22 results on '"Vitale M"'
Search Results
2. In vitro exposure to human immunodeficiency virus type 1 induces apoptotic cell death of the factor-dependent TF-1 hematopoietic cell line
- Author
-
Zauli, G, primary, Vitale, M, additional, Re, MC, additional, Furlini, G, additional, Zamai, L, additional, Falcieri, E, additional, Gibellini, D, additional, Visani, G, additional, Davis, BR, additional, and Capitani, S, additional
- Published
- 1994
- Full Text
- View/download PDF
3. The Induction of Megakaryocyte Differentiation Is Accompanied by Selective Ser133 Phosphorylation of the Transcription Factor CREB in Both HEL Cell Line and Primary CD34+Cells
- Author
-
Alessandra Bassini, Claudio Celeghini, Marco Marchisio, Giorgio Zauli, Marco Vitale, Davide Gibellini, Paola Secchiero, Sabina Pierpaoli, Lia Guidotti, Silvano Capitani, Zauli, G, Gibellini, D, Vitale, M, Secchiero, P, Celeghini, Claudio, Bassini, A, Pierpaoli, S, Marchisio, M, Guidotti, L, and Capitani, S.
- Subjects
Megakaryocyte differentiation ,Cellular differentiation ,Blotting, Western ,Immunology ,Antigens, CD34 ,CREB ,Biochemistry ,Cell Line ,megakaryocyte differentiation ,CD34 ,Serine ,Humans ,Enzyme Inhibitors ,Phosphorylation ,Cyclic AMP Response Element-Binding Protein ,Protein kinase A ,Protein Kinase Inhibitors ,Thrombopoietin ,Protein kinase C ,Flavonoids ,biology ,Kinase ,Cell Differentiation ,hemic and immune systems ,Cell Biology ,Hematology ,Flow Cytometry ,Hematopoietic Stem Cells ,Cell biology ,megakaryocyte differentiation, CREB, CD34 ,biology.protein ,Cancer research ,Megakaryocytes ,Protein Kinases ,Signal Transduction - Abstract
The addition of thrombopoietin (TPO) to HEL cells, cultured in a chemically defined serum-free medium, induced a rapid and dose-dependent phosphorylation of the transcription factor CREB on serine133 (PSer133), as detected by Western blot analysis. TPO also significantly increased the transactivation of CRE-dependent promoter, as determined in transient transfection experiments. On the other hand, neither erythropoietin (Epo; 1 to 10 U) nor hemin (10−7 mol/L) were able to significantly stimulate CREB-PSer133 or to activate CRE-promoter in HEL cells. Although pharmacological inhibitors of protein kinase C (chelerytrine and BIM) and protein kinase A (H-89) failed to block the TPO-mediated CREB phosphorylation, a specific inhibitor of the mitogen-activated protein kinases (PD98059) completely blocked the ability of TPO to stimulate CREB-PSer133. Moreover, PD98059 significantly decreased the ability of TPO to upregulate the surface expression of the αIIbβ3 megakaryocytic marker in HEL cells. In parallel, primary CD34+ hematopoietic cells were seeded in liquid cultures supplemented with 100 ng/mL of TPO and examined by immunofluorescence for the coexpression of αIIbβ3 and CREB-PSer133 at various time points. High levels of nuclear CREB-PSer133 were unequivocally demonstrated in αIIbβ3+cells, including morphologically recognizable megakaryocytes. Taken together, these data suggest that CREB plays a role in modulating the expression of genes critical for megakaryocyte differentiation and that the TPO-mediated CREB phosphorylation seems to be regulated via mitogen-activated protein kinases.
- Published
- 1998
- Full Text
- View/download PDF
4. PKCepsilon controls protection against TRAIL in erythroid progenitors
- Author
-
Prisco Mirandola, Lucio Cocco, Ivonne Sponzilli, Giuliana Gobbi, Cristina Ponti, Marco Vitale, Mirandola, P, Gobbi, G, Ponti, Cristina, Sponzilli, I, Cocco, L, and Vitale, M.
- Subjects
medicine.medical_treatment ,Receptor expression ,Cellular differentiation ,Immunology ,Blotting, Western ,TRAIL ,Antigens, CD34 ,Antineoplastic Agents ,Apoptosis ,Protein Kinase C-epsilon ,Biology ,Biochemistry ,TNF-Related Apoptosis-Inducing Ligand ,medicine ,Humans ,Erythropoiesis ,RNA, Messenger ,RNA, Small Interfering ,Erythropoietin ,Erythroid Precursor Cells ,Membrane Glycoproteins ,Reverse Transcriptase Polymerase Chain Reaction ,Tumor Necrosis Factor-alpha ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Flow Cytometry ,Cell biology ,PKCepsilon ,Up-Regulation ,Haematopoiesis ,Kinetics ,Cytokine ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,Cell culture ,CD34 ,Apoptosis Regulatory Proteins - Abstract
Apoptosis plays a central role in the regulation of the size of the hematopoietic stem cell pool as well as in the processes of cell differentiation along the various hematopoietic lineages. TRAIL is a member of the TNF family of cytokines with a known apoptogenic role against a variety of malignant cells and an emerging role in the modulation of normal hematopoiesis. Here we worked on the hypothesis that PKCϵ could act as a switch of the cellular response to TRAIL during erythropoiesis. We demonstrate that EPO-induced erythroid CD34 cells are insensitive to the apoptogenic effect of TRAIL at day 0 due to the lack of specific receptor expression. From day 3 onward, erythroid cells express surface death receptors and become sensitive to TRAIL up to day 7/8 when, notwithstanding death-receptor expression, the EPO-driven up-regulation of PKCϵ intracellular levels renders differentiating erythroid cells resistant to TRAIL likely via Bcl-2 up-regulation. Our conclusion is that in human CD34 cells, EPO promotes a series of events that, being finely regulated in their kinetics, restricts the sensitivity of these cells to TRAIL to a specific period of time, which therefore represents the “TRAIL window” for the negative regulation of erythroid-cell numbers.
- Published
- 2005
5. Proplatelet generation in the mouse requires PKCε-dependent RhoA inhibition.
- Author
-
Gobbi G, Mirandola P, Carubbi C, Masselli E, Sykes SM, Ferraro F, Nouvenne A, Thon JN, Italiano JE Jr, and Vitale M
- Subjects
- Animals, Blood Platelets metabolism, Blotting, Western, Cell Differentiation, Cells, Cultured, Fetus cytology, Fetus metabolism, Flow Cytometry, Fluorescent Antibody Technique, Humans, Liver cytology, Liver metabolism, Megakaryocytes metabolism, Mice, RNA, Small Interfering genetics, rhoA GTP-Binding Protein genetics, rhoA GTP-Binding Protein metabolism, Blood Platelets cytology, Megakaryocytes cytology, Protein Kinase C-epsilon metabolism, Thrombopoiesis physiology, Tubulin metabolism, rhoA GTP-Binding Protein antagonists & inhibitors
- Abstract
During thrombopoiesis, megakaroycytes undergo extensive cytoskeletal remodeling to form proplatelet extensions that eventually produce mature platelets. Proplatelet formation is a tightly orchestrated process that depends on dynamic regulation of both tubulin reorganization and Rho-associated, coiled-coil containing protein kinase/RhoA activity. A disruption in tubulin dynamics or RhoA activity impairs proplatelet formation and alters platelet morphology. We previously observed that protein kinase Cepsilon (PKCε), a member of the protein kinase C family of serine/threonine-kinases, expression varies during human megakaryocyte differentiation and modulates megakaryocyte maturation and platelet release. Here we used an in vitro model of murine platelet production to investigate a potential role for PKCε in proplatelet formation. By immunofluorescence we observed that PKCε colocalizes with α/β-tubulin in specific areas of the marginal tubular-coil in proplatelets. Moreover, we found that PKCε expression escalates during megakarocyte differentiation and remains elevated in proplatelets, whereas the active form of RhoA is substantially downregulated in proplatelets. PKCε inhibition resulted in lower proplatelet numbers and larger diameter platelets in culture as well as persistent RhoA activation. Finally, we demonstrate that pharmacological inhibition of RhoA is capable of reversing the proplatelet defects mediated by PKCε inhibition. Collectively, these data indicate that by regulating RhoA activity, PKCε is a critical mediator of mouse proplatelet formation in vitro.
- Published
- 2013
- Full Text
- View/download PDF
6. Abnormal VWF modifies megakaryocytopoiesis: studies of platelets and megakaryocyte cultures from patients with von Willebrand disease type 2B.
- Author
-
Nurden P, Gobbi G, Nurden A, Enouf J, Youlyouz-Marfak I, Carubbi C, La Marca S, Punzo M, Baronciani L, De Marco L, Vitale M, and Federici AB
- Subjects
- Amino Acid Substitution, Blood Platelets ultrastructure, Cell Membrane ultrastructure, Cell Size, Cells, Cultured ultrastructure, Cytoplasmic Granules ultrastructure, Humans, Megakaryocytes drug effects, Microscopy, Confocal, Microscopy, Electron, Mutation, Missense, Platelet Glycoprotein GPIb-IX Complex, Point Mutation, Protein Interaction Mapping, Thrombopoiesis drug effects, Thrombopoietin pharmacology, von Willebrand Disease, Type 2 genetics, von Willebrand Factor genetics, von Willebrand Factor pharmacology, Blood Platelets pathology, Megakaryocytes pathology, Membrane Glycoproteins physiology, Thrombopoiesis physiology, von Willebrand Disease, Type 2 blood, von Willebrand Factor physiology
- Abstract
von Willebrand factor (VWF) is an essential mediator of platelet adhesion to the vessel wall, but little is known about its role in megakaryocytopoiesis. VWF and its platelet receptor, glycoprotein Ibalpha (GPIbalpha), are both expressed during megakaryocyte (MK) maturation. This study was designed to evaluate whether the enhanced VWF-GPIbalpha interactions typical of patients with von Willebrand disease type 2B (VWD2B) modify platelet production. Platelets from 9 patients with VWD2B with 7 different gain-of-function mutations were examined by electron microscopy (EM) and immunofluorescence labeling. For the patients with VWD2B, EM characteristically showed variable numbers of structurally abnormal giant platelets, sometimes in agglutinates. Cultures of MKs from controls performed with or without purified VWF confirmed a positive influence of VWF on platelet production with specific inhibition by an antibody blocking VWF binding to GPIbalpha. VWD2B MK cultures examined by EM showed a disorganized demarcation membrane system and abnormal granule distribution. They produced platelets with structural abnormalities typical of VWD2B. Confocal examination of MK revealed limited extension of pseudopods with few large proplatelets. These results confirm that megakaryocytopoiesis is modified by the enhanced VWF-GPIbalpha interactions. These data obtained for controls and patients with VWD2B suggest a novel regulatory role of VWF-GPIbalpha interactions in platelet production.
- Published
- 2010
- Full Text
- View/download PDF
7. Phorbol ester-induced PKCepsilon down-modulation sensitizes AML cells to TRAIL-induced apoptosis and cell differentiation.
- Author
-
Gobbi G, Mirandola P, Carubbi C, Micheloni C, Malinverno C, Lunghi P, Bonati A, and Vitale M
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Apoptosis genetics, CASP8 and FADD-Like Apoptosis Regulating Protein genetics, CASP8 and FADD-Like Apoptosis Regulating Protein metabolism, Caspase 3 genetics, Caspase 3 metabolism, Caspase 8 genetics, Caspase 8 metabolism, Cell Culture Techniques, Cell Differentiation genetics, Down-Regulation drug effects, Drug Resistance, Neoplasm genetics, Drug Synergism, Gene Expression Regulation, Enzymologic drug effects, Gene Expression Regulation, Leukemic drug effects, Humans, K562 Cells, Leukemia, Myeloid, Acute enzymology, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Phorbol 12,13-Dibutyrate administration & dosage, Phorbol 12,13-Dibutyrate pharmacology, Phorbol Esters administration & dosage, Protein Kinase C-epsilon metabolism, TNF-Related Apoptosis-Inducing Ligand administration & dosage, TNF-Related Apoptosis-Inducing Ligand pharmacology, Tumor Cells, Cultured, Apoptosis drug effects, Cell Differentiation drug effects, Drug Resistance, Neoplasm drug effects, Leukemia, Myeloid, Acute drug therapy, Phorbol Esters pharmacology, Protein Kinase C-epsilon genetics, TNF-Related Apoptosis-Inducing Ligand therapeutic use
- Abstract
Despite the relevant therapeutic progresses made in these last 2 decades, the prognosis of acute myeloid leukemia (AML) remains poor. Phorbol esters are used at very low concentrations as differentiating agents in the therapy of myeloid leukemias. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), in turn, is a death ligand that spares normal cells and is therefore currently under clinical trials for cancer therapy. Emerging evidence, however, suggests that TRAIL is also involved in nonapoptotic functions, like cell differentiation. PKCepsilon is differentially modulated along normal hematopoiesis, and its levels modulate the response of hematopoietic precursors to TRAIL. Here, we investigated the effects of the combination of phorbol esters (phorbol ester 4-beta-phorbol-12,13-dibutyrate [PDBu]) and TRAIL in the survival/differentiation of AML cells. We demonstrate here that PDBu sensitizes primary AML cells to both the apoptogenic and the differentiative effects of TRAIL via PKCepsilon down-modulation, without affecting TRAIL receptor surface expression. We believe that the use of TRAIL in combination with phorbol esters (or possibly more specific PKCepsilon down-modulators) might represent a significative improvement of our therapeutic arsenal against AML.
- Published
- 2009
- Full Text
- View/download PDF
8. Calmodulin-dependent kinase IV links Toll-like receptor 4 signaling with survival pathway of activated dendritic cells.
- Author
-
Illario M, Giardino-Torchia ML, Sankar U, Ribar TJ, Galgani M, Vitiello L, Masci AM, Bertani FR, Ciaglia E, Astone D, Maulucci G, Cavallo A, Vitale M, Cimini V, Pastore L, Means AR, Rossi G, and Racioppi L
- Subjects
- Animals, CREB-Binding Protein genetics, CREB-Binding Protein immunology, CREB-Binding Protein metabolism, Calcium Signaling drug effects, Calcium-Calmodulin-Dependent Protein Kinase Type 4 genetics, Cell Survival drug effects, Cell Survival genetics, Cell Survival immunology, Cyclic AMP Response Element-Binding Protein genetics, Cyclic AMP Response Element-Binding Protein immunology, Cyclic AMP Response Element-Binding Protein metabolism, Dendritic Cells cytology, Dendritic Cells immunology, Humans, Lipopolysaccharides immunology, Lipopolysaccharides metabolism, Lipopolysaccharides pharmacology, Mice, Mice, Knockout, Monocytes cytology, Monocytes immunology, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins immunology, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-bcl-2, Spleen cytology, Spleen immunology, Spleen metabolism, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 immunology, bcl-X Protein genetics, bcl-X Protein immunology, bcl-X Protein metabolism, Calcium Signaling physiology, Calcium-Calmodulin-Dependent Protein Kinase Type 4 immunology, Calcium-Calmodulin-Dependent Protein Kinase Type 4 metabolism, Dendritic Cells metabolism, Monocytes metabolism, Toll-Like Receptor 4 metabolism
- Abstract
Microbial products, including lipopolysaccharide (LPS), an agonist of Toll-like receptor 4 (TLR4), regulate the lifespan of dendritic cells (DCs) by largely undefined mechanisms. Here, we identify a role for calcium-calmodulin-dependent kinase IV (CaMKIV) in this survival program. The pharmacologic inhibition of CaMKs as well as ectopic expression of kinase-inactive CaMKIV decrease the viability of monocyte-derived DCs exposed to bacterial LPS. The defect in TLR4 signaling includes a failure to accumulate the phosphorylated form of the cAMP response element-binding protein (pCREB), Bcl-2, and Bcl-xL. CaMKIV null mice have a decreased number of DCs in lymphoid tissues and fail to accumulate mature DCs in spleen on in vivo exposure to LPS. Although isolated Camk4-/- DCs are able to acquire the phenotype typical of mature cells and release normal amounts of cytokines in response to LPS, they fail to accumulate pCREB, Bcl-2, and Bcl-xL and therefore do not survive. The transgenic expression of Bcl-2 in CaMKIV null mice results in full recovery of DC survival in response to LPS. These results reveal a novel link between TLR4 and a calcium-dependent signaling cascade comprising CaMKIV-CREB-Bcl-2 that is essential for DC survival.
- Published
- 2008
- Full Text
- View/download PDF
9. The tryptophan catabolite L-kynurenine inhibits the surface expression of NKp46- and NKG2D-activating receptors and regulates NK-cell function.
- Author
-
Della Chiesa M, Carlomagno S, Frumento G, Balsamo M, Cantoni C, Conte R, Moretta L, Moretta A, and Vitale M
- Subjects
- Antigens, CD biosynthesis, Antigens, CD immunology, Cells, Cultured, GPI-Linked Proteins, Gene Expression Regulation drug effects, Humans, Immunity, Cellular drug effects, Immunity, Cellular immunology, Indoleamine-Pyrrole 2,3,-Dioxygenase metabolism, Killer Cells, Natural cytology, Killer Cells, Natural enzymology, Kynurenine immunology, Kynurenine metabolism, Lymphocyte Activation drug effects, Lymphocyte Activation immunology, Membrane Glycoproteins biosynthesis, NK Cell Lectin-Like Receptor Subfamily K, Natural Cytotoxicity Triggering Receptor 1, Natural Cytotoxicity Triggering Receptor 3, Receptors, IgG biosynthesis, Receptors, IgG immunology, Receptors, Immunologic biosynthesis, Receptors, Natural Killer Cell, Tryptophan immunology, Tryptophan metabolism, Gene Expression Regulation immunology, Indoleamine-Pyrrole 2,3,-Dioxygenase immunology, Killer Cells, Natural immunology, Kynurenine pharmacology, Membrane Glycoproteins immunology, Receptors, Immunologic immunology
- Abstract
Tryptophan (Trp) catabolism mediated by indoleamine 2,3-dioxygenase (IDO) plays a central role in the regulation of T-cell-mediated immune responses. In this study, we also demonstrate that natural killer (NK)-cell function can be influenced by IDO. Indeed, l-kynurenine, a Trp-derived catabolite resulting from IDO activity, was found to prevent the cytokine-mediated up-regulation of the expression and function of specific triggering receptors responsible for the induction of NK-cell-mediated killing. The effect of l-kynurenine appears to be restricted to NKp46 and NKG2D, while it does not affect other surface receptors such as NKp30 or CD16. As a consequence, l-kynurenine-treated NK cells display impaired ability to kill target cells recognized via NKp46 and NKG2D. Instead, they maintain the ability to kill targets, such as dendritic cells (DCs), that are mainly recognized via the NKp30 receptor. The effect of l-kynurenine, which is effective at both the transcriptional and the protein level, can be reverted, since NK cells were found to recover their functional competence after washing.
- Published
- 2006
- Full Text
- View/download PDF
10. PKCepsilon controls protection against TRAIL in erythroid progenitors.
- Author
-
Mirandola P, Gobbi G, Ponti C, Sponzilli I, Cocco L, and Vitale M
- Subjects
- Antigens, CD34 metabolism, Blotting, Western, Erythroid Precursor Cells cytology, Erythropoietin pharmacology, Flow Cytometry, Humans, Kinetics, Protein Kinase C-epsilon antagonists & inhibitors, Protein Kinase C-epsilon genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Small Interfering pharmacology, Reverse Transcriptase Polymerase Chain Reaction, TNF-Related Apoptosis-Inducing Ligand, Up-Regulation, Antineoplastic Agents metabolism, Apoptosis, Apoptosis Regulatory Proteins metabolism, Erythroid Precursor Cells metabolism, Erythropoiesis physiology, Membrane Glycoproteins metabolism, Protein Kinase C-epsilon pharmacology, Tumor Necrosis Factor-alpha metabolism
- Abstract
Apoptosis plays a central role in the regulation of the size of the hematopoietic stem cell pool as well as in the processes of cell differentiation along the various hematopoietic lineages. TRAIL is a member of the TNF family of cytokines with a known apoptogenic role against a variety of malignant cells and an emerging role in the modulation of normal hematopoiesis. Here we worked on the hypothesis that PKCepsilon could act as a switch of the cellular response to TRAIL during erythropoiesis. We demonstrate that EPO-induced erythroid CD34 cells are insensitive to the apoptogenic effect of TRAIL at day 0 due to the lack of specific receptor expression. From day 3 onward, erythroid cells express surface death receptors and become sensitive to TRAIL up to day 7/8 when, notwithstanding death-receptor expression, the EPO-driven up-regulation of PKCepsilon intracellular levels renders differentiating erythroid cells resistant to TRAIL likely via Bcl-2 up-regulation. Our conclusion is that in human CD34 cells, EPO promotes a series of events that, being finely regulated in their kinetics, restricts the sensitivity of these cells to TRAIL to a specific period of time, which therefore represents the "TRAIL window" for the negative regulation of erythroid-cell numbers.
- Published
- 2006
- Full Text
- View/download PDF
11. NK-dependent DC maturation is mediated by TNFalpha and IFNgamma released upon engagement of the NKp30 triggering receptor.
- Author
-
Vitale M, Della Chiesa M, Carlomagno S, Pende D, Aricò M, Moretta L, and Moretta A
- Subjects
- Cell Differentiation, Coculture Techniques, Histiocytosis, Non-Langerhans-Cell immunology, Histiocytosis, Non-Langerhans-Cell metabolism, Histocompatibility Antigens Class I metabolism, Humans, Membrane Glycoproteins deficiency, Natural Cytotoxicity Triggering Receptor 3, Perforin, Pore Forming Cytotoxic Proteins, Dendritic Cells cytology, Dendritic Cells immunology, Interferon-gamma metabolism, Killer Cells, Natural immunology, Membrane Glycoproteins metabolism, Receptors, Immunologic metabolism, Tumor Necrosis Factor-alpha metabolism
- Abstract
Natural killer (NK) cells were recently shown to play a relevant role in the process of dendritic cell (DC) maturation. This function is exerted either by direct DC stimulation or through killing those DCs that did not properly acquire a mature phenotype. While killing of immature DCs is dependent on the function of the NKp30 triggering receptor, the mechanism by which NK cells induce DC maturation is still unclear. In this study, we show that also the NK-mediated induction of DC maturation is dependent on NKp30. Upon NK/DC interaction, resulting in NKp30 engagement, NK cells produced tumor necrosis factor alpha (TNFalpha) (and interferon gamma [IFNgamma]) that, in turn, promoted DC maturation. Masking of NKp30 with specific monoclonal antibodies (mAbs) strongly reduced maturation of DCs cocultured with NK cells. In addition, supernatant from NK cells stimulated via NKp30 induced DC maturation, and this effect was neutralized by anti-TNFalpha antibodies (Abs). This NKp30 function is controlled by the HLA-specific inhibitory NK receptors. Accordingly, the ability to promote maturation was essentially confined to NK cells expressing the killer immunoglobulin-like receptor-negative (KIR-) NKG2A(dull) phenotype. Finally, the analysis of perforin-deficient NK cells allowed the dissection of the 2 NKp30-mediated NK-cell functions, since NKp30 could induce cytokine-dependent DC maturation in the absence of NK-mediated DC killing.
- Published
- 2005
- Full Text
- View/download PDF
12. Activated human NK and CD8+ T cells express both TNF-related apoptosis-inducing ligand (TRAIL) and TRAIL receptors but are resistant to TRAIL-mediated cytotoxicity.
- Author
-
Mirandola P, Ponti C, Gobbi G, Sponzilli I, Vaccarezza M, Cocco L, Zauli G, Secchiero P, Manzoli FA, and Vitale M
- Subjects
- Apoptosis Regulatory Proteins, CASP8 and FADD-Like Apoptosis Regulating Protein, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes metabolism, Cells, Cultured, Dactinomycin pharmacology, Flow Cytometry, GPI-Linked Proteins, Humans, Interleukin-2 pharmacology, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Killer Cells, Natural cytology, Killer Cells, Natural drug effects, Killer Cells, Natural metabolism, Membrane Glycoproteins genetics, Membrane Glycoproteins pharmacology, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Receptors, TNF-Related Apoptosis-Inducing Ligand, Receptors, Tumor Necrosis Factor genetics, Receptors, Tumor Necrosis Factor immunology, Receptors, Tumor Necrosis Factor, Member 10c, TNF-Related Apoptosis-Inducing Ligand, Transcription, Genetic drug effects, Tumor Necrosis Factor Decoy Receptors, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha pharmacology, Apoptosis drug effects, CD8-Positive T-Lymphocytes immunology, Cytotoxicity, Immunologic immunology, Killer Cells, Natural immunology, Lymphocyte Activation, Membrane Glycoproteins metabolism, Receptors, Tumor Necrosis Factor metabolism, Tumor Necrosis Factor-alpha metabolism
- Abstract
The expression of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and TRAIL receptors was investigated in resting and cytokine-activated purified primary human natural killer (NK) and CD8(+) T cells. Resting NK and CD8(+) T cells expressed the mRNA for all TRAIL receptors, but TRAIL-R4 was the only receptor clearly detectable on the surface of both cell types. NK cells were activated by interleukin 2 (IL-2) or IL-15, whereas CD8(+) T cells were activated by phytohemagglutinin (PHA) + IL-2 followed by IL-2 alone for up to 10 days. On activation, both cell types rapidly expressed TRAIL-R2 and TRAIL-R3, whose expression peaked at day 10 of culture. TRAIL-R1, however, was never expressed at any time point examined, whereas the expression of TRAIL-R4, which showed a progressive increase in CD8(+) T cells, remained constant in NK cells. Notwithstanding the expression of TRAIL-R2, recombinant TRAIL did not show any cytotoxic activity on either NK or CD8(+) T cells. Both resting and activated NK and CD8(+) T cells were found to express high levels of the 2 isoforms of c-FLIP (cellular Fas-associated death domain protein [FADD]-like IL-1-converting enzyme [FLICE]-inhibitory protein). Small interference RNA-mediated inhibition of c-FLIP expression in NK cells abrogated their resistance to the apoptotic effect of soluble TRAIL. Thus, once activated the major cytotoxic effector cells are potentially sensitive to TRAIL but are physiologically protected from its apoptotic action by intracellular level of c-FLIP.
- Published
- 2004
- Full Text
- View/download PDF
13. Expression and function of KIR and natural cytotoxicity receptors in NK-type lymphoproliferative diseases of granular lymphocytes.
- Author
-
Zambello R, Falco M, Della Chiesa M, Trentin L, Carollo D, Castriconi R, Cannas G, Carlomagno S, Cabrelle A, Lamy T, Agostini C, Moretta A, Semenzato G, and Vitale M
- Subjects
- Antibodies, Monoclonal, Antigens, CD genetics, Cell Line, Female, Gene Expression immunology, Gene Expression Profiling, Genotype, HLA Antigens genetics, Humans, Lectins, C-Type genetics, Lymphoproliferative Disorders immunology, Male, NK Cell Lectin-Like Receptor Subfamily D, Natural Cytotoxicity Triggering Receptor 1, Natural Cytotoxicity Triggering Receptor 3, Receptors, KIR, Killer Cells, Natural physiology, Lymphoproliferative Disorders physiopathology, Receptors, Immunologic genetics
- Abstract
Using monoclonal antibodies (mAbs) specific for different natural killer (NK) receptors, we studied the lymphocyte population from 18 patients with NK-type lymphoproliferative disease of granular lymphocytes (LDGL). The analysis of both resting and cultured NK cell populations demonstrated that these patients are frequently characterized by NK cells displaying a homogeneous staining with given anti-killer Ig-like receptor (anti-KIR) mAb (11 of 18 patients). In most patients NK cells were characterized by the CD94/NKG2A+ phenotype, whereas only a minor fraction of the cases expressed CD94/NKG2C. In 7 of these patients we could also assess the function of the various NK receptors. Remarkably those KIR molecules that, in each patient, homogeneously marked the NK cell expansion were found to display an activating function as determined by cross-linking with specific anti-KIR mAb. The KIR genotype analysis performed in 13 of 18 cases revealed that in NK-type LDGL certain activating KIRs, as well as certain infrequent KIR genotypes, were detected with higher frequencies as compared to previously analyzed healthy donors. Moreover, most KIR genotypes included multiple genes coding for activating KIRs. The analysis of non-HLA-specific triggering receptors indicated that the natural cytotoxicity receptors (NKp46, NKp30) were expressed at significantly low levels in freshly drawn NK cells from most patients analyzed. However, in most instances the expression of NKp46 and NKp30 could be up-regulated on culture in interleukin 2. Our data indicate that in NK-LDGL the expanded subset is frequently characterized by the expression of a given activating KIR, suggesting a direct role for these molecules in the pathogenetic mechanisms of this disorder.
- Published
- 2003
- Full Text
- View/download PDF
14. Analysis of natural killer cells in TAP2-deficient patients: expression of functional triggering receptors and evidence for the existence of inhibitory receptor(s) that prevent lysis of normal autologous cells.
- Author
-
Vitale M, Zimmer J, Castriconi R, Hanau D, Donato L, Bottino C, Moretta L, de la Salle H, and Moretta A
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 3, ATP-Binding Cassette Transporters genetics, Adult, Child, Codon, Nonsense, Cytotoxicity, Immunologic, Histocompatibility Antigens Class I immunology, Histocompatibility Antigens Class I metabolism, Humans, Immunity immunology, Killer Cells, Natural pathology, Natural Cytotoxicity Triggering Receptor 1, Receptors, Immunologic immunology, Receptors, Immunologic metabolism, Tumor Cells, Cultured, ATP-Binding Cassette Transporters metabolism, Killer Cells, Natural immunology
- Abstract
Natural killer (NK) cells are characterized by the ability to kill cells that lack HLA class I molecules while sparing autologous normal (HLA class I(+)) cells. However, patients with transporter-associated antigen processing (TAP) deficiency, though displaying strong reductions of HLA class I surface expression, in most instances do not experience NK-mediated autoimmune phenomena. A possible mechanism by which TAP(-/-) NK cells avoid autoreactivity against autologous HLA class I-deficient cells could be based on either quantitative or qualitative defects of surface receptors involved in NK cell triggering. In this study we show that NK cells derived from 2 patients with TAP2(-/-) express normal levels of all known triggering receptors. As revealed by the analysis of polyclonal and clonal NK cells, these receptors display normal functional capabilities and allow the killing of a panel of NK-susceptible targets, including autologous B-LCLs. On the other hand, TAP2(-/-) NK cells were unable to kill either allogeneic (HLA class I(+)) or autologous (HLA class I(-) ) phytohemagglutinin (PHA) blasts even in the presence of anti-HLA class I monoclonal antibody. These data suggest that TAP2(-/-) NK cells express still unknown inhibitory receptor(s) capable of down-regulating the NK cell cytotoxicity on binding to surface ligand(s) expressed by T cell blasts. Functional analyses, both at the polyclonal and at the clonal level, are consistent with the concept that the putative inhibitory receptor is expressed by virtually all TAP2(-/-) NK cells, whereas it is present only in rare NK cells from healthy persons. Another possibility would be that TAP2(-/-) NK cells are missing a still unidentified triggering receptor involved in NK cell-mediated killing of PHA blasts.
- Published
- 2002
- Full Text
- View/download PDF
15. Human herpesvirus 7 induces the functional up-regulation of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) coupled to TRAIL-R1 down-modulation in CD4(+) T cells.
- Author
-
Secchiero P, Mirandola P, Zella D, Celeghini C, Gonelli A, Vitale M, Capitani S, and Zauli G
- Subjects
- Apoptosis Regulatory Proteins, Blotting, Western, CD4 Antigens genetics, CD4-Positive T-Lymphocytes virology, Cell Line, Cytotoxicity, Immunologic, Humans, Membrane Glycoproteins genetics, Recombinant Proteins metabolism, TNF-Related Apoptosis-Inducing Ligand, Tumor Necrosis Factor-alpha genetics, fas Receptor immunology, Apoptosis, CD4-Positive T-Lymphocytes immunology, Gene Expression Regulation, Herpesvirus 7, Human immunology, Membrane Glycoproteins physiology, Tumor Necrosis Factor-alpha physiology
- Abstract
Human herpesvirus 7 (HHV-7) is endemic in the adult human population. Although HHV-7 preferentially infects activated CD4(+) T lymphocytes, the consequence of T-cell infection for viral pathogenesis and immunity are still largely unknown. HHV-7 infection induces apoptosis mostly in uninfected bystander cells but not in productively infected CD4(+) T cells. To dissect the underlying molecular events, the role of death-inducing ligands belonging to the tumor necrosis factor (TNF) cytokine superfamily was investigated. HHV-7 selectively up-regulated the expression of TNF-related apoptosis-inducing ligand (TRAIL), but not that of CD95 ligand or TNF-alpha in lymphoblastoid (SupT1) or primary activated CD4(+) T cells. Moreover, in a cell-to-cell-contact assay, HHV-7-infected CD4(+) T lymphocytes were cytotoxic for bystander uninfected CD4(+) T cells through the TRAIL pathway. By contrast, HHV-7 infection caused a marked decrease of surface TRAIL-R1, but not of TRAIL-R2, CD95, TNF-R1, or TNF-R2. Of note, the down-regulation of TRAIL-R1 selectively occurred in cells coexpressing HHV-7 antigens that became resistant to TRAIL-mediated cytotoxicity. These findings suggest that the TRAIL-mediated induction of T-cell death may represent an important immune evasion mechanism of HHV-7, helping the virus to persist in the host organism throughout its lifetime.
- Published
- 2001
- Full Text
- View/download PDF
16. Infection of CD34(+) hematopoietic progenitor cells by human herpesvirus 7 (HHV-7).
- Author
-
Mirandola P, Secchiero P, Pierpaoli S, Visani G, Zamai L, Vitale M, Capitani S, and Zauli G
- Subjects
- Adult, Antigens, CD analysis, Antigens, CD34 analysis, Bone Marrow Cells cytology, Cell Differentiation, Cell Line, DNA Replication, DNA, Viral analysis, Erythrocytes cytology, Erythrocytes virology, Fetal Blood cytology, Granulocytes cytology, Granulocytes virology, Hematopoiesis, Herpesvirus 7, Human genetics, Humans, Infant, Newborn, RNA, Messenger genetics, Reverse Transcriptase Polymerase Chain Reaction, T-Lymphocytes, Virus Replication, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells virology, Herpesvirus 7, Human physiology
- Abstract
To investigate the tropism of the T-lymphotropic human herpesvirus 7 (HHV-7) for hematopoietic progenitors, cord blood CD34(+) cells were inoculated in vitro with HHV-7 and then induced to differentiate along the granulocytic and erythroid lineages by the addition of appropriate cytokine cocktails. In semisolid assays, HHV-7 modestly affected the growth of committed (granulocytic/macrophagic and erythroid) progenitors, whereas it significantly decreased the number of pluripotent (granulocytic/erythroid/ monocytic/megakaryocytic) progenitors. Such inhibitory effect was completely abrogated by incubating HHV-7 inoculum with anti-HHV-7 neutralizing serum. In liquid cultures, HHV-7 hastened maturation along the myeloid but not the erythroid lineage, as demonstrated by the up-regulation of CD33 early myeloid antigen at day 7 of culture, and of CD15 and CD14 antigens at day 15. Moreover, HHV-7 messenger RNA was detected by reverse transcriptase-polymerase chain reaction (RT-PCR) in cells maturating along both the myeloid and the erythroid lineages. To evaluate the relevance of these in vitro findings, the presence of HHV-7 was investigated in bone marrow (BM) unfractionated mononuclear cells (MCs) as well as in purified CD34(+) and CD34(-) cell subsets, obtained from 14 normal adult donors. HHV-7 DNA was detected by DNA-PCR in 4 of 7 BMMC samples, and it was found to be associated with both the CD34(-) (2 of 7) and the CD34(+ )(1 of 7) fractions. These data indicate that HHV-7 infects BM cells in vivo and shows the ability to affect the survival/differentiation of CD34(+) hematopoietic progenitors in vitro by inhibiting more ancestral progenitors and perturbing the maturation of myeloid cells.
- Published
- 2000
17. TNF-related apoptosis-inducing ligand (TRAIL) as a negative regulator of normal human erythropoiesis.
- Author
-
Zamai L, Secchiero P, Pierpaoli S, Bassini A, Papa S, Alnemri ES, Guidotti L, Vitale M, and Zauli G
- Subjects
- Adult, Antigens, CD analysis, Antigens, CD34 analysis, Apoptosis Regulatory Proteins, Cell Survival drug effects, Cells, Cultured, Colony-Forming Units Assay, Erythropoiesis drug effects, Flow Cytometry, Hematopoietic Stem Cells drug effects, Hematopoietic Stem Cells physiology, Humans, Lewis X Antigen analysis, Ligands, Recombinant Proteins pharmacology, TNF-Related Apoptosis-Inducing Ligand, Apoptosis drug effects, Bone Marrow Cells cytology, Erythropoiesis physiology, Hematopoietic Stem Cells cytology, Membrane Glycoproteins pharmacology, Tumor Necrosis Factor-alpha pharmacology
- Abstract
The impact of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) on normal hematopoietic development was investigated using adult peripheral blood CD34(+) hematopoietic progenitor cells, induced to differentiate along the erythroid, megakaryocytic, granulocytic, and monocytic lineages by the addition of specific cytokine cocktails. TRAIL selectively reduced the number of erythroblasts, showing intermediate levels of glycophorin A (glycophorin A(interm)) surface expression, which appeared in liquid cultures supplemented with stem cell factor + interleukin 3 + erythropoietin at days 7-10. However, neither immature (day 4) glycophorin A(dim) erythroid cells nor mature (day 14) glycophorin A(bright) erythroblasts were sensitive to TRAIL-mediated apoptosis. Moreover, pre-exposure to TRAIL significantly decreased the number and size of erythroid colonies in semisolid assays. These adverse effects of TRAIL were selective for erythropoiesis, as TRAIL did not significantly influence the survival of cells differentiating along the megakaryocytic, granulocytic, or monocytic lineages. Furthermore, TRAIL was detected by Western blot analysis in lysates obtained from normal bone marrow mononuclear cells. These findings indicate that TRAIL acts in a lineage- and stage of differentiation-specific manner, as a negative regulator of normal erythropoiesis. (Blood. 2000;95:3716-3724)
- Published
- 2000
18. Lineage-restricted expression of protein kinase C isoforms in hematopoiesis.
- Author
-
Bassini A, Zauli G, Migliaccio G, Migliaccio AR, Pascuccio M, Pierpaoli S, Guidotti L, Capitani S, and Vitale M
- Subjects
- Animals, Biomarkers, Blotting, Western, Cell Line, Gene Expression Regulation, Enzymologic, Hematopoietic Stem Cells cytology, Humans, Isoenzymes biosynthesis, Isoenzymes genetics, Mice, Protein Kinase C genetics, Cell Lineage, Hematopoiesis, Hematopoietic Stem Cells enzymology, Protein Kinase C biosynthesis
- Abstract
The pattern of expression of several protein kinase C (PKC) isoforms (alpha, betaI, delta, epsilon, eta, and zeta) during the course of hematopoietic development was investigated using primary human CD34(+) hematopoietic cells and stable cell lines subcloned from the growth factor-dependent 32D murine hematopoietic cell line. Each 32D cell clone shows the phenotype and growth factor dependence characteristics of the corresponding hematopoietic lineage. Clear-cut differences were noticed between erythroid and nonerythroid lineages. (1) The functional inhibition of PKC-epsilon in primary human CD34(+) hematopoietic cells resulted in a twofold increase in the number of erythroid colonies. (2) Erythroid 32D Epo1 cells showed a lower level of bulk PKC catalytic activity, lacked the expression of epsilon and eta PKC isoforms, and showed a weak or absent upregulation of the remaining isoforms, except betaI, upon readdition of Epo to growth factor-starved cells. (3) 32D, 32D GM1, and 32D G1 cell lines with mast cell, granulo-macrophagic, and granulocytic phenotype, respectively, expressed all the PKC isoforms investigated, but showed distinct responses to growth factor readdition. (4) 32D Epo 1.1, a clone selected for interleukin-3 (IL-3) responsiveness from 32D Epo1, expressed the epsilon isoform only when cultured with IL-3. On the other hand, when cultured in Epo, 32D Epo1.1 cells lacked the expression of both epsilon and eta PKC isoforms, similarly to 32D Epo1. (5) All 32D cell lines expressed the mRNA for PKC-epsilon, indicating that the downmodulation of the epsilon isoform occurred at a posttranscriptional level. In conclusion, the PKC isoform expression during hematopoiesis appears to be lineage-specific and, at least partially, related to the growth factor response.
- Published
- 1999
19. Human herpesvirus 7 infection induces profound cell cycle perturbations coupled to disregulation of cdc2 and cyclin B and polyploidization of CD4(+) T cells.
- Author
-
Secchiero P, Bertolaso L, Casareto L, Gibellini D, Vitale M, Bemis K, Aleotti A, Capitani S, Franchini G, Gallo RC, and Zauli G
- Subjects
- CD4-Positive T-Lymphocytes virology, Cell Cycle, Cell Nucleus metabolism, Cells, Cultured, DNA analysis, Flow Cytometry, G2 Phase, Herpesviridae Infections metabolism, Homeostasis, Humans, Microscopy, Confocal, Mitosis, Phosphotyrosine metabolism, S Phase, CD4-Positive T-Lymphocytes ultrastructure, CDC2 Protein Kinase metabolism, Cyclin B metabolism, Herpesviridae Infections pathology, Herpesvirus 7, Human, Polyploidy
- Abstract
Human herpesvirus 7 (HHV-7) infection of both primary CD4(+) T lymphocytes and SupT1 lymphoblastoid T-cell line induced a progressive accumulation of cells exibiting a gap 2/mitosis (G2/M) and polyploid content coupled to an increased cell size. The expression of both cyclin-dependent kinase cdc2 and cyclin B was increased in HHV-7-infected cells with respect to the uninfected ones. Moreover, the simultaneous flow cytometric analysis of cyclin B and DNA content showed that cyclin B expression was not only increased but also unscheduled with respect to its usual cell cycle pattern. However, the levels of kinase activity associated to cdc2 were decreased in HHV-7-infected cells with respect to uninfected cultures. To elucidate the origin of the enlarged HHV-7-infected cells, extensive electron and confocal microscopy analyses were performed. Membrane fusion events associated to cytoplasmic bridges, which characterize the formation of syncytia, were never observed. On the other hand, analysis of serial sections of the same cells strongly suggested that enlarged HHV-7-infected cells contained a single polylobated nucleus. This was confirmed by flow cytometry analysis performed on nuclei isolated from HHV-7-infected cells, which showed multiple peaks with a DNA content >4n. Taken together, these data indicate that giant cells, which represent the hallmark of in vitro HHV-7 infection, arise from single CD4(+) T cells undergoing a process of polyploidization., (Copyright 1998 by The American Society of Hematology.)
- Published
- 1998
20. The induction of megakaryocyte differentiation is accompanied by selective Ser133 phosphorylation of the transcription factor CREB in both HEL cell line and primary CD34+ cells.
- Author
-
Zauli G, Gibellini D, Vitale M, Secchiero P, Celeghini C, Bassini A, Pierpaoli S, Marchisio M, Guidotti L, and Capitani S
- Subjects
- Antigens, CD34, Blotting, Western, Cell Differentiation physiology, Cell Line, Enzyme Inhibitors pharmacology, Flavonoids pharmacology, Flow Cytometry, Humans, Phosphorylation, Protein Kinase Inhibitors, Protein Kinases physiology, Serine, Signal Transduction drug effects, Thrombopoietin pharmacology, Cyclic AMP Response Element-Binding Protein physiology, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells physiology, Megakaryocytes cytology, Megakaryocytes physiology, Signal Transduction physiology
- Abstract
The addition of thrombopoietin (TPO) to HEL cells, cultured in a chemically defined serum-free medium, induced a rapid and dose-dependent phosphorylation of the transcription factor CREB on serine133 (PSer133), as detected by Western blot analysis. TPO also significantly increased the transactivation of CRE-dependent promoter, as determined in transient transfection experiments. On the other hand, neither erythropoietin (Epo; 1 to 10 U) nor hemin (10 (-7) mol/L) were able to significantly stimulate CREB-PSer133 or to activate CRE-promoter in HEL cells. Although pharmacological inhibitors of protein kinase C (chelerytrine and BIM) and protein kinase A (H-89) failed to block the TPO-mediated CREB phosphorylation, a specific inhibitor of the mitogen-activated protein kinases (PD98059) completely blocked the ability of TPO to stimulate CREB-PSer133. Moreover, PD98059 significantly decreased the ability of TPO to upregulate the surface expression of the alphaIIIbbeta3 megakaryocytic marker in HEL cells. In parallel, primary CD34+ hematopoietic cells were seeded in liquid cultures supplemented with 100 ng/mL of TPO and examined by immunofluorescence for the coexpression of alphaIIIbbeta3 and CREB-PSer133 at various time points. High levels of nuclear CREB-PSer133 were unequivocally demonstrated in alphaIIIbbeta3+ cells, including morphologically recognizable megakaryocytes. Taken together, these data suggest that CREB plays a role in modulating the expression of genes critical for megakaryocyte differentiation and that the TPO-mediated CREB phosphorylation seems to be regulated via mitogen-activated protein kinases.
- Published
- 1998
21. In vitro senescence and apoptotic cell death of human megakaryocytes.
- Author
-
Zauli G, Vitale M, Falcieri E, Gibellini D, Bassini A, Celeghini C, Columbaro M, and Capitani S
- Subjects
- Antigens, CD34 analysis, Cell Differentiation drug effects, Cell Separation, DNA Fragmentation, Erythropoietin pharmacology, Humans, Interleukin-3 pharmacology, Microscopy, Electron, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Ploidies, Thrombopoietin pharmacology, Apoptosis, Cellular Senescence, Megakaryocytes cytology
- Abstract
To investigate the fate of human megakaryocytes, CD34+ hematopoietic progenitor cells were purified from the peripheral blood or bone marrow of healthy donors and seeded in serum-free chemically defined suspension cultures. In the presence of thrombopoietin (TPO; 100 ng/mL), CD34-derived cells showed an eightfold numerical expansion and a progressive maturation along the megakaryocytic lineage. Megakaryocyte maturation was characterized ultrastructurally by the presence of a demarcation membrane system and phenotypically by a high surface expression of alpha(IIb)beta3 integrin. The number of mature megakaryocytes peaked at days 12 to 15 of culture. On the other hand, the number of platelets released in the culture supernatant by CD34-derived megakaryocytes peaked at days 18 to 21, when a high percentage of megakaryocytes showed the characteristic features of apoptosis, as evaluated by electron microscopy, terminal deoxynucleotidyl transferase (TdT)-mediated d-UTP-biotin nick end-labeling technique (TUNEL) and uptake of propidium iodide. In other experiments, primary alpha(IIb)beta3+ megakaryocytic cells were directly purified from the bone marrow aspirates of normal donors and seeded in serum-free suspension cultures. In the absence of cytokines, alpha(IIb)beta3+ megakaryocytes progressively underwent apoptotic cell death. The addition of TPO but not interleukin-3 or erythropoietin showed some protection of alpha(IIb)beta3+ cells from apoptosis at early culture times (days 2 to 4), but it did not show any significant effect at later time points. These findings suggest that the terminal phase of the megakaryocyte life span is characterized by the onset of apoptosis, which can be modulated only to a certain extent by TPO.
- Published
- 1997
22. Thrombopoietin enhances the alpha IIb beta 3-dependent adhesion of megakaryocytic cells to fibrinogen or fibronectin through PI 3 kinase.
- Author
-
Zauli G, Bassini A, Vitale M, Gibellini D, Celeghini C, Caramelli E, Pierpaoli S, Guidotti L, and Capitani S
- Subjects
- Actins biosynthesis, Androstadienes pharmacology, Antigens, CD34 pharmacology, Blood Coagulation, Blood Platelets drug effects, Blood Platelets physiology, Cell Adhesion drug effects, Cell Division drug effects, Enzyme Activation drug effects, Fibrin, Fibrinogen drug effects, Fibronectins drug effects, Humans, Leukemia, Erythroblastic, Acute, Megakaryocytes enzymology, Phosphatidylinositol 3-Kinases, Phosphotransferases (Alcohol Group Acceptor) antagonists & inhibitors, Phosphotransferases (Alcohol Group Acceptor) metabolism, Platelet Glycoprotein GPIIb-IIIa Complex biosynthesis, Platelet Glycoprotein GPIIb-IIIa Complex drug effects, Protein Binding drug effects, Tumor Cells, Cultured, Up-Regulation, Wortmannin, Fibrinogen metabolism, Fibronectins metabolism, Megakaryocytes drug effects, Megakaryocytes physiology, Phosphotransferases (Alcohol Group Acceptor) physiology, Platelet Glycoprotein GPIIb-IIIa Complex physiology, Thrombopoietin pharmacology
- Abstract
The effect of thrombopoietin (TPO) on the functional activity of surface alpha IIb beta 3 (GPIIbIIIa) was investigated in both primary human megakaryocytic cells, derived from peripheral blood CD34+ cells, and HEL hematopoietic cell line. TPO (100 ng/mL) induced a sixfold to ninefold enhancement of adhesion of both primary megakaryocytic and HEL cells to plates coated with either fibrinogen or fibronectin and a parallel increase of immunoreactivity to the PAC1 monoclonal antibody (MoAb) and fluorescein isothiocyanate-fibrinogen, both of which recognize an activated state of alpha IIb beta 3. The enhanced adhesion to fibrinogen or fibronectin was mediated by the Arg-Gly-Asp (RGD) recognition sequence of alpha IIb beta 3, as it was abolished by pretreatment of cells with saturating concentrations of RGDS peptide. A MoAb specific for the alpha IIb beta subunit of alpha IIb beta 3 also inhibited cell attachment to fibrinogen or fibronectin, while MoAb to anti-alpha v beta 3 or anti-alpha 5 integrins were completely ineffective, clearly indicating that alpha IIb beta 3 participates in this association. A role for PI 3 kinase (PI 3-K) in the TPO-mediated increase in alpha IIb beta 3 function in megakaryocytic cells was suggested by the ability of the PI 3-K inhibitor wortmannin (100 nmol/L) and antisense oligonucleotides directed against the p85 regulatory subunit of PI 3-K to completely block the TPO-induced increase in alpha IIb beta 3 integrin activity upon TPO stimulation. The modulation of adhesiveness to extracellular matrix proteins containing the RGD motif mediated by TPO likely plays a physiologic role in megakaryocytopoiesis, as pretreatment of CD34+ cells with RGDS or anti-alpha IIb MoAb significantly reduced the number of megakaryocytic colonies obtained in a fibrinclot semisolid assay.
- Published
- 1997
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.