Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell ; 134(2): 256-67, 2008 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-18662541

RESUMEN

In response to DNA damage in G2, mammalian cells must avoid entry into mitosis and instead initiate DNA repair. Here, we show that, in response to genotoxic stress in G2, the phosphatase Cdc14B translocates from the nucleolus to the nucleoplasm and induces the activation of the ubiquitin ligase APC/C(Cdh1), with the consequent degradation of Plk1, a prominent mitotic kinase. This process induces the stabilization of Claspin, an activator of the DNA-damage checkpoint, and Wee1, an inhibitor of cell-cycle progression, and allows an efficient G2 checkpoint. As a by-product of APC/C(Cdh1) reactivation in DNA-damaged G2 cells, Claspin, which we show to be an APC/C(Cdh1) substrate in G1, is targeted for degradation. However, this process is counteracted by the deubiquitylating enzyme Usp28 to permit Claspin-mediated activation of Chk1 in response to DNA damage. These findings define a novel pathway that is crucial for the G2 DNA-damage-response checkpoint.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Reparación del ADN , Fosfatasas de Especificidad Dual/metabolismo , Fase G2 , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ciclosoma-Complejo Promotor de la Anafase , Línea Celular Tumoral , Núcleo Celular/metabolismo , Daño del ADN , Humanos , Mitosis , Neoplasias/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Quinasa Tipo Polo 1
2.
Genes Dev ; 28(2): 153-66, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24449270

RESUMEN

The progressive bone marrow failure syndrome dyskeratosis congenita (DC) is often caused by mutations in telomerase or the factors involved in telomerase biogenesis and trafficking. However, a subset of DC patients is heterozygous for mutations in the shelterin component TIN2. To determine how the TIN2-DC mutations affect telomere function, we generated mice with the equivalent of the TIN2 K280E DC allele (TIN2(DC)) by gene targeting. Whereas homozygous TIN2(DC/DC) mice were not viable, first-generation TIN2(+/DC) mice were healthy and fertile. In the second and third generations, the TIN2(+/DC) mice developed mild pancytopenia, consistent with hematopoietic dysfunction in DC, as well as diminished fecundity. Bone marrow telomeres of TIN2(+/DC) mice shortened over the generations, and immortalized TIN2(+/DC) mouse embryonic fibroblasts (MEFs) showed telomere shortening with proliferation. Unexpectedly, telomere shortening was accelerated in TIN2(+/DC) mTR(-/-) mice and MEFs compared with TIN2(+/+) mTR(-/-) controls, establishing that the TIN2(DC) telomere maintenance defect was not solely due to diminished telomerase action. The TIN2(DC) allele induced mild ATR kinase signaling at telomeres and a fragile telomere phenotype, suggestive of telomere replication problems. These data suggest that this TIN2-DC mutation could induce telomeric dysfunction phenotypes in telomerase-negative somatic cells and tissues that further exacerbate the telomere maintenance problems in telomerase-positive stem cell compartments.


Asunto(s)
Acortamiento del Telómero/genética , Proteínas de Unión a Telómeros/genética , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Disqueratosis Congénita/genética , Fertilidad/genética , Técnicas de Sustitución del Gen , Células HeLa , Humanos , Ratones , Mutación , Pancitopenia/genética , Transducción de Señal , Telomerasa/metabolismo , Telómero/patología
4.
Proc Natl Acad Sci U S A ; 114(9): E1668-E1677, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28193858

RESUMEN

Studying the phenomenon of cellular senescence has been hindered by the lack of senescence-specific markers. As such, detection of proteins informally associated with senescence accompanies the use of senescence-associated ß-galactosidase as a collection of semiselective markers to monitor the presence of senescent cells. To identify novel biomarkers of senescence, we immunized BALB/c mice with senescent mouse lung fibroblasts and screened for antibodies that recognized senescence-associated cell-surface antigens by FACS analysis and a newly developed cell-based ELISA. The majority of antibodies that we isolated, cloned, and sequenced belonged to the IgM isotype of the innate immune system. In-depth characterization of one of these monoclonal, polyreactive natural antibodies, the IgM clone 9H4, revealed its ability to recognize the intermediate filament vimentin. By using 9H4, we observed that senescent primary human fibroblasts express vimentin on their cell surface, and MS analysis revealed a posttranslational modification on cysteine 328 (C328) by the oxidative adduct malondialdehyde (MDA). Moreover, elevated levels of secreted MDA-modified vimentin were detected in the plasma of aged senescence-accelerated mouse prone 8 mice, which are known to have deregulated reactive oxygen species metabolism and accelerated aging. Based on these findings, we hypothesize that humoral innate immunity may recognize senescent cells by the presence of membrane-bound MDA-vimentin, presumably as part of a senescence eradication mechanism that may become impaired with age and result in senescent cell accumulation.


Asunto(s)
Anticuerpos/metabolismo , Membrana Celular/metabolismo , Senescencia Celular/fisiología , Vimentina/metabolismo , Animales , Biomarcadores/metabolismo , Células Cultivadas , Femenino , Fibroblastos/metabolismo , Inmunidad Humoral/fisiología , Inmunidad Innata/fisiología , Inmunoglobulina M/metabolismo , Filamentos Intermedios/metabolismo , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , beta-Galactosidasa/metabolismo
5.
Mol Cell ; 44(4): 647-59, 2011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-22099311

RESUMEN

To prevent ATR activation, telomeres deploy the single-stranded DNA binding activity of TPP1/POT1a. POT1a blocks the binding of RPA to telomeres, suggesting that ATR is repressed through RPA exclusion. However, comparison of the DNA binding affinities and abundance of TPP1/POT1a and RPA indicates that TPP1/POT1a by itself is unlikely to exclude RPA. We therefore analyzed the central shelterin protein TIN2, which links TPP1/POT1a (and POT1b) to TRF1 and TRF2 on the double-stranded telomeric DNA. Upon TIN2 deletion, telomeres lost TPP1/POT1a, accumulated RPA, elicited an ATR signal, and showed all other phenotypes of POT1a/b deletion. TIN2 also affected the TRF2-dependent repression of ATM kinase signaling but not to TRF2-mediated inhibition of telomere fusions. Thus, while TIN2 has a minor contribution to the repression of ATM by TRF2, its major role is to stabilize TPP1/POT1a on the ss telomeric DNA, thereby allowing effective exclusion of RPA and repression of ATR signaling.


Asunto(s)
Reparación del ADN , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/metabolismo , Expresión Génica , Transducción de Señal/genética , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Daño del ADN , ADN de Cadena Simple/química , ADN de Cadena Simple/genética , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Ratones , Ratones Noqueados , Unión Proteica/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Complejo Shelterina , Proteínas de Unión a Telómeros/genética , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo
6.
J Biol Chem ; 289(35): 24180-7, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25056954

RESUMEN

The single-stranded DNA binding proteins in mouse shelterin, POT1a and POT1b, accumulate at telomeres as heterodimers with TPP1, which binds TIN2 and thus links the TPP1/POT1 dimers with TRF1 and TRF2/Rap1. When TPP1 is tethered to TIN2/TRF1/TRF2, POT1a is thought to block replication protein A binding to the single-stranded telomeric DNA and prevent ataxia telangiectasia and Rad3-related kinase activation. Similarly, TPP1/POT1b tethered to TIN2 can control the formation of the correct single-stranded telomeric overhang. Consistent with this view, the telomeric phenotypes following deletion of POT1a,b or TPP1 are phenocopied in TIN2-deficient cells. However, the loading of TRF1 and TRF2/Rap1 is additionally compromised in TIN2 KO cells, leading to added phenotypes. Therefore, it could not be excluded that, in addition to TIN2, other components of shelterin contribute to the recruitment of TPP1/POT1a,b as suggested by previous reports. To test whether TIN2 is the sole link between TPP1/POT1a,b and telomeres, we defined the TPP1 interaction domain of TIN2 and generated a TIN2 allele that was unable to interact with TPP1 but retained its interaction with TRF1 and TRF2. We demonstrated that cells expressing TIN2ΔTPP1 instead of wild-type TIN2 phenocopy the POT1a,b knockout setting without showing additional phenotypes. Therefore, these results are consistent with TIN2 being the only mechanism by which TPP1/POT1 heterodimers bind to shelterin and function in telomere protection.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Serina Proteasas/metabolismo , Telomerasa , Homeostasis del Telómero , Proteínas de Unión a Telómeros/metabolismo , Alelos , Secuencia de Aminoácidos , Animales , Línea Celular Transformada , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Serina Proteasas/química , Complejo Shelterina , Telomerasa/metabolismo , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/genética
7.
Nature ; 452(7185): 365-9, 2008 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-18354482

RESUMEN

REST/NRSF (repressor-element-1-silencing transcription factor/neuron-restrictive silencing factor) negatively regulates the transcription of genes containing RE1 sites. REST is expressed in non-neuronal cells and stem/progenitor neuronal cells, in which it inhibits the expression of neuron-specific genes. Overexpression of REST is frequently found in human medulloblastomas and neuroblastomas, in which it is thought to maintain the stem character of tumour cells. Neural stem cells forced to express REST and c-Myc fail to differentiate and give rise to tumours in the mouse cerebellum. Expression of a splice variant of REST that lacks the carboxy terminus has been associated with neuronal tumours and small-cell lung carcinomas, and a frameshift mutant (REST-FS), which is also truncated at the C terminus, has oncogenic properties. Here we show, by using an unbiased screen, that REST is an interactor of the F-box protein beta-TrCP. REST is degraded by means of the ubiquitin ligase SCF(beta-TrCP) during the G2 phase of the cell cycle to allow transcriptional derepression of Mad2, an essential component of the spindle assembly checkpoint. The expression in cultured cells of a stable REST mutant, which is unable to bind beta-TrCP, inhibited Mad2 expression and resulted in a phenotype analogous to that observed in Mad2(+/-) cells. In particular, we observed defects that were consistent with faulty activation of the spindle checkpoint, such as shortened mitosis, premature sister-chromatid separation, chromosome bridges and mis-segregation in anaphase, tetraploidy, and faster mitotic slippage in the presence of a spindle inhibitor. An indistinguishable phenotype was observed by expressing the oncogenic REST-FS mutant, which does not bind beta-TrCP. Thus, SCF(beta-TrCP)-dependent degradation of REST during G2 permits the optimal activation of the spindle checkpoint, and consequently it is required for the fidelity of mitosis. The high levels of REST or its truncated variants found in certain human tumours may contribute to cellular transformation by promoting genomic instability.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Inestabilidad Cromosómica , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Proteínas con Repetición de beta-Transducina/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Línea Celular , Fase G2 , Regulación de la Expresión Génica , Inestabilidad Genómica , Humanos , Proteínas Mad2 , Mitosis , Unión Proteica , Proteínas Represoras/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Huso Acromático/fisiología , Factores de Transcripción/genética , Proteínas con Repetición de beta-Transducina/deficiencia , Proteínas con Repetición de beta-Transducina/genética
8.
Nature ; 450(7167): 309-13, 2007 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-17994099

RESUMEN

JHDM1B is an evolutionarily conserved and ubiquitously expressed member of the JHDM (JmjC-domain-containing histone demethylase) family. Because it contains an F-box motif, this protein is also known as FBXL10 (ref. 4). With the use of a genome-wide RNAi screen, the JHDM1B worm orthologue (T26A5.5) was identified as a gene that regulates growth. In the mouse, four independent screens have identified JHDM1B as a putative tumour suppressor by retroviral insertion analysis. Here we identify human JHDM1B as a nucleolar protein and show that JHDM1B preferentially binds the transcribed region of ribosomal DNA to repress the transcription of ribosomal RNA genes. We also show that repression of ribosomal RNA genes by JHDM1B is dependent on its JmjC domain, which is necessary for the specific demethylation of trimethylated lysine 4 on histone H3 in the nucleolus. In agreement with the notion that ribosomal RNA synthesis and cell growth are coupled processes, we show a JmjC-domain-dependent negative effect of JHDM1B on cell size and cell proliferation. Because aberrant ribosome biogenesis and the disruption of epigenetic control mechanisms contribute to cellular transformation, these results, together with the low levels of JHDM1B expression found in aggressive brain tumours, suggest a role for JHDM1B in cancer development.


Asunto(s)
Nucléolo Celular/metabolismo , ADN Ribosómico/genética , Proteínas F-Box/metabolismo , Genes de ARNr/genética , Proteínas Nucleares/metabolismo , Oxidorreductasas N-Desmetilantes/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Proliferación Celular , ADN Ribosómico/metabolismo , Regulación hacia Abajo , Proteínas F-Box/química , Células HeLa , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji , Metilación , Proteínas Nucleares/química , Oxidorreductasas N-Desmetilantes/química , Estructura Terciaria de Proteína , Interferencia de ARN , Proteínas Represoras/química
9.
J Cell Biol ; 173(2): 219-30, 2006 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-16636144

RESUMEN

Drosophila melanogaster embryogenesis begins with 13 nuclear division cycles within a syncytium. This produces >6,000 nuclei that, during the next division cycle, become encased in plasma membrane in the process known as cellularization. In this study, we investigate how the secretory membrane system becomes equally apportioned among the thousands of syncytial nuclei in preparation for cellularization. Upon nuclear arrival at the cortex, the endoplasmic reticulum (ER) and Golgi were found to segregate among nuclei, with each nucleus becoming surrounded by a single ER/Golgi membrane system separate from adjacent ones. The nuclear-associated units of ER and Golgi across the syncytial blastoderm produced secretory products that were delivered to the plasma membrane in a spatially restricted fashion across the embryo. This occurred in the absence of plasma membrane boundaries between nuclei and was dependent on centrosome-derived microtubules. The emergence of secretory membranes that compartmentalized around individual nuclei in the syncytial blastoderm is likely to ensure that secretory organelles are equivalently partitioned among nuclei at cellularization and could play an important role in the establishment of localized gene and protein expression patterns within the early embryo.


Asunto(s)
Blastodermo/ultraestructura , Membrana Celular/ultraestructura , Núcleo Celular/ultraestructura , Drosophila melanogaster/embriología , Células Gigantes/ultraestructura , Vesículas Secretoras/ultraestructura , Animales , Blastodermo/química , Compartimento Celular , Línea Celular , Membrana Celular/química , Núcleo Celular/química , Embrión no Mamífero/fisiología , Retículo Endoplásmico/química , Retículo Endoplásmico/ultraestructura , Células Gigantes/química , Aparato de Golgi/química , Aparato de Golgi/ultraestructura , Microscopía Confocal , Microtúbulos/química , Modelos Biológicos , Vesículas Secretoras/química
10.
Aging Cell ; 19(10): e13219, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32856419

RESUMEN

Adipose tissue is recognized as a major source of systemic inflammation with age, driving age-related tissue dysfunction and pathogenesis. Macrophages (Mφ) are central to these changes yet adipose tissue Mφ (ATMs) from aged mice remain poorly characterized. To identify biomarkers underlying changes in aged adipose tissue, we performed an unbiased RNA-seq analysis of ATMs from young (8-week-old) and healthy aged (80-week-old) mice. One of the genes identified, V-set immunoglobulin-domain-containing 4 (VSIG4/CRIg), encodes a Mφ-associated complement receptor and B7 family-related immune checkpoint protein. Here, we demonstrate that Vsig4 expression is highly upregulated with age in perigonadal white adipose tissue (gWAT) in two mouse strains (inbred C57BL/6J and outbred NIH Swiss) independent of gender. The accumulation of VSIG4 was mainly attributed to a fourfold increase in the proportion of VSIG4+ ATMs (13%-52%). In a longitudinal study, VSIG4 expression in gWAT showed a strong correlation with age within a cohort of male and female mice and correlated strongly with physiological frailty index (PFI, a multi-parameter assessment of health) in male mice. Our results indicate that VSIG4 is a novel biomarker of aged murine ATMs. VSIG4 expression was also found to be elevated in other aging tissues (e.g., thymus) and was strongly induced in tumor-adjacent stroma in cases of spontaneous and xenograft lung cancer models. VSIG4 expression was recently associated with cancer and several inflammatory diseases with diagnostic and prognostic potential in both mice and humans. Further investigation is required to determine whether VSIG4-positive Mφ contribute to immunosenescence and/or systemic age-related deficits.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Receptores de Complemento/metabolismo , Envejecimiento/metabolismo , Animales , Biomarcadores/metabolismo , Femenino , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
11.
Mol Biol Cell ; 15(10): 4512-21, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15292454

RESUMEN

Drosophila ovarian cysts arise through a series of four synchronous incomplete mitotic divisions. After each round of mitosis, a membranous organelle, the fusome, grows along the cleavage furrow and the remnants of the mitotic spindle to connect all cystocytes in a cyst. The fusome is essential for the pattern and synchrony of the mitotic cyst divisions as well as oocyte differentiation. Using live cell imaging, green fluorescent protein-tagged proteins, and photobleaching techniques, we demonstrate that fusomal endomembranes are part of a single continuous endoplasmic reticulum (ER) that is shared by all cystocytes in dividing ovarian cysts. Membrane and lumenal proteins of the common ER freely and rapidly diffuse between cystocytes. The fusomal ER mediates intercellular ER connectivity by linking the cytoplasmic ER membranes of all cystocytes within a cyst. Before entry into meiosis and onset of oocyte differentiation (between region 1 and region 2A), ER continuity between cystocytes is lost. Furthermore, analyses of hts and Dhc64c mutants indicate that intercellular ER continuity within dividing ovarian cysts requires the fusome cytoskeletal component and suggest a possible role for the common ER in synchronizing mitotic cyst divisions.


Asunto(s)
Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/fisiología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Membranas Intracelulares/metabolismo , Mitosis/fisiología , Animales , Biomarcadores , Diferenciación Celular , Femenino , Membranas Intracelulares/ultraestructura , Ovario/anatomía & histología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
12.
Cell Cycle ; 16(16): 1526-1533, 2017 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-28650766

RESUMEN

Age-related health decline has been attributed to the accumulation of senescent cells recognized in vivo by p16(Ink4a) expression. The pharmacological elimination of p16(Ink4a)-positive cells from the tissues of mice was shown to extend a healthy lifespan. Here, we describe a population of mesenchymal cells isolated from mice that are highly p16(INK4a)-positive are proficient in proliferation but lack other properties of cellular senescence. These data, along with earlier reports on p16(Ink4a)-positive macrophages, indicate that p16(Ink4a)-positive and senescent cell populations only partially intersect, therefore, extending the list of potential cellular targets for anti- aging therapies.


Asunto(s)
Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Animales , Proliferación Celular , Humanos , Antígenos Comunes de Leucocito/metabolismo , Ratones Endogámicos C57BL
13.
Exp Biol Med (Maywood) ; 240(8): 1065-70, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25956682

RESUMEN

The capability to modify the genome precisely and efficiently offers an extremely useful tool for biomedical research. Recent developments in genome editing technologies such as transcription activator-like effector nuclease and the clustered regularly interspaced short palindromic repeats system have made genome modification available for a number of organisms with relative ease. Here, we introduce these genome editing techniques, compare and contrast each technical approach and discuss their potential to study the underlying mechanisms of human disease using patient-derived induced pluripotent stem cells.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Marcación de Gen/métodos , Genoma Humano , Células Madre Pluripotentes Inducidas , Animales , Humanos
14.
Mol Cell Biol ; 34(7): 1349-62, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24469404

RESUMEN

The shelterin protein TIN2 is required for the telomeric accumulation of TPP1/POT1 heterodimers and for the protection of telomeres by the POT1 proteins (POT1a and POT1b in the mouse). TIN2 also binds to TRF1 and TRF2, improving the telomeric localization of TRF2 and its function. Here, we ask whether TIN2 needs to interact with both TRF1 and TRF2 to mediate the telomere protection afforded by TRF2 and POT1a/b. Using a TIN2 allele deficient in TRF1 binding (TIN2-L247E), we demonstrate that TRF1 is required for optimal recruitment of TIN2 to telomeres and document phenotypes associated with the TIN2-L247E allele that are explained by insufficient TIN2 loading onto telomeres. To bypass the requirement for TRF1-dependent recruitment, we fused TIN2-L247E to the TRF2-interacting (RCT) domain of Rap1. The RCT-TIN2-L247E fusion showed improved telomeric localization and was fully functional in terms of chromosome end protection by TRF2, TPP1/POT1a, and TPP1/POT1b. These data indicate that when sufficient TIN2 is loaded onto telomeres, its interaction with TRF1 is not required to mediate the function of TRF2 and the TPP1/POT1 heterodimers. We therefore conclude that shelterin can protect chromosome ends as a TRF2-tethered TIN2/TPP1/POT1 complex that lacks a physical connection to TRF1.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Animales , Células Cultivadas , Ratones , Mutagénesis Sitio-Dirigida , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Complejo Shelterina , Telómero/genética , Homeostasis del Telómero/genética , Homeostasis del Telómero/fisiología , Proteínas de Unión a Telómeros/deficiencia , Proteínas de Unión a Telómeros/genética , Proteína 1 de Unión a Repeticiones Teloméricas/química , Proteína 1 de Unión a Repeticiones Teloméricas/genética , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/química , Proteína 2 de Unión a Repeticiones Teloméricas/genética
15.
Nat Rev Cancer ; 8(6): 438-49, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18500245

RESUMEN

The maintenance and preservation of distinct phases during the cell cycle is a highly complex and coordinated process. It is regulated by phosphorylation--through the activity of cyclin-dependent kinases (CDKs)--and protein degradation, which occurs through ubiquitin ligases such as SCF (SKP1-CUL1-F-box protein) complexes and APC/C (anaphase-promoting complex/cyclosome). Here, we explore the functionality and biology of the F-box proteins, SKP2 (S-phase kinase-associated protein 2) and beta-TrCP (beta-transducin repeat-containing protein), which are emerging as important players in cancer biogenesis owing to the deregulated proteolysis of their substrates.


Asunto(s)
Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteínas Quinasas Asociadas a Fase-S/fisiología , Proteínas con Repetición de beta-Transducina/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Ciclo Celular/fisiología , Proteínas Co-Represoras , Proteínas Cullin/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Humanos , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Oncogenes , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Proteínas Ligasas SKP Cullina F-box/fisiología , Fosfatasas cdc25/metabolismo
16.
Cell Cycle ; 7(22): 3539-47, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19001877

RESUMEN

Heterochromatin plays an essential role in the preservation of epigenetic information, the transcriptional repression of repetitive DNA elements and inactive genes, and the proper segregation of chromosomes during mitosis. Here we identify KDM2A, a JmjC-domain containing histone demethylase, as a heterochromatin-associated and HP1-interacting protein that promotes HP1 localization to chromatin. We show that KDM2A is required to maintain the heterochromatic state, as determined using a candidate-based approach coupled to an in vivo epigenetic reporter system. Remarkably, a parallel and independent siRNA screen also detected a role for KDM2A in epigenetic silencing. Moreover, we demonstrate that KDM2A associates with centromeres and represses transcription of small non-coding RNAs that are encoded by the clusters of satellite repeats at the centromere. Dissecting the relationship between heterochromatin and centromeric RNA transcription is the basis of ongoing studies. We demonstrate that forced expression of these satellite RNA transcripts compromise the heterochromatic state and HP1 localization to chromatin. Finally, we show that KDM2A is required to sustain centromeric integrity and genomic stability, particularly during mitosis. Since the disruption of epigenetic control mechanisms contributes to cellular transformation, these results, together with the low levels of KDM2A found in prostate carcinomas, suggest a role for KDM2A in cancer development.


Asunto(s)
ADN Satélite/genética , Heterocromatina/genética , Oxidorreductasas N-Desmetilantes/fisiología , Transcripción Genética , Células 3T3 , Animales , Transformación Celular Neoplásica , Centrómero/genética , Ensamble y Desensamble de Cromatina/genética , Proteínas F-Box , Inestabilidad Genómica , Células HeLa , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji , Ratones , Transfección
17.
J Biol Chem ; 280(21): 20589-95, 2005 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-15788402

RESUMEN

Activation of NAD-dependent deacetylases, or Sirtuins, prolongs life span and mimics the effects of caloric restriction in yeast. The FoxO subfamily of forkhead transcription factors has been shown to mediate some of the effects of Sirtuins. Here we have shown that Sirtuin activation or hydrogen peroxide treatment overrides the phosphorylation-dependent nuclear exclusion of FoxO1 caused by growth factors and causes nuclear translocation of FoxO1 in hepatocytes. Kinetic measurements of nuclear fluorescence recovery after photobleaching show that FoxO1 is readily diffusible within the nucleus under normal conditions but becomes restricted within a nuclear subdomain following treatment with the prototypical Sirtuin agonist resveratrol or oxidative stress. Expression of FoxO1 target genes is accordingly increased, leading to activation of gluconeogenesis and increased glucose release from hepatocytes. Selective modulation of the FoxO/Sirtuin interaction represents a promising therapeutic modality for metabolic disorders.


Asunto(s)
Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Gluconeogénesis/genética , Sirtuinas/farmacología , Factores de Transcripción/metabolismo , Acetilación , Animales , Transporte Biológico/efectos de los fármacos , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead , Glucosa/metabolismo , Proteínas Fluorescentes Verdes/genética , Hepatocitos/metabolismo , Hepatocitos/ultraestructura , Peróxido de Hidrógeno/farmacología , Cinética , Ratones , Estrés Oxidativo , Fosforilación , Fotoblanqueo , Proteínas Recombinantes de Fusión , Resveratrol , Sirtuinas/antagonistas & inhibidores , Estilbenos/farmacología , Factores de Transcripción/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA