Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4450, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38789411

RESUMEN

Histone deacetylases (HDACs) play a crucial role in transcriptional regulation and are implicated in various diseases, including cancer. They are involved in histone tail deacetylation and canonically linked to transcriptional repression. Previous studies suggested that HDAC recruitment to cell-cycle gene promoters via the retinoblastoma (RB) protein or the DREAM complex through SIN3B is essential for G1/S and G2/M gene repression during cell-cycle arrest and exit. Here we investigate the interplay among DREAM, RB, SIN3 proteins, and HDACs in the context of cell-cycle gene repression. Knockout of SIN3B does not globally derepress cell-cycle genes in non-proliferating HCT116 and C2C12 cells. Loss of SIN3A/B moderately upregulates several cell-cycle genes in HCT116 cells but does so independently of DREAM/RB. HDAC inhibition does not induce general upregulation of RB/DREAM target genes in arrested transformed or non-transformed cells. Our findings suggest that E2F:RB and DREAM complexes can repress cell-cycle genes without relying on HDAC activity.


Asunto(s)
Factores de Transcripción E2F , Histona Desacetilasas , Proteínas Represoras , Proteína de Retinoblastoma , Humanos , Histona Desacetilasas/metabolismo , Histona Desacetilasas/genética , Células HCT116 , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Factores de Transcripción E2F/metabolismo , Factores de Transcripción E2F/genética , Proteína de Retinoblastoma/metabolismo , Proteína de Retinoblastoma/genética , Ratones , Animales , Complejo Correpresor Histona Desacetilasa y Sin3/metabolismo , Complejo Correpresor Histona Desacetilasa y Sin3/genética , Proteínas de Interacción con los Canales Kv/metabolismo , Proteínas de Interacción con los Canales Kv/genética , Ciclo Celular/genética , Regiones Promotoras Genéticas/genética , Regulación de la Expresión Génica , Genes cdc
2.
bioRxiv ; 2023 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-37961464

RESUMEN

Histone deacetylases (HDACs) are pivotal in transcriptional regulation, and their dysregulation has been associated with various diseases including cancer. One of the critical roles of HDAC-containing complexes is the deacetylation of histone tails, which is canonically linked to transcriptional repression. Previous research has indicated that HDACs are recruited to cell-cycle gene promoters through the RB protein or the DREAM complex via SIN3B and that HDAC activity is essential for repressing G1/S and G2/M cell-cycle genes during cell-cycle arrest and exit. In this study, we sought to explore the interdependence of DREAM, RB, SIN3 proteins, and HDACs in the context of cell-cycle gene repression. We found that genetic knockout of SIN3B did not lead to derepression of cell-cycle genes in non-proliferating HCT116 and C2C12 cells. A combined loss of SIN3A and SIN3B resulted in a moderate upregulation in mRNA expression of several cell-cycle genes in arrested HCT116 cells, however, these effects appeared to be independent of DREAM or RB. Furthermore, HDAC inhibition did not induce a general upregulation of RB and DREAM target gene expression in arrested transformed or non-transformed cells. Our findings provide evidence that E2F:RB and DREAM complexes can repress cell-cycle genes without reliance on HDAC activity.

3.
J Biol Chem ; 298(9): 102319, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35926712

RESUMEN

B-Myb is a highly conserved member of the vertebrate Myb family of transcription factors that plays a critical role in cell-cycle progression and proliferation. Myb proteins activate Myb-dependent promoters by interacting specifically with Myb-binding site (MBS) sequences using their DNA-binding domain (DBD). Transactivation of MBS promoters by B-Myb is repressed by its negative regulatory domain (NRD), and phosphorylation of the NRD by Cdk2-CyclinA relieves the repression to activate B-Myb-dependent promoters. However, the structural mechanisms underlying autoinhibition and activation of B-Myb-mediated transcription have been poorly characterized. Here, we determined that a region in the B-Myb NRD (residues 510-600) directly associates with the DBD and inhibits binding of the DBD to the MBS DNA sequence. We demonstrate using biophysical assays that phosphorylation of the NRD at T515, T518, and T520 is sufficient to disrupt the interaction between the NRD and the DBD, which results in increased affinity for MBS DNA and increased B-Myb-dependent promoter activation in cell assays. Our biochemical characterization of B-Myb autoregulation and the activating effects of phosphorylation provide insight into how B-Myb functions as a site-specific transcription factor.


Asunto(s)
Proteínas de Ciclo Celular , Quinasa 2 Dependiente de la Ciclina , ADN , Transactivadores , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , ADN/metabolismo , Humanos , Fosforilación , Dominios Proteicos , Transactivadores/química , Transactivadores/metabolismo , Activación Transcripcional
4.
Trends Biochem Sci ; 47(12): 1009-1022, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35835684

RESUMEN

Cell cycle-dependent gene transcription is tightly controlled by the retinoblastoma (RB):E2F and DREAM complexes, which repress all cell cycle genes during quiescence. Cyclin-dependent kinase (CDK) phosphorylation of RB and DREAM allows for the expression of two gene sets. The first set of genes, with peak expression in G1/S, is activated by E2F transcription factors (TFs) and is required for DNA synthesis. The second set, with maximum expression during G2/M, is required for mitosis and is coordinated by the MuvB complex, together with B-MYB and Forkhead box M1 (FOXM1). In this review, we summarize the key findings that established the distinct control mechanisms regulating G1/S and G2/M gene expression in mammals and discuss recent advances in the understanding of the temporal control of these genes.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas Represoras , Animales , Proteínas Represoras/genética , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Mitosis , Quinasas Ciclina-Dependientes/genética , Expresión Génica , Mamíferos
5.
Structure ; 30(9): 1340-1353.e3, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35716663

RESUMEN

The retinoblastoma protein (Rb) and its homologs p107 and p130 are critical regulators of gene expression during the cell cycle and are commonly inactivated in cancer. Rb proteins use their "pocket domain" to bind an LxCxE sequence motif in other proteins, many of which function with Rb proteins to co-regulate transcription. Here, we present binding data and crystal structures of the p107 pocket domain in complex with LxCxE peptides from the transcriptional co-repressor proteins HDAC1, ARID4A, and EID1. Our results explain why Rb and p107 have weaker affinity for cellular LxCxE proteins compared with the E7 protein from human papillomavirus, which has been used as the primary model for understanding LxCxE motif interactions. Our structural and mutagenesis data also identify and explain differences in Rb and p107 affinities for some LxCxE-containing sequences. Our study provides new insights into how Rb proteins bind their cell partners with varying affinity and specificity.


Asunto(s)
Proteínas Represoras , Proteína de Retinoblastoma , Ciclo Celular , Humanos , Proteínas Represoras/genética , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Proteína p130 Similar a la del Retinoblastoma/metabolismo
6.
Oncogene ; 41(21): 2909-2919, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35468940

RESUMEN

Proper progression through the cell-division cycle is critical to normal development and homeostasis and is necessarily misregulated in cancer. The key to cell-cycle regulation is the control of two waves of transcription that occur at the onset of DNA replication (S phase) and mitosis (M phase). MuvB complexes play a central role in the regulation of these genes. When cells are not actively dividing, the MuvB complex DREAM represses G1/S and G2/M genes. Remarkably, MuvB also forms activator complexes together with the oncogenic transcription factors B-MYB and FOXM1 that are required for the expression of the mitotic genes in G2/M. Despite this essential role in the control of cell division and the relationship to cancer, it has been unclear how MuvB complexes inhibit and stimulate gene expression. Here we review recent discoveries of MuvB structure and molecular interactions, including with nucleosomes and other chromatin-binding proteins, which have led to the first mechanistic models for the biochemical function of MuvB complexes.


Asunto(s)
Proteínas de Ciclo Celular , Neoplasias , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Humanos , Mitosis/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/radioterapia , Transactivadores/genética
7.
Nat Commun ; 13(1): 526, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35082292

RESUMEN

The chromatin architecture in promoters is thought to regulate gene expression, but it remains uncertain how most transcription factors (TFs) impact nucleosome position. The MuvB TF complex regulates cell-cycle dependent gene-expression and is critical for differentiation and proliferation during development and cancer. MuvB can both positively and negatively regulate expression, but the structure of MuvB and its biochemical function are poorly understood. Here we determine the overall architecture of MuvB assembly and the crystal structure of a subcomplex critical for MuvB function in gene repression. We find that the MuvB subunits LIN9 and LIN37 function as scaffolding proteins that arrange the other subunits LIN52, LIN54 and RBAP48 for TF, DNA, and histone binding, respectively. Biochemical and structural data demonstrate that MuvB binds nucleosomes through an interface that is distinct from LIN54-DNA consensus site recognition and that MuvB increases nucleosome occupancy in a reconstituted promoter. We find in arrested cells that MuvB primarily associates with a tightly positioned +1 nucleosome near the transcription start site (TSS) of MuvB-regulated genes. These results support a model that MuvB binds and stabilizes nucleosomes just downstream of the TSS on its target promoters to repress gene expression.


Asunto(s)
Genes cdc , Nucleosomas/metabolismo , Unión Proteica , Sitio de Iniciación de la Transcripción , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , División Celular/fisiología , Cromatina , ADN/metabolismo , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo
8.
Cell Death Differ ; 28(12): 3357-3370, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34183782

RESUMEN

Ki-67 serves as a prominent cancer marker. We describe how expression of the MKI67 gene coding for Ki-67 is controlled during the cell cycle. MKI67 mRNA and Ki-67 protein are maximally expressed in G2 phase and mitosis. Expression is dependent on two CHR elements and one CDE site in the MKI67 promoter. DREAM transcriptional repressor complexes bind to both CHR sites and downregulate the expression in G0/G1 cells. Upregulation of MKI67 transcription coincides with binding of B-MYB-MuvB and FOXM1-MuvB complexes from S phase into G2/M. Importantly, binding of B-MYB to the two CHR elements correlates with loss of CHR-dependent MKI67 promoter activation in B-MYB-knockdown experiments. In knockout cell models, we find that DREAM/MuvB-dependent transcriptional control cooperates with the RB Retinoblastoma tumor suppressor. Furthermore, the p53 tumor suppressor indirectly downregulates transcription of the MKI67 gene. This repression by p53 requires p21/CDKN1A. These results are consistent with a model in which DREAM, B-MYB-MuvB, and FOXM1-MuvB together with RB cooperate in cell cycle-dependent transcription and in transcriptional repression following p53 activation. In conclusion, we present mechanisms how MKI67 gene expression followed by Ki-67 protein synthesis is controlled during the cell cycle and upon induction of DNA damage, as well as upon p53 activation.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Antígeno Ki-67/genética , Humanos , Transfección
9.
Methods Mol Biol ; 2267: 81-90, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33786786

RESUMEN

The interaction of proteins with DNA plays a central role in gene regulation. We describe a DNA affinity purification method that allows for identification and analysis of protein complex components. For example, a DNA probe carrying a transcription factor binding site is used to purify proteins from a nuclear extract. The proteins binding to the probe are then identified by mass spectrometry. In similar experiments, proteins purified by this pulldown method can be analyzed by Western blot. Employing this method, we found that the DREAM transcriptional repressor complex binds to CHR transcriptional elements in promoters of cell cycle genes. This complex is important for cell cycle-dependent repression and as part of the p53-DREAM pathway serves as a link for indirect transcriptional repression of target genes by the tumor suppressor p53. In general, the methods described can be applied for the identification and analysis of proteins binding to DNA.


Asunto(s)
Fraccionamiento Químico/métodos , ADN/química , Inmunoprecipitación/métodos , Factores de Transcripción/metabolismo , Animales , Biotinilación/métodos , Western Blotting/métodos , Línea Celular , ADN/metabolismo , Humanos , Espectrometría de Masas/métodos , Regiones Promotoras Genéticas , Unión Proteica
10.
Cancers (Basel) ; 12(5)2020 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-32365858

RESUMEN

In search of new biomarkers suitable for the diagnosis and treatment of prostate cancer, genome-wide transcriptome sequencing was carried out with tissue specimens from 40 prostate cancer (PCa) and 8 benign prostate hyperplasia patients. We identified two intergenic long non-coding transcripts, located in close genomic proximity, which are highly expressed in PCa. Microarray studies on a larger cohort comprising 155 patients showed a profound diagnostic potential of these transcripts (AUC~0.94), which we designated as tumor associated prostate cancer increased lncRNA (TAPIR-1 and -2). To test their therapeutic potential, knockdown experiments with siRNA were carried out. The knockdown caused an increase in the p53/TP53 tumor suppressor protein level followed by downregulation of a large number of cell cycle- and DNA-damage repair key regulators. Furthermore, in radiation therapy resistant tumor cells, the knockdown leads to a renewed sensitization of these cells to radiation treatment. Accordingly, in a preclinical PCa xenograft model in mice, the systemic application of nanoparticles loaded with siRNA targeting TAPIR-1 significantly reduced tumor growth. These findings point to a crucial role of TAPIR-1 and -2 in PCa.

11.
Nucleic Acids Res ; 47(17): 9087-9103, 2019 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-31400114

RESUMEN

Most human cancers acquire mutations causing defects in the p53 signaling pathway. The tumor suppressor p53 becomes activated in response to genotoxic stress and is essential for arresting the cell cycle to facilitate DNA repair or to initiate apoptosis. p53-induced cell cycle-arrest is mediated by expression of the CDK inhibitor p21WAF1/Cip1, which prevents phosphorylation and inactivation of the pocket proteins RB, p130, and p107. In a hypophosphorylated state, pocket proteins bind to E2F factors forming RB-E2F and DREAM transcriptional repressor complexes. Here, we analyze the influence of RB and DREAM on p53-induced gene repression and cell-cycle arrest. We show that abrogation of DREAM function by knockout of the DREAM component LIN37 results in a reduced repression of cell-cycle genes. We identify the genes repressed by the p53-DREAM pathway and describe a set of genes that is downregulated by p53 independent of LIN37/DREAM. Most strikingly, p53-dependent repression of cell-cycle genes is completely abrogated in LIN37-/-;RB-/- cells leading to a loss of the G1/S checkpoint. Taken together, we show that DREAM and RB are key factors in the p53 signaling pathway to downregulate a large number of cell-cycle genes and to arrest the cell cycle at the G1/S transition.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Regulación de la Expresión Génica , Proteínas de Interacción con los Canales Kv/metabolismo , Proteínas Represoras/metabolismo , Proteína de Retinoblastoma/genética , Transactivadores/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Células Cultivadas , Proteína Sustrato Asociada a CrK/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Factores de Transcripción E2F/genética , Factores de Transcripción E2F/metabolismo , Fibroblastos/metabolismo , Genes cdc , Células HCT116 , Humanos , Proteínas de Interacción con los Canales Kv/genética , Ratones , Proteínas Represoras/genética , Proteína de Retinoblastoma/metabolismo , Proteína p107 Similar a la del Retinoblastoma/genética , Transactivadores/genética , Transactivadores/metabolismo , Proteína p53 Supresora de Tumor/genética
12.
Oncotarget ; 8(58): 97736-97748, 2017 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-29228647

RESUMEN

A central question in cell cycle control is how differential gene expression is regulated. Timing of expression is important for correct progression through the cell cycle. E2F, CDE, and CHR promoter sites have been linked to transcriptional repression in resting cells and activation during the cell cycle. Further, the DREAM complex binds CHR or CDE/CHR elements of G2/M genes resulting in repression during G0/G1. Here, we show that DREAM also binds to E2F sites of S phase genes in quiescence and upon p53 activation. Furthermore, we describe a novel class of promoter sites, the CHR-like elements (CLE), which can support binding of DREAM to E2F elements. Activation of such S phase genes is achieved through binding of E2F1-3/DP complexes to E2F sites. In contrast, the activating MuvB complexes MMB and FOXM1-MuvB bind to CHR elements and mediate peak expression in G2/M. In conclusion, data presented here in combination with earlier results leads us to propose a model that explains how DREAM can repress early cell cycle genes through E2F or E2F/CLE sites and late genes through CHR or CDE/CHR elements. Also p53-dependent indirect transcriptional repression through the p53-p21-Cyclin/CDK-DREAM-E2F/CLE/CDE/CHR pathway requires DREAM binding to E2F or E2F/CLE sites in early cell cycle genes and binding of DREAM to CHR or CDE/CHR elements of late cell cycle genes. Specific timing of activation is achieved through binding of E2F1-3/DP to E2F sites and MMB or FOXM1-MuvB complexes to CHR elements.

13.
Elife ; 62017 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-28920576

RESUMEN

The retinoblastoma Rb protein is an important factor controlling the cell cycle. Yet, mammalian cells carrying Rb deletions are still able to arrest under growth-limiting conditions. The Rb-related proteins p107 and p130, which are components of the DREAM complex, had been suggested to be responsible for a continued ability to arrest by inhibiting E2f activity and by recruiting chromatin-modifying enzymes. Here, we show that p130 and p107 are not sufficient for DREAM-dependent repression. We identify the MuvB protein Lin37 as an essential factor for DREAM function. Cells not expressing Lin37 proliferate normally, but DREAM completely loses its ability to repress genes in G0/G1 while all remaining subunits, including p130/p107, still bind to target gene promoters. Furthermore, cells lacking both Rb and Lin37 are incapable of exiting the cell cycle. Thus, Lin37 is an essential component of DREAM that cooperates with Rb to induce quiescence.


Asunto(s)
Ciclo Celular , Regulación de la Expresión Génica , Proteína de Retinoblastoma/metabolismo , Transactivadores/metabolismo , Animales , Línea Celular , Técnicas de Inactivación de Genes , Ratones , Transactivadores/genética
14.
Crit Rev Biochem Mol Biol ; 52(6): 638-662, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28799433

RESUMEN

The precise timing of cell cycle gene expression is critical for the control of cell proliferation; de-regulation of this timing promotes the formation of cancer and leads to defects during differentiation and development. Entry into and progression through S phase requires expression of genes coding for proteins that function in DNA replication. Expression of a distinct set of genes is essential to pass through mitosis and cytokinesis. Expression of these groups of cell cycle-dependent genes is regulated by the RB pocket protein family, the E2F transcription factor family, and MuvB complexes together with B-MYB and FOXM1. Distinct combinations of these transcription factors promote the transcription of the two major groups of cell cycle genes that are maximally expressed either in S phase (G1/S) or in mitosis (G2/M). In this review, we discuss recent work that has started to uncover the molecular mechanisms controlling the precisely timed expression of these genes at specific cell cycle phases, as well as the repression of the genes when a cell exits the cell cycle.


Asunto(s)
Ciclo Celular , Factores de Transcripción E2F/metabolismo , Proteínas de Interacción con los Canales Kv/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Proteína de Retinoblastoma/metabolismo , Transactivadores/metabolismo , Activación Transcripcional , Animales , Diferenciación Celular , Factores de Transcripción E2F/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de Interacción con los Canales Kv/genética , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Nucleares/genética , Mapas de Interacción de Proteínas , Proteínas Represoras/genética , Proteína de Retinoblastoma/genética , Transactivadores/genética , Transcripción Genética
15.
Nucleic Acids Res ; 42(16): 10331-50, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25106871

RESUMEN

The cell cycle genes homology region (CHR) has been identified as a DNA element with an important role in transcriptional regulation of late cell cycle genes. It has been shown that such genes are controlled by DREAM, MMB and FOXM1-MuvB and that these protein complexes can contact DNA via CHR sites. However, it has not been elucidated which sequence variations of the canonical CHR are functional and how frequent CHR-based regulation is utilized in mammalian genomes. Here, we define the spectrum of functional CHR elements. As the basis for a computational meta-analysis, we identify new CHR sequences and compile phylogenetic motif conservation as well as genome-wide protein-DNA binding and gene expression data. We identify CHR elements in most late cell cycle genes binding DREAM, MMB, or FOXM1-MuvB. In contrast, Myb- and forkhead-binding sites are underrepresented in both early and late cell cycle genes. Our findings support a general mechanism: sequential binding of DREAM, MMB and FOXM1-MuvB complexes to late cell cycle genes requires CHR elements. Taken together, we define the group of CHR-regulated genes in mammalian genomes and provide evidence that the CHR is the central promoter element in transcriptional regulation of late cell cycle genes by DREAM, MMB and FOXM1-MuvB.


Asunto(s)
Ciclo Celular/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Genes cdc , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , División Celular/genética , Línea Celular , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/metabolismo , Fase G2/genética , Genoma , Humanos , Ratones , Células 3T3 NIH , Proteínas Proto-Oncogénicas c-myb/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética
16.
Virology ; 460-461: 194-206, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25010285

RESUMEN

Human immunodeficiency virus Gag drives assembly of virions in infected cells and interacts with host factors which facilitate or restrict viral replication. Although several Gag-binding proteins have been characterized, understanding of virus-host interactions remains incomplete. In a series of six affinity purification screens, we have identified protein candidates for interaction with HIV-1 Gag. Proteins previously found in virions or identified in siRNA screens for host factors influencing HIV-1 replication were recovered. Helicases, translation factors, cytoskeletal and motor proteins, factors involved in RNA degradation and RNA interference were enriched in the interaction data. Cellular networks of cytoskeleton, SR proteins and tRNA synthetases were identified. Most prominently, components of cytoplasmic RNA transport granules were co-purified with Gag. This study provides a survey of known Gag-host interactions and identifies novel Gag binding candidates. These factors are associated with distinct molecular functions and cellular pathways relevant in host-pathogen interactions.


Asunto(s)
Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/metabolismo , Proteoma/metabolismo , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/metabolismo , Infecciones por VIH/genética , VIH-1/genética , Interacciones Huésped-Patógeno , Humanos , Unión Proteica , Mapas de Interacción de Proteínas , Proteoma/genética , Replicación Viral , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética
17.
Nucleic Acids Res ; 42(1): 163-80, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24071582

RESUMEN

Infection by oncogenic viruses is a frequent cause for tumor formation as observed in cervical cancer. Viral oncoproteins cause inactivation of p53 function and false transcriptional regulation of central cell cycle genes. Here we analyze the regulation of Plk4, serving as an example of many cell cycle- and p53-regulated genes. Cell cycle genes are often repressed via CDE and CHR elements in their promoters and activated by NF-Y binding to CCAAT-boxes. In contrast, general activation of Plk4 depends on NRF1 and CRE sites. Bioinformatic analyses imply that NRF1 and CRE are central elements of the transcriptional network controlling cell cycle genes. We identify CDE and CHR sites in the Plk4 promoter, which are necessary for binding of the DREAM (DP, RB-like, E2F4 and MuvB) complex and for mediating repression in G0/G1. When cells progress to G2 and mitosis, DREAM is replaced by the MMB (Myb-MuvB) complex that only requires the CHR element for binding. Plk4 expression is downregulated by the p53-p21(WAF1/CIP1)-DREAM signaling pathway through the CDE and CHR sites. Cell cycle- and p53-dependent repression is abrogated by HPV E7 oncoprotein. Together with genome-wide analyses our results imply that many cell cycle genes upregulated in tumors by viral infection are bound by DREAM through CDE/CHR sites.


Asunto(s)
Proteínas E7 de Papillomavirus/metabolismo , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/genética , Proteínas Represoras/metabolismo , Activación Transcripcional , Animales , Secuencia de Bases , Sitios de Unión , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , Humanos , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Factor Nuclear 1 de Respiración/metabolismo , Elementos de Respuesta , Transactivadores/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
18.
Mol Cell Biol ; 33(2): 227-36, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23109430

RESUMEN

There are nearly 50 forkhead (FOX) transcription factors encoded in the human genome and, due to sharing a common DNA binding domain, they are all thought to bind to similar DNA sequences. It is therefore unclear how these transcription factors are targeted to specific chromatin regions to elicit specific biological effects. Here, we used chromatin immunoprecipitation followed by sequencing (ChIP-seq) to investigate the genome-wide chromatin binding mechanisms used by the forkhead transcription factor FOXM1. In keeping with its previous association with cell cycle control, we demonstrate that FOXM1 binds and regulates a group of genes which are mainly involved in controlling late cell cycle events in the G(2) and M phases. However, rather than being recruited through canonical RYAAAYA forkhead binding motifs, FOXM1 binding is directed via CHR (cell cycle genes homology region) elements. FOXM1 binds these elements through protein-protein interactions with the MMB transcriptional activator complex. Thus, we have uncovered a novel and unexpected mode of chromatin binding of a FOX transcription factor that allows it to specifically control cell cycle-dependent gene expression.


Asunto(s)
Ciclo Celular , Cromatina/metabolismo , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Animales , División Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Clonación Molecular , Biología Computacional , Proteínas de Unión al ADN , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/metabolismo , Genes cdc , Células HCT116 , Células HEK293 , Humanos , Ratones , Mitosis , Células 3T3 NIH , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Cell Cycle ; 11(24): 4661-72, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23187802

RESUMEN

The tumor suppressor p53 plays an important role in cell cycle arrest by downregulating transcription. Many genes repressed by p53 code for proteins with functions in G2/M. A large portion of these genes is controlled by cell cycle-dependent elements (CDE) and cell cycle genes homology regions (CHR) in their promoters. Cyclin B2 is an example of such a gene, with a function at the transition from G2 to mitosis. We find that p53-dependent downregulation of cyclin B2 promoter activity is dependent on an intact CHR element. In the presence of high levels of p53 or p21(WAF1/CIP1), protein binding to the CHR switches from MMB to DREAM complex by shifting MuvB core-associated proteins from B-Myb to E2F4/DP1/p130. The results suggest a model for p53-dependent transcriptional repression by which p53 directly activates p21(WAF1/CIP1). The inhibitor then prevents further phosphorylation of p130 by cyclin-dependent kinases. The presence of hypophosphorylated pocket proteins shifts the equilibrium for complex formation from MMB to DREAM. In the case of promoters that do not hold CDE or E2F elements, binding of DREAM and MMB solely relies on a CHR site. Thus, p53 can repress target genes indirectly through CHR elements.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ciclo Celular/genética , Ciclina B2/genética , Ciclina B2/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Doxorrubicina/farmacología , Células HCT116 , Humanos , Proteínas de Interacción con los Canales Kv/genética , Proteínas de Interacción con los Canales Kv/metabolismo , Ratones , Células 3T3 NIH , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína p53 Supresora de Tumor/genética
20.
FASEB J ; 26(2): 668-77, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22038052

RESUMEN

The vascular type of the Ehlers-Danlos syndrome (vEDS) is caused by dominant-negative mutations in the procollagen type III (COL3A1) gene. Patients with this autosomal dominant disorder have a shortened life expectancy due to complications from ruptured vessels or hollow organs. We tested the effectiveness of allele-specific RNA interference (RNAi) to reduce the mutated phenotype in fibroblasts. Small-interfering RNAs (siRNAs) discriminating between wild-type and mutant COL3A1 allele were identified by a luciferase reporter gene assay and in primary fibroblasts from a normal donor and a patient with vEDS. The best discriminative siRNA with the mutation at position 10 resulted in >90% silencing of the mutant allele without affecting the wild-type allele. Transmission and immunogold electron microscopy of extracted extracellular matrices from untreated fibroblasts of the patient with vEDS revealed structurally abnormal fibrils. After siRNA treatment, collagen fibrils became similar to fibrils from fibroblasts of normal and COL3A1 haploinsufficient donors. In addition, it was shown that expression of mutated COL3A1 activates the unfolded protein response and that reduction of the amount of mutated protein by siRNA reduces cellular stress. Taken together, the results provide evidence that allele-specific siRNAs are able to reduce negative effects of mutated COL3A1 proteins. Thus, the application of allele-specific RNAi may be a promising direction for future personalized therapies to reduce the severity of vEDS.


Asunto(s)
Síndrome de Ehlers-Danlos/genética , Síndrome de Ehlers-Danlos/terapia , Técnicas de Silenciamiento del Gen , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Alelos , Sustitución de Aminoácidos , Secuencia de Bases , Células Cultivadas , Colágeno Tipo III/genética , Síndrome de Ehlers-Danlos/patología , Matriz Extracelular/ultraestructura , Fibroblastos/metabolismo , Fibroblastos/patología , Genes Reporteros , Terapia Genética/métodos , Haploinsuficiencia , Humanos , Luciferasas/genética , Microscopía Inmunoelectrónica , Proteínas Mutantes/genética , Mutación , Mutación Missense , Fenotipo , Medicina de Precisión , Interferencia de ARN , Respuesta de Proteína Desplegada/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...