Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
1.
J Surg Oncol ; 102(5): 539-42, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20812264

RESUMEN

Bevacizumab (Avastin™; rhuMab VEGF), a monoclonal antibody targeting vascular endothelial growth factor (VEGF), has seen increased use in the perioperative treatment of colorectal and pancreatic cancer. Little is known, however, regarding its impact on surgical outcomes in patients undergoing resection. The objective of this review was to examine if the addition of bevacizumab to existing neoadjuvant regimens increases morbidity after cancer resection.


Asunto(s)
Inhibidores de la Angiogénesis/efectos adversos , Anticuerpos Monoclonales/efectos adversos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de la Angiogénesis/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Bevacizumab , Neoplasias Colorrectales/cirugía , Humanos , Terapia Neoadyuvante , Neoplasias Pancreáticas/cirugía , Complicaciones Posoperatorias , Resultado del Tratamiento
2.
Blood ; 97(11): 3424-32, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11369633

RESUMEN

The retinoblastoma (Rb), cyclin-dependent kinase (CDK), and CDK inhibitor genes regulate cell generation, and deregulation can produce increased cell growth and tumorigenesis. Polycythemia vera (PV) is a clonal myeloproliferative disease where the mechanism producing increased hematopoiesis is still unknown. To investigate possible defects in cell-cycle regulation in PV, the expression of Rb and CDK inhibitor gene messenger RNAs (mRNAs) in highly purified human erythroid colony-forming cells (ECFCs) was screened using an RNase protection assay (RPA) and 11 gene probes. It was found that RNA representing exon 2 of p16(INK4a) and p14(ARF) was enhanced by 2.8- to 15.9-fold in 11 patients with PV. No increase of exon 2 mRNA was evident in the T cells of patients with PV, or in the ECFCs and T cells from patients with secondary polycythemia. p27 also had elevated mRNA expression in PV ECFCs, but to a lesser degree. Because the INK4a/ARF locus encodes 2 tumor suppressors, p16(INK4a) and p14(ARF) with the same exon 2 sequence, the increased mRNA fragment could represent either one. To clarify this, mRNA representing the unique first exons of INK4a and ARF were analyzed by semiquantitative reverse transcription-polymerase chain reaction. This demonstrated that mRNAs from the first exons of both genes were increased in erythroid and granulocyte-macrophage cells and Western blot analysis showed that the INK4a protein (p16(INK4a)) was increased in PV ECFCs. Sequencing revealed no mutations of INK4a or ARF in 10 patients with PV. p16(INK4a) is an important negative cell-cycle regulator, but in contrast with a wide range of malignancies where inactivation of the INK4a gene is one of the most common carcinogenetic events, in PV p16( INK4a) expression was dramatically increased without a significant change in ECFC cell cycle compared with normal ECFCs. It is quite likely that p16(INK4a) and p14(ARF) are not the pathogenetic cause of PV, but instead represent a cellular response to an abnormality of a downstream regulator of proliferation such as cyclin D, CDK4/CDK6, Rb, or E2F. Further work to delineate the function of these genes in PV is in progress. (Blood. 2001;97:3424-3432)


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Expresión Génica , Policitemia Vera/genética , Proteínas/genética , Anciano , Ciclo Celular , División Celular , Análisis Mutacional de ADN , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patología , Exones , Femenino , Granulocitos/química , Células Madre Hematopoyéticas/química , Humanos , Macrófagos/química , Masculino , Persona de Mediana Edad , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/química , Linfocitos T/metabolismo , Proteína p14ARF Supresora de Tumor
3.
Blood ; 97(4): 843, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11873810
5.
Br J Haematol ; 110(1): 63-70, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10930980

RESUMEN

Erythropoietin (EPO) and stem cell factor (SCF) are two important factors in human erythropoiesis. We have recently demonstrated that SCF and EPO synergistically activate mitogen-activated protein (MAP) kinase, thereby promoting growth of human erythroid colony-forming cells (ECFCs). In the present study, we have examined the intracellular mechanisms by which SCF and EPO maintain survival of these cells. In the absence of SCF and EPO, human ECFCs underwent rapid apoptosis. The process was significantly inhibited by addition of a single factor and was totally prevented in the presence of both factors. Treatment of ECFCs with wortmannin, a specific inhibitor of phosphoinositide 3-kinase (PI3K), inhibited the antiapoptotic effect of SCF but had no effect on that of EPO, indicating that SCF but not EPO inhibits apoptosis through the PI3K pathway. In contrast, treatment of ECFCs with PD98059, a specific inhibitor of MAP kinase/ERK kinase (MEK), inhibited cell growth but had no effect on the antiapoptotic activity of either SCF or EPO, suggesting that SCF and EPO prevent apoptosis of human ECFCs independent of the extracellular signal-regulated kinase (ERK) pathway. Interestingly, both EPO and SCF induced activation of PI3K. However, through PI3K, SCF caused activation of protein kinase B (PKB), an anti-apoptosis signal, whereas EPO led to activation of ERKs. Furthermore, the SCF- and EPO-maintained expression of antiapoptotic protein Bcl-XL was correlated with the activation of ERKs and was inhibited by PD98059, suggesting that Bcl-XL may not have a major role in preventing apoptosis of human ECFCs. Phosphorylated BAD was not affected by SCF, EPO or wortmannin. Taken together with our previous results, the present study indicates that SCF and EPO support survival and growth of human ECFCs through different signalling pathways and that they transduce distinctly different signals through activation of PI3K.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Precursoras Eritroides/fisiología , Eritropoyetina/farmacología , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Factor de Células Madre/farmacología , Androstadienos/farmacología , Células Cultivadas , Sinergismo Farmacológico , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Células Precursoras Eritroides/efectos de los fármacos , Flavonoides/farmacología , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Wortmanina
6.
Blood ; 95(12): 3742-9, 2000 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-10845905

RESUMEN

Based on the hypothesis that interferon gamma (IFN-gamma) may have stimulating effects on survival of hematopoietic progenitor cells, we examined the effect of IFN-gamma on apoptosis of mature erythroid colony-forming cells (ECFCs) derived from human peripheral blood obtained from normal, healthy volunteers. When the cells were cultured in the presence of IFN-gamma, even without erythropoietin (EPO), the viability of the cells was maintained for at least 36 hours. When apoptosis of ECFCs was assessed by flow cytometric analysis', using annexin V, IFN-gamma reduced the extent of apoptosis of the cells, as well as EPO. DNA fragmentation of ECFCs was also reduced by IFN-gamma. In cells cultured with IFN-gamma alone, expression of Bcl-x was detected but the level of expression decreased gradually during incubation for 36 hours, and the expression level was lower than incubation with EPO. Fas expression and activation of downstream caspases were assessed by flow cytometric analysis or fluorometric protease assay. IFN-gamma induced Fas expression of the cells without the activation of caspase8 or caspase3 during 16 hours of incubation, while deprivation of EPO induced expression of Fas and the activation of both caspase8 and caspase3. We propose that IFN-gamma produces a stimulating signal for the survival of mature erythroid progenitor cells by reducing apoptosis through a mechanism other than modulating Fas and one related to the expression of Bcl-x. (Blood. 2000;95:3742-3749)


Asunto(s)
Apoptosis/fisiología , Eritropoyetina/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Interferón gamma/farmacología , Anexina A5/análisis , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Medio de Cultivo Libre de Suero , Fragmentación del ADN , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Interleucina-3/farmacología , Cinética , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Proteínas Recombinantes/farmacología , Factor de Células Madre/farmacología
7.
Blood ; 94(12): 4067-76, 1999 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-10590051

RESUMEN

Erythropoietin (EPO), a major regulator of erythroid progenitor cells, is essential for the survival, proliferation, and differentiation of immature erythroid cells. To gain insight into the molecular mechanism by which EPO functions, we analyzed the activation of Jun N-terminal kinases (JNKs) and extracellular signal-regulated kinases (ERKs) in HCD-57 cells, a murine erythroid progenitor cell line that requires EPO for survival and proliferation. Withdrawal of EPO from the cell culture medium resulted in sustained activation of JNKs plus p38 MAP kinase, and inactivation of ERKs, preceding apoptosis of the cells. Addition of EPO to the EPO-deprived cells caused activation of ERKs accompanied by inactivation of JNKs and p38 MAP kinase and rescued the cells from apoptosis. Phorbol 12-myristate 13-acetate, which activated ERKs by a different mechanism, also suppressed the activation of JNKs and significantly retarded apoptosis of the cells caused by withdrawal of EPO. Furthermore, MEK inhibitor PD98059, which inhibited activation of ERKs, caused activation of JNKs, whereas suppression of JNK expression by antisense oligonucleotides and inhibition of p38 MAP kinase by SB203580 caused attenuation of the apoptosis that occurs upon withdrawal of EPO. Finally, the activation of JNKs and p38 MAP kinase and concurrent inactivation of ERKs upon withdrawal of EPO were also observed in primary human erythroid colony-forming cells. Taken together, the data suggest that activation of ERKs promotes cell survival, whereas activation of JNKs and p38 MAP kinase leads to apoptosis and EPO functions by controlling the dynamic balance between ERKs and JNKs.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Eritroblastos/patología , Eritroblastos/fisiología , Eritropoyetina/farmacología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Animales , Línea Celular , Supervivencia Celular/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos
8.
Blood ; 93(10): 3309-16, 1999 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-10233883

RESUMEN

Interferon gamma (IFNgamma) induces apoptosis in purified human erythroid colony-forming cells (ECFC) and inhibits cell growth. Fas (APO-1; CD95) and Fas ligand (FasL) mediate apoptosis induced by IFNgamma, because Fas is significantly upregulated by IFNgamma, whereas Fas ligand is constitutively present in the ECFC and neutralization of FasL greatly reduces the apoptosis. Because conversion of caspases from their dormant proenzyme forms to active enzymes has a critical role in transducing a cascade leading to apoptosis, we performed further studies of the expression and activation of caspases in normal human and IFNgamma-treated day-6 ECFC to better understand the mechanism of IFNgamma action in producing this cell death. RNase protection assays showed that the caspase-1, -2, -6, -8, and -9 mRNAs were upregulated by IFNgamma, whereas the caspase-5 and -7 mRNAs were not increased. Western blots showed that FLICE/caspase-8 was upregulated and activated by 24 hours of incubation with IFNgamma. FADD was not similarly altered by incubation with IFNgamma. Western blots of ICE/caspase-1, which might be required for amplification of the initial FLICE activation signal, showed that pro-ICE expression significantly increased after treatment with IFNgamma for 24 hours and cleavage of pro-ICE also increased. CPP32/apopain/caspase-3, responsible for the proteolytic cleavage of poly (ADP) ribose polymerase (PARP), was also studied and treatment of ECFC with IFNgamma resulted in an increased concentration of caspase-3 by 24 hours and a clear induction of enzyme activation by 48 hours, which was identified by the appearance of its p17-kD peptide fragment. The cleavage of PARP was demonstrated by an obvious increase of the 89-kD PARP cleavage product, which was observed at almost the same time as caspase-3 activation in the IFNgamma-treated cells, whereas untreated ECFC showed little change. Peptide inhibitors of the caspase proteins, DEVD-fmk, DEVD-cho, YVAD-cho, and IETD-fmk, were incubated with the ECFC to obtain further evidence for the involvement of caspases in IFNgamma-induced apoptosis. The activation of FLICE/caspase-8 and CPP32/caspase-3 and cleavage of PARP clearly were inhibited, but the reduction of cell growth due to apoptosis, induced by IFNgamma, was only partially blocked by the presence of the inhibitors. These results indicate that IFNgamma acts on ECFC not only to upregulate Fas, but also to selectively upregulate caspases-1, -3, and -8, which are activated and produce apoptosis, whereas the concentrations of FasL and FADD are not demonstrably changed.


Asunto(s)
Caspasa 1/genética , Caspasas/genética , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/fisiología , Regulación Enzimológica de la Expresión Génica , Interferón gamma/farmacología , Apoptosis/efectos de los fármacos , Caspasa 1/metabolismo , Caspasa 3 , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , Separación Celular/métodos , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Activación Enzimática , Células Precursoras Eritroides/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Cinética , ARN Mensajero/genética , Proteínas Recombinantes , Factores de Tiempo , Transcripción Genética/efectos de los fármacos
10.
Blood ; 92(4): 1142-9, 1998 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-9694701

RESUMEN

Stem cell factor (SCF) and erythropoietin (EPO) work synergistically to support erythropoiesis, but the mechanism for this synergism is unknown. By using purified human erythroid colony-forming cells (ECFC), we have found that SCF and EPO synergistically activate MAP kinase (MAPK, ERK1/2), which correlates with the cell growth and thus may be responsible for the synergistic effects. Treatment of the cells with PD98059 and wortmannin, inhibitors of MEK and PI-3 kinase, respectively, inhibited the synergistic activation of MAPK and also the cell growth, further supporting this conclusion. Wortmannin only inhibits MAPK activation induced by EPO but not that by SCF, suggesting that SCF and EPO may activate MAPK through different pathways, which would facilitate synergy. Furthermore, EPO, but not SCF, led to activation of STAT5, whereas SCF and wortmannin had no effect on the EPO-induced STAT5 activation, suggesting that STAT5 is not involved in the synergistic action of SCF and EPO. Together, the data suggest that synergistic activation of MAPK by SCF and EPO is essential for expanded erythropoiesis.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Células Precursoras Eritroides/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Eritropoyetina/farmacología , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas de la Leche , Proteínas Quinasas Activadas por Mitógenos , Transducción de Señal/efectos de los fármacos , Factor de Células Madre/farmacología , Androstadienos/farmacología , Células Cultivadas , Proteínas de Unión al ADN/fisiología , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Humanos , Proteína Quinasa 1 Activada por Mitógenos , Proteína Quinasa 3 Activada por Mitógenos , Fosfatidilinositol 3-Quinasas/efectos adversos , Fosforilación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptores de Eritropoyetina/metabolismo , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT5 , Transactivadores/fisiología , Wortmanina
11.
Blood ; 91(4): 1235-42, 1998 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9454753

RESUMEN

Interferon gamma (IFNgamma) inhibits the growth and differentiation of highly purified human erythroid colony-forming cells (ECFCs) and induces erythroblast apoptosis. These effects are dose- and time-dependent. Because the cell surface receptor known as Fas (APO-1; CD95) triggers programmed cell death after activation by its ligand and because incubation of human ECFCs with IFNgamma produces apoptosis, we have investigated the expression and function of Fas and Fas ligand (FasL) in highly purified human ECFCs before and after incubation with IFNgamma in vitro. Only a small percentage of normal human ECFCs express Fas and this is present at a low level as detected by Northern blotting for the Fas mRNA and flow cytometric analysis of Fas protein using a specific mouse monoclonal antibody. The addition of IFNgamma markedly increased the percentage of cells expressing Fas on the surface of the ECFCs as well as the intensity of Fas expression. Fas mRNA was increased by 6 hours, whereas Fas antigen on the cell surface increased by 24 hours, with a plateau at 72 hours. This increase correlated with the inhibitory effect of IFNgamma on ECFC proliferation. CH-11 anti-Fas antibody, which mimics the action of the natural FasL, greatly enhanced IFNgamma-mediated suppression of cell growth and production of apoptosis, indicating that Fas is functional. Expression of FasL was also demonstrated in normal ECFCs by reverse transcriptase-polymerase chain reaction and flow cytometric analysis with specific monoclonal antibody. FasL was constitutively expressed among erythroid progenitors as they matured from day 5 to day 8 and IFNgamma treatment did not change this expression. Apoptosis induced by IFNgamma was greatly reduced by the NOK-2 antihuman FasL antibody and an engineered soluble FasL receptor, Fas-Fc, suggesting that Fas-FasL interactions among the ECFCs produce the erythroid inhibitory effects and apoptosis initiated by IFNgamma.


Asunto(s)
Apoptosis , Eritrocitos/patología , Eritropoyesis , Interferón gamma/farmacología , Glicoproteínas de Membrana/fisiología , Receptor fas/fisiología , Animales , Apoptosis/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Proteína Ligando Fas , Humanos , Ratones , Proteínas Recombinantes
12.
Blood ; 90(6): 2244-52, 1997 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9310475

RESUMEN

Interferon gamma (IFNgamma) has been shown to inhibit proliferation and differentiation of erythroid progenitor cells and to produce apoptosis of erythroid cells, whereas stem cell factor (SCF), erythropoietin (EP), and insulin-like growth factor-I (IGF-I) have distinct roles in enhancing erythroid cell production and preventing apoptosis. The mechanism by which IFNgamma exerts an inhibitory effect on the positive roles of these growth factors is unknown. Although some inhibitory cytokines including IFNgamma have been shown to downregulate growth factor receptors, the effect of IFNgamma on SCF, EP, and IGF-I receptors of human erythroid progenitor cells has not been defined. We obtained highly purified day-5 or day-6 erythroid colony-forming cells (ECFCs) from human blood in sufficient quantity and purity for radiolabeled cytokine binding studies and analysis of mRNA. When day-5 ECFCs were incubated with increasing concentrations of recombinant human (rh) IFNgamma for 24 hours at 37 degrees C, specific binding of 125I-rhSCF to SCF receptors was significantly decreased by 25% to 40% in a dose-dependent fashion, with the maximum effect at 2,500 to 5,000 U/mL of IFNgamma. The decrease was apparent by 12 hours of incubation and was only slightly lower by 24 hours. The numbers of SCF and EP receptors, but not of IGF-I receptors, per ECFC, calculated by Scatchard analysis, were significantly decreased by 30% and 23% to 25%, respectively, after incubation with 2,500 U/mL rhIFNgamma for 24 hours at 37 degrees C, whereas the binding affinities were not affected. This decrease in SCF receptors was confirmed by flow cytometry using an anti-c-kit mouse monoclonal antibody. Northern blot analysis showed that the mRNAs for the SCF and EP receptors, but not for the IGF-I receptors, were decreased by 50% to 60% after 3 hours of incubation at 37 degrees C with 2,500 U/mL of rhIFNgamma. This persisted for 24 hours without alteration of the stability of the SCF and EP receptor mRNAs. These observations suggest that one means by which IFNgamma inhibits erythroid cell proliferation and differentiation and produces apoptosis may be through the reduction of the number of target receptors for SCF and EP and that this occurs through transcriptional inhibition of the corresponding mRNAs.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Eritropoyetina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interferón gamma/farmacología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptores de Eritropoyetina/metabolismo , Factor de Células Madre/metabolismo , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , ARN Mensajero/genética , Proteínas Recombinantes
13.
Blood ; 90(2): 651-7, 1997 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-9226165

RESUMEN

Polycythemia vera (PV) is a clonal hematologic disease characterized by hyperplasia of the three major bone marrow lineages. PV erythroid progenitor cells display hypersensitivity to several growth factors, which might be caused by an abnormality of tyrosine phosphorylation. In the present study, we have investigated protein tyrosine phosphatase (PTP) activity in highly purified erythroid progenitor cells and found that the total PTP activity in the PV cells was twofold to threefold higher than that in normal cells. Protein separation on anion-exchange and gel-filtration columns showed that the increased activity was due to a major PTP eluted at approximately 170 kD. This enzyme was sensitive to PTP inhibitors and it did not cross-react with antibodies to SHP-1, SHP-2, or CD45. Subcellular fractionation showed that the PTP localized with the membrane fraction, where its activity was increased by threefold in PV erythroid progenitors when compared with normal cells. As the erythroid progenitors progressively matured, activity of the PTP declined rapidly in the normal cells but at a much slower rate in the PV cells. These studies suggest that a potentially novel membrane or membrane-associated PTP, representing a major PTP activity, may have an important role in proliferation and/or survival of human erythroid progenitors and that its hyperactivation in PV erythroid progenitors might be responsible for the increased erythropoiesis in PV patients.


Asunto(s)
Células Precursoras Eritroides/enzimología , Policitemia Vera/enzimología , Proteínas Tirosina Fosfatasas/sangre , Adulto , Secuencia de Aminoácidos , Biomarcadores/sangre , Membrana Celular/enzimología , Células Cultivadas , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Citosol/enzimología , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/patología , Humanos , Cinética , Masculino , Persona de Mediana Edad , Peso Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Policitemia Vera/sangre , Proteínas Tirosina Fosfatasas/aislamiento & purificación , Valores de Referencia , Especificidad por Sustrato
14.
Blood ; 89(10): 3574-81, 1997 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-9160662

RESUMEN

Erythropoietin (EP) and stem cell factor (SCF) are essential growth factors for erythroid progenitor cell proliferation and differentiation in serum-free culture. It has been previously shown that burst-forming units-erythroid and colony-forming units-erythroid from patients with polycythemia vera (PV) have enhanced sensitivity to EP and SCF compared with normal erythroid progenitors, but little is known about the mechanism for this difference. In the present investigation, the effect of EP and SCF on protein tyrosine phosphorylation in day-8 normal and PV erythroid colony-forming cells, which give rise to colonies of 2-49 hemoglobinized cells, was studied. EP rapidly induced tyrosine phosphorylation of the EP receptor, whereas the most prominent phosphorylated protein induced by SCF was identified as the SCF receptor. No additional phosphorylated proteins were evident when PV cells were compared with normal cells. Culture of normal erythroid progenitors with orthovanadate, an inhibitor of protein tyrosine phosphatases, resulted in an increased number of erythroid colonies and enhanced protein tyrosine phosphorylation. However, in contrast, little enhancement was evident with PV cells. These results indicate that, although vanadate may be acting in normal erythroid progenitors as a phosphatase inhibitor that potentiates the kinase activity induced by SCF and EP, this function is diminished in PV cells. Because erythropoiesis is regulated by a balance between protein tyrosine kinase activity and protein tyrosine phosphatase activity, PV patients may have an abnormal phosphatase activity allowing increased cell proliferation.


Asunto(s)
Células Precursoras Eritroides/efectos de los fármacos , Policitemia Vera/patología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Tirosina Fosfatasas/deficiencia , Vanadatos/farmacología , División Celular/efectos de los fármacos , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Medio de Cultivo Libre de Suero , Células Precursoras Eritroides/enzimología , Células Precursoras Eritroides/patología , Eritropoyetina/farmacología , Humanos , Fosforilación/efectos de los fármacos , Policitemia Vera/enzimología , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Recombinantes/farmacología , Factor de Células Madre/farmacología
15.
Exp Hematol ; 25(3): 193-8, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9091293

RESUMEN

Interferon-gamma (IFN-gamma) has been shown to inhibit proliferation and differentiation of erythroid progenitor cells and to produce apoptosis of erythroid cells, but IFN-gamma receptors are not present on red cells and have never been demonstrated on erythroid progenitor cells. We obtained highly purified day 6 erythroid colony-forming cells (ECFCs) from human blood in sufficient quantity and purity to measure binding of radioiodinated recombinant human IFN-gamma ([125I]rhIFN-gamma). When [125I]rhIFN-gamma was incubated with day 6 ECFC, 77% of the binding was inhibited by excess unlabeled rhIFN-gamma, but no inhibition occurred with a variety of growth factors and glycoproteins. Specific binding was directly proportional to the cell concentration with a straight line passing through the origin, and equilibrium was reached at 0 degree C by 24-48 hours. Saturation of specific binding occurred at a [125I]rhIFN-gamma concentration of 1.0 nM and internalization was demonstrated with further incubation at 37 degrees C. Scatchard analysis showed a single class of binding sites and at a high ECFC cell purity of 80-89%, 1910-2070 binding sites per ECFC were present with a Kd of 0.01-0.02 nM. As day 5 ECFC developed into more mature day 7-day 12 cells, with incubation at 37 degrees C in vitro, specific binding for [125I]IFN-gamma greatly decreased. These experiments delineate specific binding sites for IFN-gamma on human erythroid progenitor cells and indicate that the enhanced sensitivity to rhIFN-gamma inhibition of mature day 3-day 6 burst-forming units-erythroid may be a result of enhanced specific binding. Human IFN-gamma is a multifunctional lymphokine, secreted by activated T lymphocytes and NK cells, which exerts antiviral, antiproliferative, and immunomodulatory activities on a wide variety of cells [1,2]. With regard to hematopoietic cells, IFN-gamma has been reported to inhibit the growth of granulocyte-macrophage colony-forming units, burst-forming units-erythroid (BFU-E) and colony-forming units-erythroid (CFU-E) in vitro [3-7]. Most recently, mature day 3 to day 6 BFU-E have been shown to be most sensitive to the inhibitory effect of recombinant human (rh) IFN-gamma, while primitive day 1 to day 2 cells and later day 7 cells were less affected [7]. Incubation of rhIFN-gamma with mature BFU-E inhibits hemoglobin accumulation and produces apoptosis of the maturing erythroid cells [7]. Moreover, since blood IFN-gamma levels are elevated and vary directly with the degree of the anemia, in patients with hematologic malignancies [8] and HIV-seropositivity [9], IFN-gamma appears to have a prominent role in producing the anemia associated with chronic disease [10,11]. Although characterization of human IFN-gamma receptors has been extensively performed for a variety of human cells including fibroblasts, lymphocytes, monocytes, granulocytes, eosinophiles, platelets, and many tumor cells [12-17], IFN-gamma receptors have not been identified on red cells [12] and the presence plus the extent of IFN-gamma receptors on progenitor cells, including human erythroid progenitor cells, remains unknown. A method has been reported from our laboratory by which human erythroid colony-forming cells (ECFC) can be highly purified, starting with peripheral blood BFU-E, in a sufficient amount for analysis of cytokine binding [18-20]. In this paper, we report the results of [125I]rhIFN-gamma binding to day 6 ECFC in vitro and demonstrate the presence of specific binding that is saturable at 1.0 nM. Scatchard analysis reveals that there are 1910-2070 rhIFN-gamma binding sites per ECFC with a Kd of 0.01-0.02 nM and, as with erythropoietin (EP) and insulin-line growth factor I (IGF-I) receptors, specific binding is highest with the earliest BFU-E studied and declines progressively as the erythroid progenitors mature.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Interferón gamma/metabolismo , Receptores de Interferón/sangre , Ensayo de Unidades Formadoras de Colonias , Endocitosis , Células Precursoras Eritroides/metabolismo , Humanos , Factores de Tiempo , Receptor de Interferón gamma
16.
J Cell Physiol ; 168(2): 255-63, 1996 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8707861

RESUMEN

The expression of c-myc was analyzed in murine and human erythroblasts throughout their differentiation in vitro into reticulocytes. The murine cells were splenic erythroblasts from animals infected with the anemia strain of Friend virus (FVA cells). In FVA cells cultured without EPO, the c-myc mRNA and protein levels decrease sharply within 3 to 4 h, showing that continual EPO stimulation is required to maintain c-myc expression. When cultured with EPO, the c-myc mRNA level of FVA cells is raised within 30 min of exposure. The c-myc mRNA and protein reach maxima at 1 to 3 h, then decline slowly to very low levels by 18 h. In contrast, c-fos and c-jun mRNA levels are not regulated by EPO in FVA cells. The human cells analyzed were colony-forming units-erythroid, CFU-E, derived in vitro by the culture of peripheral blood burst-forming units-erythroid (BFU-E). When grown in EPO and insulin-like growth factor 1 (IGF-1) these cells differentiate into reticulocytes over 6 days rather than the 2 days required for murine cells, but the c-myc mRNA kinetics and response to EPO parallel those of mouse cells at similar stages of differentiation. Both IGF-1 and c-kit ligand (SCF) cause an additive increase in c-myc mRNA in human CFU-E in conjunction with EPO. These additive effects suggest that EPO, IGF-1, and SCF affect c-myc mRNA accumulation by distinct mechanisms. Addition of an antisense oligonucleotide to c-myc in cultures of human CFU-E specifically inhibited cell proliferation but did not affect erythroid cell differentiation or apoptosis. When human cells were grown in high SCF concentrations, an environment which enhances proliferation and retards differentiation, antisense oligonucleotide to c-myc strongly inhibited proliferation, but such inhibition did not induce differentiation. This latter result indicates that differentiation requires signals other than depression of c-Myc and resultant depression of proliferation.


Asunto(s)
Eritroblastos/citología , Células Precursoras Eritroides/citología , Regulación del Desarrollo de la Expresión Génica , Genes myc/genética , Animales , Secuencia de Bases , División Celular , Supervivencia Celular , Sondas de ADN , Electroforesis en Gel de Poliacrilamida , Eritroblastos/efectos de los fármacos , Eritroblastos/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoyetina/farmacología , Virus de la Leucemia Murina de Friend/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , Hierro/metabolismo , Ratones , Datos de Secuencia Molecular , Oligonucleótidos Antisentido , Proteínas Proto-Oncogénicas c-myc/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Células Madre/farmacología
17.
Blood ; 87(6): 2558-67, 1996 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-8630423

RESUMEN

The locus control region (LCR) far upstream of the human beta-like globin genes is defined by the preferential chromatin accessibility/DNase I hypersensitivity of four constituent DNA sites HS4, 3, 2, and 1. In an attempt to understand the mechanism of LCR function during early stages of erythropoiesis, a new polymerase chain reaction (PCR) method has been developed to examine the chromatin structure/DNase I hypersensitivity of the LCR in progenitor cells logistically available in limited cell numbers. In erythroid progenitors as well as in multipotent cells with erythroid potential, hypersensitive sites HS4, 3, 2, and 1 were present and the chromatin structure of the LCR was accessible. Moreover, the chromatin structure of the LCR underwent dynamic changes during erythropoiesis. In early erythroid progenitors, the HS2 site was more accessible than the HS3 site. In more mature erythroid progenitors, HS2 became less accessible than HS3 and the other sites. The results indicate that the transcriptional program of the globin genes is encoded, at least in part, in the chromatin accessibility of the LCR. This globin program was apparently initiated in multipotent cells and maintained in erythroid progenitors. Furthermore, the program could be modulated in response to cellular changes accompanying differentiation of the progenitor cells.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Eritropoyesis/genética , Regulación de la Expresión Génica , Globinas/genética , Reacción en Cadena de la Polimerasa , Secuencias Reguladoras de Ácidos Nucleicos , Adulto , Cromatina/genética , Desoxirribonucleasa I/metabolismo , Globinas/biosíntesis , Células HL-60/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Conformación de Ácido Nucleico , Transcripción Genética , Células Tumorales Cultivadas
18.
Exp Hematol ; 24(2): 204-8, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8641342

RESUMEN

Previous investigations have demonstrated that interferons alpha, beta, and gamma (alpha-, beta-, and gamma-IFN) are potent inhibitors of erythropoiesis in vitro. By utilizing a cell population enriched for human erythroid colony-forming units (CFU-E), we have previously demonstrated that the inhibitory effects of beta- and gamma-IFNs are direct effects, not requiring the presence of accessory cells, and that the inhibitory effect of recombinant human (rh) gamma-IFN could be corrected by high concentrations of rh erythropoietin (Epo). In this study, we compared the effects of rh(alpha)-IFN on cells enriched for CFU-E to its effects on unpurified marrow cells and found that although h(beta)-IFN (which shares a common receptor with alpha-IFN) directly inhibits CFU-E colony formation, the effect of rh(alpha)-IFN is indirect and is mediated by a soluble factor released from T lymphocytes in response to rh(alpha)-IFN. However, rh(alpha)-IFN enhanced the direct inhibitory effect of rh(gamma)-IFN on CFU-E not inhibited by rh(alpha)-IFN. The inhibitory effects of neither alpha- nor beta-IFN could be overcome by high levels of rhEpo. These findings imply that alpha- and beta-IFN exert different cellular effects despite binding to the same receptor. Failure of rhEpo to correct CFU-E colony inhibition by alpha- and beta-IFNs but not by gamma-IFN also suggests a mechanism for the differing degrees of response to different doses of rhEpo in patients with the anemia of chronic disease.


Asunto(s)
Células Precursoras Eritroides/efectos de los fármacos , Interferón Tipo I/farmacología , Interferón beta/farmacología , Linfocinas/fisiología , Receptores de Interferón/efectos de los fármacos , Linfocitos T/metabolismo , Anemia/fisiopatología , Células Sanguíneas/citología , Células de la Médula Ósea , Ensayo de Unidades Formadoras de Colonias , Depresión Química , Eritropoyetina/farmacología , Humanos , Interferón gamma/farmacología , Receptores de Interferón/fisiología , Proteínas Recombinantes , Transducción de Señal
19.
J Cell Physiol ; 165(2): 323-32, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7593210

RESUMEN

Highly purified human blood burst-forming units-erythroid (BFU-E) were used to study the effects of interferon gamma (IFN gamma). IFN gamma inhibited erythroid colony formation, cell proliferation, and differentiation of day 3 to day 6 mature BFU-E in a dose-dependent manner. The primitive BFU-E (day 1 and day 2 cells) and later day 7 cells were less affected. IFN gamma dose-response experiments demonstrated that the number and size of erythroid colonies were reduced at a concentration of 500 U/ml with more complete inhibition at 1,000 U/ml. Inhibition of day 4 to day 6 erythroid progenitors was first noted by 72 h of incubation with IFN gamma, and target cell growth and differentiation continued to decrease with further incubation. IFN gamma also induced erythroblast apoptosis which was demonstrated by both nuclear condensation and fragmentation plus flow cytometry with in situ end-labelling. Because day 3 to day 6 cells need stem cell factor (SCF) for development in serum-free culture, the relationship of IFN gamma inhibition to this growth factor was investigated. The reduction in the number of erythroid colonies by IFN gamma was reversed by SCF although the colony size was not completely re-established. In contrast, interleukin-3 did not have the capacity to overcome the inhibitory effects of IFN gamma. Since IFN gamma blood levels are elevated in some anemias of chronic disease, IFN gamma may have a role in promoting this anemia and its inhibitory effect might be better overcome by SCF plus EP. However, the mechanism by which these growth factors overcome the inhibition of IFN gamma, or vice versa, is unknown at the present time.


Asunto(s)
Células Precursoras Eritroides/efectos de los fármacos , Interferón gamma/antagonistas & inhibidores , Interferón gamma/farmacología , Factor de Células Madre/farmacología , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Senescencia Celular , Eritroblastos/efectos de los fármacos , Humanos , Factores de Tiempo
20.
Blood ; 86(2): 572-80, 1995 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-7541668

RESUMEN

Stem cell factor (SCF), the ligand for the c-kit tyrosine kinase receptor, markedly stimulates the accumulation of erythroid progenitor cells in vitro. We now report that SCF delays erythroid differentiation among the progeny of individual erythroid progenitors while greatly increasing the proliferation of these progeny. These effects appear to be independent of an effect on maintenance of cell viability. Highly purified day-6 erythroid colony-forming cells (ECFC), consisting mainly of colony-forming units-erythroid (CFU-E), were generated from human peripheral blood burst-forming units-erythroid (BFU-E). Addition of SCF to the ECFC in serum-free liquid culture, together with erythropoietin (EP) and insulin-like growth factor 1 (IGF-1), resulted in a marked increase in DNA synthesis, associated with a delayed peak in cellular benzidine positivity and a delayed incorporation of 59Fe into hemoglobin compared with cultures without SCF. In the presence of SCF, the number of ECFC was greatly expanded during this culture period, and total production of benzidine-positive cells plus hemoglobin synthesis were ultimately increased. To determine the effect of SCF on individual ECFC, single-cell cultures were performed in both semisolid and liquid media. These cultures demonstrated that SCF, in the presence of EP and IGF-1, acted on single cells and their descendants to delay erythroid differentiation while substantially stimulating cellular proliferation, without an enhancement of viability of the initial cells. This was also evident when the effect of SCF was determined using clones of ECFC derived from single BFU-E. Our experiments demonstrate that SCF acts on individual day-6 ECFC to retard erythroid differentiation while simultaneously providing enhanced proliferation by a process apparently independent of an effect on cell viability or programmed cell death.


Asunto(s)
Células Precursoras Eritroides/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Factores de Crecimiento de Célula Hematopoyética/farmacología , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Células Clonales/efectos de los fármacos , Medio de Cultivo Libre de Suero , Replicación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Precursoras Eritroides/citología , Hemoglobinas/biosíntesis , Humanos , Factor de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...