Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Cell Cycle ; 18(8): 898-913, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30975033

RESUMEN

Besides TopBP1, ETAA1 has been identified more recently as an activator of the ATR-ATRIP complex in human cells. We have examined the role of ETAA1 in the Xenopus egg-extract system, which has been instrumental in the study of ATR-ATRIP. Depletion of ETAA1 from egg extracts did not noticeably reduce the activation of ATR-ATRIP in response to replication stress, as monitored by the ATR-dependent phosphorylation of Chk1 and RPA. Moreover, lack of ETAA1 did not appear to affect DNA replication during an unperturbed S-phase. Significantly, we find that TopBP1 is considerably more abundant than ETAA1 in egg extracts. We proceeded to show that ETAA1 could support the activation of ATR-ATRIP in response to replication stress if we increased its concentration in egg extracts by adding extra full-length recombinant ETAA1. Thus, TopBP1 appears to be the predominant activator of ATR-ATRIP in response to replication stress in this system. We have also explored the biochemical mechanism by which ETAA1 activates ATR-ATRIP. We have developed an in vitro system in which full-length recombinant ETAA1 supports activation of ATR-ATRIP in the presence of defined components. We find that binding of ETAA1 to RPA associated with single-stranded DNA (ssDNA) greatly stimulates its ability to activate ATR-ATRIP. Thus, RPA-coated ssDNA serves as a direct positive effector in the ETAA1-mediated activation of ATR-ATRIP.


Asunto(s)
Antígenos de Superficie/metabolismo , ADN de Cadena Simple/metabolismo , Proteína de Replicación A/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Anticuerpos/inmunología , Antígenos de Superficie/inmunología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Cromatina/metabolismo , Replicación del ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Fosforilación/inmunología , Unión Proteica , Proteínas Recombinantes/metabolismo , Fase S/inmunología , Xenopus , Proteínas de Xenopus/metabolismo
2.
Science ; 349(6248): 643-6, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26184917

RESUMEN

The germinal center (GC) is a microanatomical compartment wherein high-affinity antibody-producing B cells are selectively expanded. B cells proliferate and mutate their antibody genes in the dark zone (DZ) of the GC and are then selected by T cells in the light zone (LZ) on the basis of affinity. Here, we show that T cell help regulates the speed of cell cycle phase transitions and DNA replication of GC B cells. Genome sequencing and single-molecule analyses revealed that T cell help shortens S phase by regulating replication fork progression, while preserving the relative order of replication origin activation. Thus, high-affinity GC B cells are selected by a mechanism that involves prolonged dwell time in the DZ where selected cells undergo accelerated cell cycles.


Asunto(s)
Linfocitos B/citología , Ciclo Celular/inmunología , Replicación del ADN/inmunología , Centro Germinal/citología , Inmunidad Humoral/genética , Linfocitos T/inmunología , Animales , Ciclo Celular/genética , Proliferación Celular , Replicación del ADN/genética , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fase S/genética , Fase S/inmunología
3.
J Theor Biol ; 335: 57-78, 2013 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-23806696

RESUMEN

In this paper, we sought to identify the CD4(+) T-cell dynamics in the course of HIV infection in response to continuous and intermittent intravenous courses of interleukin-2 (IL-2), the principal cytokine responsible for progression of CD4(+) T-lymphocytes from the G1 to the S phase of the cell cycle. Based on multivariate regression models, previous literature has concluded that the increase in survival of CD4(+) T-cell appears to be the critical mechanism leading to sustained CD4(+) T-cell levels in HIV-infected patients receiving intermittent IL-2 therapy. Underscored by comprehensive mathematical modeling, a major finding of the present work is related to the fact that, rather than due to any increase in survival of CD4(+) T-cells, the expressive, selective and sustained CD4(+) T-cell expansions following IL-2 administration may be related to the role of IL-2 in modulating the dynamics of Fas-dependent apoptotic pathways, such as activation-induced cell death (AICD) or HIV-specific apoptotic routes triggered by viral proteins.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/inmunología , Síndrome de Inmunodeficiencia Adquirida/terapia , Linfocitos T CD4-Positivos/inmunología , VIH-1/inmunología , Inmunoterapia , Interleucina-2 , Síndrome de Inmunodeficiencia Adquirida/patología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Linfocitos T CD4-Positivos/patología , Femenino , Fase G1/efectos de los fármacos , Fase G1/inmunología , Humanos , Interleucina-2/inmunología , Interleucina-2/uso terapéutico , Masculino , Fase S/efectos de los fármacos , Fase S/inmunología , Proteínas Virales/inmunología , Receptor fas/inmunología
4.
Mol Immunol ; 53(3): 214-7, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22960197

RESUMEN

The activation-induced cytidine deaminase (AID) initiates Ig gene hypermutation by converting cytosine to uracil (U) and generating a U:G lesion. Genetic and biochemical studies suggest that the AID-triggered U:G lesions are processed by three mutagenic pathways to induce mutations at both C:G and A:T pairs. First, direct replication of the U:G lesion leads to C to T and G to A transitions. Second, U can be excised by the uracil DNA glycosylase (UNG) and the replication/processing of the resulting abasic site leads to transversions and transitions at C:G pairs. Third, the U:G lesion is recognized by an atypical mismatch repair (MMR) pathway which generates mutations at A:T pairs in a DNA polymerase η (POLH)-dependent manner. To further explore whether these three mutagenic pathways function competitively or independently, we have analyzed Ig gene hypermutation in mice deficient in both UNG and POLH. Compared with WT mice, UNG deficiency caused elevated frequency of C:G mutations, suggesting that UNG-mediated U excision led to error-free as well as error-prone repair. In contrast, UNG deficiency did not affect the frequency and patterns of A:T mutations, suggesting that the MMR did not target U:G lesions normally recognized and processed by UNG. In addition, POLH deficiency did not affect the frequency and patterns of C:G mutations and UNG POLH double deficiency showed an additive effect of single deficiency. Based on these observations and previous results, along with the recent finding that UNG excises AID-triggered U predominantly during G1 phase of the cell cycle, it appears that UNG and MMR targets U:G lesions generated during G1 and S phases of the cell cycle, respectively.


Asunto(s)
ADN Polimerasa Dirigida por ADN/deficiencia , ADN Polimerasa Dirigida por ADN/genética , Hipermutación Somática de Inmunoglobulina , Uracil-ADN Glicosidasa/deficiencia , Uracil-ADN Glicosidasa/genética , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Secuencia de Bases , Citidina Desaminasa/metabolismo , Reparación de la Incompatibilidad de ADN/genética , Reparación de la Incompatibilidad de ADN/inmunología , Cartilla de ADN/genética , Fase G1/genética , Fase G1/inmunología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Genéticos , Modelos Inmunológicos , Fase S/genética , Fase S/inmunología
5.
J Immunol ; 189(5): 2374-82, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22826323

RESUMEN

Activation-induced cytidine deaminase (AID) initiates DNA double-strand breaks (DSBs) in the IgH gene (Igh) to stimulate isotype class switch recombination (CSR), and widespread breaks in non-Igh (off-target) loci throughout the genome. Because the DSBs that initiate class switching occur during the G1 phase of the cell cycle, and are repaired via end joining, CSR is considered a predominantly G1 reaction. By contrast, AID-induced non-Igh DSBs are repaired by homologous recombination. Although little is known about the connection between the cell cycle and either induction or resolution of AID-mediated non-Igh DSBs, their repair by homologous recombination implicates post-G1 phases. Coordination of DNA breakage and repair during the cell cycle is critical to promote normal class switching and prevent genomic instability. To understand how AID-mediated events are regulated through the cell cycle, we have investigated G1-to-S control in AID-dependent genome-wide DSBs. We find that AID-mediated off-target DSBs, like those induced in the Igh locus, are generated during G1. These data suggest that AID-mediated DSBs can evade G1/S checkpoint activation and persist beyond G1, becoming resolved during S phase. Interestingly, DSB resolution during S phase can promote not only non-Igh break repair, but also Ig CSR. Our results reveal novel cell cycle dynamics in response to AID-initiated DSBs, and suggest that the regulation of the repair of these DSBs through the cell cycle may ensure proper class switching while preventing AID-induced genomic instability.


Asunto(s)
Citidina Desaminasa/fisiología , Roturas del ADN de Doble Cadena , Cambio de Clase de Inmunoglobulina/genética , Isotipos de Inmunoglobulinas/genética , Fase S/genética , Fase S/inmunología , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células Cultivadas , Citidina Desaminasa/deficiencia , Citidina Desaminasa/genética , Reparación del ADN/genética , Reparación del ADN/inmunología , Fase G1/genética , Fase G1/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
Immunol Res ; 51(1): 45-60, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21850539

RESUMEN

Complement system activation plays an important role in both innate and acquired immunity. Activation of the complement and the subsequent formation of C5b-9 channels (the membrane attack complex) on the cell membranes lead to cell death. However, when the number of channels assembled on the surface of nucleated cells is limited, sublytic C5b-9 can induce cell cycle progression by activating signal transduction pathways and transcription factors and inhibiting apoptosis. This induction by C5b-9 is dependent upon the activation of the phosphatidylinositol 3-kinase/Akt/FOXO1 and ERK1 pathways in a Gi protein-dependent manner. C5b-9 induces sequential activation of CDK4 and CDK2, enabling the G1/S-phase transition and cellular proliferation. In addition, it induces RGC-32, a novel gene that plays a role in cell cycle activation by interacting with Akt and the cyclin B1-CDC2 complex. C5b-9 also inhibits apoptosis by inducing the phosphorylation of Bad and blocking the activation of FLIP, caspase-8, and Bid cleavage. Thus, sublytic C5b-9 plays an important role in cell activation, proliferation, and differentiation, thereby contributing to the maintenance of cell and tissue homeostasis.


Asunto(s)
Apoptosis/inmunología , Membrana Celular/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Animales , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/inmunología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Proteína Quinasa CDC2 , Caspasa 8/inmunología , Caspasa 8/metabolismo , Proteínas de Ciclo Celular/inmunología , Proteínas de Ciclo Celular/metabolismo , Membrana Celular/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Ciclina B/inmunología , Ciclina B/metabolismo , Quinasa 2 Dependiente de la Ciclina/inmunología , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Fase G1/inmunología , Humanos , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Musculares/inmunología , Proteínas Musculares/metabolismo , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/metabolismo , Fosfatidilinositol 3-Quinasas/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fase S/inmunología , Proteína Letal Asociada a bcl/inmunología , Proteína Letal Asociada a bcl/metabolismo
7.
Mol Immunol ; 46(16): 3454-61, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19748123

RESUMEN

PPARalpha ligands are medications used clinically to prevent cardiovascular events, however studies have shown that these agents are also anti-inflammatory. Our previous studies have shown that PPARalpha ligands induce lymphocyte depletion. PPARalpha ligands also potently upregulate TRB3, a protein that has been associated with cell cycle arrest. Therefore the following studies were undertaken to determine the mechanisms associated with lymphocyte depletion. Our studies demonstrate that WY14,643, a PPARalpha ligand, decreases the amount of lymphocytes recovered after stimulation and reduces cellular divisions. Cells treated with WY14,643 also accumulate in the G2/S phase of the cell cycle. TRB3 has been shown to inhibit the phosphorylation of AKT/Protein Kinase B, and reduced activation of AKT has been associated with decreased cellular divisions and survival. However in lymphocytes, TRB3 did not reduce the phosphorylation of AKT, and WY14,643 treatment was associated with enhanced activation of AKT. Drosophila tribbles (TRB3 homolog) causes G2 arrest by decreasing the expression of a Cdc25c homolog. Lymphocytes stimulated and treated with WY14,643 have reduced expression of Cdc25c, however this is not associated with enhanced expression of phosphorylated-Cdc2 which induces G2 arrest. Instead we observed that WY14,643 consistently reduces the protein and mRNA expression of Cyclin B1. Moreover, TRB3 inhibits activation of a Cyclin B1 promoter construct. In summary, we propose that PPARalpha ligands may reduce cellular number by augmenting TRB3 expression, which in turn induces cell cycle arrest by reducing the expression of Cyclin B1. Reduced cellular divisions and cell cycle arrest may be responsible for some of the immunomodulatory effects of these agents that have been consistently observed in human trials.


Asunto(s)
Proteínas de Ciclo Celular/inmunología , Fase G2/efectos de los fármacos , Linfocitos/inmunología , PPAR alfa/agonistas , PPAR alfa/inmunología , Proliferadores de Peroxisomas/farmacología , Pirimidinas/farmacología , Fase S/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Animales , Proteínas de Ciclo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Ciclina B/biosíntesis , Ciclina B/inmunología , Ciclina B1 , Fase G2/inmunología , Humanos , Ligandos , Depleción Linfocítica , Ratones , Ratones Endogámicos BALB C , PPAR alfa/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/inmunología , Regiones Promotoras Genéticas/inmunología , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-akt/inmunología , Fase S/inmunología , Regulación hacia Arriba/inmunología , Fosfatasas cdc25/inmunología
8.
Blood ; 113(22): 5506-15, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19339697

RESUMEN

The canonical mode of transcriptional activation by both the Epstein-Barr viral protein, Epstein-Barr virus-encoded nuclear antigen 2 (EBNA2), and an activated Notch receptor (Notch-IC) requires their recruitment to RBPJ, suggesting that EBNA2 uses the Notch pathway to achieve B-cell immortalization. To gain further insight into the biologic equivalence between Notch-IC and EBNA2, we performed a genome-wide expression analysis, revealing that Notch-IC and EBNA2 exhibit profound differences in the regulation of target genes. Whereas Notch-IC is more potent in regulating genes associated with differentiation and development, EBNA2 is more potent in inducing viral and cellular genes involved in proliferation, survival, and chemotaxis. Because both EBNA2 and Notch-IC induced the expression of cell cycle-associated genes, we analyzed whether Notch1-IC or Notch2-IC can replace EBNA2 in B-cell immortalization. Although Notch-IC could drive quiescent B cells into the cell cycle, B-cell immortalization was not maintained, partially due to an increased apoptosis rate in Notch-IC-expressing cells. Expression analysis revealed that both EBNA2 and Notch-IC induced the expression of proapoptotic genes, but only in EBNA2-expressing cells were antiapoptotic genes strongly up-regulated. These findings suggest that Notch signaling in B cells and B-cell lymphomas is only compatible with proliferation if pathways leading to antiapototic signals are active.


Asunto(s)
Linfocitos B/fisiología , Proliferación Celular , Infecciones por Virus de Epstein-Barr/inmunología , Antígenos Nucleares del Virus de Epstein-Barr/fisiología , Receptor Notch1/fisiología , Receptor Notch2/fisiología , Proteínas Virales/fisiología , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/virología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Transformación Celular Viral/inmunología , Células Cultivadas , Infecciones por Virus de Epstein-Barr/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Modelos Biológicos , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Proteínas Recombinantes/farmacología , Fase S/efectos de los fármacos , Fase S/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Proteínas Virales/metabolismo , Proteínas Virales/farmacología
9.
Hybridoma (Larchmt) ; 27(2): 107-11, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18642675

RESUMEN

The effect of mild hypothermic (32 degrees C) conditions on cell growth, cell-cycle progress, and antibody production of hybridoma C2E7 cells was investigated in the present study. The growth of hybridoma cells was slower during the mild hypothermic condition compared to that at 37 degrees C; this led to about 10% decrease in maximum viable cell density and volumetric antibody productivity. However, under mild hypothermic growth conditions, the culture viability was substantially improved and the specific antibody productivity was enhanced compared to that at 37 degrees C. The average specific productivity for the entire batch culture at 32 degrees C is about 5% higher than that at 37 degrees C. Cell-cycle analysis data showed that there was no growth arrestment during the mild hypothermic growth of hybridoma cells. The G1-phase cells were increased, while the S-phase cells were decreased gradually as the culture time progressed. Further analysis showed that the specific antibody productivity of hybridoma cells was correlated to the fraction of S-phase cells.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Ciclo Celular/inmunología , Proliferación Celular , Frío , Hibridomas/inmunología , Hibridomas/metabolismo , Inmunoglobulina M/biosíntesis , Animales , Especificidad de Anticuerpos , Células CHO , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Cricetinae , Cricetulus , Humanos , Ratones , Ratones Endogámicos BALB C , Fase S/inmunología
10.
J Microsc ; 231(Pt 1): 28-37, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18638187

RESUMEN

Current understanding of the mechanisms involved in osseointegration following implantation of a biomaterial has led to adhesion quantification being implemented as an assay of cytocompatibility. Such measurement can be hindered by intra-sample variation owing to morphological changes associated with the cell cycle. Here we report on a new scanning electron microscopical method for the simultaneous immunogold labelling of cellular focal adhesions and S-phase nuclei identified by BrdU incorporation. Prior to labelling, cellular membranes are removed by tritonization and antigens of non-interest blocked by serum incubation. Adhesion plaque-associated vinculin and S-phase nuclei were both separately labelled with a 1.4 nm gold colloid and visualized by subsequent colloid enhancement via silver deposition. This study is specifically concerned with the effects microgroove topographies have on adhesion formation in S-phase osteoblasts. By combining backscattered electron (BSE) imaging with secondary electron (SE) imaging it was possible to visualize S-phase nuclei and the immunogold-labelled adhesion sites in one energy 'plane' and the underlying nanotopography in another. Osteoblast adhesion to these nanotopographies was ascertained by quantification of adhesion complex formation.


Asunto(s)
Fémur/citología , Adhesiones Focales , Microscopía Electrónica de Rastreo/métodos , Osteoblastos/ultraestructura , Fase S , Anciano de 80 o más Años , Adhesión Celular , Ciclo Celular , Células Cultivadas , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Osteoblastos/citología , Fase S/inmunología , Fase S/fisiología
11.
J Immunol ; 179(12): 8305-12, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18056375

RESUMEN

Heat shock factor-1 (HSF1) is a transcription factor that serves as the major temperature-inducible sensor for eukaryotic cells. In most cell types, HSF1 becomes activated to the DNA binding form at 42 degrees C and mediates the classical heat shock response, protecting the cells from subsequent lethal temperatures. We have recently demonstrated that HSF1 is activated at a lower temperature in T lymphocytes than in most other cell types (39 degrees C vs 42 degrees C), within the physiological range of fever. In this study, we show that T cell activation at fever temperatures not only activates HSF1 but induces the up-regulation of the HSF1 protein and the HSF1-regulated protein, HSP70i. T cells from HSF1 knockout mice proliferate normally under optimal conditions but are impaired in proliferation at physiological fever temperatures and low CO2 concentrations, conditions that do not impair wild-type T cells. This defect in proliferation appears to be mediated by a block in the G1/S transition of the cell cycle and is independent of HSP70. Elevated temperature and low CO2 concentrations resulted in a dramatic reduction of the intracellular reactive oxygen species (ROS) levels in both normal and knockout T cells. Wild-type T cells were able to restore ROS levels to normal within 5 h, whereas HSF1-/- T cells were not. These results suggest that the proliferation defect seen in T cells from HSF1-/- mice at fever temperatures was because of dysregulated ROS levels and that HSF1 is important in maintaining ROS homeostasis and cell cycle progression under the stressful conditions encountered during fever.


Asunto(s)
Temperatura Corporal/inmunología , Proteínas de Unión al ADN/fisiología , Fiebre/inmunología , Linfocitos T/inmunología , Factores de Transcripción/fisiología , Animales , Proliferación Celular , Proteínas de Unión al ADN/genética , Fase G1/genética , Fase G1/inmunología , Proteínas HSP70 de Choque Térmico/metabolismo , Factores de Transcripción del Choque Térmico , Activación de Linfocitos , Ratones , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Fase S/genética , Fase S/inmunología , Factores de Transcripción/genética
12.
J Immunol ; 179(8): 5291-300, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17911615

RESUMEN

Fas-associated death domain protein (FADD) constitutes an essential component of TNFR-induced apoptotic signaling. Paradoxically, FADD has also been shown to be crucial for lymphocyte development and activation. In this study, we report that FADD is necessary for long-term maintenance of S6 kinase (S6K) activity. S6 phosphorylation at serines 240 and 244 was only observed after long-term stimulation of wild-type cells, roughly corresponding to the time before S-phase entry, and was poorly induced in T cells expressing a dominantly interfering form of FADD (FADDdd), viral FLIP, or possessing a deficiency in caspase-8. Defects in S6K1 phosphorylation were also observed. However, defective S6K1 phosphorylation was not a consequence of a wholesale defect in mammalian target of rapamycin function, because 4E-BP1 phosphorylation following T cell activation was unaffected by FADDdd expression. Although cyclin D3 up-regulation and retinoblastoma hypophosphorylation occurred normally in FADDdd T cells, cyclin E expression and cyclin-dependent kinase 2 activation were markedly impaired in FADDdd T cells. These results demonstrate that a FADD/caspase-8-signaling axis promotes T cell cycle progression and sustained S6K activity.


Asunto(s)
Caspasa 8/fisiología , Proteína de Dominio de Muerte Asociada a Fas/fisiología , Interleucina-2/fisiología , Proteínas Quinasas S6 Ribosómicas/metabolismo , Fase S/inmunología , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/enzimología , Síndrome de Alstrom , Animales , Caspasa 8/genética , Células Cultivadas , Quinasa 2 Dependiente de la Ciclina/deficiencia , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 2 Dependiente de la Ciclina/fisiología , Activación Enzimática/genética , Activación Enzimática/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Proteínas Quinasas S6 Ribosómicas/deficiencia , Fase S/genética , Transducción de Señal/genética , Subgrupos de Linfocitos T/inmunología
13.
Eur J Immunol ; 36(6): 1583-97, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16703564

RESUMEN

Twenty-four hours of TCR engagement and CD28 costimulation was found sufficient to elicit an optimal rate of cell division over a 72-h period only when a high concentration of IL-2 was produced in the culture and remained readily available to the CD4(+) T cells. The cell division response could be aborted following 24 h of stimulation by the simultaneous abrogation of IL-2R signaling and the blockade of CD28 or TCR ligands. Biochemical and pharmacologic studies indicated that a phosphatidylinositol 3-kinase-Akt signaling cascade costimulated by the TCR and CD28 maintained the blasting cell division rate at a maximal level beyond 24 h even when IL-2 was withdrawn, neutralized, or exhausted. These data show that CD4(+) T cells remain sensitive to antigens (Ag) and costimulatory signals throughout the clonal expansion response. Furthermore, only those T cells that perceive the presence of a continued threat in the form of Ag/MHC complexes and B7 costimulatory ligands or a high concentration of a growth factor are directed to remain in cell cycle.


Asunto(s)
Antígeno B7-1/metabolismo , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/citología , Fase G1/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Fase S/fisiología , Animales , Antígeno B7-1/inmunología , Western Blotting , Antígenos CD28/inmunología , Linfocitos T CD4-Positivos/inmunología , Procesos de Crecimiento Celular/fisiología , Ciclosporina/farmacología , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Fase G1/inmunología , Interleucina-2/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Proteínas Quinasas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Fase S/inmunología , Transducción de Señal , Serina-Treonina Quinasas TOR
14.
Blood ; 107(12): 4807-16, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16484589

RESUMEN

CD74 is an integral membrane protein that was thought to function mainly as an MHC class II chaperone. However, CD74 was recently shown to have a role as an accessory-signaling molecule. Our studies demonstrated that CD74 regulates B-cell differentiation by inducing a pathway leading to the activation of transcription mediated by the NF-kappaB p65/RelA homodimer and its coactivator, TAF(II)105. Here, we show that CD74 stimulation with anti-CD74 antibody leads to an induction of a signaling cascade resulting in NF-kappaB activation, entry of the stimulated cells into the S phase, elevation of DNA synthesis, cell division, and augmented expression of BCL-X(L). These studies therefore demonstrate that surface CD74 functions as a survival receptor.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/inmunología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Activación de Linfocitos/inmunología , Fase S/inmunología , Transducción de Señal/inmunología , Animales , Antígenos de Diferenciación de Linfocitos B/genética , Linfocitos B/citología , Diferenciación Celular/genética , Supervivencia Celular/genética , Supervivencia Celular/inmunología , ADN/genética , ADN/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Ratones , Ratones Noqueados , Transducción de Señal/genética , Factores Asociados con la Proteína de Unión a TATA/genética , Factores Asociados con la Proteína de Unión a TATA/inmunología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/inmunología , Factor de Transcripción TFIID/genética , Factor de Transcripción TFIID/inmunología , Transcripción Genética/genética , Transcripción Genética/inmunología , Células Tumorales Cultivadas , Proteína bcl-X/genética , Proteína bcl-X/inmunología
15.
Atherosclerosis ; 187(2): 301-8, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16289073

RESUMEN

The pleiotropic effects of statin, including its anti-inflammatory effects, via chemokines may be independent of statin-induced cholesterol reduction. Therefore, we examined the effect of pitavastatin on cell proliferation and the association between chemokine receptors (CCR2 and CCR5) and their ligands, RANTES (regulated upon activation, normal T cell-expressed and secreted) and monocyte chemotactic protein-1 (MCP-1), in monocytes. Pitavastatin but not pravastatin inhibited cell proliferation in a dose-dependent manner and showed S-phase arrest associated with the downregulation of CCR2 and CCR5 expression in human monocytic tumor cells (U937 cells). Although the anti-proliferative effects of pitavastatin were not inhibited by lower concentrations of RANTES and MCP-1, overexpression of CCR2/CCR5 significantly blocked the anti-proliferation with a low concentration of RANTES or MCP-1. Pitavastatin upregulated p21(waf1) but not p27(kip1), and did not change the expression levels of cyclin D1 or cdk4. In addition, RANTES and MCP-1 upregulated cyclin D1 in the presence of pitavastatin. In conclusion, the anti-proliferative effect of pitavastatin, but not pravastatin, through the downregulation of CCR2/CCR5 may be a pleiotropic effect. This effect may be anti-atherogenic in monocytes.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Monocitos/efectos de los fármacos , Quinolinas/farmacología , Receptores CCR5/metabolismo , Receptores de Quimiocina/metabolismo , Anticolesterolemiantes/farmacología , División Celular/efectos de los fármacos , División Celular/inmunología , Quimiocina CCL2/metabolismo , Quimiocina CCL2/farmacología , Quimiocina CCL5/metabolismo , Quimiocina CCL5/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Humanos , Monocitos/citología , Monocitos/metabolismo , Pravastatina/farmacología , Receptores CCR2 , Fase S/efectos de los fármacos , Fase S/inmunología , Células U937 , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
16.
J Leukoc Biol ; 78(6): 1378-85, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16204634

RESUMEN

Interleukin-3 (IL-3) mediates hematopoietic cell survival and proliferation via several signaling pathways such as the Janus kinase/signal transducer and activator of transcription pathway, mitogen-activated protein kinase (MAPK) pathway, and phosphoinositide-3 kinase (PI-3K) pathway. Mammalian target of rapamycin (mTOR) is one of the downstream targets of the PI-3K pathway, and it plays an important role in hematopoiesis and immune cell function. To better elucidate how mTOR mediates proliferation signals from IL-3, we assessed the role of S6 kinase 2 (S6K2), one of the downstream targets of mTOR, in IL-3 signaling. We show that S6K2 is activated by IL-3 in the IL-3-dependent Ba/F3 cell line and that this is mediated by mTOR and its upstream activator PI-3K but not by the MAPK kinase/extracellular signal-regulated kinase pathway. S6K2 is also activated in primary mouse bone marrow-derived mast cells upon IL-3 stimulation. Expression of a rapamycin-resistant form of S6K2, T388E, in Ba/F3 cells provides a proliferation advantage in the absence or presence of rapamycin, indicating that S6K2 can potentiate IL-3-mediated mitogenic signals. In cells expressing T388E, rapamycin still reduces proliferation at all doses of rapamycin, showing that mTOR targets other than S6K2 play an important role in IL-3-dependent proliferation. Cell-cycle analysis shows that T388E-expressing Ba/F3 cells enter S phase earlier than the control cells, indicating that the proliferation advantage may be mediated by a shortened G1 phase. This is the first indication that S6K2 plays a role in IL-3-dependent cell proliferation.


Asunto(s)
Células de la Médula Ósea/enzimología , Proliferación Celular/efectos de los fármacos , Interleucina-3/metabolismo , Mastocitos/enzimología , Proteínas Quinasas/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , División Celular/efectos de los fármacos , División Celular/inmunología , Línea Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Fase G1/efectos de los fármacos , Fase G1/inmunología , Humanos , Interleucina-3/farmacología , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Ratones , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Fase S/efectos de los fármacos , Fase S/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR
17.
J Immunol ; 175(2): 647-55, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16002659

RESUMEN

Homeostasis-driven T cell proliferation occurs in response to a lymphopenic environment and is mediated by TCR and IL-7 signaling. In this report, we demonstrate a defect in the proliferation of murine naive and memory T cells lacking both E2F1 and E2F2 in response to lymphopenic conditions, suggesting that E2F1 and E2F2 function redundantly downstream of TCR and/or IL-7 signaling during homeostasis-driven proliferation. In contrast, T cell proliferation in response to antigenic stimulation is either unaffected (in vivo) or potentiated (ex vivo) by loss of E2F1 and E2F2, indicating divergent requirements for these E2F factors in T cell proliferation mediated by distinct stimuli. E2F1/E2F2 double knockout (DKO) T cells enter S phase in response to homeostatic signaling, but fail to divide, suggesting that S phase progression is either incomplete or defective. In addition, E2F1/E2F2 DKO mice do not recover normal T cell numbers following exposure to a sublethal dose of radiation, indicating that this defect in homeostasis-driven proliferation is physiologically relevant. Consistent with their failure in cell cycle progression, the differentiation of DKO T cells into memory T cells in response to homeostatic signals is significantly reduced. These observations support the idea that proliferation is required for memory T cell formation and also have implications for the development of clinical strategies to minimize the occurrence of lymphopenia-induced autoimmunity.


Asunto(s)
Antígenos/administración & dosificación , Antígenos/inmunología , Proteínas de Ciclo Celular/fisiología , Proliferación Celular , Proteínas de Unión al ADN/fisiología , Homeostasis/inmunología , Linfocitos T/inmunología , Factores de Transcripción/fisiología , Animales , Proteínas de Ciclo Celular/genética , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Factor de Transcripción E2F2 , Homeostasis/genética , Memoria Inmunológica/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Fase S/genética , Fase S/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/citología , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
18.
J Infect Dis ; 192(1): 62-70, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15942895

RESUMEN

Heightened proliferation and death of T lymphocytes may play a key role in human immunodeficiency virus (HIV) pathogenesis; however, the mechanism that mediates this effect and the phenotype of the proliferating T cells have not been clearly determined. We assessed S-phase cell frequencies and phenotype by ex vivo bromodeoxyuridine incorporation and flow-cytometric analysis in a group of 35 HIV-infected individuals. Frequencies of S-phase T cells were increased in HIV disease and were related to plasma HIV RNA levels but not to CD4 cell, total T cell, or total lymphocyte counts. S-phase cells were phenotypically defined as "central memory" cells (CD45RO+CD62L+CCR7+). Although activated (CD38+), S-phase cells lacked CD69 expression, rarely expressed CD25, and were not overrepresented among HIV-specific cells, as might have been expected if these cells had recently been activated by HIV antigens. Thus, in HIV infection, central memory T cells may be highly susceptible to bystander mechanisms of immune activation, leading to S-phase entry.


Asunto(s)
Infecciones por VIH/inmunología , Memoria Inmunológica/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Fase S/inmunología , Linfocitos T/inmunología , Adulto , Antígenos CD/fisiología , Terapia Antirretroviral Altamente Activa , Bromodesoxiuridina , Proliferación Celular , Humanos , Activación de Linfocitos , Recuento de Linfocitos , Fenotipo , Replicación Viral/fisiología
19.
J Immunol ; 174(11): 7226-33, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15905568

RESUMEN

Phagocytosis of microorganisms by macrophages is an important host defense mechanism. While studying the phagocytosis of the human pathogenic fungus Cryptococcus neoformans, we noted that macrophage-like J774 cells with ingested fungal cells had frequent mitotic figures. By analyzing the relative proportion of phagocytic cells as a function of cell cycle phase, we observed an increase in S phase cells after Fc-mediated phagocytosis of polystyrene beads, live or heat-killed C. neoformans. This result was confirmed by increased nuclear BrdU incorporation after Fc-mediated phagocytosis. The induced progression to S phase was observed after both Fc- and complement-mediated phagocytosis of live yeasts. Fc-mediated stimulation of cell division did not require ingestion, because it could be triggered by incubating cells in IgG1-coated plates. Phagocytosis-mediated stimulation of replication was confirmed in vitro using primary bone marrow macrophages and in vivo for peritoneal macrophages. We conclude that phagocytosis of microbes or inert particles can stimulate macrophages to enter S phase and commence cell division. This observation suggests a potential mechanism for increasing the number of effector cells after microbial ingestion, but can also promote the spread of infection.


Asunto(s)
Ciclo Celular/inmunología , Fase G1/inmunología , Macrófagos/citología , Macrófagos/inmunología , Receptores de Complemento/metabolismo , Receptores Fc/metabolismo , Fase S/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Bromodesoxiuridina/metabolismo , Recuento de Células , Línea Celular Tumoral , Células Cultivadas , Activación de Complemento/inmunología , Cryptococcus neoformans/inmunología , Cryptococcus neoformans/metabolismo , Inmunoglobulina G/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fagocitosis/inmunología , Receptores de Complemento/fisiología , Receptores Fc/fisiología
20.
Blood ; 105(7): 2900-7, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15585652

RESUMEN

Recurrent chromosomal rearrangements are associated with the development of acute myeloid leukemia (AML). The frequent inversion of chromosome 16 creates the CBFB-MYH11 fusion gene that encodes the fusion protein CBFbeta-SMMHC. This fusion protein inhibits the core-binding factor (CBF), resulting in a block of hematopoietic differentiation, and induces leukemia upon the acquisition of additional mutations. A recent genetic screen identified Plag1 and Plagl2 as CBF beta-SMMHC candidate cooperating proteins. In this study, we demonstrate that Plag1 and Plagl2 independently cooperate with CBF beta-SMMHC in vivo to efficiently trigger leukemia with short latency in the mouse. In addition, Plag1 and Plagl2 increased proliferation by inducing G1 to S transition that resulted in the expansion of hematopoietic progenitors and increased cell renewal in vitro. Finally, PLAG1 and PLAGL2 expression was increased in 20% of human AML samples. Interestingly, PLAGL2 was preferentially increased in samples with chromosome 16 inversion, suggesting that PLAG1 and PLAGL2 may also contribute to human AML. Overall, this study shows that Plag1 and Plagl2 are novel leukemia oncogenes that act by expanding hematopoietic progenitors expressing CbF beta-SMMHC.


Asunto(s)
Proteínas de Unión al ADN/genética , Leucemia Mieloide/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Unión al ARN/genética , Factores de Transcripción/genética , Enfermedad Aguda , Adolescente , Adulto , Animales , Proteínas de Unión al ADN/metabolismo , Femenino , Fase G1/inmunología , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/citología , Humanos , Leucemia Mieloide/fisiopatología , Masculino , Ratones , Ratones Mutantes , Persona de Mediana Edad , Mutagénesis Insercional , Proteínas de Fusión Oncogénica/metabolismo , Proteínas de Unión al ARN/metabolismo , Retroviridae/genética , Fase S/inmunología , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA