Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 572: 65-71, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34358965

RESUMEN

Previously, we reported that glyceraldehyde 3-phosphate dehydrogenase (GAPDH) is a binding partner of prolyl oligopeptidase (POP) in neuroblastoma NB-1 cells and that the POP inhibitor, SUAM-14746, inhibits cytosine arabinoside (Ara-C)-induced nuclear translocation of GAPDH and protects against Ara-C cytotoxicity. To carry out a more in-depth analysis of the interaction between POP and GAPDH, we generated POP-KO NB-1 cells and compared the nuclear translocation of GAPDH after Ara-C with or without SUAM-14746 treatment to wild-type NB-1 cells by western blotting and fluorescence immunostaining. Ara-C did not induce the nuclear translocation of GAPDH and SUAM-14746 did not protect against Ara-C cytotoxicity in POP-KO cells. These results indicate that the anticancer effects of Ara-C not only include the commonly known antimetabolic effects, but also the induction of cell death by nuclear transfer of GAPDH through interaction with POP.


Asunto(s)
Núcleo Celular/efectos de los fármacos , Citarabina/farmacología , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Prolil Oligopeptidasas/metabolismo , Muerte Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Citarabina/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Humanos , Prolina/análogos & derivados , Prolina/farmacología , Prolil Oligopeptidasas/antagonistas & inhibidores , Prolil Oligopeptidasas/deficiencia , Tiazolidinas/farmacología , Células Tumorales Cultivadas
2.
Neurotoxicology ; 28(1): 83-92, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16973216

RESUMEN

Intact cerebellum cytoarchitecture and cellular communication are indispensable for successful motor coordination and certain forms of memory. Cytosine arabinoside (AraC), often used as an anti-neoplastic agent in humans, can have cerebellum-targeting adverse effects. In order to characterize the nature of AraC-induced cerebellar lesions in an adult rodent model, we have administered AraC (400 mg/kg b.w., i.p.) in adult male Wistar rats for 5 days. The animals' walking pattern, motor coordination, locomotion, spatial navigation and cognition were evaluated, along with neurofilament- and calbindin-like distribution in the cerebellum. AraC-treated rats demonstrated a disturbed walking pattern and a reduced ability of motor learning and coordination, indicative of a mild cerebellar deficit. Although the general locomotion and spatial cognition of AraC-treated rats was not significantly altered, their navigation into the water, in terms of swimming velocity, was irregular, compared to vehicle-treated animals. Neurofilament-like immunostaining was reduced in the molecular cerebellar layer, while calbindin D 28 kDa levels were increased in Purkinje neurons, following AraC treatment. Administration of the antioxidant N-acetylcysteine (NAC) (200 mg/kg b.w., p.o.), for 14 days (prior to and during AraC treatment) largely prevented the AraC-induced behavioral deficits. Our in vivo model of neurotoxicity provides data on the AraC-induced behavioral and cellular alterations concerning the adult rat cerebellum. Furthermore, it provides evidence of a possible neuroprophylactic role of the antioxidant N-acetylcysteine in this model of chemotherapy-induced toxicity.


Asunto(s)
Acetilcisteína/farmacología , Antimetabolitos Antineoplásicos/toxicidad , Cerebelo/citología , Citarabina/antagonistas & inhibidores , Citarabina/toxicidad , Fármacos Neuroprotectores , Desempeño Psicomotor/efectos de los fármacos , Animales , Calcio/metabolismo , Cerebelo/efectos de los fármacos , Cognición/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/patología , Relación Dosis-Respuesta a Droga , Inmunohistoquímica , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Proteínas de Neurofilamentos/metabolismo , Equilibrio Postural/efectos de los fármacos , Ratas , Ratas Wistar , Percepción Espacial/efectos de los fármacos , Caminata
3.
Toxicol In Vitro ; 20(8): 1446-54, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16942854

RESUMEN

We studied the ability of prolyl oligopeptidase (POP) inhibitors, Z-Pro-Prolinal and JTP-4819, to prevent translocation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and formation of reactive oxygen species (ROS), in 6-hydroxydopamine (6-OHDA) and cytosine arabinoside (Ara-C) treated monkey fibroblast (CV1-P) and human neuroblastoma (SH-SY5Y) cells. The cells were pretreated with POP inhibitors (30 min) before addition of toxicants. GAPDH was analyzed by Western hybridization, ROS by fluorescent 2'7'-dichlorodihydro-fluorescein diacetate, and viability by the MTT method. Both toxicants induced GAPDH translocation to the particulate fraction (mitochondria and nuclei). Z-Pro-Prolinal was able to inhibit the translocation in 6-OHDA-exposed CV1-P cells. In SH-SY5Y cells and in JTP-4819 pretreated cells, no prevention of translocation was seen. However, the intensity of GAPDH in cytosolic fraction increased. Both inhibitors blocked 6-OHDA-induced ROS-production to the control level in CV1-P but, not in SH-SY5Y cells, without affecting their viability. In conclusion, POP inhibitors are able to prevent certain cell stress related factors such as ROS production or GAPDH translocation.


Asunto(s)
Dipéptidos/farmacología , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Oxidopamina/antagonistas & inhibidores , Oxidopamina/toxicidad , Inhibidores de Proteasas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Serina Endopeptidasas/metabolismo , Actinas/biosíntesis , Actinas/genética , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citarabina/antagonistas & inhibidores , Citarabina/toxicidad , Humanos , Prolil Oligopeptidasas , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Pirrolidinas/farmacología , Sales de Tetrazolio , Tiazoles , Proteína p53 Supresora de Tumor/metabolismo
4.
Hum Exp Toxicol ; 25(6): 319-24, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16866189

RESUMEN

beta-Glucan (BG) was tested in vitro to determine its potential clastogenic and/or anti-clastogenic activity, and attempts were made to elucidate its possible mechanism of action by using combinations with an inhibitor of DNA polymerase. The study was carried out on cells deficient (CHO-k1) and cells proficient (HTC) in phases I and II enzymes, and the DNA damage was assessed by the chromosomal aberration assay. BG did not show a clastogenic effect, but was anti-clastogenic in both cell lines used, and at all concentrations tested (2.5, 5 and 10 microg/mL) in combination with damage inducing agents (methylmethane sulfonate in cell line CHO-k1, and methylmethane sulfonate or 2-aminoanthracene in cell line HTC). BG also showed a protective effect in the presence of a DNA polymerase beta inhibitor (cytosine arabinoside-3-phosphate, Ara-C), demonstrating that BG does not act through an anti-mutagenic mechanism of action involving DNA polymerase beta.


Asunto(s)
Antimutagênicos/farmacología , Aberraciones Cromosómicas/efectos de los fármacos , Citarabina/antagonistas & inhibidores , Daño del ADN/efectos de los fármacos , Hordeum , Metilmetanosulfonato/antagonistas & inhibidores , Mutágenos/toxicidad , beta-Glucanos/farmacología , Animales , Antracenos/toxicidad , Antimetabolitos Antineoplásicos/toxicidad , Antimutagênicos/aislamiento & purificación , Antimutagênicos/uso terapéutico , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Citarabina/toxicidad , Metilmetanosulfonato/toxicidad , beta-Glucanos/aislamiento & purificación , beta-Glucanos/uso terapéutico
5.
Leukemia ; 20(4): 610-9, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16482209

RESUMEN

17-Allylamino-17-demethoxygeldanamycin (17-AAG) is a new anticancer agent currently in clinical trials. The ability of 17-AAG to abrogate the function of heat-shock protein Hsp90 and modulate cellular sensitivity to anticancer agents has prompted recent research to use this compound in drug combination therapy. Here we report that 17-AAG has striking opposite effects on the activity of arsenic trioxide (ATO) and ara-C. Combination of 17-AAG with ATO exhibited a synergistic effect in leukemia cells, whereas coincubation of 17-AAG and ara-C showed antagonistic activity. Mechanistic studies revealed that ATO exerted cytotoxic action by reactive oxygen species generation, and activated Akt survival pathway. 17-AAG abrogated Akt activation and enhanced the activity of ATO. In contrast, treatment of leukemia cells with 17-AAG caused a G1 arrest, a decrease in DNA synthesis and reduced ara-C incorporation into DNA, leading to antagonism. The ability of 17-AAG to enhance the antileukemia activity of ATO was further demonstrated in primary leukemia cells isolated from patients with acute myeloid leukemia and chronic lymphocytic leukemia, including cells from refractory patients. Our data suggest that combination of 17-AAG and ATO may be an effective therapeutic regimen. Caution should be exercised in using 17-AAG together with ara-C, as their combination effects are schedule dependent.


Asunto(s)
Antineoplásicos/farmacología , Arsenicales/farmacología , Benzoquinonas/farmacología , Citarabina/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Lactamas Macrocíclicas/farmacología , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Mieloide/metabolismo , Óxidos/farmacología , Enfermedad Aguda , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Arsenicales/antagonistas & inhibidores , Citarabina/antagonistas & inhibidores , ADN/biosíntesis , ADN/efectos de los fármacos , ADN/metabolismo , Esquema de Medicación , Antagonismo de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Quimioterapia Combinada , Fase G1/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/fisiología , Células HeLa , Humanos , Células Jurkat , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Mieloide/tratamiento farmacológico , Óxidos/antagonistas & inhibidores , Sensibilidad y Especificidad , Relación Estructura-Actividad , Células Tumorales Cultivadas
6.
Cancer Res ; 65(15): 6874-81, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16061671

RESUMEN

2'-C-cyano-2'-deoxy-1-beta-D-arabino-pentofuranosylcytosine (CNDAC) is a nucleoside analogue with a novel mechanism of action that is currently being evaluated in clinical trials. Incorporation of CNDAC triphosphate into DNA and extension during replication leads to single-strand breaks directly caused by beta-elimination. These breaks, or the lesions that arise from further processing, cause cells to arrest in G2. The purpose of this investigation was to define the molecular basis for G2 checkpoint activation and to delineate the sequelae of its abrogation. Cell lines derived from diverse human tissues underwent G2 arrest after CNDAC treatment, suggesting a common mechanism of response to the damage created. CNDAC-induced G2 arrest was instituted by activation of the Chk1-Cdc25C-Cdk1/cyclin B checkpoint pathway. Neither Chk2, p38, nor p53 was required for checkpoint activation. Inhibition of Chk1 kinase with 7-hydroxystaurosporine (UCN-01) abrogated the checkpoint pathway as indicated by dephosphorylation of checkpoint proteins and progression of cells through mitosis and into G1. Cell death was first evident in hematologic cell lines after G1 entry. As indicated by histone H2AX phosphorylation, DNA damage initiated by CNDAC incorporation was transformed into double-strand breaks when ML-1 cells arrested in G2. Some breaks were manifested as chromosomal aberrations when the G2 checkpoint of CNDAC-arrested cells was abrogated by UCN-01 but also in a minor population of cells that escaped to mitosis during treatment with CNDAC alone. These findings provide a mechanistic rationale for the design of new strategies, combining CNDAC with inhibitors of cell cycle checkpoint regulation in the therapy of hematologic malignancies.


Asunto(s)
Citarabina/análogos & derivados , Fase G2/efectos de los fármacos , Neoplasias Hematológicas/tratamiento farmacológico , Neoplasias Hematológicas/patología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Aberraciones Cromosómicas , Citarabina/antagonistas & inhibidores , Citarabina/farmacología , Daño del ADN , Interacciones Farmacológicas , Fase G2/genética , Neoplasias Hematológicas/genética , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Mitosis/efectos de los fármacos , Proteínas Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Estaurosporina/análogos & derivados , Estaurosporina/farmacología
7.
Neurochem Int ; 46(1): 61-72, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15567516

RESUMEN

Neurotrophin-induced neuroprotection against apoptosis was investigated using immature cultured cerebellar granule cells (CGC) from newborn rat pups. Apoptotic cell death induced by treatment with cytosine arabinoside (AraC) was confirmed by DNA fragmentation and quantified by cell survival assays. AraC was most effective in inducing apoptosis when added to CGC on the day of culture preparation, while less or no effect was observed when added at 24 or 48h after plating, respectively. Pretreatment of CGC cultures for 24h with brain-derived neurotrophic factor (BDNF) or neurotrophin-4 (NT-4), but not neurotrophin-3 (NT-3), robustly protected against AraC neurotoxicity. K252a, an inhibitor of the tropomyosin-related kinase (Trk) tyrosine kinase receptor family which showed no toxicity by itself, blocked BDNF protection of AraC-induced apoptosis in a concentration-dependent manner. Neither protein kinase C activation nor inhibition mimicked or affected BDNF protection against AraC neurotoxicity. BDNF, but not NT-3, treatment of immature CGC caused a marked, but transient activation of Akt through phosphatidylinositol (PI) 3-kinase. The neuroprotective effects of BDNF were suppressed by pretreatment with LY 294002 (a PI 3-kinase inhibitor). BDNF neuroprotection was also preceded by activation of mitogen activated protein kinase (MAPK) and suppressed by two MAPK/ERK (MEK)-selective inhibitors, PD 98059 and U-0126. Moreover, inhibitors of PI 3-kinase and MEK potentiated AraC-induced neurotoxicity. These results show that neurotrophins protect against AraC-induced apoptosis, at least in part, through TrkB-mediated activation of the PI 3-kinase/Akt and MEK signaling pathways.


Asunto(s)
Antimetabolitos Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Cerebelo/citología , Citarabina/antagonistas & inhibidores , Citarabina/toxicidad , Factores de Crecimiento Nervioso/farmacología , Neuronas/efectos de los fármacos , Animales , Western Blotting , Factor Neurotrófico Derivado del Encéfalo/farmacología , Supervivencia Celular/efectos de los fármacos , Cerebelo/efectos de los fármacos , Gránulos Citoplasmáticos/efectos de los fármacos , Fragmentación del ADN , Quinasas Quinasa Quinasa PAM/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Sprague-Dawley
8.
Biochem J ; 387(Pt 1): 231-8, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15540985

RESUMEN

The chimaeric protein Bcr/Abl, the hallmark of chronic myeloid leukaemia, has been connected with several signalling pathways, such as those involving protein kinase B/Akt, JNK (c-Jun N-terminal kinase) or ERKs (extracellular-signal-regulated kinases) 1 and 2. However, no data about the p38 MAPK (mitogen-activated protein kinase) have been reported. Here, we present evidence showing that Bcr/Abl is able to modulate this signalling pathway. Transient transfection experiments indicated that overexpression of Bcr/Abl in 293T cells is able to activate p38 MAPK or induce p73 stabilization, suggesting that c-Abl and Bcr/Abl share some biological substrates. Interestingly, the control exerted by Bcr/Abl on the p38 MAPK pathway was not only mediated by the tyrosine kinase activity of Bcr/Abl, as the use of STI571 demonstrated. In fact, Bcr alone was able to induce p38 MAPK activation specifically through MKK3 (MAP kinase kinase 3). Supporting these observations, chronic myeloid leukaemia-derived K562 cells or BaF 3 cells stably transfected with Bcr/Abl showed higher levels of phosphorylated p38 MAPK compared with Bcr/Abl-negative cells. While Bcr/Abl-negative cells activated p38 MAPK in response to Ara-C (1-beta-D-arabinofuranosylcytosine), Bcr/Abl-positive cells were unable to activate p38 MAPK, suggesting that the p38 MAPK pathway is not sensitive to Abl-dependent stimuli in Bcr/Abl-positive cells. Our results demonstrate that the involvement of Bcr/Abl in the p38 MAPK pathway is a key mechanism for explaining resistance to Ara-C, and could provide a clue for new therapeutic approaches based on the use of specific Abl inhibitors.


Asunto(s)
Proteínas de Fusión bcr-abl/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Benzamidas , Línea Celular , Línea Celular Tumoral , Citarabina/antagonistas & inhibidores , Citarabina/metabolismo , Citarabina/farmacología , Proteínas de Unión al ADN/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Proteínas de Fusión bcr-abl/fisiología , Genes Supresores de Tumor , Humanos , Mesilato de Imatinib , Riñón/química , Riñón/citología , Riñón/embriología , Riñón/metabolismo , Proteínas Nucleares/metabolismo , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-abl/metabolismo , Pirimidinas/farmacología , Proteína Tumoral p73 , Proteínas Supresoras de Tumor , Células U937/enzimología , Células U937/metabolismo , Células U937/patología
9.
Mol Pharmacol ; 62(6): 1446-55, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12435813

RESUMEN

Induction of apoptosis by chemotherapeutic drugs involves the sphingomyelin-ceramide (SM-CER) pathway. This signaling is critically dependent on reactive oxygen species (ROS) generation and p53/p56 Lyn activation. In this study, we have investigated the influence of protein kinase C (PKC) zeta overexpression on the SM-CER pathway in U937 human leukemia cell line. We show that PKCzeta overexpression resulted in delayed apoptosis and significant resistance to both 1-beta-D-arabinofuranosylcytosine (ara-C) and daunorubicin (DNR), but there was no significant protection against cell-permeant C(6)-CER. Moreover, PKCzeta overexpression abrogated drug-induced neutral sphingomyelinase stimulation and CER generation by inhibiting ROS production. We further investigated p53/p56 Lyn activation in PKCzeta-overexpressing U937 cells treated with ara-C or DNR. We demonstrate that PKCzeta inhibited p53/p56 Lyn phosphorylation and stimulation in drug- or H(2)O(2)-treated cells, suggesting that p53/p56 Lyn redox regulation is altered in PKCzeta-overexpressing cells. Finally, we show that PKCzeta-overexpressing U937 cells displayed accelerated H(2)O(2) detoxification. Altogether, our study provides evidence for the role of PKCzeta in the negative regulation of drug-induced SM-CER pathway.


Asunto(s)
Antineoplásicos/antagonistas & inhibidores , Sustancias Protectoras/farmacología , Proteína Quinasa C/farmacología , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Supervivencia Celular/efectos de los fármacos , Ceramidas/farmacología , Citarabina/antagonistas & inhibidores , Citarabina/farmacología , Daunorrubicina/antagonistas & inhibidores , Daunorrubicina/farmacología , Humanos , Peróxido de Hidrógeno/antagonistas & inhibidores , Peróxido de Hidrógeno/farmacología , Inactivación Metabólica , Oxidación-Reducción , Fosforilación , Proteína Quinasa C/biosíntesis , Proteína Quinasa C/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Esfingomielina Fosfodiesterasa/metabolismo , Transfección , Proteína p53 Supresora de Tumor/metabolismo , Células U937 , Familia-src Quinasas/metabolismo
10.
Leukemia ; 16(9): 1808-17, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12200697

RESUMEN

Methotrexate (MTX) and cytarabine have been widely used for the treatment of acute leukemias and lymphomas for over 30 years. However, the optimal schedule of this combination is yet to be determined and a variety of schedules of the combination has been used. We studied the cytotoxic effects of MTX and cytarabine in combination against human leukemia cell lines at various schedules in vitro. The effects of the combinations at the concentration of drug that produced 80% cell growth inhibition (IC(80)) were analyzed using the isobologram method of Steel and Peckham. Simultaneous exposure to MTX and cytarabine for 3 days produced antagonistic effects in human T cell leukemia, MOLT-3 and CCRF-CEM, B cell leukemia, BALL-1, Burkitt's lymphoma, Daudi, promyelocytic leukemia, HL-60 and Philadelphia chromosome-positive leukemia, K-562 cells. Simultaneous exposure to MTX and cytarabine for 24 h produced antagonistic effects, sequential exposure to MTX for 24 h followed by cytarabine for 24 h produced synergistic effects, and the reverse sequence produced additive effects in both CCRF-CEM and HL-60 cells. Sequential exposure to MTX for 24 h followed by cytarabine for 3 days also produced synergistic effects in MOLT-3 cells. Cell cycle analysis supported these observations. Our findings suggest that the simultaneous administration of MTX and cytarabine is not appropriate and the sequential administration of MTX followed by cytarabine may be the optimal schedule of this combination.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Citarabina/farmacología , Leucemia/tratamiento farmacológico , Metotrexato/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Citarabina/administración & dosificación , Citarabina/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Humanos , Metotrexato/administración & dosificación , Metotrexato/antagonistas & inhibidores , Sales de Tetrazolio , Tiazoles
11.
Cancer Chemother Pharmacol ; 50(2): 163-6, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12172984

RESUMEN

PURPOSE: Chemokines are a family of small proteins that regulate leukocyte infiltration into inflamed tissue and play key roles in the pathogenesis of many diseases. Some chemokines can also reversibly inhibit the proliferation of hematopoietic progenitors. We have previously found that the chemokine CCL21 (Exodus-2/SLC/6Ckine/TCA4) is a potent inhibitor of the proliferation of normal hematopoietic progenitors. In this study we sought to determine whether this inhibition of proliferation could be therapeutically exploited by protecting normal marrow progenitors from the cytotoxicity of the S phase-active chemotherapeutic agent Ara-C. METHODS: Untreated and CCL21-pretreated mice were given doses of Ara-C that are toxic to marrow myeloid progenitors. The recovery of these myeloid progenitors was analyzed by colony formation assays. RESULTS: It was found that pretreatment with small doses of CCL21 prevented the death of normal murine marrow progenitors from the toxic effects of Ara-C. CONCLUSIONS: The chemokine CCL21 may be able to prevent Ara-C myelosuppression during acute leukemia induction chemotherapy, and thereby decrease morbidity and mortality of such therapy, and shorten hospital stays.


Asunto(s)
Antimetabolitos Antineoplásicos/toxicidad , Enfermedades de la Médula Ósea/prevención & control , Médula Ósea/efectos de los fármacos , Quimiocinas CC/uso terapéutico , Citarabina/antagonistas & inhibidores , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Enfermedades de la Médula Ósea/inducido químicamente , División Celular/efectos de los fármacos , Quimiocina CCL21 , Ensayo de Unidades Formadoras de Colonias , Citarabina/toxicidad , Evaluación Preclínica de Medicamentos , Ratones , Fase S/efectos de los fármacos
12.
Blood ; 99(4): 1373-80, 2002 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11830489

RESUMEN

Development of resistance to cytarabine (AraC) is a major problem in the treatment of patients with acute myeloid leukemia (AML). Inactivation of deoxycytidine kinase (dCK) plays an important role in AraC resistance in vitro. We have identified inactive, alternatively spliced dCK forms in leukemic blasts from patients with resistant AML. Because these dCK-spliced variants were only detectable in resistant AML, it was hypothesized that they might play a role in AraC resistance in vivo. In the current study, the biologic role of the alternatively spliced dCK forms in AraC resistance was further investigated by retroviral transductions in rat leukemic cells. Introduction of inactive, alternatively spliced dCK forms into AraC-resistant K7 cells, with no endogenous wild-type (wt) dCK activity, could not restore AraC sensitivity, whereas wt dCK fully restored the AraC-sensitive phenotype. Transfection of alternatively spliced dCK forms into AraC-sensitive KA cells, as well as in human leukemic U937 cells and in phytohemagglutinin-stimulated T cells, did not significantly change sensitivity toward AraC. In addition, cotransduction of wt dCK with alternatively spliced dCK in K7 cells did not result in altered sensitivity to AraC compared with K7 cells only transduced with wt dCK. These data indicate that the alternatively spliced dCK forms cannot act as a dominant-negative inhibitor on dCK wt activity when they are coexpressed in a single cell. However, a cell expressing alternatively spliced dCK forms that has lost wt dCK expression is resistant to the cytotoxic effects of AraC.


Asunto(s)
Empalme Alternativo/fisiología , Citarabina/antagonistas & inhibidores , Desoxicitidina Quinasa/farmacología , Leucemia Mieloide/tratamiento farmacológico , Enfermedad Aguda , Animales , Células de la Médula Ósea/patología , Supervivencia Celular/efectos de los fármacos , Citarabina/farmacología , Citarabina/uso terapéutico , Desoxicitidina Quinasa/genética , Desoxicitidina Quinasa/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos , Humanos , Leucemia Mieloide/patología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacología , Ratas , Transducción Genética , Células Tumorales Cultivadas/efectos de los fármacos
13.
Oncol Res ; 10(6): 313-24, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9848102

RESUMEN

Overexpression of bcl-2 or bcl-XL has been found to inhibit the induction of apoptosis in malignant cells by a large number of agents including a wide variety of chemotherapeutic drugs. CD437 ¿6-[3-(1-adamantyl)-4 hydroxyphenyl]-2-naphthalene carboxylic acid¿ is a novel retinoid that induces apoptosis in a number of malignant cells through a unique mechanism of action. The addition of 1 microM CD437 to HL-60/NEO cells resulted in capase 3 (CPP32) activation and poly(ADP-ribose) polymerase (PARP) cleavage in 3 h whereas in bcl-2- or bcl-XL-overexpressing HL-60 cells CD437 induced CPP32 activation and PARP cleavage in 6 h. Although 50 and 300 nM CD437 were required to induce PARP cleavage in HL-60/NEO and HL-60/bcl-2, HL-60/bcl-XL cells, respectively, maximal apoptosis in both cell lines was achieved utilizing 300 nM CD437. All three cell lines, however, share identical dose-response curves in terms of their growth inhibition, suggesting that CD437-mediated inhibition of growth and induction of apoptosis represent two distinct and separable processes. In addition, CD437 induces GI arrest as well as p21WAFI/CIPI mRNA expression in these cells despite the overexpression of bcl-2 or bcl-XL. CD437 induced mitochondrial instability as indicated by cytochrome c leakage into the cytoplasm in all three cell lines. CD437 also induced growth inhibition and apoptosis of an apoptosis-resistant variant of the HL-60 cell line (HCW-2), which switched expression from bcl-2 to bcl-XL. CD437-mediated apoptosis is not accompanied by downregulation of bcl-2 or bcl-XL or upregulation of bax. The reason for the inability of bcl-2 or bcl-XL overexpression to inhibit CD437-mediated apoptosis is unclear. The ability of CD437 to initiate apoptosis in a spectrum of malignant cells without interference from bcl-2 or bcl-XL overexpression suggests that CD437 may possess significant therapeutic potential in the treatment of malignancy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Retinoides/farmacología , Antineoplásicos/antagonistas & inhibidores , Caspasa 3 , Caspasas/efectos de los fármacos , Caspasas/metabolismo , División Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/biosíntesis , Citarabina/antagonistas & inhibidores , Citarabina/farmacología , Grupo Citocromo c/metabolismo , Activación Enzimática/efectos de los fármacos , Células HL-60/efectos de los fármacos , Células HL-60/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , ARN Mensajero/metabolismo , Retinoides/antagonistas & inhibidores , Regulación hacia Arriba , Proteína X Asociada a bcl-2 , Proteína bcl-X
14.
Cancer Res ; 58(15): 3202-8, 1998 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-9699642

RESUMEN

Taxol, 1-beta-D-arabinofuranosylcytosine (ara-C), and etoposide induce apoptosis in HL-60 cells that is blocked by overexpression of Bcl-2 or Bcl-xL.A 60-amino acid "loop" domain of Bcl-2 and Bcl-xL that contains phosphorylation sites is known to negatively regulate their antiapoptotic function. In the present studies, Taxol-, ara-C-, or etoposide-induced apoptosis was examined in HL-60/Bcl-2delta and HL-60/Bcl-xLdelta cells that express the loop-deletional mutant cDNA constructs p19Bcl-2delta32-80 and p18Bcl-xLdelta26-83, respectively. This was compared with control HL-60/neo cells as well as HL-60/Bcl-2 and HL-60/Bcl-xL cells. The latter two cell lines overexpress full-length Bcl-2 and Bcl-xL, respectively. Immunoblot analyses showed that HL-60/neo and HL-60/Bcl-2delta cells express similar levels of p26Bcl-2. In contrast, as compared with HL-60/neo, HL-60/Bcl-xLdelta cells expressed significantly lower levels of p26Bcl-2. p29Bcl-xL and p21Bax levels were similar in all cell types. Exposure to etoposide (50 microM) or ara-C (100 microM) for 4 h induced apoptosis in HL-60/neo cells, but not in HL-60/Bcl-2, HL-60/Bcl-xL, HL-60/Bcl-2delta, or HL-60/Bcl-xLdelta cells. In contrast, Taxol treatment (500 nM for 24 h) triggered the molecular cascade of apoptosis, represented by the cytosolic increase of cytochrome c and poly(ADP-ribose) polymerase or the DNA fragmentation factor cleavage activity of caspase-3 in HL-60/neo cells as well as in HL-60/Bcl-xLdelta and HL-60/Bcl-2delta cells, but not in their counterparts overexpressing full-length Bcl-2 and Bcl-xL. Equal amounts of p26Bcl-2 were coimmunoprecipitated with apoptosis protease-activating factor 1 (APAF-1) in HL-60/neo and HL-60/Bcl-2delta cells, whereas a markedly higher level of p26Bcl-2 coimmunoprecipitated with APAF-1 in HL-60/Bcl-2 cells. In association with Taxol-induced apoptosis, the levels of Bcl-2 that were coimmunoprecipitated with APAF-1 declined in HL-60/neo and HL-60/Bcl-2delta cells. This was not observed in HL-60/Bcl-2 cells, in which Taxol-induced apoptosis was blocked. Previous studies have demonstrated that Taxol induces phosphorylation of Bcl-2 in association with Taxol-induced apoptosis of HL-60/neo cells. Immunoblot analysis demonstrated a Taxol-induced mobility shift of Bcl-2 but not p19Bcl-2delta. Taxol also increased [32P]Pi incorporation in p26Bcl-2, but not in p19Bcl-2delta or p18Bcl-xL. These findings indicate that the loop domain is necessary for the Taxol-induced mobility shift and phosphorylation of Bcl-2. Loop domain also seems to be necessary for the antiapoptotic effect of Bcl-2 against Taxol-induced apoptosis but not ara-C- or etoposide-induced apoptosis.


Asunto(s)
Antineoplásicos Fitogénicos/antagonistas & inhibidores , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Grupo Citocromo c/metabolismo , Paclitaxel/antagonistas & inhibidores , Paclitaxel/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Factor Apoptótico 1 Activador de Proteasas , Sitios de Unión , Western Blotting , Citarabina/antagonistas & inhibidores , Citarabina/farmacología , Citosol/metabolismo , ADN de Neoplasias/metabolismo , Etopósido/antagonistas & inhibidores , Etopósido/farmacología , Células HL-60/citología , Células HL-60/efectos de los fármacos , Células HL-60/metabolismo , Humanos , Mutación , Fosforilación , Poli(ADP-Ribosa) Polimerasas/metabolismo , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Transfección , Proteína X Asociada a bcl-2 , Proteína bcl-X
15.
Neurosci Lett ; 223(3): 141-4, 1997 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-9080452

RESUMEN

Cytosine arabinoside (1-beta-D-arabinofuranosylcytosine, AraC) is a commonly used antimitotic agent that kills proliferating cells by inhibiting DNA synthesis. We report that AraC is toxic to cultured chick embryo spinal cord motoneurons (MTNs) in a concentration-dependent fashion with an EC50 of about 2 microM. Interestingly, this type of MTN death is specific, resembles that occurring upon muscle extract (MEX) trophic deprivation regarding its morphological and temporal characteristics, and has apoptotic features, as judged by observation of nuclear morphology. The death of AraC-treated MTNs can be blocked by 2'-deoxycytidine (dC), a pyrimidine metabolite AraC is structurally related to. Overall, these findings suggest that dC may participate in a pathway, different from inhibition of DNA synthesis, that is necessary for cultured MTNs to respond to the trophic activities present in MEX.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Citarabina/farmacología , Neuronas Motoras/efectos de los fármacos , Neurotoxinas/farmacología , Médula Espinal/efectos de los fármacos , Animales , Células Cultivadas , Embrión de Pollo , Citarabina/antagonistas & inhibidores , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Concentración Osmolar , Médula Espinal/citología
16.
Int J Cancer ; 68(2): 245-50, 1996 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-8900436

RESUMEN

Fatty acid ester surfactants Cremophor EL and Solutol HS 15 were described earlier as modulators of multidrug resistance mediated by MDR1 P-glycoprotein (Pgp). We have shown that the most active components of these polydisperse surfactants are fatty acid-polyethylene glycol-fatty acid diesters (FA-PEG-FA). A new generation of Pgp-surfactant inhibitors of defined structure was therefore synthesized. In the present study we show that these compounds are also able to inhibit up-regulation of MDR1 gene expression caused by cytarabine (ARA-C) and doxorubicin in human tumor cell lines H9 and KB 3-1, which express minimal levels of MDR1 mRNA. The surfactant inhibitors, however, had no effect on the induction of MDR1 gene expression by protein kinase C agonists. Using a set of FA-PEG-FA diesters with various fatty acids and different lengths of the PEG domain, we demonstrated that the activity of diester preparations as inhibitors of drug-induced MDR1 activation was in proportion to their activity as inhibitors of Pgp function. Oleic and stearic acid diesters with PEG 900 (20 ethylene oxide units) were the most potent. The poloxamer analogs of these diesters demonstrated similar effects. In contrast, the well-known, structurally unrelated inhibitors of Pgp activity, verapamil, cyclosporin A and PSC 833, had no inhibitory effect on drug-induced MDR1 activation. The ability of FA-PEG-FA diesters to inhibit both Pgp function and drug-induced MDR1 activation suggests that these chemomodulators may be uniquely useful for the prophylaxis of Pgp-mediated multidrug resistance in drug-treated tumors.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Antineoplásicos/antagonistas & inhibidores , Citarabina/antagonistas & inhibidores , Doxorrubicina/antagonistas & inhibidores , Ácidos Grasos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes MDR/efectos de los fármacos , Neoplasias/genética , Polietilenglicoles/farmacología , Tensoactivos/farmacología , Antineoplásicos/farmacología , Citarabina/farmacología , Doxorrubicina/farmacología , Genes MDR/genética , Humanos , Activación Transcripcional , Células Tumorales Cultivadas
17.
Biochem Biophys Res Commun ; 218(3): 753-8, 1996 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-8579586

RESUMEN

Cytosine arabinoside, a potent antimitotic agent, is used clinically as an anticancer drug with the side effect of severe neurotoxicity. Earlier reports suggested that this agent blocks the signaling pathway of nerve growth factor in sympathetic neurons, which results in neuronal death. The present study demonstrated that neuronal PC12 cells deprived of nerve growth factor showed a decrease of neurite outgrowth, which was not seen in cytosine arabinoside-treated neuronal PC12 cells, suggesting that this agent may not interfere with the part of the NGF signaling pathway that leads to neurite outgrowth. On the other hand, treatment of neuronal PC12 cells with cytosine arabinoside led to a dose- and time-dependent decrease of viability. These results suggest that there is a distinction between the neurite outgrowth and the survival promoting activities of nerve growth factor.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Citarabina/toxicidad , Neuritas/ultraestructura , Células PC12/citología , Animales , Ácido Aurintricarboxílico/farmacología , Diferenciación Celular/efectos de los fármacos , Citarabina/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Factores de Crecimiento Nervioso/farmacología , Cloruro de Potasio/farmacología , Ratas , Transducción de Señal/efectos de los fármacos
18.
Leukemia ; 9(11): 1851-6, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7475274

RESUMEN

By suppressing apoptosis, hemopoietic growth factors (HGFs) promote the survival of CD34+, HLA-DR+ marrow cells that are enriched for hemopoietic progenitor cells (HPC). In the present studies, we have examined the effects of pIXY321, a genetically engineered fusion protein of GM-CSF and IL-3 (GM-CSF/IL-3), on high-dose Ara-C (HIDAC) and taxol-induced apoptosis and survival of a multilineage HPC, the CFU-GEMM. Exposure to 1.0 mumol/l taxol for 24 h or HIDAC > or = 10 mumol/l for 4 h induced internucleosomal DNA fragmentation and the morphologic features of apoptosis in CD34+, HLA-DR+ cells. These treatments were associated with > or = 50% inhibition of the assayable CFU-GEMM colony numbers. Incubation in serum-free medium (SFM) alone for 24 h also induced apoptosis of CD34+, HLA-DR+ cells, which was associated with reduced intracellular levels of the bcl-2 gene product p26BCL-2. Co-treatment with pIXY321 (10 ng/ml) inhibited apoptosis of CD34+, HLA-DR+ cells incubated in SFM, without significantly increasing the intracellular p26BCL-2 levels. Furthermore, co-treatment with pIXY321 significantly reduced taxol- and Ara-C-induced apoptosis and promoted the survival of CFU-GEMM (P < 0.05). Taxol and Ara-C mediated apoptosis of CD34+, HLA-DR+ cells, and its inhibition by pIXY321, was not accompanied by any significant alteration in the intracellular p26BCL-2 levels. By demonstrating that co-treatment with pIXY321 confers significant protection against apoptosis of CD34+, HLA-DR+ cells as well as promotes survival of normal HPC exposed to clinically relevant schedules and concentrations of taxol of Ara-C, these results support the design of chemotherapy regimens incorporating pIXY321 plus taxol and/or high-dose Ara-C for solid tumors and/or acute leukemias. It is hoped that the use of such a cytokine might maintain normal HPC numbers following chemotherapy, therefore avoiding prolonged suppression.


Asunto(s)
Apoptosis/efectos de los fármacos , Citarabina/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Interleucina-3/farmacología , Paclitaxel/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/farmacología , Antígenos CD34/análisis , Células de la Médula Ósea , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Antígenos HLA-DR/análisis , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Humanos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2
19.
Neurodegeneration ; 4(1): 99-106, 1995 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7600190

RESUMEN

We have measured the effects of cytosine-beta-D-arabinofuranoside (AraC) on cell survival and neurite outgrowth in cultures of dissociated 4-6 day embryonic chick neural tube cells. High concentrations of AraC (greater than 100 microM) reduced neuronal cell survival and neurite outgrowth from viable cells. Concentrations normally used to inhibit mitotic cell division (1-10 microM) were toxic to the neurones cultured in serum free medium on a poly-DL-ornithine/laminin substrate. AraC does not appear to have a neurite promoting effect on dissociated neurones that are cultured in the presence of low numbers of non-neuronal cells. This suggests that the neurite promoting effects of AraC reported by others is likely to be through the non-neuronal cells that were an inherent feature of the culturing systems in these studies. AraC cytotoxicity was completely blocked by the addition of the competitive antagonist: 2'deoxycytidine (2'DC) but not by its metabolic precursor cytosine (cyt). We suggest that the acute effects of AraC on neurones which are actively growing neurites are the result of interference with lipid metabolism.


Asunto(s)
Sistema Nervioso Central/efectos de los fármacos , Citarabina/toxicidad , Neuritas/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Bioensayo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Sistema Nervioso Central/citología , Sistema Nervioso Central/embriología , Embrión de Pollo , Citarabina/antagonistas & inhibidores , Citosina/farmacología , Desoxicitidina/farmacología , Laminina
20.
Curr Eye Res ; 13(11): 839-43, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7851120

RESUMEN

Antifibrosis agents have improved the success of glaucoma filtration surgery, although undesired side effects are not readily reversible and may present a major limitation in the use of these agents. Our purpose was to study the efficacy of cytosine arabinoside (Ara-C) as an adjunctive antimetabolite in glaucoma surgery in the rabbit, and reversal of toxicity due to this agent with the competitive inhibitor 2'-deoxycytidine. Posterior lip sclerectomy was performed in rabbit eyes treated with 15 mg subconjunctival Ara-C daily for 7 d then every other day for 7 d. Mean intraocular pressure was lower in eyes treated with Ara-C compared with controls at all time points following filtration surgery. On the 10th postoperative day, the mean intraocular pressure of control eyes (25.0 +/- 1.9 mm Hg) had returned to baseline levels, whereas the intraocular pressure of eyes treated with Ara-C was significantly lower (16.0 +/- 1.7 mm Hg) (P < 0.001). Bleb survival was also prolonged in the Ara-C-treated eyes. The major ocular side effect of Ara-C was corneal toxicity, with epithelial defects in 40% of eyes after 8 daily injections of 15 mg Ara-C. Reversal of toxicity was enhanced with 2'-deoxycytidine, with complete resolution of epithelial toxicity after 6.5 +/- 1.7 d following daily topical 10% 2'-deoxycytidine compared with 12.7 +/- 0.58 for control (P < 0.002). These results demonstrate that postoperative subconjunctival injection of Ara-C results in improved bleb function after filtration surgery in the rabbit. Recovery from corneal epithelial toxicity due to Ara-C is markedly enhanced with the competitive inhibitor 2'-deoxycytidine.


Asunto(s)
Córnea/efectos de los fármacos , Enfermedades de la Córnea/prevención & control , Citarabina/toxicidad , Citarabina/uso terapéutico , Desoxicitidina/farmacología , Cirugía Filtrante , Animales , Quimioterapia Adyuvante , Enfermedades de la Córnea/inducido químicamente , Citarabina/antagonistas & inhibidores , Epitelio/efectos de los fármacos , Glaucoma/tratamiento farmacológico , Glaucoma/cirugía , Inyecciones , Presión Intraocular , Conejos , Escleroplastia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...