Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
BMC Bioinformatics ; 18(1): 15, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-28056778

RESUMEN

BACKGROUND: The cyclin-dependent kinase 2 (CDK2) together with its cyclin E and A partners is a central regulator of cell growth and division. Deregulation of CDK2 activity is associated with diseases such as cancer. The analysis of substrates identified S/T-P-X-R/K/H as the CDK2 consensus sequence. The crystal structure of cyclin A/CDK2 with a short model peptide supports this sequence and identifies key interactions. However, CDKs use additional determinants to recognize substrates, including the RXL motif that is read by the cyclin subunits. We were interested to determine whether additional amino acids beyond the minimal consensus sequence of the well-studied substrate and tumor suppressor p27KIP1 were relevant for catalysis. RESULTS: To address whether additional amino acids, close to the minimal consensus sequence, play a role in binding, we investigate the interaction of cyclin A/CDK2 with an in vivo cellular partner and CDK inhibitor p27KIP1. This protein is an intrinsically unfolded protein and, in particular, the C-terminal half of the protein has not been accessible to structural analysis. This part harbors the CDK2 phosphorylation site. We used bioinformatics tools, including MODELLER, iTASSER and HADDOCK, along with partial structural information to build a model of the C-terminal region of p27KIP1 with cyclin A/CDK2. This revealed novel interactions beyond the consensus sequence with a proline and a basic amino acid at the P + 1 and the P + 3 sites, respectively. We suggest that the lysine at P + 2 might regulate the reversible association of the second counter ion in the active site of CDK2. The arginine at P + 7 interacts with both cyclin A and CDK2 and is important for the catalytic turnover rate. CONCLUSION: Our modeling identifies additional amino acids in p27KIP1 beyond the consensus sequence that contribute to the efficiency of substrate phosphorylation.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/química , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/química , Secuencia de Aminoácidos , Animales , Biología Computacional , Ciclina A/química , Ciclina E/química , Humanos , Fosforilación , Conformación Proteica , Spodoptera
2.
Nucleic Acids Res ; 42(11): 6901-20, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24782528

RESUMEN

The appropriate expression of the roughly 30,000 human genes requires multiple layers of control. The oncoprotein MYC, a transcriptional regulator, contributes to many of the identified control mechanisms, including the regulation of chromatin, RNA polymerases, and RNA processing. Moreover, MYC recruits core histone-modifying enzymes to DNA. We identified an additional transcriptional cofactor complex that interacts with MYC and that is important for gene transcription. We found that the trithorax protein ASH2L and MYC interact directly in vitro and co-localize in cells and on chromatin. ASH2L is a core subunit of KMT2 methyltransferase complexes that target histone H3 lysine 4 (H3K4), a mark associated with open chromatin. Indeed, MYC associates with H3K4 methyltransferase activity, dependent on the presence of ASH2L. MYC does not regulate this methyltransferase activity but stimulates demethylation and subsequently acetylation of H3K27. KMT2 complexes have been reported to associate with histone H3K27-specific demethylases, while CBP/p300, which interact with MYC, acetylate H3K27. Finally WDR5, another core subunit of KMT2 complexes, also binds directly to MYC and in genome-wide analyses MYC and WDR5 are associated with transcribed promoters. Thus, our findings suggest that MYC and ASH2L-KMT2 complexes cooperate in gene transcription by controlling H3K27 modifications and thereby regulate bivalent chromatin.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Línea Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Humanos , Péptidos y Proteínas de Señalización Intracelular , Metilación , Proteínas Nucleares/antagonistas & inhibidores , Regiones Promotoras Genéticas , Factores de Transcripción/antagonistas & inhibidores
3.
Proc Natl Acad Sci U S A ; 109(4): E187-96, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22190494

RESUMEN

Silent information regulator 1 (SIRT1) represents an NAD(+)-dependent deacetylase that inhibits proapoptotic factors including p53. Here we determined whether SIRT1 is downstream of the prototypic c-MYC oncogene, which is activated in the majority of tumors. Elevated expression of c-MYC in human colorectal cancer correlated with increased SIRT1 protein levels. Activation of a conditional c-MYC allele induced increased levels of SIRT1 protein, NAD(+), and nicotinamide-phosphoribosyltransferase (NAMPT) mRNA in several cell types. This increase in SIRT1 required the induction of the NAMPT gene by c-MYC. NAMPT is the rate-limiting enzyme of the NAD(+) salvage pathway and enhances SIRT1 activity by increasing the amount of NAD(+). c-MYC also contributed to SIRT1 activation by sequestering the SIRT1 inhibitor deleted in breast cancer 1 (DBC1) from the SIRT1 protein. In primary human fibroblasts previously immortalized by introduction of c-MYC, down-regulation of SIRT1 induced senescence and apoptosis. In various cell lines inactivation of SIRT1 by RNA interference, chemical inhibitors, or ectopic DBC1 enhanced c-MYC-induced apoptosis. Furthermore, SIRT1 directly bound to and deacetylated c-MYC. Enforced SIRT1 expression increased and depletion/inhibition of SIRT1 reduced c-MYC stability. Depletion/inhibition of SIRT1 correlated with reduced lysine 63-linked polyubiquitination of c-Myc, which presumably destabilizes c-MYC by supporting degradative lysine 48-linked polyubiquitination. Moreover, SIRT1 enhanced the transcriptional activity of c-MYC. Taken together, these results show that c-MYC activates SIRT1, which in turn promotes c-MYC function. Furthermore, SIRT1 suppressed cellular senescence in cells with deregulated c-MYC expression and also inhibited c-MYC-induced apoptosis. Constitutive activation of this positive feedback loop may contribute to the development and maintenance of tumors in the context of deregulated c-MYC.


Asunto(s)
Apoptosis/fisiología , Senescencia Celular/fisiología , Citocinas/metabolismo , Retroalimentación Fisiológica/fisiología , Nicotinamida Fosforribosiltransferasa/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sirtuina 1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Cicloheximida , Cartilla de ADN/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Células HEK293 , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , NAD/metabolismo , Proteínas del Tejido Nervioso , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/aislamiento & purificación , Ubiquitinación
4.
Proc Natl Acad Sci U S A ; 107(1): 58-63, 2010 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-19966300

RESUMEN

The MYC and RAS oncogenes are frequently activated in cancer and, together, are sufficient to transform rodent cells. The basis for this cooperativity remains unclear. We found that although Ras interfered with Myc-induced apoptosis, Myc repressed Ras-induced senescence, together abrogating two main barriers of tumorigenesis. Inhibition of cellular senescence required phosphorylation of Myc at Ser-62 by cyclin E/cyclin-dependent kinase (Cdk) 2. Cdk2 interacted with Myc at promoters, where it affected Myc-dependent regulation of genes, including Bmi-1, p16, p21, and hTERT, which encode proteins known to control senescence. Repression of senescence by Myc was abrogated by the Cdk inhibitor p27Kip1, which is induced by antiproliferative signals like IFN-gamma or by pharmacological inhibitors of Cdk2 but not by inhibitors of other Cdks. In contrast, a phospho-mimicking Myc-S62D mutant was resistant to these manipulations. Inhibition of cyclin E/Cdk2 reversed the senescence-associated gene expression pattern imposed by Myc/cyclin E/Cdk2. This indicates a role of Cdk2 as a transcriptional cofactor and activator of the antisenescence function of Myc and provides mechanistic insight into the Myc-p27Kip1 antagonism. Finally, our findings highlight that pharmacological inhibition of Cdk2 activity is a potential therapeutical principle for cancer therapy, in particular for tumors with activated Myc or Ras.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Senescencia Celular/fisiología , Quinasa 2 Dependiente de la Ciclina/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas ras/metabolismo , Animales , Línea Celular Tumoral , Ciclina E/genética , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Interferón gamma/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/genética , Ratas , Serina/metabolismo , Proteínas ras/genética
5.
Nat Commun ; 11(1): 1032, 2020 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32098961

RESUMEN

The AMP-activated protein kinase (AMPK) is a master sensor of the cellular energy status that is crucial for the adaptive response to limited energy availability. AMPK is implicated in the regulation of many cellular processes, including autophagy. However, the precise mechanisms by which AMPK controls these processes and the identities of relevant substrates are not fully understood. Using protein microarrays, we identify Cyclin Y as an AMPK substrate that is phosphorylated at Serine 326 (S326) both in vitro and in cells. Phosphorylation of Cyclin Y at S326 promotes its interaction with the Cyclin-dependent kinase 16 (CDK16), thereby stimulating its catalytic activity. When expressed in cells, Cyclin Y/CDK16 is sufficient to promote autophagy. Moreover, Cyclin Y/CDK16 is necessary for efficient AMPK-dependent activation of autophagy. This functional interaction is mediated by AMPK phosphorylating S326 of Cyclin Y. Collectively, we define Cyclin Y/CDK16 as downstream effector of AMPK for inducing autophagy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia/fisiología , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Beclina-1/metabolismo , Compuestos de Bifenilo , Quinasas Ciclina-Dependientes/genética , Ciclinas/genética , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Células 3T3 NIH , Fosforilación/efectos de los fármacos , Análisis por Matrices de Proteínas , Pironas/farmacología , Serina/metabolismo , Tiofenos/farmacología
6.
Methods Mol Biol ; 1732: 373-391, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29480488

RESUMEN

AMPK is an energy-sensing kinase and is required for the induction and progression of the autophagy process. In this chapter, we describe experimental approaches to study the steady state and flux of autophagy in response to AMPK activation. For this purpose, we provide detailed protocols for the measurement of general as well as AMPK-specific autophagy markers by immunoblot and immunofluorescence analysis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia/efectos de los fármacos , Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/farmacología , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Biomarcadores/análisis , Compuestos de Bifenilo , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Fibroblastos , Técnica del Anticuerpo Fluorescente/instrumentación , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Immunoblotting/instrumentación , Immunoblotting/métodos , Macrólidos/farmacología , Ratones , Células 3T3 NIH , Pironas/farmacología , Ribonucleótidos/farmacología , Tiofenos/farmacología
7.
Mol Neurobiol ; 55(3): 2524-2546, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28401474

RESUMEN

SIL1 acts as a co-chaperone for the major ER-resident chaperone BiP and thus plays a role in many BiP-dependent cellular functions such as protein-folding control and unfolded protein response. Whereas the increase of BiP upon cellular stress conditions is a well-known phenomenon, elevation of SIL1 under stress conditions was thus far solely studied in yeast, and different studies indicated an adverse effect of SIL1 increase. This is seemingly in contrast with the beneficial effect of SIL1 increase in surviving neurons in neurodegenerative disorders such as amyotrophic lateral sclerosis and Alzheimer's disease. Here, we addressed these controversial findings. Applying cell biological, morphological and biochemical methods, we demonstrated that SIL1 increases in various mammalian cells and neuronal tissues upon cellular stress. Investigation of heterozygous SIL1 mutant cells and tissues supported this finding. Moreover, SIL1 protein was found to be stabilized during ER stress. Increased SIL1 initiates ER stress in a concentration-dependent manner which agrees with the described adverse SIL1 effect. However, our results also suggest that protective levels are achieved by the secretion of excessive SIL1 and GRP170 and that moderately increased SIL1 also ameliorates cellular fitness under stress conditions. Our immunoprecipitation results indicate that SIL1 might act in a BiP-independent manner. Proteomic studies showed that SIL1 elevation alters the expression of proteins including crucial players in neurodegeneration, especially in Alzheimer's disease. This finding agrees with our observation of increased SIL1 immunoreactivity in surviving neurons of Alzheimer's disease autopsy cases and supports the assumption that SIL1 plays a protective role in neurodegenerative disorders.


Asunto(s)
Rastreo Celular , Cerebro/metabolismo , Factores de Intercambio de Guanina Nucleótido/biosíntesis , Factores de Intercambio de Guanina Nucleótido/genética , Animales , Rastreo Celular/métodos , Células Cultivadas , Cerebro/química , Cerebro/citología , Chaperón BiP del Retículo Endoplásmico , Femenino , Expresión Génica , Factores de Intercambio de Guanina Nucleótido/análisis , Células HEK293 , Humanos , Masculino , Ratones , Ratones Transgénicos , Proteómica/métodos
8.
Cell Death Differ ; 24(10): 1655-1671, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28622300

RESUMEN

Amyotrophic lateral sclerosis (ALS) is characterized by the selective degeneration of motor neurons (MNs) and their target muscles. Misfolded proteins which often form intracellular aggregates are a pathological hallmark of ALS. Disruption of the functional interplay between protein degradation (ubiquitin proteasome system and autophagy) and RNA-binding protein homeostasis has recently been suggested as an integrated model that merges several ALS-associated proteins into a common pathophysiological pathway. The E102Q mutation in one such candidate gene, the endoplasmic reticulum (ER) chaperone Sigma receptor-1 (SigR1), has been reported to cause juvenile ALS. Although loss of SigR1 protein contributes to neurodegeneration in several ways, the molecular mechanisms underlying E102Q-SigR1-mediated neurodegeneration are still unclear. In the present study, we showed that the E102Q-SigR1 protein rapidly aggregates and accumulates in the ER and associated compartments in transfected cells, leading to structural alterations of the ER, nuclear envelope and mitochondria and to subsequent defects in proteasomal degradation and calcium homeostasis. ER defects and proteotoxic stress generated by E102Q-SigR1 aggregates further induce autophagy impairment, accumulation of stress granules and cytoplasmic aggregation of the ALS-linked RNA-binding proteins (RBPs) matrin-3, FUS, and TDP-43. Similar ultrastructural abnormalities as well as altered protein degradation and misregulated RBP homeostasis were observed in primary lymphoblastoid cells (PLCs) derived from E102Q-SigR1 fALS patients. Consistent with these findings, lumbar α-MNs of both sALS as well as fALS patients showed cytoplasmic matrin-3 aggregates which were not co-localized with pTDP-43 aggregates. Taken together, our results support the notion that E102Q-SigR1-mediated ALS pathogenesis comprises a synergistic mechanism of both toxic gain and loss of function involving a vicious circle of altered ER function, impaired protein homeostasis and defective RBPs.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Estrés del Retículo Endoplásmico/genética , Homeostasis/genética , Mutación/genética , Proteínas de Unión al ARN/metabolismo , Receptores sigma/genética , Animales , Retículo Endoplásmico/metabolismo , Humanos , Ratones , Neuronas Motoras/metabolismo , ARN/metabolismo , Receptor Sigma-1
9.
Oncogene ; 24(12): 1982-93, 2005 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-15674325

RESUMEN

The proto-oncoprotein c-Myc functions as a transcriptional regulator that controls different aspects of cell behavior, including proliferation, differentiation, and apoptosis. In addition, Myc proteins have the potential to transform cells and are deregulated in the majority of human cancers. Several Myc-interacting factors have been described that mediate part of Myc's functions in the control of cell behavior. Here, we describe the isolation of a novel 150 kDa protein, designated PARP-10, that interacts with Myc. PARP-10 possesses domains with homology to RNA recognition motifs and to poly(ADP-ribose) polymerases (PARP). Molecular modeling and biochemical analysis define a PARP domain that is capable of ADP-ribosylating PARP-10 itself and core histones, but neither Myc nor Max. PARP-10 is localized to the nuclear and cytoplasmic compartments that is controlled at least in part by a Leu-rich nuclear export sequence (NES). Functionally, PARP-10 inhibits c-Myc- and E1A-mediated cotransformation of rat embryo fibroblasts, a function that is independent of PARP activity but that depends on a functional NES. Together, our findings define a novel PARP enzyme involved in the control of cell proliferation.


Asunto(s)
Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Secuencia de Aminoácidos , División Celular , Línea Celular , Mapeo Cromosómico , Humanos , Hibridación Fluorescente in Situ , Datos de Secuencia Molecular , Plásmidos , Biosíntesis de Proteínas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transcripción Genética
11.
PLoS One ; 10(4): e0123736, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25860957

RESUMEN

Inhibitor of growth (ING) proteins have multiple functions in the control of cell proliferation, mainly by regulating processes associated with chromatin regulation and gene expression. ING5 has been described to regulate aspects of gene transcription and replication. Moreover deregulation of ING5 is observed in different tumors, potentially functioning as a tumor suppressor. Gene transcription in late G1 and in S phase and replication is regulated by cyclin-dependent kinase 2 (CDK2) in complex with cyclin E or cyclin A. CDK2 complexes phosphorylate and regulate several substrate proteins relevant for overcoming the restriction point and promoting S phase. We have identified ING5 as a novel CDK2 substrate. ING5 is phosphorylated at a single site, threonine 152, by cyclin E/CDK2 and cyclin A/CDK2 in vitro. This site is also phosphorylated in cells in a cell cycle dependent manner, consistent with it being a CDK2 substrate. Furthermore overexpression of cyclin E/CDK2 stimulates while the CDK2 inhibitor p27KIP1 represses phosphorylation at threonine 152. This site is located in a bipartite nuclear localization sequence but its phosphorylation was not sufficient to deregulate the subcellular localization of ING5. Although ING5 interacts with the tumor suppressor p53, we could not establish p53-dependent regulation of cell proliferation by ING5 and by phospho-site mutants. Instead we observed that the knockdown of ING5 resulted in a strong reduction of proliferation in different tumor cell lines, irrespective of the p53 status. This inhibition of proliferation was at least in part due to the induction of apoptosis. In summary we identified a phosphorylation site at threonine 152 of ING5 that is cell cycle regulated and we observed that ING5 is necessary for tumor cell proliferation, without any apparent dependency on the tumor suppressor p53.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Sustitución de Aminoácidos , Sitios de Unión , Línea Celular , Proliferación Celular/fisiología , Ciclina A/antagonistas & inhibidores , Ciclina A/genética , Ciclina A/metabolismo , Ciclina E/antagonistas & inhibidores , Ciclina E/genética , Ciclina E/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Fosforilación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Treonina/química , Factores de Transcripción/química , Factores de Transcripción/genética , Transfección , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética
12.
Cell Signal ; 26(9): 2051-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24882689

RESUMEN

COP9 signalosome subunit 5 (CSN5) plays a decisive role in cellular processes such as cell cycle regulation and apoptosis via promoting protein degradation, gene transcription, and nuclear export. CSN5 regulates cullin-RING-E3 ligase (CRL) activity through its deNEDDylase function. It is overexpressed in several tumor entities, but its role in colorectal cancer (CRC) is poorly understood. Wnt/ß-catenin signaling is aberrant in most CRC cells, resulting in increased levels of oncogenic ß-catenin and thus tumor progression. Under physiological conditions, ß-catenin levels are tightly regulated by continuous proteasomal degradation. We recently showed that knockdown of CSN5 in model and CRC cells results in decreased (phospho)-ß-catenin levels. Reduced ß-catenin levels were associated with an attenuated proliferation rate of different CRC cell types after CSN5 knockdown. The canonical Wnt pathway involves degradation of ß-catenin by a ß-TrCP1-containing E3 ligase, but is mostly non-functional in CRC cells. We thus hypothesized that alternative ß-catenin degradation mediated by SIAH-1 (seven in absentia homolog-1), is responsible for the effect of CSN5 on ß-catenin signaling in CRC cells. We found that SIAH-1 plays an essential role in ß-catenin degradation in HCT116 CRC cells and that CSN5 affects ß-catenin target gene expression in these cells. Of note, CSN5 affected SIAH-1 mRNA and SIAH-1 protein levels. Moreover, ß-catenin and SIAH-1 form protein complexes with CSN5 in HCT116 cells. Lastly, we demonstrate that CSN5 promotes SIAH-1 degradation in HCT116 and SW480 cells and that this is associated with its deNEDDylase activity. In conclusion, we have identified a CSN5/ß-catenin/SIAH-1 interaction network that might control ß-catenin degradation in CRC cells.


Asunto(s)
Neoplasias Colorrectales/enzimología , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Complejo del Señalosoma COP9 , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Cicloheximida/farmacología , Células HCT116 , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína NEDD8 , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Inhibidores de la Síntesis de la Proteína/farmacología , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Ubiquitinas/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
13.
Gene ; 494(2): 145-60, 2012 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-22227497

RESUMEN

The proteins of the MYC/MAX/MAD network are central regulators of many key processes associated with basic cell physiology. These include the regulation of protein biosynthesis, energy metabolism, proliferation, and apoptosis. Molecularly the MYC/MAX/MAD network achieves these broad activities by controlling the expression of many target genes, which are primarily responsible for the diverse physiological consequences elicited by the network. The MYC proteins of the network possess oncogenic activity and their functional deregulation is associated with the majority of human tumors. Over the last years we have witnessed the accumulation of a considerable number of molecular observations that suggest many different biochemical means and tools by which MYC controls gene expression. We will summarize the more recent findings and discuss how these different building blocks might come together to explain how MYC regulates gene transcription. We note that despite the many molecular details known, we do not have an integrated view of how MYC uses the different tools, neither in a spatial nor in a temporal order.


Asunto(s)
Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas c-myc/genética , Acetilación , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN/metabolismo , Humanos , Procesamiento Proteico-Postraduccional , Ubiquitinación
14.
J Cell Biol ; 180(5): 915-29, 2008 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-18332217

RESUMEN

Cyclin-dependent kinases (Cdks) fulfill key functions in many cellular processes, including cell cycle progression and cytoskeletal dynamics. A limited number of Cdk substrates have been identified with few demonstrated to be regulated by Cdk-dependent phosphorylation. We identify on protein expression arrays novel cyclin E-Cdk2 substrates, including SIRT2, a member of the Sirtuin family of NAD(+)-dependent deacetylases that targets alpha-tubulin. We define Ser-331 as the site phosphorylated by cyclin E-Cdk2, cyclin A-Cdk2, and p35-Cdk5 both in vitro and in cells. Importantly, phosphorylation at Ser-331 inhibits the catalytic activity of SIRT2. Gain- and loss-of-function studies demonstrate that SIRT2 interfered with cell adhesion and cell migration. In postmitotic hippocampal neurons, neurite outgrowth and growth cone collapse are inhibited by SIRT2. The effects provoked by SIRT2, but not those of a nonphosphorylatable mutant, are antagonized by Cdk-dependent phosphorylation. Collectively, our findings identify a posttranslational mechanism that controls SIRT2 function, and they provide evidence for a novel regulatory circuitry involving Cdks, SIRT2, and microtubules.


Asunto(s)
Movimiento Celular/genética , Quinasas Ciclina-Dependientes/metabolismo , Sirtuinas/metabolismo , Secuencia de Aminoácidos , Animales , Dominio Catalítico , Diferenciación Celular/genética , Línea Celular , Ciclina A/genética , Ciclina A/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/genética , Conos de Crecimiento/metabolismo , Conos de Crecimiento/ultraestructura , Células HeLa , Hipocampo/embriología , Hipocampo/metabolismo , Hipocampo/ultraestructura , Humanos , Ratones , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional/genética , Serina/metabolismo , Sirtuina 2 , Sirtuinas/genética
15.
Cancer Res ; 68(3): 749-58, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18245475

RESUMEN

Regulation of chromatin is an important aspect of controlling promoter activity and gene expression. Posttranslational modifications of core histones allow proteins associated with gene transcription to access chromatin. Closely associated with promoters of actively transcribed genes, trimethylation of histone H3 at lysine 4 (H3K4me3) is a core histone mark set by several protein complexes. Some of these protein complexes contain the trithorax protein ASH2 combined with the MLL oncoproteins. We identified human ASH2 in a complex with the oncoprotein MYC. This finding, together with the observation that hASH2 interacts with MLL, led us to test whether hASH2 itself is involved in transformation. We observed that hASH2 cooperates with Ha-RAS to transform primary rat embryo fibroblasts (REF). Furthermore, transformation of REFs by MYC and Ha-RAS required the presence of rAsh2. In an animal model, the hASH2/Ha-RAS-transformed REFs formed rapidly growing tumors characteristic of fibrosarcomas that, compared with tumors derived from MYC/Ha-RAS transformed cells, were poorly differentiated. This finding suggests that ASH2 functions as an oncoprotein. Although hASH2 expression at the mRNA level was generally not deregulated, hASH2 protein expression was increased in most human tumors and tumor cell lines. In addition, knockdown of hASH2 inhibited tumor cell proliferation. Taken together, these observations define hASH2 as a novel oncoprotein.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/genética , Neoplasias/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética , Animales , Procesos de Crecimiento Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Proteínas de Unión al ADN/biosíntesis , Fibroblastos , Regulación Neoplásica de la Expresión Génica , Genes ras , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/biosíntesis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Endogámicas F344 , Factores de Transcripción/biosíntesis , Transfección
16.
J Biol Chem ; 281(46): 34725-9, 2006 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-16987807

RESUMEN

The proteins of the MYC family are key regulators of cell behavior. MYC, originally identified as an oncoprotein, affects growth, proliferation, differentiation, and apoptosis of cells through its ability to regulate a significant number of genes. In addition MYC governs events associated with tumor progression, including genetic stability, migration, and angiogenesis. The pleiotropic activities attributed to MYC and their balanced control requires that the expression and function of MYC is tightly controlled. Indeed many different pathways and factors have been identified that impinge on MYC gene expression and protein function. In particular the protein is subject to different posttranslational modifications, including phosphorylation, ubiquitinylation, and acetylation. Here we discuss the latest developments regarding these modifications that control various aspects of MYC function, including its stability, the interaction with partner proteins, and the transcriptional potential.


Asunto(s)
Procesamiento Proteico-Postraduccional/fisiología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sitios de Unión , Fosforilación
17.
EMBO J ; 25(21): 5159-70, 2006 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17053782

RESUMEN

Entry of cells into the cell division cycle requires the coordinated activation of cyclin-dependent kinases (cdks) and the deactivation of cyclin kinase inhibitors. Degradation of p27kip1 is known to be a central component of this process as it allows controlled activation of cdk2-associated kinase activity. Turnover of p27 at the G1/S transition is regulated through phosphorylation at T187 and subsequent SCF(skp2)-dependent ubiquitylation. However, detailed analysis of this process revealed the existence of additional pathways that regulate the abundance of the protein in early G1 and as cells exit quiescence. Here, we report on a molecular mechanism that regulates p27 stability by phosphorylation at T198. Phosphorylation of p27 at T198 prevents ubiquitin-dependent degradation of free p27. T198 phosphorylation also controls progression through the G1 phase of the cell cycle by regulating the association of p27 with cyclin-cdk complexes. Our results unveil the molecular composition of a pathway, which regulates the abundance and activity of p27kip1 during early G1. They also explain how the T187- and the T198-dependent turnover systems synergize to allow cell cycle progression in G1.


Asunto(s)
Fase G1/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Ubiquitina/metabolismo , Línea Celular Transformada , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Activación Enzimática/fisiología , Humanos , Fosforilación , Proteínas Quinasas Asociadas a Fase-S/metabolismo
18.
Mol Cell Biochem ; 276(1-2): 159-67, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16132697

RESUMEN

Human TSPY is a candidate oncogene and is supposed to function as a proliferation factor during spermatogenesis. It is the only mammalian protein-coding gene known to be organized as a tandem repeat gene family. It is expressed at highest level in spermatogonia and to a lower amount in primary spermatocytes. To characterize the human TSPY promoter we used the luciferase reporter system in a mouse spermatogonia derived cell line (GC-1 spg) and in a GC-4 spc cell line, that harbour prophase spermatocytes of the preleptotene and early pachytene stage. We isolated a 1303 bp fragment of the 5'-flanking region of exon 1 that shows significant promoter activity in GC-1 spg and reduced activity in GC-4 spc cells. In order to gain further insight into the organization of the TSPY-promoter, stepwise truncations of the putative promoter sequence were performed. The resulting fragments were cloned into the pGL 3-vector and analysed for reporter gene activity in the murine germ cell lines GC-1 spg and GC-4 spc, leading to the characterization of a core promoter (--159 to--1), an enhancing region (--673 to--364) and a silencing region (--1262 to--669). Database research for cis-active elements yielded two putative SOX-like binding sites in the enhancing region and reporter gene activity was drastically reduced when three nucleotides of the AACAAT SOX core sequence were mutated. Our findings strongly suggest that testis-specific expression of human TSPY is mediated by Sox proteins.


Asunto(s)
Proteínas de Ciclo Celular/genética , Regiones Promotoras Genéticas/genética , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Secuencia Conservada , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Especificidad de Órganos , Testículo/metabolismo , Transcripción Genética
19.
EMBO Rep ; 4(5): 484-90, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12776737

RESUMEN

The c-MYC oncoprotein regulates various aspects of cell behaviour by modulating gene expression. Here, we report the identification of the cAMP-response-element-binding protein (CBP) as a novel c-MYC binding partner. The two proteins interact both in vitro and in cells, and CBP binds to the carboxy-terminal region of c-MYC. Importantly, CBP, as well as p300, is associated with E-box-containing promoter regions of genes that are regulated by c-MYC. Furthermore, c-MYC and CBP/p300 function synergistically in the activation of reporter-gene constructs. Thus, CBP and p300 function as positive cofactors for c-MYC. In addition, c-MYC is acetylated in cells. This modification does not require MYC box II, suggesting that it is independent of TRRAP complexes. Instead, CBP acetylates c-MYC in vitro, and co-expression of CBP with c-MYC stimulates in vivo acetylation. Functionally, this results in a decrease in ubiquitination and stabilization of c-MYC proteins. Thus, CBP and p300 are novel functional binding partners of c-MYC.


Asunto(s)
Acetiltransferasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Acetilcoenzima A/metabolismo , Acetilación , Acetiltransferasas/genética , Proteínas Adaptadoras Transductoras de Señales , Sitios de Unión , Proteínas de Ciclo Celular/genética , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/química , Genes Reporteros , Histona Acetiltransferasas , Humanos , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-myc/química , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción , Transcripción Genética , Activación Transcripcional , Factores de Transcripción p300-CBP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA