Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Mol Cell Biol ; 17(7): 3556-65, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9199291

RESUMEN

To study the mechanisms by which catecholamines regulate hepatocyte proliferation after partial hepatectomy (PHX), hepatocytes were isolated from adult male rats 24 h after sham operation or two-thirds PHX and treated with catecholamines and other agonists. In freshly isolated sham cells, p42 mitogen-activated protein (MAP) kinase activity was stimulated by the alpha1-adrenergic agonist phenylephrine (PHE). Activation of p42 MAP kinase by growth factors was blunted by pretreatment of sham hepatocytes with glucagon but not by that with the beta2-adrenergic agonist isoproterenol (ISO). In PHX cells, the ability of PHE to activate p42 MAP kinase was dramatically reduced, whereas ISO became competent to inhibit p42 MAP kinase activation. PHE treatment of sham but not PHX and ISO treatment of PHX but not sham hepatocytes also activated the stress-activated protein (SAP) kinases p46/54 SAP kinase and p38 SAP kinase. These data demonstrate that an alpha1- to beta2-adrenergic receptor switch occurs upon PHX and results in an increase in SAP kinase versus MAP kinase signaling by catecholamines. In primary cultures of hepatocytes, ISO treatment of PHX but not sham cells inhibited [3H]thymidine incorporation. In contrast, PHE treatment of sham but not PHX cells stimulated [3H]thymidine incorporation, which was reduced by approximately 25 and approximately 95% with specific inhibitors of p42 MAP kinase and p38 SAP kinase function, respectively. Inhibition of the p38 SAP kinase also dramatically reduced basal [3H]thymidine incorporation. These data suggest that p38 SAP kinase plays a permissive role in liver regeneration. Alterations in the abilities of catecholamines to modulate the activities of protein kinase A and the MAP and SAP kinase pathways may represent one physiological mechanism by which these agonists can regulate hepatocyte proliferation after PHX.


Asunto(s)
Agonistas Adrenérgicos/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Regeneración Hepática , Hígado/fisiología , Proteínas Quinasas Activadas por Mitógenos , Receptores Adrenérgicos alfa 1/fisiología , Receptores Adrenérgicos beta 2/fisiología , Estrés Fisiológico/fisiopatología , Animales , División Celular , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos , Hígado/citología , Masculino , Fosforilasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-raf , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos
2.
Cancer Res ; 54(7): 1707-14, 1994 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-7511048

RESUMEN

The present studies were undertaken to characterize further the potential role of protein kinase C (PKC) in the regulation of apoptosis in HL-60 promyelocytic leukemia cells. The capacity of acute exposure to specific and nonspecific pharmacological inhibitors of PKC to promote apoptotic DNA fragmentation was examined both quantitatively and qualitatively and correlated with effects on cellular differentiation and proliferation. Incubation of HL-60 cells for 6 h with chelerythrine and calphostin C (highly specific inhibitors that act at the regulatory domain) or H7 and gossypol (nonspecific inhibitors that act at the PKC catalytic domain) produced concentration-dependent increases in DNA fragmentation. Induction of DNA fragmentation by chelerythrine, calphostin C, and gossypol was biphasic, resulting in a sharp decline in effect at concentrations above 5 microM, 0.1 microM, and 100 microM, respectively, whereas maximal and more stable effects were observed in response to H7 (100 microM). A 6-h exposure to staurosporine, a nonspecific but potent PKC inhibitor, failed to induce DNA fragmentation at concentrations generally used to achieve maximal inhibition of enzyme activity (e.g., 50 nM) but promoted fragmentation at considerably higher concentrations (e.g., > or = 200 nM). In contrast, 6-h exposures to the nonspecific protein kinase inhibitor hypericin (0.1 to 100 microM) or to the nonspecific inhibitor of protein kinase A, HA1004 (50 microM), were without effect on DNA fragmentation. DNA obtained from cells exposed to chelerythrine (5 microM), calphostin C (100 nM), H7 (50 microM), gossypol (50 microM), and staurosporine (200 nM)--but not hypericin (25 microM)--exhibited clear evidence of internucleosomal DNA cleavage on agarose gel electrophoresis; moreover, these cells exhibited the classical morphological features of apoptosis (cell shrinkage, nuclear condensation, and the formation of apoptotic bodies). All of the PKC inhibitors that induced apoptosis, and one of the inhibitors that did not (hypericin), substantially inhibited HL-60 cell clonogenicity at the concentrations evaluated. None of the agents tested induced cellular maturation as assessed by nonspecific esterase and nitro-blue tetrazolium positivity. DNA fragments obtained from cells exposed to specific and nonspecific PKC inhibitors possessed predominantly 5'-phosphate termini, consistent with the action of a Ca(2+)-/Mg(2+)-dependent endonuclease. Finally, Northern blot analysis revealed that exposure to calphostin C at a concentration that induced apoptosis (100 nM) failed to alter expression of bcl-2, an oncogene known to block apoptosis in both lymphoid and myeloid leukemia cells.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Naftalenos , Proteína Quinasa C/antagonistas & inhibidores , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina , Alcaloides/toxicidad , Antracenos , Benzofenantridinas , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Gosipol/toxicidad , Humanos , Isoquinolinas/toxicidad , Leucemia Promielocítica Aguda , Perileno/análogos & derivados , Perileno/toxicidad , Fenantridinas/toxicidad , Piperazinas/toxicidad , Compuestos Policíclicos/toxicidad , Estaurosporina , Factores de Tiempo , Células Tumorales Cultivadas
3.
Cancer Res ; 52(22): 6270-8, 1992 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-1423273

RESUMEN

We have examined the interaction between 1-beta-D-arabinofuranosylcytosine (ara-C) and the macrocyclic lactone protein kinase C activator bryostatin 1 in the human promyelocytic leukemia cell line HL-60. Preexposure of cells to 10 nM bryostatin 1 for 24 h, followed by an additional 24-h incubation with 10 microM ara-C, resulted in greater than additive inhibitory effects toward clonogenic HL-60 cells. In a series of alkaline elution assays, cells preincubated with bryostatin 1 and prelabeled with [3H]thymidine exhibited a significant increase in DNA fragmentation following exposure to ara-C in comparison to cells exposed to ara-C alone. This increase in DNA damage was apparent at both neutral and alkaline pH and was not protein associated. In contrast, studies using cells pulse-labeled with [3H]thymidine immediately before analysis suggested that bryostatin 1 pretreatment did not increase the ability of ara-C to interfere with DNA replicative intermediates. Additional studies demonstrated that the increase in DNA fragmentation induced by bryostatin 1 and ara-C preceded both loss of cell membrane integrity (as determined by trypan blue exclusion) as well as depletion of intracellular ATP and NAD pools. Furthermore, the enhanced inhibitory effects of bryostatin 1 and ara-C toward clonogenic HL-60 cells did not appear to result from the induction of cellular differentiation. Finally, agarose gel electrophoresis of DNA obtained from cells exposed to both bryostatin 1 and ara-C revealed a pattern of integer multiples of 180- to 200-base pair fragments commonly associated with endonucleolytic cleavage; the extent of this fragmentation was considerably greater than that observed in cells exposed to ara-C alone. Taken together, these findings suggest that exposure of HL-60 cells to bryostatin 1 renders them more susceptible to ara-C-related DNA damage and that this phenomenon contributes to the cytotoxic effects of this drug combination. They also raise the possibility that bryostatin 1, perhaps through modulation of intracellular signaling events in leukemic cells, has the capacity to potentiate ara-C-related apoptosis or programmed cell death.


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Citarabina/farmacología , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , Lactonas/farmacología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Adenosina Trifosfato/metabolismo , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Brioestatinas , Calcitriol/farmacología , Citarabina/administración & dosificación , ADN/efectos de los fármacos , ADN de Neoplasias/metabolismo , Sinergismo Farmacológico , Humanos , Lactonas/administración & dosificación , Macrólidos , NAD/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Timidina/metabolismo , Factores de Tiempo , Tritio , Células Tumorales Cultivadas
4.
Oncogene ; 18(50): 7016-25, 1999 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-10597302

RESUMEN

Determinants of differentiation and apoptosis in myelomonocytic leukemia cells (U937) exposed to the novel hybrid polar compound SAHA (suberoylanilide hydroxamic acid) have been examined. In contrast to hexamethylenbisacetamide (HMBA), SAHA-related maturation was limited and accompanied by marked cytoxicity. SAHA-mediated apoptosis occurred within the G0G1 and S phase populations, and was associated with decreased mitochondrial membrane potential, caspase-3 activation, PARP degradation, hypophosphorylation/cleavage of pRB, and down-regulation of c-Myc, c-Myb, and B-Myb. Enforced expression of Bcl-2 or Bcl-XL inhibited SAHA-induced apoptosis, but only modestly potentiated differentiation. While SAHA induced the cyclin-dependent kinase inhibitor p21CIP1, antisense ablation of this CDKI increased, rather than decreased, SAHA-related lethality. In contrast, conditional expression of wild-type p53 failed to modify SAHA actions, but markedly potentiated HMBA-induced apoptosis. Finally, SAHA modestly increased expression/activation of the stress-activated protein kinase (SAPK/JNK); moreover, SAHA-related lethality was partially attenuated by a dominant-negative c-Jun mutant protein (TAM67). SAHA did not stimulate mitogen-activated protein kinase (MAPK), nor was lethality diminished by the specific MEK/MAPK inhibitor PD98059. These findings indicate that SAHA potently induces apoptosis in human leukemia cells via a pathway that is p53-independent but at least partially regulated by Bcl-2/Bcl-XL, p21CIP1, and the c-Jun/AP-1 signaling cascade.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclinas/metabolismo , Ácidos Hidroxámicos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Regulación hacia Abajo , Humanos , Leucemia/metabolismo , Leucemia/patología , Proteínas Proto-Oncogénicas c-myb/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Células U937 , Vorinostat , Proteína bcl-X
5.
Biochim Biophys Acta ; 1452(1): 1-11, 1999 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-10525155

RESUMEN

Over two dozen alternative splice variants of CaMK-II, the type II Ca(2+)/CaM-dependent protein kinase, are encoded from four genes (alpha, beta, gamma and delta) in mammalian cells. Isozymes of alpha and beta CaMK-II are well characterized in brain; however, an understanding of the relative endogenous levels of CaMK-II isozymes in a wide variety of non-neuronal cells has not yet been described. In this study, we have demonstrated that CaMK-II consists primarily of the 54 kDa delta CaMK-II (delta(2) or delta(C)) isozyme in rodent fibroblasts. beta and gamma CaMK-II isozymes are minor and alpha CaMK-II was not expressed. The primary delta CaMK-II in human fibroblasts and the MCF10A mammary epithelial cell line was the 52 kDa delta(4) CaMK-II, an isozyme identical to delta(2) except for a missing 21-amino-acid C-terminal tail. delta CaMK-II levels were diminished in both human and rodent fibroblasts after SV40 transformation and in the mammary adenocarcinoma MCF7 cell line when compared to MCF10A cells. In fact, most tumor cells exhibited CaMK-II specific activities which were two- to tenfold lower than in untransformed fibroblasts. We conducted complementary CaMK-II studies on the NGF-induced differentiation of rat PC-12 cells. Although no new synthesis of CaMK-II occurs, neurite outgrowth in these cells is accompanied by a preferential activation of delta CaMK-II. Endogenous delta CaMK-II has a perinuclear distribution in fibroblasts and extends along neurites in PC-12 cells. These findings point to a role for delta CaMK-II isozymes in cellular differentiation.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Células 3T3 , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/biosíntesis , Diferenciación Celular , Línea Celular Transformada , Regulación hacia Abajo , Humanos , Immunoblotting , Isoenzimas/metabolismo , Ratones , Células PC12 , Ratas , Células Tumorales Cultivadas
6.
Cell Death Differ ; 4(4): 294-303, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-16465244

RESUMEN

We have previously reported that pretreatment of HL-60 human promyelocytic leukemia cells with the non-tumor-promoting protein kinase C (PKC) activator bryostatin 1 potentiates induction of apoptosis by the antimetabolite 1-[beta-D-arabinofuranosyl]cytosine (ara-C) (Biochem Pharmacol 47:839,1994). To determine whether this phenomenon results from altered expression of Bcl-2 or related proteins, Northern and Western analysis was employed to assess the effects of bryostatin 1 and other PKC activators on steady-state levels of Bcl-2, Bax, Bcl-x, and Mcl-1 mRNA and protein. Pretreatment of cells for 24 h with 10 nM bryostatin 1, or, to a lesser extent, the stage-1 tumor-promoter phorbol dibutyrate (PDB) significantly potentiated apoptosis induced by ara-C (100 microM; 6 h); in contrast, equivalent exposure to the stage-2 tumor promoter, mezerein (MZN), which, unlike bryostatin 1, is a potent inducer of differentiation in this cell line, failed to modify ara-C-related cell death. Neither bryostatin 1 nor PDB altered expression of bcl-2/Bcl-2 over this time frame. In contrast, MZN down-regulated bcl-2 mRNA levels, but this effect was not accompanied by altered expression of Bcl-2 protein. None of the PKC activators modified expression of Bax or Bcl-x(L) mRNA or protein; levels of Bcl-x(S) were undetectable in both treated and untreated cells. However, expression of Mcl-1 mRNA and protein increased modestly after treatment with either bryostatin 1 or PDB, and to a greater extent following exposure to MZN. Combined treatment of cells with bryostatin 1 and MZN resulted in undiminished potentiation of ara-C-mediated apoptosis and by antagonism of cellular maturation. These effects were accompanied by unaltered expression of Bcl-2, Bax, and Bcl-x(L), and by a further increase in Mcl-1 protein levels. When cells were co-incubated with bryostatin 1 and calcium ionophore (A23187), an identical pattern of expression of Bcl-2 family members was observed, despite the loss of bryostatin 1's capacity to potentiate apoptosis, and the restoration of its ability to induce differentiation. Finally, treatment of cells with bryostatin 1+/-ara-C (but not ara-C alone) resulted in a diffuse broadening of the Bcl-2 protein band, whereas exposure of cells to taxol (250 nM, 6 h) led to the appearance of a distinct Bcl-2 species with reduced mobility, phenomena compatible with protein phosphorylation. Together, these findings indicate that the ability of bryostatin 1 to facilitate drug-induced apoptosis in human myeloid leukemia cells involves factors other than quantitative changes in the expression of Bcl-2 family members, and raise the possibility that qualitative alterations in the Bcl-2 protein, such as phosphorylation status, may contribute to this capacity. They also suggest that increased expression of Mcl-1 occurs early in the pre-commitment stage of myeloid cell differentiation, and that this event does not protect cells from drug-induced apoptosis.

7.
Leukemia ; 17(11): 2140-8, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12970763

RESUMEN

The present studies examined performance of SAPK cascades and apoptotic commitment following ribosomal trauma in REH lymphoid leukemia cells. Ribostatic insults included disruption of ribosomal activity by mechanistically dissimilar agents such as blasticidin-S (BCS) (which binds 28S-rRNA to block peptidyl bond formation), kasugamycin (KSM) (which binds 18S-rRNA to prevent translational initiation), and cycloheximide (CHX) (which blocks A-site to P-site translocation of peptidyl-tRNA). Exposure of REH cells to BCS elicited DNA degradation and apoptotic cytolysis. BCS stimulated JNK1/JNK2 and p38, and their shared targets c-Jun and ATF2. Inhibition of JNK1/JNK2 (but not of p38) antagonized blasticidin-induced apoptosis, whereas targeting alternative ribosomal sites with KSM or CHX limited translation, but failed to activate the SAPK cascade or initiate apoptosis. Our findings indicate that interference with 28S-rRNA by BCS initiates apoptosis in REH cells through recruitment of SAPK-JNK signaling. Disparities between the lethal actions of BCS, KSM, and CHX appear to reflect established differences in the subribosomal targets of these agents. We propose that the SAPK cascade comprises an essential mechanism for the transduction of specific lethal stress signals emanating from active ribosomes, and that interference with the 28S-rRNA, rather than the peptidyl transfer center of the large subunit, is critical to apoptotic commitment.


Asunto(s)
Apoptosis/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Caspasas/metabolismo , Cicloheximida/farmacología , ADN de Neoplasias/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Leucemia Linfoide , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Nucleósidos/toxicidad , Peptidil Transferasas/antagonistas & inhibidores , Células Tumorales Cultivadas
8.
Leukemia ; 12(12): 1843-50, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9844914

RESUMEN

During the last 10 years, multiple signal transduction pathways within cells have been discovered. These pathways have been linked to the regulation of many diverse cellular events such as proliferation, senescence, differentiation and apoptosis. This review will focus upon the many roles of signaling by the p42/p44 mitogen-activated protein (MAP) kinase pathway. Recent evidence suggests that signaling by the MAP kinase pathway can both enhance proliferation by increased expression of molecules such as cyclin D1, but also cause growth arrest by increased expression of molecules such as the cyclin kinase inhibitor protein p21(Cip-1/MDA6/WAF1). These differential effects on growth have been correlated to the amplitude and duration of the MAP kinase activity signal. Furthermore several laboratories are reporting data suggesting that inhibition of the MAP kinase pathway, as well as a family of upstream MAP kinase activators, the protein kinase C family, represent an important route to both radio- and chemo-sensitization of tumor cells. Herein, we describe the historical discovery and characterization of the MAP kinase pathway. In addition we describe potential mechanisms by which inhibition of protein kinase C, the MAP kinase pathway, and potentially of p21(Cip-1/MDA6/WAF1) expression, may alter the sensitivities of leukemic and carcinoma cells to cytotoxic insults, leading to increased apoptosis and loss of clonogenicity.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Ciclinas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteínas Quinasas Activadas por Mitógenos , Transducción de Señal/fisiología , Ciclo Celular/fisiología , División Celular/fisiología , Supervivencia Celular/fisiología , Supervivencia Celular/efectos de la radiación , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Activación Enzimática , Leucemia/tratamiento farmacológico , Leucemia/enzimología , Leucemia/patología , Leucemia/radioterapia , Proteína Quinasa 3 Activada por Mitógenos , Proteína Quinasa C/fisiología , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/efectos de la radiación
9.
Leukemia ; 10(7): 1150-8, 1996 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8683995

RESUMEN

Based upon earlier reports of synergism in cells of lymphoid origin, we have examined interactions between the organotellurium compound AS101 and the protein kinase C (PKC) activator bryostatin 1 with respect to differentiation and Ara-C-induced apoptosis in human myeloid leukemia cells (HL-60). Although preincubation with bryostatin 1 (10 nM) for 24 h significantly increased DNA fragmentation and apoptosis in cells subsequently treated with 10 microM Ara-C for 6 h, this effect was not enhanced by co-administration of AS101 (1.5 microM). However, while exposure of cells to AS101 or bryostatin 1 alone for 72 h was ineffective in inducing cellular maturation, combined treatment resulted in the induction of differentiated features in a subset of cells, manifested by an increase in cell adherence, CD11b expression, cytoplasmic granularity and cell spreading. In addition, cells exposed to the combination of AS101 and bryostatin 1, in contrast to cells incubated with these agents individually, displayed a significant decline in the S-phase and a corresponding increase in the G0/G1 cell populations. These events were accompanied by an increase in protein expression of the cyclin-dependent kinase inhibitor, p21 (WAF1/CIP1/MDA6), and a decline in expression of the c-myc protein. AS101 failed to increase intracellular free Ca2+ ([Ca2+]i) in HL-60 cells, or reverse the profound PKC down-regulation induced by bryostatin 1. Whereas treatment of cells with 1.5 microM AS101 or 10 nM bryostatin 1 for 24 h exerted minimal growth inhibitory effects, combined exposure to these agents reduced colony formation by over 70%. Finally, although addition of AS101 did not potentiate apoptosis induced by the bryostatin 1/Ara-C combination, it did lead to a further reduction in clonogenicity. Together, these findings demonstrate that AS101 partially restores the ability of bryostatin 1 to trigger a differentiation program in an otherwise unresponsive HL-60 cell line, possibly by facilitating bryostatin 1-mediated G1 arrest. They also indicate that AS101 potentiates the antiproliferative effects of bryostatin 1 administered alone or in combination with Ara-C through a mechanism other than, or in addition to, induction of apoptosis.


Asunto(s)
Antineoplásicos/farmacología , Etilenos/farmacología , Lactonas/farmacología , Leucemia Promielocítica Aguda/patología , Apoptosis/efectos de los fármacos , Western Blotting , Brioestatinas , Calcio/metabolismo , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Citarabina/farmacología , Daño del ADN , Sinergismo Farmacológico , Células HL-60/efectos de los fármacos , Células HL-60/metabolismo , Células HL-60/patología , Humanos , Leucemia Promielocítica Aguda/inmunología , Leucemia Promielocítica Aguda/metabolismo , Macrólidos , Antígeno de Macrófago-1/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ensayo de Tumor de Célula Madre
10.
Cell Calcium ; 8(3): 189-96, 1987 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-3111713

RESUMEN

The 7315a tumour secretes prolactin, but is refractory to enhancement of prolactin release by thyrotrophin-releasing hormone (TRH). In order to investigate further this refractoriness of the 7315a tumour cell, we compared cells from the tumour and from the normal pituitary with regard to TRH-enhanced fractional 45Ca2+ efflux and inositol phosphate production. TRH caused a large efflux of calcium from normal pituitary cells, but only mildly enhanced calcium efflux from the tumour cells. In contrast, TRH enhanced total inositol phosphate generation in both groups of cells to a similar degree. We therefore conclude that prolactin release from 7315a tumour cells is refractory to TRH due, at least in part, to impaired mobilisation of intracellular calcium by inositol phosphates.


Asunto(s)
Calcio/metabolismo , Neoplasias Hipofisarias/metabolismo , Prolactina/metabolismo , Animales , Femenino , Técnicas In Vitro , Fosfatos de Inositol/biosíntesis , Adenohipófisis/metabolismo , Ratas , Hormona Liberadora de Tirotropina
11.
Endocrinology ; 137(10): 4419-26, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8828503

RESUMEN

Interleukin-6 (IL-6) is a B-cell differentiation-inducing cytokine that affects the secretion of several neuroendocrine hormones. Normal rat anterior pituitary (AP) cells synthesize and release IL-6, suggesting a paracrine role for the stimulation of AP hormone release by this cytokine. We have previously reported that IL-1 beta enhances IL-6 release and phospholipase A2 (PLA2)-mediated hydrolysis of phosphatidylcholine (PC) in AP cells. Because lysophosphatidylcholine (LPC) may function as a second messenger for IL-1 beta, we have investigated the effects of exogenous LPC on IL-6 release from AP cells in vitro. AP cells from male Long-Evans rats were dispersed and cultured for 5-6 days in 96-well (100,000 cells/well) culture plates. Cells were rinsed and incubated in the absence or presence of 1.25-40 microM LPC 18:0 (stearoyl) for 6 h, and IL-6 concentrations determined using the 7-TD1 cell bioassay. LPC 18:0 significantly (P < 0.01) stimulated IL-6 release up to 10-fold in a concentration-related manner. In contrast, LPC 18:0 did not affect PRL release. LPC species substituted with progressively shorter saturated 1-acyl chains (16:0-10:0) were less effective for IL-6 induction. Examination of structurally related glycerophospholipid species revealed the specificity of the LPC stimulation of IL-6 release. Thus, 1.25-40 microM lysophosphatidylethanolamine (LPE; 18:0) and lysophosphatidic acid (LPA; 18:0) were without significant effect on AP IL-6 release, demonstrating the specific functional requirement for the phosphorylcholine headgroup. Hydrolysis of the structurally related choline-linked phospholipid sphingomyelin (SM) has been implicated in IL-1 beta action in certain cell types. Similarly, 1.25-20 microM lysosphingomyelin (sphingosylphosphorylcholine; SPC) also significantly (P < or = 0.01) stimulated IL-6 release from AP cells, although SPC exhibited discernibly lower potency and efficacy than LPC. An acyl analog of platelet-activation factor (PAF), i.e. 18:0-2:0 PC (1-stearoyl-2-acetoyl-sn-glycero-3-phosphorylcholine), differs from LPC by an acetyl group in the sn-2 position; PAF was at least as effective as LPC for the stimulation of IL-6 release from AP cells in vitro. Stimulation of IL-6 release by LPC 18:0 was completely suppressed by pharmacological inhibitors of protein kinase C such as H7 (20 microM) and chelerythrine (5 microM). In addition, H7 (20 microM) abolished the stimulation of IL-6 release by IL-1 beta (0.16-100 ng/mL). These findings demonstrate that LPC, acyl PAF, or SPC (but not other lysophospholipids) stimulate IL-6 release from AP cells in vitro. We conclude that LPC-mediated activation of protein kinase C is involved in the stimulatory actions of IL-1 beta in AP cells.


Asunto(s)
Interleucina-6/metabolismo , Lisofosfatidilcolinas/farmacología , Adenohipófisis/metabolismo , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Alcaloides , Animales , Benzofenantridinas , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Interleucina-1/farmacología , Masculino , Fenantridinas/farmacología , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacología , Adenohipófisis/citología , Adenohipófisis/efectos de los fármacos , Factor de Activación Plaquetaria/farmacología , Prolactina/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Ratas Endogámicas , Esfingosina/análogos & derivados , Esfingosina/farmacología
12.
Endocrinology ; 123(6): 2793-9, 1988 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2904359

RESUMEN

We have examined the influences of dopamine and the D2 receptor agonist bromocriptine on phosphoinositide metabolism in primary cultures of rat anterior pituitary cells, monitoring changes in the levels of phosphatidylinositol (PtdIns), phosphatidylinositol-4-phosphate [PtdIns(4)P], and phosphatidylinositol-4,5-bisphosphate [PtdIns(4,5)P2]. Basal incorporation of [3H]inositol ([3H]Ins) into phosphoinositides was progressive, and radioisotopic equilibrium was attained in all three species within 48 h. The inclusion of dopamine or bromocriptine in the incubation medium promoted concentration-dependent reductions in the rate, but not the magnitude, of phosphoinositide radiolabeling. The onset of this effect was rapid; inhibition of [3H]Ins incorporation by dopamine (500 nM) and bromocriptine (100 nM) could be detected within 2 h. This treatment also produced a comparable reduction in the incorporation of [32P]orthophosphate into PtdIns(4,5)P2. In extended time-course studies, bromocriptine dramatically retarded the radiolabeling of PtdIns(4)P and PtdIns(4,5)P2, and apparent equilibria in these species were attained only after 96 h. We also assessed the ability of dopamine to modify the concentration-response characteristics of [3H]Ins-labeled inositol phosphate ([3H]InsPx) production by TRH, angiotensin II (AII), neurotensin (NTS), bombesin (BBS), and vasoactive intestinal polypeptide (VIP). Neither dopamine nor bromocriptine altered the rate or magnitude of TRH-, AII-, NTS-, or BBS-related InsPx generation. VIP was completely ineffective in stimulating InsPx generation. PRL release was significantly reduced in all dopamine-treated groups. That the InsPx concentration-response relationships for each of these peptides remained unimpaired by exposure to dopamine or bromocriptine extends our previous observation that the phosphoinositide-specific phospholipase-C is insensitive to dopaminergic tone. Consistent with our earlier findings, these data indicate that activation of the D2 dopamine receptor attenuates the activity of mechanisms associated with the serial phosphorylations of PtdIns and PtdIns(4)P, reactions that give rise to PtdIns(4)P and PtdIns(4,5)P2, respectively. It is our conclusion that dopamine, in addition to its other actions, attenuates the phosphorylation, rather than the hydrolysis, of anterior pituitary phosphoinositide. This attenuation appears to be mediated by an inhibitory coupling of the D2 receptor with the phosphoryltransferase activities that catalyze PtdIns(4)P and PtdIns(4,5)P2 formation.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Fosfatos de Fosfatidilinositol , Fosfotransferasas (Aceptor de Grupo Alcohol) , Fosfotransferasas/metabolismo , Adenohipófisis/enzimología , Receptores Dopaminérgicos/fisiología , 1-Fosfatidilinositol 4-Quinasa , Angiotensina II/farmacología , Animales , Bombesina/farmacología , Bromocriptina/farmacología , Dopamina/farmacología , Femenino , Hormona del Crecimiento/metabolismo , Inositol/metabolismo , Cinética , Neurotensina/farmacología , Fosfatidilinositol 4,5-Difosfato , Fosfatidilinositoles/metabolismo , Adenohipófisis/efectos de los fármacos , Prolactina/metabolismo , Ratas , Ratas Endogámicas , Receptores de Dopamina D2 , Somatostatina/farmacología , Hormona Liberadora de Tirotropina/farmacología
13.
Endocrinology ; 129(6): 2886-94, 1991 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1954875

RESUMEN

We have reported previously that a subpopulation(s) of anterior pituitary cells released IL-6 and that this release was stimulated by interleukin-1 (IL-1) through a non-cAMP-dependent mechanism. We now report that IL-1 induces IL-6 release from anterior pituitary cells in an eicosanoid-dependent manner. Dispersed rat anterior pituitary cells were briefly prelabeled (2-3 h) with [3H]arachidonic acid (AA) to esterify the fatty acid within the lipid pool. Incubation of these prelabeled cells with 25 ng/ml IL-1 beta caused an increase only within 1-2 min in the amount of free [3H]AA detected in the extracts of the cells. During 15- to 30-min incubations, IL-1 beta (25 ng/ml) caused an increased accumulation of [3H]AA in the incubation medium which reached levels similar to those induced by 100 nM TRH. Perifused anterior pituitary cells responded to IL-1 beta (25 ng/ml) with a rapid (less than 2 min), biphasic, and reversible efflux of [3H]AA. The [3H]AA appears to have been derived from choline phospholipids, as formation of [3H]glycerophosphorylcholine was substantially increased by exposure of [3H]choline-prelabeled cells to either IL-1 alpha (171%) or IL-1 beta (236%); in addition, the complete deacylation of phosphatidylcholine suggests that other fatty acid species are liberated as a consequence of IL-1 receptor activation and, thus, may also contribute to the actions of IL-1 alpha and IL-1 beta. However, the levels of [3H]phosphorylcholine and [3H]choline were unchanged as well as those of catabolites of other lipid species. These data suggested an involvement of phospholipase-A2 (PLA2) in mediating the IL-1 induction of IL-6 release. Subsequently, we used inhibitors of the PLA2, cyclooxygenase, and lipoxygenase enzymes to investigate a possible role for the generation of AA and its subsequent enzymatic conversion in the signal transduction pathway activated by IL-1. The PLA2 inhibitor aristolochic acid (10 microM) blocked IL-1 beta-induced IL-6 release and the release of IL-6 caused by Pyrularia pubera thionin (5 micrograms/ml), a stimulator of PLA2 activity. The cyclooxygenase inhibitor indomethacin (10 microM) did not inhibit IL-1 beta-induced IL-6 release. In contrast, the general lipoxygenase inhibitor nordihydroguaiaretic acid (10 microM) and the more specific 5-lipoxygenase inhibitors AA861 and RHC5901 (both 10 microM) reduced basal and blocked IL-1 beta-induced IL-6-release.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Ácidos Aristolóquicos , Eicosanoides/fisiología , Interleucina-1/farmacología , Interleucina-6/metabolismo , Adenohipófisis/metabolismo , Animales , Ácido Araquidónico/metabolismo , Benzoquinonas/farmacología , Células Cultivadas , Inhibidores de la Ciclooxigenasa/farmacología , Ácido Etacrínico/farmacología , Femenino , Glicerilfosforilcolina/metabolismo , Inhibidores de la Lipooxigenasa/farmacología , Masoprocol/farmacología , Compuestos Orgánicos , Fenantrenos/farmacología , Fosfolipasas A/antagonistas & inhibidores , Fosfolipasas A2 , Ratas , Ratas Endogámicas , Transducción de Señal
14.
Endocrinology ; 123(5): 2341-50, 1988 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-2844508

RESUMEN

Although dopamine inhibits PRL release from the normal anterior pituitary lactotroph, a conclusive demonstration of the mechanisms involved in this response has been impeded by the presence of other cell types in the anterior pituitary. To circumvent this problem, we have isolated a clonal cell line, designated MMQ, from the 7315a rat pituitary tumor. The MMQ cell is an exemplary model for our use because it only secretes PRL. Our studies show that dopamine inhibits secretagogue-induced PRL release from these cells. In addition, dopamine decreases the intracellular cAMP concentration in MMQ cells that have been exposed to forskolin, cholera toxin, or vasoactive intestinal polypeptide, each a stimulator of cAMP generation. This inhibition is, in turn, reversed by the dopamine antagonist haloperidol and by pertussis toxin, an inactivator of the GTP-binding coupling protein. Dopamine also decreases the uptake and fractional efflux of 45Ca2+ by MMQ cells that have been exposed to the calcium channel activator maitotoxin. It seems, therefore, that dopamine decreases PRL release from MMQ cells at least in part by decreasing intracellular cAMP levels and calcium uptake. In additional experiments, we have found that MMQ cells are responsive to somatostatin, estrogen, progesterone, and acetylcholine, but not to TRH, angiotensin II, neurotensin, or bombesin. Furthermore, these cells possess a functional protein kinase-C system, as evidenced by the increase in PRL release and decrease in stimulated intracellular cAMP levels that occur in response to treatment with phorbol diesters. We suggest that the MMQ cell line will prove a useful model system for study of the biochemical effects of dopamine and other factors that modify PRL release.


Asunto(s)
Dopamina/farmacología , Oxocinas , Neoplasias Hipofisarias/metabolismo , Prolactina/metabolismo , Calcio/metabolismo , Toxina del Cólera/farmacología , Colforsina/farmacología , AMP Cíclico/metabolismo , Activación Enzimática , Estradiol/farmacología , Haloperidol/farmacología , Inmunohistoquímica , Toxinas Marinas/farmacología , Toxina del Pertussis , Neoplasias Hipofisarias/ultraestructura , Proteína Quinasa C/metabolismo , Células Tumorales Cultivadas , Péptido Intestinal Vasoactivo/farmacología , Factores de Virulencia de Bordetella/farmacología
15.
Endocrinology ; 127(1): 191-9, 1990 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2163310

RESUMEN

Calcitonin is present in both the hypothalamus and pituitary of the rat, and normal rat anterior pituitary cells express calcitonin receptors. Calcitonin has been reported to inhibit or to stimulate PRL release from rat anterior pituitary cells. We have investigated the effects of salmon calcitonin on basal and stimulated PRL release from rat anterior pituitary cells and have studied the effects of this peptide on the intracellular biochemical pathways involved in PRL release. Salmon calcitonin had no significant effect on basal PRL release, but inhibited (P less than 0.01) TRH-stimulated PRL release without affecting PRL release promoted by angiotensin II, neurotensin, phorbol myristate acetate (a protein kinase C activator), or maitotoxin (a calcium channel activator). Salmon calcitonin had no effect on the increase in PRL release and intracellular cAMP concentration after exposure of pituitary cells to vasoactive intestinal peptide or forskolin. Salmon calcitonin significantly decreased (P less than 0.01) the TRH-stimulated rise in inositol phosphates without affecting the angiotensin II-stimulated increase in inositol phosphates. Similarly, salmon calcitonin decreased the TRH-stimulated increase in cytosolic calcium and arachidonate liberation by pituitary cells. We conclude that salmon calcitonin selectively decreases TRH-stimulated PRL release by a mechanism that involves a decrease in inositol phosphate production, as well as a subsequent reduction in cytosolic calcium levels and in arachidonate liberation.


Asunto(s)
Calcitonina/farmacología , Fosfatos de Inositol/metabolismo , Oxocinas , Adenohipófisis/metabolismo , Prolactina/metabolismo , Hormona Liberadora de Tirotropina/farmacología , Angiotensina II/farmacología , Animales , Ácido Araquidónico , Ácidos Araquidónicos/metabolismo , Calcio/metabolismo , Células Cultivadas , Colforsina/farmacología , AMP Cíclico/metabolismo , Citosol/metabolismo , Femenino , Toxinas Marinas/farmacología , Neurotensina/farmacología , Adenohipófisis/efectos de los fármacos , Ratas , Ratas Endogámicas , Acetato de Tetradecanoilforbol/farmacología , Péptido Intestinal Vasoactivo/farmacología
16.
Endocrinology ; 121(6): 2035-43, 1987 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2824178

RESUMEN

Thymosin fraction 5 (TF5) is a partially purified extract of bovine thymus containing 40-60 peptides. In addition to its well documented immunopotentiating effects, TF5 reportedly modulates the secretion of some hypothalamic peptides and pituitary hormones. In this study, TF5 (10-100 micrograms/ml) stimulated PRL release from normal, MtTW15, and 7315a cells and GH release from normal and MtTW15 cells, but had no apparent effect on LH release. No changes in intracellular cAMP or cGMP levels could be correlated with these responses. Stimulation of PRL release from perifused normal anterior pituitary cells was rapid, sustained, and concentration related. Although it had no apparent effect on normal prelabeled anterior pituitary cells with respect to 45Ca2+ efflux, the calcium channel blocker D-600 inhibited TF5-mediated hormone release from these cells. Additive increases in TRH-stimulated PRL release and GRF-stimulated GH release by TF5 suggested independent mechanisms of action. Dopamine (500 nM) blocked TF5-stimulated PRL release, but somatostatin (10-100 nM) had no effect on TF5-stimulated PRL or GH release. TF5 failed to affect either basal or TRH-induced polyphosphoinositide hydrolysis. Perifused normal anterior pituitary cells prelabeled with [3H]arachidonate responded to TF5 treatment with a liberation of radioactive arachidonate and/or its metabolites. BW755c, an inhibitor of all known catabolic pathways of arachidonic acid, blocked the ability of TF5 to stimulate PRL and GH release. Reversed phase HPLC separation of TF5 into five fractions resulted in two fractions that exhibited hormone-releasing activity. These data suggest that TF5 stimulates pituitary hormone release through a mechanism different from that ascribed to TRH or GRF. The stimulus-secretion coupling mechanism involves neither polyphosphoinositide hydrolysis nor cAMP generation, but appears to be dependent on the generation of arachidonate metabolites.


Asunto(s)
Hormona del Crecimiento/metabolismo , Adenohipófisis/metabolismo , Prolactina/metabolismo , Timosina/análogos & derivados , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Femenino , Fosfatos de Inositol/metabolismo , Cinética , Masculino , Adenohipófisis/efectos de los fármacos , Ratas , Ratas Endogámicas , Timosina/farmacología
17.
FEBS Lett ; 412(1): 9-14, 1997 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-9257680

RESUMEN

The role of protein kinase C (PKC) and p42(MAP kinase) signaling in the regulation of proliferation and apoptosis was investigated in freshly isolated and primary cultured rat hepatocytes. Acute treatment of freshly isolated hepatocytes with phenylephrine and EGF caused rapid phasic activations of p42(MAP kinase) and JNK1. Acute pre-treatment of hepatocytes with the PKC inhibitors sphingosine, chelerythrine and bis-indolylmaleimide abolished the ability of phenylephrine, but not EGF, to activate p42(MAP kinase) and JNK1. Acute pretreatments with all of the PKC inhibitors alone increased JNK1 basal activity approximately 2-fold. Acute treatments of primary cultures of hepatocytes with an inhibitor of MEK1 activation (PD98059) also caused inhibition of p42(MAP kinase) and a approximately 2-fold activation of JNK1. These data demonstrate that PKC can function as both a proximal activator and a distal inhibitor of signaling through the JNK1/SAP kinase pathway. Treatments (4 h) of primary cultured hepatocytes with sphingosine, chelerythrine, bis-indolylmaleimide and PD98059 did not induce apoptosis as judged by propidium iodide staining. Similar acute treatments of HepG2 cells rapidly induced cell death. These data demonstrate that acute inhibition of either PKC or p42(MAP kinase) function is sufficient to rapidly induce apoptosis in transformed, but not in non-transformed hepatocytes.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Hígado/enzimología , Quinasas de Proteína Quinasa Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos , Proteína Quinasa C/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Animales , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos , Cinética , MAP Quinasa Quinasa 1 , Masculino , Proteína Quinasa 1 Activada por Mitógenos , Fenilefrina/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley
18.
J Endocrinol ; 110(3): 389-93, 1986 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-3020141

RESUMEN

The hydrolysis of membrane phosphatidylinositol to yield [3H]labelled inositol phosphates by anterior pituitary cells was stimulated significantly by angiotensin II, TRH and neurotensin over a broad range of concentrations. These secretagogues also stimulated release of prolactin. Although the coincident incubation of dopamine with these agents resulted in a marked diminution of prolactin release, no concomitant reduction in inositol phosphate production was observed. In addition, bromocriptine, a potent agonist of dopamine, also proved ineffective in blunting stimulated phosphatidylinositol catabolism. Although it slightly inhibited basal rates of inositol tris-, bis- and monophosphate production, these results show that the secretagogue-mediated enhancement of phosphatidylinositol catabolism may be correlated with an increased release of prolactin and that the inhibition of hormone release produced by dopamine is not achieved by reducing basal or secretagogue-mediated inositol phosphate production.


Asunto(s)
Dopamina/farmacología , Fosfatidilinositoles/metabolismo , Adenohipófisis/metabolismo , Angiotensina II/farmacología , Animales , Bromocriptina/farmacología , Membrana Celular/metabolismo , Células Cultivadas , Femenino , Hidrólisis , Fosfatos de Inositol/metabolismo , Neurotensina/farmacología , Adenohipófisis/citología , Adenohipófisis/efectos de los fármacos , Prolactina/metabolismo , Ratas , Ratas Endogámicas , Hormona Liberadora de Tirotropina/farmacología
19.
Biochem Pharmacol ; 54(5): 563-73, 1997 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-9337072

RESUMEN

The role of the nuclear phosphoprotein c-Myc has been examined with respect to the regulation of 1-beta-D-arabinofuranosylcytosine (ara-C)-induced apoptosis in human leukemia cells exposed to bryostatin 1 and other pharmacologic protein kinase C (PKC) activators. Pretreatment of HL-60 cells for 24 hr with 10 nM bryostatin 1 significantly potentiated the ability of ara-C (10 microM; 6 hr) to induce apoptosis without reducing the expression of c-Myc protein. In contrast, equivalent exposure to the stage 2 tumor-promoting PKC activator mezerein (10 nM) in conjunction with ara-C reduced c-Myc levels by 87% and failed to potentiate apoptosis. Co-administration of bryostatin 1 with mezerein before ara-C prevented down-regulation of c-Myc and augmented cell death, whereas co-treatment with the calcium ionophore A23187 (250 nM) and bryostatin 1 reduced c-Myc levels by 80% and abrogated the increase in ara-C-induced apoptosis. When cells were exposed for 24 hr to a c-myc antisense oligonucleotide (AS-ODN;10 microM) but not to a scrambled sequence ODN (SS-ODN) prior to ara-C, c-Myc expression was reduced by 81%, and apoptosis and cell viability were unperturbed. However, AS-ODN (but not SS-ODN) reduced c-Myc protein in cells pre-exposed to bryostatin 1 by 74% and abrogated potentiation of ara-C-induced apoptosis. The actions of c-myc AS-ODN did not stem from proximal G1 arrest/differentiation or biochemical events, since they were not associated with a reduction in the S-phase cell fraction, p21(WAF1/CIP1) induction, pRb hypophosphorylation, or alterations in ara-C metabolism. Together, these findings indicate that HL-60 cell apoptosis proceeds by both c-Myc-dependent and -independent pathways, and that only the former are involved in the potentiation of ara-C-mediated cell death by bryostatin 1.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Citarabina/farmacología , Diterpenos , Células HL-60/efectos de los fármacos , Lactonas/farmacología , Oligonucleótidos Antisentido/farmacología , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Antineoplásicos/farmacología , Brioestatinas , Fragmentación del ADN , Sinergismo Farmacológico , Activación Enzimática , Humanos , Cinética , Macrólidos , Forbol 12,13-Dibutirato/farmacología , Proteínas Proto-Oncogénicas c-myc/genética , Terpenos/farmacología , Acetato de Tetradecanoilforbol/farmacología , Tionucleótidos
20.
Biochem Pharmacol ; 47(5): 839-52, 1994 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-8135859

RESUMEN

We have demonstrated previously that bryostatin 1, a macrocylic lactone with putative protein kinase C (PKC)-activating properties, synergistically augments the antileukemic actions of the deoxycytidine analog 1-[beta-D-arabinofuranosyl]cytosine (ara-C) in HL-60 human promyelocytic leukemia cells (Grant et al., Biochem Pharmacol 42: 853-867, 1991), and that this effect appears to be related to sensitization to ara-C-induced apoptosis (Grant et al., Cancer Res 52: 6270-6278, 1992). In the present studies, we have assessed the extent of this damage by quantitative spectrofluorophotometry of small molecular weight, double-stranded DNA fragments in order to provide: (a) a more complete characterization of the interaction between ara-C and bryostatin 1, and (b) a direct comparison of the relative effects of bryostatin 1 treatment with other pharmacological manipulations known to modulate protein kinase C activity. Exposure of cells to ara-C (10(-9) to 10(-4) M; 1-24 hr) induced time- and concentration-related increases in the extent of DNA fragmentation. Treatment with bryostatin 1 (10(-11) to 10(-7) M; 1-24 hr) alone failed to induce DNA damage, but promoted substantial time- and concentration-related increases in the extent of fragmentation induced by a subsequent 6-hr exposure to ara-C. Maximal potentiation of fragmentation (e.g. 2- to 3-fold greater than that obtained with ara-C alone) was observed following a 24-hr pretreatment with 10(-8) M or 10(-7) M bryostatin 1, and correlated closely with enhanced inhibition of HL-60 cell clonogenicity. The stage-1 tumor-promoter phorbol dibutyrate potentiated the effects of ara-C in a biphasic manner, maximally augmenting the response at 2.5 x 10(-8) M, but exerting no effect at 10(-7) M, whereas the stage-2 tumor-promoter mezerein failed to augment ara-C-related DNA fragmentation at low concentrations, and antagonized ara-C action at high concentrations. In contrast, ara-C-related DNA fragmentation was attenuated or abolished either by continual preexposure to synthetic diglyceride or by pretreatment with exogenous phospholipase C at all concentrations tested. Increased DNA fragmentation was not specifically related to recruitment of cells into S-phase or enhancement of ara-C-related cellular differentiation. Finally, concentrations of bryostatin 1 that maximally potentiated ara-C-related DNA fragmentation were associated with virtually complete down-regulation of total cellular PKC activity, whereas diglyceride and phospholipase C, which suppressed the response to ara-C, moderately increased total PKC activity.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , Lactonas/farmacología , Leucemia Mieloide/genética , Proteína Quinasa C/efectos de los fármacos , Antineoplásicos/administración & dosificación , Apoptosis/genética , Brioestatinas , Citarabina/administración & dosificación , Citarabina/farmacología , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos , Lactonas/administración & dosificación , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/patología , Macrólidos , Proteína Quinasa C/análisis , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda