446 results on '"Rafii S"'
Search Results
402. Brain derived neurotrophic factor is an endothelial cell survival factor required for intramyocardial vessel stabilization.
- Author
-
Donovan MJ, Lin MI, Wiegn P, Ringstedt T, Kraemer R, Hahn R, Wang S, Ibañez CF, Rafii S, and Hempstead BL
- Subjects
- Animals, Animals, Newborn, Apoptosis, Brain-Derived Neurotrophic Factor deficiency, Brain-Derived Neurotrophic Factor genetics, Capillaries growth & development, Capillaries physiology, Cell Communication, Cell Survival, Coronary Circulation, Coronary Vessels growth & development, Crosses, Genetic, Endothelium, Vascular physiology, Heart growth & development, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptor, trkB genetics, Reverse Transcriptase Polymerase Chain Reaction, Brain-Derived Neurotrophic Factor physiology, Coronary Vessels physiology, Endothelium, Vascular cytology, Heart Defects, Congenital genetics
- Abstract
Brain derived neurotrophic factor, BDNF, is a neurotrophin best characterized for its survival and differentiative effects on neurons expressing the trk B receptor tyrosine kinase. Although many of these neurons are lost in the BDNF(-)(/)(- )mouse, the early postnatal lethality of these animals suggests a wider function for this growth factor. Here, we demonstrate that deficient expression of BDNF impairs the survival of endothelial cells in intramyocardial arteries and capillaries in the early postnatal period, although the embryonic vasculature can remodel into arteries, capillaries and veins. BDNF deficiency results in a reduction in endothelial cell-cell contacts and in endothelial cell apoptosis, leading to intraventricular wall hemorrhage, depressed cardiac contractility and early postnatal death. Vascular hemorrhage is restricted to cardiac vessels, reflecting the localized expression of BDNF and trk B by capillaries and arterioles in this vascular bed. Conversely, ectopic BDNF overexpression in midgestational mouse hearts results in an increase in capillary density. Moreover, BDNF activation of endogenous trk B receptors supports the survival of cardiac microvascular endothelial cells cultured from neonatal mice. These results establish an essential role for BDNF in maintaining vessel stability in the heart through direct angiogenic actions on endothelial cells.
- Published
- 2000
- Full Text
- View/download PDF
403. Downregulation of CXCR4 gene expression in primary human endothelial cells following infection with E1(-)E4(+) adenovirus gene transfer vectors.
- Author
-
Ramalingam R, Worgall S, Rafii S, and Crystal RG
- Subjects
- Blotting, Northern, Cells, Cultured, Down-Regulation, Flow Cytometry, Gene Expression, Gene Transfer Techniques, Genetic Therapy, Genetic Vectors, Humans, Neovascularization, Pathologic therapy, Receptors, CXCR4 metabolism, Transfection, beta-Galactosidase metabolism, Adenoviridae genetics, Adenovirus E1 Proteins genetics, Adenovirus E4 Proteins genetics, Endothelium, Vascular metabolism, RNA, Messenger metabolism, Receptors, CXCR4 genetics
- Abstract
Infection of human endothelial cells with first-generation E1(-)E4(+) adenovirus (Ad) vectors leads to prolonged cell survival and changes in the cell phenotype to a more quiescent stage. Based on the concept that the CXCR4, the receptor for the endothelial chemoattractant stromal-derived factor-&alpha (SDF-alpha), is constitutively expressed by quiescent, resting endothelial cells, the present study analyzes the effect of Ad vector infection on CXCR4 expression and SDF-alpha responses of human umbilical vein endothelial cells (HUVEC). CXCR4 transcripts were markedly downregulated in E1(-)E4(+) Ad-infected cells 48 h following infection, but not in uninfected control cells or when the cells were infected with an E1(-)E4(-) Ad vector. Analysis of surface CXCR4 expression by flow cytometry demonstrated marked reduction of the CXCR4 receptor on cells infected with E1(-)E4(+) Ad compared to uninfected control cells or E1(-)E4(-) Ad-infected cells. Infection of other cell types which express CXCR4, such as dendritic cells and myeloma cells, did not exhibit CXCR4 receptor downregulation following infection with E1(-)E4(+) Ad. Consistent with the observed downregulation of CXCR4 mRNA and surface protein, infection of the endothelial cells with an E1(-)E4(+) Ad rendered the cells unresponsive to the chemoattractant SDF-alpha compared to naive or E1(-)E4(-) Ad-infected cells. Together, the data suggest that first-generation Ad vectors, likely the E4 region, modify the ability of endothelial cells to respond to at least one important chemoattractant.
- Published
- 2000
- Full Text
- View/download PDF
404. Arsenic trioxide induces dose- and time-dependent apoptosis of endothelium and may exert an antileukemic effect via inhibition of angiogenesis.
- Author
-
Roboz GJ, Dias S, Lam G, Lane WJ, Soignet SL, Warrell RP Jr, and Rafii S
- Subjects
- Arsenic Trioxide, Cells, Cultured, Dose-Response Relationship, Drug, Humans, Leukemia drug therapy, Leukemia pathology, Neovascularization, Pathologic drug therapy, Time Factors, Antineoplastic Agents pharmacology, Apoptosis drug effects, Arsenicals pharmacology, Endothelium, Vascular drug effects, Endothelium, Vascular pathology, Oxides pharmacology
- Abstract
Arsenic trioxide (As(2)O(3)) has recently been used successfully in the treatment of acute promyelocytic leukemia and has been shown to induce partial differentiation and apoptosis of leukemic cells in vitro. However, the mechanism by which As(2)O(3) exerts its antileukemic effect remains uncertain. Emerging data suggest that the endothelium and angiogenesis play a seminal role in the proliferation of liquid tumors, such as leukemia. We have shown that activated endothelial cells release cytokines that may stimulate leukemic cell growth. Leukemic cells, in turn, can release endothelial growth factors, such as vascular endothelial growth factor (VEGF). On the basis of these observations, we hypothesized that As(2)O(3) may interrupt a reciprocal loop between leukemic cells and the endothelium by direct action on both cell types. We have shown that treatment of proliferating layers of human umbilical vein endothelial cells (HUVECs) with a variety of concentrations of As(2)O(3) results in a reproducible dose- and time-dependent sequence of events marked by change to an activated morphology, up-regulation of endothelial cell adhesion markers, and apoptosis. Also, treatment with As(2)O(3) caused inhibition of VEGF production in the leukemic cell line HEL. Finally, incubation of HUVECs with As(2)O(3) prevented capillary tubule and branch formation in an in vitro endothelial cell-differentiation assay. In conclusion, we believe that As(2)O(3 )interrupts a reciprocal stimulatory loop between leukemic cells and endothelial cells by causing apoptosis of both cell types and by inhibiting leukemic cell VEGF production. (Blood. 2000;96:1525-1530)
- Published
- 2000
405. Drip and ship: a new strategy for the treatment of acute coronary syndromes.
- Author
-
Chadow HL, Hauptman RE, VanAuker M, Rafii SE, Gunsburg MY, Giarraffa L, and Strom JA
- Subjects
- Abciximab, Acute Disease, Adult, Aged, Aged, 80 and over, Angina, Unstable, Antibodies, Monoclonal administration & dosage, Anticoagulants administration & dosage, Aspirin administration & dosage, Coronary Angiography, Electrocardiography, Female, Fibrinolytic Agents administration & dosage, Heparin administration & dosage, Hospitalization, Humans, Immunoglobulin Fab Fragments administration & dosage, Male, Middle Aged, Myocardial Infarction, Nitrates administration & dosage, Platelet Aggregation Inhibitors administration & dosage, Prospective Studies, Stents, Tirofiban, Treatment Outcome, Tyrosine administration & dosage, Tyrosine analogs & derivatives, Clinical Protocols standards, Coronary Disease drug therapy
- Abstract
Glycoprotein (GP) IIb/IIIa inhibitors block the final common pathway of platelet aggregation by preventing fibrinogen from binding to the GP IIb/IIIa platelet receptor. In patients with unstable angina (UA) or a non-Q wave myocardial infarction (NQWMI), including those with UA refractory to medical therapy, these agents decrease the risk of death, myocardial infarction (MI), and recurrent ischemia. Most patients with acute coronary syndromes are managed in hospitals without on-site angioplasty capabilities and often require transfer for an interventional procedure. We propose that GP IIb/IIIa inhibitors can be safely initiated at the referring hospital. We studied 20 patients with UA/NQWMI in whom therapy with a GP IIb/IIIa inhibitor, in addition to standard medical therapy, was initiated prior to transfer for an urgent percutaneous coronary intervention (PCI) ("drip and ship"). The primary end point was a composite of death, MI, and recurrent ischemia at 30 days. Twelve patients were treated with abciximab, 5 patients were treated with tirofiban, and 3 patients initially treated with tirofiban were converted to abciximab. Procedural success occurred in 33 out of 36 (92%) lesions and 18 out of 20 (90%) patients. At 30 days, 4 out of 20 (20%) patients had recurrent ischemia. The PTCA sites were widely patent in the 3 patients who underwent repeat angiography. The fourth patient had an unsuccessful PCI and was referred for coronary artery bypass surgery. There were no MIs or deaths. Patients who require transfer for an urgent PCI can be managed safely and efficaciously by initiating a GP IIb/IIIa inhibitor, in addition to standard medical therapy, prior to transfer.
- Published
- 2000
- Full Text
- View/download PDF
406. Autocrine stimulation of VEGFR-2 activates human leukemic cell growth and migration.
- Author
-
Dias S, Hattori K, Zhu Z, Heissig B, Choy M, Lane W, Wu Y, Chadburn A, Hyjek E, Gill M, Hicklin DJ, Witte L, Moore MA, and Rafii S
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Base Sequence, Cell Division physiology, Cell Movement drug effects, Cell Movement physiology, DNA Primers genetics, Endothelial Growth Factors genetics, Endothelial Growth Factors metabolism, Endothelial Growth Factors pharmacology, Gene Expression, Graft Survival, Humans, Leukemia genetics, Leukemia pathology, Lymphokines genetics, Lymphokines metabolism, Lymphokines pharmacology, Matrix Metalloproteinase 9 biosynthesis, Mice, Mice, Inbred NOD, Neoplasm Transplantation, Neoplastic Cells, Circulating, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Neoplasm genetics, RNA, Neoplasm metabolism, Receptor Protein-Tyrosine Kinases genetics, Receptors, Growth Factor genetics, Receptors, Vascular Endothelial Growth Factor, Signal Transduction, Transplantation, Heterologous, Tumor Cells, Cultured, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Leukemia metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Growth Factor metabolism
- Abstract
Emerging data suggest that VEGF receptors are expressed by endothelial cells as well as hematopoietic stem cells. Therefore, we hypothesized that functional VEGF receptors may also be expressed in malignant counterparts of hematopoietic stem cells such as leukemias. We demonstrate that certain leukemias not only produce VEGF but also express functional VEGFR-2 in vivo and in vitro, resulting in the generation of an autocrine loop that may support leukemic cell survival and proliferation. Approximately 50% of freshly isolated leukemias expressed mRNA and protein for VEGFR-2. VEGF(165) induced phosphorylation of VEGFR-2 and increased proliferation of leukemic cells, demonstrating these receptors were functional. VEGF(165) also induced the expression of MMP-9 by leukemic cells and promoted their migration through reconstituted basement membrane. The neutralizing mAb IMC-1C11, specific to human VEGFR-2, inhibited leukemic cell survival in vitro and blocked VEGF(165)-mediated proliferation of leukemic cells and VEGF-induced leukemic cell migration. Xenotransplantation of primary leukemias and leukemic cell lines into immunocompromised nonobese diabetic mice resulted in significant elevation of human, but not murine, VEGF in plasma and death of inoculated mice within 3 weeks. Injection of IMC-1C11 inhibited proliferation of xenotransplanted human leukemias and significantly increased the survival of inoculated mice. Interruption of signaling by VEGFRs, particularly VEGFR-2, may provide a novel strategy for inhibiting leukemic cell proliferation.
- Published
- 2000
- Full Text
- View/download PDF
407. Expression and secretion of vascular endothelial growth factor-A by cytokine-stimulated hematopoietic progenitor cells. Possible role in the hematopoietic microenvironment.
- Author
-
Bautz F, Rafii S, Kanz L, and Möhle R
- Subjects
- Antigens, CD34 analysis, Blotting, Northern, Bone Marrow Cells cytology, Bone Marrow Cells drug effects, Bone Marrow Cells metabolism, Capillary Permeability, Cell Differentiation drug effects, Cell Division drug effects, Cell Movement drug effects, Cells, Cultured, Culture Media, Conditioned, Endothelial Growth Factors genetics, Endothelial Growth Factors metabolism, Endothelial Growth Factors pharmacology, Endothelial Growth Factors physiology, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Endothelium, Vascular metabolism, Enzyme-Linked Immunosorbent Assay, Gene Expression Regulation drug effects, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Hematopoietic Stem Cells metabolism, Humans, RNA, Messenger biosynthesis, RNA, Messenger genetics, Vascular Endothelial Growth Factor A, Endothelial Growth Factors biosynthesis, Hematopoiesis, Hematopoietic Cell Growth Factors pharmacology, Hematopoietic Stem Cells drug effects
- Abstract
In the hematopoietic microenvironment, bone marrow endothelial cells may play an important role in trafficking and maintenance of progenitor and stem cells due to adhesive interactions and paracrine secretion of hematopoietic growth factors. However, it is unknown whether progenitors in turn modulate endothelial proliferation and function. We analyzed mRNA expression (Northern blot) and release of vascular endothelial growth factor-A (VEGF-A), which specifically acts on endothelial cells, by cytokine-stimulated peripheral blood-derived CD34+ hematopoietic progenitor cells. While unstimulated CD34+ cells expressed VEGF-A mRNA weakly without cytokine release in vitro, incubation for 24 hours with a single cytokine (e.g., kit ligand [KL]) resulted in increased VEGF-A mRNA expression and significant secretion of VEGF-A into the supernatant. The amount of VEGF released was substantially augmented by incubation with a combination of cytokines (e.g., KL, IL-3, GM-CSF, G-CSF), or by exposure to hematopoietic cytokines for a longer time period. In addition, we show that VEGF induced the release of hematopoietic growth factors (GM-CSF) by bone marrow endothelial cells and that in vitro stromal cell-derived factor-1 (SDF-1) driven transendothelial progenitor cell migration was increased by the presence of VEGF, which might be due to pore formation (increased endothelial fenestration). In vivo, release of VEGF by progenitor cells may result in a paracrine loop supporting proliferation of both endothelium and progenitors and may facilitate transendothelial migration during cytokine-induced progenitor cell mobilization.
- Published
- 2000
- Full Text
- View/download PDF
408. Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors.
- Author
-
Peichev M, Naiyer AJ, Pereira D, Zhu Z, Lane WJ, Williams M, Oz MC, Hicklin DJ, Witte L, Moore MA, and Rafii S
- Subjects
- AC133 Antigen, Antibodies, Monoclonal immunology, Antigens, CD, Antigens, CD34 analysis, Biomarkers, Cadherins analysis, Cell Differentiation, Cell Lineage, Chemokine CXCL12, Chemokines, CXC pharmacology, E-Selectin analysis, Endothelial Growth Factors pharmacology, Fetal Blood cytology, Gene Expression Regulation, Developmental, Glycoproteins genetics, Hematopoietic Stem Cells classification, Humans, Infant, Newborn, Lymphokines pharmacology, Neovascularization, Physiologic genetics, Peptides genetics, Receptor Protein-Tyrosine Kinases genetics, Receptor Protein-Tyrosine Kinases immunology, Receptors, Growth Factor genetics, Receptors, Growth Factor immunology, Receptors, Vascular Endothelial Growth Factor, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Endothelium, Vascular cytology, Glycoproteins metabolism, Hematopoietic Stem Cells metabolism, Peptides metabolism, Receptor Protein-Tyrosine Kinases biosynthesis, Receptors, Growth Factor biosynthesis
- Abstract
Emerging data suggest that a subset of circulating human CD34(+) cells have phenotypic features of endothelial cells. Whether these cells are sloughed mature endothelial cells or functional circulating endothelial precursors (CEPs) is not known. Using monoclonal antibodies (MoAbs) to the extracellular domain of the human vascular endothelial receptor-2 (VEGFR-2), we have shown that 1.2 +/- 0.3% of CD34(+) cells isolated from fetal liver (FL), 2 +/- 0.5% from mobilized peripheral blood, and 1.4 +/- 0.5% from cord blood were VEGFR-2(+). In addition, most CD34(+)VEGFR-2(+) cells express hematopoietic stem cell marker AC133. Because mature endothelial cells do not express AC133, coexpression of VEGFR-2 and AC133 on CD34(+) cells phenotypically identifies a unique population of CEPs. CD34(+)VEGFR-2(+) cells express endothelial-specific markers, including VE-cadherin and E-selectin. Also, virtually all CD34(+)VEGFR-2(+) cells express the chemokine receptor CXCR4 and migrate in response to stromal-derived factor (SDF)-1 or VEGF. To quantitate the plating efficiency of CD34(+) cells that give rise to endothelial colonies, CD34(+) cells derived from FL were incubated with VEGF and fibroblast growth factor (FGF)-2. Subsequent isolation and plating of nonadherent FL-derived VEGFR-2(+) cells with VEGF and FGF-2 resulted in differentiation of AC133(+ )VEGFR-2(+) cells into adherent AC133(-)VEGFR-2(+)Ac-LDL(+ )(acetylated low-density lipoprotein) colonies (plating efficiency of 3%). In an in vivo human model, we have found that the neo-intima formed on the surface of left ventricular assist devices is colonized with AC133(+)VEGFR-2(+) cells. These data suggest that circulating CD34(+) cells expressing VEGFR-2 and AC133 constitute a phenotypically and functionally distinct population of circulating endothelial cells that may play a role in neo-angiogenesis.
- Published
- 2000
409. Chemotaxis of primitive hematopoietic cells in response to stromal cell-derived factor-1.
- Author
-
Jo DY, Rafii S, Hamada T, and Moore MA
- Subjects
- Animals, Antigens, CD34 analysis, Cell Line, Cell Movement drug effects, Chemokine CXCL12, Endothelium, Vascular cytology, Hematopoietic Stem Cells physiology, Membrane Proteins pharmacology, Mice, Receptors, CXCR4 analysis, Receptors, CXCR4 physiology, Stem Cell Factor pharmacology, Thrombopoietin pharmacology, Chemokines, CXC pharmacology, Chemotaxis drug effects, Hematopoietic Stem Cells drug effects
- Abstract
Stromal cell-derived factor-1 (SDF-1) provides a potent chemotactic stimulus for CD34(+) hematopoietic cells. We cultured mobilized peripheral blood (PB) and umbilical cord blood (CB) for up to 5 weeks and examined the migratory activity of cobblestone area-forming cells (CAFCs) and long-term culture-initiating cells (LTC-ICs) in a transwell assay. In this system, SDF-1 or MS-5 marrow stromal cells placed in the lower chamber induced transmembrane and transendothelial migration by 2- and 5-week-old CAFCs and LTC-ICs in 3 hours. Transmigration was blocked by preincubation of input CD34(+) cells with antibody to CXCR4. Transendothelial migration of CB CAFCs and LTC-ICs was higher than that of PB. We expanded CD34(+) cells from CB in serum-free medium with thrombopoietin, flk-2 ligand, and c-kit ligand, with or without IL-3 and found that CAFCs cultured in the absence of IL-3 had a chemotactic response equivalent to noncultured cells, even after 5 weeks. However, addition of IL-3 to the culture reduced this response by 20-50%. These data indicate that SDF-1 induces chemotaxis of primitive hematopoietic cells signaling through CXCR4 and that the chemoattraction could be downmodulated by culture ex vivo.
- Published
- 2000
- Full Text
- View/download PDF
410. Circulating endothelial precursors: mystery, reality, and promise.
- Author
-
Rafii S
- Subjects
- Bone Marrow Transplantation, Humans, Neovascularization, Physiologic, Endothelium, Vascular cytology, Stem Cells physiology
- Published
- 2000
- Full Text
- View/download PDF
411. Stromal derived factor-1-induced chemokinesis of cord blood CD34(+) cells (long-term culture-initiating cells) through endothelial cells is mediated by E-selectin.
- Author
-
Naiyer AJ, Jo DY, Ahn J, Mohle R, Peichev M, Lam G, Silverstein RL, Moore MA, and Rafii S
- Subjects
- Cell Movement physiology, Cells, Cultured, Chemokine CXCL12, Chemokines physiology, Fetal Blood, Humans, Cell Communication physiology, Chemokines, CXC physiology, E-Selectin physiology, Endothelium, Vascular cytology, Endothelium, Vascular physiology, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells physiology
- Abstract
Homing of hematopoietic stem cells to the bone marrow (BM) involves sequential interaction with adhesion molecules expressed on BM endothelium (BMEC) and chemokine stromal derived factor-1 (SDF-1). However, the mechanism whereby adhesion molecules regulate the SDF-1-induced transendothelial migration process is not known. E-selectin is an endothelial-specific selectin that is constitutively expressed by the BMEC in vivo. Hence, we hypothesized that E-selectin may mediate SDF-1-induced transendothelial migration of CD34(+) cells. We show that CD34(+) cells express both E-selectin ligand and fucosyltransferase-VII (FucT-VII). Soluble E-selectin-IgG chimera binds avidly to 75% +/- 10% of CD34(+) cells composed mostly of progenitors and cells with long-term culture-initiating cell (LTC-IC) potential. To assess the functional capacity of E-selectin to mediate CD34(+) cell migration in a transendothelial migration system, CD34(+) cells were placed on transwell plates coated with interleukin-1beta-activated BMEC. In the absence of SDF-1, there was spontaneous migration of 7.0% +/- 1.4% of CD34(+) cells and 14.1% +/- 2.2% of LTC-IC. SDF-1 induced migration of an additional 23.0% +/- 4.4% of CD34(+) cells and 17.6% +/- 3.6% of LTC-IC. Blocking MoAb to E-selectin inhibited SDF-1-induced migration of CD34(+) cells by 42.0% +/- 2.5% and LTC-IC by 90.9% +/- 16.6%. To define the mechanism of constitutive expression of E-selectin by the BMEC in vivo, we have found that vascular endothelial growth factor (VEGF(165)) induces E-selectin expression by cultured endothelial cells. VEGF-stimulated endothelial cells support transendothelial migration of CD34(+) cells that could be blocked by MoAb to E-selectin. These results suggest that trafficking of subsets of CD34(+) cells with LTC-IC potential is determined in part by sequential interactions with E-selectin and SDF-1.
- Published
- 1999
412. Induction of endogenous genes following infection of human endothelial cells with an E1(-) E4(+) adenovirus gene transfer vector.
- Author
-
Ramalingam R, Rafii S, Worgall S, Hackett NR, and Crystal RG
- Subjects
- Adenovirus E1 Proteins genetics, Adenovirus E4 Proteins genetics, Adenoviruses, Human genetics, Capsid metabolism, Cells, Cultured, DNA, Complementary, Endothelium, Vascular cytology, Gene Deletion, Genetic Vectors genetics, Humans, Kinetics, Adenovirus E1 Proteins physiology, Adenovirus E4 Proteins physiology, Adenoviruses, Human physiology, Gene Expression Regulation, Viral, Gene Transfer Techniques, Genetic Vectors physiology
- Abstract
Recombinant adenovirus (Ad) gene transfer vectors are effective at transferring exogenous genes to a variety of cells and tissue types both in vitro and in vivo. However, in the process of gene transfer, the Ad vectors induce the expression of target cell genes, some of which may modify the function of the target cell and/or alter the local milieu. To develop a broader understanding of Ad vector-mediated induction of endogenous gene expression, genes induced by first-generation E1(-) E4(+) Ad vectors in primary human umbilical vein endothelial cells were identified by cDNA subtraction cloning. The identified cDNAs included signaling molecules (lymphoid blast crisis [LBC], guanine nucleotide binding protein alpha type S [Galpha-S], and mitogen kinase [MEK5]), calcium-regulated/cytoskeletal proteins (calpactin p11 and p36 subunits, vinculin, and spinocerebellar ataxia [SCA1]), growth factors (insulin-like growth factor binding protein 4 and transforming growth factor beta2), glyceraldehyde-6-phosphate dehydrogenase, an expressed sequence tag, and a novel cDNA showing homology to a LIM domain sequence. Two- to sevenfold induction of the endogenous gene expression was observed at 24 h postinfection, and induction continued up to 72 h, although the timing of gene expression varied among the identified genes. In contrast to that observed in endothelial cells, the Ad vector-mediated induction of gene expression was not found following Ad vector infection of primary human dermal fibroblasts or human alveolar macrophages. Empty Ad capsids did not induce endogenous gene expression in endothelial cells. Interestingly, additional deletion of the E4 gene obviated the upregulation of genes in endothelial cells by the E1(-) E3(-) Ad vector, suggesting that genes carried by the E4 region play a central role in modifying target cell gene expression. These findings are consistent with the notion that efficient transfer of exogenous genes to endothelial cells by first-generation Ad vectors comes with the price that these vectors also induce the expression of a variety of cellular genes.
- Published
- 1999
- Full Text
- View/download PDF
413. Endothelial trophic support of neuronal production and recruitment from the adult mammalian subependyma.
- Author
-
Leventhal C, Rafii S, Rafii D, Shahar A, and Goldman SA
- Subjects
- Animals, Brain-Derived Neurotrophic Factor antagonists & inhibitors, Brain-Derived Neurotrophic Factor biosynthesis, Brain-Derived Neurotrophic Factor metabolism, Cell Differentiation drug effects, Cell Differentiation physiology, Cell Movement drug effects, Cell Movement physiology, Cell Survival drug effects, Cell Survival physiology, Cells, Cultured, Coculture Techniques, Culture Media, Conditioned pharmacology, Endothelium, Vascular metabolism, Extracellular Space metabolism, Neurons drug effects, RNA, Messenger biosynthesis, Rats, Rats, Sprague-Dawley, Receptor Protein-Tyrosine Kinases physiology, Receptor, Ciliary Neurotrophic Factor, Receptors, Nerve Growth Factor physiology, Recombinant Fusion Proteins pharmacology, Endothelium, Vascular cytology, Ependyma cytology, Neurons cytology
- Abstract
Vascular endothelial cells are among the first cells that ventricular zone neuroblasts encounter during early development. The ventricular zone cells promote angiogenesis by the invading vasculature, with the release of endothelial mitogens. Yet the feedback support of young neurons by endothelial cells (ECs) has not hitherto been explored. We therefore asked whether ECs might participate in neuronal recruitment, by providing neurotrophic support to newly generated neurons. We used the neurogenic subependymal zone (SZ) of the adult rat forebrain as a model system, because of its well-characterized and relatively homogeneous population of neuronal precursor cells. We found that explants of the adult rat SZ raised on ECs generated more neurons, which survived longer, than explants raised on astrocytes, fibroblasts, or laminin. This endothelial trophic effect was humoral, in that it was also noted in SZ explants raised in noncontiguous coculture with ECs grown on porous inserts. RT-PCR for neurotrophin family members revealed that cultures of both human brain- and umbilical cord-derived ECs produced brain-derived neurotrophic factor (BDNF) mRNA, but no detectable NGF, NT-3, or NT-4 mRNA. ELISA revealed that BDNF protein was secreted by ECs into the medium at >1 ng/ml. The neurotrophic effect of ECs could be replaced by added BDNF, and was blocked by addition of 5 microg/ml trkB-Fc to endothelial-SZ cocultures. Thus, endothelial cells can act as sources of secreted BDNF, through which the capillary microvasculature may act to support neuronal recruitment and survival in the CNS., (Copyright 1999 Academic Press.)
- Published
- 1999
- Full Text
- View/download PDF
414. E1(-)E4(+) adenoviral gene transfer vectors function as a "pro-life" signal to promote survival of primary human endothelial cells.
- Author
-
Ramalingam R, Rafii S, Worgall S, Brough DE, and Crystal RG
- Subjects
- Adenovirus E1 Proteins genetics, Adenovirus E4 Proteins genetics, Apoptosis, Cell Division, Cells, Cultured, Culture Media, Gene Deletion, Genetic Vectors, Glucuronidase biosynthesis, Glucuronidase genetics, Growth Substances pharmacology, Humans, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-bcl-2 genetics, Transfection methods, Umbilical Veins, bcl-2-Associated X Protein, Adenoviridae genetics, Cell Survival, Endothelium, Vascular cytology, Endothelium, Vascular physiology
- Abstract
Although endothelial cells are quiescent and long-lived in vivo, when they are removed from blood vessels and cultured in vitro they die within days to weeks. In studies of the interaction of E1(-)E4(+) replication-deficient adenovirus (Ad) vectors and human endothelium, the cells remained quiescent and were viable for prolonged periods. Evaluation of these cultures showed that E1(-)E4(+) Ad vectors provide an "antiapoptotic" signal that, in association with an increase in the ratio of Bcl2 to Bax levels, induces the endothelial cells to enter a state of "suspended animation," remaining viable for at least 30 days, even in the absence of serum and growth factors. Although the mechanisms initiating these events are unclear, the antiapoptoic signal requires the presence of E4 genes in the vector genome, suggesting that one or more E4 open reading frames of subgroup C Ad initiate a "pro-life" program that modifies cultured endothelial cells to survive for prolonged periods.
- Published
- 1999
415. Interleukin-5 and the regulation of eosinophil production.
- Author
-
Roboz GJ and Rafii S
- Subjects
- Animals, Hematopoiesis drug effects, Humans, Rhinitis, Allergic, Perennial physiopathology, Rhinitis, Allergic, Seasonal physiopathology, Eosinophils cytology, Interleukin-5 physiology
- Abstract
Eosinophils play important roles in adaptive immune responses, inflammatory processes, and disease states. Recently, considerable research has been devoted to better defining the normal and abnormal biology of these cells, specifically their origin and mechanisms of stimulation, chemotaxis, regulation, and activation. Interleukin-5 has been identified as a major regulator of eosinophil development and function. This review highlights current literature on interleukin-5 and eosinophil production. Areas covered include molecular signaling, physiologic sources of interleukin-5, and interactions between interleukin-5, eosinophils, and the bone marrow microenvironment. Clinical correlates are also presented.
- Published
- 1999
- Full Text
- View/download PDF
416. Regulation of transendothelial migration of hematopoietic progenitor cells.
- Author
-
Möhle R, Bautz F, Rafii S, Moore MA, Brugger W, and Kanz L
- Subjects
- Animals, Bone Marrow Cells cytology, Bone Marrow Cells physiology, Cell Adhesion Molecules physiology, Cell Communication, Chemokines physiology, Cytokines physiology, Humans, Models, Biological, Stromal Cells cytology, Stromal Cells physiology, Endothelium, Vascular physiology, Hematopoietic Stem Cells physiology
- Abstract
Transendothelial migration of hematopoietic progenitor cells occurs in the bone marrow during mobilization and homing, and may therefore play a key role in the trafficking of hematopoietic stem cells. We hypothesize that adhesion molecules, chemokines, and paracrine cytokines are involved in this multifactorial process. As suggested in several studies, downregulation of adhesion molecules (e.g., integrins) may contribute to mobilization of progenitors due to a decreased avidity to bone marrow stromal and endothelial cells, which express the corresponding ligands. Using an in vitro model of transendothelial migration, we have shown that only a small number of more mature, committed progenitors migrates spontaneously under the control of adhesion molecules of the beta-2-integrin family and their corresponding endothelial/stromal ligands. However, transendothelial migration of progenitors in vitro is substantially enhanced by the chemokine stromal cell-derived factor-1 (SDF-1), which is constitutively produced by bone marrow stromal cells. More primitive progenitors also respond to this chemokine. In addition, the ligand for SDF-1, the chemokine receptor CXCR-4, is expressed in greater levels on bone marrow CD34+ cells as compared to mobilized progenitors, suggesting that downregulation of chemokine receptors occurs during progenitor mobilization. Indeed, bone marrow CD34+ cells migrate more avidly in response to SDF-1 than mobilized progenitors. Paracrine cytokines may also play a role in hematopoietic stem cell trafficking, since growth factor-stimulated hematopoietic cells produce cytokines that act on endothelial cells (e.g., vascular endothelial growth factor, VEGF), modifying their proliferation, motility, permeability, and fenestration. We conclude that transendothelial migration of hematopoietic progenitor cells is regulated by adhesion molecules, paracrine cytokines, and chemokines. Cytotoxic therapy as well as exogenously administered hematopoietic growth factors may affect adhesion molecule expression, the local cytokine and chemokine milieu, and chemokine receptor expression, which indirectly results in mobilization of hematopoietic stem cells.
- Published
- 1999
- Full Text
- View/download PDF
417. Access to coronary artery bypass surgery by race/ethnicity and gender among patients who are appropriate for surgery.
- Author
-
Hannan EL, van Ryn M, Burke J, Stone D, Kumar D, Arani D, Pierce W, Rafii S, Sanborn TA, Sharma S, Slater J, and DeBuono BA
- Subjects
- Aged, Coronary Angiography, Female, Health Services Accessibility standards, Health Services Research, Humans, Male, Middle Aged, Multivariate Analysis, New York, Patient Selection, Practice Guidelines as Topic, Prospective Studies, Referral and Consultation statistics & numerical data, Severity of Illness Index, Sex Factors, Black or African American statistics & numerical data, Coronary Artery Bypass statistics & numerical data, Health Services Accessibility statistics & numerical data, Hispanic or Latino statistics & numerical data, White People statistics & numerical data
- Abstract
Objective: The study sought to determine if there were race/ethnicity or gender differences in access to coronary artery bypass graft (CABG) surgery among patients who have been designated as appropriate and as necessary for that surgery according to the RAND methodology., Methods: RAND appropriateness and necessity criteria were used to identify a race/gender stratified sample of postangiography patients who would benefit from coronary artery bypass graft surgery. These patients were tracked for 3 months to determine if they had undergone coronary artery bypass graft surgery in New York State. Subjects were a total of 1,261 postangiography patients in eight New York hospitals in 1994 to 1996. Measures included percentages of patients for whom coronary artery bypass graft surgery was appropriate and necessary undergoing surgery by race/ethnicity and gender, as well as multivariate odds ratios for race/ethnicity and gender., Results: After controlling for age, payer, number of vessels diseased, and presence of left main disease, African-American and Hispanic patients were found to be significantly less likely to undergo coronary artery bypass graft surgery than white non-Hispanic patients (respective odds ratios 0.64 and 0.60). When "necessity" was used as a criterion instead of "appropriateness," significant differences in access for African-American patients remained. The gatekeeper physician recommended surgery only 10% of the time that patients did not undergo "appropriate" coronary artery bypass graft surgery, and this percentage did not vary significantly by race/ethnicity or gender of the patient., Conclusions: Even after controlling for appropriateness and necessity for coronary artery bypass graft surgery in a prospective study, African-American patients had significant access problems in obtaining coronary artery bypass graft surgery. These problems appeared not to be related to patient refusals.
- Published
- 1999
- Full Text
- View/download PDF
418. [Adenovirus long-term expression of thrombopoietin in vivo: a new model for myeloproliferative syndrome and osteomyelofibrosis].
- Author
-
Frey BM, Rafii S, Crystal RG, and Moore MA
- Subjects
- Animals, Bone Marrow pathology, Female, Gene Expression physiology, Humans, Liver pathology, Lung pathology, Male, Mice, Mice, Inbred BALB C, Mice, Inbred NOD, Mice, SCID, Spleen pathology, Mastadenovirus genetics, Myeloproliferative Disorders genetics, Primary Myelofibrosis genetics, Thrombopoietin genetics
- Abstract
Using a new adenoviral vector (Ad) construct, we expressed human thrombopoietin (TPO) cDNA (AdTPO) in mice with various inherited immune deficiency syndromes such as nude, SCID and NOD-SCID mice. Immune normal Balb/c mice and a vector construct without TPOcDNA (AdNull), respectively, were used for controls. All animals (3 per group) were treated with a single application of 10(9) PFU (plaque forming unit) of Ad (AdTPO or AdNull) intraperitoneally on day 0. Four to 5 weeks following AdTPO administration, SCID and NOD-SCID mice demonstrated peak concentration of PLT of 12- to 14-fold normal value simultaneously with maximum concentration of PMNs (10- to 12-fold normal value). Later on these animals had a chronic thrombocytosis. In contrast, Balb/c mice and nude mice experienced PLT peak concentration of 4- to 6-fold normal value without granulocytosis 1 to 2 weeks following AdTPO treatment. Only nude mice had chronically elevated PLTs. In contrast, Balb/c mice developed thrombocytopenia due to cross-reacting anti-TPO antibodies. Animals with chronic thrombocytosis revealed increased content of CFU-G/GM, CFU-GEMM and CFU-Meg in bone marrow compared with controls. In contrast, Balb/c mice showed decreased content of CFUs if anti-TPO-antibodies were present. Histologically, only SCID mice developed severe osteomyelofibrosis and osteomyelosclerosis, hepato-splenomegaly, extramedullary hematopoiesis in liver and lung and ultimately suffered of progressive pancytopenia, anisocytosis, fragmentocytosis and a lethal wasting syndrome. In contrast, NOD-SCID mice which demonstrated similar extent of TPO overexpression and in addition to the B- and T-cellular immune deficiency harbour defective monocytes and macrophages, did not develop fibrotic changes of the bone marrow. From these results, we conclude (1) chronic TPO overexpression in vivo may lead to thrombocytosis and granulocytosis with expansion of CFU-GM, -GEMM and -Meg; (2) in vivo expression of adenovirally mediated TPOcDNA depends on immune competency of the host; (3) functionally normal monocytes and macrophages are indispensable for development of secondary osteomyelofibrosis and (4) adenovirally mediated expression of xenogeneic transgenes may brake immune tolerance for the respective self protein leading to autoimmune phenomena. Our in vivo model might provide further insights into the pathophysiology of secondary osteomyelofibrosis and may prove useful in designing new strategies for immune therapies of cancer.
- Published
- 1998
419. Transformation of primary human endothelial cells by Kaposi's sarcoma-associated herpesvirus.
- Author
-
Flore O, Rafii S, Ely S, O'Leary JJ, Hyjek EM, and Cesarman E
- Subjects
- Antigens, Viral biosynthesis, Cell Adhesion, Cell Division, Cell Survival, Cells, Cultured, DNA, Viral analysis, Endothelial Growth Factors physiology, Endothelium, Vascular pathology, Humans, Lymphokines physiology, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Growth Factor metabolism, Receptors, Vascular Endothelial Growth Factor, Telomerase metabolism, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Virus Replication, Cell Transformation, Neoplastic, Cell Transformation, Viral, Endothelium, Vascular virology, Herpesvirus 8, Human physiology, Sarcoma, Kaposi virology
- Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV), or human herpesvirus 8, is invariably present in Kaposi's sarcoma lesions. KSHV contains several viral oncogenes and serological evidence suggests that KSHV infection is necessary for the development of Kaposi's sarcoma, but cellular transformation by this virus has not so far been demonstrated. KSHV is found in the microvascular endothelial cells in Kaposi's sarcoma lesions and in the spindle 'tumour' cells, which are also thought to be of endothelial origin. Here we investigate the biological consequences of infecting human primary endothelial cells with purified KSHV particles. We find that infection causes long-term proliferation and survival of these cells, which are associated with the acquisition of telomerase activity and anchorage-independent growth. KSHV was present in only a subset of cells, and paracrine mechanisms were found to be responsible for the survival of uninfected cells. Their survival may have been mediated by upregulation of a receptor for vascular endothelial growth factor. Our results indicate that transformation of endothelial cells by KSHV, as well as paracrine mechanisms that are induced by this virus, may be critical in the pathogenesis of Kaposi's sarcoma.
- Published
- 1998
- Full Text
- View/download PDF
420. Transendothelial migration of megakaryocytes in response to stromal cell-derived factor 1 (SDF-1) enhances platelet formation.
- Author
-
Hamada T, Möhle R, Hesselgesser J, Hoxie J, Nachman RL, Moore MA, and Rafii S
- Subjects
- Cell Line, Chemokine CXCL12, Humans, Megakaryocytes drug effects, Polyploidy, Receptors, CXCR4 biosynthesis, Blood Platelets physiology, Bone Marrow physiology, Chemokines, CXC physiology, Chemotaxis physiology, Endothelium, Vascular physiology, Megakaryocytes physiology
- Abstract
Although thrombopoietin has been shown to promote megakaryocyte (MK) proliferation and maturation, the exact mechanism and site of platelet formation are not well defined. Studies have shown that MKs may transmigrate through bone marrow endothelial cells (BMEC), and release platelets within the sinusoidal space or lung capillaries. In search for chemotactic factor(s) that may mediate transmigration of MKs, we have discovered that mature polyploid MKs express the G protein-coupled chemokine receptor CXCR4 (Fusin, LESTR). Therefore, we explored the possibility that stromal cell-derived factor 1 (SDF-1), the ligand for CXCR4, may also induce transendothelial migration of mature MKs. SDF-1, but not other CXC or CC chemokines, was able to mediate MK migration (ED50 = 125 pmol/liter). The MK chemotaxis induced by SDF-1 was inhibited by the CXCR4-specific mAb (12G5) and by pertussis toxin, demonstrating that signaling via the G protein-coupled receptor CXCR4 was necessary for migration. SDF-1 also induced MKs to migrate through confluent monolayers of BMEC by increasing the affinity of MKs for BMEC. Activation of BMEC with interleukin 1beta resulted in a threefold increase in the migration of MKs in response to SDF-1. Neutralizing mAb to the endothelial-specific adhesion molecule E-selectin blocked the migration of MKs by 50%, suggesting that cellular interaction of MKs with BMEC is critical for the migration of MKs. Light microscopy and ploidy determination of transmigrated MKs demonstrated predominance of polyploid MKs. Virtually all platelets generated in the lower chamber also expressed CXCR4. Platelets formed in the lower chamber were functional and expressed P-selectin (CD62P) in response to thrombin stimulation. Electron microscopy of the cells that transmigrated through the BMEC monolayers in response to SDF-1 demonstrated the presence of intact polyploid MKs as well as MKs in the process of platelet formation. These results suggest that SDF-1 is a potent chemotactic factor for mature MKs. Expression of CXCR4 may be the critical cellular signal for transmigration of MKs and platelet formation.
- Published
- 1998
- Full Text
- View/download PDF
421. Evidence for circulating bone marrow-derived endothelial cells.
- Author
-
Shi Q, Rafii S, Wu MH, Wijelath ES, Yu C, Ishida A, Fujita Y, Kothari S, Mohle R, Sauvage LR, Moore MA, Storb RF, and Hammond WP
- Subjects
- Animals, Antigens, CD34, Cell Differentiation drug effects, Cells, Cultured, Dogs, Endothelium, Vascular metabolism, Lipoproteins, LDL metabolism, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, von Willebrand Factor metabolism, Endothelial Growth Factors pharmacology, Endothelium, Vascular cytology, Fibroblast Growth Factor 2 pharmacology, Hematopoietic Stem Cells cytology, Insulin-Like Growth Factor I pharmacology, Lymphokines pharmacology
- Abstract
It has been proposed that hematopoietic and endothelial cells are derived from a common cell, the hemangioblast. In this study, we demonstrate that a subset of CD34(+) cells have the capacity to differentiate into endothelial cells in vitro in the presence of basic fibroblast growth factor, insulin-like growth factor-1, and vascular endothelial growth factor. These differentiated endothelial cells are CD34(+), stain for von Willebrand factor (vWF), and incorporate acetylated low-density lipoprotein (LDL). This suggests the possible existence of a bone marrow-derived precursor endothelial cell. To demonstrate this phenomenon in vivo, we used a canine bone marrow transplantation model, in which the marrow cells from the donor and recipient are genetically distinct. Between 6 to 8 months after transplantation, a Dacron graft, made impervious to prevent capillary ingrowth from the surrounding perigraft tissue, was implanted in the descending thoracic aorta. After 12 weeks, the graft was retrieved, and cells with endothelial morphology were identified by silver nitrate staining. Using the di(CA)n and tetranucleotide (GAAA)n repeat polymorphisms to distinguish between the donor and recipient DNA, we observed that only donor alleles were detected in DNA from positively stained cells on the impervious Dacron graft. These results strongly suggest that a subset of CD34+ cells localized in the bone marrow can be mobilized to the peripheral circulation and can colonize endothelial flow surfaces of vascular prostheses.
- Published
- 1998
422. The chemokine receptor CXCR-4 is expressed on CD34+ hematopoietic progenitors and leukemic cells and mediates transendothelial migration induced by stromal cell-derived factor-1.
- Author
-
Möhle R, Bautz F, Rafii S, Moore MA, Brugger W, and Kanz L
- Subjects
- Antigens, CD34, Chemokine CXCL12, Endothelium, Vascular pathology, Flow Cytometry, Humans, Tumor Cells, Cultured, Cell Movement drug effects, Cell Movement physiology, Chemokines, CXC pharmacology, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells pathology, Leukemia metabolism, Leukemia pathology, Receptors, CXCR4 physiology
- Abstract
The chemokine stromal cell-derived factor-1 (SDF-1) and its receptor CXCR-4 (fusin, LESTR) are likely to be involved in the trafficking of hematopoietic progenitor and stem cells, as suggested by the reduced bone marrow hematopoiesis in SDF-1-deficient mice and the chemotactic effect of SDF-1 on CD34+ progenitor cells. Migration of leukemic cells might also depend on the expression of chemokine receptors. Therefore, we analyzed expression of CXCR-4 on mobilized normal CD34+ progenitors and leukemic cells. In addition, SDF-1-induced transendothelial migration across a bone marrow endothelial cell layer was assessed in vitro. By flow cytometry, CXCR-4 was found to be expressed in significant amounts on circulating CD34+ hematopoietic progenitor cells, including more primitive subsets (CD34+/CD38- and CD34+/Thy-1+ cells). In accordance with the immunofluorescence data, CD34+ progenitors efficiently migrated across endothelium in response to SDF-1 containing conditioned medium from the stromal cell line MS-5. Leukemic blasts (mostly CD34+) from patients with acute myeloblastic leukemia (AML) expressed variable amounts of CXCR-4, which was functionally active, as demonstrated by a positive correlation between the SDF-1-induced transendothelial migration and the cell surface density of CXCR-4 (r = 0.97). Also recombinant SDF-1beta induced migration of CXCR-4-positive leukemic blasts. The effect of both conditioned medium and recombinant SDF-1 was inhibited by a CXCR-4 blocking antibody. In contrast, CD34+ leukemic cell lines (KG1, KG1a, Kasumi-1, MOLM-1) expressed low levels or were negative for CXCR-4, and did not migrate. By reverse transcriptase-polymerase chain reaction (RT-PCR), however, basal levels of CXCR-4 mRNA were also detected in all leukemic cell lines. We conclude that CXCR-4 is expressed on CD34+ cells including more primitive, pluripotent progenitors, and may therefore play a role in the homing of hematopoietic stem cells. CXCR-4 expressed in variable amounts on primary AML leukemic cells is functionally active and may be involved in the trafficking of malignant hematopoietic cells.
- Published
- 1998
423. High-efficiency gene transfer into ex vivo expanded human hematopoietic progenitors and precursor cells by adenovirus vectors.
- Author
-
Frey BM, Hackett NR, Bergelson JM, Finberg R, Crystal RG, Moore MA, and Rafii S
- Subjects
- Cells, Cultured, Genes, Reporter, Green Fluorescent Proteins, Humans, Luminescent Proteins genetics, Adenoviridae, Gene Transfer Techniques, Genetic Vectors, Hematopoietic Stem Cells physiology
- Abstract
Replication-deficient adenoviral vectors (AdVec), which infect cycling and noncycling cells with high efficiency, low toxicity, and ease of delivery, provide ideal vehicles to study the expression of regulatory genes controlling different stages of hematopoiesis. To examine the infection efficiency of AdVec in hematopoietic precursor and progenitor cells, we used a replication-deficient adenovector expressing the humanized form of the cDNA for green fluorescent protein (AdGFP), permitting assessment of infection efficiency and kinetics of transgene expression in viable hematopoietic cells using flow cytometry and fluorescence microscopy. Flow-cytometric analysis of ex vivo expanded hematopoietic precursor cells infected with a multiplicity of infection (MOI) of 100 of AdGFP show that 78% of megakaryocytic (CD41a+ and CD42b+) cells, 82% of dendritic (CD1a+) cells, 41% of RBC precursors (glycophorin A+), and 32% of monocytic (CD14(+)) cells expressed GFP. Nineteen percent +/- 1% of freshly isolated CD34(+) cells from peripheral blood leukapheresis products infected under the same conditions expressed GFP. Morphologic evaluation of ex vivo expanded, AdGFP-infected CD34(+) cells showed normal maturation. The functional capacity of AdGFP-infected CD34(+) cells was analyzed by quantifying clonogeneic efficiency and proliferative capacity. Infection of CD34(+) progenitor cells with MOIs of 1 to 100 did not impair clonogeneic efficiency of CD34(+ )cells. However, MOI greater than 100 resulted in a significant inhibition of colony-forming unit-granulocyte/granulocyte-macrophage (CFU-G/GM) formation. In sequential dilution expansion over 3 weeks (Delta assay), the cytokine-driven proliferative potential of CD34(+) cells was not impaired following exposure to AdGFP at MOIs of 1 to 1,000. The GFP+ population expanded 10- to 15-fold at high MOIs (500 to 1,000), indicating multiple copies of the transgene in the initially infected CD34(+) cells, which were expressed in subsequent progenies. These data show that AdVec deliver transgenes with high efficiency and low toxicity to hematopoietic progenitor and precursor cells. Introduction of marker genes such as GFP into hematopoietic cells by AdVec will provide a valuable system for study of development, homing, and trafficking of hematopoietic precursor and progenitor cells in vitro and in vivo. Furthermore, these results provide insights into the design of gene therapy strategies for treatment of hematologic disorders by AdVec.
- Published
- 1998
424. Adenovector-mediated expression of human thrombopoietin cDNA in immune-compromised mice: insights into the pathophysiology of osteomyelofibrosis.
- Author
-
Frey BM, Rafii S, Teterson M, Eaton D, Crystal RG, and Moore MA
- Subjects
- Adenoviruses, Human immunology, Animals, Blood Platelets pathology, Bone Marrow Cells pathology, Clone Cells, Erythrocytes pathology, Gene Expression Regulation immunology, Genetic Vectors administration & dosage, Genetic Vectors metabolism, Granulocytes pathology, Humans, Injections, Intraperitoneal, Mice, Mice, Inbred BALB C, Mice, Inbred NOD, Mice, Nude, Mice, SCID, Platelet Count, Primary Myelofibrosis genetics, Primary Myelofibrosis pathology, Thrombopoietin administration & dosage, Thrombopoietin biosynthesis, Adenoviruses, Human genetics, DNA, Complementary biosynthesis, Genetic Vectors immunology, Immunocompromised Host genetics, Primary Myelofibrosis immunology, Primary Myelofibrosis physiopathology, Thrombopoietin genetics
- Abstract
Thrombopoietin (TPO) cDNA can be effectively delivered in vivo by adenovectors. Immune normal mice (BALB/c) and syngeneic mice with variable degrees of immune dysfunction nu, SCID, and NOD-SCID) were treated with an adenovirus vector expressing the human TPO cDNA (AdTPO). Platelet peaks were significantly higher in SCID and NOD-SCID mice compared with BALB/c and nu mice. Human plasma TPO concentration correlated with the platelet counts. SCID and NOD-SCID mice exhibited also granulocytosis and increased numbers of hemopoietic progenitors in bone marrow. Following platelet peak, BALB/c mice developed autoantibodies against murine TPO leading to thrombocytopenia and depletion of megakaryocytes and hemopoietic progenitors in bone marrow. AdTPO-treated SCID mice developed osteomyelofibrosis and extramedullary/extrasplenal hemopoiesis. In contrast, NOD-SCID mice with a similar magnitude of TPO overexpression did not show fibrotic changes in bone marrow. We conclude, first, that a chronic high level of TPO overexpression stimulates megakaryocytopoiesis and myelopoiesis leading to thrombocytosis and granulocytosis. Second, increased megakaryocytopoiesis is not sufficient for development of secondary osteomyelofibrosis. The functionally deficient monocytes and macrophages of NOD-SCID mice probably prevented fibrotic marrow changes. Third, immune deficiency enhances expression of adenovirally mediated transgenes, and fourth, xenogeneic transgene delivered by adenovector to a host with normal immune functions may induce loss of immune tolerance and autoimmune phenomenon.
- Published
- 1998
425. The role of endothelium in the regulation of hematopoietic stem cell migration.
- Author
-
Möhle R, Rafii S, and Moore MA
- Subjects
- Animals, Bone Marrow blood supply, Cell Movement, Cytokines pharmacology, Endothelium, Vascular drug effects, Growth Substances pharmacology, Hematopoietic Stem Cell Mobilization, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells drug effects, Humans, Microcirculation physiology, Endothelium, Vascular physiology, Hematopoietic Stem Cells physiology
- Abstract
Mobilization of hematopoietic progenitor cells appears to be a multifactorial process which is at least partially regulated at the level of bone marrow microvascular endothelium (BMEC). In order to study the regulation of progenitor cell migration by endothelium in vitro, methods have been developed to isolate BMEC from bone marrow aspirates. In addition, immortalized BMEC cell lines have been generated. Using an in vitro model of migration across bone marrow endothelium, we demonstrate that only a small number of more mature, committed progenitors migrate spontaneously. In this model, adhesion molecules of the beta2-integrin family and the corresponding endothelial ligands are involved. The low spontaneous migratory capacity suggests that, in addition to adhesion molecules which mediate direct cellular contacts, paracrine cytokines and chemokines may play a role in progenitor migration across endothelium. Growth-factor-stimulated hematopoietic cells can produce cytokines which act on endothelial cells (e.g., vascular endothelial growth factor, VEGF), modifying their motility, growth, permeability, and fenestration. Therefore, VEGF might be involved in the mobilization and homing of hematopoietic progenitor cells. Furthermore, transendothelial migration of progenitors in vitro is substantially enhanced by the chemokine stromal-cell-derived factor-1 (SDF-1), which is produced by bone marrow stromal cells. More primitive progenitors, which do not migrate spontaneously, also respond to this chemokine. We conclude that transendothelial progenitor cell migration is regulated by adhesion molecules, paracrine cytokines, and chemokines. Mobilizing hematopoietic growth factors stimulate proliferation of hematopoietic cells, which may indirectly result in changes of the local cytokine and chemokine milieu, adhesion molecule expression, and eventually the mobilization of hematopoietic progenitor cells.
- Published
- 1998
- Full Text
- View/download PDF
426. Expression of interleukin-5 by human bone marrow microvascular endothelial cells: implications for the regulation of eosinophilopoiesis in vivo.
- Author
-
Möhle R, Salemi P, Moore MA, and Rafii S
- Subjects
- Bone Marrow metabolism, Bone Marrow Cells metabolism, Cell Division, Cells, Cultured, Eosinophils physiology, Humans, Microcirculation, Polymerase Chain Reaction, RNA, Messenger metabolism, Sensitivity and Specificity, Bone Marrow blood supply, Endothelium, Vascular metabolism, Eosinophils cytology, Interleukin-5 metabolism
- Abstract
We have shown that bone marrow microvascular endothelial cells (BMEC) support growth and differentiation of haemopoietic progenitors in vitro by elaboration of haemopoietic cytokines. Since generation of eosinophils can be observed in these coculture experiments, and BMEC do not produce interleukin (IL)-3, we evaluated BMEC for expression of IL-5, a specific growth factor for the eosinophilic lineage. Using RT-PCR, IL-5 mRNA was expressed by BMEC after stimulation (12 h) with lipopolysaccharide (LPS), IL-1, IL-2 and phorbol myristate acetate (PMA), but not by resting BMEC, after stimulation with TNF-alpha or interferon (IFN)-gamma. Moreover, IFN-gamma suppressed expression of IL-5 in response to LPS and IL-2. The identity of the PCR products was confirmed by restriction enzyme digestion, which resulted in fragments of the predicted size. T lymphocytes were not present in the endothelial cultures as demonstrated by absence of CD2 mRNA. Using a sensitive (1 pg/ml) ELISA assay. IL-5 was detected after 48 h incubation of BMEC with IL-2 (4.1 pg/10(6) cells) or with a combination of LPS and IL-2 (4.8 pg). However, the number of eosinophils generated after 4 weeks coculture of CD34+ haemopoietic cells with BMEC was not increased by addition of IL-2. RT-PCR revealed that BMEC in coculture with haemopoietic cells expressed IL-5 even without addition of exogenous cytokines or stimulating agents. In conclusion, expression of IL-5 by BMEC can be stimulated by cytokines (IL-1, IL-2), LPS, PMA, and coculture with proliferating haemopoietic cells. Thus, BMEC may support proliferation and differentiation of eosinophils in the bone marrow. IFN-gamma represents a cytokine with an inhibitory effect on IL-5 expression by BMEC. In addition, eosinophilia in response to circulating IL-2 or bacterial products (LPS) in vivo may be partially mediated by BMEC or vascular endothelium.
- Published
- 1997
- Full Text
- View/download PDF
427. Regulation of hematopoiesis by microvascular endothelium.
- Author
-
Rafii S, Mohle R, Shapiro F, Frey BM, and Moore MA
- Subjects
- Antigens, CD34 metabolism, Blood Platelets physiology, Bone Marrow Cells cytology, Bone Marrow Cells physiology, Cell Communication, Cell Culture Techniques, Cell Differentiation, Cell Division, Endothelium, Vascular cytology, Hematopoietic Stem Cells physiology, Humans, Leukemia physiopathology, Megakaryocytes physiology, Microcirculation, Cytokines physiology, Endothelium, Vascular physiology, Hematopoiesis, Hematopoietic Stem Cells cytology
- Abstract
The bone marrow microenvironment is a complex three dimensional structure where hematopoietic stem cells proliferate, mature, migrate into the sinusoidal space, and enter the circulation in an exquisitely regulated fashion. Stromal cells within the BM microenvironment provide a suitable environment for self-renewal, proliferation and differentiation of hematopoietic stem cells. Within the hematopoietic microenvironment, whether it is embryonic yolk sac, fetal liver, or adult bone marrow, microvascular endothelium not only acts as a gatekeeper controlling the trafficking and homing of hematopoietic progenitors, but also provides cellular contact and secretes cytokines that allows for the preservation of the steady state hematopoiesis. Recently, homogenous monolayers of bone marrow endothelial cells (BMEC) have been isolated and cultivated in tissue culture. Long-term coculture studies have shown that BMEC monolayers are unique type of endothelium and can support long-term proliferation of hematopoietic progenitor cells particularly megakaryocytic and myeloid progenitor cells by constitutive elaboration of lineage-specific cytokines such as G-CSF, GM-CSF, M-CSF, Kit-ligand, IL6, FLK-2 ligand, and leukemia inhibitory factor. Direct cellular contact between hematopoietic progenitor cells and BMEC monolayers through specific adhesion molecules including beta1, beta2 integrins and selectins play a critical role in trafficking and possibly proliferation of hematopoietic stem cells. Dysfunction of microvascular endothelial cells within the hematopoietic microenvironment may result in stem cell disorders and progression to aplastic anemias, and contribute to graft failure during bone marrow transplantation. Further studies on the role of microvascular endothelium in the regulation of hematopoietic stem cell homing and proliferation may enhance our understanding of the pathophysiology of stem cell and leukemic disorders.
- Published
- 1997
- Full Text
- View/download PDF
428. Dendritic cells genetically modified with an adenovirus vector encoding the cDNA for a model antigen induce protective and therapeutic antitumor immunity.
- Author
-
Song W, Kong HL, Carpenter H, Torii H, Granstein R, Rafii S, Moore MA, and Crystal RG
- Subjects
- Adenocarcinoma prevention & control, Adenocarcinoma therapy, Adenoviridae immunology, Animals, Bone Marrow Cells immunology, Bone Marrow Cells virology, Bone Marrow Transplantation, Cell Line, Colonic Neoplasms prevention & control, Colonic Neoplasms therapy, Dendritic Cells virology, Gene Transfer Techniques, Genetic Vectors immunology, Humans, Lung Neoplasms immunology, Lung Neoplasms prevention & control, Lung Neoplasms secondary, Lymphocyte Activation genetics, Male, Mice, Mice, Inbred BALB C, Models, Immunological, Neoplasm Transplantation, T-Lymphocytes, Cytotoxic immunology, Adenocarcinoma immunology, Adenoviridae genetics, Colonic Neoplasms immunology, DNA, Complementary immunology, Dendritic Cells immunology, Dendritic Cells transplantation, Immunotherapy, Adoptive methods, beta-Galactosidase immunology
- Abstract
Dendritic cells (DCs) are potent antigen-presenting cells that play a critical role in the initiation of antitumor immune responses. In this study, we show that genetic modifications of a murine epidermis-derived DC line and primary bone marrow-derived DCs to express a model antigen beta-galactosidase (betagal) can be achieved through the use of a replication-deficient, recombinant adenovirus vector, and that the modified DCs are capable of eliciting antigen-specific, MHC-restricted CTL responses. Importantly, using a murine metastatic lung tumor model with syngeneic colon carcinoma cells expressing betagal, we show that immunization of mice with the genetically modified DC line or bone marrow DCs confers potent protection against a lethal tumor challenge, as well as suppression of preestablished tumors, resulting in a significant survival advantage. We conclude that genetic modification of DCs to express antigens that are also expressed in tumors can lead to antigen-specific, antitumor killer cells, with a concomitant resistance to tumor challenge and a decrease in the size of existing tumors.
- Published
- 1997
- Full Text
- View/download PDF
429. Constitutive production and thrombin-induced release of vascular endothelial growth factor by human megakaryocytes and platelets.
- Author
-
Möhle R, Green D, Moore MA, Nachman RL, and Rafii S
- Subjects
- Alternative Splicing, Cells, Cultured, Endothelial Growth Factors genetics, Endothelium, Vascular cytology, Gene Expression, Hematopoiesis, Humans, Lymphokines genetics, Neovascularization, Physiologic, Platelet Glycoprotein GPIIb-IIIa Complex analysis, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Growth Factor metabolism, Receptors, Vascular Endothelial Growth Factor, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Blood Platelets metabolism, Endothelial Growth Factors metabolism, Lymphokines metabolism, Megakaryocytes metabolism, Thrombin pharmacology
- Abstract
We have shown that coculture of bone marrow microvascular endothelial cells with hematopoietic progenitor cells results in proliferation and differentiation of megakaryocytes. In these long-term cultures, bone marrow microvascular endothelial cell monolayers maintain their cellular integrity in the absence of exogenous endothelial growth factors. Because this interaction may involve paracrine secretion of cytokines, we evaluated megakaryocytic cells for secretion of cytokines, we evaluated megakaryocytic cells for secretion of vascular endothelial growth factor (VEGF). Megakaryocytes (CD41a+) were generated by ex vivo expansion of hematopoietic progenitor cells with kit-ligand and thrombopoietin for 10 days and further purified with immunomagnetic microbeads. Using reverse transcription-PCR, we showed that megakaryocytic cell lines (Dami, HEL) and purified megakaryocytes expressed mRNA of the three VEGF isoforms (121, 165, and 189 amino acids). Large quantities of VEGF (> 1 ng/10(6) cells/3 days) were detected in the supernatant of Dami cells, ex vivo-generated megakaryocytes, and CD41a+ cells isolated from bone marrow. The constitutive secretion of VEGF by CD41a+ cells was stimulated by growth factors of the megakaryocytic lineage (interleukin 3, thrombopoietin). Western blotting of heparin-Sepharose-enriched supernatant mainly detected the isoform VEGF165. In addition, immunohistochemistry showed intracytoplasmic VEGF in polyploid megakaryocytes. Thrombin stimulation of megakaryocytes and platelets resulted in rapid release of VEGF within 30 min. We conclude that human megakaryocytes produce and secrete VEGF in an inducible manner. Within the bone marrow microenvironment, VEGF secreted by megakaryocytes may contribute to the proliferation of endothelial cells. VEGF delivered to sites of vascular injury by activated platelets may initiate angiogenesis.
- Published
- 1997
- Full Text
- View/download PDF
430. Transendothelial migration of CD34+ and mature hematopoietic cells: an in vitro study using a human bone marrow endothelial cell line.
- Author
-
Möhle R, Moore MA, Nachman RL, and Rafii S
- Subjects
- Cell Adhesion Molecules immunology, Cell Adhesion Molecules metabolism, Cell Differentiation, Cell Division, Cell Line, Transformed, Cell Lineage, Cell Movement, Endothelium cytology, Hematopoietic Stem Cells classification, Hematopoietic Stem Cells metabolism, Humans, Immunoglobulin G immunology, Immunoglobulin G pharmacology, Immunophenotyping, Monocytes cytology, Antigens, CD34 analysis, Bone Marrow Cells, Hematopoietic Stem Cells cytology
- Abstract
To study the role of bone marrow endothelial cells (BMEC) in the regulation of hematopoietic cell trafficking, we have designed an in vitro model of transendothelial migration of hematopoietic progenitor cells and their progeny. For these studies, we have taken advantage of a human BMEC-derived cell line (BMEC-1), which proliferates independent of growth factors, is contact inhibited, and expresses adhesion molecules similar to BMEC in vivo. BMEC-1 monolayers were grown to confluency on 3 microns microporous membrane inserts and placed in 6-well tissue culture plates. Granulocytecolony stimulating factor (G-CSF)-mobilized peripheral blood CD34+ cells were added to the BMEC-1 monolayer in the upper chamber of the 6-well plate. After 24 hours of coincubation, the majority of CD34+ cells remained nonadherent in the upper chamber, while 1.6 +/- 0.3% of the progenitor cells had transmigrated. Transmigrated CD34 cells expressed a higher level of CD38 compared with nonmigrating CD34+ cells and may therefore represent predominantly committed progenitor cells. Accordingly, the total plating efficiency of the transmigrated CD34+ cells for lineage-committed progenitors was higher (14.0 +/- 0.1 v 7.8% +/- 1.5%). In particular, the plating efficiency of transmigrated cells for erythroid progenitors was 27-fold greater compared with nonmigrating cells (8.0% +/- 0.8% v 0.3% +/- 0.1%) and 5.5-fold compared with unprocessed CD34+ cells (2.2% +/- 0.4%). While no difference in the expression of the beta 1-integrin very late activation antigen (VLA)-4 and beta 2-integrin lymphocyte function-associated antigen (LFA)-1 was found, L-selectin expression on transmigrated CD34+ cells was lost, suggesting that shedding had occurred during migration. The number of transmigrated cells was reduced by blocking antibodies to LFA-1, while L-selectin and VLA-4 antibodies had no inhibitory effect. Continuous coculture of the remaining CD34+ cells in the upper chamber of the transwell inserts resulted in proliferation and differentiation into myeloid and megakaryocytic cells. While the majority of cells in the upper chamber comprised proliferating myeloid precursors such as promyelocytes and myelocytes, only mature monocytes and granulocytes were detected in the lower chamber. In conclusion, BMEC-1 cells support transmigration of hematopoietic progenitors and mature hematopoietic cells. Therefore, this model may be used to study mechanisms involved in mobilization and homing of CD34+ cells during peripheral blood progenitor cell transplantation and trafficking of mature hematopoietic cells.
- Published
- 1997
431. The effects of Flk-2/flt3 ligand as compared with c-kit ligand on short-term and long-term proliferation of CD34+ hematopoietic progenitors elicited from human fetal liver, umbilical cord blood, bone marrow, and mobilized peripheral blood.
- Author
-
Shapiro F, Pytowski B, Rafii S, Witte L, Hicklin DJ, Yao TJ, and Moore MA
- Subjects
- Antigens, CD34, Cell Division drug effects, Cells, Cultured, Female, Hematopoietic Stem Cells drug effects, Humans, Liver embryology, Organ Specificity, Pregnancy, Blood Cells cytology, Bone Marrow Cells, Fetal Blood cytology, Hematopoietic Stem Cells cytology, Liver cytology, Membrane Proteins pharmacology, Stem Cell Factor pharmacology
- Abstract
The Flk-2/flt3 ligand (FL) was evaluated and compared with c-kit ligand (KL) for its in vitro proliferative effects on CD34+ cells from human fetal liver, umbilical cord blood, bone marrow, and mobilized peripheral blood. Using a 7-day liquid culture system, FL in combination with interleukin-3 (IL-3), interleukin-6 (IL-6), and granulocyte colony-stimulating factor (G-CSF) was comparable with KL in combination with IL-3, IL-6, and G-CSF for the expansion of hematopoietic progenitors. When FL-containing cultures were assayed after 21 or 28 days, a greater number of progenitors were generated as compared with KL-containing cultures. Using bone marrow microvascular endothelial cells as support stroma, cultures supplemented with FL generated a greater number of progenitors in both the nonadherent and adherent layers at day 35. These data suggest that FL ligand, in combination with other cytokines, can be used for short-term ex vivo expansion of hematopoietic progenitors and facilitates the preservation and possible expansion of primitive cells capable of long-term generation of progenitors.
- Published
- 1996
- Full Text
- View/download PDF
432. BMEC-1: a human bone marrow microvascular endothelial cell line with primary cell characteristics.
- Author
-
Candal FJ, Rafii S, Parker JT, Ades EW, Ferris B, Nachman RL, and Kellar KL
- Subjects
- Cell Adhesion Molecules metabolism, Endothelium, Vascular immunology, Endothelium, Vascular metabolism, Humans, Immunophenotyping, Bone Marrow blood supply, Cell Line, Endothelium, Vascular cytology, Microcirculation cytology
- Abstract
Bone marrow microvascular endothelial cells (BMEC) are a functional component of the bone marrow stroma and have been shown to release hematopoietic regulatory factors as well as to selectively adhere and support the proliferation and differentiation of CD34+ hematopoietic progenitors. An early passage of these cells was immortalized by transfection with a vector (pSVT) encoding the large T antigen of SV40. The transformed cell line (CDC/CU.BMEC-1) expresses the SV40 transcript, retains the primary cell expression of Ulex europeaus and vWF/ FVIII, and incorporates acetylated low-density lipoprotein. In addition, BMEC-1 mirrors the phenotype of the primary cells with only a few exceptions. Both cell populations express the cellular adhesion molecules ICAM-1 and PECAM and also VCAM-1 and ELAM-1 after upregulation by tumor necrosis factor-alpha. The fibronectin receptor, hyaluronate receptor, collagen receptor, integrins VLA-alpha 3, VLA-alpha 4, and beta 4, endoglin, collagen IV, CD58, and CD61 are also expressed. The only differences are that BMEC-1 expresses higher levels of ICAM-1, CD58, CD34, CD36, and c-kit than the primary cells. The supernatants of primary cell and BMEC-1 contain stem cell factor, interleukin-6 (IL-6), granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-1 alpha, IL-11, and G-CSF. The functional significance of these hematopoietic cytokines was demonstrated in transwell cultures. Both cell populations supported the expansion of progeny from CD34+ cell-enriched cord blood mononuclear cells suspended in the upper chamber. These characteristics, plus the fact that BMEC-1 can be maintained independently of exogenous growth factors and exhibit contact inhibition, indicate that this cell line can be used to further define the role of BMEC in hematopoiesis.
- Published
- 1996
- Full Text
- View/download PDF
433. In vivo adenovirus vector-mediated transfer of the human thrombopoietin cDNA maintains platelet levels during radiation-and chemotherapy-induced bone marrow suppression.
- Author
-
Ohwada A, Rafii S, Moore MA, and Crystal RG
- Subjects
- Animals, Bone Marrow pathology, DNA, Complementary genetics, Female, Humans, Male, Mice, Mice, Inbred BALB C, Pancytopenia chemically induced, Pancytopenia etiology, Platelet Count, Recombinant Fusion Proteins metabolism, Recombinant Fusion Proteins toxicity, Spleen pathology, Thrombocytopenia etiology, Thrombopoietin biosynthesis, Thrombopoietin toxicity, Adenoviruses, Human genetics, Carboplatin toxicity, Genetic Therapy, Genetic Vectors genetics, Pancytopenia therapy, Radiation Injuries, Experimental therapy, Thrombocytopenia prevention & control, Thrombopoietin genetics, Whole-Body Irradiation adverse effects
- Abstract
Thrombopoietin (TPO, c-mpl ligand) has emerged as a major hematopoietic cytokine stimulating megakaryocyte proliferation, endomitosis, and platelet production. This study shows that a single administration of an adenovirus (Ad) vector encoding TPO (AdCMV.TPO) abrogates thrombocytopenia induced in mice by carboplatin and irradiation. Normal Balb/c mice receiving the vector had increased platelet counts peaking at 7 days and returning to baseline by day 15. Mice rendered pancytopenic with 500 rads and 1.2 mg of carboplatin had a nadir platelet count of five percent of the baseline. Mice receiving AdCMV.TPO 3 days before receiving irradiation and chemotherapy achieved a platelet nadir fourfold higher, and had significant reduction in duration of thrombocytopenia, than mice receiving the control Ad vector. Introduction of AdCMV.TPO the same day of chemotherapy and irradiation was equally effective in acceleration of platelet recovery, but administration of AdCMV.TPO 3 days after chemotherapy-radiation had little effect on platelet recovery. At 30 days after therapy bone marrow and spleen of mice treated with AdCMV.TPO were populated with a large number of polyploid megakaryocytes, but there was no evidence of circulating megakaryocytes in the liver or lungs and no pathologic bone abnormalities such as osteosclerosis or myelofibrosis. These observations suggest that an Ad vector may be an excellent delivery system to provide adequate TPO production to maintain platelet levels in circumstances associated with life-threatening thrombocytopenia.
- Published
- 1996
434. Human herpesvirus-8/Kaposi's sarcoma-associated herpesvirus is a new transmissible virus that infects B cells.
- Author
-
Mesri EA, Cesarman E, Arvanitakis L, Rafii S, Moore MA, Posnett DN, Knowles DM, and Asch AS
- Subjects
- Blotting, Southern, Cell Line, DNA Probes, DNA, Viral analysis, Fetal Blood, Genome, Viral, Herpesviridae isolation & purification, Herpesvirus 2, Saimiriine classification, Herpesvirus 4, Human classification, Humans, Polymerase Chain Reaction, Sarcoma, Kaposi etiology, Acquired Immunodeficiency Syndrome complications, B-Lymphocytes virology, Herpesviridae classification, Herpesviridae physiology, Sarcoma, Kaposi virology
- Abstract
Herpesviral DNA fragments isolated from AIDS-associated Kaposi's sarcoma (KS) tissue (KSHV-DNA) share homology with two lymphotropic oncogenic gamma-herpesviruses, Epstein-Barr virus and Herpesvirus saimiri, and are present in the lesions of more than 95% of HIV and non-HIV-associated forms of KS, AIDS-related body cavity-based lymphomas, and AIDS-related multicentric Castleman's disease. Here we show that BC-1, a KSHV-DNA-positive, body cavity-based lymphoma cell line, produces infective herpesviral particles carrying a linear 270-kb genome that specifically transmits KSHV-DNA to CD19+ B cells. Transmission of KSHV-DNA is dependent upon a biologically active, replicating virus, since it is blocked by UV irradiation and foscarnet, an inhibitor of viral DNA-polymerase. This study represents the first isolation and transmission of the human herpesvirus-8/KS-associated herpesvirus.
- Published
- 1996
- Full Text
- View/download PDF
435. Density enrichment and characterization of hematopoietic progenitors and stem cells from umbilical cord blood.
- Author
-
Yurasov SV, Flasshove M, Rafii S, and Moore MA
- Subjects
- Cell Culture Techniques methods, Cell Separation economics, Colony-Forming Units Assay, Cost-Benefit Analysis, Drug Resistance, Ficoll, Flow Cytometry, Hematopoiesis, Humans, Infant, Newborn, Iohexol, Methotrexate pharmacology, Povidone, Reproducibility of Results, Silicon Dioxide, Tetrahydrofolate Dehydrogenase genetics, Cell Separation methods, Centrifugation, Density Gradient methods, Fetal Blood cytology, Hematopoietic Stem Cells
- Abstract
Umbilical cord blood (UCB) is being used for hematopoietic rescue after myeloablative therapy in a rapidly growing number of patients. Recent developments of cord blood banking, ex vivo progenitor expansion and gene therapy techniques have raised the issue of efficient progenitor and stem cell enrichment procedures using UCB. We have used discontinuous density gradient techniques to analyze progenitor distribution in the mononuclear cell fraction of cord blood. This resulted in establishment of a highly reproducible, rapid, cost-effective single-step density separation method that generates a light density fraction, which when compared to conventional mononuclear cells has a high number of clonogenic progenitors, can be extensively expanded in vitro for up to 21 days and has the ability to sustain long-term hematopoiesis when inoculated on a preformed stromal layer. It can also serve as an efficient target for retrovirally mediated gene transfer, utilizing a vector expressing a mutated dihydrofolate reductase gene that confers methotrexate resistance.
- Published
- 1996
436. Characterization of hematopoietic cells arising on the textured surface of left ventricular assist devices.
- Author
-
Rafii S, Oz MC, Seldomridge JA, Ferris B, Asch AS, Nachman RL, Shapiro F, Rose EA, and Levin HR
- Subjects
- Antigens, CD analysis, Cell Division, Cells, Cultured, Cytokines pharmacology, Female, Flow Cytometry, Hematopoietic Stem Cells classification, Humans, Immunohistochemistry, Male, Middle Aged, Surface Properties, Heart-Assist Devices, Hematopoietic Stem Cells cytology
- Abstract
Background: Textured biomaterial surfaces in implantable left ventricular assist devices induce development of a nonthrombotic neointimal surface and allow elimination of anticoagulation therapy in device recipients. Characterization of the hematopoietic cells formed within the neointimal surfaces of these devices will contribute to our understanding of this unique neointima., Methods: The blood-contacting surface of seven ThermoCardiosystems left ventricular assist devices was removed, washed with phosphate-buffered saline solution, and digested with 0.1% collagenase for 15 to 20 minutes. The hematopoietic cells released from the explants were isolated and analyzed by flow cytometry and immuno-histochemical staining., Results: More than 80% +/- 6% of hematopoietic cells isolated in this fashion are of myelomonocytic origin and express CD14, CD15, and CD33 surface molecules. Four percent of cells express the CD34 surface marker, which suggests that the neointima is colonized by pluripotent hematopoietic stem cells. Continuous culture of these hematopoietic cells in the presence of the cytokines interleukin-3, c-kit ligand, granulocyte colony-stimulating factor resulted in tenfold expansion by day 7 and 25-fold expansion by day 14., Conclusions: Pluripotent hematopoietic cells with a high proliferative capacity colonize textured surfaces of left ventricular assist devices and may contribute to the development of a biologically nonthrombogenic neointima.
- Published
- 1995
- Full Text
- View/download PDF
437. Human bone marrow microvascular endothelial cells support long-term proliferation and differentiation of myeloid and megakaryocytic progenitors.
- Author
-
Rafii S, Shapiro F, Pettengell R, Ferris B, Nachman RL, Moore MA, and Asch AS
- Subjects
- Antigens, CD analysis, Base Sequence, Cell Differentiation, Cell Division, Cell Lineage, Cells, Cultured, Coculture Techniques, DNA, Complementary genetics, Endothelium, Vascular cytology, Hematopoietic Cell Growth Factors pharmacology, Hematopoietic Stem Cells drug effects, Humans, Megakaryocytes cytology, Megakaryocytes drug effects, Molecular Sequence Data, Polymerase Chain Reaction, Bone Marrow blood supply, Endothelium, Vascular metabolism, Hematopoiesis physiology, Hematopoietic Cell Growth Factors physiology, Hematopoietic Stem Cells cytology
- Abstract
Endothelial cells are a major component of the bone marrow (BM) microenvironment that regulate the trafficking and homing of hematopoietic progenitor and stem cells. In this paper, we provide evidence that BM endothelial cells (BMECs) also support multilineage hematopoiesis by elaboration of soluble cytokines. Hematopoietic progenitor cells incubated in direct contact with BMEC monolayers, or physically separated by microporous membrane, expanded five-fold to sevenfold at 7 days, in the absence of exogenous cytokines. Flow cytometric analysis of proliferating progenitor cells grown in the presence of BMEC monolayers showed that by day 14 of coculture, 70% to 80% of hematopoietic cells were myeloid, expressing CD15 or CD14, and 14% to 19% were megakaryocytic, expressing GPIIb/IIIa or GPIb. CD34+ cells derived from umbilical cord blood, cultured in the upper chamber of transwell culture plates, as well as the cells grown in direct contact with BMEC monolayers, generated progenitors for up to 70 days. Unstimulated BMEC monolayers constitutively produce interleukin-6, Kit-ligand, granulocyte colony-stimulating factor, and granulocyte macrophage colony-stimulating factor. These data suggest that BMEC regulate proliferation of hematopoietic progenitor cells and long-term culture initiating cells by elaboration of lineage-specific cytokines.
- Published
- 1995
438. Isolation and characterization of human bone marrow microvascular endothelial cells: hematopoietic progenitor cell adhesion.
- Author
-
Rafii S, Shapiro F, Rimarachin J, Nachman RL, Ferris B, Weksler B, Moore MA, and Asch AS
- Subjects
- Antigens, CD analysis, Antigens, CD34, Cell Adhesion, Endothelium, Vascular cytology, Endothelium, Vascular ultrastructure, Humans, Interleukin-1 pharmacology, Microcirculation cytology, Bone Marrow Cells, Endothelium, Vascular physiology, Hematopoietic Stem Cells physiology
- Abstract
To examine potential mechanisms by which hematopoiesis may be regulated by endothelial cells within the bone marrow (BM) microenvironment, we have devised a technique for the in vitro study of the interaction of human BM microvascular endothelial cells (BMEC) with hematopoietic cells. Microvessels isolated by collagenase digestion of spicules obtained from filtered BM aspirate were plated on gelatin-coated plastic dishes, and colonies of endothelial cells grown from microvessel explants were further purified by Ulex europaeus lectin affinity separation. BMEC monolayers isolated by this technique grew in typical cobblestone fashion, stained positively with antibody to factor VIII/von Willebrand factor, and incorporated acetylated LDL. Immunohistochemical studies showed that BM microvessels and BMEC monolayers express CD34, PECAM, and thrombospondin. Incubation of resting BMEC with BM mononuclear hematopoietic cells resulted in the selective adhesion of relatively large numbers of CD34+ progenitor cells and megakaryocytes. The binding of purified BM-derived CD34+ progenitor cells to BMEC was dependent on divalent cations and was partially blocked by antibodies to CD34. IL-1 beta treatment of BMEC monolayers resulted in an increase of CD34+ progenitor cell adhesion by mechanisms independent of CD34 or divalent cations. BMEC exhibit specific affinity for CD34+ progenitor cells and megakaryocytes, suggesting that the BM microvasculature may play a role in regulating the trafficking, proliferation, and differentiation of lineage specific hematopoietic elements, and possibly of pluripotent stem cells within the CD34+ population.
- Published
- 1994
439. Perforin gene expression in stimulated human peripheral blood T cells studied by in situ hybridization and northern blotting analysis.
- Author
-
Liu CC, Rafii S, Koizumi H, Granelli-Piperno A, and Young JD
- Subjects
- Blotting, Northern, Concanavalin A pharmacology, Humans, Interleukin-2 pharmacology, Lymphocyte Activation drug effects, Membrane Glycoproteins genetics, Muromonab-CD3, Nucleic Acid Hybridization, Perforin, Phytohemagglutinins, Pore Forming Cytotoxic Proteins, RNA, Messenger biosynthesis, T-Lymphocytes drug effects, Tetradecanoylphorbol Acetate pharmacology, Membrane Glycoproteins biosynthesis, T-Lymphocytes metabolism
- Abstract
In situ hybridization was used here to monitor the mRNA level of the pore-forming protein perforin in mitogen-stimulated primary peripheral blood human T cells. In situ hybridization was performed using sense and antisense ribonucleotide probes specific for this granule mediator. After IL-2 treatment, an increase in perforin mRNA could be detected by 4 h; they peaked at 12 h, and decreased after 24 h. The perforin mRNA was also induced in T cells treated with a combination of phorbol ester PMA plus lectin or OKT3 mAb. This latter induction followed slower kinetics, peaking at 48 h. For all three mitogens used, even at peak induction times less than 10% of T cells were labeled with perforin probe. Similar patterns of mRNA expression were observed for both unprimed T cells and lectin-primed T blasts. The induction response of mRNA due to IL-2 stimulation is probably mediated by the IL-2 receptor p75 chain since its mRNA was upregulated by IL-2 with a kinetics comparable to that associated with an increase of perforin mRNA. The p55 IL-2 receptor chain increased much more slowly than p75.
- Published
- 1992
- Full Text
- View/download PDF
440. How lymphocytes kill.
- Author
-
Young LH, Liu CC, Joag S, Rafii S, and Young JD
- Subjects
- Animals, Cells, Cultured, Humans, Membrane Proteins physiology, Perforin, Pore Forming Cytotoxic Proteins, Immunity, Cellular physiology, Killer Cells, Natural physiology, Membrane Glycoproteins, T-Lymphocytes, Cytotoxic physiology
- Abstract
Cytotoxic T lymphocytes and natural killer cells are potent killers of target cells. These lymphocytes have large cytoplasmic granules containing cytotoxic peptides and other factors. Several of these molecules have been isolated and their functions elucidated. These molecules may be directly involved in the killing of virus-infected and transformed cells as well as in the development of cell-mediated autoimmune disorders.
- Published
- 1990
- Full Text
- View/download PDF
441. Progression of the milking effect of the coronary artery.
- Author
-
Traube C, Rafii S, Greenfield DH, Levine RS, and Fox P
- Subjects
- Angina Pectoris diagnosis, Constriction, Pathologic diagnosis, Coronary Disease diagnosis, Female, Humans, Middle Aged, Coronary Disease physiopathology, Coronary Vessels physiopathology, Myocardial Contraction, Systole
- Abstract
Myocardial bridging and systolic milking may be involved in myocardial ischemia, myocardial infarction, and sudden death. We describe a patient whose milking effect progressed significantly over a two-year period. We conclude that bridging and milking effect can progress over a relatively short period. In those patients with prior reports of insignificant bridging and milking on coronary arteriograms and continued angina-like chest pain, a repeated catheterization may be warranted.
- Published
- 1981
- Full Text
- View/download PDF
442. Letter: Acute myocardial infarction in 'miliary tuberculosis'.
- Author
-
Kossowsky WA, Rafii S, and Gomez-Leon G
- Subjects
- Acute Disease, Adult, Humans, Male, Thromboembolism etiology, Tuberculosis, Cardiovascular etiology, Myocardial Infarction etiology, Tuberculosis, Miliary complications
- Published
- 1975
- Full Text
- View/download PDF
443. Arterial thromboembolism: a complication after insertion of Mobin-Uddin vena cava filter.
- Author
-
Flores L, Caldera F, Lotlikar U, Greenfield DH, Levine R, Rafii S, and Levowitz BS
- Subjects
- Adult, Aorta, Abdominal injuries, Female, Filtration adverse effects, Humans, Vena Cava, Inferior, Filtration instrumentation, Pulmonary Embolism prevention & control, Thromboembolism etiology
- Published
- 1982
444. Perforin and serine esterase gene expression in stimulated human T cells. Kinetics, mitogen requirements, and effects of cyclosporin A.
- Author
-
Liu CC, Rafii S, Granelli-Piperno A, Trapani JA, and Young JD
- Subjects
- DNA isolation & purification, Gene Expression, Humans, Interleukin-2 pharmacology, Kinetics, Perforin, Pore Forming Cytotoxic Proteins, RNA, Messenger analysis, Tetradecanoylphorbol Acetate pharmacology, Cyclosporins pharmacology, Esterases genetics, Lymphocyte Activation, Membrane Glycoproteins, Membrane Proteins genetics, T-Lymphocytes metabolism
- Abstract
A pore-forming protein (PFP; perforin) and various serine esterases (SE) have been identified in the cytoplasmic granules of CTL and NK cells. Perforin and several SE have recently been cloned. Northern blotting analysis was performed here using cDNA probes specific for human perforin and two SE (SE 1/HS and SE 2/GB) to monitor the levels of specific mRNAs in mitogen-stimulated primary human T cells. These mRNAs were rapidly induced by IL-2 with optimal responses at 300 U/ml. After IL-2 treatment, mRNAs for perforin, SE 1, and SE 2 peaked at 12-24 h and decreased after 48 h. The three mRNAs were also induced in T cells treated with a combination of PMA plus lectin, OKT3 mAb, or plastic-adherent accessory cells. However, the induction induced by PMA/mitogen followed a slower kinetics, peaking at 48 h. In general, we found that SE 1 mRNA was more readily induced by IL-2, while SE 2 responded better to PMA/mitogen. Similar patterns of mRNA expression were observed for both unprimed T cells and PHA-primed T blasts. After stimulation with IL-2 and PMA/mitogen, the T8+ subset was shown to be the main producer of perforin, SE 1, and SE 2. Low levels of all three mRNAs, however, were also detected in the T4+ subset. The induction of all three mRNAs by either IL-2 or PMA/mitogen was partially blocked by the immunosuppressive drug cyclosporin A (CsA), but not by the biologically inactive analogue cyclosporin H. Together, these results point to some similarities and differences with upregulation of granule mediator mRNAs relative to lymphokine mRNAs. Both sets of genes require two signals for their induction by mitogens. In contrast to lymphokines, there is a strong response of granule mRNAs to IL-2, and the induction of these transcripts is only partially blocked by CsA.
- Published
- 1989
- Full Text
- View/download PDF
445. Iodine-131 MIBG scintigraphy in small cell lung cancer.
- Author
-
Wadler S, Tai K, Chervu LR, Rafii S, Landau L, Blaufox MD, and Wiernik PH
- Subjects
- 3-Iodobenzylguanidine, APUD Cells metabolism, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Radionuclide Imaging, Carcinoma, Small Cell diagnostic imaging, Iodine Radioisotopes, Iodobenzenes, Lung Neoplasms diagnostic imaging
- Abstract
There is a well documented relationship between small cell carcinoma of the lung and the amine precursor uptake and decarboxylation system of endocrine cells (APUD). We attempted to exploit this association by employing the unique radiopharmaceutical, 131I-MIBG, which is recognized and taken up by the APUD system to monitor disease activity in patients with small cell carcinoma of the lung. A total of eight patients with biopsy proven, metastatic small cell carcinoma of the lung were studied. 131I-MIBG was synthesized in our laboratory by reacting metaiodobenzylamine hydrochloride with cyanamide with subsequent solid phase radioiodination. A dose of 0.5 mCi radiopharmaceutical was injected and images obtained on a large field of view gamma camera with a high energy parallel hole collimator at 2, 24, and either 48 or 72 h. Images were compared with known focal areas of metastatic disease demonstrable on computed tomographic scan, chest roentgenogram or bone scan. We were unable to detect reproducible correlations between the images produced by conventional radiographic techniques and the images produced by our radiopharmaceutical. We conclude that this agent will probably not be useful for localization of metastatic small cell lung carcinoma.
- Published
- 1989
- Full Text
- View/download PDF
446. Silo Filler's disease. Relapse following latent period.
- Author
-
RAFII S and GODWIN MC
- Subjects
- Humans, Chronic Disease, Lung Abscess, Medical Records, Recurrence, Silo Filler's Disease
- Published
- 1961
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.