Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genes Cells ; 29(6): 451-455, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38553254

RESUMEN

The 10th International MDM2 Workshop was held at the National Cancer Center Research Institute (NCCRI) in Tokyo, Japan, from October 15 to 18, 2023. It attracted 166 participants from 12 countries. The meeting featured 52 talks and 41 poster presentations. In the first special session, six invited speakers gave educational and outstanding talks on breakthroughs in MDM2 research. Three keynote speakers presented emerging p53-independent functions of MDM2/MDM4, functional association of MDM2/p53 with cancer immunity, and drug discovery targeting the MDM2/MDM4-p53 pathway. Additionally, 19 invited speakers introduced their new findings. Twenty-one presenters, many of whom were young investigators, postdocs, and students, were selected from submitted abstracts and reported their exciting and unpublished results. For poster presenters, outstanding poster awards were given to the best presenters. There were many inspiring questions and discussions throughout the meeting. Social events like a welcome party, a workshop dinner, and an optional tour enabled further scientific interactions among the participants. The meeting successfully provided an exciting platform for scientific exchange. The experience gained from organizing this meeting will be handed over to the next organizers of the 11th International MDM2 Workshop.


Asunto(s)
Proteínas Proto-Oncogénicas c-mdm2 , Proteína p53 Supresora de Tumor , Animales , Humanos , Asia , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética
2.
J Cell Sci ; 135(15)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35775474

RESUMEN

Accelerated aerobic glycolysis is a distinctive metabolic property of cancer cells that confers dependency on glucose for survival. However, the therapeutic strategies targeting this vulnerability are still inefficient and have unacceptable side effects in clinical trials. Therefore, developing biomarkers to predict therapeutic efficacy would be essential to improve the selective targeting of cancer cells. Here, we found that cell lines that are sensitive to glucose deprivation have high expression of cystine/glutamate antiporter xCT (also known as SLC7A11). We found that cystine uptake and glutamate export through xCT contributed to rapid NADPH depletion under glucose deprivation. This collapse of the redox system oxidized and inactivated AMP-activated protein kinase (AMPK), a major regulator of metabolic adaptation, resulting in a metabolic catastrophe and cell death. Although this phenomenon was prevented by pharmacological or genetic inhibition of xCT, overexpression of xCT sensitized resistant cancer cells to glucose deprivation. Taken together, these findings suggest a novel crosstalk between AMPK and xCT that links metabolism and signal transduction, and reveal a metabolic vulnerability to glucose deprivation in cancer cells expressing high levels of xCT.


Asunto(s)
Cistina , Neoplasias , Proteínas Quinasas Activadas por AMP/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Línea Celular Tumoral , Cistina/metabolismo , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Neoplasias/genética , Oxidación-Reducción
3.
Br J Cancer ; 124(1): 299-312, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33144694

RESUMEN

BACKGROUND: Mutant TP53 interacts with other proteins to produce gain-of-function properties that contribute to cancer metastasis. However, the underlying mechanisms are still not fully understood. METHODS: Using immunoprecipitation and proximity ligation assays, we evaluated breast cancer anti-estrogen resistance 1 (BCAR1) as a novel binding partner of TP53R273H, a TP53 mutant frequently found in human cancers. The biological functions of their binding were examined by the transwell invasion assay. Clinical outcome of patients was analysed based on TP53 status and BCAR1 expression using public database. RESULTS: We discovered a novel interaction between TP53R273H and BCAR1. We found that BCAR1 translocates from the cytoplasm into the nucleus and binds to TP53R273H in a manner dependent on SRC family kinases (SFKs), which are known to enhance metastasis. The expression of full-length TP53R273H, but not the BCAR1 binding-deficient mutant TP53R273HΔ102-207, promoted cancer cell invasion. Furthermore, among the patients with mutant TP53, high BCAR1 expression was associated with a poorer prognosis. CONCLUSIONS: The interaction between TP53R273H and BCAR1 plays an important role in enhancing cancer cell invasion. Thus, our study suggests a disruption of the TP53R273H-BCAR1 binding as a potential therapeutic approach for TP53R273H-harbouring cancer patients.


Asunto(s)
Proteína Sustrato Asociada a CrK/metabolismo , Invasividad Neoplásica/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Humanos , Mutación
5.
J Immunol ; 201(2): 451-464, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29848755

RESUMEN

The ability of cells to induce the appropriate transcriptional response to inflammatory stimuli is crucial for the timely induction of host defense mechanisms. Although a role for tumor suppressor p14ARF (ARF) in the innate immune response was previously demonstrated, the underlying mechanism is still unclear. ARF is a potent upregulator of protein SUMOylation; however, no association of this function with the immune system has been made. In this study, we show the unique role of ARF in IFN-γ-induced immune response using human cell lines. Through a systematic search of proteins SUMOylated by ARF, we identified PIAS1, an inhibitor of IFN-activated transcription factor STAT1, as a novel ARF-binding partner and SUMOylation target. In response to IFN-γ treatment, ARF promoted PIAS1 SUMOylation to inhibit the ability of PIAS1 to attenuate IFN-γ response. Wild-type, but not ARF mutants unable to enhance PIAS1 SUMOylation, prevented the PIAS1-mediated inhibition of IFN-γ response. Conversely, the SUMO-deconjugase SENP1 deSUMOylated PIAS1 to reactivate PIAS1 that was inhibited by ARF. These findings suggest that PIAS1 function is negatively modulated by SUMO modification and that SUMOylation by ARF is required to inhibit PIAS1 activity and restore IFN-γ-induced transcription. In the presence of ARF, in which case PIAS1 is inhibited, depletion of PIAS1 did not have an additive effect on IFN-γ response, suggesting that ARF-mediated enhancement of IFN-γ response is mainly due to PIAS1 inhibition. Our findings reveal a novel function of ARF to inhibit PIAS1 by enhancing SUMOylation to promote the robust induction of IFN-γ response.


Asunto(s)
Inmunidad Innata/inmunología , Interferón gamma/inmunología , Proteínas Inhibidoras de STAT Activados/inmunología , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/inmunología , Sumoilación/inmunología , Proteína p14ARF Supresora de Tumor/inmunología , Línea Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Inflamación/inmunología , Factor de Transcripción STAT1/inmunología , Transcripción Genética/inmunología , Regulación hacia Arriba/inmunología
6.
FASEB J ; 32(7): 3892-3902, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29465311

RESUMEN

Sustained endoplasmic reticulum (ER) stress plays a major role in the development of many metabolic diseases, including cardiovascular disease, nonalcoholic fatty liver disease, insulin resistance, obesity, and diabetes. p32 is a multicompartmental protein involved in the regulation of oxidative phosphorylation and glucose oxidation. p32 ablation is associated with resistance to age-associated and diet-induced obesity through a mechanism that remains largely unknown. Here, we show that p32 promotes lipid biosynthesis by modulating fatty acid-induced ER stress. We found that p32 interacts with endoplasmic reticulum-anchored enzyme mannosyl-oligosaccharide glucosidase I (GCS1), an ER lumen-anchored glucosidase that is essential for the processing of N-linked glycoproteins, and reduces GCS1 in a lysosome-dependent manner. We demonstrate that increased GCS1 expression alleviates fatty acid-induced ER stress and is critical for suppressing ER stress-associated lipogenic gene activation, as demonstrated by the down-regulation of Srebp1, Fasn, and Acc. Consistently, suppression of p32 leads to increased GCS1 expression and alleviates fatty acid-induced ER stress, resulting in reduced lipid accumulation. Thus, p32 and GCS1 are regulators of ER function and lipid homeostasis and are potential therapeutic targets for the treatment of obesity and diabetes.-Liu, Y., Leslie, P. L., Jin, A., Itahana, K., Graves, L. M., Zhang, Y. p32 regulates ER stress and lipid homeostasis by down-regulating GCS1 expression.


Asunto(s)
Estrés del Retículo Endoplásmico , Metabolismo de los Lípidos , Proteínas Mitocondriales/metabolismo , alfa-Glucosidasas/metabolismo , Células 3T3 , Acetil-CoA Carboxilasa/genética , Acetil-CoA Carboxilasa/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Regulación hacia Abajo , Acido Graso Sintasa Tipo I/genética , Acido Graso Sintasa Tipo I/metabolismo , Homeostasis , Humanos , Ratones , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , alfa-Glucosidasas/genética
7.
Int J Mol Sci ; 19(3)2018 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-29518025

RESUMEN

Glucose is the key source for most organisms to provide energy, as well as the key source for metabolites to generate building blocks in cells. The deregulation of glucose homeostasis occurs in various diseases, including the enhanced aerobic glycolysis that is observed in cancers, and insulin resistance in diabetes. Although p53 is thought to suppress tumorigenesis primarily by inducing cell cycle arrest, apoptosis, and senescence in response to stress, the non-canonical functions of p53 in cellular energy homeostasis and metabolism are also emerging as critical factors for tumor suppression. Increasing evidence suggests that p53 plays a significant role in regulating glucose homeostasis. Furthermore, the p53 family members p63 and p73, as well as gain-of-function p53 mutants, are also involved in glucose metabolism. Indeed, how this protein family regulates cellular energy levels is complicated and difficult to disentangle. This review discusses the roles of the p53 family in multiple metabolic processes, such as glycolysis, gluconeogenesis, aerobic respiration, and autophagy. We also discuss how the dysregulation of the p53 family in these processes leads to diseases such as cancer and diabetes. Elucidating the complexities of the p53 family members in glucose homeostasis will improve our understanding of these diseases.


Asunto(s)
Glucosa/metabolismo , Glucólisis , Proteína p53 Supresora de Tumor/metabolismo , Animales , Humanos , Mutación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Tumoral p73/genética , Proteína Tumoral p73/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
8.
Int J Med Sci ; 14(1): 13-17, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28138304

RESUMEN

Globally, morbidity and mortality due to cancer are predicted to increase in both men and women in the coming decades. Furthermore, it is estimated that two thirds of these cancer-related deaths will occur in low-and middle-income countries (LMIC). In addition to morbidity and mortality, cancer also causes an enormous economic burden, especially in developing countries. There are several treatment and management options for cancer including chemotherapy, radiation therapy, surgery, and palliative care. Radiotherapy or radiation therapy (RT) can be an effective treatment, especially for localized or solid cancers; about half of cancer patients receive radiation as a curative or palliative treatment. Because of its low cost, for patients from LMIC with inoperable tumors, RT may be the only option. With the overall increase in the number of cancer patients especially in resource-starved LMIC, the need for more RT facilities further affects the economic growth of those countries. Therefore, an advanced molecular-targeted and more integrated approach involving either RT alone or with surgery and improved cancer drug access worldwide are urgent needs for cancer care.


Asunto(s)
Neoplasias/radioterapia , Radioterapia/métodos , Muerte Celular/efectos de la radiación , Países en Desarrollo , Humanos , Neoplasias/economía , Neoplasias/epidemiología , Radioterapia/economía , Radioterapia/estadística & datos numéricos
9.
Biochem Biophys Res Commun ; 458(1): 104-9, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25637534

RESUMEN

Cells in many organs exist in both proliferating and quiescent states. Proliferating cells are more radio-sensitive, DNA damage pathways including p53 pathway are activated to undergo either G1/S or G2/M arrest to avoid entering S and M phase with DNA damage. On the other hand, quiescent cells are already arrested in G0, therefore there may be fundamental difference of irradiation response between proliferating and quiescent cells, and this difference may affect their radiosensitivity. To understand these differences, proliferating and quiescent human normal lung fibroblasts were exposed to 0.10-1 Gy of γ-radiation. The response of key proteins involved in the cell cycle, cell death, and metabolism as well as histone H2AX phosphorylation were examined. Interestingly, p53 and p53 phosphorylation (Ser-15), as well as the cyclin-dependent kinase inhibitors p21 and p27, were induced similarly in both proliferating and quiescent cells after irradiation. Furthermore, the p53 protein half-life, and expression of cyclin A, cyclin E, proliferating cell nuclear antigen (PCNA), Bax, or cytochrome c expression as well as histone H2AX phosphorylation were comparable after irradiation in both phases of cells. The effect of radioprotection by a glycogen synthase kinase 3ß inhibitor on p53 pathway was also similar between proliferating and quiescent cells. Our results showed that quiescence does not affect irradiation response of key proteins involved in stress and DNA damage at least in normal fibroblasts, providing a better understanding of the radiation response in quiescent cells, which is crucial for tissue repair and regeneration.


Asunto(s)
Fibroblastos/fisiología , Fibroblastos/efectos de la radiación , Pulmón/citología , Proteína p53 Supresora de Tumor/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Fibroblastos/efectos de los fármacos , Rayos gamma , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Semivida , Humanos , Indoles/farmacología , Maleimidas/farmacología , Fosforilación
10.
Cancer Cell ; 12(4): 355-66, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17936560

RESUMEN

It is believed that Mdm2 suppresses p53 in two ways: transcriptional inhibition by direct binding, and degradation via its E3 ligase activity. To study these functions physiologically, we generated mice bearing a single-residue substitution (C462A) abolishing the E3 function without affecting p53 binding. Unexpectedly, homozygous mutant mice died before E7.5, and deletion of p53 rescued the lethality. Furthermore, reintroducing a switchable p53 by crossing with p53ER(TAM) mice surprisingly demonstrated that the mutant Mdm2(C462A) was rapidly degraded in a manner indistinguishable from that of the wild-type Mdm2. Hence, our data indicate that (1) the Mdm2-p53 physical interaction, without Mdm2-mediated p53 ubiquitination, cannot control p53 activity sufficiently to allow early mouse embryonic development, and (2) Mdm2's E3 function is not required for Mdm2 degradation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Mutagénesis Sitio-Dirigida , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Sustitución de Aminoácidos , Animales , Células Cultivadas , Daño del ADN , Regulación hacia Abajo , Embrión de Mamíferos , Fibroblastos/enzimología , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Rayos gamma , Regulación del Desarrollo de la Expresión Génica/efectos de la radiación , Genotipo , Edad Gestacional , Homocigoto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-mdm2/química , Proteínas Proto-Oncogénicas c-mdm2/deficiencia , Proteínas Proto-Oncogénicas c-mdm2/genética , Transcripción Genética/efectos de la radiación , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
11.
Elife ; 132024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39037770

RESUMEN

Bats have unique characteristics compared to other mammals, including increased longevity and higher resistance to cancer and infectious disease. While previous studies have analyzed the metabolic requirements for flight, it is still unclear how bat metabolism supports these unique features, and no study has integrated metabolomics, transcriptomics, and proteomics to characterize bat metabolism. In this work, we performed a multi-omics data analysis using a computational model of metabolic fluxes to identify fundamental differences in central metabolism between primary lung fibroblast cell lines from the black flying fox fruit bat (Pteropus alecto) and human. Bat cells showed higher expression levels of Complex I components of electron transport chain (ETC), but, remarkably, a lower rate of oxygen consumption. Computational modeling interpreted these results as indicating that Complex II activity may be low or reversed, similar to an ischemic state. An ischemic-like state of bats was also supported by decreased levels of central metabolites and increased ratios of succinate to fumarate in bat cells. Ischemic states tend to produce reactive oxygen species (ROS), which would be incompatible with the longevity of bats. However, bat cells had higher antioxidant reservoirs (higher total glutathione and higher ratio of NADPH to NADP) despite higher mitochondrial ROS levels. In addition, bat cells were more resistant to glucose deprivation and had increased resistance to ferroptosis, one of the characteristics of which is oxidative stress. Thus, our studies revealed distinct differences in the ETC regulation and metabolic stress responses between human and bat cells.


Asunto(s)
Quirópteros , Fibroblastos , Quirópteros/metabolismo , Humanos , Fibroblastos/metabolismo , Animales , Metabolómica , Especies Reactivas de Oxígeno/metabolismo , Proteómica/métodos , Línea Celular , Consumo de Oxígeno , Multiómica
12.
Commun Chem ; 7(1): 158, 2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39003409

RESUMEN

Chemotherapy-induced drug resistance remains a major cause of cancer recurrence and patient mortality. ATP binding cassette subfamily B member 1 (ABCB1) transporter overexpression in tumors contributes to resistance, yet current ABCB1 inhibitors have been unsuccessful in clinical trials. To address this challenge, we propose a new strategy using tryptophan as a lead molecule for developing ABCB1 inhibitors. Our idea stems from our studies on bat cells, as bats have low cancer incidences and high ABCB1 expression. We hypothesized that potential ABCB1 substrates in bats could act as competitive inhibitors in humans. By molecular simulations of ABCB1-substrate interactions, we generated a benzylated Cyclo-tryptophan (C3N-Dbn-Trp2) that inhibits ABCB1 activity with efficacy comparable to or better than the classical inhibitor, verapamil. C3N-Dbn-Trp2 restored chemotherapy sensitivity in drug-resistant human cancer cells with no adverse effect on cell proliferation. Our unique approach presents a promising lead toward developing effective ABCB1 inhibitors to treat drug-resistant cancers.

13.
Cell Stress Chaperones ; 24(4): 835-849, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31230214

RESUMEN

Bats, unique among mammals with powered flight, have many species with the longest size-proportionate lifespan of all mammals. Evolutionary adaptations would have been required to survive the elevated body temperatures during flight. Heat shock protein (HSP), highly conserved master regulators of cell stress, expression was examined across tissues and various cell lines in bats. Basal expression level of major HSPs (HSP70 and HSP90) is significantly higher in two different bat species compared to other mammals. This HSP expression could be a bat-unique, key factor to modulate cellular stress and death. Consequently, bat cells survive prolonged heat treatment, along with other stress stimuli, in a HSP-dependent manner, whereas other mammalian cells succumbed. This suggests HSP expression in bats could be an important adaption to intrinsic metabolic stresses like flight and therefore an important model to study stress resilience and longevity in general.


Asunto(s)
Quirópteros/metabolismo , Vuelo Animal/fisiología , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Respuesta al Choque Térmico/fisiología , Longevidad/fisiología , Estrés Oxidativo/fisiología , Adaptación Fisiológica/fisiología , Animales , Línea Celular , Humanos
14.
Nat Commun ; 10(1): 2820, 2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31249297

RESUMEN

Bats are unusual mammals, with the ability to fly, and long lifespans. In addition, bats have a low incidence of cancer, but the mechanisms underlying this phenomenon remain elusive. Here we discovered that bat cells are more resistant than human and mouse cells to DNA damage induced by genotoxic drugs. We found that bat cells accumulate less chemical than human and mouse cells, and efficient drug efflux mediated by the ABC transporter ABCB1 underlies this improved response to genotoxic reagents. Inhibition of ABCB1 triggers an accumulation of doxorubicin, DNA damage, and cell death. ABCB1 is expressed at higher levels in several cell lines and tissues derived from bats compared to humans. Furthermore, increased drug efflux and high expression of ABCB1 are conserved across multiple bat species. Our findings suggest that enhanced efflux protects bat cells from DNA damage induced by genotoxic compounds, which may contribute to their low cancer incidence.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Quirópteros/genética , Quirópteros/metabolismo , Daño del ADN/efectos de los fármacos , Mutágenos/toxicidad , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Doxorrubicina/toxicidad , Humanos , Ratones
15.
Science ; 366(6472): 1486-1492, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31857479

RESUMEN

Disruptions in the ubiquitin protein ligase E3A (UBE3A) gene cause Angelman syndrome (AS). Whereas AS model mice have associated synaptic dysfunction and altered plasticity with abnormal behavior, whether similar or other mechanisms contribute to network hyperactivity and epilepsy susceptibility in AS patients remains unclear. Using human neurons and brain organoids, we demonstrate that UBE3A suppresses neuronal hyperexcitability via ubiquitin-mediated degradation of calcium- and voltage-dependent big potassium (BK) channels. We provide evidence that augmented BK channel activity manifests as increased intrinsic excitability in individual neurons and subsequent network synchronization. BK antagonists normalized neuronal excitability in both human and mouse neurons and ameliorated seizure susceptibility in an AS mouse model. Our findings suggest that BK channelopathy underlies epilepsy in AS and support the use of human cells to model human developmental diseases.


Asunto(s)
Síndrome de Angelman/metabolismo , Canales de Calcio Tipo N/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Síndrome de Angelman/fisiopatología , Animales , Epilepsia/metabolismo , Humanos , Ratones , Modelos Neurológicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Organoides , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico , Convulsiones/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
16.
Leuk Res ; 32(1): 131-41, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17462731

RESUMEN

Inosine monophosphate dehydrogenase (IMPDH) is a pivotal enzyme in the de novo pathway of guanine nucleotide biosynthesis. Inhibitors of this enzyme decrease intracellular guanine nucleotide levels by 50-80% and have potential as anti-neoplastic agents. Both mycophenolic acid (MPA) and AVN-944 are highly specific inhibitors of IMPDH that cause cell cycle arrest or apoptosis in lymphocytes and leukemic cell lines. We have examined the mechanisms by which these two agents cause cytotoxicity. Both MPA and AVN-944 inhibit the growth of K562 cells, and induce apoptosis in Raji B and CCRF-CEM T cells. Both compounds strikingly inhibit RNA synthesis within 2 h of exposure. Depletion of guanine nucleotides by MPA and AVN-944 also causes an early and near-complete reduction in levels of the 45S precursor rRNA synthesis and the concomitant translocation of nucleolar proteins including nucleolin, nucleophosmin, and nucleostemin from the nucleolus to the nucleoplasm. This efflux correlates temporally with the sustained induction of p53 in cell lines with wild-type p53. We conclude that inhibition of IMPDH causes a primary reduction in rRNA synthesis and secondary nucleolar disruption and efflux of nucleolar proteins that most likely mediate cell cycle arrest or apoptosis. The ability of AVN-944 to induce apoptosis in a number of leukemic cell lines supports its potential utility in the treatment of hematologic malignancies.


Asunto(s)
Carbamatos/farmacología , Nucléolo Celular/metabolismo , Nucleótidos de Guanina/metabolismo , IMP Deshidrogenasa/antagonistas & inhibidores , Ácido Micofenólico/farmacología , Compuestos de Fenilurea/farmacología , Precursores del ARN/biosíntesis , ARN Ribosómico/biosíntesis , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica , Genes p53 , Humanos , Células K562 , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Sci Signal ; 11(512)2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29317521

RESUMEN

Cancer cells increase glucose metabolism to support aerobic glycolysis. However, only some cancer cells are acutely sensitive to glucose withdrawal, and the underlying mechanism of this selective sensitivity is unclear. We showed that glucose deprivation initiates a cell death pathway in cancer cells that is dependent on the kinase RIPK1. Glucose withdrawal triggered rapid plasma membrane depolarization and an influx of extracellular calcium into the cell through the L-type calcium channel Cav1.3 (CACNA1D), followed by activation of the kinase CAMK1. CAMK1 and the demethylase PPME1 were required for the subsequent demethylation and inactivation of the catalytic subunit of the phosphatase PP2A (PP2Ac) and the phosphorylation of RIPK1. Plasma membrane depolarization, PP2Ac demethylation, and cell death were prevented by glucose and, unexpectedly, by its nonmetabolizable analog 2-deoxy-d-glucose (2-DG), a glycolytic inhibitor. These findings reveal a previously unknown function of glucose as a signaling molecule that protects cells from death induced by plasma membrane depolarization, independently of its role in glycolysis. Components of this cancer cell death pathway represent potential therapeutic targets against cancer.


Asunto(s)
Calcio/metabolismo , Muerte Celular , Desmetilación , Glucosa/metabolismo , Glucólisis , Neoplasias/patología , Proteína Fosfatasa 2/metabolismo , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 1 Dependiente de Calcio Calmodulina/metabolismo , Humanos , Neoplasias/metabolismo , Fosforilación , Proteína Fosfatasa 2/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Células Tumorales Cultivadas
18.
Mol Cell Biol ; 24(17): 7669-80, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15314174

RESUMEN

The importance of coordinating cell growth with proliferation has been recognized for a long time. The molecular basis of this relationship, however, is poorly understood. Here we show that the ribosomal protein L23 interacts with HDM2. The interaction involves the central acidic domain of HDM2 and an N-terminal domain of L23. L23 and L11, another HDM2-interacting ribosomal protein, can simultaneously yet distinctly interact with HDM2 together to form a ternary complex. We show that, when overexpressed, L23 inhibits HDM2-induced p53 polyubiquitination and degradation and causes a p53-dependent cell cycle arrest. On the other hand, knocking down L23 causes nucleolar stress and triggers translocation of B23 from the nucleolus to the nucleoplasm, leading to stabilization and activation of p53. Our data suggest that cells may maintain a steady-state level of L23 during normal growth; alternating the levels of L23 in response to changing growth conditions could impinge on the HDM2-p53 pathway by interrupting the integrity of the nucleolus.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Ribosómicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ciclo Celular/fisiología , Línea Celular , Dactinomicina/metabolismo , Regulación hacia Abajo , Humanos , Sustancias Macromoleculares , Proteínas Nucleares/genética , Unión Proteica , Inhibidores de la Síntesis de la Proteína/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2 , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Ribosómicas/genética , Ubiquitina/metabolismo
19.
Mol Cell Biol ; 23(1): 389-401, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12482990

RESUMEN

The polycomb protein Bmi-1 represses the INK4a locus, which encodes the tumor suppressors p16 and p14(ARF). Here we report that Bmi-1 is downregulated when WI-38 human fibroblasts undergo replicative senescence, but not quiescence, and extends replicative life span when overexpressed. Life span extension by Bmi-1 required the pRb, but not p53, tumor suppressor protein. Deletion analysis showed that the RING finger and helix-turn-helix domains of Bmi-1 were required for life span extension and suppression of p16. Furthermore, a RING finger deletion mutant exhibited dominant negative activity, inducing p16 and premature senescence. Interestingly, presenescent cultures of some, but not all, human fibroblasts contained growth-arrested cells expressing high levels of p16 and apparently arrested by a p53- and telomere-independent mechanism. Bmi-1 selectively extended the life span of these cultures. Low O(2) concentrations had no effect on p16 levels or life span extension by Bmi-1 but reduced expression of the p53 target, p21. We propose that some human fibroblast strains are more sensitive to stress-induced senescence and have both p16-dependent and p53/telomere-dependent pathways of senescence. Our data suggest that Bmi-1 extends the replicative life span of human fibroblasts by suppressing the p16-dependent senescence pathway.


Asunto(s)
Senescencia Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Fibroblastos/fisiología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , División Celular/genética , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Replicación del ADN , Fibroblastos/citología , Secuencias Hélice-Giro-Hélice , Humanos , Mutación , Proteínas Nucleares/genética , Oxígeno/metabolismo , Complejo Represivo Polycomb 1 , Proteínas del Grupo Polycomb , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
20.
Methods Mol Biol ; 371: 21-31, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17634571

RESUMEN

Most normal human cells undergo cellular senescence after accruing a fixed number of cell divisions, or are challenged by a variety of potentially oncogenic stimuli, in culture and most likely in vivo. Cellular senescence is characterized by an irreversible growth arrest and certain altered functions. Senescent cells in culture are identified by their inability to undergo DNA synthesis, a property also shared by quiescent cells. Several years ago, we described a biomarker associated with the senescent phenotype, a senescence associated beta-galactosidase (SA-beta-gal), which is detected by histochemical staining of cells using the artificial substrate X-gal. The presence of the SA-beta-gal biomarker is independent of DNA synthesis and generally distinguishes senescent cells from quiescent cells. The method to detect SA-beta-gal is a convenient, single cell-based assay, which can identify senescent cells even in heterogeneous cell populations and aging tissues, such as skin biopsies from older individuals. Because it is easy to detect, SA-beta-gal is currently a widely used biomarker of senescence. Here we describe a method to detect SA-beta-gal in detail, including some recent modifications.


Asunto(s)
Envejecimiento/metabolismo , Senescencia Celular/fisiología , Fibroblastos/enzimología , Piel/enzimología , beta-Galactosidasa/metabolismo , Biomarcadores/análisis , Biomarcadores/metabolismo , Línea Celular , ADN/biosíntesis , Fibroblastos/citología , Galactósidos/química , Humanos , Indoles/química , beta-Galactosidasa/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA