Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Blood ; 119(1): 217-26, 2012 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-22053107

RESUMEN

Extracellular ATP and UTP nucleotides increase the proliferation and engraftment potential of normal human hematopoietic stem cells via the engagement of purinergic receptors (P2Rs). In the present study, we show that ATP and UTP have strikingly opposite effects on human acute myeloblastic leukemia (AML) cells. Leukemic cells express P2Rs. ATP-stimulated leukemic cells, but not normal CD34+ cells, undergo down-regulation of genes involved in cell proliferation and migration, whereas cell-cycle inhibitors are up-regulated. Functionally, ATP induced the inhibition of proliferation and accumulation of AML cells, but not of normal cells, in the G0 phase of the cell cycle. Exposure to ATP or UTP inhibited AML-cell migration in vitro. In vivo, xenotransplantation experiments demonstrated that the homing and engraftment capacity of AML blasts and CD34+CD38- cells to immunodeficient mice BM was significantly inhibited by pretreatment with nucleotides. P2R-expression analysis and pharmacologic profiling suggested that the inhibition of proliferation by ATP was mediated by the down-regulation of the P2X7R, which is up-regulated on untreated blasts, whereas the inhibition of chemotaxis was mainly mediated via P2Y2R and P2Y4R subtypes. We conclude that, unlike normal cells, P2R signaling inhibits leukemic cells and therefore its pharmacologic modulation may represent a novel therapeutic strategy.


Asunto(s)
Adenosina Trifosfato/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Trasplante de Células , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Receptores Purinérgicos/metabolismo , Uridina Trifosfato/farmacología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal
2.
Blood ; 117(2): 419-28, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-20585044

RESUMEN

Mechanisms governing stress-induced hematopoietic progenitor cell mobilization are not fully deciphered. We report that during granulocyte colony-stimulating factor-induced mobilization c-Met expression and signaling are up-regulated on immature bone marrow progenitors. Interestingly, stromal cell-derived factor 1/CXC chemokine receptor-4 signaling induced hepatocyte growth factor production and c-Met activation. We found that c-Met inhibition reduced mobilization of both immature progenitors and the more primitive Sca-1(+)/c-Kit(+)/Lin(-) cells and interfered with their enhanced chemotactic migration to stromal cell-derived factor 1. c-Met activation resulted in cellular accumulation of reactive oxygen species by mammalian target of rapamycin inhibition of Forkhead Box, subclass O3a. Blockage of mammalian target of rapamycin inhibition or reactive oxygen species signaling impaired c-Met-mediated mobilization. Our data show dynamic c-Met expression and function in the bone marrow and show that enhanced c-Met signaling is crucial to facilitate stress-induced mobilization of progenitor cells as part of host defense and repair mechanisms.


Asunto(s)
Movimiento Celular/fisiología , Factor Estimulante de Colonias de Granulocitos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Animales , Quimiocina CXCL12/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Factor de Crecimiento de Hepatocito/metabolismo , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
Blood ; 116(20): 4328-37, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-20686121

RESUMEN

Severe malaria anemia is characterized by inhibited/altered erythropoiesis and presence of hemozoin-(HZ)-laden bone-marrow macrophages. HZ mediates peroxidation of unsaturated fatty acids and production of bioactive aldehydes such as 4-hydroxynonenal (HNE). HZ-laden human monocytes inhibited growth of cocultivated human erythroid cells and produced HNE that diffused to adjacent cells generating HNE-protein adducts. Cocultivation with HZ or treatment with low micromolar HNE inhibited growth of erythroid cells interfering with cell cycle without apoptosis. After HZ/HNE treatment, 2 critical proteins in cell-cycle regulation, p53 and p21, were increased and the retinoblastoma protein, central regulator of G1-to-S-phase transition, was consequently hypophosphorylated, while GATA-1, master transcription factor in erythropoiesis was reduced. The resultant decreased expression of cyclin A and D2 retarded cell-cycle progression in erythroid cells and the K562 cell line. As a second major effect, HZ and HNE inhibited protein expression of crucial receptors (R): transferrinR1, stem cell factorR, interleukin-3R, and erythropoietinR. The reduced receptor expression and the impaired cell-cycle activity decreased the production of cells expressing glycophorin-A and hemoglobin. Present data confirm the inhibitory role of HZ, identify HNE as one HZ-generated inhibitory molecule and describe molecular targets of HNE in erythroid progenitors possibly involved in erythropoiesis inhibition in malaria anemia.


Asunto(s)
Aldehídos/farmacología , Anemia/complicaciones , Anemia/fisiopatología , Eritropoyesis/efectos de los fármacos , Hemoproteínas/farmacología , Malaria/complicaciones , Malaria/fisiopatología , Anemia/patología , Biomarcadores/metabolismo , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Ensayo de Unidades Formadoras de Colonias , Ciclina A/metabolismo , Ciclina D2/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Eritroides/efectos de los fármacos , Células Eritroides/metabolismo , Células Eritroides/patología , Factor de Transcripción GATA1/metabolismo , Glicoforinas/metabolismo , Hemoglobinas/metabolismo , Humanos , Malaria/patología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Monocitos/patología , Receptores Inmunológicos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
4.
Stem Cells ; 26(6): 1620-7, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18369098

RESUMEN

As mobilized peripheral blood (MPB) represents an attractive cell source for gene therapy, we investigated the ability of third-generation lentiviral vectors (LVs) to transfer the enhanced green fluorescent protein gene into MPB CD34(+) cells in culture conditions allowing expansion of transplantable human hematopoietic stem cells. To date, few studies have reported transduction of MPB cells with vesicular stomatitis virus G pseudotyped LVs. The critical issue remains whether primitive, hematopoietic repopulating cells have, indeed, been transduced. In vitro (5 weeks' culture in FLT3 ligand + thrombopoietin + stem cell factor + interleukin 6) and in vivo (serial transplantation in NOD/SCID mice) experiments show that MPB CD34(+) cells can be effectively long-term transduced by LV and maintain their proliferation, self-renewal, and multilineage differentiation potentials. We show that expansion following transduction improves the engraftment of transduced MPB CD34(+) (4.6-fold expansion of SCID repopulating cells by limiting dilution studies). We propose ex vivo expansion after transduction as an effective tool to improve gene therapy protocols with MPB. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Antígenos CD34/análisis , Terapia Genética/métodos , VIH-1/genética , Animales , Células Sanguíneas/fisiología , Células de la Médula Ósea/fisiología , Citometría de Flujo , Genes Reporteros , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Proteínas Fluorescentes Verdes/genética , Movilización de Célula Madre Hematopoyética , Humanos , Inmunofenotipificación , Interleucina-3/farmacología , Ratones , Ratones SCID , Proteínas Recombinantes/farmacología , Trombopoyetina/farmacología
5.
Exp Hematol ; 39(3): 360-74, 374.e1-5, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21145936

RESUMEN

OBJECTIVE: Extracellular adenosine triphosphate (ATP) is a well-recognized mediator of cell-to-cell communication. Here we show ATP effects on bone marrow (BM)-derived human mesenchymal stem cell (hMSCs) functions. MATERIALS AND METHODS: ATP-induced modification of hMSCs gene expression profile was assessed by Affymetrix technology. Clonogenic and migration assays in vitro, as well as xenotransplant experiments in vivo, were performed to evaluate the effects of ATP on hMSCs proliferation and BM homing. Enzyme-linked immunosorbent assays were used to assess hMSCs cytokines production, whereas T-cell cultures demonstrated the immunoregulatory activity of ATP-treated hMSCs. RESULTS: hMSCs were resistant to the cytotoxic effects of ATP, as demonstrated by the lack of morphological and mitochondrial changes or release of intracellular markers of cell death. Gene expression profiling revealed that ATP-stimulated hMSCs underwent a downregulation of genes involved in cell proliferation, whereas those involved in cell migration were strongly upregulated. The inhibitory activity of ATP on hMSCs proliferation was confirmed by assessing clonogenic stromal progenitors. ATP potentiated the chemotactic response of hMSCs to the chemokine CXCL12, and increased their spontaneous migration. In vivo, the homing capacity of hMSCs to the BM of immunodeficient mice was significantly increased by pretreatment with ATP. Moreover, ATP increased the production of the proinflammatory cytokines interleukin-2, interferon-γ, and interleukin-12p70, while decreasing the anti-inflammatory cytokine interleukin-10, and this finding was associated with the reduced ability of MSCs to inhibit T-cell proliferation. CONCLUSIONS: Our data show that purinergic signaling modulates hMSCs functions and highlights a role for extracellular nucleotides in hMSCs biology.


Asunto(s)
Adenosina Trifosfato/farmacología , Células de la Médula Ósea , Quimiocina CXCL12/metabolismo , Quimiotaxis/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Interleucinas/biosíntesis , Células Madre Mesenquimatosas , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Quimiocina CXCL12/farmacología , Quimiotaxis/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/fisiología , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Trasplante Heterólogo
6.
J Exp Med ; 205(7): 1673-85, 2008 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-18559453

RESUMEN

Increased evidence suggests that cancer-associated inflammation supports tumor growth and progression. We have previously shown that semaphorin 4D (Sema4D), a ligand produced by different cell types, is a proangiogenic molecule that acts by binding to its receptor, plexin B1, expressed on endothelial cells (Conrotto, P., D. Valdembri, S. Corso, G. Serini, L. Tamagnone, P.M. Comoglio, F. Bussolino, and S. Giordano. 2005. Blood. 105:4321-4329). The present work highlights the role of Sema4D produced by the tumor microenvironment on neoplastic angiogenesis. We show that in an environment lacking Sema4D, the ability of cancer cells to generate tumor masses and metastases is severely impaired. This condition can be explained by a defective vascularization inside the tumor. We demonstrate that tumor-associated macrophages (TAMs) are the main cells producing Sema4D within the tumor stroma and that their ability to produce Sema4D is critical for tumor angiogenesis and vessel maturation. This study helps to explain the protumoral role of inflammatory cells of the tumor stroma and leads to the identification of an angiogenic molecule that might be a novel therapeutic target.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Macrófagos/metabolismo , Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Semaforinas/metabolismo , Animales , Línea Celular , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Proteínas del Tejido Nervioso/genética , Receptores de Superficie Celular/genética , Semaforinas/genética
7.
Stem Cells ; 22(2): 135-43, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14990853

RESUMEN

We have previously established a stroma-free culture with Flt-3 ligand (FL), stem cell factor (SCF), and thrombopoietin (TPO) that allows the maintenance and the expansion for several weeks of a cord blood (CB) CD34+ cell population capable of multilineage and long-lasting hematopoietic repopulation in non-obese diabetic/ severe combined immunodeficient (NOD/SCID) mice. In this work the kinetics of megakarocyte (Mk)-engraftment that is often poor and delayed in CB transplantation, and human platelet (HuPlt) generation in NOD/SCID mice of baseline CD34+ cells (b34+), and of CD34+ cells reisolated after a 4-week expansion with FL+SCF+TPO (4w34+) were compared. With b34+ cells Mk-engraftment was first seen at week 3 (CD41+: 0.4%); 4w34+ cells allowed a more rapid Mk-engraftment (at weeks 2 and 3 the CD41+ cells were 0.3% and 0.8%). Circulating HuPlts were first seen at weeks 2 and 1, respectively. Mk-engraftment levels of b34+ and 4w34+ cells 6-8 weeks after transplantation were similar (12 +/- 3.5 versus 15 +/- 5% CD45+; 1.3 +/- 0.5 versus 1.8 +/- 0.5% CD41+ cells). Also serial transplant experiments were performed with expanded and reselected CB cells. In secondary and tertiary recipients the Mk population was detected with bone marrow fluorescence-activated cell sorter analysis; these experiments indicate the effective long-term repopulation of expanded cells. Selected CD34+ cells after a 4-week expansion with FL+SCF+TPO are more efficient in Mk engraftment than the same number of unmanipulated cells.


Asunto(s)
Sangre Fetal/trasplante , Trasplante de Células Madre Hematopoyéticas/métodos , Megacariocitos/trasplante , Transfusión de Plaquetas , Animales , Antígenos CD34/inmunología , Médula Ósea/inmunología , Sangre Fetal/inmunología , Humanos , Megacariocitos/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA