Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Biosci ; 14(1): 15, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38291538

RESUMEN

BACKGROUND: Sexual dimorphism represents a key concept in the comprehension of molecular processes guiding several sex-specific physiological and pathological mechanisms. It has been reported that genes involved in many disorders show a sex-dependent expression pattern. Moreover, the loss of Y chromosome (LOY), found to be a physiological age-driven phenomenon, has been linked to many neurodegenerative and autoimmune disorders, and to an increased cancer risk. These findings drove us towards the consideration that LOY may cause the de-regulation of disease specific networks, involving genes located in both autosomal and sex chromosomes. RESULTS: Exploiting the CRISPR/Cas9 and RNA-sequencing technologies, we generated a Y-deficient human cell line that has been investigated for its gene expression profile. Our results showed that LOY can influence the transcriptome displaying relevant enriched biological processes, such as cell migration regulation, angiogenesis and immune response. Interestingly, the ovarian follicle development pathway was found enriched, supporting the female-mimicking profile of male Y-depleted cells. CONCLUSION: This study, besides proposing a novel approach to investigate sex-biased physiological and pathological conditions, highlights new roles for the Y chromosome in the sexual dimorphism characterizing human health and diseases. Moreover, this analysis paves the way for the research of new therapeutic approaches for sex dimorphic and LOY-related diseases.

2.
Cell Death Dis ; 14(3): 197, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36918555

RESUMEN

Females have a lower probability to develop somatic cancers and a better response to chemotherapy than males. However, the reasons for these differences are still not well understood. The X-linked gene TSPY-Like 2 (TSPYL2) encodes for a putative tumor suppressor protein involved in cell cycle regulation and DNA damage response (DDR) pathways. Here, we demonstrate that in unstressed conditions TSPYL2 is maintained at low levels by MDM2-dependent ubiquitination and proteasome degradation. Upon genotoxic stress, E2F1 promotes TSPYL2 expression and protein accumulation in non-transformed cell lines. Conversely, in cancer cells, TSPYL2 accumulates only in females or in those male cancer cells that lost the Y-chromosome during the oncogenic process. Hence, we demonstrate that while TSPYL2 mRNA is induced in all the tested tumor cell lines after DNA damage, TSPYL2 protein stability is increased only in female cancer cells. Indeed, we found that TSPYL2 accumulation, in male cancer cells, is prevented by the Y-encoded protein SRY, which modulates MDM2 protein levels. In addition, we demonstrated that TSPYL2 accumulation is required to sustain cell growth arrest after DNA damage, possibly contributing to protect normal and female cancer cells from tumor progression. Accordingly, TSPYL2 has been found more frequently mutated in female-specific cancers. These findings demonstrate for the first time a sex-specific regulation of TSPYL2 in the DDR of cancer cells and confirm the existence of sexual dimorphism in DNA surveillance pathways.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Neoplasias , Femenino , Humanos , Masculino , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Daño del ADN/genética , Neoplasias/genética , Proteínas Supresoras de Tumor/genética , Proteínas de Unión al ADN/genética
3.
iScience ; 25(3): 103875, 2022 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-35243237

RESUMEN

Cancer incidence and survival are different between men and women. Indeed, females have a lesser risk and a better prognosis than males in many tumors unrelated to reproductive functions. Although the reasons for these disparities are still unknown, they constitute an important starting point for the development of personalized cancer therapies. One of the mechanisms that fuels carcinogenesis is the accumulation of defects in DNA damage response (DDR) pathways, a complex signaling cascade that senses DNA lesions and, depending on the severity, coordinates transient cell-cycle arrest, DNA replication, repair, apoptosis, and senescence, preventing genomic instability and cancer. Recently, evidence of sexual dimorphisms is emerging in these pathways, therefore providing new opportunities for precision medicine. Here, we will discuss current knowledge about sexual disparities in the DDR, their role in tumorigenesis and cancer progression, and the importance of considering sex contribution in both research and cancer therapies.

4.
Cell Death Differ ; 26(5): 918-931, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30050056

RESUMEN

Protein acetylation and deacetylation events are finely regulated by lysine-acetyl-transferases and lysine-deacetylases and constitute an important tool for the activation or inhibition of specific cellular pathways. One of the most important lysine-acetyl-transferases is p300, which is involved in the regulation of gene expression, cell growth, DNA repair, differentiation, apoptosis, and tumorigenesis. A well-known target of p300 is constituted by the tumor suppressor protein p53, which plays a critical role in the maintenance of genomic stability and whose activity is known to be controlled by post-translational modifications, among which acetylation. p300 activity toward p53 is negatively regulated by the NAD-dependent deacetylase SIRT1, which deacetylates p53 preventing its transcriptional activation and the induction of p53-dependent apoptosis. However, the mechanisms responsible for p53 regulation by p300 and SIRT1 are still poorly understood. Here we identify the nucleosome assembly protein TSPY-Like 2 (TSPYL2, also known as TSPX, DENTT, and CDA1) as a novel regulator of SIRT1 and p300 function. We demonstrate that, upon DNA damage, TSPYL2 inhibits SIRT1, disrupting its association with target proteins, and promotes p300 acetylation and activation, finally stimulating p53 acetylation and p53-dependent cell death. Indeed, in response to DNA damage, cells silenced for TSPYL2 were found to be defective in p53 activation and apoptosis induction and these events were shown to be dependent on SIRT1 and p300 function. Collectively, our results shed new light on the regulation of p53 acetylation and activation and reveal a novel TSPYL2 function with important implications in cancerogenesis.


Asunto(s)
Proteína p300 Asociada a E1A/genética , Neoplasias Pulmonares/genética , Proteínas Nucleares/genética , Sirtuina 1/genética , Proteína p53 Supresora de Tumor/genética , Células A549 , Acetilación/efectos de los fármacos , Apoptosis/efectos de los fármacos , Camptotecina/farmacología , Proliferación Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina/genética , Daño del ADN/efectos de los fármacos , Proteínas de Unión al ADN , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Etopósido/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Procesamiento Proteico-Postraduccional/genética , Activación Transcripcional/efectos de los fármacos , Gemcitabina
5.
Mutat Res Rev Mutat Res ; 776: 1-9, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29807573

RESUMEN

Cell cycle and apoptosis regulator 2 (CCAR2 or DBC1) is a human protein recently emerged as a novel and important player of the DNA damage response (DDR). Indeed, upon genotoxic stress, CCAR2, phosphorylated by the apical DDR kinases ATM and ATR, increases its binding to the NAD+-dependent histone deacetylase SIRT1 and inhibits SIRT1 activity. This event promotes the acetylation and activation of p53, a SIRT1 target, and the subsequent induction of p53 dependent apoptosis. In addition, CCAR2 influences DNA repair pathway choice and promotes the chromatin relaxation necessary for the repair of heterochromatic DNA lesions. However, besides DDR, CCAR2 is involved in several other cellular functions. Indeed, through the interaction with transcription factors, nuclear receptors, epigenetic modifiers and RNA polymerase II, CCAR2 regulates transcription and transcript elongation. Moreover, promoting Rev-erbα protein stability and repressing BMAL1 and CLOCK expression, it was reported to modulate the circadian rhythm. Through SIRT1 inhibition, CCAR2 is also involved in metabolism control and, suppressing RelB and p65 activities in the NFkB pathway, it restricts B cell proliferation and immunoglobulin production. Notably, CCAR2 expression is deregulated in several tumors and, compared to the non-neoplastic counterpart, it may be up- or down-regulated. Since its up-regulation in cancer patients is usually associated with poor prognosis and its depletion reduces cancer cell growth in vitro, CCAR2 was suggested to act as a tumor promoter. However, there is also evidence that CCAR2 functions as a tumor suppressor and therefore its role in cancer formation and progression is still unclear. In this review we discuss CCAR2 functions in the DDR and its multiple biological activities in unstressed cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Daño del ADN/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Apoptosis/genética , Apoptosis/fisiología , Linfocitos B/inmunología , Linfocitos B/fisiología , Senescencia Celular/genética , Senescencia Celular/fisiología , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Relojes Circadianos/genética , Relojes Circadianos/fisiología , Daño del ADN/genética , Reparación del ADN/genética , Reparación del ADN/fisiología , Epigénesis Genética , Humanos , Modelos Biológicos , Mutación , Neoplasias/etiología , Sirtuina 1/genética , Sirtuina 1/fisiología , Transcripción Genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/fisiología
6.
Cell Death Dis ; 7(11): e2453, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27809307

RESUMEN

Human CCAR2 has recently emerged as having a pivotal role in the DNA damage response, promoting apoptosis and repair of heterochromatic DNA breaks. However, less is known about the function of CCAR2 in tumor formation and cancer progression. Here, we demonstrate, for the first time, that CCAR2 loss inhibits the proliferation of cancer cells, but preserves the growth of normal cells. Investigating the mechanisms responsible for this differential effect, we found that CCAR2 depletion specifically impairs the activation of AKT pathway in cancer cells, but not in normal cells, by reducing AKT phosphorylation on Ser473. This effect is achieved through the transcriptional upregulation of TRB3 gene and accumulation of TRB3 protein, which then binds to and inhibits the phosphorylation and activation of AKT. The defective activation of AKT finally results in reduced GSK3ß phosphorylation, prevention of G1/S transition and inhibition of cancer cell growth. These results establish an important role for CCAR2 in cancer cells proliferation and could shed new light on novel therapeutic strategies against cancer, devoid of detrimental side effects.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Activación Enzimática , Fase G1 , Humanos , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Fase S
7.
Oncotarget ; 6(19): 17817-31, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-26158765

RESUMEN

Cell cycle and apoptosis regulator 2 (CCAR2, formerly known as DBC1) is a nuclear protein largely involved in DNA damage response, apoptosis, metabolism, chromatin structure and transcription regulation. Upon DNA lesions, CCAR2 is phosphorylated by the apical kinases ATM/ATR and this phosphorylation enhances CCAR2 binding to SIRT1, leading to SIRT1 inhibition, p53 acetylation and p53-dependent apoptosis. Recently, we found that also the checkpoint kinase Chk2 and the proteasome activator REGγ are required for efficient CCAR2-mediated inhibition of SIRT1 and induction of p53-dependent apoptosis.Here, we report that CCAR2 is required for the repair of heterochromatic DNA lesions, as cells knock-out for CCAR2 retain, at late time-points after genotoxic treatment, abnormal levels of DNA damage-associated nuclear foci, whose timely resolution is reinstated by HP1ß depletion. Conversely, repair of DNA damages in euchromatin are not affected by CCAR2 absence.We also report that the impairment in heterochromatic DNA repair is caused by defective Chk2 activation, detectable in CCAR2 ablated cells, which finally impacts on the phosphorylation of the Chk2 substrate KAP1 that is required for the induction of heterochromatin relaxation and DNA repair.These studies further extend and confirm the role of CCAR2 in the DNA damage response and DNA repair and illustrate a new mechanism of Chk2 activity regulation. Moreover, the involvement of CCAR2 in the repair of heterochromatic DNA breaks suggests a new role for this protein in the maintenance of chromosomal stability, which is necessary to prevent cancer formation.


Asunto(s)
Quinasa de Punto de Control 2/metabolismo , Daño del ADN/fisiología , Reparación del ADN/fisiología , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Western Blotting , Proteínas de Ciclo Celular , Línea Celular Tumoral , Homólogo de la Proteína Chromobox 5 , Técnica del Anticuerpo Fluorescente , Técnicas de Inactivación de Genes , Humanos , Inmunoprecipitación , Proteínas del Tejido Nervioso , Fosforilación , ARN Interferente Pequeño , Transfección , Proteína 28 que Contiene Motivos Tripartito
8.
Nucleic Acids Res ; 42(21): 13150-60, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25361978

RESUMEN

Human DBC1 (Deleted in Breast Cancer 1; KIAA1967; CCAR2) is a protein implicated in the regulation of apoptosis, transcription and histone modifications. Upon DNA damage, DBC1 is phosphorylated by ATM/ATR on Thr454 and this modification increases its inhibitory interaction with SIRT1, leading to p53 acetylation and p53-dependent apoptosis. Here, we report that the inhibition of SIRT1 by DBC1 in the DNA damage response (DDR) also depends on Chk2, the transducer kinase that is activated by ATM upon DNA lesions and contributes to the spreading of DNA damage signal. Indeed we found that inactivation of Chk2 reduces DBC1-SIRT1 binding, thus preventing p53 acetylation and DBC1-induced apoptosis. These events are mediated by Chk2 phosphorylation of the 11S proteasome activator REGγ on Ser247, which increases REGγ-DBC1 interaction and SIRT1 inhibition. Overall our results clarify the mechanisms underlying the DBC1-dependent SIRT1 inhibition and link, for the first time, Chk2 and REGγ to the ATM-DBC1-SIRT1 axis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Autoantígenos/metabolismo , Quinasa de Punto de Control 2/metabolismo , Daño del ADN , Complejo de la Endopetidasa Proteasomal/metabolismo , Sirtuina 1/metabolismo , Línea Celular Tumoral , Humanos , Sirtuina 1/antagonistas & inhibidores
9.
J Mol Cell Biol ; 6(6): 442-57, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25404613

RESUMEN

The serine/threonine kinase CHK2 is a key component of the DNA damage response. In human cells, following genotoxic stress, CHK2 is activated and phosphorylates >20 proteins to induce the appropriate cellular response, which, depending on the extent of damage, the cell type, and other factors, could be cell cycle checkpoint activation, induction of apoptosis or senescence, DNA repair, or tolerance of the damage. Recently, CHK2 has also been found to have cellular functions independent of the presence of nuclear DNA lesions. In particular, CHK2 participates in several molecular processes involved in DNA structure modification and cell cycle progression. In this review, we discuss the activity of CHK2 in response to DNA damage and in the maintenance of the biological functions in unstressed cells. These activities are also considered in relation to a possible role of CHK2 in tumorigenesis and, as a consequence, as a target of cancer therapy.


Asunto(s)
Puntos de Control del Ciclo Celular , Transformación Celular Neoplásica/metabolismo , Quinasa de Punto de Control 2/metabolismo , Daño del ADN , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Quinasa de Punto de Control 2/genética , Humanos , Fosforilación/genética
10.
J Mol Cell Biol ; 4(5): 294-303, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22735644

RESUMEN

Human DBC1 (deleted in breast cancer-1; KIAA1967) is a nuclear protein that, in response to DNA damage, competitively inhibits the NAD(+)-dependent deacetylase SIRT1, a regulator of p53 apoptotic functions in response to genotoxic stress. DBC1 depletion in human cells increases SIRT1 activity, resulting in the deacetylation of p53 and protection from apoptosis. However, the mechanisms regulating this process have not yet been determined. Here, we report that, in human cell lines, DNA damage triggered the phosphorylation of DBC1 on Thr454 by ATM (ataxia telangiectasia-mutated) and ATR (ataxia telangiectasia and Rad3-related) kinases. Phosphorylated DBC1 bound to and inhibited SIRT1, resulting in the dissociation of the SIRT1-p53 complex and stimulating p53 acetylation and p53-dependent cell death. Indeed, DBC1-mediated genotoxicity, which was shown in knockdown experiments to be dependent on SIRT1 and p53 expression, was defective in cells expressing the phospho-mutant DBC1(T454A). This study describes the first post-translational modification of DBC1 and provides new mechanistic insight linking ATM/ATR to the DBC1-SIRT1-p53 apoptotic axis triggered by DNA damage.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Sirtuina 1/antagonistas & inhibidores , Acetilación , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Daño del ADN/fisiología , Células HEK293 , Humanos , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Sirtuina 1/genética , Sirtuina 1/metabolismo
11.
DNA Repair (Amst) ; 9(11): 1200-8, 2010 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-20947454

RESUMEN

We have previously shown that whereas T-cells from normal individuals undergo accumulation of p53 and apoptosis when treated with the genotoxic agent Actinomycin D (ActD), those from Ataxia Telangiectasia (AT) and Nijmegen Breakage Syndrome (NBS) patients resist ActD-induced apoptosis [1]. We have now found similar resistance by the p53-null Jurkat T-cell line and by siRNA p53-knockdown normal T-cells. This evidence that ActD initiates a p53-dependent apoptotic responce prompted us to look for defective p53 accumulation by AT and NBS T-cells. Surprisingly the total p53 level was only slightly reduced compared to normal T cells but its intracellular localization was highly defective: p53 was poorly accumulated in the cytosol and nearly undetectable in mitochondria. In accordance with the dependence of ActD-induced apoptosis on a mitochondrial p53 function, in control T-cells specific inhibition of mitochondrial p53 translocation with µ pifithrin reduced apoptosis by 86%, whereas treatment with α pifithrin, which blocks p53-mediated transcription, had no effect. We also showed that nuclear export is not required for mitochondrial p53 translocation. Observation of an altered p53 ubiquitination pattern and Mdm2 accumulation in ActD-treated AT and NBS T-cells provided a mechanistic link to their defective extranuclear p53 localization. Our results disclose an undescribed defect in mitochondrial p53 accumulation in AT and NBS T-cells that makes them resistant to apoptosis following unrepairable DNA damage.


Asunto(s)
Apoptosis , Ataxia Telangiectasia/patología , Daño del ADN , Mitocondrias/metabolismo , Síndrome de Nijmegen/patología , Linfocitos T/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Dactinomicina/toxicidad , Humanos , Células Jurkat , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Síndrome de Nijmegen/genética , Síndrome de Nijmegen/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Ubiquitinación/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
12.
Cell Cycle ; 8(15): 2399-407, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19556897

RESUMEN

REGgamma (also called PA28gamma or PSME3) is a proteasome activator involved in the degradation of several proteins that regulate cell cycle and transcription. Recently, we demonstrated that this protein has a role also in the maintenance of chromosomal stability and in the response to spindle damaging agents. Here we report for the first time that REGgamma interacts with the promyelocytic leukemia protein (PML), accumulates in PML nuclear bodies (PML-NBs), but it does not play any role in normal or arsenic-induced PML degradation. However, REGgamma seems to regulate PML-NBs number, since its deficiency causes an increase in PML-NBs, which can be overcome by increased levels of SUMO1, and its overexpression has the opposite effect. We additionally found that REGgamma interacts with the DNA damage checkpoint kinase Chk2, whose presence is necessary for the increase of PML-NBs induced by REGgamma deficiency, and that REGgamma depletion resulted in a partial restoration of PML-NBs in APL derived cells. Altogether, these results underline a new role for REGgamma in the control and regulation of PML subnuclear structures.


Asunto(s)
Autoantígenos/metabolismo , Núcleo Celular/enzimología , Cuerpos de Inclusión Intranucleares/metabolismo , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Línea Celular Tumoral , Núcleo Celular/ultraestructura , Quinasa de Punto de Control 2 , Fibroblastos/enzimología , Humanos , Cuerpos de Inclusión Intranucleares/ultraestructura , Proteína de la Leucemia Promielocítica , Proteína SUMO-1/metabolismo
13.
Curr Biol ; 19(10): 874-9, 2009 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-19375317

RESUMEN

The shelterin complex [1] shapes and protects telomeric DNA from being processed as double strand breaks (DSBs) [2, 3]. Here we show that in human undamaged cells, a fraction of the kinase Chk2, a downstream target of ATM and mediator of checkpoint responses and senescence [4, 5], physically interacts with the shelterin subunit TRF2 and colocalizes with this complex at chromosome ends. This interaction, enhanced by TRF2 binding to telomeric DNA, inhibits the activation and senescence-induced function of Chk2 by a mechanism in which TRF2 binding to the N terminus of Chk2 surrounding Thr68 hinders the phosphorylation of this priming site. In response to radiation-induced DSBs, but not chromatin-remodelling agents, the telomeric Chk2-TRF2 binding dissociates in a Chk2 activity-dependent manner. Moreover, active Chk2 phosphorylates TRF2 and decreases its binding to telomeric DNA repeats, corroborating the evidences on the specific TRF2 relocalization in presence of DSBs [6]. Altogether, the capacity of TRF2 to locally repress Chk2 provides an additional level of control by which shelterin restrains the DNA damage response from an unwanted activation [6, 7] and may explain why TRF2 overexpression acts as a telomerase-independent oncogenic stimulus [8].


Asunto(s)
Daño del ADN , Proteínas Serina-Treonina Quinasas/metabolismo , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Senescencia Celular , Quinasa de Punto de Control 2 , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Telomerasa/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
14.
Cell Cycle ; 7(4): 504-12, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18235248

RESUMEN

REGgamma is a member of the 11S regulatory particle that activates the 20S proteasome. Studies in REGgamma deficient mice indicated an additional role for this protein in cell cycle regulation and proliferation control. In this paper we demonstrate that REGgamma protein is equally expressed throughout the cell cycle, but undergoes a distinctive subcellular localization at mitosis. Thus, while in interphase cells REGgamma is nuclear, in telophase cells it localizes on chromosomes, suggesting a role in mitotic progression. Furthermore, we found that REGgamma overexpression weakens the mitotic arrest induced by spindle damage, allowing premature exit from mitosis, whereas REGgamma depletion has the opposite effect, thus reflecting a new REGgamma function, unrelated to its role as proteasome activator. Additionally, we found that primary cells from REGgamma-/- mice and human fibroblasts with depleted expression of REGgamma or overexpressing a dominant negative mutant unable to activate the 20S proteasome, demonstrated a marked aneuploidy (chromosomal gains and losses), supernumerary centrosomes and multipolar spindles. These findings thus underscore a previously uncharacterized function of REGgamma in centrosome and chromosomal stability maintenance.


Asunto(s)
Autoantígenos/metabolismo , Centrosoma/metabolismo , Inestabilidad Cromosómica/fisiología , Mitosis/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Autoantígenos/genética , Autoantígenos/fisiología , Western Blotting , Inestabilidad Cromosómica/genética , Cartilla de ADN/genética , Fibroblastos , Citometría de Flujo , Humanos , Ratones , Ratones Noqueados , Microscopía Fluorescente , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/fisiología
15.
Cancer Cell ; 10(6): 473-86, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17157788

RESUMEN

Che-1 is a RNA polymerase II-binding protein involved in the transcription of E2F target genes and induction of cell proliferation. Here we show that Che-1 contributes to DNA damage response and that its depletion sensitizes cells to anticancer agents. The checkpoint kinases ATM/ATR and Chk2 interact with Che-1 and promote its phosphorylation and accumulation in response to DNA damage. These Che-1 modifications induce a specific recruitment of Che-1 on the TP53 and p21 promoters. Interestingly, it has a profound effect on the basal expression of p53, which is preserved following DNA damage. Notably, Che-1 contributes to the maintenance of the G2/M checkpoint induced by DNA damage. These findings identify a mechanism by which checkpoint kinases regulate responses to DNA damage.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/fisiología , Genes p53 , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/fisiología , Animales , Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada , División Celular , Quinasa de Punto de Control 2 , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Daño del ADN , Fase G2 , Humanos , Ratones , Células 3T3 NIH , Fosforilación , Regiones Promotoras Genéticas , Transcripción Genética
16.
Mol Cell Biol ; 26(21): 7832-45, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16940182

RESUMEN

Chk2 kinase is activated by DNA damage to regulate cell cycle arrest, DNA repair, and apoptosis. Phosphorylation of Chk2 in vivo by ataxia telangiectasia-mutated (ATM) on threonine 68 (T68) initiates a phosphorylation cascade that promotes the full activity of Chk2. We identified three serine residues (S19, S33, and S35) on Chk2 that became phosphorylated in vivo rapidly and exclusively in response to ionizing radiation (IR)-induced DNA double-strand breaks in an ATM- and Nbs1-dependent but ataxia telangiectasia- and Rad3-related-independent manner. Phosphorylation of these residues, restricted to the G(1) phase of the cell cycle, was induced by a higher dose of IR (>1 Gy) than that required for phosphorylation of T68 (0.25 Gy) and declined by 45 to 90 min, concomitant with a rise in Chk2 autophosphorylation. Compared to the wild-type form, Chk2 with alanine substitutions at S19, S33, and S35 (Chk2(S3A)) showed impaired dimerization, defective auto- and trans-phosphorylation activities, and reduced ability to promote degradation of Hdmx, a phosphorylation target of Chk2 and regulator of p53 activity. Besides, Chk2(S3A) failed to inhibit cell growth and, in response to IR, to arrest G(1)/S progression. These findings underscore the critical roles of S19, S33, and S35 and argue that these phosphoresidues may serve to fine-tune the ATM-dependent response of Chk2 to increasing amounts of DNA damage.


Asunto(s)
Ciclo Celular/fisiología , Daño del ADN , Proteínas Serina-Treonina Quinasas , Serina/metabolismo , 4-Nitroquinolina-1-Óxido/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Ciclo Celular/efectos de la radiación , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Quinasa de Punto de Control 2 , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/metabolismo , Humanos , Hidroxiurea/metabolismo , Complejos Multiproteicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Quinolonas/metabolismo , Interferencia de ARN , Radiación Ionizante , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
17.
Int J Cancer ; 119(7): 1599-606, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16671099

RESUMEN

N-(4-Hydroxyphenyl)retinamide (4-HPR) is a nonclassical retinoid with cancer preventive effects in vivo and antiproliferative and apoptotic activities in vitro. Examining the transcriptional profile of human breast cancer cell lines, MCF7 and T47D, treated with 4-HPR, we identified the lipocalin member LCN2 (NGAL or 24p3) as a gene, markedly induced by the retinoid. Because of its presumed function in apoptosis, LCN2 was examined more thoroughly in response to 4-HPR. Like mRNA, the expression of LCN2 protein in MCF7 and T47D cells was highly induced in a time-dependent manner by 4-HPR, but not by its inactive metabolite 4-MPR and, to some extent, this event was linked to the free radicals normally generated by 4-HPR. All-trans retinoic acid also induced LCN2 protein, particularly in T47D cells. Ectopic LCN2 compromised cell viability, and the few MCF7 clones that survived LCN2 overexpression were less sensitive than do mock cells to 4HPR, indicating that selective pressure for survival to LCN2 confers cross-resistance to 4-HPR. Significantly, ablation of LCN2 induction by siRNA did not modify the response to 4-HPR, implying that LCN2 is not critical for apoptosis by 4-HPR. Our results indicate that 4-HPR markedly induces LCN2 expression, but this event may not represent an apoptotic response.


Asunto(s)
Proteínas de Fase Aguda/genética , Antineoplásicos/farmacología , Fenretinida/farmacología , Neoplasias/genética , Proteínas Proto-Oncogénicas/genética , Proteínas de Fase Aguda/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citosol/efectos de los fármacos , Citosol/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Radicales Libres/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lipocalina 2 , Lipocalinas , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Proto-Oncogénicas/metabolismo , ARN Interferente Pequeño/genética , Regulación hacia Arriba
18.
J Biol Chem ; 278(43): 42346-51, 2003 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-12909615

RESUMEN

Chk2 is a nuclear protein kinase involved in the DNA damage-induced ataxia telangiectasia mutated-dependent checkpoint arrest at multiple cell cycle phases. Searching for Chk2-binding proteins by a yeast two-hybrid system, we identified a strong interaction with karyopherin-alpha2 (KPNA-2), a gene product involved in active nuclear import of proteins bearing a nuclear localization signal (NLS). This finding was confirmed by glutathione S-transferase pull-down and co-immunoprecipitation assays. Of the three predicted Chk2 NLSs, located at amino acids 179-182 (NLS-1), 240-256 (NLS-2), and 515-522 (NLS-3), only the latter mediated the interaction with KPNA-2 in the yeast two-hybrid system, and in particular with its C terminus. Unlike mutations in NLS-1 or NLS-2, which left the nuclear localization of Chk2 unaffected, mutations in NLS-3 caused a cytoplasmic relocalization, indicating that the NLS-3 motif acts indeed as NLS for Chk2 in vivo. Finally, co-transfection experiments with green fluorescent protein (GFP)-Chk2 and wild type or mutant KPNA-2 confirmed the role of KPNA-2 in nuclear import of Chk2.


Asunto(s)
Transporte Activo de Núcleo Celular , Proteínas Serina-Treonina Quinasas/metabolismo , alfa Carioferinas/metabolismo , Secuencia de Aminoácidos , Quinasa de Punto de Control 2 , Humanos , Mutación , Señales de Localización Nuclear/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Transfección , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos
19.
Blood ; 101(9): 3622-7, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12511424

RESUMEN

The possible involvement of germline mutation of the ataxia telangiectasia mutated (ATM) gene in childhood acute leukemia with mixed lineage leukemia (MLL) gene rearrangement (MLL(+)) was investigated. Of the 7 patients studied, 1 showed a germline missense ATM mutation (8921C>T; Pro2974Leu), located in the phosphatidylinositol-3 (PI-3) kinase domain. In reconstitution assays, the ATM mutant 8921T could only partially rescue the radiosensitive phenotype of AT fibroblasts, and in an in vitro kinase assay, it showed a defective phosphorylation of p53-Ser15. Furthermore, the introduction of 8921T in U2OS cells, characterized by a normal ATM/p53 signal transduction, caused a significant reduction of in vivo p53-Ser15 phosphorylation, suggesting a dominant-negative effect of the mutant ATM over the wild-type protein. Our finding in this patient suggests that altered function of ATM plays some pathogenic roles in the development of MLL(+) leukemia.


Asunto(s)
Proteínas de Unión al ADN/genética , Mutación Missense , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Serina-Treonina Quinasas/genética , Proto-Oncogenes , Factores de Transcripción , Ataxia Telangiectasia/patología , Proteínas de la Ataxia Telangiectasia Mutada , Neoplasias Óseas/patología , Proteínas de Ciclo Celular , Línea Celular/efectos de la radiación , Transformación Celular Neoplásica/genética , Cromosomas Humanos Par 11/genética , Análisis Mutacional de ADN , ADN de Neoplasias/genética , Femenino , Genes Dominantes , Prueba de Complementación Genética , Mutación de Línea Germinal , N-Metiltransferasa de Histona-Lisina , Humanos , Lactante , Leucemia Monocítica Aguda/genética , Masculino , Proteína de la Leucemia Mieloide-Linfoide , Osteosarcoma/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Tolerancia a Radiación/genética , Transducción de Señal , Translocación Genética , Células Tumorales Cultivadas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...