Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nucleic Acids Res ; 52(12): 7063-7080, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38808662

RESUMEN

Cohesin plays a crucial role in the organization of topologically-associated domains (TADs), which influence gene expression and DNA replication timing. Whether epigenetic regulators may affect TADs via cohesin to mediate DNA replication remains elusive. Here, we discover that the histone demethylase PHF2 associates with RAD21, a core subunit of cohesin, to regulate DNA replication in mouse neural stem cells (NSC). PHF2 loss impairs DNA replication due to the activation of dormant replication origins in NSC. Notably, the PHF2/RAD21 co-bound genomic regions are characterized by CTCF enrichment and epigenomic features that resemble efficient, active replication origins, and can act as boundaries to separate adjacent domains. Accordingly, PHF2 loss weakens TADs and chromatin loops at the co-bound loci due to reduced RAD21 occupancy. The observed topological and DNA replication defects in PHF2 KO NSC support a cohesin-dependent mechanism. Furthermore, we demonstrate that the PHF2/RAD21 complex exerts little effect on gene regulation, and that PHF2's histone-demethylase activity is dispensable for normal DNA replication and proliferation of NSC. We propose that PHF2 may serve as a topological accessory to cohesin for cohesin localization to TADs and chromatin loops, where cohesin represses dormant replication origins directly or indirectly, to sustain DNA replication in NSC.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona , Cohesinas , Replicación del ADN , Proteínas de Unión al ADN , Células-Madre Neurales , Animales , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Ratones , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Cromatina/metabolismo , Origen de Réplica , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Genoma/genética , Factor de Unión a CCCTC/metabolismo , Factor de Unión a CCCTC/genética , Ratones Noqueados
2.
Int J Mol Sci ; 22(19)2021 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-34638761

RESUMEN

Niclosamide is an oral anthelmintic drug, approved for use against tapeworm infections. Recent studies suggest however that niclosamide may have broader clinical applications in cancers, spurring increased interest in the functions and mechanisms of niclosamide. Previously, we reported that niclosamide targets a metabolic vulnerability in p53-deficient tumours, providing a basis for patient stratification and personalised treatment strategies. In the present study, we functionally characterised the contribution of the aniline 4'-NO2 group on niclosamide to its cellular activities. We demonstrated that niclosamide induces genome-wide DNA damage that is mechanistically uncoupled from its antitumour effects mediated through mitochondrial uncoupling. Elimination of the nitro group in ND-Nic analogue significantly reduced γH2AX signals and DNA breaks while preserving its antitumour mechanism mediated through a calcium signalling pathway and arachidonic acid metabolism. Lipidomics profiling further revealed that ND-Nic-treated cells retained a metabolite profile characteristic of niclosamide-treated cells. Notably, quantitative scoring of drug sensitivity suggests that elimination of its nitro group enhanced the target selectivity of niclosamide against p53 deficiency. Importantly, the results also raise concern that niclosamide may impose a pleiotropic genotoxic effect, which limits its clinical efficacy and warrants further investigation into alternative drug analogues that may ameliorate any potential unwanted side effects.


Asunto(s)
Daño del ADN , Mitocondrias/metabolismo , Neoplasias , Niclosamida , Células HCT116 , Humanos , Mitocondrias/patología , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Niclosamida/análogos & derivados , Niclosamida/farmacología
3.
Nat Commun ; 11(1): 2086, 2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32350249

RESUMEN

Gain of function (GOF) DNA binding domain (DBD) mutations of TP53 upregulate chromatin regulatory genes that promote genome-wide histone methylation and acetylation. Here, we therapeutically exploit the oncogenic GOF mechanisms of p53 codon 158 (Arg158) mutation, a DBD mutant found to be prevalent in lung carcinomas. Using high throughput compound screening and combination analyses, we uncover that acetylating mutp53R158G could render cancers susceptible to cisplatin-induced DNA stress. Acetylation of mutp53R158G alters DNA binding motifs and upregulates TRAIP, a RING domain-containing E3 ubiquitin ligase which dephosphorylates IĸB and impedes nuclear translocation of RelA (p65), thus repressing oncogenic nuclear factor kappa-B (NF-ĸB) signaling and inducing apoptosis. Given that this mechanism of cytotoxic vulnerability appears inapt in p53 wild-type (WT) or other hotspot GOF mutp53 cells, our work provides a therapeutic opportunity specific to Arg158-mutp53 tumors utilizing a regimen consisting of DNA-damaging agents and mutp53 acetylators, which is currently being pursued clinically.


Asunto(s)
Codón/genética , Mutación/genética , Neoplasias/genética , Proteína p53 Supresora de Tumor/genética , Acetilación/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Epigénesis Genética/efectos de los fármacos , Mutación con Ganancia de Función/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ácidos Hidroxámicos/farmacología , Ratones SCID , Modelos Biológicos , Proteínas Mutantes/metabolismo , FN-kappa B/metabolismo , Neoplasias/tratamiento farmacológico , Motivos de Nucleótidos/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Unión Proteica/efectos de los fármacos , Isoformas de Proteínas/genética , Sulfonamidas/farmacología , Topotecan/farmacología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Mol Cell Biol ; 11(3): 255-264, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30865230

RESUMEN

Inactivation of the p53 gene is a key driver of tumorigenesis in various cancer cohorts and types. The quest for a successful p53-based therapy that holds the promise of treating more than half of the cancer population has culminated in extensive knowledge about the role and function of p53 and led to new proposed innovative strategies against p53-defective cancers. We will discuss some of these latest studies with a focus on metabolic regulation and DNA damage response and also highlight novel functions of p53 in these pathways that may provide a contemporary rationale for targeting p53 loss in tumors.


Asunto(s)
Neoplasias/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Daño del ADN/genética , Daño del ADN/fisiología , Humanos , Neoplasias/genética , Proteína p53 Supresora de Tumor/genética
5.
Sci Rep ; 8(1): 5236, 2018 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-29568071

RESUMEN

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

6.
Sci Rep ; 7(1): 17367, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29234018

RESUMEN

RIF1 plays a key role in inhibiting DNA end resection and promoting NHEJ mediated DNA double stand break repair in G1. However, whether SUMOlyation may regulate RIF1 functions is still largely unknown. Here, we report that RIF1 is SUMOlyated in response to DNA damage. We identified PIAS4 as the primary SUMO E3 ligase required for the SUMOylation of RIF1 protein. Mammalian cells compromised of PIAS4 expression, show impaired RIF1 SUMOylation and defective for the disassembly of DNA damage responsive RIF1 foci. Mechanistically, we show that PIAS4 knockdown abrogates UHRF1-dependent ubiquitination of RIF1, compromising RIF1 protein turnover. We detected intense RPA foci that colocalize with RIF1 foci in PIAS4 knockdown cells. These data highlight an important role of PIAS4-dependent regulation of RIF1, likely mediated by SUMOylation, in the disassembly of RIF1 DNA damage response (DDR) foci. We propose that unresolved RIF1 protein at sites of DNA damage in PIAS4-depleted cells largely accumulates in S phase, and subsequently leads to DNA double strand breaks. Therefore, PIAS4 promotes genomic stability by regulating the timely removal of RIF1 from sites of DNA damage.


Asunto(s)
Reparación del ADN/fisiología , Inestabilidad Genómica/fisiología , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Sumoilación/fisiología , Proteínas de Unión a Telómeros/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas Inhibidoras de STAT Activados/genética , ARN Interferente Pequeño/metabolismo , Ubiquitina-Proteína Ligasas
7.
Mol Cell Biol ; 37(16)2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28584195

RESUMEN

Sox2 is known to be important for neuron formation, but the precise mechanism through which it activates a neurogenic program and how this differs from its well-established function in self-renewal of stem cells remain elusive. In this study, we identified a highly conserved cyclin-dependent kinase (Cdk) phosphorylation site on serine 39 (S39) in Sox2. In neural stem cells (NSCs), phosphorylation of S39 enhances the ability of Sox2 to negatively regulate neuronal differentiation, while loss of phosphorylation is necessary for chromatin retention of a truncated form of Sox2 generated during neurogenesis. We further demonstrated that nonphosphorylated cleaved Sox2 specifically induces the expression of proneural genes and promotes neurogenic commitment in vivo Our present study sheds light on how the level of Cdk kinase activity directly regulates Sox2 to tip the balance between self-renewal and differentiation in NSCs.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Neurogénesis , Fosfoserina/metabolismo , Factores de Transcripción SOXB1/metabolismo , Secuencia de Aminoácidos , Animales , Diferenciación Celular , ADN/metabolismo , Regulación de la Expresión Génica , Ratones , Modelos Biológicos , Proteínas Mutantes/metabolismo , Células 3T3 NIH , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo , Fosforilación , Unión Proteica , Estabilidad Proteica , Factores de Transcripción SOXB1/química , Serina Proteasas/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-28193768

RESUMEN

The excitement around the entry into the clinic of the first generation of p53-specific drugs has become muted as the hoped-for dramatic clinical responses have not yet been seen. However, these pioneer molecules have become exceptionally powerful tools in the analysis of the p53 pathway and, as a result, a whole spectrum of new interventions are being explored. These include entirely novel and innovative approaches to drug discovery, such as the use of exon-skipping antisense oligonucleotides and T-cell-receptor-based molecules. The extraordinary resources available to the p53 community in terms of reagents, models, and collaborative networks are generating breakthrough approaches to medicines for oncology and also for other diseases in which aberrant p53 signaling plays a role.


Asunto(s)
Neoplasias/diagnóstico , Neoplasias/terapia , Proteína p53 Supresora de Tumor/fisiología , Animales , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Perfilación de la Expresión Génica , Terapia Genética/métodos , Humanos , Ratones , Terapia Molecular Dirigida/métodos , Mutación , Neoplasias/genética , Oligonucleótidos Antisentido/uso terapéutico , Receptores de Antígenos de Linfocitos T , Proteína p53 Supresora de Tumor/genética
9.
Cell Rep ; 15(1): 132-146, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-27052176

RESUMEN

p53 tumor suppressor maintains genomic stability, typically acting through cell-cycle arrest, senescence, and apoptosis. We discovered a function of p53 in preventing conflicts between transcription and replication, independent of its canonical roles. p53 deficiency sensitizes cells to Topoisomerase (Topo) II inhibitors, resulting in DNA damage arising spontaneously during replication. Topoisomerase IIα (TOP2A)-DNA complexes preferentially accumulate in isogenic p53 mutant or knockout cells, reflecting an increased recruitment of TOP2A to regulate DNA topology. We propose that p53 acts to prevent DNA topological stress originating from transcription during the S phase and, therefore, promotes normal replication fork progression. Consequently, replication fork progression is impaired in the absence of p53, which is reversed by transcription inhibition. Pharmacologic inhibition of transcription also attenuates DNA damage and decreases Topo-II-DNA complexes, restoring cell viability in p53-deficient cells. Together, our results demonstrate a function of p53 that may underlie its role in tumor suppression.


Asunto(s)
Replicación del ADN , Inestabilidad Genómica , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Antígenos de Neoplasias/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Células HCT116 , Humanos , Proteínas de Unión a Poli-ADP-Ribosa , Inhibidores de Topoisomerasa/farmacología , Proteína p53 Supresora de Tumor/genética
10.
DNA Repair (Amst) ; 15: 54-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24462468

RESUMEN

DNA double strand breaks (DSBs) are highly toxic to the cells and accumulation of DSBs results in several detrimental effects in various cellular processes which can lead to neurological, immunological and developmental disorders. Failure of the repair of DSBs spurs mutagenesis and is a driver of tumorigenesis, thus underscoring the importance of the accurate repair of DSBs. Two major canonical DSB repair pathways are the non-homologous end joining (NHEJ) and homologous recombination (HR) pathways. 53BP1 and BRCA1 are the key mediator proteins which coordinate with other components of the DNA repair machinery in the NHEJ and HR pathways respectively, and their exclusive recruitment to DNA breaks/ends potentially decides the choice of repair by either NHEJ or HR. Recently, Rap1 interacting factor 1 has been identified as an important component of the DNA repair pathway which acts downstream of the ATM/53BP1 to inhibit the 5'-3' end resection of broken DNA ends, in-turn facilitating NHEJ repair and inhibiting homology directed repair. Rif1 is conserved from yeast to humans but its function has evolved from telomere length regulation in yeast to the maintenance of genome integrity in mammalian cells. Recently its role in the maintenance of genomic integrity has been expanded to include the regulation of chromatin structure, replication timing and intra-S phase checkpoint. We present a summary of these important findings highlighting the various aspects of Rif1 functions and discuss the key implications for genomic integrity.


Asunto(s)
Roturas del ADN de Doble Cadena , Replicación del ADN , Proteínas de Unión a Telómeros/fisiología , Animales , Reparación del ADN por Unión de Extremidades , Inestabilidad Genómica , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Transducción de Señal , Telómero/genética , Proteína 1 de Unión al Supresor Tumoral P53
11.
Discov Med ; 14(77): 263-71, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23114582

RESUMEN

Targeting the p53-MDM2 pathway is regarded as a viable therapeutic strategy and is supported by several preclinical mouse models which show that the restoration of p53 activity leads to tumor regression in vivo. Given that a large proportion of cancers, including hematological malignancies, retain the expression of the wildtype p53 allele, reactivating wildtype p53 in these cancers could lead to selective apoptosis and is regarded as a potential therapeutic strategy. The exploration of inhibitors and peptides targeting the p53-MDM2 pathway led to the discoveries of specific small molecule inhibitors that disrupt the MDM2-mediated inhibition of p53 transcriptional activity and protein stability. Nutlin is one of the specific small molecule that is well tolerated in vivo in mice but has been used in combinations with conventional chemotherapy and radiotherapy, as well as molecularly targeted drugs to further increase its specificity and potency in vivo. We attempt to identify pathways or new targets which when inhibited may synergize with nutlin in its activation of p53 transcriptional activity. Our previous results show that CDK inhibition synergizes with nutlin in p53 activation and p53-dependent apoptosis, converting a cell cycle arrest response to apoptosis. Here, using a siRNA screen against 726 human kinases, we identified several pathways, including the MAP kinase pathway, the sphingosine kinase pathway, and the CDK pathway which may have crosstalk with the p53 pathway. Selective inhibition of these pathways may synergize with nutlin in the induction of p53 transcriptional activity.


Asunto(s)
Neoplasias/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/fisiología , Ciclo Celular/fisiología , Humanos , Proteínas Proto-Oncogénicas c-mdm2/metabolismo
13.
EMBO Rep ; 13(7): 638-44, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22653443

RESUMEN

The invasiveness of tumour cells depends on changes in cell shape, polarity and migration. Mutant p53 induces enhanced tumour metastasis in mice, and human cells overexpressing p53R273H have aberrant polarity and increased invasiveness, demonstrating the 'gain of function' of mutant p53 in carcinogenesis. We hypothesize that p53R273H interacts with mutant p53-specific binding partners that control polarity, migration or invasion. Here we analyze the p53R273H interactome using stable isotope labelling by amino acids in cell culture and quantitative mass spectrometry, and identify at least 15 new potential mutant p53-specific binding partners. The interaction of p53R273H with one of them--nardilysin (NRD1)--promotes an invasive response to heparin binding-epidermal growth factor-like growth factor that is p53R273H-dependant but does not require Rab coupling protein or p63. Advanced proteomics has thus allowed the detection of a new mechanism of p53-driven invasion.


Asunto(s)
Metaloendopeptidasas/metabolismo , Invasividad Neoplásica , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Polaridad Celular , Factor de Crecimiento Epidérmico/metabolismo , Histidina , Espectrometría de Masas/métodos , Ratones , Mutación Missense , Unión Proteica , Proteómica
14.
Discov Med ; 12(63): 107-17, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21878188

RESUMEN

A common step in human cancer is the inactivation of the p53 tumor suppressor pathway. This occurs either by mutations in the coding region of the p53 gene itself, or equally commonly, by inactivation of pathways that are required for p53 to exert its cellular function. Dramatic new results from animal models and the widespread availability of p53 activating small molecules are yielding important new insights into the therapeutic and toxic effects of p53 and how these can be exploited for improving therapy of cancer and other diseases.


Asunto(s)
Neoplasias/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Humanos , Modelos Biológicos , Neoplasias/genética , Proteína p53 Supresora de Tumor/genética
15.
Nat Rev Clin Oncol ; 8(1): 25-37, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20975744

RESUMEN

Mutations in the TP53 gene are a feature of 50% of all reported cancer cases. In the other 50% of cases, the TP53 gene itself is not mutated but the p53 pathway is often partially inactivated. Cancer therapies that target specific mutant genes are proving to be highly active and trials assessing agents that exploit the p53 system are ongoing. Many trials are aimed at stratifying patients on the basis of TP53 status. In another approach, TP53 is delivered as a gene therapy; this is the only currently approved p53-based treatment. The p53 protein is overexpressed in many cancers and p53-based vaccines are undergoing trials. Processed cell-surface p53 is being exploited as a target for protein-drug conjugates, and small-molecule drugs that inhibit the activity of MDM2, the E3 ligase that regulates p53 levels, have been developed by several companies. The first MDM2 inhibitors are being trialed in both hematologic and solid malignancies. Finally, the first agent found to restore the active function of mutant TP53 has just entered the clinic. Here we discuss the basis of these trials and the future of p53-based therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Terapia Genética , Neoplasias/genética , Neoplasias/terapia , Proteína p53 Supresora de Tumor/genética , Ensayos Clínicos como Asunto , Humanos , Mutación/genética , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo
16.
Cold Spring Harb Perspect Biol ; 2(9): a001222, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20463003

RESUMEN

Inactivation of p53 functions is an almost universal feature of human cancer cells. This has spurred a tremendous effort to develop p53 based cancer therapies. Gene therapy using wild-type p53, delivered by adenovirus vectors, is now in widespread use in China. Other biologic approaches include the development of oncolytic viruses designed to replicate and kill only p53 defective cells and also the development of siRNA and antisense RNA's that activate p53 by inhibiting the function of the negative regulators Mdm2, MdmX, and HPV E6. The altered processing of p53 that occurs in tumor cells can elicit T-cell and B-cell responses to p53 that could be effective in eliminating cancer cells and p53 based vaccines are now in clinical trial. A number of small molecules that directly or indirectly activate the p53 response have also reached the clinic, of which the most advanced are the p53 mdm2 interaction inhibitors. Increased understanding of the p53 response is also allowing the development of powerful drug combinations that may increase the selectivity and safety of chemotherapy, by selective protection of normal cells and tissues.


Asunto(s)
Terapia Molecular Dirigida/métodos , Neoplasias/terapia , Proteína p53 Supresora de Tumor/metabolismo , Animales , Vacunas contra el Cáncer/uso terapéutico , Terapia Genética/métodos , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , ARN Interferente Pequeño/administración & dosificación , Proteína p53 Supresora de Tumor/genética
17.
Cell Cycle ; 9(4): 748-54, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20160485

RESUMEN

The p53 protein and its negative regulator the ubiquitin E3 ligase Mdm2 have been shown to be conserved from the T. adhaerens to man. In common with D. melanogaster and C. elegans, there is a single copy of the p53 gene in T. adhaerens, while in the vertebrates three p53-like genes can be found: p53, p63 and p73. The Mdm2 gene is not present within the fully sequenced and highly annotated genomes of C. elegans and D. melanogaster. However, it is present in Placazoanand the presence of multiple distinct p53 genes in the Sea anemone N. vectensis led us to examine the genomes of other phyla for p53 and Mdm2-like genes. We report here the discovery of an Mdm2-like gene and two distinct p53-like genes in the Arachnid Ioxodes scapularis (Northern Deer Tick). The two predicted Deer Tick p53 proteins are much more highly related to the human p53 protein in sequence than are the fruit fly and nematode proteins. One of the Deer Tick genes encodes a p53 protein that is initiated within the DNA binding domain of p53 and shows remarkable homology to the newly described N-terminally truncated delta isoforms of human and zebrafish p53.


Asunto(s)
Arácnidos/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteína p53 Supresora de Tumor/genética , Secuencia de Aminoácidos , Animales , Proteínas de Unión al ADN/química , Evolución Molecular , Humanos , Ratones , Datos de Secuencia Molecular , Placozoa/genética , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-mdm2/química , Alineación de Secuencia , Proteína p53 Supresora de Tumor/química , Pez Cebra
18.
Cell Cycle ; 9(3): 540-7, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20081368

RESUMEN

The p53 protein is the most commonly mutated tumor suppressor gene in man. Understanding of its evolutionary origins have been enhanced by the recent discovery of p53 family genes in the Sea Anemone Nematostella vectensis. This amino acid sequence conservation has been reflected in biological activity since the early p53 proteins, like their human counterparts, are responsible for DNA damage-induced cellular apoptosis, albeit restricted to the germ cell compartment in model organisms such as the nematode and fruit fly. In vertebrates from zebrafish to man the function of p53 is tightly and absolutely constrained by a negative regulator Mdm2. However the Mdm2 gene has not been detected in the genome of the model nematode (C. elegans) and insect (D. melanogaster) species. We have found that the p53 gene and the Mdm2 gene are present in Placozoans, one of the simplest of all free living multi-cellular organisms, implying that both proteins arose much earlier in evolution than previously thought. Detailed sequence analysis shows the exceptional retention of key features of both proteins from man to Placazoan implying that the p53-Mdm2 interaction and its regulation have been conserved from a basal eumetazoan since the pre-cambrian era over 1 billion years ago.


Asunto(s)
Secuencia Conservada , Placozoa/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/química , Proteína p53 Supresora de Tumor/química , Secuencia de Aminoácidos , Animales , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Alineación de Secuencia
19.
Mol Cancer Res ; 5(11): 1133-45, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18025259

RESUMEN

Current chemotherapy focuses on the use of genotoxic drugs that may induce general DNA damage in cancer cells but also high levels of toxicity in normal tissues. Nongenotoxic activation of p53 by targeting specific molecular pathways therefore provides an attractive therapeutic strategy in cancers with wild-type p53. Here, we explored the antitumor potential of cyclin-dependent kinase (CDK) inhibitors in combination with a small molecule inhibitor of p53-murine double minute 2 (MDM2) interaction. We show that low doses of CDK inhibitors roscovitine and DRB synergize with the MDM2 antagonist nutlin-3a in the induction of p53 activity and promote p53-dependent apoptosis in a dose- and time-dependent manner. Statistical measurement of the combination effects shows that the drug combination is additive on the reduction of cell viability and synergistic on inducing apoptosis, a critical end point of cytotoxic drugs. The degree of apoptosis observed 24 to 48 h after drug treatment correlated with the accumulation of p53 protein and concomitant induction of proapoptotic proteins Puma and PIG3. The antiproliferative and cytotoxic effects of this drug combination are validated in a range of tumor-derived cells including melanoma, colon carcinoma, breast adenocarcinoma, and hepatocarcinoma cells. Furthermore, this drug combination does not induce phosphorylation of Ser(15) on p53 and does not induce genotoxic stress in the cell. Given that many cytotoxic drugs rely on their ability to induce apoptosis via DNA damage-mediated activation of p53, the data presented here may provide a new therapeutic approach for the use of CDK inhibitors and MDM2 antagonists in combinatorial drug therapy.


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Imidazoles/farmacología , Neoplasias/enzimología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Purinas/farmacología , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias/patología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Roscovitina , Proteína p53 Supresora de Tumor/metabolismo
20.
Nucleic Acids Res ; 33(12): 3932-41, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16024743

RESUMEN

The product of the gene mutated in Bloom's syndrome, BLM, is a 3'-5' DNA helicase belonging to the highly conserved RecQ family. In addition to a conventional DNA strand separation activity, BLM catalyzes both the disruption of non-B-form DNA, such as G-quadruplexes, and the branch migration of Holliday junctions. Here, we have characterized a new activity for BLM: the promotion of single-stranded DNA (ssDNA) annealing. This activity does not require Mg(2+), is inhibited by ssDNA binding proteins and ATP, and is dependent on DNA length. Through analysis of various truncation mutants of BLM, we show that the C-terminal domain is essential for strand annealing and identify a 60 amino acid stretch of this domain as being important for both ssDNA binding and strand annealing. We present a model in which the ssDNA annealing activity of BLM facilitates its role in the processing of DNA intermediates that arise during repair of damaged replication forks.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , ADN Helicasas/metabolismo , ADN de Cadena Simple/metabolismo , Adenosina Trifosfatasas/química , ADN Helicasas/química , Reparación del ADN , ADN de Cadena Simple/química , Proteínas de Unión al ADN/metabolismo , Modelos Genéticos , Estructura Terciaria de Proteína , RecQ Helicasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA