Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
1.
Toxicol Appl Pharmacol ; 260(3): 232-40, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22414680

RESUMEN

Embryo implantation is a crucial step in human reproduction and depends on the timely development of a receptive endometrium. The human endometrium is unique among adult tissues due to its dynamic alterations during each menstrual cycle. It hosts the implantation process which is governed by progesterone, whereas 17ß-estradiol regulates the preceding proliferation of the endometrium. The receptors for both steroids are targets for drugs and endocrine disrupting chemicals. Chemicals with unwanted antigestagenic actions are potentially hazardous to embryo implantation since many pharmaceutical antiprogestins adversely affect endometrial receptivity. This risk can be addressed by human tissue-specific in vitro assays. As working basis we compiled data on chemicals interacting with the PR. In our experimental work, we developed a flexible in vitro model based on human endometrial Ishikawa cells. Effects of antiprogestin compounds on pre-selected target genes were characterized by sigmoidal concentration-response curves obtained by RT-qPCR. The estrogen sulfotransferase (SULT1E1) was identified as the most responsive target gene by microarray analysis. The agonistic effect of progesterone on SULT1E1 mRNA was concentration-dependently antagonized by RU486 (mifepristone) and ZK137316 and, with lower potency, by 4-nonylphenol, bisphenol A and apigenin. The negative control methyl acetoacetate showed no effect. The effects of progesterone and RU486 were confirmed on the protein level by Western blotting. We demonstrated proof of principle that our Ishikawa model is suitable to study quantitatively effects of antiprogestin-like chemicals on endometrial target genes in comparison to pharmaceutical reference compounds. This test is useful for hazard identification and may contribute to reduce animal studies.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Implantación del Embrión/efectos de los fármacos , Endometrio/efectos de los fármacos , Progesterona/metabolismo , Pruebas de Toxicidad/métodos , Adulto , Western Blotting , Células Cultivadas , Disruptores Endocrinos/toxicidad , Endometrio/metabolismo , Femenino , Antagonistas de Hormonas/toxicidad , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Progesterona/farmacología , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Sulfotransferasas/genética
3.
Oncogene ; 30(41): 4275-88, 2011 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-21532628

RESUMEN

We have studied the effects of carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) on tumor angiogenesis in murine ductal mammary adenocarcinomas. We crossed transgenic mice with whey acidic protein promoter-driven large T-antigen expression (WAP-T mice) with oncogene-induced mammary carcinogenesis with CEACAM1null mice, and with Tie2-Ceacam1 transgenics, in which the Tie2 promoter drives endothelial overexpression of CEACAM1 (WAP-T × CEACAM1(endo+) mice), and analyzed tumor vascularization, angiogenesis and vessel maturation in these mice. Using flat-panel volume computed tomography (fpVCT) and histology, we found that WAP-T × CEACAM1(endo+) mice exhibited enhanced tumoral vascularization owing to CEACAM1(+) vessels in the tumor periphery, and increased intratumoral angiogenesis compared with controls. In contrast, vascularization of CEACAM1null/WAP-T-derived tumors was poor, and tumor vessels were dilated, leaky and showed poor pericyte coverage. Consequently, the tumoral vasculature could not be visualized in CEACAM1null/WAP-T mice by fpVCT, and we observed poor organization of the perivascular extracellular matrix (ECM), accompanied by the accumulation of collagen IV-degrading matrix metalloproteinase 9(+) (MMP9(+)) leukocytes and stromal cells. Vascular instability and alterations in ECM structure were accompanied by a significant increase in pulmonary metastases in CEACAM1null/WAP-T mice, whereas only occasional metastases were observed in CEACAM1(+) hosts. In CEACAM1(+) hosts, intratumoral vessels did not express CEACAM1, but they were intact, extensively covered with pericytes and framed by a well-organized perivascular ECM. MMP9(+) accessory cells were largely absent. Orthotopic transplantation of primary WAP-T- and CEACAM1null/WAP-T tumors into all three mouse lines confirmed that a CEACAM1(+) host environment is a prerequisite for productive angiogenic remodeling of the tumor microenvironment. Hence, CEACAM1 expression in the tumor periphery determines the vascular phenotype in a tumor, whereas systemic absence of CEACAM1 interferes with the formation of an organized tumor matrix and intratumoral vessel maturation.


Asunto(s)
Antígenos CD/genética , Moléculas de Adhesión Celular/genética , Neoplasias Mamarias Experimentales/genética , Neovascularización Patológica/genética , Microambiente Tumoral/genética , Animales , Antígenos CD/metabolismo , Antígenos Transformadores de Poliomavirus/genética , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Western Blotting , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular/metabolismo , Matriz Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Leucocitos/metabolismo , Leucocitos/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Masculino , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de la Leche/genética , Neovascularización Patológica/metabolismo , Regiones Promotoras Genéticas/genética
4.
Reprod Toxicol ; 30(1): 89-93, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20172022

RESUMEN

The human endometrium is a fertility-determining factor. Its receptivity during the implantation window may be altered by chemicals. Since human embryo implantation is unique chemical risk assessment cannot be based solely on animal studies. We established a tissue-specific in vitro test based on human endometrial adenocarcinoma (Ishikawa) cells. Progesterone receptor (PR) was selected as primary target gene for estrogenic effects. Changes of mRNA levels were investigated by reverse transcription quantitative real-time PCR. Sigmoidal dose-response curves for up-regulation of PR mRNA and EC(50) values were established for 17beta-estradiol, diethylstilbestrol and the weak xenoestrogen bisphenol A. Nonylphenol also had a clear PR mRNA up-regulating effect. Several other chemicals were characterized as negative compounds. Among them was methoxyacetic acid which may produce false positive results in reporter gene assays. Up-regulation of PR protein by 17beta-estradiol, diethylstilbestrol, bisphenol A and nonylphenol was confirmed by Western Blotting.


Asunto(s)
Disruptores Endocrinos/toxicidad , Endometrio/efectos de los fármacos , Receptores de Progesterona/metabolismo , Reproducción/efectos de los fármacos , Pruebas de Toxicidad/métodos , Alternativas a las Pruebas en Animales , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Endometrio/metabolismo , Endometrio/patología , Femenino , Humanos , Técnicas In Vitro , ARN Mensajero/genética , Receptores de Progesterona/genética , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Pruebas de Toxicidad/normas
5.
Oncogene ; 28(39): 3456-67, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19648966

RESUMEN

The different pocket proteins are established as negative cell cycle regulators. With regard to the repressor functions of pocket proteins in cellular senescence, studies so far have mainly focused on pRb/p105. Here, we show that in a broad range of wild-type p53-expressing human tumor cells, and in human diploid fibroblasts, Rb2/p130 is the dominating pocket protein in replicative and in accelerated senescence. Senescent cells are arrested at the transition from late G1- to early S-phase, as indicated by the absence of S- and G2-phase cyclins A and B. Expression of cyclin A and entry into S-phase resumed after RNA interference-mediated knockdown of Rb2/p130. Activation of different upstream pathways by overexpression of either p21 or p16 converged on Rb2/p130 accumulation and induced senescence. In contrast, p53- or p21-negative cells treated with DNA-damaging agents failed to accumulate Rb2/p130 and to enter senescence. Our data suggest that Rb2/p130 is a member of the p53-p21 DNA damage signaling cascade, and represents the essential pocket protein family member needed for the induction of any type of senescence.


Asunto(s)
Reparación del ADN , Proteína p130 Similar a la del Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Quinasas p21 Activadas/metabolismo , Línea Celular Tumoral , Células Cultivadas , Senescencia Celular , Daño del ADN , Fibroblastos/metabolismo , Fase G1 , Humanos , Transducción de Señal
7.
Cell Death Differ ; 15(2): 398-407, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18049480

RESUMEN

Signaling mediated by activation of the transmembrane receptor Notch influences cell-fate decisions, differentiation, proliferation, and cell survival. Activated Notch reduces proliferation by altering cell-cycle kinetics and promotes differentiation in hematopoietic progenitor cells. Here, we investigated if the G(1) arrest and differentiation induced by activated mNotch1 are dependent on tumor suppressor p53, a critical mediator of cellular growth arrest. Multipotent wild-type p53-expressing (p53(wt)) and p53-deficient (p53(null)) hematopoietic progenitor cell lines (FDCP-mix) carrying an inducible mNotch1 system were used to investigate the effects of proliferation and differentiation upon mNotch1 signaling. While activated Notch reduced proliferation of p53(wt)-cells, no change was observed in p53(null)-cells. Activated Notch upregulated the p53 target p21(cip/waf) in p53(wt)-cells, but not in p53(null)-cells. Induction of the p21(cip/waf) gene by activated Notch was mediated by increased binding of p53 to p53-binding sites in the p21(cip/waf) promoter and was independent of the canonical RBP-J binding site. Re-expression of p53(wt) in p53(null) cells restored the inhibition of proliferation by activated Notch. Thus, activated Notch inhibits proliferation of multipotent hematopoietic progenitor cells via a p53-dependent pathway. In contrast, myeloid and erythroid differentiation was similarly induced in p53(wt) and p53(null) cells. These data suggest that Notch signaling triggers two distinct pathways, a p53-dependent one leading to a block in proliferation and a p53-independent one promoting differentiation.


Asunto(s)
Eritropoyesis , Células Madre Hematopoyéticas/fisiología , Células Madre Multipotentes/fisiología , Mielopoyesis , Receptor Notch1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proliferación Celular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Mutantes , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Transducción de Señal
8.
Oncogene ; 27(10): 1429-38, 2008 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-17891184

RESUMEN

Tumors that acquire resistance against death stimuli constitute a severe problem in the context of cancer therapy. To determine genetic alterations that favor the development of stress-resistant tumors in vivo, we took advantage of polyclonal tumors generated after retroviral infection of newborn Elambda-MYC mice, in which the retroviral integration acts as a mutagen to enhance tumor progression. Tumor cells were cultivated ex vivo and exposed to gamma-irradiation prior to their transplantation into syngenic recipients, thereby providing a strong selective pressure for pro-survival mutations. Secondary tumors developing from stress-resistant tumor stem cells were analysed for retroviral integration sites to reveal candidate genes whose dysregulation confer survival. In addition to the gene encoding the antiapoptotic Bcl-x(L) protein, we identified the gadd45b locus to be a novel common integration site in these tumors, leading to enhanced expression. In accord with a thus far undocumented role of Gadd45beta in tumorigenesis, we showed that NIH3T3 cells overexpressing Gadd45beta form tumors in NOD/SCID mice. Interestingly and differently to other known 'classical' antiapoptotic factors, high Gadd45beta levels did not protect against MYC-, UV- or gamma-irradiation-induced apoptosis, but conferred a strong and specific survival advantage to serum withdrawal.


Asunto(s)
Antígenos de Diferenciación/fisiología , Linfoma de Células B/metabolismo , Estrés Oxidativo/fisiología , Sobrevida/fisiología , Animales , Modelos Animales de Enfermedad , Linfoma de Células B/mortalidad , Linfoma de Células B/virología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Células 3T3 NIH
10.
Oncogene ; 26(15): 2185-90, 2007 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-17401427

RESUMEN

Since the very early days of p53 research, the gain of oncogenic activities by some mutant p53 proteins had been suspected as an important factor contributing to cancer progression. Considerable progress towards understanding the biology of mutant p53 has been made during the last years, the quintessence being the realization that the impact of mutant p53 proteins on the transcriptome of a tumor cell is much more global than previously thought. The emerging role of mutant p53 proteins in coordinating oncogenic signaling and chromatin modifying activities reveals an until now unsuspected function of these proteins as important modifiers of the oncogenic transcriptional response. Notwithstanding the fact that the sequence-specific DNA binding activity of mutant p53 proteins is impaired, they are still able to associate with specific loci on DNA by utilizing different mechanisms. The ability to associate with DNA appears to be crucial for the master role of mutant p53 proteins in coordinating oncogenic transcriptional responses.


Asunto(s)
Neoplasias/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , ADN/metabolismo , Humanos , Mutación , Transcripción Genética
11.
Oncogene ; 25(38): 5257-62, 2006 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-16936745

RESUMEN

Growth regulatory functions of Rb2/p130, which aim at a sustained arrest such as in quiescent or differentiated cells, qualify the protein also to act as a central regulator of growth arrest in cellular senescence. In this respect, Rb2/p130 functions are connected to signaling pathways induced by p53, which is a master regulator in cellular senescence. Here, we summarize the pathways, which specify pRb2/p130 to control this arrest program and distinguish its functions from those of pRb/p105.


Asunto(s)
Senescencia Celular/fisiología , Proteína de Retinoblastoma/fisiología , Proteína p130 Similar a la del Retinoblastoma/fisiología , Animales , Diferenciación Celular , Humanos , Lesiones Precancerosas/patología , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/fisiología
13.
Cell Death Differ ; 13(2): 324-34, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16123778

RESUMEN

To determine pathways cooperating with p53 in cellular senescence when the retinoblastoma protein (pRb)/p16INK4a pathway is defunct, we stably transfected the p16INK4a-negative C6 rat glioma cell line with a temperature-sensitive mutant p53. Activation of p53(Val-135) induces a switch in pocket protein expression from pRb and p107 to p130(Rb2) and stalls the cells in late G1, early S-phase at high levels of cyclin E. Maintenance of the arrest depends on the functions of p130(Rb2) repressing cyclin A. Inactivation of p53 in senescent cultures restores the pocket proteins to initial levels and initiates progression into S-phase, but the cells fail to resume proliferation, likely due to DNA damage becoming apparent in the arrest and activating apoptosis subsequent to the release from p53-dependent growth suppression. The data indicate that p53 can cooperate selectively with p130(Rb2) to induce cellular senescence, a pathway that may be relevant when the pRb/p16INK4a pathway is defunct.


Asunto(s)
Senescencia Celular , Proteína p130 Similar a la del Retinoblastoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Ciclina A/genética , Ciclina A/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Daño del ADN , Regulación Neoplásica de la Expresión Génica , Glioma/química , Glioma/patología , Glioma/fisiopatología , Ratones , Mutación , Fenotipo , Ratas , Proteína p130 Similar a la del Retinoblastoma/genética , Fase S , Transfección , Proteína p53 Supresora de Tumor/genética
14.
Oncogene ; 25(6): 940-53, 2006 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-16247471

RESUMEN

Following genotoxic stress, p53 either rescues a damaged cell or promotes its elimination. The parameters determining a specific outcome of the p53 response are largely unknown. In mouse fibroblasts treated with different irradiation schemes, we monitored transcriptional and non-transcriptional p53 activities and identified determinants that initiate an anti- or a pro-apoptotic p53 response within the context of p53-independent stress signaling. The primary, transcription-mediated p53 response in these cells is anti-apoptotic, while induction of p53-dependent apoptosis requires an additional, transcription-independent p53 activity, provided by high intracellular levels of activated p53. High intracellular levels of p53 were selectively generated after apoptosis-inducing high-dose UV-irradiation, and correlated with a strongly delayed upregulation of Mdm2. Following high-dose UV-irradiation, p53 accumulated in the cytoplasm and led to activation of the pro-apoptotic protein Bax. As p53-dependent Bax-activation is transcription-independent, we postulated that certain transcription-deficient mutant p53 proteins might also exert this activity. Indeed we found an endogenous, transcription-inactive mutant p53 that upon genotoxic stress induced Bax-activation in vivo. Our results demonstrate the impact and in vivo relevance of non-transcriptional mechanisms for wild-type and mutant p53-mediated apoptosis.


Asunto(s)
Apoptosis/genética , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/efectos de la radiación , Células 3T3/efectos de la radiación , Animales , Apoptosis/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/efectos de la radiación , Citosol/metabolismo , Relación Dosis-Respuesta en la Radiación , Rayos gamma , Péptidos y Proteínas de Señalización Intracelular , Ratones , Pruebas de Mutagenicidad , Mutación , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/efectos de la radiación , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/efectos de la radiación , Estrés Fisiológico , Proteína p53 Supresora de Tumor/genética , Rayos Ultravioleta , Regulación hacia Arriba , Proteína X Asociada a bcl-2/metabolismo , Proteína X Asociada a bcl-2/efectos de la radiación
15.
Anticancer Res ; 25(1A): 213-24, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15816541

RESUMEN

BACKGROUND: p53 is the most frequently mutated gene in human cancers and its functional integrity is an important predictor of treatment response and clinical outcome. The majority of mutations found in different types of cancer cluster within the DNA binding domain encoded by exons 5-8. In clinical specimens the functional status of p53 is, therefore, often evaluated by direct mutation analysis of exons 5-8 or indirectly by immunostaining and evaluation of the subcellular localization pattern or protein accumulation. MATERIALS AND METHODS: In a panel of glioma cell lines, the status of the P53 gene was analyzed by temperature gradient gel electrophoresis (TGGE) of exons 5-8 and direct sequencing of all p53 exons. The nuclear accumulation of p53 in unstressed cells was assessed by immunostaining. These data were correlated with stress induction of the p53 protein, nuclear translocation and a direct determination of the transcriptional activity of endogenous p53 protein and induction of p53 target genes. RESULTS: Our analysis demonstrated that a p53 gene mutation analysis limited to exons 5-8 and analysis of immunostaining patterns can not serve as reliable predictors of functional p53 in tumor cells. Conversely, in some presumably rare cases, the transcriptional activity of p53 may be retained in tumor cells in the presence of a mutation and a pathological immunostaining pattern. In our analysis, the constitutive dephosphorylation at Ser 376 correlated with the nuclear accumulation of p53, but not with the transcriptional activity of the protein. This suggests that constitutive dephosphorylation at Ser376 may be one of the factors determining stabilization of mutant and wild-type p53, which is frequently observed in glial tumors. CONCLUSION: The incidence of a dysfunctional p53 protein in gliomas may be higher than expected, based on a single parameter evaluation by mutation analysis of exons 5-8 or assessment of p53 accumulation and subcellular localization by immunostaining.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Glioma/genética , Glioma/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Electroforesis/métodos , Exones/genética , Genes p53/genética , Humanos , Mutación , Fosforilación , Fracciones Subcelulares/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
16.
Exp Clin Endocrinol Diabetes ; 111(3): 154-61, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12784189

RESUMEN

Recent research suggests a significant role for placental corticotropin-releasing hormone (CRH) in controlling human parturition. This paper describes the expression of CRH, CRH receptors 1 and 2, and CRH binding protein (CRH-BP) in gestational tissue in late pregnancy. Placenta, myometrium, decidua, and fetal membranes were collected after uncomplicated pregnancies at term caesarian section before the onset of labour. The localisation and mRNA expression of CRH, CRH receptors, and CRH-BP were studied by immunohistochemistry and reverse transcription (RT)-PCR. CRH receptors were detected in placenta, myometrium, decidua, and fetal membranes. We demonstrated for the first time the presence of CRH receptors on resident macrophages and on endothelial cells. CRH receptor 1 mRNA was detected in all tissues investigated by RT-PCR, whereas CRH receptor 2 mRNA was restricted to myometrium and decidua. CRH mRNA was widely expressed in all tissue under study. Novel findings are also presented on the expression of CRH-BP in the myometrium. This widespread expression of the CRH system in gestational tissue suggests a paracrine role for CRH in the birth process (e.g. effects on macrophages and endothelial cells).


Asunto(s)
Proteínas Portadoras/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Embarazo/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Proteínas Portadoras/genética , Hormona Liberadora de Corticotropina/genética , Decidua/citología , Decidua/metabolismo , Endotelio/citología , Endotelio/metabolismo , Membranas Extraembrionarias/citología , Membranas Extraembrionarias/metabolismo , Femenino , Edad Gestacional , Humanos , Inmunohistoquímica , Macrófagos/metabolismo , Miometrio/citología , Miometrio/metabolismo , Placenta/citología , Placenta/metabolismo , Tercer Trimestre del Embarazo , Receptores de Hormona Liberadora de Corticotropina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Prostaglandins Leukot Essent Fatty Acids ; 67(6): 397-404, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12468260

RESUMEN

Cytochrome P450 metabolism of arachidonic acid (AA) was investigated in human peritoneal macrophages which play a central role in chronic pelvic diseases in women (for example in endometriosis). The formation of eicosanoids other than prostaglandins (PGs) by these cells is still unknown. In non-activated macrophages obtained from women in the reproductive age, the main [(3)H]-AA metabolites coeluted with epoxyeicosatrienoic acids, dihydroxyeicosatrienoic acids (DHETs) and hydroxyeicosatetraenoic acids (HETEs) in reverse-phase HPLC. After zymosan activation a shift to PGs pathway was observed. Treatment with low doses of 2,3,7,8-tetrachlorodibenzo- p -dioxin increased the formation of a metabolite coeluting with 5,6-DHET. By gas chromatography/mass spectrometry 5,6-DHET (after beta-naphthoflavone induction), and 14,15-DHET as well as 11,12-DHET (after AA stimulation) were identified as major epoxygenase metabolites, respectively. The enantioselective formation of 12(S)-HETE was demonstrated by chiral-phase HPLC. Our findings demonstrate that non-activated peritoneal macrophages produce substantial amounts of bioactive cytochrome P450 metabolites of AA.


Asunto(s)
Ácido Araquidónico/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Fondo de Saco Recto-Uterino/patología , Macrófagos Peritoneales/metabolismo , Ácido Araquidónico/análisis , Ácido Araquidónico/química , Cromatografía Líquida de Alta Presión , Femenino , Cromatografía de Gases y Espectrometría de Masas , Humanos , Ácidos Hidroxieicosatetraenoicos/análisis , Ácidos Hidroxieicosatetraenoicos/química , Ácidos Hidroxieicosatetraenoicos/metabolismo , Marcaje Isotópico , Macrófagos Peritoneales/enzimología , Conformación Molecular
18.
J Virol ; 75(22): 10593-602, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11602701

RESUMEN

Protective immunity of BALB/c mice immunized with simian virus 40 (SV40) large T antigen (TAg) against SV40-transformed, TAg-expressing mKSA tumor cells is critically dependent on both CD8(+) and CD4(+) T lymphocytes. By depleting mice of T-cell subsets at different times before and after tumor challenge, we found that at all times, CD4(+) and CD8(+) cells both were equally important in establishing and maintaining a protective immune response. CD4(+) cells do not contribute to tumor eradication by directly lysing mKSA cells. However, CD4(+) lymphocytes provide help to CD8(+) cells to proliferate and to mature into fully active cytotoxic T lymphocytes (CTL). Depletion of CD4(+) cells by a single injection of CD4-specific monoclonal antibody at any time from directly before injection of the vaccinating antigen to up to 7 days after tumor challenge inhibited the generation of cytolytic CD8(+) lymphocytes. T helper cells in this system secrete the typical Th-1 cytokines interleukin 2 (IL-2) and gamma interferon. Because in this system TAg-specific CD8(+) cells secrete only minute amounts of IL-2, it appears that T helper cells provide these cytokines for CD8(+) T cells. Moreover, this helper effect of CD4(+) T cells in mKSA tumor rejection in BALB/c mice does not simply improve the activity of TAg-specific CD8(+) CTL but actually enables them to mature into cytolytic effector cells. Beyond this activity, the presence of T helper cells is necessary even in the late phase of tumor cell rejection in order to maintain protective immunity. However, despite the support of CD4(+) T helper cells, the tumor-specific CTL response is so weak that only at the site of tumor cell inoculation and not in the spleen or in the regional lymph nodes can TAg-specific CTL be detected.


Asunto(s)
Antígenos Transformadores de Poliomavirus/inmunología , Linfocitos T CD4-Positivos/inmunología , Rechazo de Injerto , Virus 40 de los Simios/inmunología , Linfocitos T Citotóxicos/inmunología , Infecciones Tumorales por Virus/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Femenino , Inmunización , Inmunoterapia , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Depleción Linfocítica , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Infecciones Tumorales por Virus/terapia
19.
Mol Cell Biol ; 21(7): 2581-93, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11259605

RESUMEN

Metabolic labeling of primate cells revealed the existence of phosphorylated and hypophosphorylated DNA polymerase alpha-primase (Pol-Prim) populations that are distinguishable by monoclonal antibodies. Cell cycle studies showed that the hypophosphorylated form was found in a complex with PP2A and cyclin E-Cdk2 in G1, whereas the phosphorylated enzyme was associated with a cyclin A kinase in S and G2. Modification of Pol-Prim by PP2A and Cdks regulated the interaction with the simian virus 40 origin-binding protein large T antigen and thus initiation of DNA replication. Confocal microscopy demonstrated nuclear colocalization of hypophosphorylated Pol-Prim with MCM2 in S phase nuclei, but its presence preceded 5-bromo-2'-deoxyuridine (BrdU) incorporation. The phosphorylated replicase exclusively colocalized with the BrdU signal, but not with MCM2. Immunoprecipitation experiments proved that only hypophosphorylated Pol-Prim associated with MCM2. The data indicate that the hypophosphorylated enzyme initiates DNA replication at origins, and the phosphorylated form synthesizes the primers for the lagging strand of the replication fork.


Asunto(s)
ADN Primasa/genética , Replicación del ADN , Animales , Anticuerpos Monoclonales/inmunología , Línea Celular , ADN Primasa/inmunología , Humanos , Isoenzimas/genética , Isoenzimas/inmunología , Microscopía Confocal , Fosforilación
20.
Genes Dev ; 15(3): 286-93, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11159909

RESUMEN

This study describes a tumor progression model for ductal pancreatic cancer in mice overexpressing TGF-alpha. Activation of Ras and Erk causes induction of cyclin D1-Cdk4 without increase of cyclin E or PCNA in ductal lesions. Thus, TGF-alpha is able to promote progression throughout G1, but not S phase. Crossbreeding with p53 null mice accelerates tumor development in TGF-alpha transgenic mice dramatically. In tumors developing in these mice, biallelic deletion of Ink4a/Arf or LOH of the Smad4 locus is found suggesting that loci in addition to p53 are involved in antitumor activities. We conclude that these genetic events are critical for pancreatic tumor formation in mice. This model recapitulates pathomorphological features and genetic alterations of the human disease.


Asunto(s)
Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas , Factor de Crecimiento Transformador alfa/genética , Animales , Proteínas Portadoras/genética , Ciclina D1/genética , Quinasa 4 Dependiente de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Fase G1/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Invasividad Neoplásica , Neoplasias Pancreáticas/patología , Fenotipo , Fase S/fisiología , Factor de Crecimiento Transformador alfa/biosíntesis , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...