Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Psychiatry ; 21(2): 178-88, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26347316

RESUMEN

Increased dosage of methyl-CpG-binding protein-2 (MeCP2) results in a dramatic neurodevelopmental phenotype with onset at birth. We generated induced pluripotent stem cells (iPSCs) from patients with the MECP2 duplication syndrome (MECP2dup), carrying different duplication sizes, to study the impact of increased MeCP2 dosage in human neurons. We show that cortical neurons derived from these different MECP2dup iPSC lines have increased synaptogenesis and dendritic complexity. In addition, using multi-electrodes arrays, we show that neuronal network synchronization was altered in MECP2dup-derived neurons. Given MeCP2 functions at the epigenetic level, we tested whether these alterations were reversible using a library of compounds with defined activity on epigenetic pathways. One histone deacetylase inhibitor, NCH-51, was validated as a potential clinical candidate. Interestingly, this compound has never been considered before as a therapeutic alternative for neurological disorders. Our model recapitulates early stages of the human MECP2 duplication syndrome and represents a promising cellular tool to facilitate therapeutic drug screening for severe neurodevelopmental disorders.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/fisiología , Red Nerviosa/metabolismo , Diferenciación Celular , Dendritas/metabolismo , Dosificación de Gen/fisiología , Duplicación de Gen/genética , Estudios de Asociación Genética , Humanos , Células Madre Pluripotentes Inducidas , Masculino , Neurogénesis , Neuronas
2.
Biotechnol Bioeng ; 108(2): 364-75, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20830682

RESUMEN

Many applications of stem cell technologies require a large quantity of cells for which scalable processes of cell expansion and differentiation are essential. Multipotent adult progenitor cells (MAPCs) are adult stem cells isolated from the bone marrow with extensive self-renewal and broad differentiation capabilities. MAPCs are typically cultured surface adherent (2D) and at low cell density, making the large surface required for cell expansion a hindrance for many applications. This study demonstrates that MAPCs can be cultivated as aggregates in an undifferentiated state for at least 16 days, as levels of a number of transcripts, including Oct4, remained similar, Oct4 protein was unchanged, and differentiation to neural progenitor, endothelial cell and hepatocyte like cells was retained. Cultivation of these aggregates in stirred bioreactor lead to a 70-fold expansion in 6 days with final cell densities of close to 106/mL. Importantly, the MAPC aggregates recovered from stirred bioreactors could be differentiated to hepatocyte-like cells that expressed albumin, alpha-1-antitrypsin (AAT), and tyrosine amino transferase (TAT) transcripts and also secreted albumin and urea. This method of scalable expansion combined with differentiation of MAPCs can potentially be used for generating large numbers of MAPC and MAPC-derived differentiated cells.


Asunto(s)
Células Madre Multipotentes/fisiología , Animales , Reactores Biológicos , Técnicas de Cultivo de Célula/métodos , Proliferación Celular , Células Cultivadas , Perfilación de la Expresión Génica , Ratas , Factores de Tiempo
3.
J Exp Med ; 174(3): 693-703, 1991 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-1875168

RESUMEN

We have previously demonstrated that primitive progenitors from human bone marrow termed long term bone marrow culture initiating cells (LTBMC-IC) adhere avidly to irradiated bone marrow stroma, while more mature clonogenic progenitors fail to do so. In this study we examine the interaction between these progenitors and components of the bone marrow stroma. (a) We demonstrate that both primitive LTBMC-IC and more mature clonogenic progenitors adhere to intact fibronectin. (b) Primitive LTBMC-IC and multi-lineage CFU-MIX progenitors adhere to the 33/66 kD COOH-terminal heparin-binding cell-adhesion promoting fragment of fibronectin, but adhere significantly less to its 75 kD RGDS-dependent cell-binding fragment. In contrast, more differentiated single-lineage progenitors adhere equally well to the 33/66 kD RGDS independent and the 75 kD RGDS-dependent cell-adhesion fragments of fibronectin. (c) Both primitive LTBMC-IC and clonogenic progenitors adhere to the three known cell-attachment sites in the 33/66 kD cell-adhesion promoting fragment, FN-C/H I, FN-C/H II and CS1. However, LTBMC-IC and CFU-MIX progenitors adhere significantly better to FN-C/H II than to the flanking FN-C/H I and CS1 cell-attachment sites. In contrast, single-lineage progenitors adhere equally well to all three cell attachment sites in the 33/66 kD cell-adhesion promoting fragment. (d) Finally, adhesion of primitive LTBMC-IC to intact irradiated stroma can be inhibited partially by peptide FN-C/H II and almost completely by a combination of FN-C/H II and peptide FN-C/H I and CS1. This study demonstrates that adhesive interactions between primitive hematopoietic progenitors and the extracellular matrix component fibronectin can occur. Specific changes in adhesion to the 33/66 kD cell-adhesion promoting fragment and the 75 kD RGDS-dependent cell-adhesion fragment of fibronectin are associated with differentiation of primitive multi-lineage progenitors into committed single-lineage progenitors. Such differences in adhesive interaction with fibronectin may allow hematopoietic progenitors at various stages of differentiation to interact with specific supportive loci of the bone marrow microenvironment. Finally, the ability to block adhesion of LTBMC-IC to intact irradiated stroma with peptides FN-C/H II, FN-C/H I and CS1 suggests that receptors responsible for this interaction may be important in the homing of primitive progenitors to the bone marrow.


Asunto(s)
Células de la Médula Ósea , Fibronectinas/metabolismo , Células Madre Hematopoyéticas/citología , Secuencia de Aminoácidos , Sitios de Unión , Adhesión Celular , Diferenciación Celular , Células Clonales , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Heparina/metabolismo , Humanos , Datos de Secuencia Molecular , Oligopéptidos/metabolismo , Fragmentos de Péptidos/metabolismo , Unión Proteica
4.
J Exp Med ; 179(2): 643-9, 1994 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-8294873

RESUMEN

Factors that induce proliferation of the human hematopoietic stem cell are ill-defined. Primitive hematopoietic progenitors can be maintained and differentiate in stroma-dependent, long-term bone marrow cultures (LTBMC), originally described by Dexter et al. (Dexter, T. M., L. H. Coutinho, E. Spooncer, C. M. Heyworth, C. P. Daniel, R. Schiro, J. Chang, and T. D. Allen. 1990. Molecular Control of Haemopoiesis). However, 70-80% of primitive progenitors capable of reinitiating secondary stromal cultures (LTBMC-initiating cells [IC]) are lost over a period of 5 wk in such cultures. We have recently described a novel "stroma-noncontact" culture system, in which hematopoietic progenitors are separated from the stromal layer by a 0.4-micron microporous filter membrane. Primitive progenitors in such cultures can not only differentiate into committed progenitors, but are also maintained to a greater extent than in "Dexter" cultures. However, still only 50% of the originally seeded LTBMC-IC are recovered at week 5. Since maintenance of primitive progenitors may depend not only on growth-promoting factors but also on factors that inhibit differentiation and/or proliferation, we evaluated the effect of macrophage inflammatory protein 1 alpha (MIP-1 alpha) or "stem cell inhibitor" in combination with the growth-inducing factor interleukin 3 (IL-3) on the recovery of LTBMC-IC from stroma-noncontact cultures. We demonstrate that addition of MIP-1 alpha alone to stroma-noncontact cultures does not change the number of LTBMC-IC present after 8 wk, indicating that this factor may not directly inhibit or stimulate proliferation of primitive progenitors. Addition of the growth stimulatory cytokine, IL-3, alone results in exhaustion of LTBMC-IC after 8 wk of culture, possibly as a result of their terminal differentiation. However, LTBMC-IC can be maintained for at least 8 wk when grown in stroma-noncontact cultures supplemented with both MIP-1 alpha plus IL-3. This effect depends on soluble (ill-defined) stromal factors, and results from a direct interaction of these cytokines with the progenitor population or its progeny, but not the stroma.


Asunto(s)
Citocinas/metabolismo , Células Madre Hematopoyéticas/citología , Interleucina-3/metabolismo , Monocinas/metabolismo , Médula Ósea/metabolismo , Células de la Médula Ósea , División Celular , Células Cultivadas , Quimiocina CCL3 , Quimiocina CCL4 , Medios de Cultivo , Citometría de Flujo , Humanos , Proteínas Inflamatorias de Macrófagos , Células del Estroma/citología
5.
Gut ; 58(4): 594-603, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19091822

RESUMEN

There has been recent progress in the isolation and characterisation of stem/progenitor cells that may differentiate towards the hepatic lineage. This has raised expectations that therapy of genetic or acquired liver disease might be possible by transplanting stem/progenitor cells or their liver-committed progeny. However, it is currently impossible to determine from the many documented studies which of the stem/progenitor cell populations are the best for therapy of a given disease. This is largely because of the great variability in methods used to characterise cells and their differentiation ability, variability in transplantation models and inconsistent methods to determine the effect of cell grafting in vivo. This manuscript represents a first proposal, created by a group of investigators ranging from basic biologists to clinical hepatologists. It aims to define standardised methods to assess stem/progenitor cells or their hepatic lineage-committed progeny that could be used for cell therapy in liver disease. Furthermore standardisation is suggested both for preclinical animal models to evaluate the ability of such cells to repopulate the liver functionally, and for the ongoing clinical trials using mature hepatocytes. Only when these measures have been put in place will the promise of stem/progenitor-derived hepatocyte-based therapies become reality.


Asunto(s)
Hepatocitos/trasplante , Hepatopatías/terapia , Trasplante de Células Madre/normas , Células Madre/citología , Células Madre Adultas/trasplante , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Células Madre Embrionarias/trasplante , Rechazo de Injerto , Humanos , Regeneración Hepática , Trasplante de Células Madre/métodos
6.
J Clin Invest ; 100(4): 912-20, 1997 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-9259591

RESUMEN

Hematopoiesis occurs in different organs during fetal development. Several studies suggest that the growth of hematopoietic progenitors at one stage of ontogenic maturation may not be supported by a microenvironment from a different ontogenic stage. To determine if human fetal liver (FL) clonogenic progenitors can develop in an adult bone marrow (ABM) microenvironment, we compared growth of BFU-E and CFU-GM from 7-14-wk-old FL, 11-20-wk-old fetal bone marrow (FBM), umbilical cord blood (UCB), or ABM in clonogenic medium with or without ABM stroma. In contrast to BFU-E from FBM, UCB, or ABM, soluble factor(s) produced by ABM stroma severely suppressed growth of 98% of FL BFU-E by inducing apoptosis of cells beyond early erythroblast stage. The nature of the soluble factor remains unknown, although we have evidence that it is heat labile with molecular mass < 10 kD. Antibody neutralization studies indicate that TGF-beta1, IL-1, TNF-alpha, macrophage inflammatory protein (MIP)-1alpha, or IFN-gamma are not responsible. The observation that FL progenitors may not be capable of differentiating when transferred to an ABM microenvironment may have important implications for FL transplantation into postnatal recipients. Further, this demonstrates that ontogenic stage-specific interactions between hematopoietic progenitors and their microenvironment are important for the normal development of hematopoiesis.


Asunto(s)
Apoptosis , Médula Ósea/metabolismo , Células Precursoras Eritroides/fisiología , Matriz Extracelular/metabolismo , Hígado/metabolismo , Factores de Edad , Antígenos CD34 , Diferenciación Celular , División Celular , Ensayo de Unidades Formadoras de Colonias , Medios de Cultivo Condicionados/farmacología , Sangre Fetal/fisiología , Citometría de Flujo , Células Madre Hematopoyéticas/fisiología , Humanos , Subgrupos de Linfocitos T/fisiología
7.
J Clin Invest ; 90(4): 1232-41, 1992 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1383271

RESUMEN

We studied the adhesion of primitive and committed progenitors from chronic myelogenous leukemia (CML) and normal bone marrow to stroma and to several extracellular matrix components. In contrast to benign primitive progenitors from CML or normal bone marrow, Ph1-positive primitive progenitors from CML bone marrow fail to adhere to normal stromal layers and to fibronectin and its proteolytic fragments, but do adhere to collagen type IV, an extracellular matrix component of basement membranes. Similarly, multilineage colony-forming unit (CFU-MIX) progenitors from CML bone marrow do not adhere to fibronectin or its adhesion promoting fragments but adhere to collagen type IV. Unlike committed progenitors from normal bone marrow, CML single-lineage burst-forming units-erythroid and granulocyte/macrophage colony-forming units fail to adhere to fibronectin or its components but do adhere to both collagen type IV and laminin. Evaluation of adhesion receptor expression demonstrates that fibronectin receptors (alpha 4, alpha 5, and beta 1) are equally present on progenitors from normal and CML bone marrow. However, a fraction of CML progenitors express alpha 2 and alpha 6 receptors, associated with laminin and collagens, whereas these receptors are absent from normal progenitors. These observations indicate that the premature release of malignant Ph1-positive progenitors into the circulation may be caused by loss of adhesive interactions with stroma and/or fibronectin and acquisition of adhesive interactions with basement membrane components. Further study of the altered function of cell-surface adhesion receptors characteristic of the malignant clone in CML may lead to a better understanding of the mechanisms underlying both abnormal expansion and abnormal circulation of malignant progenitors in CML.


Asunto(s)
Células de la Médula Ósea , Colágeno/fisiología , Fibronectinas/fisiología , Laminina/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/patología , Adolescente , Antígenos CD/análisis , Antígenos CD34 , Membrana Basal/fisiología , Adhesión Celular , Niño , Preescolar , Antígenos HLA-DR/análisis , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Células del Estroma/patología
8.
J Clin Invest ; 96(1): 511-9, 1995 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7542285

RESUMEN

In long-term bone marrow cultures, stroma-adherent progenitors proliferate significantly less than nonadherent progenitors. Thus, close progenitor-stroma interactions may serve to regulate or restrict rather than promote hematopoietic progenitor proliferation. We hypothesized that signaling through adhesion receptors on hematopoietic cells may contribute to the inhibition of proliferation observed when progenitors are in contact with stroma. We demonstrate that progenitors cultured physically separated from stroma in a transwell proliferate significantly more than progenitors adherent to stroma. Furthermore, proliferation of colony forming cells (CFC) is reduced after specific adhesion to stroma, metabolically inactivated glutaraldehyde-fixed stroma, stromal-extracellular matrix, or the COOH-terminal heparin-binding domain of fibronectin. Nonspecific adhesion to poly-L-lysine fails to inhibit CFC proliferation. That the VLA-4 integrin is one of the receptors that transfers proliferation inhibitory signals was shown using blocking anti-alpha 4 monomeric F(ab) fragments. Furthermore, when synthetic peptides representing specific cell attachment sites within the heparin-binding domain of fibronectin were added to Dexter-type marrow cultures, significantly increased recovery and proliferation of CFC was observed, suggesting that these peptides disrupt adhesion-mediated proliferation inhibitory events. Thus, negative regulation of hematopoiesis may not only depend on the action of growth inhibitory cytokines but also on growth inhibitory signals resulting from direct adhesive interactions between progenitors and marrow stroma.


Asunto(s)
Células de la Médula Ósea , Células Madre Hematopoyéticas/fisiología , Receptores de Fibronectina/fisiología , Secuencia de Aminoácidos , Adhesión Celular , Moléculas de Adhesión Celular/fisiología , División Celular , Células Cultivadas , Heparina/metabolismo , Humanos , Integrina alfa4beta1 , Integrinas/fisiología , Datos de Secuencia Molecular , Células del Estroma/fisiología , Molécula 1 de Adhesión Celular Vascular
9.
J Clin Invest ; 96(2): 931-9, 1995 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7543495

RESUMEN

The mechanisms by which interferon-alpha (IFN-alpha) restores normal hematopoiesis in chronic myelogenous leukemia (CML) are not well understood. We have recently demonstrated that IFN-alpha acts directly on CML hematopoietic progenitors to restore their adhesion to marrow stroma by modulating beta 1 integrin receptor function. In the present study we examined the effect of IFN-alpha treatment of marrow stroma on subsequent adhesion of CML progenitors. Stromal layers were preincubated with IFN-alpha (10,000 microns/ml) for 48 h. Subsequent coincubation with CML progenitors for 2 h resulted in significantly increased adhesion of CML progenitors. We demonstrated that alpha 4 beta 1 and alpha 5 beta 1 integrin receptors were involved in the enhanced adhesion of CML progenitors, suggesting that IFN-alpha-treated stroma can upregulate CML integrin function. This effect is due, at least in part, to IFN-alpha-induced increased stromal production of the chemokine macrophage inflammatory protein-1 alpha (MIP-1 alpha), which upregulates beta 1 integrin-dependent adhesion of CML progenitors to stroma. Thus, IFN-alpha treatment of marrow stroma restores beta 1 integrin-dependent adhesion of CML progenitors, at least in part through induction of MIP-1 alpha production. These observations provide further insights into mechanisms by which IFN-alpha may restore normal hematopoiesis in CML.


Asunto(s)
Médula Ósea/efectos de los fármacos , Tejido Conectivo/efectos de los fármacos , Citocinas/fisiología , Células Madre Hematopoyéticas/fisiología , Integrinas/fisiología , Interferón-alfa/farmacología , Leucemia Mieloide de Fase Crónica/patología , Monocinas/fisiología , Células Madre Neoplásicas/fisiología , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Células de la Médula Ósea , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL4 , Tejido Conectivo/metabolismo , Células del Tejido Conectivo , Citocinas/biosíntesis , Humanos , Integrina beta1 , Interferón alfa-2 , Proteínas Inflamatorias de Macrófagos , Monocinas/biosíntesis , Proteínas Recombinantes
10.
J Clin Invest ; 94(1): 384-91, 1994 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7518835

RESUMEN

Treatment of chronic myelogenous leukemia (CML) with interferon-alpha frequently results in normalization of peripheral blood counts and, in up to 20% of patients, reestablishment of normal hematopoiesis. We hypothesize that interferon-alpha may restore normal adhesive interactions between CML progenitors and the bone marrow microenvironment and restore normal growth regulatory effects resulting from these progenitor-stroma interactions. We demonstrate that treatment with interferon-alpha induces a significant, dose-dependent increase in the adhesion of primitive long-term culture initiating cells and committed colony-forming cells (CFC) from CML bone marrow to normal stroma. Adhesion of CFC seen after interferon-alpha treatment could be inhibited by blocking antibodies directed at the alpha 4, alpha 5, and beta 1 integrins and vascular cell adhesion molecule, but not CD44 or intracellular adhesion molecule, suggesting that interferon-alpha induces normalization of progenitor-stroma interactions in CML. Because FACS analysis showed that the level of alpha 4, alpha 5, and beta 1 integrin expression after interferon-alpha treatment is unchanged, this suggests that interferon-alpha may restore normal beta 1 integrin function. Normalization of interactions between CML progenitors and the bone marrow microenvironment may then result in the restoration of normal regulation of CML progenitor proliferation, and explain, at least in part, the therapeutic efficacy of interferon-alpha in CML.


Asunto(s)
Células de la Médula Ósea , Células Madre Hematopoyéticas/efectos de los fármacos , Integrinas/fisiología , Interferón-alfa/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Antígenos CD/análisis , Antígenos CD34 , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Fusión bcr-abl/análisis , Humanos , Integrina beta1 , Interferón-alfa/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Células del Estroma/fisiología
11.
J Clin Invest ; 101(11): 2456-67, 1998 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-9616217

RESUMEN

Despite the wide use of mobilized peripheral blood (PB) progenitor cells (PBPC) for clinical transplantation the mechanism(s) underlying their mobilization and subsequent engraftment are still unknown. We compared the adhesive phenotype of CD34(+) colony-forming cells (CFC) in bone marrow (BM) and PB of normal donors before and after administration of granulocyte colony-stimulating factor (G-CSF) for 5 d. G-CSF-mobilized PB CFC cells adhered significantly less to BM stroma, fibronectin, and to the alpha4 beta1 binding fibronectin peptide, CS1, because of decreased expression of the alpha4 integrin. Since incubation of BM CD34(+) cells for 4 d with G-CSF at concentrations found in serum of G-CSF- treated individuals did not affect alpha4-dependent adhesion, G-CSF may not be directly responsible for the decreased alpha4-mediated adhesion of PB CFC. Culture of G-CSF-mobilized PB CD34(+) cells with cytokines at concentrations found in BM stromal cultures upregulated alpha4 expression and restored adhesion of mobilized PB CFC to stroma, fibronectin, and CS1. Adhesion of cultured, mobilized PB CFC to stroma and CS1 could not be further upregulated by the beta1 activating antibody, 8A2. This indicates acquisition of a maximally activated alpha4 beta1 integrin once PB CFC have been removed from the in vivo mobilizing milieu. Thus, decreased alpha4 expression on CD34(+) CFC in PB may be responsible for the aberrant circulation of mobilized PB CD34(+) cells. Reexpression of a maximally activated alpha4 beta1 integrin on mobilized PB CFC removed from the mobilizing in vivo milieu may contribute to the early engraftment of mobilized PBPC.


Asunto(s)
Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/fisiología , Integrinas/fisiología , Receptores Mensajeros de Linfocitos/fisiología , Antígenos CD34/análisis , Células de la Médula Ósea/fisiología , Adhesión Celular , Células Cultivadas , Regulación hacia Abajo , Fibronectinas/fisiología , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Integrina alfa4beta1 , Células del Estroma/fisiología
12.
Leukemia ; 19(3): 373-80, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15674360

RESUMEN

The mechanism underlying p210(BCR/ABL) oncoprotein-mediated transformation in chronic myelogenous leukemia (CML) is not fully understood. We hypothesized that p210(BCR/ABL) suppresses expression of genes which may explain at least some of the pathogenetic features of CML. A subtractive cDNA library was created between BCR/ABL-enhanced-green-fluorescent-protein (GFP)-transduced umbilical cord blood (UCB) CD34+ cells and GFP-transduced UCB CD34+ cells to identify genes whose expression is downregulated by p210(BCR/ABL). At least 100 genes were identified. We have confirmed for eight of these genes that expression was suppressed by quantitative real-time-RT-PCR (Q-RT-PCR) of additional p210(BCR/ABL)-transduced CD34+ UCB cells as well as primary early chronic phase (CP) bone marrow (BM) CML CD34+ cells. Imatinib mesylate reversed downregulation of some genes, to approximately normal levels. Several of the genes are implicated in cell adhesion and motility, including L-selectin, intercellular adhesion molecule-1 (ICAM-1), and the chemokine receptor, CCR7, consistent with the known defect in adhesion and migration of CML cells. Compared with GFP UCB or normal (NL) BM CD34+ cells, p210 UCB and CML CD34+ cells migrated poorly towards the CCR7 ligands, CCL19 and CCL21, suggesting a possible role for CCR7 in the abnormal migratory behavior of CML CD34+ cells.


Asunto(s)
Movimiento Celular/fisiología , Quimiocinas CC/fisiología , Proteínas de Fusión bcr-abl/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Receptores de Quimiocina/fisiología , Adhesión Celular/genética , Adhesión Celular/fisiología , Línea Celular , Movimiento Celular/genética , Quimiocina CCL19 , Quimiocina CCL21 , Quimiocinas CC/genética , Regulación hacia Abajo , Proteínas de Fusión bcr-abl/genética , Humanos , Molécula 1 de Adhesión Intercelular/genética , Células K562 , Selectina L/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ligandos , ARN Mensajero/genética , Receptores CCR7 , Receptores de Quimiocina/genética
13.
Leukemia ; 12(2): 136-8, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9519773

RESUMEN

CML, characterized by the BCR/ABL gene rearrangement has been more extensively studied than any other malignancy. Over the last decade, significant progress has been made in our understanding of BCR-ABL-induced alterations in intracellular signaling. Unfortunately, we still only poorly understand the correlation between the clinical symptoms of chronic phase CML and the BCR-ABL oncoprotein. This is in part due to lack of a good in vivo animal model of chronic phase CML. In vivo and in vitro studies from the Clarkson group, recently reviewed in this journal (Leukemia 1997; 11: 1404-1428), have significantly enhanced our understanding of the pathophysiology of CML. However, further characterization of the effect of the BCR-ABL oncoprotein on signal molecules involved with cell differentiation, cell proliferation, cell survival and cell adhesion in primary Ph+ CML progenitors or in vivo models of CML will be needed to provide a full understanding of the pathophysiology of chronic phase CML.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Animales , Proteínas de Fusión bcr-abl/genética , Reordenamiento Génico , Humanos
14.
Leukemia ; 10(3): 498-504, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8642869

RESUMEN

Significantly less long-term culture initiating cells (LTC-IC) are recovered from cultures in which progenitors are cultured in contact with stroma (stroma-contact) than when cultured separated from stroma by a transwell (stroma-noncontact). This suggests that direct contact with stroma inhibits either proliferation or survival of LTC-IC. Using the membrane intercallating fluorochrome, PKH-26, we demonstrated that significantly less LTC-IC plated for 14 days in stroma-contact cultures proliferated than in stroma-noncontact cultures (16+/-7 vs 50+/-10%). Furthermore, when LTC-IC were sorted singly in stroma-contact cultures for 2 weeks, only 25+/-4% of individual LTC-IC progeny could initiate two secondary stromal cultures and had therefore proliferated, whereas 45+/-6% of single sorted LTC-IC progeny proliferated when cultured in stroma-conditioned medium without stromal feeder. However, LTC-IC survival was similar in both culture systems. Finally, proliferation inhibition occurred even when LTC-IC were cultured in contact with glutaraldehyde-fixed stroma, which is no longer capable of producing growth inhibitory or stimulatory cytokines. Thus, direct adhesive interactions between LTC-IC and stromal components inhibits their proliferation.


Asunto(s)
Células de la Médula Ósea , Células Madre Hematopoyéticas/citología , Compuestos Orgánicos , Adhesión Celular , División Celular , Supervivencia Celular , Células Cultivadas , Colorantes Fluorescentes , Humanos , Células del Estroma/citología , Factores de Tiempo
15.
Leukemia ; 14(9): 1614-20, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10995008

RESUMEN

To develop a model of early human adult B lymphopoiesis, we cultured CD34+CD38+CD10+ pro-B cells in contact with AFT024 stroma in X-VIVO10 media with 5% serum. The cytokines FLT3L + SCF + IL7 + IGF1 were added at day 0, IL4 + IL5 + IL6 + IL10 and soluble CD40 ligand at day 14, and Staph. aureus Cowan particles on day 21. Greater than 25-fold expansion of CD34+CD38+CD10+ cells was seen at 2 weeks, the majority being CD34-CD19+ pre-B cells. Differentiation to immature IgM+ B cells was seen at 3 weeks and mature IgD+ B cells at 4 weeks, with secretion of IgM into the media. Immature and mature B cells could also be generated from culture of CD34+CD10+CD19- and CD34+CD10+CD19+ cells under similar conditions. In conclusion, we have demonstrated in vitro differentiation of early pro-B cells, and possibly common lymphoid progenitor cells, to mature B cells. Additional stimuli, provided by T helper cells or dendritic cells for example, may be required for the generation of IgG+ B cells or plasma cells. However, our culture system should be a valuable tool to further investigate B cell biology and B cell malignancies such as multiple myeloma and lymphoma.


Asunto(s)
Linfocitos B/citología , Leucopoyesis/fisiología , Adolescente , Adulto , Antígenos CD19/análisis , Antígenos CD34/análisis , División Celular , Células Cultivadas , Herpesvirus Humano 4 , Humanos , Inmunoglobulinas/metabolismo , Técnicas In Vitro , Interleucina-2/fisiología , Interleucina-3/fisiología , Persona de Mediana Edad , Neprilisina/análisis
16.
Leukemia ; 18(4): 727-33, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14961028

RESUMEN

Chronic myelogenous leukemia (CML) is a malignancy of the human hematopoietic stem cell (HSC) caused by the p210BCR/ABL oncoprotein. Although alternative splicing of pre-mRNA is a critical determinant of a cell's protein repertoire, it has not been associated with CML pathogenesis. We identified a BCR/ABL-dependent increase in expression of multiple genes involved in pre-mRNA splicing (eg SRPK1, RNA Helicase II/Gu, and hnRNPA2/B1) by subtractive hybridization of cDNA from p210BCR/ABL-eGFP vs eGFP-transduced umbilical cord blood CD34+ cells. beta1-integrin signaling is important to HSC maintenance and proliferation/differentiation, and is abnormal in CML. As an example of how changes in pre-mRNA processing might contribute to CML pathogenesis, we observed alternative splicing of a gene for a beta1-integrin-responsive nonreceptor tyrosine kinase (PYK2), resulting in increased expression of full-length Pyk2 in BCR/ABL-containing cells. Treatment of p210BCR/ABL-positive cells with the Abl-specific tyrosine kinase inhibitor STI571 reverted PYK2 splicing to a configuration more consistent with normal cells, and correlated with decreased expression of BCR/ABL-induced proteins involved in pre-mRNA processing. Whether altered PYK2 splicing contributes to CML pathogenesis remains undetermined; however, we propose that generic changes in pre-mRNA splicing as a result of p210BCR/ABL kinase activity may contribute to CML pathogenesis.


Asunto(s)
Proteínas de Fusión bcr-abl/fisiología , Células Madre Hematopoyéticas/patología , Proteínas Tirosina Quinasas/genética , Empalme del ARN , Antígenos CD34 , Quinasa 2 de Adhesión Focal , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/etiología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Mieloide de Fase Crónica/etiología , Leucemia Mieloide de Fase Crónica/genética , Leucemia Mieloide de Fase Crónica/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Precursores del ARN/genética
17.
Leukemia ; 12(11): 1708-17, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9823945

RESUMEN

Abnormal beta1 integrin receptor function may contribute to the continuous proliferation and abnormal circulation of malignant hematopoietic progenitors in chronic myelogenous leukemia (CML). Previous studies suggest that abnormal integrin function in CML progenitors is related to the presence of the BCR/ABL oncogene. BCR/ABL may alter integrin function in CML by phosphorylating cytoskeletal and/or signaling proteins important for normal integrin function. We evaluated the effect of Tyrphostin AG957, a protein tyrosine kinase (PTK) inhibitor which has activity against the p210BCR/ABL kinase, on beta1 integrin function in CML progenitors. Incubation of CML marrow CD34+HLA-DR+ cells with Tyrphostin AG957 at concentrations that did not affect colony-forming cells (CFC) viability, but which partly inhibited p210BCR/ABL kinase activity, significantly increased CML CFC adhesion to stroma and alpha4beta1 and alpha5beta1 integrin binding fragments of fibronectin (FN). CML CFC proliferation, unlike that of normal CFC, is not inhibited following integrin receptor engagement with FN or anti-integrin antibodies. AG957 did not alter CML CFC proliferation by itself, but resulted in significant inhibition of CML CFC proliferation following integrin engagement. Another PTK inhibitor, Tyrphostin AG555, which does not have anti-p210BCR/ABL kinase activity, did not affect CML CFC adhesion or proliferation. Neither AG957 nor AG555 affected normal CFC adhesion or proliferation. In BCR/ABL expressing cells, AG957 partially inhibited phosphorylation of several proteins that are BCR/ABL PTK substrates and are involved in normal integrin signaling. These studies suggest that abnormal tyrosine phosphorylation may play an important role in defective integrin function in CML progenitors.


Asunto(s)
Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Células Madre Hematopoyéticas/efectos de los fármacos , Integrina beta1/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Tirfostinos/farmacología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , División Celular/efectos de los fármacos , Células Cultivadas , Fibronectinas/metabolismo , Proteínas de Fusión bcr-abl/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Tumorales Cultivadas
18.
Leukemia ; 14(2): 307-11, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10673749

RESUMEN

One of the factors required for successful retroviral transduction is contact between viral particles and target cells. We hypothesized that combining agents that improve virus-target cell interaction via different mechanisms will increase transduction efficiency. We examined the transduction efficiency of leukemic K562 cells, primary normal and chronic myelogenous leukemia CD34+ cells with the amphotropic retroviral vector, G1Na, packaged in PA317 by enumerating G418-resistant colonies in semisolid media. We evaluated the ability of the recombinant fibronectin fragment, CH296, cationic lipids, or a transwell flow-through system, alone or in combination to improve retroviral transduction. Transduction of K562 cells improved 1.5 to two-fold with lipids or CH296, while their combination improved transduction 2.5-fold. Transduction of K562 cells in the transwell flow-through system improved transduction three-fold. Transduction of normal (NL) CD34+ CFC improved 10-fold with lipids and 20-fold with CH296. Lipid and CH296 had synergistic effects. The transwell flow-through system improved transduction of normal CD34+ CFC 30-fold. Finally, similar to what was seen for K562 cells, transduction of CML CFC improved two- to three-fold with either CH296 or lipids, whereas the combination had synergistic effects. We conclude that any physical means that enhances contact between viral particles and target cells improves transduction. Two such methods that have different action mechanisms have additive or synergistic effects on transduction.


Asunto(s)
Fibronectinas/farmacología , Células Madre Hematopoyéticas/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Lípidos/farmacología , Retroviridae/metabolismo , Transducción de Señal/efectos de los fármacos , Antígenos CD34/metabolismo , Cationes , Sinergismo Farmacológico , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Células K562/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Lípidos/química , Fragmentos de Péptidos , Proteínas Recombinantes/farmacología , Retroviridae/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
19.
Leukemia ; 13(7): 1079-84, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10400424

RESUMEN

Soluble factors produced by human marrow stroma or the murine marrow derived M2-10B4 cell line support ex vivo maintenance for 5-8 weeks of 50% of human long-term culture initiating cells (LTC-IC). As the AFT024 cell line supports LTC-IC cultured in contact conditions better than M2-10B4 feeders, we evaluated LTC-IC support in non-contact conditions above AFT024 feeders. We show that only 15% of LTC-IC were maintained for 5 weeks in AFT024 non-contact cultures (n=6, P<0.05). As AFT024-conditioned media added to M2-10B4 non-contact cultures did not inhibit LTC-IC maintenance, AFT024 cells do not secrete factors that inhibit LTC-IC growth. We next characterized heparan sulfate glycosaminoglycans (HS-GAGs) and cytokines produced by AFT024 cells, which are both required for LTC-IC maintenance in M2-10B4 non-contact cultures. The size and extent of O-sulfation of HS-GAGs in AFT024 and M2-10B4 conditioned medium were similar, indicating that absence of hematopoietic specific HS-GAGs is not responsible for the lack of hematopoietic in AFT024 non-contact cultures. Levels of 13 different cytokines secreted in AFT024- and M2-10B4-conditioned medium were similar. However, addition of human SCF, G-CSF, GM-CSF, LIF, MIP-1alpha and IL-6 in concentrations found in human marrow stroma-conditioned medium to AFT024 non-contact cultures increased LTC-IC-maintenance to 72% at 5 weeks. These cytokines improved LTC-IC maintenance in part through interaction with the progenitors and in part, through interaction with the AFT024 feeder. Thus, although LTC-IC maintenance is poor in AFT024 non-contact cultures, addition of human cytokines enhances LTC-IC maintenance in part through indirect effects on the AFT024 feeder. Characterization of known or novel growth factors secreted by AFT024 cells before and after cytokine stimulation may lead to the identification of cytokines that support growth of human hematopoietic stem cells.


Asunto(s)
Factores Biológicos/metabolismo , Citocinas/farmacología , Hígado/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Humanos , Hígado/embriología , Hígado/metabolismo , Estimulación Química , Factores de Tiempo
20.
Leukemia ; 16(8): 1402-11, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12145676

RESUMEN

Chronic myelogenous leukemia (CML), characterized by the BCR-ABL gene rearrangement, has been extensively studied. Significant progress has been made in the area of BCR-ABL-mediated intracellular signaling, which has led to a better understanding of BCR-ABL-mediated clinical features in chronic phase CML. Disease progression and blast crisis CML is associated with characteristic non-random cytogenetic and molecular events. These can be viewed as increased oncogenic activity or loss of tumor suppressor activity. However, what causes transformation and disease progression to blast crisis is only poorly understood. This is in part due to the lack of a good in vivo model of chronic phase CML even though animal models developed over the last few years have started to provide insights into blast crisis development. Thus, additional in vitro and in vivo studies will be needed to provide a complete understanding of the contribution of BCR-ABL and other genes to disease progression and to improve therapeutic approaches for blast crisis CML.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Animales , Apoptosis , Crisis Blástica/genética , Crisis Blástica/patología , Diferenciación Celular , Aberraciones Cromosómicas , Reparación del ADN , Progresión de la Enfermedad , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/fisiología , Genes Supresores de Tumor , Células Madre Hematopoyéticas/patología , Humanos , Vigilancia Inmunológica , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Ratones , Ratones Noqueados , Modelos Animales , Modelos Biológicos , Células Madre Neoplásicas/patología , Oncogenes , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA