Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Eur Acad Dermatol Venereol ; 33 Suppl 8: 52-56, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31833606

RESUMEN

Evasion of immune recognition by the innate and acquired immune system is a major principle of tumour cells and belongs to the hallmarks of cancer. Immune checkpoint inhibitor-based cancer therapies targeting the co-inhibitory receptors CTLA-4 or PD-1 have received enormous scientific and clinical attention during the last few years, because of promising clinical results observed in the treatment of different cancer entities including melanoma and cutaneous squamous cell carcinoma. However, the enthusiasm about the effects of the immune checkpoint inhibitors is muted as only a subfraction of patients shows a stable clinical response. To predefine the patient cohorts that may benefit from immune checkpoint therapy, rigorous biomarker analyses, which predict the response to these novel therapies, need to be performed. In addition, combination of immune checkpoint therapy with classical DNA-damaging chemotherapy or radiotherapy, which positively affects tumour neo-antigen presentation, appears to be a promising approach in optimizing patients' response. In this review, we briefly summarize important biomarkers for patient stratification and discuss the current limitations of these biomarkers in defining responders vs. non-responders to immune checkpoint therapy.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/terapia , Biomarcadores de Tumor , Humanos , Resultado del Tratamiento
2.
Transplant Proc ; 49(8): 1947-1955, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28923653

RESUMEN

BACKGROUND: The influence of immunosuppressants on hepatitis C virus (HCV) re-infection after liver transplantation, particularly mammalian target of rapamycin inhibitors, remains unclear. The aim of our study was to analyze the influence of everolimus (EVR) on HCV replication activity in the context of underlying molecular mechanisms, with focus on the pro-myelocytic leukemia protein (PML). METHODS: HCV viral load was recorded in 40 patients with post-transplant HCV re-infection before and 8 weeks after introduction of EVR. An HCV cell culture replicon system for genotype (GT) 1b, GT2b, and GT3a was used to compare the influence of EVR on HCV replication for the respective genotypes in vitro. Fluorescence-activated cell-sorting analysis was used to test for effects on cell proliferation. PML expression was silenced with the use of small hairpin RNA constructs, and PML expression was quantified by means of quantitative real-time polymerase chain reaction. RESULTS: In patients with HCV, the viral load of GT1a and GT1b was hardly affected by EVR, whereas the viral load was reduced in patients with GT2a (P ≤ .0001) or GT3 infection (P ≤ .05). In vitro EVR impairs HCV replication activity of GT2a and GT3a up to 60% (P ≤ .0005), whereas in GT1b cells, HCV replication activity is increased by 50% (P ≤ .005). Replicon cell viability was not impaired. HCV replication activity is impaired in the absence of PML, which can be reversed by overexpression of PML isoforms. Furthermore, in the absence of PML, the effect of EVR on HCV replication activity is nearly abrogated. CONCLUSIONS: The mammalian target of rapamycin inhibitor EVR influences HCV replication via PML. The herein presented results suggest a genotype-dependent benefit for an EVR-based immunosuppressive regimen in patients with GT2a or GT3 re-infection after liver transplantation.


Asunto(s)
Everolimus/farmacología , Hepacivirus/efectos de los fármacos , Hepatitis C Crónica/virología , Inmunosupresores/farmacología , Trasplante de Hígado , Sirolimus/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Genotipo , Hepacivirus/genética , Hepacivirus/fisiología , Humanos , Técnicas In Vitro , ARN Viral , Reacción en Cadena en Tiempo Real de la Polimerasa , Carga Viral/efectos de los fármacos
3.
Cell Death Differ ; 23(1): 110-22, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26113041

RESUMEN

Upon severe DNA damage a cellular signalling network initiates a cell death response through activating tumour suppressor p53 in association with promyelocytic leukaemia (PML) nuclear bodies. The deacetylase Sirtuin 1 (SIRT1) suppresses cell death after DNA damage by antagonizing p53 acetylation. To facilitate efficient p53 acetylation, SIRT1 function needs to be restricted. How SIRT1 activity is regulated under these conditions remains largely unclear. Here we provide evidence that SIRT1 activity is limited upon severe DNA damage through phosphorylation by the DNA damage-responsive kinase HIPK2. We found that DNA damage provokes interaction of SIRT1 and HIPK2, which phosphorylates SIRT1 at Serine 682 upon lethal damage. Furthermore, upon DNA damage SIRT1 and HIPK2 colocalize at PML nuclear bodies, and PML depletion abrogates DNA damage-induced SIRT1 Ser682 phosphorylation. We show that Ser682 phosphorylation inhibits SIRT1 activity and impacts on p53 acetylation, apoptotic p53 target gene expression and cell death. Mechanistically, we found that DNA damage-induced SIRT1 Ser682 phosphorylation provokes disruption of the complex between SIRT1 and its activator AROS. Our findings indicate that phosphorylation-dependent restriction of SIRT1 activity by HIPK2 shapes the p53 response.


Asunto(s)
Apoptosis/genética , Proteínas Portadoras/biosíntesis , Leucemia Promielocítica Aguda/genética , Proteínas Serina-Treonina Quinasas/biosíntesis , Sirtuina 1/biosíntesis , Proteína p53 Supresora de Tumor/biosíntesis , Acetilación , Proteínas Portadoras/genética , Línea Celular Tumoral , Daño del ADN/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Leucemia Promielocítica Aguda/patología , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Sirtuina 1/genética , Proteína p53 Supresora de Tumor/genética
4.
Cell Death Differ ; 16(2): 187-94, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18974774

RESUMEN

Homeodomain-interacting protein kinase 2 (HIPK2) is an emerging regulator of cell growth and apoptosis in various cell types, tissues and organisms. Previous work indicates that HIPK2 is a potential tumour suppressor and DNA damage-responsive kinase, which phosphorylation-dependently activates the apoptotic programme by engaging diverse downstream targets, including tumour suppressor p53 and the anti-apoptotic transcriptional corepressor C-terminal binding protein. The regulation of HIPK2, however, remained largely obscure. Recent studies show that HIPK2 activity is mainly controlled at the post-transcriptional level through targeted proteolysis. Caspase-dependent processing triggers HIPK2 hyperactivation, whereas the ubiquitin-proteasome system (UPS) keeps HIPK2 in check by targeting it for degradation. Both HIPK2 hyperactivation and HIPK2 degradation are under the control of transcription factor p53. Negative regulation of HIPK2 by the UPS is abolished in response to DNA damage, which facilitates HIPK2 stabilization and activation. Here we discuss these findings in the context of DNA damage signalling and tumour suppression.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis , Caspasas/metabolismo , Daño del ADN , Humanos , Proteínas Nucleares/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo
5.
Oncogene ; 26(11): 1557-66, 2007 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-16924230

RESUMEN

Apoptin, a protein of the chicken anemia virus (CAV), represents a novel potential anticancer therapeutic, because it induces apoptotic death specifically in tumor but not normal cells. The cellular localization appears to be crucial for apoptin's selective toxicity. In normal cells apoptin remains in the cytoplasm, whereas in transformed cells it migrates into the nucleus and kills the cell. However, the manner by which apoptin is able to distinguish between tumor and normal cells is unknown. Here, we report for the first time that apoptin interacts directly with the promyelocytic leukemia protein (PML) in tumor cells and accumulates in PML nuclear bodies (NBs), which are involved in apoptosis induction and viral replication. We also demonstrate that apoptin is sumoylated and that a sumoylation-deficient apoptin mutant is no longer recruited to PML-NBs, but localizes in the nuclear matrix. This mutant fails to bind PML, but can still induce apoptosis as efficiently as wild-type apoptin. Moreover, apoptin kills also PML-/- cells and promyelocytic leukemia cells with defective PML expression. Our results therefore suggest that apoptin kills tumor cells independently of PML and sumoylation, however, the interaction of apoptin with PML and small ubiquitin-like modifier (SUMO) proteins might be relevant for CAV replication.


Asunto(s)
Proteínas de la Cápside/metabolismo , Núcleo Celular/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Proteína SUMO-1/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Línea Celular , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Proteína de la Leucemia Promielocítica
6.
Proc Natl Acad Sci U S A ; 103(40): 14808-12, 2006 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-17003125

RESUMEN

Cajal bodies are nuclear subdomains that are involved in maturation of small ribonucleoproteins and frequently associate with small nuclear RNA and histone gene clusters in interphase cells. We have recently identified FADD-like IL-1beta-converting enzyme (FLICE) associated huge protein (FLASH) as an essential component of Cajal bodies. Here we show that FLASH associates with nuclear protein, ataxia-telangiectasia, a component of the cell-cycle-dependent histone gene transcription machinery. Reduction of FLASH expression by RNA interference results in disruption of the normal Cajal body architecture and relocalization of nuclear protein, ataxia-telangiectasia. Furthermore, FLASH down-regulation results in a clear reduction of histone transcription and a dramatic S-phase arrest of the cell cycle. Chromatin immunoprecipitation reveals that FLASH interacts with histone gene promoter sequences. These results identify FLASH as an important component of the machinery required for histone precursor mRNA expression and cell-cycle progression.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Histonas/genética , Fase S/fisiología , Transcripción Genética , Animales , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Regulación hacia Abajo/genética , Células HeLa , Histonas/metabolismo , Humanos , Ratones , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo
7.
Cell Death Differ ; 10(12): 1290-9, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12934066

RESUMEN

Promyelocytic leukaemia (PML) nuclear bodies (NBs) are macromolecular nuclear domains present in virtually every mammalian cell. PML nuclear bodies (PML-NBs) were functionally linked to various fundamental cellular processes, including transcriptional control, tumour suppression and apoptosis regulation. Supporting the important function of PML and its associated NBs in apoptosis regulation, several apoptotic regulators localise to PML-NBs, and cells from PML-deficient mice show severe apoptotic defects, including induction of genotoxic stress and death receptor CD95 (Fas/APO-1) activation. Based on the current literature, we hypothesise that PML-NBs regulate apoptosis through different molecular mechanisms, on the one hand by acting as macromolecular scaffolds for recruitment and post-translational modification of the apoptotic key regulator p53, and on the other by regulating the subcellular bioavailability and quality of some apoptotic signal transducers.


Asunto(s)
Apoptosis , Núcleo Celular/metabolismo , Núcleo Celular/patología , Proteínas de Neoplasias/fisiología , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Animales , Ratones , Modelos Biológicos , Fosforilación , Proteína de la Leucemia Promielocítica , Procesamiento Proteico-Postraduccional , Proteína SUMO-1/metabolismo , Transducción de Señal , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor , Receptor fas/metabolismo
8.
Int J Cancer ; 93(2): 185-91, 2001 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-11410864

RESUMEN

Here we investigated CD95-mediated JNK activation pathways and their physiological relevance by employing a variety of cell lines with deficiencies in individual signal transmitting proteins. JNK activation was completely dependent on the activation of caspases in type I and type II cells, as revealed by the inhibitory effects of the caspase inhibitors zVAD-fmk or the cowpoxvirus-encoded CrmA protein. Jurkat cells deficient in caspase-8 or expressing a dominant negative (DN) form of FADD were unable to induce JNK in response to CD95 ligation, indicating that these death-inducing signaling complex (DISC) proteins are required for signal transmission. Activation of caspases, JNK and apoptosis occurred with a markedly slower kinetics in cells expressing a DN version of ASK1, revealing an important contribution of ASK1 for these processes. A C-terminally truncated version of Daxx impaired CD95-mediated apoptosis without affecting the JNK signal. DN forms of FADD, MKK4 and MKK7 completely inhibited CD95-mediated JNK activation but remained without impact on cell killing, indicating that JNK activation is not required for the execution process of CD95-mediated cell killing.


Asunto(s)
Apoptosis , Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas Quinasas JNK Activadas por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Nucleares , Transducción de Señal/fisiología , Receptor fas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Caspasas/metabolismo , Proteínas Co-Represoras , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Células Jurkat , MAP Quinasa Quinasa 4 , MAP Quinasa Quinasa Quinasa 5 , Quinasas Quinasa Quinasa PAM/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Chaperonas Moleculares , Células Tumorales Cultivadas
9.
Biochim Biophys Acta ; 1518(1-2): 168-72, 2001 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-11267674

RESUMEN

HIPK2 (homeodomain-interacting protein kinase 2) is a CD95 binding partner in yeast. Its primary amino acid sequence is highly conserved between human and mouse. The highest HIPK2 mRNA expression is found in neuronal tissue. The HIPK2 gene is located on human chromosome 7q33-35 and the protein is mainly localized in the nucleus. HIPK2 has been described to play a role as a co-repressor for homeodomain transcription factors.


Asunto(s)
Proteínas Portadoras/genética , Cromosomas Humanos Par 7 , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/genética , Receptor fas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/metabolismo , Codón Iniciador , ADN Complementario , Femenino , Expresión Génica , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero , Homología de Secuencia de Aminoácido , Distribución Tisular , Células Tumorales Cultivadas
10.
Biochimie ; 82(12): 1123-7, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11120354

RESUMEN

Here we identified the human serine/threonine kinase HIPK2 as a novel member of the DYRK kinase subfamily. Alignment of several DYRK family proteins including the kinases minibrain, MJAK, PKY, the Dictyostelium kinase YakA and Saccharomyces YAK1 allowed the identification of several evolutionary conserved DYRK consensus motifs within the kinase domain. A lysine residue conserved between all DYRK kinase family members was found to be essential for the kinase function of HIPK2. Human HIPK2 was mapped to chromosome 7q32-q34 and murine HIPK2 to chromosome 6B, the homologue to human chromosome 7.


Asunto(s)
Proteínas Portadoras/genética , Cromosomas Humanos Par 7 , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/metabolismo , Secuencia Conservada , Evolución Molecular , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas Quinasas/metabolismo , Homología de Secuencia de Aminoácido , Quinasas DyrK
11.
Eur J Biochem ; 267(12): 3828-35, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10849002

RESUMEN

To test whether the behaviour of transcription factor NF-kappaB as a promoter or antagonist of apoptosis depends on the apoptotic stimulus, we determined the influence of NF-kappaB on cell killing elicited by a variety of inducers within a given cell type. Inhibition of NF-kappaB by genetic and pharmacological approaches rendered HeLa cells more susceptible to TNF-alpha-induced cell killing, but protected them almost completely from H2O2- and pervanadate-induced apoptosis. TNF-alpha was unable to protect HeLa from H2O2- and pervanadate-induced apoptosis and further enhanced the cytotoxicity induced by these two adverse agents. Supernatants from HeLa cells stably overexpressing a transdominant negative form of IkappaB-alpha selectively increased the cytotoxicity of TNF-alpha for HeLa cells, suggesting that the enhanced susceptibility of these cells can be attributed to one or more secretable factors. Supershift experiments showed that the various apoptotic stimuli induced the same subset of DNA-binding subunits. Therefore, the nature of the signals elicited by the respective death inducers determines whether NF-kappaB induction leads to apoptosis or survival, suggesting that the manipulation of NF-kappaB activity may provide a new approach to adjuvant therapy in cancer treatment.


Asunto(s)
Apoptosis/fisiología , FN-kappa B/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Activación Enzimática , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Interleucina-6/metabolismo , FN-kappa B/efectos de los fármacos , FN-kappa B/genética , Transfección , Factor de Necrosis Tumoral alfa/farmacología , Vanadatos/farmacología
12.
J Biol Chem ; 275(24): 18160-71, 2000 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-10849438

RESUMEN

In this study we identified tyrosine-phosphorylated Vav1 as an early point of integration between the signaling routes triggered by the T-cell receptor and CD28 in human T-cell leukemia cells. Costimulation resulted in a prolonged and sustained phosphorylation and membrane localization of Vav1 in comparison to T-cell receptor activation alone. T-cell stimulation induced the recruitment of Vav1 to an inducible multiprotein T-cell activation signaling complex at the plasma membrane. Vav1 activated the mitogen-activated protein kinases JNK and p38. The Vav1-mediated activation of JNK employed a pathway involving Rac, HPK1, MLK3, and MKK7. The costimulation-induced activation of p38 was inhibited by dominant negative forms of Vav1, Rac, and MKK6. Here we show that Vav1 also induces transcription factors that bind to the CD28RE/AP element contained in the interleukin-2 promoter. A detailed mutational analysis of Vav1 revealed a series of constitutively active and nonfunctional forms of Vav1. Almost all inactive versions were mutated in their Dbl homology domain and behaved as dominant negative mutants that impaired costimulation-induced activation of JNK, p38, and CD28RE/AP-dependent transcription. In contrast to NF-AT-dependent transcription, Vav1-mediated transcriptional induction of the CD28RE/AP element in the interleukin-2 promoter could only partially be inhibited by cyclosporin A, suggesting a dual role of Vav1 for controlling Ca(2+)-dependent and -independent events.


Asunto(s)
Antígenos CD28/metabolismo , Proteínas de Ciclo Celular , Interleucina-2/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transcripción Genética , Tirosina/metabolismo , Calcio/metabolismo , Activación Enzimática , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Células Jurkat , Modelos Biológicos , Fosforilación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-vav , Transducción de Señal , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos
13.
Oncogene ; 19(9): 1153-63, 2000 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-10713703

RESUMEN

Here we identify the hematopoietic proto-oncogene Vav1 as a caspase substrate during apoptosis in lymphoid cells. Cleavage of Vav1 is prevented by the caspase inhibitors zDEVD and zVAD as well as by expression of CrmA. Vav1 is cleaved in vivo at the evolutionary conserved caspase consensus cleavage site DLYD161C, generating the carboxy-terminal cleavage product Vav1p76 of intermediate stability. In vitro caspase assays reveal cleavage of Vav1 at position 161 either by apoptotic cell lysates or by recombinant caspase-3. Mutation of Asp 161 to Ala leads to the usage of the adjacent alternative cleavage sequence DQID150D. Mutation of both cleavage sites at position 150 and 161 protects Vav1 from caspase-mediated proteolysis in vitro and in vivo. The cleavage product Vav1p76 is capable of activating JNK in T-cells, but fails to induce the phosphorylation of p38/HOG1. Vav1p76 displays a diminished capacity to activate the transcription factors NF-AT, AP-1 and NF-kappaB, and thus completely fails to activate IL-2 transcription. Since Vav1 is essential for IL-2 production and plays a central role for cytoskeletal reorganization, its proteolytic inactivation during apoptosis affects multiple downstream targets.


Asunto(s)
Apoptosis , Caspasas/fisiología , Proteínas de Ciclo Celular , Interleucina-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Transcripción Genética , Proteínas Virales , Clorometilcetonas de Aminoácidos/farmacología , Apoptosis/genética , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Inhibidores de Caspasas , Fraccionamiento Celular , Sistema Libre de Células/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Activación Enzimática , Células HeLa , Humanos , Hidrólisis , Interleucina-2/genética , Interleucina-2/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos , Células Jurkat/citología , Células Jurkat/enzimología , Células Jurkat/metabolismo , Linfocitos/citología , Linfocitos/enzimología , Linfocitos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutagénesis Sitio-Dirigida , Oligopéptidos/farmacología , Fragmentos de Péptidos/fisiología , Fosforilación , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-vav , Serpinas/farmacología , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos
14.
Mol Cell Biol ; 20(7): 2556-68, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10713178

RESUMEN

The phosphorylation of IkappaB by the multiprotein IkappaB kinase complex (IKC) precedes the activation of transcription factor NF-kappaB, a key regulator of the inflammatory response. Here we identified the mixed-lineage group kinase 3 (MLK3) as an activator of NF-kappaB. Expression of the wild-type form of this mitogen-activated protein kinase kinase kinase (MAPKKK) induced nuclear immigration, DNA binding, and transcriptional activity of NF-kappaB. MLK3 directly phosphorylated and thus activated IkappaB kinase alpha (IKKalpha) and IKKbeta, revealing its function as an IkappaB kinase kinase (IKKK). MLK3 cooperated with the other two IKKKs, MEKK1 and NF-kappaB-inducing kinase, in the induction of IKK activity. MLK3 bound to components of the IKC in vivo. This protein-protein interaction was dependent on the central leucine zipper region of MLK3. A kinase-deficient version of MLK3 strongly impaired NF-kappaB-dependent transcription induced by T-cell costimulation but not in response to tumor necrosis factor alpha or interleukin-1. Accordingly, endogenous MLK3 was phosphorylated and activated by T-cell costimulation but not by treatment of cells with tumor necrosis factor alpha or interleukin-1. A dominant negative version of MLK3 inhibited NF-kappaB- and CD28RE/AP-dependent transcription elicited by the Rho family GTPases Rac and Cdc42, thereby providing a novel link between these GTPases and the IKC.


Asunto(s)
Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Antígenos CD28/metabolismo , Línea Celular , Activación Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Quinasa I-kappa B , Leucina Zippers , FN-kappa B/metabolismo , Fosforilación , Linfocitos T/metabolismo , Activación Transcripcional , Transfección , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
15.
J Immunol ; 163(10): 5617-23, 1999 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-10553091

RESUMEN

The transcription factor NF-kappa B is a key regulator of the cellular inflammatory and immune response. Therefore, components of the NF-kappa B-activating signaling pathways are frequent targets for antiinflammatory agents. This study shows that the sesquiterpene lactone parthenolide inhibits a common step in NF-kappa B activation by preventing the TNF-alpha-induced induction of I kappa B kinase (IKK) and IKK beta, without affecting the activation of p38 and c-Jun N-terminal kinase. Parthenolide impairs NF-kappa B-dependent transcription triggered by expression of TNFR-associated factor-2, mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEKK1), and NF-kappa B-inducing kinase. This compound also prevents activation of both IKKs and DNA binding of NF-kappa B induced by MEKK and NF-kappa B-inducing kinase. Parthenolide targets a component of the I kappa B kinase complex without directly inhibiting IKK alpha, IKK beta, or MEKK1. Therefore, this sesquiterpene lactone could serve as a lead compound for the development of antiinflammatory remedies and is suitable as a molecular tool, allowing the dissection of TNF-alpha-derived signaling pathways leading to the activation of NF-kappa B, c-Jun N-terminal kinase, and p38.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Proteínas I-kappa B/antagonistas & inhibidores , Quinasa 1 de Quinasa de Quinasa MAP , FN-kappa B/antagonistas & inhibidores , Sesquiterpenos/farmacología , Activación Enzimática/efectos de los fármacos , Células HeLa , Humanos , Quinasa I-kappa B , Proteínas I-kappa B/metabolismo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/metabolismo , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Quinasa de Factor Nuclear kappa B
16.
Cell Death Differ ; 6(9): 833-41, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10510465

RESUMEN

The inhibition of protein tyrosine phosphatases by pervanadate, a potent activator of B- and T-cells through the induction of tyrosine phosphorylation and downstream signaling events in different activation cascades, efficiently induced apoptosis in lymphoid cell lines. Pervanadate-elicited apoptosis could be blocked by the tyrosine kinase inhibitor herbimycin A. This apoptotic process involved the activation of caspases 3, 8 and 9, the induction of mitochondrial permeability transition, the release of cytochrome C and the fragmentation of chromosomal DNA. T-cells lacking the CD95 receptor or caspase-8 and T-cells stably overexpressing a transdominant negative form of the adaptor protein FADD were still susceptible to pervanadate-induced apoptosis, excluding the involvement of the CD95 system or other FADD-dependent death receptors. The apoptotic program initiated by the inhibition of tyrosine phosphatases did not require the presence of the tyrosine kinase p56lck or phosphatase CD45, whereas Bcl-2 overexpression protected T-cells from pervanadate-induced cytochrome C release, caspase-8 cleavage and apoptosis.


Asunto(s)
Apoptosis , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Transducción de Señal , Receptor fas/metabolismo , Caspasa 3 , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , Grupo Citocromo c/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Membranas Intracelulares/fisiología , Células Jurkat , Mitocondrias/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Células Tumorales Cultivadas , Vanadatos/farmacología
17.
Eur J Biochem ; 264(1): 132-9, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10447681

RESUMEN

Ligation of the CD95 receptor resulted in a transient increase of cellular tyrosine phosphorylation. The inhibition of protein tyrosine phosphatases by pervanadate, a potent activator of B cells and T cells through the induction of tyrosine phosphorylation and downstream signaling events in the activation cascade, antagonized CD95-triggered apoptosis. Pervanadate exerted its inhibitory effect only during the early phase of apoptosis prior to the CD95-induced decrease of the mitochondrial transmembrane potential. Inhibition of tyrosine phosphatases delayed the cleavage and activation of caspase-8 and caspase-3 and antagonized the tyrosine dephosphorylation of the CD95 receptor-associated phosphoproteins p61 and p89/92. In contrast, ligation of the tumor necrosis factor (TNF) receptor resulted in a continuous tyrosine dephosphorylation of cellular proteins. Pervanadate-induced tyrosine phosphorylation increased the TNF-alpha-induced cytotoxicity and NF-kappaB activation, suggesting that it stimulates early signaling events prior to the separation of the two signaling pathways.


Asunto(s)
Apoptosis/fisiología , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Receptor fas/fisiología , Secuencia de Bases , Línea Celular , Cartilla de ADN , Humanos , Células Jurkat , FN-kappa B/metabolismo , Fosforilación , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Tirosina/metabolismo
18.
Oncogene ; 18(3): 747-57, 1999 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-9989825

RESUMEN

Reactive oxygen species (ROS) play an important role in cell death induced by many different stimuli. This study shows that hydrogen peroxide-induced apoptosis in T-cells did not require tyrosine kinase p561ck, phosphatase CD45, the CD95 receptor and its associated Caspase-8. H2O2-triggered cell death led to the induced cleavage and activation of Caspase-3. Hydrogen peroxide-treatment of T-cells resulted in the formation of mitochondrial permeability transition pores, a rapid decrease of the mitochondrial transmembrane potential delta psi(m) and the release of Cytochrome C. Inhibition of the mitochondrial permeability transition by bongkrekic acid (BA), or interference with the mitochondrial electron transport system by rotenone or menadione prevented the cytotoxic effect of H2O2. Antimycin A, a mitochondrial inhibitor that increases the release of mitochondrial ROS (MiROS), enhanced apoptosis. Overexpression of Bcl-2 and the viral anti-apoptotic proteins BHRF-1 and E1B 19K counteracted H2O2-induced apoptosis. Pharmacological and genetic inhibition of transcription factor NF-kappaB protected cells from hydrogen peroxide-elicited cell death. This detrimental effect of NF-kappaB mediating hydrogen peroxide-induced cell death presumably relies on the induced expression of death effector genes such as p53, which was NF-kappaB-dependently upregulated in the presence of H2O2.


Asunto(s)
Apoptosis , Peróxido de Hidrógeno/farmacología , Proteínas I-kappa B , FN-kappa B/metabolismo , Receptor fas/metabolismo , Proteínas E1B de Adenovirus/metabolismo , Caspasa 3 , Caspasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática , Humanos , Células Jurkat , Antígenos Comunes de Leucocito/metabolismo , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Mitocondrias/metabolismo , Inhibidor NF-kappaB alfa , Especies Reactivas de Oxígeno/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Virales/metabolismo
19.
EMBO J ; 17(20): 5974-86, 1998 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-9774341

RESUMEN

The CD95 signaling pathway comprises proteins that contain one or two death effector domains (DED), such as FADD/Mort1 or caspase-8. Here we describe a novel 37 kDa protein, DEDD, that contains an N-terminal DED. DEDD is highly conserved between human and mouse (98. 7% identity) and is ubiquitously expressed. Overexpression of DEDD in 293T cells induced weak apoptosis, mainly through its DED by which it interacts with FADD and caspase-8. Endogenous DEDD was found in the cytoplasm and translocated into the nucleus upon stimulation of CD95. Immunocytological studies revealed that overexpressed DEDD directly translocated into the nucleus, where it co-localizes in the nucleolus with UBF, a basal factor required for RNA polymerase I transcription. Consistent with its nuclear localization, DEDD contains two nuclear localization signals and the C-terminal part shares sequence homology with histones. Recombinant DEDD binds to both DNA and reconstituted mononucleosomes and inhibits transcription in a reconstituted in vitro system. The results suggest that DEDD is a final target of a chain of events by which the CD95-induced apoptotic signal is transferred into the nucleolus to shut off cellular biosynthetic activities.


Asunto(s)
Apoptosis , Proteínas de Arabidopsis , Nucléolo Celular/metabolismo , Proteínas de Unión al ADN/genética , Fragmentos de Péptidos/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Secuencia Conservada , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/fisiología , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Ácido Graso Desaturasas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Células Jurkat , Linfoma , Ratones , Datos de Secuencia Molecular , Proteínas de Plantas/metabolismo , Unión Proteica , Células Tumorales Cultivadas
20.
Eur J Immunol ; 28(8): 2276-82, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9710205

RESUMEN

Here we investigated the effects of the second messenger molecule NO at various concentrations on the activation of transcription factor NF-kappaB, IkappaB-alpha kinase (IKK-alpha), Jun N-terminal kinase (JNK) and apoptosis in murine endothelial cells. Low concentrations of NO alone failed to activate NF-kappaB, IKK-alpha and JNK. When NF-kappaB was prestimulated by TNF-alpha or phorbol 12-myristate 13-acetate, the addition of NO at low concentrations enhanced the activation of NF-kappaB. This provides a mechanism for a self-amplifying signal in the inflammatory response, since the inducible NO synthase in endothelial cells is regulated by NF-kappaB. The co-stimulatory effect of NO on NF-kappaB activation was also evident from IKK-alpha kinase assays and reporter gene experiments in endothelial cells. High doses of NO impaired the TNF-alpha-induced DNA-binding activity of NF-kappaB. Accordingly, these high amounts of NO also repressed the TNF-alpha-induced transactivation by NF-kappaB as efficient as dexamethasone. The doses of NO required for the inhibition of NF-kappaB are not cytotoxic for the endothelial cells, enabling the establishment of an autoregulatory loop for NF-kappaB signaling.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos , FN-kappa B/metabolismo , Óxido Nítrico/farmacología , Animales , Apoptosis , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular , Endotelio/citología , Endotelio/efectos de los fármacos , Endotelio/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Quinasa I-kappa B , Inflamación/etiología , Proteínas Quinasas JNK Activadas por Mitógenos , Luciferasas/genética , Ratones , FN-kappa B/fisiología , Óxido Nítrico/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Transfección , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...