Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell Biol ; 15(11): 6398-405, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7565792

RESUMEN

Human pro-tumor necrosis factor (pro-TNF) is a type II transmembrane protein with a highly conserved 76-residue leader sequence. We have analyzed the behavior, both in a microsomal translocational system and by transfection, of a series of mutants with deletions from the cytoplasmic, transmembrane, and linking domains. Cytoplasmic deletions included the Arg doublet at -49 and -48 and/or the Lys doublet at -58 and -57; additional mutants included deletion of residues -73 to -55 and -73 to -55, -49, and -48. The transmembrane and linking domain mutants included deletions in the -42 to -35 region, combined with the deletion of residues -32 to -1. Two hybrid mutants combined the cytoplasmic deletions with the deletion of residues -32 to -1. All of the cytoplasmic deletion mutants were properly translocated, as were the transmembrane deletion mutants with deletions up to residues -36, -35, -32 to -1, although the last one exhibited reduced efficiency; further incremental deletions, including deletions of residues -38 to -35 and -32 to -1, completely blocked translocation. Both hybrid mutants were effectively translocated; furthermore, transfection analysis revealed competent expression and maturation of both the cytoplasmic and hybrid mutants. Thus, proper expression and maturation of human pro-TNF can be accomplished with as few as approximately 12 of the 26 residues of the native transmembrane domain and with a net negative charge in the cytoplasmic domain flanking the transmembrane region.


Asunto(s)
Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Señales de Clasificación de Proteína/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico , Membrana Celular/metabolismo , Sistema Libre de Células , Células Cultivadas , Chlorocebus aethiops , Perros , Retículo Endoplásmico Rugoso/metabolismo , Humanos , Iones , Datos de Secuencia Molecular , ARN Mensajero/genética , Eliminación de Secuencia , Relación Estructura-Actividad
2.
J Natl Cancer Inst ; 69(4): 889-94, 1982 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-6750198

RESUMEN

Thioglycollate-elicited C57BL/6 peritoneal exudate macrophage monolayers (PEMM) stimulated with poly I . poly C or LPS released a macrophage cytotoxin (MCT) that rapidly bound to syngeneic (EL 4) or allogeneic (NS-1, YAC-1) tumor cells but did not bind to normal splenocytes. No binding to human (K562) tumor cells was observed. PEMM stimulated with poly I . poly C destroyed allogeneic tumor cells (NS-1) when separated by cell-impermeable Millipore filters in vitro; in contrast, PEMM not stimulated with poly I . poly C were incapable of lysing targets when separated by membranes. The reversible inhibitors N alpha-p-tosyl-L-arginine methyl ester and soybean trypsin inhibitor and the irreversible inhibitors N alpha-p-tosyl-L-lysine chloromethyl ketone and phenylmethylsulfonyl fluoride, of trypsin-like proteases, significantly or totally inhibited MCT cell-lytic activity for L-929 cells in vitro. Furthermore, modification of MCT-associated arginine residues by 1,2-cyclohexanedione completely blocked lytic activity. MCT was concluded to be an inducible nonspecific cell-lytic effector molecule elaborated by activated macrophages, which could bind to potential target cells, and was itself or was associated with a protease.


Asunto(s)
Citotoxinas/biosíntesis , Activación de Macrófagos , Macrófagos/efectos de los fármacos , Neoplasias Experimentales/inmunología , Péptido Hidrolasas/inmunología , Animales , Línea Celular , Citotoxinas/aislamiento & purificación , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos , Neoplasias Experimentales/enzimología , Péptido Hidrolasas/metabolismo , Poli I-C/farmacología , Inhibidores de Proteasas , Unión Proteica , Bazo/inmunología
3.
J Natl Cancer Inst ; 74(1): 1-9, 1985 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-3918210

RESUMEN

A cloned subline of the human monocytic leukemia cell line, THP-1, was induced to produce high levels of cytotoxic activity following an 18-hour phorbol myristate acetate (CAS: 16561-29-8) stimulation in vitro. This activity, termed monocyte cell line cytotoxin(s) (MCCT), was tested in vitro on different human continuous cell lines (Chang, ESH-7, GM3104A, HeLa, HT-1080, K562, Mel, T-24) and on primary human fibroblasts (GM3468, Manz). The continuous cell lines exhibited a spectrum of sensitivity to MCCT-containing supernatants whereas the primary fibroblasts were resistant to lysis. Enzymatic degradation and heat denaturation studies indicate that MCCT is a protein. Its bioactivity can be resolved into three lytic peaks after molecular sieving on Ultrogel AcA 44. The major peak, designated alpha MCCT, eluted with a molecular weight of 100,000-140,000 daltons. A minor peak, beta MCCT, was seen at 60,000-80,000 daltons, and a third, unstable minor peak, gamma MCCT, eluted at less than 10,000 daltons. The alpha-lytic peak was examined further and was found to migrate as a single peak in 7% native polyacrylamide gel electrolysis tube gels with an rf of 0.25-0.30. None of the MCCT forms were immunologically cross-reactive with human alpha-lymphotoxin. Various protease inhibitors known to inhibit monokine- and macrophage-mediated direct cell lysis in vitro were tested for their inhibitory effects on alpha MCCT activity. The irreversible binding inhibitor N alpha-p-tosyl-L-lysyl chloromethyl ketone inhibited the biologic activity of alpha MCCT. The reversible binding inhibitors N alpha-p-tosyl-L-arginine methyl ester and soybean trypsin inhibitor were able to block in vitro lytic activity when added to alpha MCCT in the presence of the target cell. In contrast, the inhibitors phenylmethylsulfonyl fluoride, L-1-tosylamide, 2-phenylethyl chloromethyl ketone, and N alpha-acetyl-L-lysine methyl ester were not effective in blocking cytolysis. Finally, the hydrogen peroxide scavenger catalase inhibited alpha MCCT lytic activity in vitro; however, the hypochlorous acid scavengers alanine, serine, and valine were without effect.


Asunto(s)
Citotoxicidad Inmunológica , Leucemia Mieloide/inmunología , Proteínas/aislamiento & purificación , Alanina/farmacología , Catalasa/farmacología , Línea Celular , Clonación Molecular , Electroforesis en Gel de Poliacrilamida , Humanos , Cinética , Peso Molecular , Monocitos/análisis , Monocitos/inmunología , Monocinas , Inhibidores de Proteasas/farmacología , Serina/farmacología , Valina/farmacología
4.
J Natl Cancer Inst ; 70(4): 717-24, 1983 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-6339778

RESUMEN

The cytotoxic effects of activated peritoneal macrophages and the J774 reticulum cell sarcoma cell line on B16 melanoma cells of differing metastatic potential were investigated in vitro. The melanoma target cells were sublines of low (B16-F1) or high (B16-F10) lung colonization potential as well as a subline of high (B16-B14b) brain colonization ability. Thioglycolate-elicited peritoneal macrophages from syngeneic C57BL/6 mice and J774 cells were activated in vitro with polyinosinic-polycytidylic acid (poly I:C) and used as effector cells. Macrophage-mediated cytolysis was determined by means of 24- to 72-hour radioactivity release assays with [125I]5-iodo-2'-deoxyuridine-labeled melanoma cells; the results indicated that the more metastatic sublines B16-F10 and B16-B14b were less susceptible to cytolysis by activated macrophages and J774 cells than was the poorly metastatic B16-F1 subline. The poly I:C-activated effector cells also released soluble cytotoxin(s), which resulted in melanoma cell lysis and growth inhibition. Cytotoxin-mediated melanoma cell cytolysis was determined by counting the number of viable mitomycin-treated target cells remaining after a 40-hour incubation period in the absence or presence of various concentrations of cell-released factors, and cytotoxin-mediated cytostasis was performed with the use of the same procedures without mitomycin pretreatment of targets. The factors released from both activated macrophages and J774 cells were more effective against the poorly metastatic B16-F1 cells than against the highly metastatic B16-F10 or B16-F14b cells. In addition, the activity of the factors from both activated effector cells was inhibited by fetal bovine serum. The J774 cells and the activated peritoneal macrophages demonstrated similar activities against B16 melanoma variants, indicating that the J774 cell line may be suitable as a model for the study of macrophage cytotoxicity. The results suggested that the potential of the highly metastatic melanoma cells to implant, survive, and grow at secondary sites may be due, in part, to their ability to circumvent host antitumor mechanisms.


Asunto(s)
Linfoma de Células B Grandes Difuso/fisiopatología , Macrófagos/fisiología , Melanoma/fisiopatología , Animales , Línea Celular , Supervivencia Celular , Femenino , Variación Genética , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Neoplasias Experimentales/fisiopatología
5.
J Natl Cancer Inst ; 69(4): 879-87, 1982 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-6956764

RESUMEN

Peritoneal exudate macrophage monolayers (PEMM) from C57BL/6 and DBA/2 mice inoculated ip with tumor allografts were induced to release in vitro labile cell toxin(s), herein called "macrophage cytotoxin(s)" (MCT). Macrophages released MCT spontaneously for a short interval when initially established as monolayers, and they were reinduced to secrete MCT by exposure to allogeneic and syngeneic tumor cells (but not to normal cells) and by exposure to polyinosinic-polycytidylic acid (poly I . poly C) and lipopolysaccharide (LPS). PEMM from normal mice treated ip 3 days previously with thioglycollate were also induced to release toxins in vitro. These cells did not release MCT spontaneously before or after treatment with neoplastic cells but were induced to release MCT by exposure to poly I . poly C or LPS. Resident peritoneal macrophages did not release MCT either spontaneously or after treatment with tumor cells, poly I . poly C, or LPS. MCT released from alloimmune mice stimulated with syngeneic or allogeneic tumor cells were resolved by molecular sieving into a major peak at 140,000--160,000 daltons, called "alpha-MCT," and into a minor peak at 60,000 daltons, called "beta-MCT." However, supernatants from thioglycollate-induced PEMM, stimulated with poly I . poly C or LPS, appeared to be composed entirely of the alpha-class. alpha-MCT from poly I . poly C-stimulated PEMM caused 31--56% lysis of syngeneic EL-4 and allogeneic L-929, NS-1, and YAC-1 tumor cells in vitro but was not cytotoxic for normal cells. Secretion of the MCT by PEMM derived from thioglycollate-treated animals stimulated with poly I . poly C was inhibited by colchicine, emetine, iodoacetic acid, trypan blue, and cytochalasin B.


Asunto(s)
Citotoxinas/biosíntesis , Activación de Macrófagos , Macrófagos/inmunología , Animales , Línea Celular , Cromatografía en Gel , Colchicina/farmacología , Citocalasina B/farmacología , Citotoxinas/análisis , Yodoacetatos/farmacología , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos , Trasplante de Neoplasias , Neoplasias Experimentales/inmunología , Peritoneo/inmunología , Poli I-C/farmacología , Factores de Tiempo
6.
Cancer Res ; 51(13): 3362-6, 1991 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-2054776

RESUMEN

The liposomal delivery of recombinant human tumor necrosis factor (rHuTNF) has been shown to be effective in reducing its toxic effects and in its targeting of organs rich in cells of the reticuloendothelial system. However, native recombinant human TNF shows only poor affinity for liposomes, presumably due to its low hydrophobicity. In an attempt to increase the efficiency of association with liposomes, we have modified TNF to increase its hydrophobicity by selective substitution of its amino groups with fatty acid side chains. N-hydroxysuccinimide esters of saturated fatty acids ranging from C8 to C18 were reacted with recombinant human TNF. Modification with esters of C8 to C14 acids occurred as determined by consumption of positively charged amino groups monitored by native polyacrylamide gel electrophoresis; however, esters of longer chain lengths (C16, C18) were much less capable of introducing these chains via amide linkages, and thus these adducts were not further characterized. Biological assays revealed that retention of activity was inversely dependent both on the number of chains introduced and on their length; activity was most conserved (greater than 50%) in a TNF preparation modified with only approximately 1-2.5 caprylic acid (C8) residues/trimer. This preparation was found to bind with high efficiency (approximately 50%) to preformed dipalmitoylphosphatidylcholine-small unilamellar vesicles. The extent of binding closely paralleled both the number of chains introduced and their length; binding was even more efficient (80-90%) for TNF modified either with approximately 3.5 caprylic acid residues/trimer or with approximately 1.5 residues of myristic acid (C14). However, the biological activity of these acylated TNFs was further reduced by this more extensive chemical modification (less than 50% activity for C8 and less than 10% for C14). The biological activity of dipalmitoylphosphatidylcholine-small unilamellar vesicle-C8-TNF was found to be comparable to that of the nonliposomal C8-TNF. Thus, biologically active preparations of liposomal-lipophilic TNF can be prepared with high efficiency.


Asunto(s)
Factor de Necrosis Tumoral alfa/química , 1,2-Dipalmitoilfosfatidilcolina/química , Acilación , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Células L , Liposomas , Ratones , Proteínas Recombinantes , Solubilidad , Relación Estructura-Actividad , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor de Necrosis Tumoral alfa/farmacología
7.
Cancer Res ; 49(22): 6252-7, 1989 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-2680061

RESUMEN

Hyperthermia modulated the cytotoxic activities of murine macrophages cocultured with EMT-6 tumor cells, altered the production of the monokines RIF (respiratory inhibition factor) and FeRF (iron-releasing factor) by these effector cells, and perturbed the activities of these monokines against EMT-6 cells. Cytotoxic activities of heated murine macrophages activated by Bacillus Calmette-Guérin were inhibited by heat doses of 40.5 degrees C for greater than or equal to 1 h if heating preceded triggering by the endotoxin lipopolysaccharide; however, cytotoxic activities were better retained if triggering preceded heating by 2 h. Treatment-sequence dependencies were also found in the secretion of some monokines that participate in these macrophage cytotoxic effects. Secretion of both RIF and FeRF by macrophages was slightly augmented or at least better retained if triggering of macrophages occurred several hours before heating for 1 h at 40.5-43 degrees C or for 24 h at 39-40.5 degrees C. If heating was nearly simultaneous with triggering or preceded it by several hours, there was a dose-dependent decrease in monokine secretion. The sensitivity of tumor cell targets to these monokines was also modified by the treatment sequence. When EMT-6 cells were treated with RIF- or FeRF-containing macrophage conditioned supernatants 1 h before heating at 40.5-43 degrees C, the RIF-treated cells became sensitized and the FeRF-treated cells retained better cytotoxic response than if cells had been treated 1 h after heating. Chronic heating for 24 h at 39-40.5 degrees C showed less dependence of tumor cell response on treatment sequence. Similar observations have recently been made in our laboratory for the interaction of tumor necrosis factor cytotoxic pathways with hyperthermia (Klostergaard et al., J. Biol. Response Modif., in press, 1989; Tomasovic et al., Int. J. Hyperthermia, in press, 1989). Those results and the present observations with the monokines RIF and FeRF support our hypothesis that the cytotoxic actions of macrophages and the cytotoxicity of endogenously added monokines can be augmented by appropriately constructed sequences combining hyperthermia with either macrophage priming/triggering or monokine treatment of tumor cells.


Asunto(s)
Calor , Activación de Macrófagos , Macrófagos/inmunología , Monocinas/metabolismo , Adenocarcinoma , Animales , Línea Celular , Supervivencia Celular , Células Cultivadas , Cromo/metabolismo , Femenino , Hierro/metabolismo , Cinética , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Masculino , Neoplasias Mamarias Experimentales , Ratones , Ratones Endogámicos , Monocinas/inmunología , Mycobacterium bovis/inmunología
8.
Cancer Res ; 46(2): 662-9, 1986 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-3079667

RESUMEN

Human peripheral blood monocytes, isolated in high purity by centrifugal counterflow elutriation from normal donors, were stimulated in vitro to release cell toxins, herein termed human monocyte toxin(s) (HMT). Bacterial lipopolysaccharide, the lipophilic 6-O-stearoyl derivative of muramyl dipeptide, and 4 beta-phorbol-12 beta-myristate-13 alpha-acetate served as effective induction signals. Induction involved a sequence of transcription, translation, and secretion, all necessary for HMT synthesis and release into the supernatant as determined by blocking of these functions with the drugs actinomycin D, cycloheximide, and monensin, respectively; HMT levels reached a peak within 4-6 h and thereafter declined. The levels of HMT produced varied considerably from donor to donor; one parameter causing this variability appeared to be the plateletapheresis history of the donor. Monocytes from donors subjected to pheresis for the first time were responsive to induction signals immediately after adherence and could not be brought to a higher state of priming for HMT production by further in vitro culture for up to 9 days, with or without recombinant human gamma-interferon. In contrast, monocytes from donors who had recently undergone pheresis (up to 1 wk earlier) were poorly responsive initially to triggering with lipopolysaccharide; however, these cells could be brought to a highly primed state for HMT production by a combination of culture in vitro for several days and a subsequent 24-h exposure to recombinant gamma-interferon (0.1-1.0 units/ml). These primed cells could then be effectively triggered by lipopolysaccharide to release HMT. HMT was found to be cytotoxic (cytostatic/cytolytic) for human and murine tumor cells in vitro.


Asunto(s)
Citotoxicidad Inmunológica , Citotoxinas/biosíntesis , Monocitos/inmunología , Animales , Adhesión Celular , Separación Celular , Citotoxinas/genética , Humanos , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Ratones , Biosíntesis de Proteínas , Transcripción Genética
9.
Cancer Res ; 47(8): 2014-9, 1987 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-3828989

RESUMEN

Murine Bacillus Calmette-Guérin-activated macrophages mediate discrete cytotoxic effects in cocultured tumor target cells in vitro. These effects include: the loss of intracellular iron, in part associated with reversible inhibition of the Kreb's cycle enzyme, aconitase; cytostasis, associated with reversible lesions inflicted in the electron transport chain (ETC) of the mitochondria resulting in reversible loss of proliferative capacity; and cytolysis, manifested by eventual gross perturbation of the integrity of the plasma membrane. We demonstrate that these manifestations of cytotoxicity are the result of three independent mechanisms employing apparently distinct macromolecules for their commission. Analysis of target cells that are highly susceptible (L-929), highly resistant (L-1210), or have incomplete resistance (EMT-6) to the cytolytic effects of cocultured activated macrophages indicates that there is no consistent relationship between the release of intracellular 59Fe and 51Cr. Thus, perturbation of intracellular iron pools did not appear to be an obligatory step on the pathway to cytolysis. Further evidence for this dissociation was obtained by employing a specific heteroantiserum reactive with cytolytic molecule(s). This antiserum could block the cytolytic response (51Cr release of cocultured L-929 and EMT-6 targets) but had no effect on the extent of iron release from viable EMT-6 or L-1210 targets. Furthermore, the cytolytic factor itself was incapable of mediating effects on the ETC or in causing release of intracellular iron. Two lines of evidence suggested that effects on the ETC are not linked with loss of intracellular iron. First, the monokine respiration inhibitory factor was incapable of causing release of intracellular iron from target cells in which the mitochondria were strongly suppressed. Second, the kinetics of release of respiration inhibitory factor from endotoxin-triggered Bacillus Calmette-Guérin-activated macrophages indicate a retarded appearance compared to the time at which a factor mediating release of intracellular iron was detectable. Our results strongly suggest that these three distinct cytotoxic reactions are under differential control by the effector cell.


Asunto(s)
Vacuna BCG/farmacología , Citotoxicidad Inmunológica , Hierro/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Mitocondrias/metabolismo , Neoplasias Experimentales/inmunología , Consumo de Oxígeno , Animales , Cromo/metabolismo , Endotoxinas/farmacología , Masculino , Ratones , Ratones Endogámicos
10.
Cancer Res ; 52(19): 5271-7, 1992 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-1394131

RESUMEN

We have investigated the cytotoxic responses in vitro of three human colon tumor cell lines with epithelial-like morphology, DLD-1, HCT-15, and HT-29, to thermochemoimmunotherapy with hyperthermia (42 degrees C for 2 h), carboplatin, and recombinant human tumor necrosis factor (TNF). Dose ranges of carboplatin and recombinant human TNF were administered essentially simultaneously and were followed 1 h later by hyperthermia. A two-tiered approach was used to evaluate cytotoxicity. In the first tier, a 5-day microcytotoxicity assay using vital dye staining was done; the effect on surviving fraction of simultaneously varying carboplatin and recombinant human TNF doses was evaluated by response surface methodology. From this analysis doses were selected for use in the second-tier clonogenic survival assays. A similar treatment protocol was used in clonogenic assays. Both assays revealed significant interline treatment response heterogeneity. Only the HCT-15 cells were sensitive to TNF alone; carboplatin activity against all three tumor cell lines was enhanced by TNF. Hyperthermia had minimal effect as a sole agent but enhanced the effects of carboplatin and TNF in DLD-1 and HCT-15 cells. Triple modality treatment resulted in 3-4-log decreased survival and could reduce cytotoxic resistance expressed against single- or dual-modality treatments by some of these cells.


Asunto(s)
Adenocarcinoma/terapia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Cisplatino/farmacología , Neoplasias del Colon/terapia , Hipertermia Inducida , Factor de Necrosis Tumoral alfa/farmacología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/microbiología , Cisplatino/administración & dosificación , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/microbiología , Terapia Combinada , Humanos , Factores Inmunológicos/farmacología , Inmunoterapia/métodos , Rojo Neutro/farmacocinética , Fenotipo , Sensibilidad y Especificidad , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/administración & dosificación , Ensayo de Tumor de Célula Madre
11.
Cancer Res ; 46(6): 2871-5, 1986 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-3698012

RESUMEN

Human peripheral blood monocytes, isolated in high purity by centrifugal counterflow elutriation from normal donors, release cell toxins, herein termed human monocyte toxins (HMTs) upon further stimulation in vitro. The principal form of HMTs produced by these human peripheral blood monocytes has been subjected to biochemical, functional, and serological characterization. By molecular sieving on Sephacryl S-200, HMTs can be resolved into two molecular weight classes. The larger, termed alpha, has a molecular weight of about 120,000, and the smaller, termed beta, has a molecular weight of about 65,000. The beta class is by far the most predominant species and has been further characterized. Chromatofocusing of beta-HMT indicates a slightly acidic nature, since this species is eluted at pH 5.8. Functional characterization of beta-HMT suggests that it is not a trypsin-like protease, since neither alpha,N-tosyl-L-lysylchloromethylketone nor alpha,N-tosyl-L-arginyl methyl ester are capable of causing significant inhibition of the cell-lytic activity of the molecule. Furthermore, cell lysis induced by beta-HMT appears to be independent of oxygen-dependent mechanisms, since catalase is incapable of blocking lysis, and since hydrogen peroxide and superoxide anion are not produced in detectable amounts during lysis. Finally, beta-HMT does not appear to be an arginase, since it is active in arginine-containing medium and further addition of arginine to the assay medium does not inhibit lysis significantly. beta-HMT is serologically related to recombinant human tumor necrosis factor (rHuTNF), since its cell lytic activity can be blocked by a rabbit antiserum against rHuTNF. However, much higher levels of this antiserum are required to achieve neutralization than are required to neutralize a comparable number of cell lytic units of rHuTNF. Furthermore, the results of preliminary immunoprecipitation experiments using the rabbit anti-rHuTNF antiserum suggest that a peptide in the Mr 60,000-70,000 range is recognized by this serum, whereas no signal at Mr 17,000 corresponding to rHuTNF is detectable. Thus, human peripheral blood monocytes can be triggered to release cell toxins, the principal form of which, beta-HMT, appears to be functionally distinct from the cytotoxic proteases reported in the murine system and appears to be molecularly distinct from, but serologically related to rHuTNF.


Asunto(s)
Citotoxinas/aislamiento & purificación , Monocitos/análisis , Neoplasias/inmunología , Cromatografía en Gel , Citotoxicidad Inmunológica , Citotoxinas/análisis , Citotoxinas/inmunología , Glicoproteínas/inmunología , Humanos , Sueros Inmunes/inmunología , Técnicas In Vitro , Peso Molecular , Monocitos/inmunología , Factor de Necrosis Tumoral alfa
12.
Cancer Res ; 52(15): 4096-101, 1992 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-1638521

RESUMEN

This study examined the effect of a trimodality therapy of the combination of recombinant human tumor necrosis factor alpha (TNF), whole-body hypertheria (WBH), and cis-diamminedichloroplatinum(II) (CDDP) or cis-diammine-1,1-cyclobutane dicarboxylate platinum(II) (CBDCA) on a fibrosarcoma and normal tissue in F344 rats. TNF (1 x 10(5) units/kg) increased the antitumor effect of both CDDP (1.5 mg/kg) + WBH (2 h at 41.5 degrees C) and CBDCA (30 mg/kg) + WBH. Tumor growth delay, which was 1.9 days for CDDP + WBH and 2.7 days for CBDCA + WBH (P less than 0.01 compared to control), was significantly increased to 2.9 days with TNF + CDDP + WBH and 5.4 days with TNF + CBDCA + WBH (P less than 0.05). WBH, TNF, CDDP or CBDCA alone, TNF + CDDP, TNF + CBDCA, or TNF + WBH had no significant effect on tumor growth. In contrast, administration of TNF did not enhance the CDDP- or CBDCA-mediated dose limiting normal tissue toxicity. CDDP + WBH-mediated acute renal injury and CBDCA + WBH-mediated acute myelosuppression, as determined by blood urea nitrogen and peripheral blood cell counts, respectively, were not increased with the addition of TNF to either dual modality therapy. Histopathologically, addition of TNF produced no significant alterations in the kidney and the bone marrow as compared to CDDP + WBH or CBDCA + WBH. These data show that TNF enhanced the platinum + WBH-mediated antitumor effect without increasing normal tissue toxicity, suggesting that TNF may increase the therapeutic efficacy of CDDP or CBDCA combined with WBH.


Asunto(s)
Cisplatino/uso terapéutico , Fibrosarcoma/terapia , Hipertermia Inducida , Factor de Necrosis Tumoral alfa/uso terapéutico , Animales , Peso Corporal/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Cisplatino/toxicidad , Terapia Combinada , Femenino , Fibrosarcoma/patología , Hipertermia Inducida/efectos adversos , Riñón/efectos de los fármacos , Riñón/patología , Ratas , Ratas Endogámicas F344 , Factor de Necrosis Tumoral alfa/toxicidad
13.
Oncogene ; 13(8): 1693-700, 1996 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-8895515

RESUMEN

The retinoblastoma gene product, Rb, has previously been implicated as an obligatory component in the antiproliferative effects mediated by the lipid second messenger, ceramide. We have evaluated both the apoptotic effects and the effects on cell cycle distribution of the exogenous cell-permeable ceramide, N-hexanoyl-D-sphingosine, in an Rb-null human bladder tumor cell line, 5637, as well as in retrovirally infected, Rb(+) clones derived therefrom. These cell lines demonstrated comparable sensitivity to N-hexanoyl-D-sphingosine in a neutral red dye uptake assay. Exposure of the Rb-null parental cell line to 20 microM N-hexanoyl-D-sphingosine for 24 h resulted in a classical pattern of DNA fragmentation that was accompanied by apoptotic nuclear morphological alterations. In contrast, the Rb(+) clones demonstrated suppression of DNA fragmentation in response to N-hexanoyl-D-sphingosine. Similarly, the frequency and degree of alteration of nuclear morphology in Rb(+) cells was also suppressed. Flow cytometric analysis of the parental and infected clones indicated that expression of Rb was without effect on their cell cycle distribution, with or without exposure to N-hexanoyl-D-sphingosine for 25 h; tunel assay confirmed that in this time frame apoptotic cells were far less frequent in the Rb(+) clones than in the parental 5637 cells. Human tumor cell lines derived from three other histological origins, breast and prostatic carcinomas and osteogenic sarcoma, also demonstrated very similar cytotoxic sensitivities to N-hexanoyl-D-sphingosine, irrespective of the expression of Rb. We conclude that Rb is not required for ceramide-induced apoptosis and that Rb can actually inhibit the DNA fragmentation and nuclear morphological changes associated with classical apoptosis.


Asunto(s)
Apoptosis/fisiología , Ceramidas/antagonistas & inhibidores , Proteína de Retinoblastoma/fisiología , Neoplasias de la Vejiga Urinaria/patología , Ciclo Celular , Ceramidas/fisiología , Fragmentación del ADN , Humanos , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/genética
14.
Biochim Biophys Acta ; 1268(1): 65-72, 1995 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-7626664

RESUMEN

The urokinase-type plasminogen activator plays a central role in tissue remodeling by controlling the synthesis of the extracellular matrix-degrading plasmin. Urokinase expression is transcriptionally regulated by a variety of cytokines including TNF-alpha. The present study was undertaken to identify key transcription factor binding sites in the urokinase promoter necessary for the TNF-alpha-dependent induction of urokinase expression. TNF-alpha treatment of a squamous cell carcinoma cell line, UM-SCC-1, which produces no detectable TNF-alpha, led to a dose-dependent increase in urokinase secretion, thus reflecting a more abundant mRNA. Transient transfections of UM-SCC-1 cells with a CAT reporter driven by 5' deletion fragments of the urokinase promoter indicated that a sequence spanning -2109 to -1870, which contained binding sites for AP-1 and PEA3 was required for the stimulation by TNF-alpha. Mutation of an AP-1 binding site at -1967 and a PEA3 motif at -1973 completely abrogated the inductive effect of TNF-alpha on urokinase promoter activity. Mobility shift assays indicated the presence of a jun-containing factor(s) which bound specifically to the AP-1 sequence present in the urokinase promoter. The amount and/or activity of this factor(s) was greatly enhanced by TNF-alpha treatment. UM-SCC-1 cells transiently transfected with a CAT reporter driven by 3 tandem AP-1 binding sites demonstrated increased CAT activity following TNF-alpha treatment. Thus, the induction of urokinase expression by TNF-alpha is likely to involve the altered expression and/or activity of transcription factors which bind to the AP-1 and PEA3 target sequences in the urokinase promoter.


Asunto(s)
Regiones Promotoras Genéticas/genética , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Secuencia de Bases , Sitios de Unión , Cloranfenicol O-Acetiltransferasa/biosíntesis , Cloranfenicol O-Acetiltransferasa/genética , ADN/metabolismo , Inducción Enzimática/efectos de los fármacos , Genes Reporteros/genética , Humanos , Datos de Secuencia Molecular , Mutación/fisiología , ARN Mensajero/biosíntesis , Células Tumorales Cultivadas , Activador de Plasminógeno de Tipo Uroquinasa/genética
15.
J Leukoc Biol ; 40(6): 755-68, 1986 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-3465837

RESUMEN

The mechanism(s) with which tumor target cells actively resist the lethal lesion induced by murine macrophage cytolytic factory (CF) has been probed by drug-induced sensitization of these cells. We have examined whether drug-induced sensitization is attributable to the action of these drugs on cellular DNA, RNA, or protein synthetic rates. The murine mammary adenocarcinoma cell line EMT-6 pretreated with a dose range of actinomycin-D (.03-3.0 micrograms/ml) for 4 hr was inhibited from 66 to 98% in DNA synthesis rate, from 81 to 93% in RNA synthesis rate, and from 38 to 52% in protein synthesis rate. The maximum sensitization toward lysis by CF was achieved with a drug dose of 1.0 micrograms/ml. The lack of correlation between the drug-induced effects on sensitization and effects on these metabolic rates was evident from the correlation coefficients for the percentage of maximum sensitization of the target toward CF-induced lysis, and the percent of inhibition of DNA, RNA, and protein synthesis. These were 0.11, -0.11, -0.44, respectively. Similarly, target cells treated with a dose range of cycloheximide (0.3-30 micrograms/ml) were inhibited from 81 to 95% in DNA synthesis rate, 68 to 81% in RNA synthesis rate, and 82 to 93% in protein synthesis rate. However, at none of the drug levels employed was significant sensitization of the target cell to CF-induced lysis observed. This was reflected in the correlation coefficients of -0.55, -0.28, and -0.34 for DNA, RNA, and protein synthetic rates, respectively. The essential role of cellular microtubule-dependent events early in the lytic mechanism was demonstrated by exposing colchicone-treated targets to CF. While colchicone could markedly inhibit lysis if introduced before CF, this inhibitory effect was not detected if the drug were withheld for 4 hr after CF exposure. The importance of the repair mechanism(s) early in the cellular response to lesion formation was demonstrated by altering the schedule for CF and actinomycin D administration to the target. If actinomycin D treatment was withheld from the targets for as little as 3 hr following introduction of CF, the lytic mechanism had already bypassed the drug-sensitive steps, manifest in markedly decreased susceptibility to lysis. Collectively, the data show that the ability of the EMT-6 target cells to resist CF-induced lysis does not depend solely on the ability of the cells to synthesize DNA, RNA, or protein.


Asunto(s)
Citotoxinas/toxicidad , Macrófagos/fisiología , Proteínas/toxicidad , Animales , Células Cultivadas , Colchicina/farmacología , Cicloheximida/farmacología , Citotoxicidad Inmunológica/efectos de los fármacos , Dactinomicina/farmacología , Técnicas In Vitro , Activación de Macrófagos , Ratones , Monocinas , Ácidos Nucleicos/biosíntesis , Biosíntesis de Proteínas
16.
J Leukoc Biol ; 48(3): 220-8, 1990 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-2167918

RESUMEN

The roles of secreted and membrane-associated TNFs were investigated in activated macrophage cytolysis of L929, EMT-6, and P815 targets. While all three targets were susceptible to cytolysis in coculture, an anti-TNF antiserum blocked lysis of L929 and EMT-6 but not of the P815 targets. Of the three targets, recombinant human or mouse TNF could only lyse the L929 target; despite the fact that a role for TNF was invoked in lysis of EMT-6 targets in coculture, the latter was strongly resistant to soluble rTNF, even at concentrations 30-40-fold higher than the Ka for its TNF-receptor. Cytolysis of the L929 target occurred when it was cocultured with BCG-activated macrophages even when these effector cells did not secrete TNF, either due to prior chemical crosslinking or to lack of exposure to a triggering level of lipopolysaccharide. Furthermore, by introduction of the anti-TNF antiserum over a dose-range, it was shown that macrophage cytolysis both of L929 and EMT-6 targets occurred in the absence of bioavailable, fluid-phase TNF. Thus, even for targets susceptible to fluid-phase TNF, TNF-dependent, direct macrophage-mediated cytolysis appears to be a function independent of secreted TNF and one that utilizes effector-target contact to express the action of a membrane form of the molecule.


Asunto(s)
Macrófagos/fisiología , Mycobacterium bovis/fisiología , Animales , Conjugación Genética/fisiología , Citotoxinas/fisiología , Fibroblastos/fisiología , Fibroblastos/ultraestructura , Sueros Inmunes/inmunología , Interferón Tipo I/inmunología , Activación de Macrófagos , Macrófagos/ultraestructura , Masculino , Ratones , Fenotipo , Receptores de Superficie Celular/metabolismo , Receptores del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/fisiología
17.
J Leukoc Biol ; 35(2): 229-38, 1984 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-6584519

RESUMEN

Macrophage cytotoxin (MCT) can be induced from peritoneal exudate macrophage monolayers (PEMM) obtained from thioglycollate-treated mice, by exposure of PEMM to lipopolysaccharide (LPS) or to double-stranded polyinosinic: poly-cytidylic acid (poly-l:poly-C). MCT is highly labile even upon storage at 4 degrees C, and is irreversibly denatured by isolectricfocusing, polyacrylamide gel electrophoresis in the absence of sodium dodecyl sulfate (SDS), or by exposure to ethylene glycol. alpha-MCT [150,000 daltons (d)] has been highly purified (2,000- to 5,000-fold) from serum-free, PEMM supernatants by a scheme of concentration, molecular sieving on Ultrogel AcA 44, negative hydrophobic affinity chromatography on benzyl-agarose, and ion exchange chromatography on aminoethyl-agarose. The scheme results in high yield of MCT, in part because of the rapidity with which the labile toxin is manipulated due to the tandemization of the chromatographic steps.


Asunto(s)
Citotoxinas/aislamiento & purificación , Macrófagos , Animales , Cromatografía de Afinidad , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos C57BL , Temperatura
18.
J Leukoc Biol ; 50(5): 509-16, 1991 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-1748844

RESUMEN

The L-arginine-dependent tumor cell cytotoxicity produced by activated macrophages (M phi) may be mediated either directly by production of nitric oxide (NO), or by induction of NO synthesis in the tumor cell. The influence of M phi NO synthesis on the release of soluble cytotoxic mediators was investigated in this study. The synthesis of M phi NO, measured as nitrite, was detected 6 h after lipopolysaccharide (LPS)-triggering and reached a peak level by 44 h. A concurrent decrease in M phi viability beginning at 18 h after triggering was detected during a period of 72 h in culture. Both the production of NO and loss of viability correlated with the presence of L-arginine in the incubation media and was inhibited by NG-monomethyl-L-arginine (NMA). The medium in which LPS-triggered adherent peritoneal exudate cells were incubated was examined for the presence of tumor necrosis factor (TNF), gamma interferon (IFN-gamma), and the soluble mediators that induce mitochondrial respiratory inhibition in tumor cells. All of these effector molecules were released at peak levels into the conditioned supernatants within 12 h after LPS-triggering. The peak level obtained for each effector molecule was influenced by the media in which the M phi was incubated; however, no correlation was detected between the level of cytokines produced and the synthesis of nitrite by the M phi indicating that NO synthesis has no inhibiting effect on the initial burst of cytotoxic factors released.


Asunto(s)
Citocinas/metabolismo , Homeostasis/efectos de los fármacos , Óxido Nítrico/farmacología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Arginina/análogos & derivados , Arginina/farmacología , Medios de Cultivo/química , Femenino , Interferón gamma/metabolismo , Células L , Lipopolisacáridos/fisiología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Óxido Nítrico/metabolismo , Nitritos/análisis , Cavidad Peritoneal/citología , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/patología , Factor de Necrosis Tumoral alfa/metabolismo , omega-N-Metilarginina
19.
J Leukoc Biol ; 53(6): 715-21, 1993 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8315355

RESUMEN

We have previously demonstrated that in vivo activation or inhibition of Kupffer cell (KC) cytotoxic function can reduce or enhance, respectively, the hepatic tumor burden in a syngeneic murine colon adenocarcinoma (MCA26) tumor model. In the current study, we have performed in vitro experiments to define the possible mechanisms of KC cytotoxicity against MCA26 cells. Addition of either anti-tumor necrosis factor (TNF) or anti-interleukin-1 alpha (IL-1 alpha) antisera reduced KC cytotoxicity in coculture against MCA26 targets in a dose-dependent fashion; addition of these sera together resulted in approximately additive inhibition, suggesting the existence of parallel pathways for these effector molecules. Nitric oxide as a mediator of cytotoxicity by KCs in coculture with MCA26 cells was evaluated by two approaches. Activated KCs produced detectable levels of nitric oxide; however, activated KC exerted cytotoxicity against MCA26 targets in the absence of exogenous free L-arginine. Thus, TNF and IL-1 play major roles in producing murine KC cytotoxicity against MCA26 colon cancer cells in vitro, whereas reactive nitric oxides do not.


Asunto(s)
Adenocarcinoma/inmunología , Neoplasias del Colon/inmunología , Citotoxicidad Inmunológica , Macrófagos del Hígado/inmunología , Animales , Arginina/farmacología , Femenino , Sueros Inmunes/inmunología , Interleucina-1/fisiología , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico/metabolismo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/fisiología
20.
Mol Immunol ; 29(1): 77-81, 1992 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1731193

RESUMEN

The role of amino functions in the expression of the biological activity of recombinant human TNF (rHuTNF) was studied by chemical modification. rHuTNF is a homotrimer of 17 kD subunits, each of which contains an N-terminal valine and six lysyl residues: two of these lysyl residues are known to be involved in intra- or intersubunit interactions. Chemically reactive amino functions were modified with the N-hydroxysuccinimide ester of acetic acid; modification of amino groups to amide, and the concomitant loss of charge, was monitored by native PAGE. When rHuTNF was reacted with the active ester at increasing mole ratios, up to 12 amino groups per trimer could be modified. When the biological activity of acetylated rHuTNF was determined, a strong correlation between the extent of modification and loss of biological activity was observed. One to three amino groups per trimer could be modified with nearly complete retention (approximately 80-95%) of biological activity; activity was essentially completely destroyed at the highest levels of modification. These results reveal important functions for the amino groups of rHuTNF and significant constraints on strategies involving their modification in development of second-generation-TNF variants.


Asunto(s)
Factor de Necrosis Tumoral alfa/química , Acetilación , Aminas/química , Muerte Celular , Humanos , Técnicas In Vitro , Proteínas Recombinantes/química , Relación Estructura-Actividad , Factor de Necrosis Tumoral alfa/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA