Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
Oncogene ; 35(29): 3807-16, 2016 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-26592445

RESUMEN

Infection of human B lymphocytes by Epstein-Barr virus (EBV) leads to the establishment of immortalized lymphoblastoid cell lines (LCLs) that are widely used as a model of viral oncogenesis. An early consequence of infection is the induction of DNA damage and activation of the DNA damage response, which limits the efficiency of growth transformation. The cause of the DNA damage remains poorly understood. We have addressed this question by comparing the response of B lymphocytes infected with EBV or stimulated with a potent B-cell mitogen. We found that although the two stimuli induce comparable proliferation during the first 10 days of culture, the EBV-infected blasts showed significantly higher levels of DNA damage, which correlated with stronger and sustained accumulation of reactive oxygen species (ROS). Treatment with ROS scavengers decreased DNA damage in both mitogen-stimulated and EBV-infected cells. However, while mitogen-induced proliferation was slightly improved, the proliferation of EBV-infected cells and the establishment of LCLs were severely impaired. Quenching of ROS did not affect the kinetics and magnitude of viral gene expression but was associated with selective downregulation of the viral LMP1 and phosphorylated cellular transcription factor STAT3 that have key roles in transformation. Analysis of the mechanism by which high levels of ROS support LMP1 expression revealed selective inhibition of viral microRNAs that target the LMP1 transcript. Our study provides novel insights into the role of EBV-induced oxidative stress in promoting B-cell immortalization and malignant transformation.


Asunto(s)
Linfocitos B/virología , Transformación Celular Viral/fisiología , Herpesvirus Humano 4/fisiología , Estrés Oxidativo , Linfocitos B/metabolismo , Linfocitos B/patología , Proliferación Celular/genética , Transformación Celular Viral/genética , Células Cultivadas , Daño del ADN , Antígenos Nucleares del Virus de Epstein-Barr/genética , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Regulación de la Expresión Génica , Herpesvirus Humano 4/genética , Interacciones Huésped-Patógeno , Humanos , Immunoblotting , Microscopía Fluorescente , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
2.
Oncogene ; 32(49): 5522-30, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-23708666

RESUMEN

Malignant cells achieve replicative immortality by two alternative mechanisms, a common one dependent on de novo synthesis of telomeric DNA by telomerase, and a rare one based on telomere recombination known as alternative lengthening of telomeres (ALT). Epstein-Barr virus (EBV) transforms human B-lymphocytes into lymphoblastoid cell lines with unlimited growth potential in vitro and in vivo. Here we show that newly EBV-infected cells exhibit multiple signs of telomere dysfunction, including the occurrence of extra-chromosomal telomeres, telomere fusion and telomere length heterogeneity, and undergo progressive increase in telomere length without a parallel increase in telomerase activity. This phenotype is accompanied by the accumulation of telomere-associated promyelocytic leukemia nuclear bodies and telomeric-sister chromatid exchange, suggesting that EBV infection promotes the activation of ALT. Newly infected cells also display a significant reduction of telomere-associated TRF2 and express low levels of TRF1, TRF2, POT1 and ATRX, pointing to telomere de-protection as an important correlate of ALT activation. Collectively, these findings highlight the involvement of recombination-dependent mechanisms for maintenance of telomere homeostasis in EBV-induced B-cell immortalization.


Asunto(s)
Linfocitos B/metabolismo , Linfocitos B/virología , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4 , Homeostasis del Telómero/genética , Telómero/genética , ADN Helicasas/metabolismo , Activación Enzimática , Humanos , Proteínas Nucleares/metabolismo , Complejo Shelterina , Intercambio de Cromátides Hermanas , Telomerasa/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Proteína Nuclear Ligada al Cromosoma X
3.
Leukemia ; 25(6): 1017-25, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21394098

RESUMEN

The Epstein-Barr virus (EBV) nuclear antigen (EBNA)-1 promotes the accumulation of chromosomal aberrations in malignant B cells by inducing oxidative stress. Here we report that this phenotype is associated with telomere dysfunction. Stable or conditional expression of EBNA1 induced telomere abnormalities including loss or gain of telomere signals, telomere fusion and heterogeneous length of telomeres. This was accompanied by the accumulation of extrachromosomal telomeres, telomere dysfunction-induced foci (TIFs) containing phosphorylated histone H2AX and the DNA damage response protein 53BP1, telomere-associated promyelocytic leukemia nuclear bodies (APBs), telomeric-sister chromatid exchanges and displacement of the shelterin protein TRF2. The induction of TIFs and APBs was inhibited by treatment with scavengers of reactive oxygen species (ROS) that also promoted the relocalization of TRF2 at telomeres. These findings highlight a novel mechanism by which EBNA1 may promote malignant transformation and tumor progression.


Asunto(s)
Transformación Celular Viral , Antígenos Nucleares del Virus de Epstein-Barr/fisiología , Herpesvirus Humano 4/fisiología , Estrés Oxidativo , Telómero/patología , Línea Celular Tumoral , Aberraciones Cromosómicas , Reparación del ADN , Histonas/metabolismo , Humanos , Fosforilación , Especies Reactivas de Oxígeno
4.
Oncogene ; 28(45): 3997-4008, 2009 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-19718051

RESUMEN

Epstein-Barr virus (EBV) has been implicated in the pathogenesis of human malignancies, but its contribution to tumorigenesis is not well understood. EBV carriage is associated with increased genomic instability in Burkitt's lymphoma, suggesting that viral products may induce this tumor phenotype. Using a panel of transfected sublines of the B-lymphoma line BJAB expressing the viral genes associated with latent infection, we show that the EBV nuclear antigens, EBNA-1 and EBNA-3C, and the latent membrane protein 1, LMP-1, independently promote genomic instability, as detected by nonclonal chromosomal aberrations, DNA breaks and phosphorylation of histone H2AX. EBNA-1 promotes the generation of DNA damage by inducing reactive oxygen species (ROS), whereas DNA repair is inhibited in LMP-1-expressing cells through downregulation of the DNA damage-sensing kinase, ataxia telangiectasia mutated (ATM), reduction of phosphorylation of its downstream targets Chk2 and inactivation of the G(2) checkpoint. EBNA-3C enhances the propagation of damaged DNA through inactivation of the mitotic spindle checkpoint and transcriptional downregulation of BubR1. Thus, multiple cellular functions involved in the maintenance of genome integrity seem to be independently targeted by EBV, pointing to the induction of genomic instability as a critical event in viral oncogenesis.


Asunto(s)
Antígenos Virales/genética , Linfoma de Burkitt/genética , Linfoma de Burkitt/virología , Daño del ADN , Reparación del ADN , Inestabilidad Genómica , Herpesvirus Humano 4/fisiología , Proteínas de la Ataxia Telangiectasia Mutada , Linfoma de Burkitt/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Antígenos Nucleares del Virus de Epstein-Barr/genética , Herpesvirus Humano 4/genética , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Huso Acromático , Transfección , Proteínas Supresoras de Tumor/metabolismo , Proteínas de la Matriz Viral/genética , Latencia del Virus
5.
Oncogene ; 26(35): 5115-23, 2007 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-17325665

RESUMEN

Epstein-Barr virus (EBV) has been implicated in the pathogenesis of human malignancies but the mechanisms of oncogenesis remain largely unknown. Genomic instability and chromosomal aberrations are hallmarks of malignant transformation. We report that EBV carriage promotes genomic instability in Burkitt's lymphoma (BL). Cytogenetic analysis of EBV- and EBV+ BL lines and their sublines derived by EBV conversion or spontaneous loss of the viral genome revealed a significant increase in dicentric chromosomes, chromosome fragments and chromatid gaps in EBV-carrying cells. Expression of EBV latency I was sufficient for this effect, whereas a stronger effect was observed in cells expressing latency III. Telomere analysis by fluorescent in situ hybridization revealed an overall increase of telomere size and prevalence of telomere fusion and double strand-break fusion in dicentric chromosomes from EBV+ cells. Phosphorylated H2AX, a reporter of DNA damage and ongoing repair, was increased in virus-carrying cells in the absence of exogenous stimuli, whereas efficient activation of DNA repair was observed in both EBV+ and EBV- cells following treatment with etoposide. These findings point to induction of telomere dysfunction and DNA damage as important mechanisms for EBV oncogenesis.


Asunto(s)
Linfoma de Burkitt/genética , Linfoma de Burkitt/virología , Roturas del ADN de Doble Cadena , Inestabilidad Genómica , Herpesvirus Humano 4 , Telómero/ultraestructura , Línea Celular Tumoral , Cromosomas Humanos/ultraestructura , Daño del ADN , Histonas/metabolismo , Humanos , Hibridación Fluorescente in Situ , Fosforilación , Latencia del Virus
6.
Cell Mol Life Sci ; 63(6): 723-34, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16501887

RESUMEN

The biological functions of the more than one hundred genes coding for deubiquitinating enzymes in the human genome remain mostly unknown. The USP25 gene, located at 21q11.2, encodes three protein isoforms produced by alternative splicing. While two of the isoforms are expressed nearly ubiquituously, the expression of the longer USP25 isoform (USP25m) is restricted to muscular tissues and is upregulated during myogenesis. USP25m interacts with three sarcomeric proteins: actin alpha-1 (ACTA1), filamin C (FLNC), and myosin binding protein C1 (MyBPC1), which are critically involved in muscle differentiation and maintenance, and have been implicated in the pathogenesis of severe myopathies. Biochemical analyses demonstrated that MyBPC1 is a short-lived proteasomal substrate, and its degradation is prevented by over-expression of USP25m but not by other USP25 isoforms. In contrast, ACTA1 and FLNC appear to be stable proteins, indicating that their interaction with USP25m is not related to their turnover rate.


Asunto(s)
Actinas/metabolismo , Empalme Alternativo , Proteínas Portadoras/metabolismo , Proteínas Contráctiles/metabolismo , Endopeptidasas/metabolismo , Proteínas de Microfilamentos/metabolismo , Sarcómeros/metabolismo , Animales , Diferenciación Celular , Línea Celular , Células Cultivadas , Endopeptidasas/genética , Filaminas , Humanos , Ratones , Sarcómeros/química , Ubiquitina Tiolesterasa
7.
Scand J Immunol ; 60(6): 592-9, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15584970

RESUMEN

Modulation of cytotoxic responses by viral immunoevasins plays an important role in the establishment of latent and persistent viral infections. Together with MHC class-I-restricted CD8T-lymphocytes, non-MHC-restricted natural killer (NK) and lymphokine-activated killer (LAK) cells participate in this anti-viral control. The Us3 protein kinase of herpes simplex virus-1 (HSV-1) inhibits CD8T-cell cytotoxicity, which correlates with the inhibition of granzyme-B (GrB)-induced activation of pro-apoptotic Bid. We have investigated the effect of Us3 on NK and LAK cytotoxicity, because these effectors are believed to share common mechanisms for inducing cell death. We show that, in contrast to their lower sensitivity to CD8T-cell lysis, HSV-1-infected cells are lysed by NK cells or LAK cells as efficiently as the uninfected controls. Both CD8T and NK/LAK effectors were dependent on the activity of GrB and were efficiently blocked by means of treatment with a GrB inhibitor. However, unlike CD8T cells, LAK cells and NK cells failed to induce Bid cleavage, suggesting that various GrB downstream targets be involved in the induction of cell lysis. This finding explains their various sensitivities to viral modulation, which is likely to be important for the respective role of MHC-restricted and non-restricted effectors in the control of HSV-1 infection.


Asunto(s)
Herpes Simple/enzimología , Herpesvirus Humano 1/enzimología , Antígenos de Histocompatibilidad Clase I/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Linfocitos T CD8-positivos/inmunología , Herpes Simple/inmunología , Herpesvirus Humano 1/patogenicidad , Humanos , Células Asesinas Activadas por Linfocinas/inmunología , Células Asesinas Naturales/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Virales
8.
Cell Death Differ ; 10(12): 1320-8, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12934063

RESUMEN

The Us3 kinase is part of the antiapoptotic arsenal that salvages herpes simplex virus (HSV)-1-infected cells from damage caused by different stimuli. We demonstrate that Us3 protects HSV-1-infected cells from lysis by MHC class I-restricted CD8T cells without affecting antigen presentation. Expression of Us3 was associated with inhibition of caspase activation and reduced cleavage of the proapoptotic protein Bid. Recombinant granzyme B (GrB) failed to cleave Bid in cytosolic extracts from Us3 positive cells, while recombinant Bid served as substrate for Us3 phosphorylation, suggesting that modification of Bid by Us3 blocks its processing by GrB. Our data illustrate a new strategy of viral escape, where modification of a cellular proapoptotic substrate may prevent lysis of the infected cells without affecting other T-cell functions.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Serina Endopeptidasas/farmacología , Presentación de Antígeno , Apoptosis , Proteína Proapoptótica que Interacciona Mediante Dominios BH3 , Western Blotting , Caspasas/metabolismo , Línea Celular , Citosol/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática , Granzimas , Humanos , Linfocitos/metabolismo , Complejo Mayor de Histocompatibilidad , Microscopía Fluorescente , Mutación , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Linfocitos T Citotóxicos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Virales
9.
Scand J Immunol ; 57(2): 99-106, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12588655

RESUMEN

Accumulating evidence strongly supports the role of lipid rafts in the regulation of T-lymphocyte activation, but the organization and molecular composition of these cholesterol- and sphingolipid-rich membrane microdomains in different subsets of T cells remain poorly investigated. Here, we show that pharmacological disruption of lipid rafts in human CD8+ cytotoxic T-lymphocyte (CTL) clones disturbs the integrity of CD3 complex and CD8 heterodimer, without affecting the reactivity with T-cell receptor (TCR)-specific antibodies. This demonstrates that interaction with completely assembled CD3 complex is not required for the stable expression of TCR at the cell surface. The effect of raft disruption on CD3 and CD8 expression correlates with failure to bind specific tetrameric complexes by a proportion of surface TCR molecules. However, the interaction of specific tetramer with the rest of surface TCR pools appears to be unaffected, demonstrating that TCR-signalling complexes may differ in their requirement for cholesterol to stably maintain their composition and to rearrange for efficient tetramer binding. Together with previously published data, our results support the existence of molecular and/or structural heterogeneity of lipid rafts that may play an important role in controlling distinct functional properties of T-cell subsets.


Asunto(s)
Antifúngicos/farmacología , Complejo CD3/metabolismo , Antígenos CD8/metabolismo , Ciclodextrinas/farmacología , Filipina/farmacología , Activación de Linfocitos/inmunología , Microdominios de Membrana/metabolismo , Linfocitos T Citotóxicos/metabolismo , beta-Ciclodextrinas , Complejo CD3/inmunología , Antígenos CD8/inmunología , Colesterol/inmunología , Colesterol/metabolismo , Citometría de Flujo , Antígenos HLA-A/inmunología , Antígenos HLA-A/metabolismo , Antígeno HLA-A11 , Humanos , Microdominios de Membrana/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
10.
Curr Top Microbiol Immunol ; 269: 23-36, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12224511

RESUMEN

Ubiquitin/proteasome-dependent proteolysis is involved in the regulation of a large variety of cellular processes including cell cycle progression, tissue development and atrophy, flux of substrates through metabolic pathways, selective elimination of abnormal proteins and processing of intracellular antigens for major histocompatibility complex (MHC) class I-restricted T-cell responses. Many viruses tamper with this proteolytic machinery by encoding proteins that interact with various components of the pathway. A particularly interesting example of a viral protein that interferes with proteasomal processing is the Epstein-Barr virus (EBV) nuclear antigen-1 (EBNA1). EBNA1 contains an internal repeat exclusively composed of glycines and alanines that inhibits in cis the presentation of MHC class I-restricted T-cell epitopes and prevents ubiquitin/proteasome-dependent proteolysis in vitro and in vivo. The glycine-alanine repeat acts as a transferable element on a variety of proteasomal substrates and may therefore provide a new approach to the modification of cellular proteins for therapeutic purposes.


Asunto(s)
Infecciones por Virus de Epstein-Barr/virología , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/inmunología , Ubiquitinas/metabolismo , Alanina , Presentación de Antígeno , Cisteína Endopeptidasas/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/genética , Glicina , Antígenos de Histocompatibilidad Clase I/inmunología , Complejos Multienzimáticos/metabolismo , Complejo de la Endopetidasa Proteasomal , Secuencias Repetitivas de Aminoácido , Secuencias Repetitivas de Ácidos Nucleicos , Linfocitos T/inmunología
11.
Antimicrob Agents Chemother ; 45(9): 2616-22, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11502538

RESUMEN

The human immunodeficiency virus type 1 (HIV-1) protease is essential for production of infectious virus and is therefore a major target for the development of drugs against AIDS. Cellular proteins are also cleaved by the protease, which explains its cytotoxic activity and the consequent failure to establish convenient cell-based protease assays. We have exploited this toxicity to develop a new protease assay that relies on transient expression of an artificial protease precursor harboring the green fluorescent protein (GFP-PR). The precursor is activated in vivo by autocatalytic cleavage, resulting in rapid elimination of protease-expressing cells. Treatment with therapeutic doses of HIV-1 protease inhibitors results in a dose-dependent accumulation of the fluorescent precursor that can be easily detected and quantified by flow cytometric and fluorimetric assays. The precursor provides a convenient and noninfectious model for high-throughput screenings of substances that can interfere with the activity of the protease in living cells.


Asunto(s)
Proteasa del VIH/análisis , Catálisis , Relación Dosis-Respuesta a Droga , Activación Enzimática , Fluorescencia , Proteínas Fluorescentes Verdes , Proteasa del VIH/metabolismo , Inhibidores de la Proteasa del VIH/análisis , Inhibidores de la Proteasa del VIH/farmacología , Células HeLa , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Volumetría/métodos , Transfección
12.
FEBS Lett ; 499(1-2): 137-42, 2001 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-11418128

RESUMEN

The Gly-Ala repeat (GAr) of the Epstein-Barr virus nuclear antigen 1 is a cis acting inhibitor of ubiquitin-proteasome proteolysis. We have investigated the capacity of various repeats to inhibit the turnover of the proteasomal substrate IkappaBalpha. Inhibition of TNFalpha-induced degradation was achieved by insertion of octamers containing three alanines or valines, interspersed by no more then three consecutive glycines. The inhibitory activity was abolished by increasing the length of the spacer, by eliminating the spacers, or by substitution of a single hydrophobic residue with a polar or charged residue. A serine containing octamer was inactive but inhibition was partially restored by insertion of three consecutive repeats. These findings suggest a model where inhibition requires the interaction of at least three alanine residues of the GAr in a beta-strand conformation with adjacent hydrophobic binding pockets of a putative receptor.


Asunto(s)
Antígenos Nucleares del Virus de Epstein-Barr/química , Antígenos Nucleares del Virus de Epstein-Barr/farmacología , Proteínas I-kappa B , Complejos Multienzimáticos/antagonistas & inhibidores , Secuencias Repetitivas de Aminoácido , Ubiquitinas/antagonistas & inhibidores , Alanina/genética , Alanina/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Sitios de Unión , Cisteína Endopeptidasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/genética , Femenino , Glicina/genética , Glicina/metabolismo , Células HeLa , Humanos , Datos de Secuencia Molecular , Peso Molecular , Complejos Multienzimáticos/metabolismo , Inhibidor NF-kappaB alfa , Complejo de la Endopetidasa Proteasomal , Conformación Proteica , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Serina/genética , Serina/metabolismo , Transfección , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología , Ubiquitinas/metabolismo , Valina/genética , Valina/metabolismo , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/farmacología
13.
Nat Cell Biol ; 3(3): 283-8, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11231578

RESUMEN

Burkitt's lymphoma (BL) is a highly malignant B-cell tumour characterized by chromosomal translocations that constitutively activate the c-myc oncogene. Here we show that BL cells are resistant to apoptosis and do not accumulate ubiquitin conjugates in response to otherwise toxic doses of inhibitors of the proteasome. Deubiquitinating enzymes and the cytosolic subtilisin-like protease tripeptidylpeptidase II are upregulated in BLs, and could be rapidly induced by the overexpression of c-myc in normal B cells carrying oestrogen-driven recombinant Epstein-Barr virus. Apoptosis was induced by inhibiting tripeptidylpeptidase II, suggesting that the activity of this protease may be required for the survival of BL cells. We thus show that there is a regulatory link between c-myc activation and changes in proteolysis that may affect malignant transformation.


Asunto(s)
Apoptosis/fisiología , Linfocitos B/citología , Linfoma de Burkitt/metabolismo , Cisteína Endopeptidasas/metabolismo , Genes myc , Complejos Multienzimáticos/metabolismo , Oligopéptidos/farmacología , Proteínas/metabolismo , Sulfonas/farmacología , Ubiquitinas/metabolismo , Aminopeptidasas , Linfocitos B/metabolismo , Linfoma de Burkitt/genética , Linfoma de Burkitt/patología , Inhibidores de Cisteína Proteinasa/farmacología , ADN/metabolismo , Fragmentación del ADN , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas , Endopeptidasas/metabolismo , Herpesvirus Humano 4/metabolismo , Humanos , Immunoblotting , Complejos Multienzimáticos/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Complejo de la Endopetidasa Proteasomal , Serina Endopeptidasas/química , Serina Endopeptidasas/metabolismo
14.
J Immunol ; 166(2): 989-95, 2001 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11145677

RESUMEN

Activation-induced cell death (AICD) of mature T cells plays an important role in the control of immune homeostasis and peripheral tolerance. TNFRs and Fas have been implicated in the induction of AICD. However, these molecules were shown to be dispensable, at least in some experimental systems, for downsizing of Ag-induced T cell expansions and development of tolerance in vivo. The conditions of T cell activation leading to T cell deletion in a death receptor-independent manner are not well characterized. Here we show that human CTLs die through a death receptor-independent apoptotic program upon triggering with a partially agonistic peptide ligand. This apoptotic process exhibits some features of T cell death due to lymphokine deprivation and is blocked by exogenous IL-2. Our data demonstrate that engagement of TCR by MHC-peptide complexes can trigger diverse apoptotic programs of AICD and that the choice between these programs is determined by the agonistic potency of MHC-peptide ligand.


Asunto(s)
Apoptosis/inmunología , Activación de Linfocitos , Fragmentos de Péptidos/agonistas , Fragmentos de Péptidos/inmunología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Animales , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Muerte Celular/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Células Clonales , Ciclosporina/farmacología , Antígenos Nucleares del Virus de Epstein-Barr/inmunología , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Proteína Ligando Fas , Antígenos HLA-A/inmunología , Antígeno HLA-A11 , Humanos , Interleucina-2/antagonistas & inhibidores , Interleucina-2/farmacología , Cinética , Ligandos , Activación de Linfocitos/efectos de los fármacos , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/metabolismo , Ratones , Fragmentos de Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/metabolismo , Células Tumorales Cultivadas , Regulación hacia Arriba/inmunología , Proteína bcl-X , Receptor fas/metabolismo
15.
Hum Immunol ; 61(10): 972-84, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11082510

RESUMEN

We have investigated the presentation and CTL recognition of an HLA A*1101-restricted CTL peptide epitope AVFDRKSDAK (AVF)(3), derived from the EBV nuclear antigen (EBNA) 4, in the context of alleles belonging to the A3-supertype, A*0101, 0301, 1101, 3101, 3301, and 6801. The peptide binds to a A*6801 molecule as efficiently as to A*1101. The A*6801:AVF complex is recognized by some A*1101-restricted AVF- specific CTL clones. However, A*6801-positive (A*6801+) EBV-transformed lymphoblastoid cell lines (LCLs) are not killed by the same effectors. Furthermore, two A*6801+ donors did not mount an AVF-specific CTL response in vitro and lacked detectable AVF-specific effectors. Thus, this epitope is either subdominant, or non-immunogenic in the context of A*6801. These characteristics correlate with low stability of this MHC:peptide complex in living cells. We also demonstrate that a highly conserved AVF-specific TCR that dominates the AVF-specific CTL response in the majority of A*1101+ individuals recognizes the A*6801 molecule as a crossreactive alloantigen. Therefore, deletion of AVF-specific T cells may contribute to the non-immunogenicity or subdominance of the peptide in A*6801+ individuals.


Asunto(s)
Antígenos Nucleares del Virus de Epstein-Barr/inmunología , Antígenos HLA-A/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Alelos , Secuencias de Aminoácidos , Presentación de Antígeno , Línea Celular Transformada , Infecciones por Virus de Epstein-Barr/inmunología , Antígenos Nucleares del Virus de Epstein-Barr/química , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Antígenos HLA-A/química , Antígenos HLA-A/metabolismo , Herpesvirus Humano 4/química , Herpesvirus Humano 4/inmunología , Humanos , Inmunoterapia/métodos , Péptidos/química , Péptidos/genética , Péptidos/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo
16.
Int J Cancer ; 88(6): 881-8, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11093809

RESUMEN

We investigated the expression of interferon gamma (IFN-gamma)-regulated subunits and the enzymatic activity of proteasomes purified from tumor-derived and normal B lymphocytes representing different stages of B-cell activation/differentiation. The catalytic beta subunits (Lmp2 and Lmp7) and the regulatory subunits (PA28alpha and PA28beta) were expressed at equally high levels in Epstein-Barr virus (EBV)-transformed lymphoblastoid cell lines (LCLs), freshly isolated B-chronic lymphocytic leukemia (B-CLL) cells and normal CD23(-) B lymphocytes. Lmp2 and Lmp7 were selectively down-regulated in germinal center cell-derived Burkitt's lymphoma (BL) and Hodgkin's lymphoma (HD) cell lines. There was a direct correlation between the expression of Lmp2/7 and the chymotrypsin and trypsin-like activities in proteasomes purified from LCLs, BLs and CLL cells, whereas 5 HD cell lines expressing B or T-cell markers exhibited a variable pattern of subunit expression and enzymatic activity. Poor hydrolysis of the fluorogenic substrates by proteasomes from BL cells correlated with a distinct pattern of cleavage of a reference 50mer peptide, production of different sets of degradation products and significantly reduced recovery of a known cytotoxic T-lymphocyte (CTL) target epitope. The enzymatic activity of proteasomes from normal CD23(-) "resting" B lymphocytes resembled that of BL cells in spite of high Lmp2/7 expression. This pattern was not reversed by treatment with the B-cell mitogen, lipopolysaccharide (LPS). The results suggest that different stages of B-cell activation/differentiation are associated with distinct profiles of IFN-gamma-regulated subunit composition and enzymatic activity of the proteasome. This may have important implications for the analysis and manipulation of tumor-specific immune responses.


Asunto(s)
Linfocitos B/química , Quimotripsina/análisis , Cisteína Endopeptidasas , Linfoma de Células B/metabolismo , Complejos Multienzimáticos , Proteínas Musculares , Proteínas/análisis , Tripsina/análisis , Linfocitos B/enzimología , Linfocitos B/fisiología , Humanos , Activación de Linfocitos/fisiología , Linfoma de Células B/enzimología , Complejo de la Endopetidasa Proteasomal , Células Tumorales Cultivadas
17.
Proc Natl Acad Sci U S A ; 97(15): 8381-5, 2000 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-10890896

RESUMEN

The Gly-Ala repeat (GAr) of the Epstein-Barr virus nuclear antigen-1 is a transferable element that inhibits in cis ubiquitin/proteasome-dependent proteolysis. We have investigated this inhibitory activity by using green fluorescent protein-based reporters that have been targeted for proteolysis by N end rule or ubiquitin-fusion degradation signals, resulting in various degrees of destabilization. Degradation of the green fluorescent protein substrates was inhibited on insertion of a 25-aa GAr, but strongly destabilized reporters were protected only partially. Protection could be enhanced by increasing the length of the repeat. However, reporters containing the Ub-R and ubiquitin-fusion degradation signals were degraded even in the presence of a 239-aa GAr. In accordance, insertion of a powerful degradation signal relieved the blockade of proteasomal degradation in Epstein-Barr virus nuclear antigen-1. Our findings suggest that the turnover of natural substrates may be finely tuned by GAr-like sequences that counteract targeting signals for proteasomal destruction.


Asunto(s)
Alanina/metabolismo , Cisteína Endopeptidasas/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Glicina/metabolismo , Herpesvirus Humano 4/metabolismo , Complejos Multienzimáticos/metabolismo , Secuencias Repetitivas de Aminoácido , Antígenos Nucleares del Virus de Epstein-Barr/genética , Genes Reporteros , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Proteínas Luminiscentes/genética , Complejo de la Endopetidasa Proteasomal , Ubiquitinas/genética
18.
Nat Biotechnol ; 18(5): 538-43, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10802622

RESUMEN

The ubiquitin/proteasome-dependent proteolytic pathway is an attractive target for therapeutics because of its critical involvement in cell cycle progression and antigen presentation. However, dissection of the pathway and development of modulators are hampered by the complexity of the system and the lack of easily detectable authentic substrates. We have developed a convenient reporter system by producing N-end rule and ubiquitin fusion degradation (UFD)-targeted green fluorescent proteins that allow quantification of ubiquitin/proteasome-dependent proteolysis in living cells. Accumulation of these reporters serves as an early predictor of G2/M arrest and apoptosis in cells treated with proteasome inhibitors. Comparison of reporter accumulation and cleavage of fluorogenic substrates demonstrates that the rate-limiting chymotrypsin-like activity of the proteasome can be substantially curtailed without significant effect on ubiquitin-dependent proteolysis. These reporters provide a new powerful tool for elucidation of the ubiquitin/proteasome pathway and for high throughput screening of compounds that selectively modify proteolysis in vivo.


Asunto(s)
Cisteína Endopeptidasas/efectos de los fármacos , Cisteína Endopeptidasas/metabolismo , Genes Reporteros , Proteínas Luminiscentes/metabolismo , Complejos Multienzimáticos/efectos de los fármacos , Complejos Multienzimáticos/metabolismo , Ubiquitinas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Leupeptinas/farmacología , Proteínas Luminiscentes/genética , Oligopéptidos/farmacología , Complejo de la Endopetidasa Proteasomal , Proteínas Recombinantes de Fusión/metabolismo , Especificidad por Sustrato , Sulfonas/farmacología , Transfección , Ubiquitinas/genética
19.
Scand J Immunol ; 51(4): 361-71, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10736108

RESUMEN

We have compared the cell phenotype and functional properties of monocyte/macrophage derived dendritic cells (DCs) obtained by culture of human adherent peripheral blood mononuclear cells (PBMCs) in medium containing granulocyte macrophage colony stimulating factor (GM-CSF) either alone (GM-CSF-DCs), or in combination with interleukin (IL)-4 (IL4-DCs) or IL-7 (IL7-DCs). The cell surface phenotype of GM-CSF-DCs and IL-7-DCs was characterized by a high expression of major histocompatibility complex (MHC) class I and II, CD80, CD86 and CD40. In contrast to 'classical' IL-4-DCs, these two types of DCs expressed CD14 and a CD21-like molecule detected by two out of four CD21-specific monoclonal antibodies (MoAb) tested. The same pattern of reactivity with CD21 specific antibodies was observed in freshly isolated adherent PBMCs but not in B lymphocytes. This reactivity was upregulated by IL-7 in a dose dependent manner. Lipopolysaccharide (LPS) treatment induced the upregulation of CD40, CD80, CD86 and the T-cell stimulatory capacity in IL-4-DCs and, to a lesser extent, in the IL-7-DCs whereas GM-CSF-DCs responded very poorly to such treatment. Our data indicate that, together with GM-CSF, the IL-7 drives macrophage precursors to a differentiation stage that is close to but distinct from the phenotype of IL-4-DCs. Comparison of DC development in the presence of IL-7 or IL-4 may help in dissecting signalling pathways that regulate the expression of functionally relevant DC markers.


Asunto(s)
Células Dendríticas/fisiología , Interleucina-7/farmacología , Monocitos/fisiología , Antígenos CD/biosíntesis , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Antígenos de Histocompatibilidad Clase I/biosíntesis , Humanos , Inmunofenotipificación , Interleucina-4/farmacología , Monocitos/citología , Monocitos/efectos de los fármacos , Proteínas Recombinantes/farmacología , Regulación hacia Arriba
20.
Blood ; 95(3): 807-14, 2000 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-10648390

RESUMEN

A semiquantitative polymerase chain reaction assay was used to monitor the blood levels of Epstein-Barr virus (EBV)-DNA in 9 patients receiving allogeneic bone marrow transplants (BMT). Four of 5 recipients of HLA-mismatched T-cell-depleted grafts showed a 4- to 5-log increase of EBV-DNA within 1 to 3 months after BMT. Administration of 2 to 4 infusions of 10(7) EBV-specific cytotoxic T-lymphocytes (CTLs)/m(2) starting from the time of maximal virus load resulted in a 2- to 3-log decrease of virus titers in 3 patients. One patient, who received a T-cell culture lacking a major EBV-specific component, progressed to fatal EBV-positive lymphoma. Administration of EBV-CTLs before the onset of the EBV-DNA peak resulted in stabilization of the virus titers within 2 to 3 logs above the normal levels in the fifth patient. A moderate increase of virus titers was also detected in 3 of 4 patients receiving unmanipulated HLA-matched grafts, whereas 1 patient with Wiskott-Aldrich syndrome reached a 5-log increase of EBV-DNA load within 70 days after BMT. Our results suggest that a rapid increase of circulating EBV-DNA occurs in the absence of EBV-specific T-cell precursors or in the presence of congenital immune defects that prevent the reestablishment of virus-specific immunity. Prophylactic administration of EBV-CTLs early after BMT appears to provide the most effective protection against the development of EBV-associated lymphoproliferative disease.


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Infecciones por Virus de Epstein-Barr/terapia , Herpesvirus Humano 4/aislamiento & purificación , Inmunoterapia Adoptiva , Trastornos Linfoproliferativos/prevención & control , Linfocitos T Citotóxicos/trasplante , Infecciones Tumorales por Virus/terapia , Viremia/virología , Adolescente , Niño , Preescolar , ADN Viral/sangre , Progresión de la Enfermedad , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/epidemiología , Femenino , Enfermedades Genéticas Congénitas/terapia , Antígenos HLA/inmunología , Neoplasias Hematológicas/complicaciones , Neoplasias Hematológicas/terapia , Herpesvirus Humano 4/inmunología , Histocompatibilidad , Humanos , Huésped Inmunocomprometido , Terapia de Inmunosupresión/efectos adversos , Lactante , Linfoma/etiología , Linfoma/prevención & control , Linfoma/virología , Trastornos Linfoproliferativos/etiología , Trastornos Linfoproliferativos/virología , Masculino , Reacción en Cadena de la Polimerasa , Prevalencia , Riesgo , Linfocitos T Citotóxicos/inmunología , Resultado del Tratamiento , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/epidemiología , Carga Viral , Viremia/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA