RESUMEN
Cytotoxic T lymphocytes and natural killer cells are essential effectors of anti-tumor immune responses in vivo. Dendritic cells (DC) 'prime' tumor antigen-specific cytotoxic T lymphocytes; thus, we investigated whether DC might also trigger the innate, NK cell-mediated anti-tumor immunity. In mice with MHC class I-negative tumors, adoptively transferred- or Flt3 ligand-expanded DC promoted NK cell-dependent anti-tumor effects. In vitro studies demonstrated a cell-to-cell contact between DC and resting NK cells that resulted in a substantial increase in both NK cell cytolytic activity and IFN-gamma production. Thus, DC are involved in the interaction between innate and adaptive immune responses.
Asunto(s)
Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Neoplasias Experimentales/inmunología , Traslado Adoptivo , Animales , Comunicación Celular , Técnicas de Cocultivo , Proteínas de Unión al ADN , Ligandos , Activación de Linfocitos , Complejo Mayor de Histocompatibilidad/inmunología , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos , Ratones Mutantes , Neoplasias Experimentales/clasificaciónRESUMEN
The initiation of T-cell-mediated antitumor immune responses requires the uptake and processing of tumor antigens by dendritic cells and their presentation on MHC-I molecules. Here we show in a human in vitro model system that exosomes, a population of small membrane vesicles secreted by living tumor cells, contain and transfer tumor antigens to dendritic cells. After mouse tumor exosome uptake, dendritic cells induce potent CD8+ T-cell-dependent antitumor effects on syngeneic and allogeneic established mouse tumors. Therefore, exosomes represent a novel source of tumor-rejection antigens for T-cell cross priming, relevant for immunointerventions.
Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias Mamarias Experimentales/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Células Dendríticas/inmunología , Humanos , Neoplasias Mamarias Experimentales/ultraestructura , Ratones , Microscopía Inmunoelectrónica , Células Tumorales CultivadasRESUMEN
The HNK-1 antibody known to define a subpopulation of human lymphocytes with natural killer and killer cell activities was shown to detect a common neuroectodermal antigen. Most tumor lines and paraffin-embedded tumors and normal tissues of neuroectodermal origin were specifically stained by HNK-1. Lines and tissues of other derivations were negative except a trophoblastic tumor line and a percentage of Ewing's sarcomas, whose histogenesis is poorly understood. These data indicate that HNK-1 antibody could be of interest in clinical histopathology but cannot be considered as specific for a lymphocyte subset.
Asunto(s)
Anticuerpos Monoclonales/inmunología , Ectodermo/inmunología , Epítopos/inmunología , Tejido Nervioso/inmunología , Especificidad de Anticuerpos , Línea Celular , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Linfocitos/inmunología , Neoplasias de Tejido Nervioso/inmunologíaRESUMEN
Peripheral T lymphocytes from patients with infectious mononucleosis (IM) are sensitized in vivo against the Epstein-Barr virus (EBV). The expression of HLA-A, B, or C molecules at the target cell surface is necessary for the cytotoxic reaction because (a) EBV-positive Daudi cells lacking HLA-A, B, and C determinants are resistant to anti-EBV T-cell lysis, (b) cytolysis of EBV-positive target cells can be consistently inhibited by anti-HLA-A, B, and C and anti-beta 2 microglobulin antibodies. However, no evidence for allogeneic restriction in this system was apparent as (a) cytotoxic T lymphocytes (CTL) from one given individual could exert a cytotoxicity of a similar magnitude on different EBV-positive target cells, regardless of the number of HLA-A or B specificities shared by the effectors and targets; (b) CTL from IM patients were able to kill target cells without any HLA-A or B antigen in common; and (c) T5-1 variants lacking one or two HLA antigens at the A, B, or D locus are killed to the same extent as the parental cells. 7 of the 9 IM patients with detectable circulating anti-EBV CTL carried the HLA-A1 antigen, whereas none of the 16 IM patients lacking detectable peripheral CTL were HLA-A1 positive (mean specific lysis of T5-1 target cells by T cells from HLA-A1 positive patients: 29.3 vs. 0.6% in HLA-A1-negative patients) (P less than 10(-9)). These data suggest an HLA-A1-linked gene control of the magnitude of the anti-EBV CTL response. Thus, the HLA region appears to act at two different level sin the T-cell-mediated lysis of EBV-infected cells by controlling first, the development of anti-EBV and second, the expression of HLA-A, B, and C molecules involved as recognition structures at the target cell surface.
Asunto(s)
Citotoxicidad Inmunológica , Antígenos HLA/genética , Herpesvirus Humano 4/inmunología , Inmunidad Celular , Mononucleosis Infecciosa/inmunología , Linfocitos T/inmunología , Reacciones Antígeno-Anticuerpo , Genes MHC Clase II , Ligamiento Genético , Humanos , Isoanticuerpos , Complejo Mayor de Histocompatibilidad , Microglobulina beta-2/inmunologíaRESUMEN
Given the broad expression of H-2 class Ib molecules on hematopoietic cells, antigen presentation pathways among CD1d expressing cells might tightly regulate CD1d-restricted natural killer T (NKT) cells. Bone marrow-derived dendritic cells (BM-DCs) and not adherent splenocytes become capable of triggering NK1.1(+)/T cell receptor (TCR)(int) hepatic NKT cell activation when (a) immature BM-DCs lack H-2D(b)-/- molecules or (b) BM-DCs undergo a stress signal of activation. In such conditions, BM-DCs promote T helper type 1 predominant CD1d-restricted NKT cell stimulation. H-2 class Ia-mediated inhibition involves more the direct H-2D(b) presentation than the indirect Qa-1(b) pathway. Such inhibition can be overruled by B7/CD28 interactions and marginally by CD40/CD40L or interleukin 12. These data point to a unique regulatory role of DCs in NKT cell innate immune responses and suggest that H-2 class Ia and Ib pathways differentially control NKT cell recognition of DC antigens.
Asunto(s)
Antígenos CD1/inmunología , Antígeno B7-1/inmunología , Células Dendríticas/inmunología , Antígenos H-2/inmunología , Interferón gamma/biosíntesis , Células Asesinas Naturales/inmunología , Linfocitos T/inmunología , Animales , Antígenos/genética , Antígenos/inmunología , Antígenos CD1/genética , Antígenos CD1d , Antígenos Ly , Antígenos de Superficie , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Antígenos CD28/inmunología , Diferenciación Celular , Células Cultivadas , Células Dendríticas/citología , Femenino , Antígenos H-2/genética , Antígeno de Histocompatibilidad H-2D , Lectinas Tipo C , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subfamilia B de Receptores Similares a Lectina de Células NK , Proteínas/genética , Proteínas/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Transducción de Señal/inmunologíaRESUMEN
The antigen defined by a rat monoclonal antibody directed to a Burkitt lymphoma cell line was identified as globotriaosylceramide [Gal alpha (1 leads to 4)-Gal beta (1 leads to 4)-Glc beta (1 leads to 1)-ceramide]. The antibody demonstrated a strict steric specificity since it did not react with globoisotriaosylceramide [Gal alpha (1 leads to 3)-Gal beta (1 leads to 4)-Glc beta (1 leads to 1)-ceramide], the positional isomer of the antigen associated with the Burkitt lymphoma. Chemical analysis of various Burkitt lymphoma cell lines revealed that the Burkitt lymphoma cells contained more than 100 times as much of the glycolipid antigen as was found in other human lymphoma and leukemia cell lines.
Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos de Neoplasias/inmunología , Linfoma de Burkitt/inmunología , Globósidos/inmunología , Glicoesfingolípidos/inmunología , Trihexosilceramidas , Animales , Línea Celular , Transformación Celular Neoplásica/metabolismo , Eritrocitos/inmunología , Humanos , Conejos , RatasRESUMEN
BACKGROUND: Recombinant tumor necrosis factor-alpha (TNF-alpha) combined to melphalan is clinically administered through isolated limb perfusion (ILP) for regionally advanced soft tissue sarcomas of the limbs. In preclinical studies, wild-type p53 gene is involved in the regulation of cytotoxic action of TNF-alpha and loss of p53 function contributes to the resistance of tumour cells to TNF-alpha. The relationship between p53 status and response to TNF-alpha and melphalan in patients undergoing ILP is unknown. PATIENTS AND METHODS: We studied 110 cases of unresectable limbs sarcomas treated by ILP. Immunohistochemistry was carried out using DO7mAb, which reacts with an antigenic determinant from the N-terminal region of both the wild-type and mutant forms of the p53 protein, and PAb1620mAb, which reacts with the 1620 epitope characteristic of the wild-type native conformation of the p53 protein. The immunohistochemistry data were then correlated with various clinical parameters. RESULTS: P53DO7 was found expressed at high levels in 28 patients, whereas PAb1620 was negative in 20. The tumours with poor histological response to ILP with TNF-alpha and melphalan showed significantly higher levels of p53-mutated protein. CONCLUSIONS: Our results might be a clue to a role of p53 protein status in TNF-alpha and melphalan response in clinical use.
Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Biomarcadores de Tumor/análisis , Quimioterapia del Cáncer por Perfusión Regional , Sarcoma/química , Sarcoma/tratamiento farmacológico , Proteína p53 Supresora de Tumor/análisis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/inmunología , Niño , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Imagen por Resonancia Magnética , Masculino , Melfalán/administración & dosificación , Persona de Mediana Edad , Mutación Missense , Sarcoma/patología , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/administración & dosificación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/inmunologíaRESUMEN
In a young woman with ulcerative colitis, hypoimmunoglobulinemia, and humoral immunodeficiency, lymphocyte counts vary between 600 and 1,000 per mm(3) with 0.5-1.5% bone marrow-derived (B) cells and 98-99% thymus-derived (T) cells. Anti-lymphocyte antibodies were detected by immunofluorescence and by microlymphocytotoxicity with increased reactivity at +4 degrees C. They belonged to the IgM class and were polyclonal. Studies performed with various normal lymphocyte subpopulations, several lymphoblastoid cell lines and lymphocytes from immunodeficiency patients showed that these antibodies reacted with B cells. The corresponding antigen(s) is distinct from membrane-bound immunoglobulins, is not an alloantigen, and is probably unrelated to the la-like molecules. Pokeweed mitogen stimulated B cells appear to lose this antigen. Cells from various lymphoproliferative disorders were tested. T-derived and "non T-non-B" leukemic cells did not react with the antibody. Malignant cells from B-derived lymphomas and prolymphocytic leukemias were reactive. The incidence of positivity of the leukemic cells among patients with common B chronic lymphocytic leukemia was surprisingly low (one-third of the patients). The autoantibody nature of the anti-B-cell antibodies and their pathogenic role in the genesis of the patient's hypoimmunoglobulinemia was demonstrated by the effect of removal of antibodies by massive plasmaphereses which were followed by a dramatic and transitory increase of B-cell figures. Whereas most primary immunodeficiency syndromes appear to result from an arrest in the differentiation capabilities of immunologically competent cells, autoantibodies to circulating B lymphocytes may be incriminated in the pathogenesis of some cases of hypogammaglobulinemia.
Asunto(s)
Autoanticuerpos , Linfocitos B/inmunología , Síndromes de Inmunodeficiencia/inmunología , Adulto , Colitis Ulcerosa/inmunología , Femenino , Humanos , Reacción de Inmunoadherencia , Síndromes de Inmunodeficiencia/fisiopatología , Tonsila Palatina/fisiopatología , Linfocitos T/inmunología , Timo/fisiologíaRESUMEN
Animal studies indicate that the use of replication-deficient adenovirus for human gene therapy is limited by host antivector immune responses that result in transient recombinant protein expression and blocking of gene transfer when rechallenged. Therefore, we have examined immune responses to an adenoviral vector and to the beta-galactosidase protein in four patients with lung cancer given a single intratumor injection of 10(9) plaque-forming units of recombinant adenovirus. The beta-galactosidase protein was expressed in day-8 tumor biopsies from all patients at variable levels. Recombinant virus DNA was detected by PCR in day-30 and day-60 tumor biopsies from all patients except patient 1. A high level of neutralizing antiadenovirus antibodies was detected in patient 1 before Ad-beta-gal injection whereas it was low (patient 3) or undetectable in the other two patients. All patients developed potent CD4 type 1 helper T cell (Th1) responses to adenoviral particles which increased gradually over time after injection. Antiadenovirus cytotoxic T lymphocyte responses were consistently boosted in the two patients examined (patients 3 and 4). Sustained production of anti-beta-galactosidase IgG was observed in all patients except patient 1. Consistent with anti-beta-gal antibody production, all patients except patient 1 developed intense, dose-dependent Th1 responses to soluble beta-galactosidase which increased over time. Strong beta-galactosidase-specific cytotoxic T lymphocyte responses were detected in patients 2, 3, and 4. Our results clearly show that despite the intensity of antiadenovirus responses, transgene protein expression was sufficient to induce strong and prolonged immunity in three patients. Recombinant adenovirus injected directly into the tumor is a highly efficient vector for immunizing patients against the transgene protein.
Asunto(s)
Neoplasias Pulmonares/terapia , Adenoviridae/genética , Anticuerpos Antivirales/biosíntesis , Citotoxicidad Inmunológica , ADN Viral/análisis , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Activación de Linfocitos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Factores de Tiempo , beta-Galactosidasa/genéticaRESUMEN
BACKGROUND: Despite vigorous efforts at curbing tobacco consumption and aggressive combined-modality treatment programs, both the incidence of and the mortality from lung cancer have remained virtually unchanged in the last 10 years. More effective innovative therapies are clearly needed. The direct transfer into tumor cells of tumor suppressor genes or toxic gene products that specifically promote tumor cell death and spare nonmalignant cells is a potentially novel anticancer treatment approach that should be investigated. PURPOSE: On the basis of compelling preclinical data, we initiated a phase I study involving six patients with inoperable lung cancer and an endobronchial lesion accessible by bronchoscopy. Our purpose was to evaluate the feasibility, tolerance, and clinical, biologic, and immunologic effects of the intratumoral administration of a recombinant, replication-deficient adenovirus (rAd.RSV beta-gal), using the Rous sarcoma virus promoter to drive transcription of the Escherichia coli lacZ marker gene that encodes for the bacterial enzyme beta-galactosidase (beta-gal). METHODS: From June 1994 through April 1995, six patients (five males and one female) were enrolled in the trial. A single dose of recombinant virus suspension containing 10(7) or 10(8) plaque-forming units (PFU) was injected intratumorally into two successive cohorts of three patients. Eligible patients received concomitant chemotherapy. Patients were kept under isolation conditions from 3 days before the injection was given until virus excretion was undetectable. Biopsy specimens of the tumor and surrounding mucosa were collected on the 8th day and at 1, 2, and 3 months after injection. They were analyzed by cell culture, polymerase chain reaction (PCR), and beta-gal expression for the presence of recombinant adenovirus. So that the risk of virus recombination or complementation could be minimized, wildtype adenovirus carriers among the hospital staff (identified by PCR) were excluded from contact with patients who were potentially excreting recombinant virus. RESULTS: beta-gal was expressed in tumor biopsy specimens of three patients (one who received the 10(7) PFU dose level and two who received 10(8)). Bronchoalveolar lavage specimens collected immediately after injection were positive for recombinant adenovirus when analyzed in culture and by PCR. All biologic fluids were negative for recombinant virus as judged by PCR after day 12, with the exception of bronchoalveolar lavage specimens (positive PCR up to 90 days in two of three patients treated with 10(8) PFU). The blood samples obtained from the three patients treated with 10(8) PFU showed positive PCR results immediately after virus injection. Patients were kept in isolation for a median of 17 days. The most common toxic effects were moderate bleeding (occurring in two patients) during bronchoscopy and fever (seen in four patients). Endoscopic and clinically objective antitumor responses were seen in four patients, including one patient who showed a complete response by pathologic evaluation. The median survival for the patients was 12.5 months (range, 3-16+ months). Throughout the study, hospital staff remained negative for recombinant adenovirus infection. CONCLUSIONS: This ongoing phase I study has demonstrated that a recombinant adenovirus-mediated marker gene, such as rAd.RSV beta-gal, can be safely introduced into humans and that the gene product is expressed by lung tumor cells of the host.
Asunto(s)
Neoplasias de los Bronquios/terapia , Carcinoma/terapia , Terapia Genética/métodos , Neoplasias Pulmonares/terapia , beta-Galactosidasa/genética , Adenoviridae , Neoplasias de los Bronquios/enzimología , Líquido del Lavado Bronquioalveolar , Broncoscopía , Carcinoma/enzimología , Estudios de Factibilidad , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Neoplasias Pulmonares/enzimología , Reacción en Cadena de la PolimerasaRESUMEN
A monoclonal immunoglobulin M (IgM) antibody (38-13) which recognizes Burkitt's lymphoma (BL) cells, by reacting with the neutral glycolipid Gal alpha 1 leads to 4-Gal beta 1 leads to 4-Glc beta 1 leads to 1-ceramide, was recently characterized. This monoclonal IgM was coupled to either ricin A chain or gelonin. The two different immunotoxins obtained retained the apparent immunological specificity of 38-13 IgM, as shown by flow cytofluorometry analysis and complement-dependent cytotoxicity test. The BL Ramos cells and the apparently irrelevant Epstein-Barr virus-containing lymphoblastoid Priess cells were used as targets in in vitro assays of the cytotoxic properties of the two immunotoxins by measuring the inhibition of protein synthesis. Isolated ricin A chain, gelonin, and 38-13 IgM exhibited very low intrinsic cytotoxicity on both target cells. 38-13 ricin A chain and 38-13 gelonin conjugates exerted toxic effects on both target cells which were about 6000-fold and 3000-fold higher than uncoupled ricin A chain and gelonin, respectively. The toxicity of these conjugates almost reached that of intact ricin. On Ramos BL cells, the kinetics of action of the 38-13 ricin A chain conjugate was almost as fast as that of intact ricin, because 50% protein synthesis inhibition was reached after 3 hr. In contrast, the kinetics of action in the non-BL Priess was much slower (50% protein synthesis inhibition after 10 hr). An obviously irrelevant immunotoxin (anti-trinitrophenol IgM-ricin A chain) had no significant cytotoxic effect on BL Ramos and non-BL Priess cells. An excess of D-galactose was shown previously to inhibit the 38-13 IgM from binding to the reactive glycolipid antigen bearing a terminal galactose. An excess of D-galactose (0.1 M) inhibited the cytotoxic effect of the two 38-13 immunotoxins, whereas it did not prevent the cytotoxic effect of the anti-trinitrophenol immunotoxin on the same trinitrophenol labeled target cells. These data suggest that the cytotoxic effect observed with 38-13 immunotoxins on non-BL Priess cells was mediated through their binding to a very low number of antigenic sites undetectable by conventional immunological methods. The main characteristics of 38-13 immunotoxins appear to be their fast kinetics of action and the very low number of antigenic sites required for the expression of their toxic effects. These properties could be related to the glycolipid nature of the reacting antigen. Such glycolipid antigens would represent valuable targets for therapeutic use of immunotoxins.
Asunto(s)
Antígenos de Neoplasias/análisis , Linfoma de Burkitt/inmunología , Glicoesfingolípidos/análisis , Proteínas de Plantas/toxicidad , Ricina/toxicidad , Anticuerpos Monoclonales , Línea Celular , Citometría de Flujo , Humanos , Inmunoglobulina M , Cinética , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Inactivadoras de Ribosomas Tipo 1RESUMEN
The kinetics of protein synthesis inhibition was studied in the choriocarcinoma-derived BeWo cell line treated with ricin and an immunotoxin (IT) constructed by linking a specific antibody to the A chain of ricin. The IT was specifically cytotoxic to BeWo and other choriocarcinoma cells. The multistep process underlying this kinetics was explored using two mathematical models where the protein synthesis-inactivation step is preceded by one or two processing rate-limiting steps. Theoretical curves were computed for different concentrations of toxic reagents. Two processing steps were found necessary to predict the duration of the observed latent period before initiation of protein synthesis inhibition. With this model, a satisfactory fit was obtained. A mathematical modeling of the intoxication induced by ricin or ricin A chain IT thus requires two processing steps to account for the data observed. In addition, the data suggest that the cytoplasmic internalization of ricin is a slow process compensated by an extremely fast enzymatic inactivation of ribosomal activity. This implies that in this and similar systems, endocytosis might not be involved in the cytoplasmic internalization of the few molecules of ricin A chain responsible for cell intoxication.
Asunto(s)
Supervivencia Celular/efectos de los fármacos , Coriocarcinoma/patología , Ricina/toxicidad , Neoplasias Uterinas/patología , Anticuerpos Monoclonales , Línea Celular , Femenino , Humanos , Cinética , Modelos Biológicos , Proteínas de Neoplasias/biosíntesis , EmbarazoRESUMEN
The molecular basis of cross-resistance to tumor necrosis factor (TNF) and Adriamycin has been investigated using the breast adenocarcinoma cell line MCF7/p, its Adriamycin-resistant counterpart MCF7AdrR, and the MDR1 gene-transduced MCF-7 cells (MCF7/MDR1). While the parental cell line MCF7/p was TNF-sensitive, MCF7AdrR was TNF-resistant. The TNF resistance exhibited by MCF7AdrR cells was not due to a lack of TNF receptor expression because both cell lines express comparable levels of p75 and p55 receptors as revealed by immunofluorescence analysis. NF-kappa B translocation, which is an essential transducing signal of the TNF-induced lysis pathway, does not appear to be involved in this resistance as assessed by gel shift experiments. In order to determine the role of MDR1 gene expression in the development of this cross-resistance, MCF7/p cells transfected by the MDR1 gene were examined. Our data showed that the expression of the MDR1 gene in these cells resulted in a relative resistance of these cells to Adriamycin without affecting their susceptibility to TNF killing. The implication of the manganese superoxide dismutase and endogenous TNF expression in the cross-resistance by MCF7AdrR cells to Adriamycin and TNF has also been investigated. Northern blot analysis indicated that following TNF stimulation, the expression of 4-kilobase and 1-kilobase manganese superoxide dismutase mRNAs were 9- to 10-fold induced in MCF7AdrR cells as compared to MCF7/p and MCF7/MDR1 cells. This suggests a possible involvement of this enzyme in the Adriamycin-induced resistance to TNF. Although TNF-treatment of MCF7/p and MDR-cells induced endogenous TNF expression in these cells, the level of mRNA induction was selectively enhanced in MCF7AdrR cells (7- to 8-fold greater in MCF7AdrR cells as compared to MCF7/p and MCF7/MDR1 cells). Collectively, these data indicate that the expression of the MDR1 gene in MCF7/p cells following gene transfection is not sufficient for the acquisition of TNF resistance by MCF7/MDR1 cells. Furthermore, our data provide the first evidence that Adriamycin-induced resistance to TNF in MCF7AdrR cells may, in part, involve an overexpression of endogenous TNF and manganese superoxide dismutase genes.
Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Doxorrubicina/farmacología , Expresión Génica/genética , Superóxido Dismutasa/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Proteínas Portadoras/fisiología , Regulación hacia Abajo/fisiología , Resistencia a Medicamentos/genética , Resistencia a Medicamentos/fisiología , Ensayos de Selección de Medicamentos Antitumorales , Inducción Enzimática , Femenino , Regulación Enzimológica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Transferencia de Gen , Humanos , Glicoproteínas de Membrana/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal/fisiología , Superóxido Dismutasa/biosíntesis , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/biosíntesisRESUMEN
Gb3/CD77 is a glycolipid antigen, specifically expressed on two different B-cell populations, Burkitt's lymphoma and a subset of tonsillar B-lymphocytes located in germinal centers, which could be the normal counterpart of Burkitt cells. Both Gb3/CD77(+) populations have recently been shown to enter programmed cell death (apoptosis) readily. Here we show that verotoxin, also called Shiga-like toxin, which is known to bind to the carbohydrate moiety of Gb3/CD77, induces cell death in Gb3/CD77(+) Burkitt's lymphoma cells, not only by inhibiting protein synthesis as classically described but also through an additional mechanism, namely apoptosis. Furthermore a recombinant B-subunit of verotoxin, which carries only the binding property of the holotoxin, also induces apoptosis in Gb3/CD77(+) cells. Gb3/CD77 could thus represent the first example of a glycolipid antigen able to transduce a signal leading to apoptosis.
Asunto(s)
Antígenos CD/metabolismo , Apoptosis/fisiología , Toxinas Bacterianas/toxicidad , Linfoma de Burkitt/patología , Daño del ADN , Trihexosilceramidas/metabolismo , Antígenos CD/biosíntesis , Antígenos CD/efectos de los fármacos , Apoptosis/efectos de los fármacos , Toxinas Bacterianas/metabolismo , Linfoma de Burkitt/ultraestructura , Línea Celular , Supervivencia Celular/efectos de los fármacos , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/aislamiento & purificación , Humanos , Peso Molecular , Proteínas Recombinantes/toxicidad , Mapeo Restrictivo , Toxina Shiga I , Trihexosilceramidas/biosíntesis , Células Tumorales CultivadasRESUMEN
The histogenesis of Ewing's sarcoma, the second most frequent primary bone tumor in humans, remains controversial. Ten Ewing cell lines were analyzed by immunological methods. Surface antigens recognized on Ewing cells were found to be related to the neuroectoderm lineage. They included ganglioside GD2, a marker of neuroectodermal tissues and tumors, and an acidic glycolipid detected by monoclonal antibody HNK-1 in the nervous system. The P61 rat monoclonal antibody that reacts with a peptide moiety of neural cell adhesion molecule (N-CAM) and a rabbit antiserum raised to purified mouse N-CAM also stained Ewing cells. Flow cytometry analysis performed using these reagents allowed the definition of four distinct Ewing phenotypes: all reagents equally stained group 1 lines; group 2 lines were strongly reactive with anti-N-CAM reagents, by contrast with a fainter staining with HNK-1 and anti-GD2 antibodies; all reagents but P61 were strongly reactive with group 3 lines; in group 4, Ewing lines were stained by P61 but only poorly by the anti-N-CAM antiserum. Several antibodies to melanoma and neuroblastoma associated antigens including two monoclonal antibodies to the nerve growth factor receptor were also found to react with Ewing cells. By contrast, all antibodies detecting antigens specifically expressed in hematopoietic cell lineages were totally unreactive. HLA class II antigens were never detected while the level of expression of class I antigens varied to a large extent. Ewing cells are characterized by a specific t(11;22)(q23-24;q12) translocation also observed in neuroepithelioma, a neuroectodermal tumor. Thus, Ewing's sarcoma cells share antigenic and karyotypic features with derivatives of the neuroectoderm possibly indicating a related histogenesis.
Asunto(s)
Antígenos de Neoplasias/análisis , Antígenos de Superficie/análisis , Ectodermo/inmunología , Sarcoma de Ewing/inmunología , Antígenos de Superficie/genética , Moléculas de Adhesión Celular , Línea Celular , Gangliósidos/análisis , Humanos , Receptores de Superficie Celular/análisis , Receptores de Factor de Crecimiento Nervioso , Sarcoma de Ewing/etiología , Translocación GenéticaRESUMEN
Following immunizations of BALB/c mice with K562 cells, we have obtained seven original monoclonal antibodies (MoAbs): (a) One MoAb, GA3, defines an antigen essentially restricted to the red cell series. This antigen is expressed on immature erythroblasts but is not detectable on the surface of early and late erythroid progenitors. GA3 MoAb immunoprecipitates a Mr 105,000 glycoprotein on K562 cells. (b) Two MoAbs, 14B6 and 12B1, react with cells of the monocytic series. MoAb 14B6, which also faintly stains platelets, is reactive with immature myeloid cells and the majority of hematopoietic progenitors. The 14B6 antigen has been immunoprecipitated from 12-O-tetradecanoylphorbol-13-acetate treated K562 cells as a Mr 130,000-100,000 protein. Antigen 12B1 is expressed only on cultured monocyte/macrophages and is restricted to a subpopulation of monocytes and to follicular dendritic cells. It is not detected on hematopoietic progenitors. Immunoprecipitation experiments performed on 12-O-tetradecanoylphorbol-13-acetate treated K562 cells revealed a glycoprotein with a molecular weight of 93,000-86,000. (c) Two anti-K562 MoAbs, CF4 and HE10, recognize a myeloid differentiation antigen expressed from the granulomonocytic colony forming unit stage to polymorphonuclear neutrophils. These MoAbs detect an apparently original glycolipid moiety distinct from LeX. (d) Two MoAbs recognize antigens expressed on the granulomonocytic series. 2E1 recognizes the monocyte low affinity Fc receptor (Mr 40,000) and defines a new cluster of myeloid differentiation (CDw32). The antigen is expressed on a small portion of immature hematopoietic progenitors. 8F5 identifies a Mr 95,000 protein which is also present on plasma cells. In some experiments, it is detected on erythroid colony forming unit analysis. Immunizations with K562 cells thus resulted in the production of antibodies recognizing antigens of the monocytic, granulocytic, as well as erythroid series. However, three of them are also detected on hematopoietic progenitors.
Asunto(s)
Anticuerpos Monoclonales , Antígenos de Diferenciación/análisis , Inmunización Pasiva , Animales , Línea Celular , Femenino , Fluorometría , Células Madre Hematopoyéticas/inmunología , Ratones , Ratones Endogámicos BALB C , Peso Molecular , Acetato de Tetradecanoilforbol/farmacologíaRESUMEN
The expression and function of CD95 and CD40 were investigated in malignant cells from EBV-positive undifferentiated nasopharyngeal carcinomas (NPCs). Large amounts of CD95 and CD40 expression were detected in 15 of 16 EBV-positive NPC specimens. In contrast, CD95 was not detected in two biopsies from patients with EBV-negative differentiated NPCs. We tested whether the CD95 apoptotic pathway was functional in NPC cells by treating two EBV-positive NPC tumor lines in vitro with a CD95 agonist. In both cases, NPC cells were extremely susceptible to CD95-mediated apoptosis, despite strong constitutive expression of Bcl-x. Combined CD40 and CD95 stimulation was used to investigate the possible anti-apoptotic activity mediated by CD40. The CD40 receptor was activated by incubating NPC cells with murine L cells producing CD154, the CD40 ligand. This treatment resulted in a strong inhibition of CD95-related cytotoxicity. Such an anti-apoptotic effect of CD40 is well known for B lymphocytes, but has not previously been reported for epithelial cells. These data suggest that NPC tumor-infiltrating lymphocytes, which often produce the CD40 ligand in situ, may increase the survival of malignant cells, thereby enhancing tumor growth in patients.
Asunto(s)
Apoptosis , Antígenos CD40/fisiología , Herpesvirus Humano 4/aislamiento & purificación , Neoplasias Nasofaríngeas/patología , Receptor fas/fisiología , Adolescente , Antígenos CD40/análisis , Supervivencia Celular , Femenino , Humanos , Neoplasias Nasofaríngeas/virología , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Células Tumorales Cultivadas , Proteína bcl-X , Receptor fas/análisisRESUMEN
Serum antibodies reacting with the tumor suppressor protein p53 have been detected previously in cancer patients with a variety of neoplasms. Two initial (although insufficient) prerequisites for a B-cell response to occur have been proposed: p53 protein accumulation in the tumor or a mutant p53 gene, or both. We have examined 65 esophageal cancer cases (42 from Guangzhou and Shenyang, People's Republic of China, and 23 from Paris, France) to obtain a prevalence estimate of anti-p53 antibodies for this type of cancer and to define the relationship of p53 tumor status to B-cell immune response. Sera were analyzed in a triplicate assay (enzyme-linked immunoassay, immunoprecipitation, and immunoblot) for anti-p53 antibodies. Tumor DNA was screened for mutations in exons 5-8, and tumor tissue was examined by immunohistochemistry for abnormal p53 protein accumulation. p53 mutations were found in 36 (58%) of 62 cases analyzed. Sixteen patients (25%) had circulating antibodies to the tumor suppressor protein. All but two (88%) of the tumors from seropositive cases had a mutation in the DNA binding region of the p53 gene, and with one exception, these tumors also showed nuclear accumulation of the p53 protein. In contrast, tumor mutations were found in just 22 (46%) of the 48 individuals in whom we did not detect anti-p53 antibodies. Among the 22 seronegative cases for which we found no tumor mutations, 11 revealed p53 protein accumulation by immunohistochemical analysis. Thus, circulating anti-p53 antibodies may be present in one-fourth of esophageal cancer patients, most of whom also would be expected to have a p53 gene mutation in their tumors. Patients without such mutations appear considerably less likely to mount a B-cell response to the p53 tumor suppressor protein than those that do (P < 0.01).
Asunto(s)
Anticuerpos/sangre , Neoplasias Esofágicas/inmunología , Genes p53 , Mutación , Proteína p53 Supresora de Tumor/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Esofágicas/genética , Femenino , Humanos , Masculino , Persona de Mediana EdadRESUMEN
Comprehensive Cancer Centres (CCCs) serve as critical drivers for improving cancer survival. In Europe, we have developed an Excellence Designation System (EDS) consisting of criteria to assess "excellence" of CCCs in translational research (bench to bedside and back), with the expectation that many European CCCs will aspire to this status.
Asunto(s)
Instituciones Oncológicas , Neoplasias/terapia , Calidad de la Atención de Salud , Investigación Biomédica Traslacional , Instituciones Oncológicas/normas , Europa (Continente) , Humanos , Calidad de la Atención de Salud/normas , Investigación Biomédica Traslacional/métodos , Investigación Biomédica Traslacional/normasRESUMEN
Numerous studies have indicated that inactivation of p53 is one of the essential requirements for the unrestrained growth of tumoral cells. When the status of the p53 gene was examined in various types of lymphoid malignancies, mutations in p53 have been predominantly detected in Burkitt's lymphoma (BL) cells, therefore suggesting that alteration of p53 could specifically contribute to the malignant phenotype of these tumoral cells. In addition to mutations, functional inactivation of p53 can also occur through interaction of the wild-type gene product with various viral or cellular proteins. The cellular MDM2 protein, for example, is able to inhibit p53 tumor suppressor function by concealing its transactivation domain. Mdm2 gene amplification has been described in several types of sarcomas, resulting in overexpression of the MDM2 protein. In this study, we have examined the status of MDM2 and p53 in 20 BL cell lines. Four were found to contain wild-type p53 and to overexpress MDM2 protein. Within these BL cells, both molecules are physically associated since they can be co-precipitated and p53 is inactivated as cells neither arrest in G1 nor enter apoptosis following gamma-radiation. We also report that the high level of the MDM2 protein in BL cells is neither associated with an amplification of the mdm2 gene nor with an elevated level of RNA or an increased protein stability, but is rather due to an enhanced translation ability of the mdm2 RNA. These results indicate that in certain BL cells, overexpression of MDM2 protein regulated at the posttranscriptional level, induces an escape from p53-controlled cell growth.