Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Blood Cells Mol Dis ; 47(3): 176-81, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21839656

RESUMEN

Studies of hemolytic agents on G6PD-deficient subjects have been extensively performed on red blood cells obtained from donors, only using in vitro methods. However, there has been no adequate G6PD-deficient animal model for in vivo assessment of potentially hemolytic agents. The objective of this study is to establish a novel mouse model of severe G6PD-deficiency, with high susceptibility to hemolytic damage upon oxidative agents. To create this model, G6PD mutant Gpdx allele was introduced into the C57L/J mouse strain background by breeding program. The hemolytic toxicity of naphthalene and its metabolite α-naphthol on G6PD-deficient red blood cells was evaluated. Our data showed that the F2 homozygous Gpdx mutant with C57L/J background exhibiting the G6PD activity was 0.9±0.1 U/g Hb, level similar to those of G6PD deficiency in human. A significantly negative correlation was demonstrated between GSH percentage reduction and G6PD activity (r=-0.51, p<0.001) upon challenge of the red blood cells with alpha-naphthol in vitro. Similar correlation was also found between GSSG elevation and G6PD activity. Our in vivo studies showed that the administration of naphthalene at 250 mg/kg inflicted significant oxidative damage to the G6PD-deficient mice, as illustrated by the decrease of the GSH-to-GSSG ratio (by 34.2%, p=0.005) and the increase of the methemoglobin level (by 1.9 fold, p<0.001). Hemolytic anemia was also found in G6PD-deficient mice at this dosage of naphthalene. In summary, this novel mouse model could be utilized as a screening platform to more accurately determine the hemolytic toxicity of pharmacological agents on G6PD-deficient subjects.


Asunto(s)
Modelos Animales de Enfermedad , Eritrocitos , Deficiencia de Glucosafosfato Deshidrogenasa/enzimología , Glucosafosfato Deshidrogenasa , Hemolíticos/farmacología , Anemia Hemolítica/inducido químicamente , Animales , Cruzamiento/métodos , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Femenino , Glucosafosfato Deshidrogenasa/genética , Glucosafosfato Deshidrogenasa/metabolismo , Deficiencia de Glucosafosfato Deshidrogenasa/metabolismo , Glutatión/metabolismo , Disulfuro de Glutatión/metabolismo , Humanos , Masculino , Metahemoglobina/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación , Naftalenos/farmacología , Naftoles/farmacología , Estrés Oxidativo
2.
Biochem Biophys Res Commun ; 391(3): 1522-5, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20035718

RESUMEN

BRE, also known as TNFRSF1A modulator and BRCC45, is an evolutionarily highly conserved protein. It is a death receptor-associated protein in cytoplasm and a component of BRCA1/2-containing DNA repair complex in nucleus. BRE was found to have anti-apoptotic activity. Over-expression of BRE by transfection promoted survival of cell lines against apoptotic induction; whereas depletion of the protein by siRNA resulted in the opposite. In vivo anti-apoptotic activity of BRE was demonstrated by significant attenuation of Fas-induced acute fulminant hepatitis in transgenic mice expressing the human protein specifically in the liver. BRE was also implicated in tumor promotion by the accelerated tumor growth of Lewis Lung carcinoma transfected with human BRE; and by high expression of BRE specifically in the tumoral regions of human hepatocellular carcinoma (HCC). The present study was to test directly if transgenic expression of BRE in livers could promote HCC development in neonatal diethylnitrosamine model. By 8months after tumor induction, the maximal sizes of tumor nodules of transgenic mice were significantly larger than those of the non-transgenic controls, although the numbers of tumor nodules between the two groups did not significantly differ. Importantly, as in human HCC, the mouse endogenous BRE level was up-regulated in mouse HCC nodules. These results show that BRE over-expression can indeed promote growth, though not initiation, of liver tumors. Furthermore, the common occurrence of BRE over-expression in human and mouse HCC suggests that up-regulation of BRE is functionally important in liver tumor development.


Asunto(s)
Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Hígado/patología , Proteínas del Tejido Nervioso/biosíntesis , Animales , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/metabolismo , Proliferación Celular , Dietilnitrosamina/toxicidad , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Transfección , Regulación hacia Arriba
3.
Stem Cells Dev ; 14(2): 223-30, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15910249

RESUMEN

Stem cell expansion remains an elusive but highly desirable goal. Here we show that platelet-derived growth factor (PDGF), along with cultured endothelial or stromal cells, significantly enhances expansion of human CD34+ cells in vitro. In media supplemented with thrombopoietin, stem cell factor, flt-3 ligand, and granulocyte-colony stimulating factor, CD34+ cells, as well as CFU-GM, BFU-CFU-E, CFU-GEMM, and CFU-MK, increased by 34.3-, 138-, 59.7-, 38.4-, and 86.0-fold, respectively. Co-culturing of CD34+ cells with cultured stromal cells or human umbilical cord vein endothelial cells (HUVECs) greatly enhanced expansion efficiency. The presence of PDGF (50 ng/ml) further augmented expansion, such that increases of 77.0-, 262-, 90.0-, 93.0-, and 200-fold, respectively, were achieved. Six weeks after infusion of expanded cells into NOD/SCID mice, human CD45+ cells were detected in recipients' bone marrow, spleen, and peripheral blood. Our results provide a rationale for development of a stem cell expansion protocol for clinical applications.


Asunto(s)
Antígenos CD34/biosíntesis , Sangre Fetal/citología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Venas Umbilicales/metabolismo , Animales , Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula/métodos , Membrana Celular/metabolismo , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Medio de Cultivo Libre de Suero/farmacología , Endotelio Vascular/citología , Citometría de Flujo , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Sistema Hematopoyético , Humanos , Integrina beta3/biosíntesis , Antígenos Comunes de Leucocito/biosíntesis , Receptores de Lipopolisacáridos/biosíntesis , Megacariocitos/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores CXCR4/metabolismo , Bazo/citología , Células Madre/citología , Células Madre/metabolismo , Células del Estroma/citología , Factores de Tiempo , Venas Umbilicales/citología
4.
Stem Cells ; 25(7): 1800-6, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17446559

RESUMEN

Serotonin is a monoamine neurotransmitter that has multiple extraneuronal functions. We previously reported that serotonin exerted mitogenic stimulation on megakaryocytopoiesis mediated by 5-hydroxytryptamine (5-HT)2 receptors. In this study, we investigated effects of serotonin on ex vivo expansion of human cord blood CD34+ cells, bone marrow (BM) stromal cell colony-forming unit-fibroblast (CFU-F) formation, and antiapoptosis of megakaryoblastic M-07e cells. Our results showed that serotonin at 200 nM significantly enhanced the expansion of CD34+ cells to early stem/progenitors (CD34+ cells, colony-forming unit-mixed [CFU-GEMM]) and multilineage committed progenitors (burst-forming unit/colony-forming unit-erythroid [BFU/CFU-E], colony-forming unit-granulocyte macrophage, colony-forming unit-megakaryocyte, CD61+ CD41+ cells). Serotonin also increased nonobese diabetic/severe combined immunodeficient repopulating cells in the expansion culture in terms of human CD45+, CD33+, CD14+ cells, BFU/CFU-E, and CFU-GEMM engraftment in BM of animals 6 weeks post-transplantation. Serotonin alone or in addition to fibroblast growth factor, platelet-derived growth factor, or vascular endothelial growth factor stimulated BM CFU-F formation. In M-07e cells, serotonin exerted antiapoptotic effects (annexin V, caspase-3, and propidium iodide staining) and reduced mitochondria membrane potential damage. The addition of ketanserin, a competitive antagonist of 5-HT2 receptor, nullified the antiapoptotic effects of serotonin. Our data suggest the involvement of serotonin in promoting hematopoietic stem cells and the BM microenvironment. Serotonin could be developed for clinical ex vivo expansion of hematopoietic stem cells for transplantation. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Antígenos CD34/metabolismo , Apoptosis/efectos de los fármacos , Sangre Fetal/citología , Hematopoyesis/efectos de los fármacos , Serotonina/farmacología , Células Madre/efectos de los fármacos , Células del Estroma/citología , Animales , Anexina A5/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Sangre Fetal/efectos de los fármacos , Sangre Fetal/enzimología , Trasplante de Células Madre Hematopoyéticas , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones SCID , Células Madre/citología , Células Madre/enzimología , Células del Estroma/efectos de los fármacos
5.
Eur J Haematol ; 74(2): 128-35, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15654904

RESUMEN

OBJECTIVES: Ex vivo expansion of granulocyte-colony stimulating factor (G-CSF)-mobilized peripheral blood stem cells (PBSC) is a promising approach for overcoming the developmental delay of bone marrow (BM) reconstitution after transplantation. This project investigated the effects of culture duration, serum-free media, cytokine combinations, and chemotherapy on the outcomes of expansion. METHODS: Enriched CD34+ cells were cultured for 8 or 10 d in serum-free media (QBSF-60 or X-Vivo 10) and four combinations of cytokines consisting of recombinant human pegylated-megakaryocyte growth and development factor, stem cell factor, flt-3 ligand, G-CSF, interleukin (IL)-6, platelet-derived growth factor (PDGF), and IL-1beta. RESULTS: Eight days of culture in QBSF-60 significantly supported efficient expansions of CD34+ cells, CD34+ CD38- cells, colony-forming units (CFU) of myeloid, erythroid, megakaryocytic, and mixed lineages to 3.76-, 14.4-, 28.3-, 24.0-, 38.1-, and 15.7-fold, respectively. Whilst PDGF or IL-6 enhanced the expansion of early, myeloid, and erythroid progenitors, IL-1beta specifically promoted the megakaryocytic lineage. Engraftment of human CD45+ cells were detectable in all non-obese diabetic/severe-combined immunodeficient mice transplanted with expanded PBSC from donor samples, being 5.80 +/- 3.34% of mouse BM cells. The expansion and engraftment capacity of CD34+ cells from subjects postchemotherapy were significantly compromised across the panel of progenitor cells. CONCLUSION: Our results provided an optimized protocol for PBSC expansion, applicable to ameliorating neutropenia and thrombocytopenia in post-BM transplant patients by the prompt provision of progenitor cells. For postchemotherapy patients, expansion products might provide committed progenitors for improving short-term engraftment, but not self-renewable stem cells.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero , Citocinas/farmacología , Factor Estimulante de Colonias de Granulocitos/farmacología , Células Progenitoras Mieloides , ADP-Ribosil Ciclasa/metabolismo , ADP-Ribosil Ciclasa 1 , Adolescente , Adulto , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Médula Ósea/patología , Médula Ósea/fisiología , Trasplante de Médula Ósea , Células Cultivadas , Niño , Humanos , Glicoproteínas de Membrana , Persona de Mediana Edad , Células Progenitoras Mieloides/metabolismo , Neoplasias/patología , Neoplasias/terapia , Células Madre/efectos de los fármacos
6.
Cell Biol Int ; 29(9): 785-91, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16087361

RESUMEN

Nitric oxide (NO), which is produced from l-arginine by three isoforms of NO synthase (NOS), has been implicated in reproductive functions. However, the specific role of NOS isoforms in gamete function and fertilization is not clear. Three types of NOS knockout mice were super ovulated and fertilized in vitro and in vivo. The sperm count and motility, in vivo and in vitro fertilization rate as indicated by two-cell embryos and blastocyst rate were examined. The sperm count and motility from all three knockout mice were not significantly different from that of the wild type. Inducible NOS (iNOS) knockout mice were found to have the largest number of two-cell embryos/mouse collected after fertilization in vivo (P<0.01), but the rate of blastocyst formation from two-cell embryos in vitro was similar for all three knockouts. The rate of in vitro fertilization using either iNOS-deficient sperm or oocytes, but not those deficient in the other two NOS isoforms, was significantly elevated when compared to that in the wild type (P<0.001). While all three types of NOS do not seem to play a significant role in pre-ejaculated sperm function, iNOS may play an inhibitory role in sperm and oocyte functions affecting the process of fertilization and early embryo development.


Asunto(s)
Fertilización/fisiología , Óxido Nítrico Sintasa de Tipo II/fisiología , Óvulo/enzimología , Espermatozoides/enzimología , Animales , Blastocisto/enzimología , Blastocisto/fisiología , Embrión de Mamíferos/enzimología , Embrión de Mamíferos/fisiología , Femenino , Fertilización In Vitro , Isoenzimas/genética , Isoenzimas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa de Tipo II/genética , Óvulo/fisiología , Embarazo , Interacciones Espermatozoide-Óvulo/fisiología , Espermatozoides/fisiología , Superovulación
7.
Br J Haematol ; 117(3): 735-46, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12028051

RESUMEN

Platelet-derived growth factor (PDGF) is a major mitogen for connective tissue cells. In this study, we investigated the effects and mechanism of PDGF on the ex vivo expansion of cord blood CD34+ cells. Our data demonstrated that among various cytokine combinations of thrombopoietin (TPO), interleukin 1 beta (IL-1beta), IL-3, IL-6 and Flt-3 ligand (Flt-3L), TPO + IL-6 + Flt-3L was most efficient in promoting the expansion of CD34+ cells, CD34+CD38- cells, mixed-lineage colony-forming units (CFU-GEMM) and long-term culture-initiating cells (LTC-IC) by 21.7 +/- 5.00-, 103 +/- 27.9-, 10.7 +/- 7.94- and 6.52 +/- 1.51-fold, respectively, after 12-14 d of culture. The addition of PDGF increased the yield of these early progenitors by 45.0%, 66.5%, 45.1% and 79.8% respectively. More significantly, PDGF enhanced the engraftment of human CD45+ cells and their myeloid subsets (CD33+, CD14+ cells) in non-obese diabetic (NOD)/severe-combined immunodeficient (SCID) mice. The expression of PDGF receptor (PDGFR)-beta was not detectable in fresh CD34+ cells but was upregulated after culture for 3 d. PDGF also enhanced the development of adherent cells/clusters that expressed the endothelial markers VE-cadherin and CD31. These findings suggest that PDGF is an effective cytokine for the ex vivo expansion of early stem and progenitor cells. The mechanism could be mediated by PDGFR-beta on committed CD34+ progenitor cells and/or secondary to the stimulation of autologous, stromal feeder cells.


Asunto(s)
Antígenos CD34/sangre , Diabetes Mellitus Experimental/terapia , Células Madre Hematopoyéticas/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Inmunodeficiencia Combinada Grave/terapia , Animales , Adhesión Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Citocinas/farmacología , Sangre Fetal/citología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA