Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cytometry A ; 89(11): 1017-1030, 2016 11.
Article in English | MEDLINE | ID: mdl-27813253

ABSTRACT

The purpose of this document is to define minimal standards for a flow cytometry shared resource laboratory (SRL) and provide guidance for best practices in several important areas. This effort is driven by the desire of International Society for the Advancement of Cytometry (ISAC) members in SRLs to define and maintain standards of excellence in flow cytometry, and act as a repository for key elements of this information (e.g. example SOPs/training material, etc.). These best practices are not intended to define specifically how to implement these recommendations, but rather to establish minimal goals for an SRL to address in order to achieve excellence. It is hoped that once these best practices are established and implemented they will serve as a template from which similar practices can be defined for other types of SRLs. Identification of the need for best practices first occurred through discussions at the CYTO 2013 SRL Forum, with the most important areas for which best practices should be defined identified through several surveys and SRL track workshops as part of CYTO 2014. © 2016 International Society for Advancement of Cytometry.


Subject(s)
Flow Cytometry/standards , Laboratories/standards , Practice Guidelines as Topic/standards
2.
J Immunol ; 182(7): 4255-66, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19299724

ABSTRACT

IL-7 is critical for B cell production in adult mice; however, its role in human B lymphopoiesis is controversial. One challenge was the inability to differentiate human cord blood (CB) or adult bone marrow (BM) hematopoietic stem cells (HSCs) without murine stroma. Here, we examine the role of IL-7 in human B cell development using a novel, human-only model based on coculturing human HSCs on primary human BM stroma. In this model, IL-7 increases human B cell production by >60-fold from both CB and adult BM HSCs. IL-7-induced increases are dose-dependent and specific to CD19(+) cells. STAT5 phosphorylation and expression of the Ki-67 proliferation Ag indicate that IL-7 acts directly on CD19(+) cells to increase proliferation at the CD34(+) and CD34(-) pro-B cell stages. Without IL-7, HSCs in CB, but not BM, give rise to a small but consistent population of CD19(lo) B lineage cells that express EBF (early B cell factor) and PAX-5 and respond to subsequent IL-7 stimulation. Flt3 ligand, but not thymic stromal-derived lymhopoietin (TSLP), was required for the IL-7-independent production of human B lineage cells. As compared with CB, adult BM shows a reduction of in vitro generative capacity that is progressively more profound in developmentally sequential populations, resulting in an approximately 50-fold reduction in IL-7-dependent B lineage generative capacity. These data provide evidence that IL-7 is essential for human B cell production from adult BM and that IL-7-induced expansion of the pro-B compartment is increasingly critical for human B cell production during the progression of ontogeny.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Fetal Blood/cytology , Hematopoietic Stem Cells/immunology , Interleukin-7/immunology , Lymphopoiesis/immunology , Adult , Animals , Bone Marrow/immunology , Cell Differentiation/immunology , Cell Line , Cell Lineage/immunology , Coculture Techniques/methods , Enzyme-Linked Immunosorbent Assay , Fetal Blood/immunology , Flow Cytometry , Humans , Interleukin-7/metabolism , Mice , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/cytology
3.
Biol Blood Marrow Transplant ; 14(1): 16-27, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18158957

ABSTRACT

Interleukin (IL)-7 promotes both thymopoiesis and mature T lymphocyte survival and proliferation in experimental murine models of hematopoietic stem cell (HSC) transplantation. Because HSC products for transplantation also may contain IL-7-responsive mature T lymphocytes, we examined whether IL-7 is necessary for the induction of GVHD after allogeneic bone marrow transplantation (BMT). Lethally irradiated C57BL6J (B6) and B6.IL-7(-/-) (both H2K(b)) recipient mice were co-transplanted with T cell-depleted (TCD) bone marrow cells and lymph nodes (LNs) from either congenic B6.SJL (CD45.1(+)) or allogeneic BALB/c (H2K(d)) donor mice. After transplantation, the recipient mice were subcutaneously injected with either human recombinant IL-7 or phosphate-buffered saline (PBS) for 60 days. No evidence of GVHD was detected in the congenic recipients or in the allogeneic B6/IL-7(-/-) recipients treated with PBS; in contrast, significantly increased rates of GVHD-related mortality and morbidity were found in the allogeneic B6.IL-7(-/-) recipients treated with IL-7. The proliferation and number of donor T cells were significantly lower at day 30 post-BMT in the PBS-treated B6.IL-7(-/-) recipients compared with the IL-7-treated B6.IL-7(-/-) mice. These experiments demonstrate that IL-7 is an important factor in the development of GVHD, presumably by supporting the survival, proliferation, and possibly activation of alloreactive donor-derived T cells in the recipients.


Subject(s)
Bone Marrow Transplantation/adverse effects , Disease Models, Animal , Graft vs Host Disease/immunology , Interleukin-7/physiology , Animals , Humans , Interleukin-7/administration & dosage , Lymphocyte Depletion , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Survival Analysis , Transplantation, Homologous/adverse effects , Transplantation, Homologous/immunology
4.
Stem Cells ; 25(10): 2419-29, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17585168

ABSTRACT

Although oval cells are postulated to be adult liver stem cells, a well-defined phenotype of a bipotent liver stem cell remains elusive. The heterogeneity of cells within the oval cell fraction has hindered lineage potential studies. Our goal was to identify an enriched population of bipotent oval cells using a combination of flow cytometry and single cell gene expression in conjunction with lineage-specific liver injury models. Expression of cell surface markers on nonparenchymal, nonhematopoietic (CD45-) cells were characterized. Cell populations were isolated by flow cytometry for gene expression studies. 3,5-Diethoxycarbonyl-1,4-dihydrocollidine toxic injury induced cell cycling and expansion specifically in the subpopulation of oval cells in the periportal zone that express CD133. CD133+CD45- cells expressed hepatoblast and stem cell-associated genes, and single cells coexpressed both hepatocyte and cholangiocyte-associated genes, indicating bilineage potential. CD133+CD45- cells proliferated in response to liver injury. Following toxic hepatocyte damage, CD133+CD45- cells demonstrated upregulated expression of the hepatocyte gene Albumin. In contrast, toxic cholangiocyte injury resulted in upregulation of the cholangiocyte gene Ck19. After 21-28 days in culture, CD133+CD45- cells continued to generate cells of both hepatocyte and cholangiocyte lineages. Thus, CD133 expression identifies a population of oval cells in adult murine liver with the gene expression profile and function of primitive, bipotent liver stem cells. In response to lineage-specific injury, these cells demonstrate a lineage-appropriate genetic response. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Antigens, CD/analysis , Cell Lineage , Glycoproteins/analysis , Liver Regeneration/physiology , Liver/cytology , Peptides/analysis , 1-Naphthylisothiocyanate/toxicity , AC133 Antigen , Animals , Biomarkers , Bone Marrow Transplantation , Carbon Tetrachloride Poisoning/pathology , Cell Division , Cells, Cultured/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dicarbethoxydihydrocollidine/toxicity , Gene Expression Profiling , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Radiation Chimera
5.
FASEB J ; 20(12): 2142-4, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16935935

ABSTRACT

Acute promyelocytic leukemia (APL) cells express promyelocytic leukemia/retinoic acid receptor alpha (PML/RARalpha) fusion protein, which leads to the blocking of APL cell differentiation. Treatment of APL with all-trans-retinoic acid (ATRA) induces disease remission by in vivo differentiation of APL cells. Differentiation requires cell cycle exit; yet how ATRA couples cell cycle exit to differentiation of APL remains largely unknown. We previously found that ATRA-induced cell differentiation accompanies ubiquitination-proteolysis of ménage à trois 1 (MAT1), an assembly factor and targeting subunit of cyclin-dependent kinase (CDK)-activating kinase (CAK) that regulates G1 exit. We report here that CAK binds to and phosphorylates PML/RARalpha in actively proliferating APL cells. In response to ATRA, PML/RARalpha is dissociated from CAK, leading to MAT1 degradation, G1 arrest, and decreased CAK phosphorylation of PML/RARalpha. CAK phosphorylation of PML/RARalpha is inhibited when MAT1 levels are reduced. Both MAT1 degradation and PML/RARalpha hypophosphorylation occur in ATRA-induced G1-arresting cells undergoing differentiation but not in the synchronized G1 cells that do not differentiate. These findings reveal a novel ATRA signaling on APL cell differentiation, in which ATRA coordinates G1 arrest and transition into differentiation by inducing MAT1 degradation and PML/RARalpha hypophosphorylation through disrupting PML/RARalpha binding and phosphorylation by CAK.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , G1 Phase/drug effects , Leukemia/pathology , Oncogene Proteins, Fusion/metabolism , Tretinoin/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Cyclin-Dependent Kinases/metabolism , Humans , Oncogene Proteins, Fusion/drug effects , Peptide Hydrolases/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Transcription Factors , Ubiquitin/metabolism , Cyclin-Dependent Kinase-Activating Kinase
6.
Exp Hematol ; 34(10): 1333-43, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16982326

ABSTRACT

OBJECTIVE: Methods of gene transfer to hematopoietic stem cells that result in stable integration may provide treatments for many inherited and acquired blood diseases. It has been demonstrated previously that a gene delivery system based on the Sleeping Beauty (SB) transposon can be derived where a plasmid transiently expressing the SB transposase can mediate the stable chromosomal integration of a codelivered second plasmid containing a gene expression unit flanked by the inverted repeats derived from the transposon. METHODS: Plasmid DNA containing the elements required for SB transposition was delivered to hematopoietic cells via electroporation. Integrated transgene (enhanced green fluorescent protein [eGFP]) expression was assessed in vitro and in vivo. RESULTS: In the K562 human hematopoietic cell line, we observed stable expression of eGFP in >60% of cells for over 2 months after electroporation of the two plasmids; in contrast, in control cells either not treated with transposase or exposed to a defective mutant transposase, the level of gene expression had fallen to near background (<0.1%) by 2 weeks. In purified human cord blood CD34(+) progenitor cells, the transposase led to stable gene transfer at levels up to 6% for over 4 weeks, but gene transfer to more primitive nonobese diabetic/severe combined immunodeficient repopulating cells or CD34(+)/CD38(-) in long-term culture was low and electroporation of the cells with plasmid DNA caused significant cell death. CONCLUSION: The long-term stable expression highlights the potential of this transposase-based gene delivery method for ameliorating diseases affecting the hematopoietic system, although further improvements in gene transfer efficacy are needed.


Subject(s)
Antigens, CD34 , DNA Transposable Elements , Electroporation , Gene Expression , Hematopoietic Stem Cells , Mutagenesis, Insertional , Transposases/biosynthesis , DNA Transposable Elements/genetics , Electroporation/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , K562 Cells , Mutagenesis, Insertional/methods , Time Factors , Transposases/genetics
7.
Stem Cell Reports ; 5(1): 125-38, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26050929

ABSTRACT

Prior efforts to identify regulators of hematopoietic stem cell physiology have relied mainly on candidate gene approaches with genetically modified mice. Here we used a genome-wide association study (GWAS) strategy with the hybrid mouse diversity panel to identify the genetic determinants of hematopoietic stem/progenitor cell (HSPC) frequency. Among 108 strains, we observed ∼120- to 300-fold variation in three HSPC populations. A GWAS analysis identified several loci that were significantly associated with HSPC frequency, including a locus on chromosome 5 harboring the homeodomain-only protein gene (Hopx). Hopx previously had been implicated in cardiac development but was not known to influence HSPC biology. Analysis of the HSPC pool in Hopx-/- mice demonstrated significantly reduced cell frequencies and impaired engraftment in competitive repopulation assays, thus providing functional validation of this positional candidate gene. These results demonstrate the power of GWAS in mice to identify genetic determinants of the hematopoietic system.


Subject(s)
Genome-Wide Association Study , Hematopoietic Stem Cells , Homeodomain Proteins/genetics , Animals , Cell Lineage/genetics , Cell Proliferation/genetics , Mice , Mice, Knockout
8.
PLoS One ; 8(8): e67861, 2013.
Article in English | MEDLINE | ID: mdl-23936296

ABSTRACT

Two distinct microenvironmental niches that regulate hematopoietic stem/progenitor cell physiology in the adult bone marrow have been proposed; the endosteal and the vascular niche. While extensive studies have been performed relating to molecular interactions in the endosteal niche, the mechanisms that regulate hematopoietic stem/progenitor cell interaction with bone marrow endothelial cells are less well defined. Here we demonstrate that endothelial cells derived from the bone marrow supported hematopoietic stem/progenitor cells to a higher degree than other endothelial or stromal cell populations. This support was dependant upon placental growth factor expression, as genetic knockdown of mRNA levels reduced the ability of endothelial cells to support hematopoietic stem/progenitor cells in vitro. Furthermore, using an in vivo model of recovery from radiation induced myelosuppression, we demonstrate that bone marrow endothelial cells were able to augment the recovery of the hematopoietic stem/progenitor cells. However, this effect was diminished when the same cells with reduced placental growth factor expression were administered, possibly owing to a reduced homing of the cells to the bone marrow vasculature. Our data suggest that placental growth factor elaborated from bone marrow endothelial cells mediates the regulatory effects of the vascular niche on hematopoietic stem/progenitor cell physiology.


Subject(s)
Bone Marrow/metabolism , Endothelium, Vascular/metabolism , Hematopoietic Stem Cells/metabolism , Pregnancy Proteins/metabolism , Animals , Blotting, Western , Cells, Cultured , Endothelium, Vascular/cytology , Fluorescent Antibody Technique , Hematopoietic Stem Cells/cytology , Immunoenzyme Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Placenta Growth Factor , Pregnancy Proteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
9.
Am J Physiol Lung Cell Mol Physiol ; 296(1): L57-70, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18952756

ABSTRACT

Shortened telomeres are a normal consequence of cell division. However, telomere shortening past a critical point results in cellular senescence and death. To determine the effect of telomere shortening on lung, four generations of B6.Cg-Terc(tm1Rdp) mice, null for the terc component of telomerase, the holoenzyme that maintains telomeres, were bred and analyzed. Generational inbreeding of terc-/- mice caused sequential shortening of telomeres. Lung histology from the generation with the shortest telomeres (terc-/- F4) showed alveolar wall thinning and increased alveolar size. Morphometric analysis confirmed a significant increase in mean linear intercept (MLI). terc-/- F4 lung showed normal elastin deposition but had significantly decreased collagen content. Both airway and alveolar epithelial type 1 cells (AEC1) appeared normal by immunohistochemistry, and the percentage of alveolar epithelial type 2 cells (AEC2) per total cell number was similar to wild type. However, because of a decrease in distal lung cellularity, the absolute number of AEC2 in terc-/- F4 lung was significantly reduced. In contrast to wild type, terc-/- F4 distal lung epithelium from normoxia-maintained mice exhibited DNA damage by terminal deoxynucleotidyltransferase (TdT)-mediated dUTP nick end labeling (TUNEL) and 8-oxoguanine immunohistochemistry. Western blotting of freshly isolated AEC2 lysates for stress signaling kinases confirmed that the stress-activated protein kinase (SAPK)/c-Jun NH(2)-terminal kinase (JNK) stress response pathway is stimulated in telomerase-null AEC2 even under normoxic conditions. Expression of downstream apoptotic/stress markers, including caspase-3, caspase-6, Bax, and HSP-25, was also observed in telomerase-null, but not wild-type, AEC2. TUNEL analysis of freshly isolated normoxic AEC2 showed that DNA strand breaks, essentially absent in wild-type cells, increased with each successive terc-/- generation and correlated strongly with telomere length (R(2) = 0.9631). Thus lung alveolar integrity, particularly in the distal epithelial compartment, depends on proper telomere maintenance.


Subject(s)
Pulmonary Alveoli/pathology , RNA/genetics , Telomerase/genetics , Telomere/genetics , Telomere/pathology , Actins/metabolism , Animals , Biomarkers/metabolism , Collagen/metabolism , DNA Damage , Elastin/metabolism , Female , In Situ Nick-End Labeling , Intercellular Signaling Peptides and Proteins , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Oxidative Stress/physiology , Peptides/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Surfactant-Associated Protein C , RNA/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Signal Transduction/physiology , Telomerase/metabolism
10.
Blood ; 111(3): 1318-26, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17959857

ABSTRACT

The identity and lineage potential of the cells that initiate thymopoiesis remain controversial. The goal of these studies was to determine, at a clonal level, the immunophenotype and differentiation pathways of the earliest progenitors in human thymus. Although the majority of human CD34(+)lin(-) thymocytes express high levels of CD7, closer analysis reveals that a continuum of CD7 expression exists, and 1% to 2% of progenitors are CD7(-). CD34(+)lin(-) thymocytes were fractionated by CD7 expression and tested for lineage potential in B-lymphoid, T-lymphoid, and myeloid-erythroid conditions. Progressive restriction in lineage potential correlated with CD7 expression, that is, the CD7(hi) fraction produced T and NK cells but lacked B and myelo-erythroid potential, the CD7(int) (CD10(+)) fraction produced B, T, and NK cells, but lacked myelo-erythroid potential. The CD7(-) fraction produced all lymphoid and myelo-erythroid lineages and expressed HSC-associated genes. However, CD34(+)lin(-)CD7(-) thymocytes also expressed early T lymphoid genes Tdt, pTalpha, and IL-7Ralpha and lacked engraftment capacity, suggesting the signals that direct lymphoid commitment and corresponding loss of HSC function are rapidly initiated on arrival of HSC in the human thymus. Thus, differential levels of CD7 identify the progressive stages of lineage commitment in human thymus, initiated from a primitive CD7(-) lympho-myeloid thymic progenitor.


Subject(s)
Antigens, CD7/immunology , Cell Lineage/immunology , Lymphocytes/immunology , Myeloid Cells/immunology , Stem Cells/cytology , Stem Cells/immunology , Thymus Gland/immunology , Animals , Antigens, CD1/metabolism , Antigens, CD34/metabolism , Antigens, CD7/metabolism , Biomarkers , Cell Line , Gene Expression Profiling , Humans , Immunophenotyping , Lymphocytes/cytology , Mice , Myeloid Cells/cytology , Phenotype , Stem Cells/metabolism , Thymus Gland/cytology
11.
Blood ; 110(8): 2803-10, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17595335

ABSTRACT

Graft-versus-host disease (GVHD) continues to be a serious complication that limits the success of allogeneic bone marrow transplantation (BMT). Using IL-7-deficient murine models, we have previously shown that IL-7 is necessary for the pathogenesis of GVHD. In the present study, we determined whether GVHD could be prevented by antibody-mediated blockade of IL-7 receptor alpha (IL-7Ralpha) signaling. C57/BL6 (H2K(b)) recipient mice were lethally irradiated and underwent cotransplantation with T-cell-depleted (TCD) BM and lymph node (LN) cells from allogeneic BALB/c (H2K(d)) donor mice. Following transplantation, the allogeneic BMT recipients were injected weekly with either anti-IL-7Ralpha antibody (100 mug per mouse per week) or PBS for 4 weeks. Anti-IL-7Ralpha antibody treatment significantly decreased GVHD-related morbidity and mortality compared with placebo (30% to 80%). IL-7Ralpha blockade resulted in the reduction of donor CD4(+) or CD8(+) T cells in the periphery by day 30 after transplantation. Paradoxically, the inhibition of GVHD by anti-IL-7Ralpha antibody treatment resulted in improved long-term thymic and immune function. Blockade of IL-7R by anti-IL-7Ralpha antibody resulted in elimination of alloreactive T cells, prevention of GVHD, and improvement of donor T-cell reconstitution.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Graft vs Host Disease/prevention & control , Receptors, Interleukin-7/antagonists & inhibitors , Animals , Antibody Formation/drug effects , Bone Marrow Transplantation/adverse effects , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Immunophenotyping , Male , Mice , Mice, Mutant Strains , T-Lymphocytes/drug effects , Thymus Gland/drug effects , Transplantation, Homologous
12.
Hepatology ; 45(5): 1250-60, 2007 May.
Article in English | MEDLINE | ID: mdl-17464997

ABSTRACT

UNLABELLED: Recent reports have provided conflicting conclusions regarding the role for bone marrow (BM)-derived cells in the regeneration of liver. Our aim was to investigate the potential of BM to contribute to liver epithelium using different BM transplant models designed to explore differentiation during normal liver development and regeneration after toxic injury. BM cells from transgenic green fluorescent protein (GFP) mice were injected into neonatal and adult immunodeficient and neonatal immune-competent mice. Three distinct models of liver injury were employed to test the contribution of marrow to the regeneration of hepatocytes, cholangiocytes, and oval cells in immune-deficient adult animals after neonatal transplant. Immunohistochemistry was combined with flow cytometry (FACS) and reverse transcription (RT)-PCR to increase the sensitivity and specificity of the analyses. Although GFP+ marrow-derived cells were observed in the livers of all transplanted animals, immunohistochemistry failed to demonstrate any marrow derived hepatocytes or cholangiocytes. FACS confirmed that GFP+ marrow-derived cells in the liver maintained expression of CD45, a leukocyte marker. Gene expression studies of GFP+ cells isolated by FACS failed to demonstrate expression of liver specific genes in these marrow-derived cells. CONCLUSION: Through highly sensitive and specific analyses, we were unable to demonstrate any evidence of transdifferentiation of BM-derived cells into epithelial hepatic tissue during the period of rapid growth in the neonatal period. Furthermore, although increased migration of hematopoietic cells to the liver occurred after toxic injury, these cells did not contribute directly to the replacement of hepatocytes, cholangiocytes, or oval cells.


Subject(s)
Bone Marrow Cells/physiology , Epithelium/physiology , Liver Regeneration/physiology , Liver/cytology , 1-Naphthylisothiocyanate/toxicity , Animals , Animals, Newborn , Carbon Tetrachloride Poisoning/pathology , Cell Cycle , Flow Cytometry , Gene Expression Profiling , Hematopoiesis , Liver/drug effects , Liver/growth & development , Male , Mice , Mice, Inbred C57BL
13.
Am J Physiol Lung Cell Mol Physiol ; 290(4): L685-L694, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16299057

ABSTRACT

In this study, C57BL/6J mice were exposed to hyperoxia and allowed to recover in room air. The sublethal dose of hyperoxia for C57BL/6J was 48 h. Distal lung cellular isolates from treated animals were characterized as 98% epithelial, with minor fibroblast and endothelial cell contaminants. Cells were then verified as 95% pure alveolar epithelial type II cells (AEC2) by surfactant protein C (SP-C) expression. After hyperoxia exposure in vivo, fresh, uncultured AEC2 were analyzed for proliferation by cell yield, cell cycle, PCNA expression, and telomerase activity. DNA damage was assessed by TdT-dUTP nick-end labeling, whereas induction of DNA repair was evaluated by GADD-153 expression. A baseline level for proliferation and damage was observed in cells from control animals that did not alter significantly during acute hyperoxia exposure. However, a rise in these markers was observed 24 h into recovery. Over 72 h of recovery, markers for proliferation remained elevated, whereas those for DNA damage and repair peaked at 48 h and then returned back to baseline. The expression of GADD-153 followed a distinct course, rising significantly during acute exposure and peaking at 48 h recovery. These data demonstrate that in healthy, adult male C57BL/6J mice, AEC2 proliferation, damage, and repair follow separate courses during hyperoxia recovery and that both proliferation and efficient repair may be required to ensure AEC2 survival.


Subject(s)
Cell Proliferation , DNA Repair , Hyperoxia/pathology , Hyperoxia/physiopathology , Pulmonary Alveoli/pathology , Pulmonary Alveoli/physiopathology , Animals , Biomarkers/metabolism , DNA Damage , Epithelial Cells/classification , Epithelial Cells/metabolism , Hyperoxia/genetics , Hyperoxia/metabolism , Keratins/metabolism , Male , Mice , Mice, Inbred C57BL , Proliferating Cell Nuclear Antigen/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Surfactant-Associated Protein C/metabolism , Recovery of Function , S Phase , Telomerase/metabolism , Time Factors , Transcription Factor CHOP/metabolism , Up-Regulation
14.
Stem Cells ; 24(4): 928-35, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16306150

ABSTRACT

In a previous investigation, we demonstrated that mesenchymal stem cells (MSCs) actively migrated to cardiac allografts and contributed to graft fibrosis and, to a lesser extent, to myocardial regeneration. The cellular/molecular mechanism responsible for MSC migration, however, is poorly understood. This paper examines the role of CD44-hyaluronan interaction in MSC migration, using a rat MSC line Ap8c3 and mouse CD44-/- or CD44+/+ bone marrow stromal cells (BMSCs). Platelet-derived growth factor (PDGF) stimulation of MSC Ap8c3 cells significantly increased the levels of cell surface CD44 detected by flow cytometry. The CD44 standard isoform was predominantly expressed by Ap8c3 cells, accounting for 90% of the CD44 mRNA determined by quantitative real-time polymerase chain reaction. Mouse CD44-/- BMSCs bonded inefficiently to hyaluronic acid (HA), whereas CD44+/+ BMSC and MSC Ap8c3 adhered strongly to HA. Adhesions of MSC Ap8c3 cells to HA were suppressed by anti-CD44 antibody and by CD44 small interfering RNA (siRNA). HA coating of the migration chamber significantly promoted passage of CD44+/+ BMSC or Ap8c3 cells, but not CD44-/- BMSCs, through the insert membranes (p < .01). Migration of MSC Ap8c3 was significantly inhibited by anti-CD44 antibodies (p < .01) and to a lesser extent by CD44 siRNA (p = .05). The data indicate that MSC Ap8c3 cells, in response to PDGF stimulation, express high levels of CD44 standard (CD44s) isoform, which facilitates cell migration through interaction with extracellular HA. Such a migratory mechanism could be critical for recruitment of MSCs into wound sites for the proposition of tissue regeneration, as well as for migration of fibroblast progenitors to allografts in the development of graft fibrosis.


Subject(s)
Hyaluronan Receptors/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Base Sequence , Cell Adhesion , Cell Line , Cell Movement , Cells, Cultured , DNA, Complementary/genetics , Extracellular Matrix/metabolism , Hyaluronan Receptors/genetics , Hyaluronic Acid/metabolism , Mesenchymal Stem Cells/drug effects , Mice , Mice, Knockout , Platelet-Derived Growth Factor/pharmacology , RNA, Small Interfering/genetics , Rats
15.
J Heart Lung Transplant ; 24(12): 2160-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16364866

ABSTRACT

BACKGROUND: Mesenchymal progenitor cells (MPC) have recently been demonstrated to actively migrate into cardiac allografts during chronic rejection. This study examines the role of MPC in tissue repair of heart allografts in a rat model of chronic rejection. METHODS: The potential of a rat MPC line (Ap8c3) to differentiate to myofibroblasts and cardiomyocytes was studied in differentiation cultures. Ap8c3 cells tagged with an enhanced green fluorescent protein (eGFP) reporter gene were engrafted into Fischer 344 (F344) recipients of Lewis (LEW) cardiac allografts. Development of intragraft MPC into scar-forming fibroblasts and cardiomyocytes was studied using immunohistochemistry. RESULTS: Ap8c3 cells contain fibroblast progenitors (FP) positive for P07 antibody. Transforming growth factor (TGF)-beta stimulation promoted FP to terminally differentiate into myofibroblasts, which express alpha-smooth muscle actin (alphaSMA). In cardiac differentiation culture, Ap8c3 cells were induced by 5-azatiditin (5-aza) to form tropomyosin+ myotubes, and to express mRNA encoding for cardiac troponin I (TnI) and alpha-myosin heavy chain (alphaMHC). Transfusion of eGFP+ Ap8c3 cells to F344 recipients resulted in migration of eGFP(+) cells into LEW heart allografts, as well as homing of the eGFP+ MPC to bone marrow. The majority of eGFP+ cells in the heart allografts appeared to be vimentin-expressing fibroblasts. Foci of eGFP+ myocardium were also detected in all heart allografts, with eGFP+ cardiomyocytes representing 4.8 +/- 1.2% of the allografted eGFP+ cells. CONCLUSIONS: The data suggest that rat MPC participate in tissue repair in heart allografts by giving rise to scar-forming myofibroblasts and cardiomyocytes.


Subject(s)
Graft Rejection/therapy , Heart Transplantation/immunology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Regeneration , Animals , Cell Differentiation , Cell Movement , Chronic Disease , Fibroblasts , Flow Cytometry , Genes, Reporter , Green Fluorescent Proteins , Immunohistochemistry , Myocytes, Cardiac , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transduction, Genetic , Transplantation, Heterotopic , Transplantation, Homologous
16.
Stem Cells ; 23(6): 852-60, 2005.
Article in English | MEDLINE | ID: mdl-15917481

ABSTRACT

The stem cell leukemia (SCL or tal-1) gene was initially identified as a translocation partner in a leukemia that possessed both lymphoid and myeloid differentiation potential. Mice that lacked SCL expression showed a complete block in hematopoiesis; thus, SCL was associated with hematopoietic stem cell (HSC) function. More recent studies show a role for SCL in murine erythroid differentiation. However, the expression pattern and the role of SCL during early stages of human hematopoietic differentiation are less clear. In this study we chart the pattern of human SCL expression from HSCs, through developmentally sequential populations of lymphoid and myeloid progenitors to mature cells of the hematopoietic lineages. Using recently defined surface immunophenotypes, we fluorescence-activated cell-sorted (FACS) highly purified populations of primary human hematopoietic progenitors for reverse transcription-polymerase chain reaction (RT-PCR) analysis of SCL expression. Our data show that SCL mRNA is easily detectable in all hematopoietic populations with erythroid potential, including HSCs, multipotential progenitors, common myeloid progenitors, megakaryocyte/erythrocyte progenitors, and nucleated erythroid lineage cells. SCL mRNA expression was present but rapidly downregulated in the common lymphoid progenitor and granulocyte/monocyte progenitor populations that lack erythroid potential. SCL expression was undetectable in immature cells of nonerythroid lineages, including pro-B cells, early thymic progenitors, and myeloid precursors expressing the M-CSF receptor. SCL expression was also absent from all mature cells of the nonerythroid lineages. Although low levels of SCL were detected in lymphoid- and myeloid-restricted progenitors, our studies show that abundant SCL expression is normally tightly linked with erythroid differentiation potential.


Subject(s)
DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/physiology , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/physiology , Transcription Factors/biosynthesis , Transcription Factors/physiology , Animals , Antigens, CD34/biosynthesis , Basic Helix-Loop-Helix Transcription Factors , Bone Marrow Cells/cytology , Cell Differentiation , Cell Line , Cell Lineage , Cell Separation , Cells, Cultured , Down-Regulation , Flow Cytometry , Hematopoietic Stem Cells/cytology , Humans , Immunophenotyping , Mice , Models, Biological , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , T-Cell Acute Lymphocytic Leukemia Protein 1 , Thymus Gland/metabolism , Time Factors
17.
Am J Physiol Lung Cell Mol Physiol ; 286(4): L658-67, 2004 Apr.
Article in English | MEDLINE | ID: mdl-12922980

ABSTRACT

Alveolar epithelial type 2 cells (AEC2) isolated from hyperoxia-treated animals exhibit increases in both proliferation and DNA damage in response to culture. AEC2 express the zonula adherens proteins E-cadherin, -, - and -catenin, desmoglein, and pp120, as demonstrated by Western blotting. Immunohistochemical analysis of cultured AEC2 showed expression of E-cadherin on cytoplasmic membranes varying from strongly to weakly staining. When cultured AEC2 placed in suspension were labeled with fluorescent-tagged antibodies to E-cadherin, cells could be sorted into at least two subpopulations, either dim or brightly staining for this marker. With the use of antibody to E-cadherin bound to magnetic beads, cells were physically separated into E-cadherin-positive and -negative subpopulations, which were then analyzed for differences in proliferation and DNA damage. The E-cadherin-positive subpopulation contained the majority of damaged cells, was quiescent, and expressed low levels of telomerase activity, whereas the E-cadherin-negative subpopulation was undamaged, proliferative, and expressed high levels of telomerase activity.


Subject(s)
Pulmonary Alveoli/cytology , Respiratory Mucosa/cytology , Stem Cells/cytology , Adherens Junctions/metabolism , Animals , Cadherins/metabolism , Cell Division , Cell Separation , Hyperoxia/pathology , In Situ Nick-End Labeling , Male , Pulmonary Alveoli/physiology , Rats , Rats, Sprague-Dawley , Regeneration , Respiratory Mucosa/physiology , Stem Cells/metabolism , Telomerase/metabolism
18.
J Immunol ; 170(6): 3091-8, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12626565

ABSTRACT

The Ikaros gene is alternately spliced to generate multiple DNA-binding and nonbinding isoforms that have been implicated as regulators of hematopoiesis, particularly in the lymphoid lineages. Although early reports of Ikaros mutant mice focused on lymphoid defects, these mice also show significant myeloid, erythroid, and stem cell defects. However, the specific Ikaros proteins expressed in these cells have not been determined. We recently described Ikaros-x (Ikx), a new Ikaros isoform that is the predominant Ikaros protein in normal human hematopoietic cells. In this study, we report that the Ikx protein is selectively expressed in human myeloid lineage cells, while Ik1 predominates in the lymphoid and erythroid lineages. Both Ik1 and Ikx proteins are expressed in early human hematopoietic cells (Lin(-)CD34(+)). Under culture conditions that promote specific lineage differentiation, Ikx is up-regulated during myeloid differentiation but down-regulated during lymphoid differentiation from human Lin(-)CD34(+) cells. We show that Ikx and other novel Ikaros splice variants identified in human studies are also expressed in murine bone marrow. In mice, as in humans, the Ikx protein is selectively expressed in the myeloid lineage. Our studies suggest that Ikaros proteins function in myeloid, as well as lymphoid, differentiation and that specific Ikaros isoforms may play a role in regulating lineage commitment decisions in mice and humans.


Subject(s)
Myeloid Cells/metabolism , Transcription Factors/biosynthesis , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Cells, Cultured , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Ikaros Transcription Factor , Lymphocytes/cytology , Lymphocytes/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
19.
J Biol Chem ; 277(45): 43369-76, 2002 Nov 08.
Article in English | MEDLINE | ID: mdl-12213824

ABSTRACT

Cell cycle G(1) exit is a critical stage where cells commonly commit to proliferate or to differentiate, but the biochemical events that regulate the proliferation/differentiation (P/D) transition at G(1) exit are presently unclear. We previously showed that MAT1 (ménage à trois 1), an assembly factor and targeting subunit of the cyclin-dependent kinase (CDK)-activating kinase (CAK), modulates CAK activities to regulate G(1) exit. Here we find that the retinoid-induced G(1) arrest and differentiation activation of cultured human leukemic cells are associated with a switch to CAK hypophosphorylation of retinoic acid receptor alpha (RARalpha) from CAK hyperphosphorylation of RARalpha. The switch to CAK hypophosphorylation of RARalpha is accompanied by decreased MAT1 expression and MAT1 fragmentation that occurs in the differentiating cells through the all-trans-retinoic acid (ATRA)-mediated proteasome degradation pathway. Because HL60R cells that harbor a truncated ligand-dependent AF-2 domain of RARalpha do not demonstrate any changes in MAT1 levels or CAK phosphorylation of RARalpha following ATRA stimuli, these biochemical changes appear to be mediated directly through RARalpha. These studies indicate that significant changes in MAT1 levels and CAK activities on RARalpha phosphorylation accompany the ATRA-induced G(1) arrest and differentiation activation, which provide new insights to explore the inversely coordinated P/D transition at G(1) exit.


Subject(s)
Cell Cycle/physiology , Cell Differentiation/drug effects , Cyclin-Dependent Kinases/metabolism , Receptors, Retinoic Acid/physiology , Retinoids/pharmacology , Bone Neoplasms , Cell Cycle/drug effects , Cell Differentiation/physiology , Cell Line , G1 Phase , Humans , Jurkat Cells , Kidney , Leukemia , Osteosarcoma , Phosphorylation , Receptors, Retinoic Acid/drug effects , Retinoic Acid Receptor alpha , Tumor Cells, Cultured , Cyclin-Dependent Kinase-Activating Kinase
20.
Int J Cancer ; 98(5): 690-7, 2002 Apr 10.
Article in English | MEDLINE | ID: mdl-11920637

ABSTRACT

Orthotopic brain tumor growth is inhibited in athymic mice by the daily systemic administration of the alpha v-integrin antagonist EMD 121974. This compound, a cyclic RGD-penta-peptide, is a potent inhibitor of angiogenesis, which induces apoptosis of growing endothelial cells through inhibition of their alpha v-integrin interaction with the matrix proteins vitronectin and tenascin. Here we show that EMD 121974 also induces apoptosis in the alpha v-integrin-expressing tumor cell lines U87 MG and DAOY by detaching them from vitronectin and tenascin, matrix proteins known to be essential for brain tumor growth and invasion. These matrix proteins are shown to be produced by the brain tumor cells in vitro and in vivo. Furthermore, only tumor cells expressing alpha v-integrins responded to the treatment with EMD 121974, after xenotransplantation into the forebrain of nude mice, supporting the importance of tumor cell-matrix interactions in tumor cell survival in the brain. Thus, the alpha v-antagonist EMD 121974 suppresses brain tumor growth through induction of apoptosis in both brain capillary and brain tumor cells by preventing their interaction with the matrix proteins vitronectin and tenascin. The dual action of this peptide explains its potent growth suppression of orthotopically transplanted brain tumors.


Subject(s)
Apoptosis/drug effects , Brain Neoplasms/pathology , Glioblastoma/pathology , Integrins/antagonists & inhibitors , Medulloblastoma/pathology , Peptides, Cyclic/pharmacology , Receptors, Vitronectin/antagonists & inhibitors , Animals , Brain Neoplasms/metabolism , Cell Adhesion/drug effects , Cell Division/drug effects , Collagen/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Glioblastoma/metabolism , Humans , Immunoenzyme Techniques , In Situ Nick-End Labeling , Integrins/metabolism , Medulloblastoma/metabolism , Mice , Mice, Nude , Receptors, Vitronectin/metabolism , Snake Venoms , Tenascin/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology , Vitronectin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL