Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Rev Med Interne ; 21(10): 863-71, 2000 Oct.
Artículo en Francés | MEDLINE | ID: mdl-11075395

RESUMEN

INTRODUCTION: Adoptive immunotherapy was first introduced in the 1980s. This new anticancer therapeutic approach has already demonstrated promising results in both animal models and humans affected by various tumors. CURRENT KNOWLEDGE AND KEY POINTS: This review summarizes the requirements of such therapies involving either activated lymphocytes, tumor-infiltrating lymphocytes or activated macrophages. It focuses more particularly on the promising approaches that represent antigen presenting cells such as macrophages and antigen-loaded dendritic cells in the development of safe and effective cancer vaccines. FUTURE PROSPECTS AND PROJECTS: Standardized procedures for macrophages and dendritic cell generation, as well as preliminary results of clinical applications in patients with either prostate cancer or melanoma, are also discussed.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Animales , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Predicción , Humanos , Inmunoterapia Adoptiva/tendencias , Linfocitos Infiltrantes de Tumor/inmunología , Macrófagos/inmunología , Masculino , Melanoma/terapia , Neoplasias de la Próstata/terapia , Resultado del Tratamiento
3.
Cancer Immunol Immunother ; 49(9): 493-503, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11092616

RESUMEN

OBJECTIVE: The aim was to determine the ability of macrophage-activated killer cells (MAK cells) obtained from peripheral blood of normal volunteers to kill glioblastoma multiforme (GBM) cell lines. Another goal was to investigate whether a bispecific antibody (bsAb) MDX-447, recognizing the high-affinity Fc receptor for IgG (FcgammaRI) and epidermal growth factor receptor (EGFR), would enhance MAK cell tumoricidal activity. METHODS: Monocytes, from leukapheresis product, were isolated by countercurrent elutriation and differentiated into MAK cells by culture with granulocyte/macrophage-colony-stimulating factor, vitamin D3 and interferon gamma. Cells were checked for sterility, endotoxin and phenotypic markers. MAK cell functional activity was measured by a flow-cytometric phagocytosis assay. Target cells, a carcinoma cell line and two glioma cell lines expressing EGFR, were stained with PKH-26. MAK cells were labeled with fluorescein-conjugated anti-CD14. Combined effectors, targets and bsAb were incubated and the percentage of MAK cells with phagocytosed targets was determined by flow cytometry. CONCLUSION: We demonstrate that a large number of highly purified monocytes, isolated from peripheral blood, can be differentiated into MAK cells for use as an adjuvant for cancer treatment. After culture these cells are sterile, endotoxin-free and comprise more than 95% MAK cells. Increased amounts of CD14, CD64 and HLA-DR, which are characteristics of macrophage activation, were expressed. MAK cells were extremely phagocytic in comparison to monocytes, even in the absence of bsAb. Moreover, bsAb enhanced the tumoricidal activity of elutriated MAK cells targeted against GBM cell lines. Therefore, intracavity MAK cells armed with MDX-447 could be an effective adoptive immunotherapy for EGFR-positive GBM.


Asunto(s)
Receptores ErbB/inmunología , Glioblastoma/inmunología , Glioblastoma/metabolismo , Células Asesinas Naturales/metabolismo , Macrófagos/metabolismo , Receptores de IgG/metabolismo , Adyuvantes Inmunológicos/uso terapéutico , Anticuerpos/metabolismo , Anticuerpos Monoclonales/metabolismo , Diferenciación Celular , Células Cultivadas , Colecalciferol/farmacología , Relación Dosis-Respuesta a Droga , Receptores ErbB/biosíntesis , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Inmunoterapia/métodos , Interferón gamma/farmacología , Receptores de Lipopolisacáridos/inmunología , Neoplasias Pulmonares/metabolismo , Microscopía Confocal , Monocitos/metabolismo , Fagocitosis , Fenotipo , Receptor ErbB-2/biosíntesis , Receptores de IgG/biosíntesis , Receptores de IgG/inmunología , Células Tumorales Cultivadas
4.
Leukemia ; 14(9): 1667-77, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10995015

RESUMEN

The recent clinical trial in lymphoma using tumor antigen-loaded DCs (Hsu et al, Nature Med 1996; 2: 52) demonstrates the efficiency of the use of professional antigen presenting cells (APCs) for taking up, processing and presenting tumor protein in a vaccine strategy in cancer. However, the production of large quantities of clinical grade APCs remains to be resolved. Here, we describe that both dendritic cells (DCs) and macrophages (MOs) can be efficiently differentiated in large numbers from lymphoma patients in spite of their disease and previous therapy. These cells were produced using the VAC and MAK cell processors according to standard operating procedures. DCs and MOs were differentiated from circulating monocytes in gas permeable hydrophobic bags, with 2% autologous serum and in the presence of GM-CSF and IL-13 or GM-CSF alone, respectively. DCs and MOs were then purified by counter flow centrifugation. Phenotypic, morphological and functional analysis showed that cells differentiated from patients with lymphoma present quite similar features to DCs and MOs produced from monocytes of healthy donors. Moreover, we show that MOs, when combined with CD20 antibody (Rituximab), can efficiently engulf tumor cells and propose that a such combination could be used for initiating a clinical trial in lymphoma. Thus, the possibility of producing functional DC and MOs in large amounts in conditions compatible with therapeutic application will allow the development of new immune strategies to eradicate lymphoma.


Asunto(s)
Células Presentadoras de Antígenos , Diferenciación Celular , Células Dendríticas , Linfoma no Hodgkin/terapia , Macrófagos , Adulto , Presentación de Antígeno/fisiología , Femenino , Humanos , Leucocitos Mononucleares/patología , Activación de Linfocitos/fisiología , Linfoma no Hodgkin/patología , Masculino , Persona de Mediana Edad , Fagocitosis , Fenotipo , Receptores Fc/fisiología , Linfocitos T/fisiología
5.
Immunol Invest ; 29(3): 319-36, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10933613

RESUMEN

The present study describes the optimization of an in vitro culture method for generating large amounts of dendritic cells (DC) in serum-free conditions from leukapheresis containing a mixed population of peripheral blood mononuclear cells (PBMC) which are cultured in the presence of GM-CSF and IL-13. Initial comparisons between the generation of DC from bulk and monocyte-enriched leukapheresis products showed that the presence of lymphocytes during the culture favors the differentiation of monocytes into DC. DC yields obtained from mixed mononuclear cell cultures were between 38 and 54% higher than yields obtained from monocyte-enriched cultures. Both types of cultures resulted in the generation of DC with an immature phenotype (CD83- and high phagocytic activity), which have been previously shown to be good stimulators for T cell responses. DC yields of bulk cultures in serum-free conditions were significantly higher than those obtained in the presence of 2% human serum. The cytokines of the supernatants of serum-free cultures comprised a significant content of pro-inflammatory cytokines such as IL-1, IL-12 and TNF-alpha. Maturation of DC generated by this method can be induced by treatment with double-stranded RNA, LPS or TNF-alpha, resulting in enhanced surface expression of CD80, CD86, CD40, CD83 and MHC molecules on the DC. The methodology described here offers the possibility for generating large amounts of clinical grade DC from bulk leukapheresis products, thus avoiding DC precursor purification steps, and thereby minimizing the risks of contamination. This culture process may be applied to cell-based therapeutic approaches for the treatment of cancer or chronic viral infections.


Asunto(s)
Técnicas de Cultivo/métodos , Células Dendríticas/citología , Comunicación Celular , Diferenciación Celular , Separación Celular/métodos , Medios de Cultivo/química , Medio de Cultivo Libre de Suero , Técnicas de Cultivo/normas , Citocinas/análisis , Humanos , Leucaféresis , Leucocitos Mononucleares/citología , Linfocitos/citología , Monocitos/citología , Fenotipo
6.
Res Immunol ; 149(7-8): 643-6, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9851516

RESUMEN

Since dendritic cells (DCs) are the most professional antigen-presenting cells, (Schuler et al., 1997), increasing interest in their use in clinical approaches has been observed. (Nestle et al., 1998; Murphy G. et al., 1996). We have developed an ex vivo standardized process for the generation of dendritic-like cells (MAC-DCs) from human blood circulating monocytes. Human monocytes can differentiate into very different functional cells according to the conditions of culture, media and cytokines used. In the present study, we demonstrate that both pure monocytes and mononuclear cells differentiate into DCs when they are grown in defined medium AIM-V in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) plus IL13 and in approved biocompatible non-adherent bags. Quality and functional controls of the immature DCs obtained rely on bacterial sterility, viability, morphology and recovery. The MAC-DCs also present an immature DC phenotype with a low expression of CD14 and CD64, and high expression of MHC-I, MHC-II and CD40. They also express B7 costimulatory molecules (CD80, CD86), CD83, and CD1a molecules. They induce strong allogenic T-cell proliferation (mixed lymphocyte reaction as well as proliferation of autologous memory T lymphocytes when incubated in the presence of recall antigens (tuberculosis, Candida albicans, and tetanus toxoid). They also show an increase in phagocytic uptake of yeast, tumour cells and debris. The global closed system which, under reproducible good medical practice (GMP) conditions, enables the production of dendritic cells of clinical quality, has been optimized ("Vac Cell Processor"). It contains all bags, connections, media, reagents, washing solutions, control antibodies, standard operating procedures, data management, traceability and help in the form of dedicated software.


Asunto(s)
Células Dendríticas/citología , Monocitos/citología , Presentación de Antígeno/inmunología , Candida albicans/inmunología , Diferenciación Celular , Supervivencia Celular , Células Dendríticas/inmunología , Humanos , Inmunofenotipificación , Prueba de Cultivo Mixto de Linfocitos , Macrófagos/citología , Macrófagos/inmunología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Fagocitosis/inmunología
7.
Res Immunol ; 149(7-8): 647-9, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9851517

RESUMEN

This project is devoted to the development of novel cellular vaccines designed to treat cancer patients. These cellular vaccines present and enhance immunogens, which will elicit a potent immune response. The goal is to achieve safe and effective immune reaction against the patient's own tumour. (1) Autologous cellular vaccines are prepared by processing circulating blood mononuclear cells outside of the patient's body (ex vivo) to differentiate them into antigen-presenting cells (APCs). Monocyte-derived APCs (MD-APCs) are then grown in the presence of exogenous target antigens (tumour cell debris, or apoptotic bodies) to become fully mature APCs. (2) Functionality for antigen presentation to T cells of ex vivo MD-APCs is evaluated in vivo. (3) Cellular vaccines are tested in selected rodent animal models. Efficiency and immune response are monitored in pertinent experimental systems for cancer. Pharmacological data are generated for clinical investigation. Tolerance and biologic effects are documented in primates. (4) The first clinical trials on cancer patients are taking place in 1998 on melanoma and prostate cancer to validate the concept. Specialized cell processors with dedicated software and standardized controls are being developed and used for the preparation of cellular vaccines. (5) The evaluation of new non-viral vectors and the validation of new non-viral transfection methods of mononuclear cells with marker genes is in progress and will lead to the ex vivo transfection of genes coding for immunostimulating cytokines or for tumour antigens in MD-APCs. Efficiency will be validated in vitro and in animal models. The ex vivo and animal model studies validate the clinical relevance of this new cellular immunotechnology. Clinical validation of individual autologous cellular vaccines in specific indications for which no treatment is presently available will allow the development of cellular and gene immunotherapy for other types of cancers.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Vacunas contra el Cáncer/inmunología , Monocitos/inmunología , Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/citología , Ensayos Clínicos como Asunto , Vectores Genéticos , Humanos , Masculino , Melanoma/prevención & control , Monocitos/citología , Neoplasias de la Próstata/prevención & control
8.
Immunobiology ; 195(4-5): 550-62, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8933156

RESUMEN

Adoptive transfer of activated macrophages has been validated in animal experimental tumor models; clinical trials are ongoing (70 patients up to now). The mechanisms involved are reviewed as well as improved and standardized procedures for macrophage differentiation and activation. New developments including specific Ag presentation and gene therapy are discussed.


Asunto(s)
Inmunoterapia Adoptiva , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/trasplante , Animales , Ensayos Clínicos como Asunto , Humanos , Inmunoterapia Adoptiva/métodos , Inmunoterapia Adoptiva/normas
9.
J Exp Ther Oncol ; 1(3): 143-54, 1996 May.
Artículo en Inglés | MEDLINE | ID: mdl-9414398

RESUMEN

Adoptive transfer of activated autologous human macrophages obtained by in vitro differentiation of monocytes in culture has successfully undergone phase I clinical trials in patients with metastatic cancer. The efficacy of these autologous macrophages in the in vivo killing of human tumors has now to be demonstrated. GM-CSF was shown to increase the number of monocytes differentiating in culture into macrophages. These were then activated with IFN gamma which was reported as the best cytokine for tumoricidal activation of macrophages. The aim of this paper was to evaluate the in vitro tumoricidal activity of macrophages grown with GM-CSF and IFN gamma. This tumoricidal function was investigated by measurement of the cytolytic (chromium-51 release assay), cytostatic (anti-proliferative activity as measured by inhibition of [3H]-thymidine incorporation in two different assays) and cytotoxic (MTT assay) activities on two tumor cells lines, U937 and K562 (respectively highly sensitive and resistant to soluble TNF alpha). Our results demonstrated that GM-CSF-grown macrophages exhibited significant cytolytic, cytotoxic and cytostatic activities on U937 cells. These were partly enhanced by IFN gamma activation. In contrast, they had no lytic and lower cytotoxic and cytostatic activities on K562 cells, and these were not modified by IFN gamma activation. Our data provide valuable information for future in vivo studies using adoptively transferred autologous macrophages.


Asunto(s)
Traslado Adoptivo , Macrófagos/fisiología , Monocitos/fisiología , Antineoplásicos/farmacología , Recuento de Células , Separación Celular , Supervivencia Celular/efectos de los fármacos , Radioisótopos de Cromo , Colorantes , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Interferón gamma/farmacología , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Proteínas Recombinantes , Sales de Tetrazolio , Tiazoles , Timidina/metabolismo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología
10.
J Immunol Methods ; 159(1-2): 29-38, 1993 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-8445262

RESUMEN

Blood monocytes collected by apheresis from healthy donors were differentiated in vitro to macrophages which were subsequently activated with recombinant human interferon-gamma. 7 day cultures were established by seeding Ficoll-separated mononuclear cells or elutriation-purified monocytes under different culture conditions. The best macrophage yields required the seeding of mononuclear cells (instead of purified monocytes) in teflon bags with a high air-liquid surface interface. The effects of GM-CSF, IL-3 and M-CSF on the macrophage yield were then evaluated. GM-CSF increased the average yield by 3.6- and 2.3-fold when purified monocytes or total mononuclear cells were seeded respectively. The corresponding increases with IL-3 were 2.5- and 2.1-fold respectively and with M-CSF 1.2- and 1.4-fold respectively. Macrophages matured under these various conditions displayed similar CD14, CD64, CD71, HLA-DR and Max 1 antigen expression and similar in vitro anti-tumoral activity against U937 cells. Culturing in the presence of cytokines permits the large scale production of activated macrophages for adoptive immunotherapy trials.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Interleucina-3/farmacología , Linfocitos/fisiología , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/fisiología , Monocitos/fisiología , Complejo CD3/análisis , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular , Células Cultivadas , Medios de Cultivo Condicionados , Humanos , Macrófagos/efectos de los fármacos
11.
Anticancer Res ; 12(6B): 2257-60, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1295473

RESUMEN

Antitumoral macrophages (MAK) were obtained by the culture of human mononuclear cells in hydrophobic bags. From one cytapheresis, up to 10(9) mature macrophages could be purified by elutriation after one week of culture in IMDM medium in the presence of 2% human AB serum. These MAK cells were used for adoptive treatment in metastatic cancer patient with no dose-limiting toxicity. The present study aimed to improve the average MAK yield by addition of GM-CSF and of dihydroxy-cholecalciferol. The differentiated macrophages obtained presented higher antitumoral functionality in response to rh-IFN gamma than in their absence. These MAK presented all the differentiation antigens of cytotoxic macrophages compared to MAK cells differentiated in standard medium. They killed human tumor targets effectively in vitro at a low (1/1) effector/tumor ratio; furthermore, the antitumoral activity reached by MAK cells after IFN gamma activation appeared to be stabilized for several days.


Asunto(s)
Calcitriol/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Activación de Macrófagos , Macrófagos/inmunología , Monocitos/inmunología , Antígenos CD/análisis , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Antígenos HLA-DR/análisis , Humanos , Interferón gamma/farmacología , Cinética , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Proteínas Recombinantes , Células Tumorales Cultivadas
12.
J Immunother (1991) ; 11(3): 209-17, 1992 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1515425

RESUMEN

Ninety-three collections of leucocytes by cytapheresis followed by separation of monocytes by centrifugal elutriation were undertaken in twelve metastatic cancer patients (four melanomas, six colon carcinomas, one ovarian carcinoma, and one lung cancer). The leucaphereses were performed aiming to collect a product, ready for introduction into the elutriation chamber, i.e., with low contamination by erythrocytes and granulocytes. The median collection of leucocytes was 7.3 x 10(9). After elutriation, purified monocytes (mean: 0.91 x 10(9)) were cultured with 3-5% autologous serum for 7 days in the presence of 250 IU/ml of recombinant human gamma-interferon (Rh-IFN gamma) for the last 18 h of culture. The median number of activated macrophages (MAK) available for reinfusion was 2.4 x 10(8) for each culture. The phenotypes and the antitumoral potentiality of MAK cells were documented. Reinfusions performed i.v. or i.p. were well tolerated with no major side effects. No complete tumor response was obtained. One partial response and two stabilizations of the disease were observed in one melanoma and two colon carcinomas.


Asunto(s)
Inmunoterapia Adoptiva , Leucocitos Mononucleares/trasplante , Activación de Macrófagos , Macrófagos/trasplante , Neoplasias/terapia , Animales , Células Cultivadas , Humanos , Interferón gamma/farmacología , Leucaféresis , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Desnudos , Persona de Mediana Edad , Neoplasias/sangre , Neoplasias/patología , Neoplasias Experimentales/terapia , Proyectos Piloto , Proteínas Recombinantes
13.
Res Immunol ; 143(1): 95-9, 1992 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1533049

RESUMEN

We report on two applications of bispecific antibodies to enhance the antitumoral function of human macrophages: (1) use of rhuIFN gamma (recombinant human IFN gamma) encapsulated in human red blood cells coated with anti-Fc gamma RI/anti-RhD+ bispecific antibodies to target and to activate human macrophages; encapsulated rhuIFN gamma was more potent than free IFN gamma in activating mature macrophages in vitro, demonstrating the efficacy of this delivery system to initiate in situ activation of macrophages and also to maintain a high antitumoral efficacy of macrophages with less side effects than after systemic injection of IFN gamma; (2) targeting of activated macrophages to tumours by bispecific antibodies directed against macrophage Fc gamma RI and against human adenocarcinoma antigen; differentiated human macrophages became cytotoxic for human adenocarcinoma in vitro and in vivo (tumours implanted in nude mice) when activated by rhuIFN gamma; this effect was increased in the presence of bispecific antibodies. These two approaches were aimed at increasing the efficacy of cellular immunotherapies using activated macrophages as effector cells (macrophage-activated killer, or MAK), an adoptive therapy which we have developed. Bispecific antibodies could increase specific homing and activation of cytotoxic MAK effectors at tumour sites.


Asunto(s)
Anticuerpos/administración & dosificación , Inmunoterapia Adoptiva , Macrófagos/inmunología , Antígenos de Diferenciación , Reactivos de Enlaces Cruzados , Eritrocitos/inmunología , Humanos , Técnicas In Vitro , Interferón gamma/administración & dosificación , Isoanticuerpos/administración & dosificación , Activación de Macrófagos , Vehículos Farmacéuticos , Receptores Fc , Receptores de IgG , Proteínas Recombinantes , Sistema del Grupo Sanguíneo Rh-Hr
14.
Anticancer Res ; 9(4): 1185-90, 1989.
Artículo en Inglés | MEDLINE | ID: mdl-2817800

RESUMEN

Treatment of C57BL/6 mice bearing Lewis lung carcinoma or of BALB/c mice bearing EMT6 sarcoma with tumor necrosis factor (TNF), lipopolysaccharides (LPS) or interferon caused necrosis of the solid tumors and regression. Toxicity was observed in tumor-bearing animals when TNF or LPS were used at effective antitumoral doses. Similar antitumoral effects could be achieved using less than 1 million macrophages from C57BL/6, lung of from BALB/c peritoneal cavity expanded in vitro, and spontaneously fully activated to cytotoxicity during culture. This effect, observed after transfer twice a week by intravenous or peritumoral route, was not dependent on histocompatibility. Additive effects were observed after combined treatment with activated macrophages and a low dose of LPS or TNF. The biodistribution of labelled LPS and of labelled cytotoxic macrophages was studied in tumor-bearing mice. Although, as expected, LPS was concentrated essentially in the liver, a slow accumulation in the center of the tumor was observed. Macrophages injected intravenously accumulated in the lung and were then redistributed towards liver, kidney and the tumor periphery. Macrophages injected locally remained essentially in the tumor periphery with a slow redistribution in the body. The complementary localization of LPS and of cytotoxic macrophages respectively in the center and periphery of solid tumors might explain their synergism.


Asunto(s)
Interferón Tipo I/uso terapéutico , Lipopolisacáridos/uso terapéutico , Neoplasias Pulmonares/terapia , Activación de Macrófagos , Macrófagos/trasplante , Sarcoma Experimental/terapia , Factor de Necrosis Tumoral alfa/uso terapéutico , Animales , Células Cultivadas , Femenino , Inmunoterapia , Lipopolisacáridos/farmacocinética , Neoplasias Pulmonares/tratamiento farmacológico , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Recombinantes , Sarcoma Experimental/tratamiento farmacológico , Especificidad de la Especie , Distribución Tisular
15.
J Leukoc Biol ; 44(5): 391-401, 1988 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3141541

RESUMEN

Murine resident macrophages can proliferate in vitro when they are grown in coculture on a layer of mesothelial or endothelial type feeder cells. Resident macrophages were obtained from lung explants of C57Bl/6 lpr/lpr mice and from spleen explants or peritoneal washing of Balb/c mice; the cells were seeded without further washing. After 3-4 weeks of culture, the macrophages began to proliferate on a confluent layer of feeder cells. The macrophages then could be collected in the fluid phase and reseeded for permanent culture after generation of a new feeder layer. These cells were characterized as macrophages by the following criteria: 1) their morphology, ultrastructure, and adherence properties; 2) more than 90% of the macrophages phagocytized yeasts compared with less than 1% of the feeder cells; 3) the presence of functional Fc and mannose receptors, nonspecific cytoplasmic esterases, and membrane ectoenzymes such as nicotinamide adenine dinucleotide (NAD) glycohydrolase and nucleotide pyrophosphatase; 4) by cytofluorographic phenotype analysis with monoclonal antibodies, characterizing a normal macrophage population (MAC1+, Fcrec+, H-2K+, THY1-, LYT2-, L3T4-). 5) by functional studies proving that the expanded macrophages could function as accessory cells in the induction of lymphocyte proliferation in response to concanavalin A (Con A), that they generated reactive oxygen radicals and that they were cytotoxic for tumor cells. During coculture, growth or activating factors such as macrophage colony-stimulating factor or gamma-interferon were released in the medium. Long-term cultured macrophages had chromosomal abnormalities. Our study suggests that tissue macrophages can proliferate in vitro and hence that it is possible to establish long-term cultured cell lines of macrophages of defined and reproducible characteristics.


Asunto(s)
Macrófagos/citología , Animales , División Celular , Línea Celular , Aberraciones Cromosómicas , Sustancias de Crecimiento/metabolismo , Interferón gamma/farmacología , Interleucinas/metabolismo , Macrófagos/fisiología , Macrófagos/ultraestructura , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA