Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Intervalo de año de publicación
1.
Phys Med Biol ; 69(12)2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38815610

RESUMEN

Objective. The distribution of hypoxia within tissues plays a critical role in tumor diagnosis and prognosis. Recognizing the significance of tumor oxygenation and hypoxia gradients, we introduce mathematical frameworks grounded in mechanistic modeling approaches for their quantitative assessment within a tumor microenvironment. By utilizing known blood vasculature, we aim to predict hypoxia levels across different tumor types.Approach. Our approach offers a computational method to measure and predict hypoxia using known blood vasculature. By formulating a reaction-diffusion model for oxygen distribution, we derive the corresponding hypoxia profile.Main results. The framework successfully replicates observed inter- and intra-tumor heterogeneity in experimentally obtained hypoxia profiles across various tumor types (breast, ovarian, pancreatic). Additionally, we propose a data-driven method to deduce partial differential equation models with spatially dependent parameters, which allows us to comprehend the variability of hypoxia profiles within tissues. The versatility of our framework lies in capturing diverse and dynamic behaviors of tumor oxygenation, as well as categorizing states of vascularization based on the dynamics of oxygen molecules, as identified by the model parameters.Significance. The proposed data-informed mechanistic method quantitatively assesses hypoxia in the tumor microenvironment by integrating diverse histopathological data and making predictions across different types of data. The framework provides valuable insights from both modeling and biological perspectives, advancing our comprehension of spatio-temporal dynamics of tumor oxygenation.


Asunto(s)
Modelos Biológicos , Oxígeno , Microambiente Tumoral , Oxígeno/metabolismo , Humanos , Hipoxia Tumoral , Neoplasias/metabolismo , Neoplasias/fisiopatología , Neoplasias/irrigación sanguínea , Hipoxia de la Célula , Hipoxia/metabolismo , Hipoxia/fisiopatología
2.
Oncogene ; 25(40): 5475-84, 2006 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-16652157

RESUMEN

The E1A-targeted transcription factor E4F1 is a key player in the control of mammalian embryonic and somatic cell proliferation and survival. Mouse embryos lacking E4F die at an early developmental stage, whereas enforced expression of E4F1 in various cell lines inhibits cell cycle progression. E4F1-antiproliferative effects have been shown to depend on its capacity to repress transcription and to interact with pRb and p53. Here we show that full-length E4F1 protein (p120(E4F1)) but not its E1A-activated and truncated form (p50(E4F1)), interacts directly in vitro and in vivo with the LIM-only protein FHL2, the product of the p53-responsive gene FHL2/DRAL (downregulated in rhabdomyosarcoma Lim protein). This E4F1-FHL2 association occurs in the nuclear compartment and inhibits the capacity of E4F1 to block cell proliferation. Consistent with this effect, ectopic expression of FHL2 inhibits E4F1 repressive effects on transcription and correlates with a reduction of nuclear E4F1-p53 complexes. Overall, these results suggest that FHL2/DRAL is an inhibitor of E4F1 activity. Finally, we show that endogenous E4F1-FHL2 complexes form in U2OS cells upon UV-light-induced nuclear accumulation of FHL2.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Proteínas Musculares/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular , Proteínas E4 de Adenovirus/metabolismo , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas con Homeodominio LIM , Ratones , Células 3T3 NIH , Unión Proteica , Proteínas Represoras/química , Transducción de Señal , Factores de Transcripción/química , Transcripción Genética , Transfección , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas , Rayos Ultravioleta
3.
Oncogene ; 20(31): 4115-27, 2001 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-11464278

RESUMEN

The expression of cyclin E and that of a few other bona fide cell cycle regulatory genes periodically oscillates every cycle in proliferating cells. Although numerous experiments have documented the role of E2F sites and E2F activities in the control of these genes as cells exit from G(0) to move through the initial G(1)/S phase transition, almost nothing is known on the role of E2Fs during the subsequent cell cycles. Here we show that a variant E2F-site that is part of the Cyclin E Repressor Module (CERM) (Le Cam et al., 1999b) accounts for the periodic down regulation of the cyclin E promoter observed between the exit from mitosis until the mid/late G(1) phase in exponentially cycling cells. This cell cycle-dependent repression correlates with the periodic binding of an atypical G(1)-specific high molecular weight p107-E2F complex (Cyclin E Repressor Complex: CERC2) that differs in both size and DNA binding behaviors from known p107-E2F complexes. Notably, affinity purified CERC2 displays a TSA-sensitive histone deacetylase activity and, consistent with this, derepression of the cyclin E promoter by trichostatin A depends on the CERM element. Altogether, this shows that the cell cycle-dependent control of cyclin E promoter in cycling cells is embroiled in acetylation pathways via the CERM-like E2F element.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Ciclina E/genética , Proteínas de Unión al ADN , Regulación hacia Abajo , Mitosis/genética , Ciclo Celular , Cromatografía de Afinidad , ADN , Factores de Transcripción E2F , Histona Desacetilasas/metabolismo , Humanos , Células K562 , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Proteínas Represoras/aislamiento & purificación , Proteínas Represoras/metabolismo , Proteína 1 de Unión a Retinoblastoma , Factores de Transcripción/metabolismo
4.
Proc Natl Acad Sci U S A ; 97(14): 7738-43, 2000 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-10869426

RESUMEN

The retinoblastoma protein pRB is involved in the transcriptional control of genes essential for cell cycle progression and differentiation. pRB interacts with different transcription factors and thereby modulates their activity by sequestration, corepression, or activation. We report that pRB, but not p107 and p130, binds to and facilitates repression by p120(E4F), a ubiquitously expressed GLI-Kruppel-related protein identified as a cellular target of E1A. The interaction involves two distinct regions of p120(E4F) and the C-terminal part of pRB. In vivo pRB-p120(E4F) complexes can only be detected in growth-arrested cells, and accordingly contain the hypophosphorylated form of pRB. Repression of an E4F-responsive promoter is strongly increased by combined expression of p120(E4F) and pRB, which correlates with pRB-dependent enhancement of p120(E4F) binding activity. Elevated levels of p120(E4F) have been shown to block growth of mouse fibroblasts in G(1). We find this requires pRB, because RB(-/-) fibroblasts are significantly less sensitive to excess p120(E4F).


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Proteínas E4 de Adenovirus/metabolismo , Proteínas Represoras/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteínas E4 de Adenovirus/genética , Animales , Sitios de Unión , División Celular , Inhibidores de Crecimiento , Ratones , Mutación , Unión Proteica , Proteínas Represoras/genética , Dedos de Zinc
5.
FEBS Lett ; 471(1): 29-33, 2000 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-10760507

RESUMEN

The bipartite repressor elements, termed cell cycle-dependent element (CDE)/cell cycle regulatory element (CCRE)-cell cycle homology region (CHR) control the growth-dependent transcription of the cyclin A, cdc25C, cdc2 genes. Here, we have identified a functional element displaying the signature of the CDE-CHR in the promoter of the mouse RB2 (p130) gene, encoding the retinoblastoma protein family (pRB)-related protein p130. This element locates close to the major transcription start site where it makes major groove contacts with proteins that can be detected in a cellular context using in vivo genomic footprinting techniques. Inactivation of either the CDE or CHR sequence strongly up-regulates the p130 promoter activity in exponentially growing cells, a situation where endogenous p130 gene expression is almost undetectable. Electrophoretic mobility shift assays suggest that two different protein complexes bind independently to the p130 CDE and CHR elements, and that the protein(s) bound to the CDE might be related to those bound on cyclin A and cdc2 promoters.


Asunto(s)
Regulación de la Expresión Génica , Fosfoproteínas/genética , Regiones Promotoras Genéticas , Proteínas , Animales , Secuencia de Bases , Clonación Molecular , ADN , Proteínas de Unión al ADN/metabolismo , Genes cdc , Humanos , Ratones , Datos de Secuencia Molecular , Mutación , Proteína p130 Similar a la del Retinoblastoma , Homología de Secuencia de Ácido Nucleico , Transcripción Genética , Regulación hacia Arriba
6.
Genes Chromosomes Cancer ; 28(1): 126-30, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10738311

RESUMEN

E2F transcription factors (E2F1 to 6) are central players in the control of animal cell proliferation as regulators of genes involved in cell cycle progression and in transformation. In this report, we have investigated the potential involvement of the E2F5 gene in tumorigenesis. We show that E2F5 can promote the formation of morphologically transformed foci in primary baby rat kidney cells (BRK) when it is overexpressed in the presence of its heterodimeric partner DP1 and activated RAS. This suggests that E2F5 behaves like a MYC-type cooperating oncogene in functional assays, prompting us to monitor potential amplifications of the E2F5 gene in primary human tumors. We mapped the human E2F5 gene to 8q21.1-21.3 equidistant from the MOS (8q12) and MYC (8q24) oncogenes. Since the long arm of chromosome 8 is frequently the site of increased gene copy number (ICN) in breast cancer, we screened 442 breast tumor DNAs for gains of E2F5, MOS, and MYC genes. The three genes showed ICN, albeit at variable incidence and levels of amplification, with the ICN of E2F5 occurring concomitantly with those of MOS and/or MYC in almost half of the cases. Moreover, a marked increase of the 2. 5-kb E2F5 transcript was also detected in some tumors and tumor cell lines. In conclusion, the evidence that sustained unregulated expression of E2F5 can cooperate with other oncogenes to promote cell transformation in functional assays, together with the detection of chromosomal amplifications and overexpressions of the E2F5 gene in breast tumors, provides the first indications that E2F5 deregulation may have a role in human tumor development.


Asunto(s)
Neoplasias de la Mama/genética , Amplificación de Genes/genética , Oncogenes/genética , Factores de Transcripción/genética , Animales , Factor de Transcripción E2F5 , Dosificación de Gen , Humanos , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/biosíntesis , Células Tumorales Cultivadas
7.
J Biol Chem ; 275(2): 949-58, 2000 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-10625632

RESUMEN

The basic helix-loop-helix tal-1 gene plays a key role in hematopoiesis, and its expression is tightly controlled through alternative promoters and complex interactions of cis-acting regulatory elements. tal-1 is not expressed in normal T cells, but its transcription is constitutive in a large proportion of human T cell leukemias. We have previously described a downstream initiation of tal-1 transcription specifically associated with a subset of T cell leukemias that leads to the production of NH(2)-truncated TAL-1 proteins. In this study, we characterize the human promoter (promoter IV), embedded within a GC-rich region in exon IV, responsible for this transcriptional activity. The restriction of promoter IV usage is assured by a novel silencer element in the 3'-untranslated region of the human gene that represses its activity in erythroid but not in T cells. The silencer activity is mediated through binding of a tissue-specific nuclear factor to a novel protein recognition motif (designated tal-RE) in the silencer. Mutation of a single residue within the tal-RE abolishes both specific protein binding and silencing activity. Altogether, our results demonstrate that the tal-1 promoter IV is actively repressed in cells of the erythro-megakaryocytic lineage and that this repression is released in leukemic T cells, resulting in the expression of the tal-1 truncated transcript.


Asunto(s)
Proteínas de Unión al ADN/genética , Silenciador del Gen , Megacariocitos/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas , Factores de Transcripción , Células 3T3 , Regiones no Traducidas 5'/genética , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Sitios de Unión , Línea Celular , Secuencia de Consenso , Proteínas de Unión al ADN/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Exones , Células HeLa , Secuencias Hélice-Asa-Hélice , Humanos , Células K562 , Leucemia de Células T/genética , Ratones , Datos de Secuencia Molecular , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas Represoras , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Proteína 1 de la Leucemia Linfocítica T Aguda , Linfocitos T/metabolismo
8.
Oncogene ; 18(30): 4357-63, 1999 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-10439043

RESUMEN

The p16-cyclin D-pRB-E2F pathway is frequently deregulated in human tumors. This critical regulatory pathway controls the G1/S transition of the mammalian cell cycle by positive and negative regulation of E2F-responsive genes required for DNA replication. To assess the value of the transcription factors E2Fs as targets for antiproliferative strategies, we have initiated a program aiming to develop inhibitors targeting specifically these proteins in vitro and in vivo. The cellular activity of E2F is the result of the heterodimeric association of two families of proteins, E2Fs and DPs, which then bind DNA. Here, we use a two hybrid approach to isolate from combinatorial libraries peptide aptamers that specifically interact with E2Fs DNA binding and dimerization domains. One of these is a potent inhibitor of E2F binding activity in vitro and in mammalian fibroblasts, blocks cells in G1, and the free variable region from this aptamer has the same effect. Our experiments argue that the variable region of this aptamer is structured, and that it functions by binding E2F with a motif that resembles a DP heterodimerization region, and blocking E2F's association with DP. These results show that cell proliferation can be inhibited using genetically-selected synthetic peptides that specifically target protein-protein interaction motifs within cell cycle regulators. These results also emphasize the critical role of the E2F pathway for cell proliferation and might allow the design of novel antiproliferative agents targeting the cyclin/CDK-pRB-E2F pathway.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , División Celular , Proteínas de Unión al ADN , Inhibidores de Crecimiento/farmacología , Péptidos/farmacología , Factores de Transcripción/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Línea Celular , Cricetinae , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Factores de Transcripción E2F , Fibroblastos , Técnica del Anticuerpo Fluorescente , Fase G1 , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Proteína 1 de Unión a Retinoblastoma , Fase S , Tiorredoxinas/química , Factor de Transcripción DP1
9.
EMBO J ; 18(7): 1878-90, 1999 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10202151

RESUMEN

Transient induction of the cyclin E gene in late G1 gates progression into S. We show that this event is controlled via a cyclin E repressor module (CERM), a novel bipartite repressor element located near the cyclin E transcription start site. CERM consists of a variant E2F-binding site and a contiguous upstream AT-rich sequence which cooperate during G0/G1 to delay cyclin E expression until late G1. CERM binds the protein complex CERC, which disappears upon progression through G0-G1 and reappears upon entry into the following G1. CERC disappearance correlates kinetically with the liberation of the CERM module in vivo and cyclin E transcriptional induction. CERC contains E2F4/DP1 and a pocket protein, and sediments faster than classical E2F complexes in a glycerol gradient, suggesting the presence of additional components in a novel high molecular weight complex. Affinity purified CERC binds to CERM but not to canonical E2F sites, thus displaying behavior different from known E2F complexes. In cells nullizygous for members of the Rb family, CERC is still detectable and CERM-dependent repression is functional. Thus p130, p107 and pRb function interchangeably in CERC. Notably, the CERC-CERM complex dissociates prematurely in pRb-/- cells in correspondence with the premature expression of cyclin E. Thus, we identify a new regulatory module that controls repression of G1-specific genes in G0/G1.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Ciclina E/genética , Proteínas de Unión al ADN , Fase G1/genética , Factores de Transcripción/metabolismo , Células 3T3 , Animales , Secuencia de Bases , Sitios de Unión/genética , ADN/genética , ADN/metabolismo , Cartilla de ADN/genética , Factores de Transcripción E2F , Factor de Transcripción E2F4 , Fase G1/fisiología , Expresión Génica , Genes Reguladores , Sustancias Macromoleculares , Ratones , Datos de Secuencia Molecular , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Proteína 1 de Unión a Retinoblastoma , Factor de Transcripción DP1 , Factores de Transcripción/química
10.
Mol Cell Biol ; 18(11): 6679-97, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9774682

RESUMEN

The E2F transcription factors are essential regulators of cell growth in multicellular organisms, controlling the expression of a number of genes whose products are involved in DNA replication and cell proliferation. In Saccharomyces cerevisiae, the MBF and SBF transcription complexes have functions similar to those of E2F proteins in higher eukaryotes, by regulating the timed expression of genes implicated in cell cycle progression and DNA synthesis. The CDC6 gene is a target for MBF and SBF-regulated transcription. S. cerevisiae Cdc6p induces the formation of the prereplication complex and is essential for initiation of DNA replication. Interestingly, the Cdc6p homolog in Schizosaccharomyces pombe, Cdc18p, is regulated by DSC1, the S. pombe homolog of MBF. By cloning the promoter for the human homolog of Cdc6p and Cdc18p, we demonstrate here that the cell cycle-regulated transcription of this gene is dependent on E2F. In vivo footprinting data demonstrate that the identified E2F sites are occupied in resting cells and in exponentially growing cells, suggesting that E2F is responsible for downregulating the promoter in early phases of the cell cycle and the subsequent upregulation when cells enter S phase. Our data also demonstrate that the human CDC6 protein (hCDC6) is essential and limiting for DNA synthesis, since microinjection of an anti-CDC6 rabbit antiserum blocks DNA synthesis and CDC6 cooperates with cyclin E to induce entry into S phase in cotransfection experiments. Furthermore, E2F is sufficient to induce expression of the endogenous CDC6 gene even in the absence of de novo protein synthesis. In conclusion, our results provide a direct link between regulated progression through G1 controlled by the pRB pathway and the expression of proteins essential for the initiation of DNA replication.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN , Regulación de la Expresión Génica/genética , Regiones Promotoras Genéticas/genética , Proteínas de Saccharomyces cerevisiae , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/fisiología , División Celular/fisiología , Clonación Molecular , Ciclina E/metabolismo , ADN/biosíntesis , Huella de ADN , Replicación del ADN/genética , Factores de Transcripción E2F , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Datos de Secuencia Molecular , ARN/metabolismo , Proteína de Retinoblastoma/fisiología , Proteína 1 de Unión a Retinoblastoma , Proteínas de Schizosaccharomyces pombe , Factor de Transcripción DP1
11.
Oncogene ; 17(5): 585-94, 1998 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-9704924

RESUMEN

E2F-1 is the prototype of a family of transcription factors playing a central role in the control of cell proliferation and apoptosis. E2F DNA binding activity is down-regulated during cellular differentiation, which is correlated with cell division arrest. We report here that the expression of E2F-1 itself is down-regulated in the developing quail neural retina between embryonic days E8-E10. This event occurs just after the massive arrest of the quail neuroretina cell division (E7-E8). To gain further insight into the regulatory mechanisms monitoring E2F-1 expression in differentiating neurons, we have cloned the quail E2F-1 promoter. In vivo DNA footprintings of this promoter have shown that a number of potential SP-1 and C/EBP response elements are constitutively occupied in the entire quail neuroretina of E5 and E14, whereas the two consensus palindromic E2F binding sites are only protected at E5. This suggests that these E2F elements participate in down-regulation of E2F-1 gene expression during avian neuroretina development. CAT reporter assays have shown that E2F-1 in association with its partner DP-1 transactivates its own promoter, whereas p105Rb inhibits the E2F-1 promoter. Both E2F-1/DP-1 and p105Rh require the presence of the E2F binding sites to mediate their effects. However, experiments performed with deletion mutants of the promoter strongly suggest that other regions located upstream of the E2F binding sites also mediate part of the E2F-1 transactivating effect on its own promoter. Altogether, these results suggest that the down-regulation of E2F-1 gene expression in differentiating neurons could be due, in part, to the E2F/Rb complexes binding to the E2F-1 promoter.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Factores de Transcripción/genética , Células 3T3 , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células Cultivadas , Huella de ADN , ADN Complementario , Regulación hacia Abajo , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Neuronas , Regiones Promotoras Genéticas , Codorniz , Retina/citología , Retina/embriología , Proteína 1 de Unión a Retinoblastoma , Factor de Transcripción DP1
12.
Oncogene ; 16(11): 1373-81, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9525736

RESUMEN

Cyclin A is a positive regulatory component of kinases required for the progression through S phase and for the transition between the G2 and M phases of the cell division cycle. Previous studies have demonstrated that the promoter of its gene is under transcriptional repression in quiescent cells. Whereas the DNA sequences mediating this effect have been clearly delineated, the nature of the proteins acting in trans is still debated. Indirect observations suggest the involvement of proteins related to the retinoblastoma tumor suppressor protein (pRb). However, the precise role of these proteins has been difficult to assess, since most experiments designed to analyse their function have been carried out in transformed cell lines. Nevertheless, a current model has emerged whereby the role of the p130 protein would be restricted to resting and early G1 cells and p107, absent in quiescent cells, would be involved later in the control of the G1/S transition, whilst pRb would be effective throughout the cell cycle. We show here that cyclin A transcriptional inhibition is relieved in primary fibroblasts from pRb(-/-) embryos and not in fibroblasts from p13O(-/-), p107(-/-) or even p130(-/-)/p107(-/-) double mutant embryos. This suggests a unique role for pRb in controlling the extinction of specific genes in G0, providing thus the first example of non-overlapping functions achieved by the different pocket proteins.


Asunto(s)
Ciclina A/genética , Regulación hacia Abajo , Proteínas , Proteína de Retinoblastoma/metabolismo , Animales , Secuencia de Bases , Células Cultivadas , ADN , Ratones , Datos de Secuencia Molecular , Mutagénesis , Proteínas Nucleares/genética , Fosfoproteínas/genética , Proteína p107 Similar a la del Retinoblastoma , Proteína p130 Similar a la del Retinoblastoma , Transcripción Genética
13.
Int Arch Allergy Immunol ; 111(4): 372-5, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8957111

RESUMEN

Tobacco smoking induces an increased nonspecific IgE response. IgE synthesis is controlled by IL-4 and IFN-gamma. The effect of nicotine (10(-10) to 10(-5) M), one of the major components of tobacco smoke, were studied on IL-4 and IFN-gamma release by peripheral blood mononuclear cells from 12 allergic patients and 12 nonallergic subjects and 16 T cell clones stimulated by nonspecific agonists (phorbol myristate acetate and calcium ionophore). The release of IL-4 and IFN-gamma was measured by ELISA in supernatants after a 48-hour culture. Nicotine did not modify IL-4 and IFN-gamma release by peripheral blood mononuclear cells or T cell clones. The effects of tobacco smoke on IgE production are unlikely to change in the T cell phenotype by nicotine.


Asunto(s)
Calcimicina/farmacología , Carcinógenos/farmacología , Interferón gamma/metabolismo , Interleucina-4/metabolismo , Ionóforos/farmacología , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , Nicotina/farmacología , Linfocitos T/inmunología , Acetato de Tetradecanoilforbol/farmacología , Células Clonales , Ensayo de Inmunoadsorción Enzimática , Humanos , Hipersensibilidad/inmunología , Inmunoglobulina E/biosíntesis , Inmunofenotipificación , Leucocitos Mononucleares/efectos de los fármacos , Linfocitos T/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA