosci Nanotechnol ; 9(12): 7359-63, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19908788

ABSTRACT

The exposure of cancer cells to ionizing radiation results in potentially lethal DNA lesions. For this reason, identification and quantification of various lesions have intensively been investigated. It has also been anticipated that DNA lesions may affect not only the chemical but also the mechanical integrity of the double helix. However, the relationship between DNA damage and mechanics has not been studied. Here, the mechanical properties of DNA damaged by ionizing radiation are examined at a single-molecule level by stretching lambda-phage DNA molecules that have been exposed to gamma-radiation. A simple-stretching method using Atomic Force Microscopy (AFM) not only identifies the types of DNA lesions but also provides information about the mechanical instability of damaged DNA against intact DNA. The results include the elastic properties of damaged DNA with single strand breaks (SSBs), double strand breaks (DSBs), and multiple-lesion clusters. The elasticity of irradiated DNA is changed compared to that of intact DNA. Specifically, consecutive stretching cycles of DNA containing multiple SSBs progressively shorten the width of the overstretching B-S transition. This originates from force-induced melting off of single-stranded DNA fragments, which upon consecutive stretching cycles converts the double helix into a hybrid structure with a growing number of single stranded gaps. Closely spaced SSBs on opposite strands, upon stretching, result in a rupture of the double helix at a decreased force of approximately 200 pN and other clustered lesions result in lowering the force at which force-induced melting of the double helix occurs. Taken together, our results suggest that single-molecule force spectroscopy may become a useful nanoscale DNA diagnostic tool.


Subject(s)
DNA Damage/physiology , DNA/chemistry , DNA/radiation effects , Nanostructures/chemistry , Nanostructures/ultrastructure , DNA/ultrastructure , Elastic Modulus , Gamma Rays , Microscopy, Atomic Force/methods , Radiation Dosage , Stress, Mechanical
6.
Blood ; 113(9): 2104-7, 2009 Feb 26.
Article in English | MEDLINE | ID: mdl-19141867

ABSTRACT

Hematopoietic stem cells (HSCs) reside in association with bone marrow (BM) sinusoidal vessels in vivo, but the function of BM endothelial cells (ECs) in regulating hematopoiesis is unclear. We hypothesized that hematopoietic regeneration following injury is regulated by BM ECs. BALB/c mice were treated with total body irradiation (TBI) and then infused with C57Bl6-derived endothelial progenitor cells (EPCs) to augment endogenous BM EC activity. TBI caused pronounced disruption of the BM vasculature, BM hypocellularity, ablation of HSCs, and pancytopenia in control mice, whereas irradiated, EPC-treated mice displayed accelerated recovery of BM sinusoidal vessels, BM cellularity, peripheral blood white blood cells (WBCs), neutrophils, and platelets, and a 4.4-fold increase in BM HSCs. Systemic administration of anti-VE-cadherin antibody significantly delayed hematologic recovery in both EPC-treated mice and irradiated, non-EPC-treated mice compared with irradiated controls. These data demonstrate that allogeneic EPC infusions can augment hematopoiesis and suggest a relationship between BM microvascular recovery and hematopoietic reconstitution in vivo.


Subject(s)
Endothelial Cells/transplantation , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Stem Cell Transplantation , Animals , Blood Cell Count , Cell Differentiation/physiology , Endothelial Cells/physiology , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/radiation effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recovery of Function , Stem Cells/physiology , Whole-Body Irradiation/adverse effects
7.
Mol Endocrinol ; 23(2): 188-201, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19106195

ABSTRACT

The retinoid X receptor (RXR) contributes to the regulation of diverse biological pathways via its role as a heterodimeric partner of several nuclear receptors. However, RXR has no established role in the regulation of hematopoietic stem cell (HSC) fate. In this study, we sought to determine whether direct modulation of RXR signaling could impact human HSC self-renewal or differentiation. Treatment of human CD34(+)CD38(-)lin(-) cells with LG1506, a selective RXR modulator, inhibited the differentiation of HSCs in culture and maintained long-term repopulating HSCs in culture that were otherwise lost in response to cytokine treatment. Further studies revealed that LG1506 had a distinct mechanism of action in that it facilitated the recruitment of corepressors to the retinoic acid receptor (RAR)/RXR complex at target gene promoters, suggesting that this molecule was functioning as an inverse agonist in the context of this heterodimer. Interestingly, using combinatorial peptide phage display, we identified unique surfaces presented on RXR when occupied by LG1506 and demonstrated that other modulators that exhibited these properties functioned similarly at both a mechanistic and biological level. These data indicate that the RAR/RXR heterodimer is a critical regulator of human HSC differentiation, and pharmacological modulation of RXR signaling prevents the loss of human HSCs that otherwise occurs in short-term culture.


Subject(s)
Hematopoietic Stem Cells/physiology , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/metabolism , Signal Transduction/physiology , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/metabolism , Animals , Benzoates/metabolism , Biomarkers/metabolism , Cell Cycle/drug effects , Cell Differentiation , Cell Lineage , Cells, Cultured , Chromans/metabolism , Dimerization , Fatty Acids, Unsaturated/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Phenyl Ethers/pharmacology , Protein Conformation , Retinoid X Receptors/chemistry , Retinoid X Receptors/genetics , Retinoids/metabolism
8.
PLoS One ; 3(4): e1912, 2008 Apr 02.
Article in English | MEDLINE | ID: mdl-18382685

ABSTRACT

BACKGROUND: Previous work has demonstrated the potential for peripheral blood (PB) gene expression profiling for the detection of disease or environmental exposures. METHODS AND FINDINGS: We have sought to determine the impact of several variables on the PB gene expression profile of an environmental exposure, ionizing radiation, and to determine the specificity of the PB signature of radiation versus other genotoxic stresses. Neither genotype differences nor the time of PB sampling caused any lessening of the accuracy of PB signatures to predict radiation exposure, but sex difference did influence the accuracy of the prediction of radiation exposure at the lowest level (50 cGy). A PB signature of sepsis was also generated and both the PB signature of radiation and the PB signature of sepsis were found to be 100% specific at distinguishing irradiated from septic animals. We also identified human PB signatures of radiation exposure and chemotherapy treatment which distinguished irradiated patients and chemotherapy-treated individuals within a heterogeneous population with accuracies of 90% and 81%, respectively. CONCLUSIONS: We conclude that PB gene expression profiles can be identified in mice and humans that are accurate in predicting medical conditions, are specific to each condition and remain highly accurate over time.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Animals , Endotoxins/metabolism , Environmental Exposure , Female , Humans , Lipopolysaccharides/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Radiation Tolerance , Radiation, Ionizing , Reproducibility of Results
9.
Small ; 4(2): 288-94, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18247386

ABSTRACT

The detection and quantification of ionizing radiation damage to DNA at a single-molecule level by atomic force microscopy (AFM) is reported. The DNA damage-detection technique combining supercoiled plasmid relaxation assay with AFM imaging is a direct and quantitative approach to detect gamma-ray-induced single- and double-strand breaks in DNA, and its accuracy and reliability are validated through a comparison with traditional agarose gel electrophoresis. In addition, the dependence of radiation-induced single-strand breaks on plasmid size and concentration at a single-molecule level in a low-dose (1 Gy) and low-concentration range (0.01 ng microL(-1)-10 ng microL(-1)) is investigated using the AFM-based damage-detection assay. The results clearly show that the number of single-strand breaks per DNA molecule is linearly proportional to the plasmid size and inversely correlated to the DNA concentration. This assay can also efficiently detect DNA damage in highly dilute samples (0.01 ng microL(-1)), which is beyond the capability of traditional techniques. AFM imaging can uniquely supplement traditional techniques for sensitive measurements of damage to DNA by ionizing radiation.


Subject(s)
DNA Damage , DNA/radiation effects , DNA/ultrastructure , Microscopy, Atomic Force/methods , DNA/isolation & purification , DNA Breaks, Single-Stranded/radiation effects , DNA, Superhelical/isolation & purification , DNA, Superhelical/radiation effects , DNA, Superhelical/ultrastructure , Dose-Response Relationship, Radiation , Electrophoresis, Agar Gel/methods , Gamma Rays/adverse effects , Nanotechnology , Plasmids/isolation & purification , Plasmids/radiation effects , Plasmids/ultrastructure
10.
PLoS Med ; 4(4): e106, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17407386

ABSTRACT

BACKGROUND: The capacity to assess environmental inputs to biological phenotypes is limited by methods that can accurately and quantitatively measure these contributions. One such example can be seen in the context of exposure to ionizing radiation. METHODS AND FINDINGS: We have made use of gene expression analysis of peripheral blood (PB) mononuclear cells to develop expression profiles that accurately reflect prior radiation exposure. We demonstrate that expression profiles can be developed that not only predict radiation exposure in mice but also distinguish the level of radiation exposure, ranging from 50 cGy to 1,000 cGy. Likewise, a molecular signature of radiation response developed solely from irradiated human patient samples can predict and distinguish irradiated human PB samples from nonirradiated samples with an accuracy of 90%, sensitivity of 85%, and specificity of 94%. We further demonstrate that a radiation profile developed in the mouse can correctly distinguish PB samples from irradiated and nonirradiated human patients with an accuracy of 77%, sensitivity of 82%, and specificity of 75%. Taken together, these data demonstrate that molecular profiles can be generated that are highly predictive of different levels of radiation exposure in mice and humans. CONCLUSIONS: We suggest that this approach, with additional refinement, could provide a method to assess the effects of various environmental inputs into biological phenotypes as well as providing a more practical application of a rapid molecular screening test for the diagnosis of radiation exposure.


Subject(s)
Environmental Exposure , Gene Expression Profiling , Gene Expression/radiation effects , Genes/radiation effects , Radiation, Ionizing , Animals , Cyclophosphamide/pharmacology , DNA/radiation effects , DNA Damage , Dose-Response Relationship, Radiation , Female , Gene Regulatory Networks/radiation effects , Humans , Leukocytes, Mononuclear/radiation effects , Mass Screening , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Particle Accelerators , Radiation Dosage , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/genetics , Sensitivity and Specificity , Single-Blind Method , Species Specificity , Transplantation Conditioning , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , Whole-Body Irradiation/adverse effects
11.
Blood ; 109(6): 2365-72, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17095624

ABSTRACT

Flk-1(+) endothelial progenitors contribute critically to the definitive onset of hematopoiesis during embryogenesis. Recent studies have suggested that adult sources of endothelial cells also possess hematopoietic activity. In this study, we sought to determine whether transplantation of primary vascular endothelial cells (ECs) could enhance the hematopoietic recovery and survival of irradiated mice. C57Bl6 mice were exposed to sublethal and lethal doses of irradiation and were subsequently given transplants of either primary murine brain-derived ECs (MBECs) or fetal blood-derived ECs (FBECs). Mice that received a transplant with MBECs alone demonstrated accelerated BM cellular recovery, radioprotection of BM c-kit(+)sca-1(-)lin(-) progenitors and enhanced regeneration of c-kit(+)sca-1(+)lin(-) (KSL) stem/progenitor cells following irradiation compared with controls. MBEC transplantation also facilitated the recovery of circulating white blood cell and platelet counts following radiation exposure. Remarkably, 57% of mice that received a transplant with MBECs alone survived long term following 1050 cGy exposure, which was 100% lethal in control mice. FBEC transplantation was also associated with increased survival compared with controls, although these mice did not survive in the long term. These data suggest that reestablishment of endothelial cell activity can improve the hematopoietic recovery and survival of irradiated mice.


Subject(s)
Cell Transplantation , Endothelial Cells/cytology , Hematopoiesis , Animals , Brain/cytology , Brain/metabolism , Cells, Cultured , Cytokines/biosynthesis , Endothelial Cells/metabolism , Gene Expression , Hematopoiesis/radiation effects , Mice , Stem Cells/cytology , Survival Rate
12.
Proc Natl Acad Sci U S A ; 103(31): 11707-12, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16857736

ABSTRACT

Aldehyde dehydrogenase (ALDH) is an enzyme that is expressed in the liver and is required for the conversion of retinol (vitamin A) to retinoic acids. ALDH is also highly enriched in hematopoietic stem cells (HSCs) and is considered a selectable marker of human HSCs, although its contribution to stem cell fate remains unknown. In this study, we demonstrate that ALDH is a key regulator of HSC differentiation. Inhibition of ALDH with diethylaminobenzaldehyde (DEAB) delayed the differentiation of human HSCs that otherwise occurred in response to cytokines. Moreover, short-term culture with DEAB caused a 3.4-fold expansion in the most primitive assayable human cells, the nonobese diabetic/severe combined immunodeficiency mouse repopulating cells, compared with day 0 CD34(+)CD38(-)lin(-) cells. The effects of DEAB on HSC differentiation could be reversed by the coadministration of the retinoic acid receptor agonist, all-trans-retinoic acid, suggesting that the ability of ALDH to generate retinoic acids is important in determining HSC fate. DEAB treatment also caused a decrease in retinoic acid receptor-mediated signaling within human HSCs, suggesting directly that inhibition of ALDH promotes HSC self-renewal via reduction of retinoic acid activity. Modulation of ALDH activity and retinoid signaling is a previously unrecognized and effective strategy to amplify human HSCs.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Cell Differentiation/physiology , Hematopoietic Stem Cells/physiology , Isoenzymes/antagonists & inhibitors , Retinoids/metabolism , Signal Transduction/physiology , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Antigens, CD/metabolism , Cells, Cultured , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Humans , Isoenzymes/metabolism , Mice , Mice, SCID , Retinal Dehydrogenase , Transcription Factors/metabolism , p-Aminoazobenzene/analogs & derivatives
13.
Biol Blood Marrow Transplant ; 12(5): 530-40, 2006 May.
Article in English | MEDLINE | ID: mdl-16635788

ABSTRACT

The risk of terrorism with nuclear or radiologic weapons is considered to be high over the coming decade. Ionizing radiation can cause a spectrum of hematologic toxicities, from mild myelosuppression to myeloablation and death. However, the potential regenerative capacity of human hematopoietic stem cells (HSCs) after radiation injury has not been well characterized. In this study, we sought to characterize the effects of ionizing radiation on human HSCs and to determine whether signals from vascular endothelial cells could promote the repair of irradiated HSCs. Exposure of human bone marrow CD34+ cells to 400 cGy caused a precipitous decline in hematopoietic progenitor cell content and primitive cells capable of repopulating nonobese diabetic/severe combined immunodeficient mice (SCID-repopulating cells), which was not retrievable via treatment with cytokines. Conversely, culture of 400 cGy-irradiated bone marrow CD34+ cells with endothelial cells under noncontact conditions supported the differential recovery of both viable progenitor cells and primitive SCID-repopulating cells. These data illustrate that vascular endothelial cells produce soluble factors that promote the repair and functional recovery of HSCs after radiation injury and suggest that novel factors with radiotherapeutic potential can be identified within this milieu.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Hematopoietic Stem Cells/radiation effects , Radiation Injuries/physiopathology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/radiation effects , Cells, Cultured/cytology , Cells, Cultured/radiation effects , Cells, Cultured/transplantation , Coculture Techniques , Colony-Forming Units Assay , Cytokines/pharmacology , Cytokines/therapeutic use , Fetal Blood/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Humans , Immunophenotyping , Mice , Mice, Inbred NOD , Mice, SCID , Transplantation, Heterologous
14.
Stem Cells ; 24(5): 1315-27, 2006 May.
Article in English | MEDLINE | ID: mdl-16373696

ABSTRACT

Recent progress has been made in the identification of the osteoblastic cellular niche for hematopoietic stem cells (HSCs) within the bone marrow (BM). Attempts to identify the soluble factors that regulate HSC self-renewal have been less successful. We have demonstrated that primary human brain endothelial cells (HUBECs) support the ex vivo amplification of primitive human BM and cord blood cells capable of repopulating non-obese diabetic/severe combined immunodeficient repopulating (SCID) mice (SCID repopulating cells [SRCs]). In this study, we sought to characterize the soluble hematopoietic activity produced by HUBECs and to identify the growth factors secreted by HUBECs that contribute to this HSC-supportive effect. Extended noncontact HUBEC cultures supported an eight-fold increase in SRCs when combined with thrombopoietin, stem cell factor, and Flt-3 ligand compared with input CD34(+) cells or cytokines alone. Gene expression analysis of HUBEC biological replicates identified 65 differentially expressed, nonredundant transcripts without annotated hematopoietic activity. Gene ontology studies of the HUBEC transcriptome revealed a high concentration of genes encoding extracellular proteins with cell-cell signaling function. Functional analyses demonstrated that adrenomedullin, a vasodilatory hormone, synergized with stem cell factor and Flt-3 ligand to induce the proliferation of primitive human CD34(+)CD38(-)lin(-) cells and promoted the expansion of CD34(+) progenitors in culture. These data demonstrate the potential of primary HUBECs as a reservoir for the discovery of novel secreted proteins that regulate human hematopoiesis.


Subject(s)
Adrenomedullin/metabolism , Brain/cytology , Endothelial Cells/cytology , Endothelial Growth Factors/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Adrenomedullin/genetics , Adrenomedullin/pharmacology , Animals , Antigens, CD34/biosynthesis , Brain/blood supply , Brain/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Fetal Blood/cytology , Fetal Blood/metabolism , Gene Expression , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Solubility , Vasodilator Agents/metabolism , Vasodilator Agents/pharmacology
15.
Blood ; 105(2): 576-83, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15345596

ABSTRACT

The CD34(+)CD38- phenotype identifies a population in the bone marrow that is enriched in the steady state for hematopoietic stem cells (HSCs). Following ex vivo culture of CD34(+) cells, HSC content is difficult to measure since committed CD34(+)CD38+ progenitors down-regulate CD38 surface expression during culture. In this study, we sought to define the phenotype of human HSCs following ex vivo culture under conditions that support the expansion of human cells capable of repopulating non-obese diabetic/severe combined immunodeficiency (SCID)-repopulating cells (SRCs). Contact coculture of fluorescence-activated cell sorter (FACS)-sorted bone marrow (BM) CD34(+)CD38- cells with human brain endothelial cells (HUBECs) supported a 4.4-fold increase in CD34(+)CD38- cells with a concordant 3.6-fold increase in SRCs over 7 days. Noncontact HUBEC cultures and the addition of thrombopoietin, stem cell factor (SCF), and macrophage colony stimulating factor I receptor (Fms)-like tyrosine kinase 3 (Flt-3) ligand supported further increases in CD34(+)CD38- cells (6.4-fold and 13.1-fold), which correlated with significant increases in SRC activity. Moreover, cell-sorting studies performed on HUBEC-cultured populations demonstrated that SRCs were significantly enriched within the CD34(+)CD38- subset compared with the CD34(-)CD38- population after culture. These results indicate that human HSCs can be identified and characterized by phenotype following expansion culture. These studies also demonstrate that HUBEC-elaborated soluble factors mediate a unique and potent expansion of human HSCs.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Brain/cytology , Endothelial Cells/cytology , Endothelial Cells/metabolism , ADP-ribosyl Cyclase/metabolism , ADP-ribosyl Cyclase 1 , Animals , Antigens, CD/metabolism , Antigens, CD34/metabolism , Cell Communication , Cell Division , Cells, Cultured , Coculture Techniques , Flow Cytometry , Humans , Membrane Glycoproteins , Mice , Mice, Inbred NOD , Mice, SCID , Solubility , Stem Cells/cytology , Stem Cells/metabolism
16.
Stem Cells ; 22(2): 202-15, 2004.
Article in English | MEDLINE | ID: mdl-14990859

ABSTRACT

Initial clinical trials examining the transplantation of ex vivo expanded cord blood (CB) cells have failed to demonstrate an impact on hematopoietic recovery compared with historical unmanipulated CB controls. In this study, we tested whether coculture with primary human brain endothelial cells (HUBECs) could increase the engraftment capacity and repopulating cell frequency within CB CD34+ cells. Quantitative analysis demonstrated that HUBEC coculture for 7 days supported a 19-fold greater number of CD34+ cells and 3.4-fold and 2.6-fold greater severe combined immunodeficient (SCID)-repopulating cell (SRC) frequencies than fresh CB CD34+ cells and liquid suspension-cultured cells. Mice transplanted with day-14 HUBEC-cultured cells showed 4.2-fold higher levels of human engraftment than mice transplanted with day-7 HUBEC-cultured cells, indicating that SRC enrichment continued to occur through day 14. Noncontact HUBEC cultures also maintained SRCs at levels comparable with contact HUBEC cultures, demonstrating that HUBEC-secreted soluble factors critically supported SRC self-renewal. Seeding efficiency studies demonstrated that HUBEC-cultured CB CD34+ cells engrafted nonobese diabetic/SCID marrow at significantly higher levels than either fresh CB CD34+ cells or liquid suspension-cultured CD34+ cells. These studies indicate that the application of HUBEC coculture or HUBEC-conditioned media can potentially improve upon current strategies for the clinical expansion of CB stem cells.


Subject(s)
Bone Marrow/metabolism , Endothelium/metabolism , Graft Survival , Stem Cell Transplantation , Animals , Antigens, CD34/metabolism , Brain/metabolism , Cells, Cultured , Fetal Blood/transplantation , Flow Cytometry , Humans , Mice
17.
Exp Hematol ; 32(3): 308-17, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15003317

ABSTRACT

OBJECTIVE: High-dose ionizing radiation can cause lethal myeloablation in exposed individuals. We examined whether ex vivo culture could rescue hematopoietic stem cells with repopulating capacity following harvest from lethally irradiated animals. METHODS: We exposed B6.SJL mice to 1050 cGy, harvested their irradiated bone marrow (BM), and examined whether ex vivo culture of the irradiated BM mononuclear cells (MNC) with porcine microvascular endothelial cells (PMVEC) or cytokines alone could rescue hematopoietic cells with in vitro colony-forming activity, in vivo radioprotective capacity, and long-term repopulating potential. RESULTS: PMVEC coculture supported the recovery of fourfold and 80-fold greater numbers of total cells and colony-forming cells (CFC) compared to cyokines alone following 1050 cGy irradiation. All control mice irradiated with 1050 cGy died by day 30, as did mice transplanted with 1050 cGy-irradiated BM MNC. In contrast, transplantation of 1050 cGy-irradiated/PMVEC-cultured BM was fully radioprotective in 12 of 16 recipient mice (75%) exposed to 1050 cGy. Six of the 12 CD45.2+ mice (50%) transplanted with 1050 cGy-irradiated/PMVEC-cultured cells showed long-term (>6 months) multilineage repopulation derived from irradiated donor CD45.1+ cells. Surprisingly, transplantation of identical doses of 1050 cGy-irradiated/cytokine-cultured BM was also radioprotective in 50% of irradiated recipient mice and 50% of these mice demonstrated donor-derived repopulation. CONCLUSIONS: Fully functional BM stem and progenitor cells can be rescued following harvest from lethally irradiated animals via ex vivo culture with PMVEC or cytokines alone. This method can serve as a model for the rapid ex vivo rescue and transplantation of autologous BM progenitors in the treatment of victims of radiation injury.


Subject(s)
Bone Marrow/radiation effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/radiation effects , Radiation Injuries, Experimental/pathology , Animals , Cell Division , Cell Lineage , Cell Separation , Cells, Cultured/cytology , Coculture Techniques , Colony-Forming Units Assay , Cytokines/pharmacology , Dose-Response Relationship, Radiation , Endothelial Cells/physiology , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Models, Animal , Radiation Chimera , Radiation Injuries/therapy , Radiation-Protective Agents/pharmacology , Swine , Transplantation, Autologous
SELECTION OF CITATIONS
SEARCH DETAIL
...