Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Int J Mol Sci ; 24(17)2023 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-37685889

RESUMEN

Through a unique genomics and drug screening platform with ~800 solid tumor cell lines, we have found a subset of SCLC cell lines are hypersensitive to venetoclax, an FDA-approved inhibitor of BCL-2. SCLC-A (ASCL1 positive) and SCLC-P (POU2F3 positive), which make up almost 80% of SCLC, frequently express high levels of BCL-2. We found that a subset of SCLC-A and SCLC-P showed high BCL-2 expression but were venetoclax-resistant. In addition, most of these SCLC cell lines have TP53 missense mutations, which make a single amino acid change. These mutants not only lose wild-type (WT) p53 tumor suppressor functions, but also acquire novel cancer-promoting activities (oncogenic, gain-of-function). A recent study with oncogenic mutant (Onc)-p53 knock-in mouse models of SCLC suggests gain-of-function activity can attenuate chemotherapeutic efficacy. Based on these observations, we hypothesize that Onc-p53 confers venetoclax resistance and that simultaneous inhibition of BCL-2 and Onc-p53 induces synergistic anticancer activity in a subset of SCLC-A and SCLC-P. We show here that (1) down-regulation of Onc-p53 increases the expression of a BH3-only pro-apoptotic BIM and sensitizes to venetoclax in SCLC-P cells; (2) targeting Onc-p53 by the HSP90 inhibitor, ganetespib, increases BIM expression and sensitizes to venetoclax in SCLC-P and SCLC-A cells. Although there are currently many combination studies for venetoclax proposed, the concept of simultaneous targeting of BCL-2 and Onc-p53 by the combination of venetoclax and HSP90 inhibitors would be a promising approach for SCLC treatment.


Asunto(s)
Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Animales , Ratones , Línea Celular Tumoral , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/genética , Proteína p53 Supresora de Tumor/genética
2.
Hum Cell ; 36(3): 1204-1210, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36737540

RESUMEN

Lung cancer remains the most chronic form of cancer and the leading cause of cancer mortality in the world. Despite significant improvements in the treatment of lung cancer, the current therapeutic interventions are only partially effective, necessitating the continued search for better, novel alternative treatments. Angiogenesis and cancer stem cells play a central role in the initiation and propagation of cancers. Tumor angiogenesis is triggered by an angiogenic switch when pro-angiogenic factors exceed anti-angiogenic components. Although many anti-angiogenic agents are used in cancer treatment, there are therapeutic limitations with significant side effects. In recent years, cannabinoids have been investigated extensively for their potential anti-neoplastic effects. Our previous findings showed that cannabidiol (CBD) could impede tumor growth in mouse models of melanoma and glioblastoma. Importantly, CBD has been suggested to possess anti-angiogenic activity. In this study, we tested, for the first time, inhalant CBD in the treatment of heterotopic lung cancer and whether such potential effects could reduce cancer stem cell numbers and inhibit tumor angiogenesis. We implanted NCI H1437 human lung cancer cells in nude mice and treated the mice with inhalant CBD or placebo. The outcomes were measured by tumor size and imaging, as well as by immunohistochemistry and flow cytometric analysis for CD44, VEGF, and P-selectin. Our findings showed that CBD decreased tumor growth rate and suppressed expression of CD44 and the angiogenic factors VEGF and P-selectin. These results suggest, for the first time, that inhalant CBD can impede lung cancer growth by suppressing CD44 and angiogenesis.


Asunto(s)
Cannabidiol , Neoplasias Pulmonares , Humanos , Animales , Ratones , Cannabidiol/farmacología , Cannabidiol/uso terapéutico , Selectina-P , Factor A de Crecimiento Endotelial Vascular , Ratones Desnudos , Neoplasias Pulmonares/patología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología
3.
Nat Commun ; 12(1): 704, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33514736

RESUMEN

p53 mutations with single amino acid changes in cancer often lead to dominant oncogenic changes. Here, we have developed a mouse model of gain-of-function (GOF) p53-driven lung cancer utilizing conditionally active LSL p53-R172H and LSL K-Ras-G12D knock-in alleles that can be activated by Cre in lung club cells. Mutation of the p53 transactivation domain (TAD) (p53-L25Q/W26S/R172H) eliminating significant transactivation activity resulted in loss of tumorigenicity, demonstrating that transactivation mediated by or dependent on TAD is required for oncogenicity by GOF p53. GOF p53 TAD mutations significantly reduce phosphorylation of nearby p53 serine 20 (S20), which is a target for PLK3 phosphorylation. Knocking out PLK3 attenuated S20 phosphorylation along with transactivation and oncogenicity by GOF p53, indicating that GOF p53 exploits PLK3 to trigger its transactivation capability and exert oncogenic functions. Our data show a mechanistic involvement of PLK3 in mutant p53 pathway of oncogenesis.


Asunto(s)
Carcinogénesis/genética , Neoplasias Pulmonares/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/genética , Animales , Carcinogénesis/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Mutación con Ganancia de Función , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Humanos , Pulmón/patología , Neoplasias Pulmonares/patología , Ratones , Ratones Transgénicos , Fosforilación/genética , Dominios Proteicos/genética , Proteínas Serina-Treonina Quinasas/genética , Serina/metabolismo , Esferoides Celulares , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor
4.
JCI Insight ; 4(20)2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31527309

RESUMEN

Depletion of epithelial cells after lung injury prompts proliferation and epithelial mesenchymal transition (EMT) of progenitor cells, and this repopulates the lost epithelial layer. To investigate the cell proliferative function of human oncoprotein MDM2, we generated mouse models targeting human MDM2 expression in either lung Club or alveolar cells after doxycycline treatment. We report that MDM2 expression in lung Club or alveolar cells activates DNA replication specifically in lung progenitor cells only after chemical- or radiation-induced lung injury, irrespective of their p53 status. Activation of DNA replication by MDM2 triggered by injury leads to proliferation of lung progenitor cells and restoration of the lost epithelial layers. Mouse lung with no Mdm2 allele loses its ability to replicate DNA, whereas loss of 1 Mdm2 allele compromises this function, demonstrating the requirement of endogenous MDM2. We show that the p53-independent ability of MDM2 to activate Akt signaling is essential for initiating DNA replication in lung progenitor cells. Furthermore, MDM2 activates the Notch signaling pathway and expression of EMT markers, indicative of epithelial regeneration. This is the first report to our knowledge demonstrating a direct p53-independent participation of MDM2 in progenitor cell proliferation and epithelial repair after lung injury, distinct from a p53-degrading antiapoptotic effect preventing injury.


Asunto(s)
Replicación del ADN , Lesión Pulmonar/patología , Pulmón/patología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Regeneración/genética , Animales , Proliferación Celular/genética , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Modelos Animales de Enfermedad , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Técnicas de Sustitución del Gen , Humanos , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/efectos de la radiación , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/genética , Ratones , Ratones Noqueados , Naftalenos/toxicidad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética , Receptores Notch/metabolismo , Transducción de Señal/genética , Células Madre/patología , Proteína p53 Supresora de Tumor/genética
5.
J Clin Invest ; 127(5): 1839-1855, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28394262

RESUMEN

Gain-of-function (GOF) p53 mutations are observed frequently in most intractable human cancers and establish dependency for tumor maintenance and progression. While some of the genes induced by GOF p53 have been implicated in more rapid cell proliferation compared with p53-null cancer cells, the mechanism for dependency of tumor growth on mutant p53 is unknown. This report reveals a therapeutically targetable mechanism for GOF p53 dependency. We have shown that GOF p53 increases DNA replication origin firing, stabilizes replication forks, and promotes micronuclei formation, thus facilitating the proliferation of cells with genomic abnormalities. In contrast, absence or depletion of GOF p53 leads to decreased origin firing and a higher frequency of fork collapse in isogenic cells, explaining their poorer proliferation rate. Following genome-wide analyses utilizing ChIP-Seq and RNA-Seq, GOF p53-induced origin firing, micronuclei formation, and fork protection were traced to the ability of GOF p53 to transactivate cyclin A and CHK1. Highlighting the therapeutic potential of CHK1's role in GOF p53 dependency, experiments in cell culture and mouse xenografts demonstrated that inhibition of CHK1 selectively blocked proliferation of cells and tumors expressing GOF p53. Our data suggest the possibility that checkpoint inhibitors could efficiently and selectively target cancers expressing GOF p53 alleles.


Asunto(s)
Puntos de Control del Ciclo Celular , Replicación del ADN , ADN de Neoplasias/biosíntesis , Mutación , Neoplasias Experimentales/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Ciclina A/genética , Ciclina A/metabolismo , ADN de Neoplasias/genética , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Ratones Mutantes , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Proteína p53 Supresora de Tumor/genética
6.
Mol Oncol ; 11(6): 696-711, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28423230

RESUMEN

Gain-of-function (GOF) mutants of p53 upregulate genes implicated in cell proliferation and oncogenesis. Here, we report that GOF p53 induces tumorigenicity through simultaneous activation of key oncogenic pathways including those controlling putative tumor-initiating cell functions. We determined that in cells expressing p53-R273H, GOF p53 simultaneously upregulates genes from multiple signaling pathways by recognizing promoters containing distinct transcription factor (TF) binding sites. Our analytical data support a model in which GOF p53 complexes with two TFs on the promoter-a mediator protein, Med17, and a histone acetyl transferase, activating histone acetylation-and enhances gene expression to signal cell proliferation and oncogenesis. Thus, therapeutic inhibition of one GOF p53-induced pathway would be insufficient to prevent tumor growth as the oncoprotein activates a multitude of parallel pathways. This discovery suggests enormous selection advantage for cancer cells with GOF p53 to induce oncogenic growth, highlighting the problems of cancer therapy.


Asunto(s)
Carcinogénesis/genética , Mutación con Ganancia de Función , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Proteína p53 Supresora de Tumor/genética , Análisis de Varianza , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Histona Acetiltransferasas/genética , Humanos , Neoplasias Pulmonares/patología , Complejo Mediador/genética , Ratones , Ratones Desnudos , Invasividad Neoplásica , Activación Transcripcional/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer Res ; 14(5): 423-36, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26965143

RESUMEN

UNLABELLED: Many mutant p53 proteins exhibit an abnormally long half-life and overall increased abundance compared with wild-type p53 in tumors, contributing to mutant p53's gain-of-function oncogenic properties. Here, a novel mechanism is revealed for the maintenance of mutant p53 abundance in cancer that is dependent on DNA damage checkpoint activation. High-level mutant p53 expression in lung cancer cells was associated with preferential p53 monoubiquitination versus polyubiquitination, suggesting a role for the ubiquitin/proteasome system in regulation of mutant p53 abundance in cancer cells. Interestingly, mutant p53 ubiquitination status was regulated by ataxia-telangectasia mutated (ATM) activation and downstream phosphorylation of mutant p53 (serine 15), both in resting and in genotoxin-treated lung cancer cells. Specifically, either inhibition of ATM with caffeine or mutation of p53 (serine 15 to alanine) restored MDM2-dependent polyubiquitination of otherwise monoubiquitinated mutant p53. Caffeine treatment rescued MDM2-dependent proteasome degradation of mutant p53 in cells exhibiting active DNA damage signaling, and ATM knockdown phenocopied the caffeine effect. Importantly, in cells analyzed individually by flow cytometry, p53 levels were highest in cells exhibiting the greatest levels of DNA damage response, and interference with DNA damage signaling preferentially decreased the relative percentage of cells in a population with the highest levels of mutant p53. These data demonstrate that active DNA damage signaling contributes to high levels of mutant p53 via modulation of ubiquitin/proteasome activity toward p53. IMPLICATION: The ability of DNA damage checkpoint signaling to mediate accumulation of mutant p53 suggests that targeting this signaling pathway may provide therapeutic gain. Mol Cancer Res; 14(5); 423-36. ©2016 AACR.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Neoplasias Pulmonares/genética , Mutación , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Daño del ADN , Regulación Neoplásica de la Expresión Génica , Genes cdc , Semivida , Humanos , Neoplasias Pulmonares/metabolismo , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Serina/metabolismo , Ubiquitinación
8.
Oncotarget ; 7(11): 12426-46, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26820293

RESUMEN

Human lung cancers harboring gain-of-function (GOF) p53 alleles express higher levels of the epidermal growth factor receptor (EGFR). We demonstrate that a number of GOF p53 alleles directly upregulate EGFR. Knock-down of p53 in lung cancer cells lowers EGFR expression and reduces tumorigenicity and other GOF p53 properties. However, addiction of lung cancer cells to GOF p53 can be compensated by overexpressing EGFR, suggesting that EGFR plays a critical role in addiction. Chromatin immunoprecipitation (ChIP) using lung cancer cells expressing GOF p53 alleles showed that GOF p53 localized to the EGFR promoter. The sequence where GOF p53 is found to interact by ChIP seq can act as a GOF p53 response element. The presence of GOF p53 on the EGFR promoter increased histone H3 acetylation, indicating a mechanism whereby GOF p53 enhances chromatin opening for improved access to transcription factors (TFs). ChIP and ChIP-re-ChIP with p53, Sp1 and CBP histone acetylase (HAT) antibodies revealed docking of GOF p53 on Sp1, leading to increased binding of Sp1 and CBP to the EGFR promoter. Up-regulation of EGFR can occur via GOF p53 contact at other novel sites in the EGFR promoter even when TAD-I is inactivated; these sites are used by both intact and TAD-I mutated GOF p53 and might reflect redundancy in GOF p53 mechanisms for EGFR transactivation. Thus, the oncogenic action of GOF p53 in lung cancer is highly dependent on transactivation of the EGFR promoter via a novel transcriptional mechanism involving coordinated interactions of TFs, HATs and GOF p53.


Asunto(s)
Receptores ErbB/genética , Neoplasias Pulmonares/genética , Proteína p53 Supresora de Tumor/genética , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Ratones SCID , Regiones Promotoras Genéticas , Factor de Transcripción Sp1/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
9.
Subcell Biochem ; 85: 71-90, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25201189

RESUMEN

p53 is a tumor suppressor protein whose key function is to maintain the integrity of the cell. Mutations in p53 have been found in up to 50 % of all human cancers and cause an increase in oncogenic phenotypes such as proliferation and tumorigenicity. Both wild-type and mutant p53 have been shown to transactivate their target genes, either through directly binding to DNA, or indirectly through protein-protein interactions. This review discusses possible mechanisms behind both wild-type and mutant p53-mediated transactivation and touches on the concept of addiction to mutant p53 of cancer cells and how that may be used for future therapies.


Asunto(s)
Genes p53 , Mutación , Transcripción Genética/genética , Animales , Humanos , Ratones , Modelos Animales , Neoplasias/genética , Oncogenes
10.
Subcell Biochem ; 85: 215-34, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25201197

RESUMEN

Frequent overexpression of MDM2 in human cancers suggests that the protein confers a survival advantage to cancer cells. However, overexpression of MDM2 in normal cells seems to restrict cell proliferation. This review discusses the cell growth regulatory functions of MDM2 in normal and genetically defective cells to assess how cancer cells evade the growth-restricting consequence of MDM2 overexpression. Similar to oncoproteins that induce a DNA damage response and oncogene induced senescence in non-transformed cells, MDM2 induces G1-arrest and intra-S phase checkpoint responses that control untimely DNA replication in the face of genetic challenges.


Asunto(s)
Proliferación Celular , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transducción de Señal , Animales , Ciclo Celular , Ciclinas/metabolismo , Replicación del ADN , Humanos , Ratones , Ratones Noqueados , Neoplasias/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética
11.
Subcell Biochem ; 85: 359-70, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25201204

RESUMEN

Over the past few decades, advances in cancer research have enabled us to understand the different mechanisms that contribute to the aberrant proliferation of normal cells into abnormal cells that result in tumors. In the pursuit to find cures, researchers have primarily focused on various molecular level changes that are unique to cancerous cells. In humans, about 50 % or more cancers have a mutated tumor suppressor p53 gene thereby resulting in accumulation of p53 protein and losing its function to activate the target genes that regulate cell cycle and apoptosis. Extensive research conducted in murine cancer models with activated p53, loss of p53, or p53 missense mutations have facilitated researchers to understand the role of this key protein. Despite the identification of numerous triggers that causes lung cancer specific cure still remain elusive. One of the primary reasons attributed to this is due to the fact that the tumor tissue is heterogeneous and contains numerous sub-populations of cells. Studies have shown that a specific sub-population of cells termed as cancer stem cells (CSCs) drive the recurrence of cancer in response to standard chemotherapy. These CSCs are mutated cells with core properties similar to those of adult stem cells. They reside in a microenvironment within the tumor tissue that supports their growth and make them less susceptible to drug treatment. These cells possess properties of symmetric self-renewal and migration thus driving tumor formation and metastasis. Therefore, research specifically targeting these cells has gained prominence towards developing new therapeutic agents against cancer. This chapter focuses on lung cancer stem cells, p53 mutations noted in these cells, and importance of MDM2 interactions. Further, research approaches for better understanding of molecular mechanisms that drive CSC function and developing appropriate therapies are discussed.


Asunto(s)
Genes p53 , Neoplasias Pulmonares/patología , Mutación , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-mdm2/genética , Humanos , Neoplasias Pulmonares/genética
12.
Oncotarget ; 5(2): 417-27, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24481480

RESUMEN

Gain-of-function mutant p53 is thought to induce gene expression in part by binding transcription factors bound to promoters for genes that mediate oncogenesis. We investigated the mechanism of mutant p53 binding by mapping the human genomic binding sites for p53 R273H using ChIP-Seq and showed them to localize to ETS DNA sequence motifs and locations with ETS1 and GABPA binding, both within promoters and distal to promoters. Strikingly, p53 R273H showed statistically significant and substantial binding to bidirectional promoters, which are enriched for inverted repeated ETS DNA sequence motifs. p53 R273H exhibited an exponential increase in probability of binding promoters with a higher number of ETS motifs. Both ETS1 and GABPA also showed an increase in the probability of binding to promoters with a higher number of ETS motifs. However, despite this increase in probability of binding by p53 R273H and ETS1, there was no increase in the binding signal, suggesting that the number of ETS1 and p53 R273H proteins bound per promoter is being limited. In contrast, GABPA did exhibit an increase in binding signal with higher numbers of ETS motifs per promoter. Analysis of the distance between inverted pairs of ETS motifs within promoters and binding by p53 R273H, ETS1 and GABPA, showed a novel coordination of binding for the three proteins. Both ETS1 and p53 R273H exhibited preference for binding promoters with distantly spaced ETS motifs in face-to-face and back-to-back orientations, and low binding preference to promoters with closely spaced ETS motifs. GABPA exhibited the inverse pattern of binding by preferring to bind promoters with closely spaced ETS motifs. Analysis of the helical phase between ETS motifs showed that ETS1 and p53 R273H exhibited a low preference for binding promoters with ETS motifs on the same face of the DNA helix. We propose a model for the binding of ETS1 and p53 R273H in which two inverted ETS motifs on a looped DNA helix are juxtaposed for ETS1 binding as a homodimer, with p53 R273H bound to ETS1. We propose that the formation of this DNA loop and protein-bound complex prevents additional binding of ETS1 and p53 R273H proteins to other proximal binding sites.


Asunto(s)
Factor de Transcripción de la Proteína de Unión a GA/metabolismo , Neoplasias Pulmonares/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Sitios de Unión , Línea Celular Tumoral , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Factor de Transcripción de la Proteína de Unión a GA/genética , Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Regiones Promotoras Genéticas , Proteína Proto-Oncogénica c-ets-1/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/genética
13.
Nucleic Acids Res ; 42(2): 926-40, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24163099

RESUMEN

Conventional paradigm ascribes the cell proliferative function of the human oncoprotein mouse double minute2 (MDM2) primarily to its ability to degrade p53. Here we report that in the absence of p53, MDM2 induces replication stress eliciting an early S-phase checkpoint response to inhibit further firing of DNA replication origins. Partially synchronized lung cells cultured from p53-/-:MDM2 transgenic mice enter S phase and induce S-phase checkpoint response earlier than lung cells from p53-/- mice and inhibit firing of DNA replication origins. MDM2 activates chk1 phosphorylation, elevates mixed lineage lymphoma histone methyl transferase levels and promotes checkpoint-dependent tri-methylation of histone H3 at lysine 4, known to prevent firing of late replication origins at the early S phase. In the absence of p53, a condition that disables inhibition of cyclin A expression by MDM2, MDM2 increases expression of cyclin D2 and A and hastens S-phase entry of cells. Consistently, inhibition of cyclin-dependent kinases, known to activate DNA replication origins during firing, inhibits MDM2-mediated induction of chk1 phosphorylation indicating the requirement of this activity in MDM2-mediated chk1 phosphorylation. Our data reveal a novel pathway, defended by the intra-S-phase checkpoint, by which MDM2 induces unscheduled origin firing and accelerates S-phase entry of cells in the absence of p53.


Asunto(s)
Replicación del ADN , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Origen de Réplica , Puntos de Control de la Fase S del Ciclo Celular/genética , Animales , Cafeína/farmacología , Línea Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Ciclina A/metabolismo , Ciclina D2/metabolismo , Genes p53 , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética , Origen de Réplica/efectos de los fármacos , Estrés Fisiológico/genética
14.
Clin Cancer Res ; 19(12): 3189-200, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23620409

RESUMEN

PURPOSE: Glioblastoma multiforme (GBM) is the most lethal form of brain cancer with a median survival of only 12 to 15 months. Current standard treatment consists of surgery followed by chemoradiation. The poor survival of patients with GBM is due to aggressive tumor invasiveness, an inability to remove all tumor tissue, and an innate tumor chemo- and radioresistance. Ataxia-telangiectasia mutated (ATM) is an excellent target for radiosensitizing GBM because of its critical role in regulating the DNA damage response and p53, among other cellular processes. As a first step toward this goal, we recently showed that the novel ATM kinase inhibitor KU-60019 reduced migration, invasion, and growth, and potently radiosensitized human glioma cells in vitro. EXPERIMENTAL DESIGN: Using orthotopic xenograft models of GBM, we now show that KU-60019 is also an effective radiosensitizer in vivo. Human glioma cells expressing reporter genes for monitoring tumor growth and dispersal were grown intracranially, and KU-60019 was administered intratumorally by convection-enhanced delivery or osmotic pump. RESULTS: Our results show that the combined effect of KU-60019 and radiation significantly increased survival of mice 2- to 3-fold over controls. Importantly, we show that glioma with mutant p53 is much more sensitive to KU-60019 radiosensitization than genetically matched wild-type glioma. CONCLUSIONS: Taken together, our results suggest that an ATM kinase inhibitor may be an effective radiosensitizer and adjuvant therapy for patients with mutant p53 brain cancers.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Neoplasias Encefálicas/terapia , Glioma/terapia , Morfolinas/administración & dosificación , Tioxantenos/administración & dosificación , Animales , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/patología , Humanos , Ratones , Mutación , Tolerancia a Radiación/efectos de los fármacos , Radiación Ionizante , Proteína p53 Supresora de Tumor/genética
15.
Methods Mol Biol ; 962: 85-94, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23150439

RESUMEN

Protein-protein interaction studies can provide valuable insight into protein function. One of the most practical and high-yielding approaches is immunoprecipitation of a bait protein followed by mass spectrometry to identify co-precipitating proteins. Here we describe an effective and simplified version of this method that can be performed in most laboratories using standard laboratory equipment (apart from the mass spectrometer). We further demonstrate the utility of this method to identify proteins that specifically interact with mutant forms of the tumor suppressor protein, p53.


Asunto(s)
Proteínas Mutantes/aislamiento & purificación , Proteómica/métodos , Proteína p53 Supresora de Tumor/aislamiento & purificación , Western Blotting , Línea Celular Tumoral , Electroforesis en Gel de Poliacrilamida , Humanos , Inmunoprecipitación , Espectrometría de Masas , Proteínas Mutantes/genética , Mutación , Transfección , Proteína p53 Supresora de Tumor/genética
16.
Methods Mol Biol ; 962: 127-33, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23150442

RESUMEN

Chemoresistance and increased growth rate are two gain-of-function functions that mutant p53 is thought to possess. Here, we describe two methods for measuring the sensitiveness of cells to chemotherapeutic drugs and the rate of cell growth. Both of which can be used with a wide range of cell types. The clonogenic assay can be used with many different chemotoxic drugs and the growth assay described here presents an alternative to the MTT assay and allows for a long-term measurement of cell growth. These protocols are both easy, flexible, require relatively little effort, and are inexpensive to carry out.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Proteína p53 Supresora de Tumor/metabolismo , Antineoplásicos/farmacología , Apoptosis , Ciclo Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias/métodos , Humanos , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
17.
Methods Mol Biol ; 962: 135-46, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23150443

RESUMEN

Pro-oncogenic properties of mutant p53 were investigated with the aid of migration assays, adhesion assays, and soft agar growth assays using cells stably expressing gain-of-function p53 mutants. To determine cell migration, "wound-healing" (scratch) assays and haptotactic (chamber) assays were used. H1299 cells expressing mutant p53 were found to migrate more rapidly than cells transfected with empty vector alone. Results from both types of migration assay were broadly similar. Migratory ability differed for different p53 mutants, suggesting allele-specific effects. Cells expressing p53 mutants also showed enhanced adhesion to extracellular matrix compare to controls. Furthermore, stable transfection of mutant p53-H179L into NIH3T3 fibroblasts was sufficient to allow anchorage-independent growth in soft agar.


Asunto(s)
Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Proteínas Mutantes/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Alelos , Animales , Adhesión Celular , Línea Celular Tumoral , Fibroblastos/metabolismo , Humanos , Ratones , Proteínas Mutantes/genética , Mutación , Células 3T3 NIH , Transfección , Proteína p53 Supresora de Tumor/genética , Técnicas de Cierre de Heridas
18.
Methods Mol Biol ; 962: 147-55, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23150444

RESUMEN

DNA replication involves a coordinated progression through S phase, and disruption of these regulated steps may cause gene abnormalities, which may lead to cancer. Different stages of DNA replication can be detected immunofluorescently that would indicate how replication is progressing in a cell population or under specific conditions. We describe a method for labeling replicating DNA with two nucleotide analogs, and then detecting the sequential patterns of incorporation using fluorescently labeled antibodies on DNA spread onto a glass slide. Quantification of the different types of replication patterns produced by this method reveals how replication is achieved under different conditions by the predominance and lengths of elongating replication forks progressing from single or clustered origins, as well as the sites of termination from two converging forks.


Asunto(s)
Replicación del ADN , Proteínas Mutantes/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , ADN/genética , Humanos , Proteínas Mutantes/genética , Fase S , Proteína p53 Supresora de Tumor/genética
19.
Methods Mol Biol ; 962: 193-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23150448

RESUMEN

In order to study the functions of a cell's endogenous mutant p53, the p53 protein levels must be knocked-down. Transient transfection of small interfering RNAs is one way to accomplish this. Another is the stable expression of short hairpin RNAs. This chapter presents a method by which a short hairpin RNA (shRNA) targeting p53 is inserted into the genome of a cell via lentivirus infection. These p53 knock-down cell lines are stable and may be grown long term for use in a wide range of applications.


Asunto(s)
Técnicas de Silenciamiento del Gen/métodos , Proteína p53 Supresora de Tumor/genética , ADN Viral/genética , Vectores Genéticos , Células HEK293 , Humanos , Lentivirus , Proteínas Mutantes/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Transfección
20.
Methods Mol Biol ; 962: 227-36, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23150451

RESUMEN

Mutant p53 may activate target genes through the interaction of transcription factors or through histone modifications. Chromatin immunoprecipitation (ChIP) is a method commonly used to study these types of protein interactions. In order to generate a list of target genes that may be activated through this mechanism, ChIP sequencing may be used. ChIP sequencing involves the mass parallel sequencing of ChIP DNA fragments. We describe a method by which to prepare chromatin immunoprecipitation sequencing libraries and how to analyze sequencing data. In this procedure, prepared libraries have been sent to a core facility. The results have been verified using quantitative PCR.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , ADN/aislamiento & purificación , Técnicas Analíticas Microfluídicas/métodos , Proteína p53 Supresora de Tumor/genética , Línea Celular Tumoral , Biología Computacional , ADN/genética , Fragmentación del ADN , Genoma Humano , Humanos , Reacción en Cadena de la Polimerasa/métodos , Análisis de Secuencia de ADN/métodos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA