Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 193
Filtrar
1.
Biochemistry ; 60(19): 1498-1505, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33870698

RESUMEN

Protein oligomerization plays a very important role in many physiological processes. p53 acts as a key tumor suppressor by regulating cell cycle arrest, DNA repair, and apoptosis, and its antitumor activity is regulated by the hetero- and homo-oligomerization of MDMX and MDM2 proteins. So far, some traditional methods have been utilized to study the oligomerization of MDMX and MDM2 in vitro, but they have not clarified some controversial issues or whether the extracellular results can represent the intracellular results. Here, we put forward an in situ method for studying protein homo- and hetero-oligomerization in single living cells by using fluorescence correlation spectroscopy. In this study, MDMX and MDM2 were labeled with fluorescent proteins using lentiviral transfection. Autocorrelation spectroscopy and cross-correlation spectroscopy methods were used to study the oligomerization of MDMX and MDM2 in situ and the effect of regulation of MDMX oligomerization on p53-MDMX interactions in single living cells. We observed the homo- and hetero-oligomerization of MDMX and MDM2 in living cells. Meanwhile, the levels of the homo-oligomers of MDMX and MDM2 were increased due to the lack of hetero-oligomerization. Finally, the binding affinity of MDMX for p53 was improved with an increase in the level of MDMX hetero-oligomerization.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Apoptosis , Proteínas de Ciclo Celular/fisiología , Fluorescencia , Humanos , Hibridación Fluorescente in Situ/métodos , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Análisis de la Célula Individual , Espectrometría de Fluorescencia/métodos , Proteína p53 Supresora de Tumor/metabolismo
2.
Atherosclerosis ; 305: 1-9, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32585463

RESUMEN

BACKGROUND AND AIMS: Murine double minute-2 (MDM2) has been poorly studied in cardiovascular diseases. The aim of the present study was to determine the biological role of MDM2 in inflammation activation and mitochondrial damage in human aortic endothelial cells (HAECs) stimulated with oxidized low-density lipoprotein (ox-LDL). METHODS: The expression of MDM2 in the aortas of atherosclerotic mice was determined. An adenoviral vector for MDM2 overexpression and siRNA for MDM2 downregulation were constructed and used to transfect HAECs. The functional changes in HAECs stimulated by ox-LDL were observed. RESULTS: The protein expression of MDM2 was increased in atherosclerotic mice and ox-LDL-treated HAECs. In addition, ox-LDL-induced mRNA expression and secretion of TNF-α, IL-6 and IL-1ß were significantly decreased by MDM2 downregulation and increased by MDM2 overexpression, and activation of NF-κB and caspase-1 was involved in the activity of MDM2. The ox-LDL-induced mitochondrial damage, indicated as increase in mitochondrial ROS production, decrease in mitochondrial membrane potential and elevation of mitochondrial DNA release, was significantly reversed by MDM2 downregulation and worsened by MDM2 overexpression. The ox-LDL-induced activation of TLR9/NF-κB and NLRP3/caspase-1 pathway was inhibited by MDM2 downregulation and worsened by MDM2 overexpression. The aggravation caused by MDM2 overexpression was abolished by mito-TEMPO. Treatment with mito-TEMPO significantly reduced the increase in mRNA expression and secretion of TNF-α, IL-6 and IL-1ß induced by MDM2 overexpression in ox-LDL treated HAECs. CONCLUSIONS: These findings suggest that MDM2 contributes to ox-LDL-induced inflammation via regulating mitochondrial damage.


Asunto(s)
Células Endoteliales , Lipoproteínas LDL/efectos adversos , Mitocondrias/patología , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Animales , Células Cultivadas , Humanos , Inflamación , Ratones , FN-kappa B , Transfección
3.
Gene ; 748: 144699, 2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-32334023

RESUMEN

Pigs have been increasingly recognized as a relevant model for studying many human diseases. However, functions and regulations of numerous critical molecules involved in human diseases are not well characterized in pigs, including the prominent tumor suppressor p53, a transcription factor involved in various anti-proliferative processes. In this study, we systematically characterized porcine p53 (p-p53) in its transcriptional activity and regulation by the E3 ligase Mdm2, in comparison with that of human p53 (h-p53). p-p53 is highly homologous to h-p53 with the N-terminal region showing relative divergence. p-p53 exhibits a comparable transcriptional activity to that of h-p53 towards a diverse range of known target genes, and is subject to ubiquitination and degradation by both human and porcine Mdm2 (h-/p-Mdm2). Utilization of the h-Mdm2 targeting compound Nutlin-3 and protein RPL11 inhibits the negative effect of p-Mdm2 on p-p53. These results suggest that the transcription activity and regulation of p-p53 is very similar to that of h-p53, and that the developed agents targeting the h-p53 pathway could be used in the study of p53 related processes and diseases in pigs.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Genes p53 , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Animales , Línea Celular , Humanos , Ratones , Porcinos
4.
Sci Rep ; 10(1): 5028, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32193420

RESUMEN

Loss-of-function mutations in the E3 ubiquitin ligase parkin have been implicated in the death of dopaminergic neurons in the substantia nigra, which is the root cause of dopamine deficit in the striatum in Parkinson's disease. Parkin ubiquitinates proteins on mitochondria that lost membrane potential, promoting the elimination of damaged mitochondria. Neuroprotective activity of parkin has been linked to its critical role in the mitochondria maintenance. Here we report a novel regulatory mechanism: another E3 ubiquitin ligase Mdm2 directly binds parkin and enhances its enzymatic activity in vitro and in intact cells. Mdm2 translocates to damaged mitochondria independently of parkin, enhances parkin-dependent ubiquitination of the outer mitochondria membrane protein mitofusin1. Mdm2 facilitates and its knockdown reduces parkin-dependent mitophagy. Thus, ubiquitously expressed Mdm2 might enhance cytoprotective parkin activity. The data suggest that parkin activation by Mdm2 could be targeted to increase its neuroprotective functions, which has implications for anti-parkinsonian therapy.


Asunto(s)
Mitofagia/genética , Mitofagia/fisiología , Fármacos Neuroprotectores , Enfermedad de Parkinson/genética , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Neuronas Dopaminérgicas/patología , GTP Fosfohidrolasas/metabolismo , Células HEK293 , Humanos , Mutación con Pérdida de Función , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Terapia Molecular Dirigida , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/terapia , Unión Proteica , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Ubiquitinación
5.
Cell Signal ; 66: 109435, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31706019

RESUMEN

Overexpression of MDM2 oncoprotein has been detected in a large number of diverse human malignancies and has been shown to play both p53-dependent and p53-independent roles in oncogenesis. Our study was designed to explore the impact of MDM2 overexpression on the levels of various cell cycle regulatory proteins including Aurora kinase-B (AURK-B), CDC25C and CDK1, which are known to promote tumor progression and increase metastatic potential. Our data from human cell cycle RT2 profiler PCR array experiments revealed significant changes in the expression profile of genes that are involved in different phases of cell cycle regulation in LNCaP-MST (MDM2 transfected) prostate cancer cells. Our current study has demonstrated a significant increase in the expression level of AURK-B, CDC25C, Cyclin A2, Cyclin B and CDK1 in LNCaP-MST cells as compared with wild type LNCaP cells that were modulated by MDM2 specific inhibitor Nutlin-3. In fact, the expression levels of the above- mentioned proteins were significantly altered at both mRNA and protein levels after treating the cells with 20 µM Nutlin-3 for 24h. Additionally, the pro-apoptotic proteins including p53, p21, and Bax were elevated with the concomitant decrease in the key anti-apoptotic proteins following MDM2 inhibitor treatment. Also, Nutlin-3 treated cells demonstrated caspase-3 activation was observed with an in-vitro caspase-3 fluorescent assay performed with caspase 3/7 specific DEVD-amc substrate. Our results offer significant evidence towards the effectiveness of MDM2 inhibition in causing cell cycle arrest via blocking the transmission of signals through AURKB-CDK1 axis and inducing apoptosis in LNCaP-MST cancer cells. It is evident from our data that MDM2 overexpression probably is the primary cause for CDK1 up-regulation in the LNCaP-MST cells, which might have occurred possibly through activation of AURK-B. However, further studies in this direction should shed more light on the intracellular mechanisms involved in the regulation of Aurora kinase-B and CDK1 axis in MDM2 positive cancers.


Asunto(s)
Adenocarcinoma/metabolismo , Puntos de Control del Ciclo Celular , Imidazoles/farmacología , Piperazinas/farmacología , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , Apoptosis , Aurora Quinasa B/metabolismo , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Masculino , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/fisiología
6.
Development ; 146(24)2019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31767619

RESUMEN

The respiratory lineage initiates from the specification of NKX2-1+ progenitor cells that ultimately give rise to a vast gas-exchange surface area. How the size of the progenitor pool is determined and whether this directly impacts final lung size remains poorly understood. Here, we show that epithelium-specific inactivation of Mdm2, which encodes an E3 ubiquitin ligase, led to lethality at birth with a striking reduction of lung size to a single vestigial lobe. Intriguingly, this lobe was patterned and contained all the appropriate epithelial cell types. The reduction of size can be traced to the progenitor stage, when p53, a principal MDM2 protein degradation target, was transiently upregulated. This was followed by a brief increase of apoptosis. Inactivation of the p53 gene in the Mdm2 mutant background effectively reversed the lung size phenotype, allowing survival at birth. Together, these findings demonstrate that p53 protein turnover by MDM2 is essential for the survival of respiratory progenitors. Unlike in the liver, in which genetic reduction of progenitors triggered compensation, in the lung, respiratory progenitor number is a key determinant factor for final lung size.


Asunto(s)
Proliferación Celular/genética , Pulmón/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Mucosa Respiratoria/citología , Células Madre/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Recuento de Células , Embrión de Mamíferos , Femenino , Pulmón/citología , Pulmón/embriología , Masculino , Ratones , Ratones Transgénicos , Tamaño de los Órganos/genética , Embarazo , Proteínas Proto-Oncogénicas c-mdm2/genética , Células Madre/citología , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/fisiología
7.
EMBO J ; 38(20): e102096, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31483066

RESUMEN

Engineered p53 mutant mice are valuable tools for delineating p53 functions in tumor suppression and cancer therapy. Here, we have introduced the R178E mutation into the Trp53 gene of mice to specifically ablate the cooperative nature of p53 DNA binding. Trp53R178E mice show no detectable target gene regulation and, at first sight, are largely indistinguishable from Trp53-/- mice. Surprisingly, stabilization of p53R178E in Mdm2-/- mice nevertheless triggers extensive apoptosis, indicative of residual wild-type activities. Although this apoptotic activity suffices to trigger lethality of Trp53R178E ;Mdm2-/- embryos, it proves insufficient for suppression of spontaneous and oncogene-driven tumorigenesis. Trp53R178E mice develop tumors indistinguishably from Trp53-/- mice and tumors retain and even stabilize the p53R178E protein, further attesting to the lack of significant tumor suppressor activity. However, Trp53R178E tumors exhibit remarkably better chemotherapy responses than Trp53-/- ones, resulting in enhanced eradication of p53-mutated tumor cells. Together, this provides genetic proof-of-principle evidence that a p53 mutant can be highly tumorigenic and yet retain apoptotic activity which provides a survival benefit in the context of cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Leucemia Mieloide Aguda/prevención & control , Linfoma/prevención & control , Mutación , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Carcinogénesis/patología , Ciclo Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Linfoma/genética , Linfoma/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Tumorales Cultivadas
8.
PLoS Genet ; 15(9): e1008364, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31557161

RESUMEN

Seizures can induce endoplasmic reticulum (ER) stress, and sustained ER stress contributes to neuronal death after epileptic seizures. Despite the recent debate on whether inhibiting ER stress can reduce neuronal death after seizures, whether and how ER stress impacts neural activity and seizures remain unclear. In this study, we discovered that the acute ER stress response functions to repress neural activity through a protein translation-dependent mechanism. We found that inducing ER stress promotes the expression and distribution of murine double minute-2 (Mdm2) in the nucleus, leading to ubiquitination and down-regulation of the tumor suppressor p53. Reduction of p53 subsequently maintains protein translation, before the onset of translational repression seen during the latter phase of the ER stress response. Disruption of Mdm2 in an Mdm2 conditional knockdown (cKD) mouse model impairs ER stress-induced p53 down-regulation, protein translation, and reduction of neural activity and seizure severity. Importantly, these defects in Mdm2 cKD mice were restored by both pharmacological and genetic inhibition of p53 to mimic the inactivation of p53 seen during ER stress. Altogether, our study uncovered a novel mechanism by which neurons respond to acute ER stress. Further, this mechanism plays a beneficial role in reducing neural activity and seizure severity. These findings caution against inhibition of ER stress as a neuroprotective strategy for seizures, epilepsies, and other pathological conditions associated with excessive neural activity.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Convulsiones/metabolismo , Animales , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Cultivo Primario de Células , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
9.
Oncogene ; 38(24): 4835-4855, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30816344

RESUMEN

Genome instability is a common feature of tumor cells, and the persistent presence of genome instability is a potential mechanism of tumorigenesis. The E3 ubiquitin ligase MDM2 is intimately involved in genome instability, but its mechanisms are unclear. Our data demonstrated that the transcription factor HBP1 is a target of MDM2. MDM2 facilitates HBP1 proteasomal degradation by ubiquitinating HBP1, regardless of p53 status, thus attenuating the transcriptional inhibition of HBP1 in the expression of its target genes, such as the DNA methyltransferase DNMT1 and histone methyltransferase EZH2, which results in global DNA hypermethylation and histone hypermethylation and ultimately genome instability. The repression of HBP1 by MDM2 finally promotes cell growth and tumorigenesis. Next, we thoroughly explored the regulatory mechanism of the MDM2/HBP1 axis in DNA damage repair following ionizing radiation. Our data indicated that MDM2 overexpression-mediated repression of HBP1 delays DNA damage repair and causes cell death in a p53-independent manner. This investigation elucidated the mechanism of how MDM2 promotes genome instability and enhances tumorigenesis in the absence of p53, thus providing a theoretical and experimental basis for targeting MDM2 as a cancer therapy.


Asunto(s)
Inestabilidad Genómica , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Proteínas Represoras/metabolismo , Ubiquitinación , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica/genética , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Masculino , Ratones , Ratones Desnudos , Procesamiento Proteico-Postraduccional/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Ubiquitinación/genética
10.
FASEB J ; 33(2): 2610-2620, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30260703

RESUMEN

Functions of tumor suppressor p53 and its negative regulator mouse double minute 2 homolog (Mdm2) in ovarian granulosa cells remain to be elucidated, and the current study aims at clarifying this issue. Mice with Mdm2 deficiency in ovarian granulosa cells [ Mdm2-loxP/ progesterone receptor ( Pgr)-Cre mice] were infertile as a result of impairment of oocyte maturation, ovulation, and fertilization, and those with Mdm2/p53 double deletion in granulosa cells ( Mdm2-loxP/ p53-loxP/ Pgr-Cre mice) showed normal fertility, suggesting that p53 induction in the ovarian granulosa cells is detrimental to ovarian function by disturbing oocyte quality. Another model of Mdm2 deletion in ovarian granulosa cells ( Mdm2-loxP/ anti-Mullerian hormone type 2 receptor-Cre mice) also showed subfertility as a result of the failure of ovulation and fertilization, indicating critical roles of ovarian Mdm2 in ovulation and fertilization. Mdm2-p53 pathway in cumulus granulosa cells transcriptionally controlled an orphan nuclear receptor steroidogenic factor 1 (SF1), a key regulator of ovarian function. Importantly, MDM2 and SF1 levels in human cumulus granulosa cells were positively associated with the outcome of oocyte maturation and fertilization in patients undergoing infertility treatment. These findings suggest that the Mdm2-p53-SF1 axis in ovarian cumulus granulosa cells directs ovarian function by affecting their neighboring oocyte quality.-Haraguchi, H., Hirota, Y., Saito-Fujita, T., Tanaka, T., Shimizu-Hirota, R., Harada, M., Akaeda, S., Hiraoka, T., Matsuo, M., Matsumoto, L., Hirata, T., Koga, K., Wada-Hiraike, O., Fujii, T., Osuga, Y. Mdm2-p53-SF1 pathway in ovarian granulosa cells directs ovulation and fertilization by conditioning oocyte quality.


Asunto(s)
Fertilización , Células de la Granulosa/fisiología , Oocitos/fisiología , Ovulación , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Factores de Empalme de ARN/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Células Cultivadas , Femenino , Células de la Granulosa/citología , Humanos , Infertilidad Femenina/etiología , Infertilidad Femenina/metabolismo , Infertilidad Femenina/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/citología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Control de Calidad , Factores de Empalme de ARN/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
11.
Cornea ; 37 Suppl 1: S55-S57, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30216333

RESUMEN

Pterygium is a fibrovascular subepithelial growth of degenerative tissue over the limbus. It is a common condition worldwide that is especially prevalent in tropical countries within the "pterygium belt." Its exact etiology remains to be elucidated; however, it is strongly associated with exposure to ultraviolet light. The high expression levels of tumor protein p53 (TP53) observed in laboratory studies of pterygium seem to contradict the fast-growing nature of its clinical behavior, and TP53 mutations have been suggested. We demonstrated that mouse double minute 2 (MDM2), a TP53-binding protein, contributes to the inhibition of TP53 activity in human pterygium. Thus, disruption of the MDM2-TP53 interaction should attenuate human pterygium cell growth. For primary pterygium, treatment is relatively straightforward and involves surgical excision. To minimize the risk of recurrence, many adjunctive therapies are adopted, including antimetabolites, such as mitomycin C and 5-fluorouracil, amniotic membrane, different variations on conjunctival and/or limbal conjunctival grafts, and other medications such as anti-vascular endothelial growth factor. In the future, MDM2 antagonists may help further lower the recurrence rates after the treatment of pterygium.


Asunto(s)
Proteínas Proto-Oncogénicas c-mdm2/fisiología , Pterigion , Proteína 1 de Unión al Supresor Tumoral P53/fisiología , Amnios/trasplante , Terapia Combinada , Conjuntiva/trasplante , Humanos , Mitomicina/uso terapéutico , Pterigion/tratamiento farmacológico , Pterigion/etiología , Pterigion/metabolismo , Pterigion/cirugía
12.
Exp Eye Res ; 175: 142-147, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29932882

RESUMEN

AIMS: To confirm that mouse double minute 2 (MDM2) could inhibit p53 activity in human pterygium. And to show the disruption of MDM2-p53 interaction could reactive the functions of p53 in pterygium. METHOD: Pterygium and corresponding conjunctiva tissues were collected for establishment of primary cell lines. Expression patterns of MDM2 and p53 were detected by immunofluorescence. Protein localization of p53 and MDM2, and transcriptional activity of p53 in both untreated and MDM2 antagonist (Nutlin) treated pterygium cells were quantified. RESULTS: In pterygium, p53 was highly expressed in cytoplasm and slightly expressed in the nuclei. MDM2 was localized in the nuclei. A p53 transcriptional regulated target gene, p21, was not expressed in pterygium tissues, suggesting the p53 transcriptional activity was not active in pterygium. After treatment with Nutlin, increased nuclear localization of p53 (4.05%-80.56%) was observed in pterygium cells along with increasing Nutlin dosages (from 0 to 50 µM, p < 0.001). The expression of p21 was increased after Nutlin treatments in pterygium cells (2.49 folds in 20 µM Nutlin treated cells compared to control treated cells, p = 0.012). CONCLUSION: We discovered a novel mechanism in pterygium whereby MDM2 suppresses p53 transcriptional activity despite abundant p53 in pterygium. Disruption of MDM2-p53 interaction by Nutlin could be a potential treatment for pterygium.


Asunto(s)
Proteínas Proto-Oncogénicas c-mdm2/fisiología , Pterigion/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular , Conjuntiva/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Imidazoles/farmacología , Immunoblotting , Masculino , Persona de Mediana Edad , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores
13.
Mol Cells ; 41(5): 381-389, 2018 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-29665672

RESUMEN

ARF is a tumor suppressor protein that has a pivotal role in the prevention of cancer development through regulating cell proliferation, senescence, and apoptosis. As a factor that induces senescence, the role of ARF as a tumor suppressor is closely linked to the p53-MDM2 axis, which is a key process that restrains tumor formation. Thus, many cancer cells either lack a functional ARF or p53, which enables them to evade cell oncogenic stress-mediated cycle arrest, senescence, or apoptosis. In particular, the ARF gene is a frequent target of genetic and epigenetic alterations including promoter hyper-methylation or gene deletion. However, as many cancer cells still express ARF, pathways that negatively modulate transcriptional or post-translational regulation of ARF could be potentially important means for cancer cells to induce cellular proliferation. These recent findings of regulators affecting ARF protein stability along with its low levels in numerous human cancers indicate the significance of an ARF post-translational mechanism in cancers. Novel findings of regulators stimulating or suppressing ARF function would provide new therapeutic targets to manage cancer- and senescence-related diseases. In this review, we present the current knowledge on the regulation and alterations of ARF expression in human cancers, and indicate the importance of regulators of ARF as a prognostic marker and in potential therapeutic strategies.


Asunto(s)
Transformación Celular Neoplásica/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/fisiología , Proteínas de Neoplasias/fisiología , Proteína p14ARF Supresora de Tumor/fisiología , Apoptosis , División Celular , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Genes p16 , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Proteína p14ARF Supresora de Tumor/deficiencia , Proteína p14ARF Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/fisiología , Ubiquitinación
14.
Sheng Li Xue Bao ; 69(6): 759-766, 2017 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-29270591

RESUMEN

Caveolin-1 (Cav-1), as an important structural protein of caveolae, has been proven to be correlated with several signal transduction pathways. Recent studies have shown that Cav-1 may play a critical role in response to DNA damage in irradiated pancreatic cancer cells. However, it is not known whether down-regulation of Cav-1 is required to enhance the damage of other kinds of human cells exposed to X-radiation. In this study, the role of Cav-1 in Chang liver cell line (CHL) exposed to X-radiation was investigated. Cav-1 knockdown cell line (CHL-CAV7) was stably established by the siRNA plasmids transfection, and Cav-1 expression was suppressed by 60%, compared with that of control group (CHL-C) which was transfected with non-targeting plasmids. Cellular survival ability and the expressions of proteins related to DNA damage and repair were examined by colony formation assay and Western blot, respectively. Down-regulation of Cav-1 expression induced a significant decrease of the survival rate in CHL-CAV7 cells exposed to 8 and 10 Gy X-radiation. Compared with CHL-C cells, CHL-CAV7 cells showed increased γH2AX expression, as well as decreased p-ATM, DNA-dependent protein kinase, catalytic subunit (DNA-PKcs) and p53 protein expressions when treated with X-radiation. Meanwhile, the colocalization of Mdm2 and Cav-1 was decreased in CHL-CAV7 cells compared with that in CHL-C cells. These results suggest that the down-regulation of Cav-1 may aggravate DNA damage of CHL cells through reducing the interaction of Cav-1 and Mdm2, which results in the promotion of p53 degradation.


Asunto(s)
Caveolina 1/fisiología , Daño del ADN , Reparación del ADN , Hepatocitos/efectos de la radiación , Línea Celular , Humanos , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Transducción de Señal/fisiología , Rayos X
15.
J Biol Chem ; 292(52): 21614-21622, 2017 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-29123033

RESUMEN

The MDM2 RING domain harbors E3 ubiquitin ligase activity critical for regulating the degradation of tumor suppressor p53, which controls many cellular pathways. The MDM2 RING domain also is required for an interaction with MDMX. Mice containing a substitution in the MDM2 RING domain, MDM2C462A, disrupting MDM2 E3 function and the MDMX interaction, die during early embryogenesis that can be rescued by p53 deletion. To investigate whether MDM2C462A, which retains p53 binding, has p53-suppressing activity, we generated Mdm2C462A/C462A ;p53ER/- mice, in which we replaced the endogenous p53 alleles with an inducible p53ER/- allele, and compared survival with that of similarly generated Mdm2-/-;p53ER/- mice. Adult Mdm2-null mice died ∼7 days after tamoxifen-induced p53 activation, indicating that in the absence of MDM2, MDMX cannot suppress p53. Surprisingly, Mdm2C462A/C462A ;p53ER/- mice died ∼5 days after tamoxifen injection, suggesting that p53 activity is higher in the presence of MDM2C462A than in the absence of MDM2. Indeed, in MDM2C462A-expressing mouse tissues and embryonic fibroblasts, p53 exhibited higher transcriptional activity than in those expressing no MDM2 or no MDM2 and MDMX. This observation indicated that MDM2C462A not only is unable to suppress p53 but may have gained the ability to enhance p53 activity. We also found that p53 acetylation, a measure of p53 transcriptional activity, was higher in the presence of MDM2C462A than in the absence of MDM2. These results reveal an unexpected role of MDM2C462A in enhancing p53 activity and suggest the possibility that compounds targeting MDM2 RING domain function could produce even more robust p53 activation.


Asunto(s)
Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ratones , Ratones Noqueados , Mutación , Proteínas Nucleares/metabolismo , Unión Proteica , Dominios Proteicos/fisiología , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Dominios RING Finger/fisiología , Activación Transcripcional/fisiología , Proteína p53 Supresora de Tumor/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
16.
JCI Insight ; 2(17)2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28878120

RESUMEN

The oncoprotein Mdm2 is a RING domain-containing E3 ubiquitin ligase that ubiquitinates G protein-coupled receptor kinase 2 (GRK2) and ß-arrestin2, thereby regulating ß-adrenergic receptor (ßAR) signaling and endocytosis. Previous studies showed that cardiac Mdm2 expression is critical for controlling p53-dependent apoptosis during early embryonic development, but the role of Mdm2 in the developed adult heart is unknown. We aimed to identify if Mdm2 affects ßAR signaling and cardiac function in adult mice. Using Mdm2/p53-KO mice, which survive for 9-12 months, we identified a critical and potentially novel role for Mdm2 in the adult mouse heart through its regulation of cardiac ß1AR signaling. While baseline cardiac function was mostly similar in both Mdm2/p53-KO and wild-type (WT) mice, isoproterenol-induced cardiac contractility in Mdm2/p53-KO was significantly blunted compared with WT mice. Isoproterenol increased cAMP in left ventricles of WT but not of Mdm2/p53-KO mice. Additionally, while basal and forskolin-induced calcium handling in isolated Mdm2/p53-KO and WT cardiomyocytes were equivalent, isoproterenol-induced calcium handling in Mdm2/p53-KO was impaired. Mdm2/p53-KO hearts expressed 2-fold more GRK2 than WT. GRK2 polyubiquitination via lysine-48 linkages was significantly reduced in Mdm2/p53-KO hearts. Tamoxifen-inducible cardiomyocyte-specific deletion of Mdm2 in adult mice also led to a significant increase in GRK2, and resulted in severely impaired cardiac function, high mortality, and no detectable ßAR responsiveness. Gene delivery of either Mdm2 or GRK2-CT in vivo using adeno-associated virus 9 (AAV9) effectively rescued ß1AR-induced cardiac contractility in Mdm2/p53-KO. These findings reveal a critical p53-independent physiological role of Mdm2 in adult hearts, namely, regulation of GRK2-mediated desensitization of ßAR signaling.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Contracción Miocárdica/fisiología , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal , Agonistas Adrenérgicos beta/farmacología , Animales , Ecocardiografía , Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Corazón/diagnóstico por imagen , Corazón/fisiología , Hemodinámica/efectos de los fármacos , Isoproterenol/farmacología , Ratones , Ratones Noqueados , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación
17.
Chin J Nat Med ; 15(6): 401-416, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28629530

RESUMEN

Inflammation is recently recognized as one of the hallmarks of human cancer. Chronic inflammatory response plays a critical role in cancer development, progression, metastasis, and resistance to chemotherapy. Conversely, the oncogenic aberrations also generate an inflammatory microenvironment, enabling the development and progression of cancer. The molecular mechanisms of action that are responsible for inflammatory cancer and cancer-associated inflammation are not fully understood due to the complex crosstalk between oncogenic and pro-inflammatory genes. However, molecular mediators that regulate both inflammation and cancer, such as NF-κB and STAT have been considered as promising targets for preventing and treating these diseases. Recent works have further demonstrated an important role of oncogenes (e.g., NFAT1, MDM2) and tumor suppressor genes (e.g., p53) in cancer-related inflammation. Natural products that target these molecular mediators have shown anticancer and anti-inflammatory activities in preclinical and clinical studies. Sesquiterpenoids (STs), a class of novel plant-derived secondary metabolites have attracted great interest in recent years because of their diversity in chemical structures and pharmacological activities. At present, we and other investigators have found that dimeric sesquiterpenoids (DSTs) may exert enhanced activity and binding affinity to molecular targets due to the increased number of alkylating centers and improved conformational flexibility and lipophilicity. Here, we focus our discussion on the activities and mechanisms of action of STs and DSTs in treating inflammation and cancer as well as their structure-activity relationships.


Asunto(s)
Antiinflamatorios/farmacología , Antineoplásicos Fitogénicos/farmacología , Sesquiterpenos/farmacología , Animales , Humanos , Inflamación/tratamiento farmacológico , Inflamación/etiología , FN-kappa B/antagonistas & inhibidores , Factores de Transcripción NFATC/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/etiología , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Sesquiterpenos/química , Relación Estructura-Actividad
18.
Cancer Cell ; 31(1): 79-93, 2017 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-28073006

RESUMEN

Chromosomal instability (CIN) contributes to cancer evolution, intratumor heterogeneity, and drug resistance. CIN is driven by chromosome segregation errors and a tolerance phenotype that permits the propagation of aneuploid genomes. Through genomic analysis of colorectal cancers and cell lines, we find frequent loss of heterozygosity and mutations in BCL9L in aneuploid tumors. BCL9L deficiency promoted tolerance of chromosome missegregation events, propagation of aneuploidy, and genetic heterogeneity in xenograft models likely through modulation of Wnt signaling. We find that BCL9L dysfunction contributes to aneuploidy tolerance in both TP53-WT and mutant cells by reducing basal caspase-2 levels and preventing cleavage of MDM2 and BID. Efforts to exploit aneuploidy tolerance mechanisms and the BCL9L/caspase-2/BID axis may limit cancer diversity and evolution.


Asunto(s)
Aneuploidia , Caspasa 2/fisiología , Neoplasias Colorrectales/genética , Cisteína Endopeptidasas/fisiología , Proteínas de Unión al ADN/fisiología , Factores de Transcripción/fisiología , Anciano , Anciano de 80 o más Años , Animales , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/fisiología , Caspasa 2/análisis , Segregación Cromosómica , Cisteína Endopeptidasas/análisis , Proteínas de Unión al ADN/genética , Células HCT116 , Humanos , Ratones , Persona de Mediana Edad , Mutación , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/fisiología
19.
Leukemia ; 31(1): 213-221, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27256803

RESUMEN

Mutations resulting in constitutive activation of signaling pathways that regulate ribosome biogenesis are among the most common genetic events in acute myeloid leukemia (AML). However, whether ribosome biogenesis presents as a therapeutic target to treat AML remains unexplored. Perturbations in ribosome biogenesis trigger the 5S ribonucleoprotein particle (RNP)-Mdm2-p53 ribosomal stress pathway, and induction of this pathway has been shown to have therapeutic efficacy in Myc-driven lymphoma. In the current study we address the physiological and therapeutic role of the 5S RNP-Mdm2-p53 pathway in AML. By utilizing mice that have defective ribosome biogenesis due to downregulation of ribosomal protein S19 (Rps19), we demonstrate that induction of the 5S RNP-Mdm2-p53 pathway significantly delays the initiation of AML. However, even a severe Rps19 deficiency that normally results in acute bone marrow failure has no consistent efficacy on already established disease. Finally, by using mice that harbor a mutation in the Mdm2 gene disrupting its binding to 5S RNP, we show that loss of the 5S RNP-Mdm2-p53 pathway is dispensable for development of AML. Our study suggests that induction of the 5S RNP-Mdm2-p53 ribosomal stress pathway holds limited potential as a single-agent therapy in the treatment of AML.


Asunto(s)
Leucemia Mieloide Aguda/etiología , Ribosomas/fisiología , Transducción de Señal/fisiología , Animales , Ratones , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Ribonucleoproteínas/metabolismo , Ribonucleoproteínas/fisiología , Proteínas Ribosómicas/deficiencia , Ribosomas/metabolismo , Estrés Fisiológico , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/fisiología
20.
Adv Exp Med Biol ; 856: 205-230, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27671724

RESUMEN

The field of toxicity testing for non-pharmaceutical chemicals is in flux with multiple initiatives in North America and the EU to move away from animal testing to mode-of-action based in vitro assays. In this arena, there are still obstacles to overcome, such as developing appropriate cellular assays, creating pathway-based dose-response models and refining in vitro-in vivo extrapolation (IVIVE) tools. Overall, it is necessary to provide assurances that these new approaches are adequately protective of human and ecological health. Another major challenge for individual scientists and regulatory agencies is developing a cultural willingness to shed old biases developed around animal tests and become more comfortable with mode-of-action based assays in human cells. At present, most initiatives focus on developing in vitro alternatives and assessing how well these alternative methods reproduce past results related to predicting organism level toxicity in intact animals. The path forward requires looking beyond benchmarking against high dose animal studies. We need to develop targeted cellular assays, new cell biology-based extrapolation models for assessing regions of safety for chemical exposures in human populations, and mode-of-action-based approaches which are constructed on an understanding of human biology. Furthermore, it is essential that assay developers have the flexibility to 'validate' against the most appropriate mode-of-action data rather than against apical endpoints in high dose animal studies. This chapter demonstrates the principles of fit-for-purpose assay development using pathway-targeted case studies. The projects include p53-mdm2-mediated DNA-repair, estrogen receptor-mediated cell proliferation and PPARα receptor-mediated liver responses.


Asunto(s)
Pruebas de Toxicidad/métodos , Toxicología , Alternativas a las Pruebas en Animales , Animales , Daño del ADN , Ensayos Analíticos de Alto Rendimiento , Humanos , Técnicas In Vitro , PPAR alfa/fisiología , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Proteína p53 Supresora de Tumor/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...