Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
2.
Mol Cell ; 70(5): 949-960.e4, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29861159

RESUMEN

The mammalian Target of Rapamycin Complex 1 (mTORC1)-signaling system plays a critical role in the maintenance of cellular homeostasis by sensing and integrating multiple extracellular and intracellular cues. Therefore, uncovering the effectors of mTORC1 signaling is pivotal to understanding its pathophysiological effects. Here we report that the transcription factor forkhead/winged helix family k1 (Foxk1) is a mediator of mTORC1-regulated gene expression. Surprisingly, Foxk1 phosphorylation is increased upon mTORC1 suppression, which elicits a 14-3-3 interaction, a reduction of DNA binding, and nuclear exclusion. Mechanistically, this occurs by mTORC1-dependent suppression of nuclear signaling by the Foxk1 kinase, Gsk3. This pathway then regulates the expression of multiple genes associated with glycolysis and downstream anabolic pathways directly modulated by Foxk1 and/or by Foxk1-regulated expression of Hif-1α. Thus, Foxk1 mediates mTORC1-driven metabolic rewiring, and it is likely to be critical for metabolic diseases where improper mTORC1 signaling plays an important role.


Asunto(s)
Reprogramación Celular , Metabolismo Energético , Factores de Transcripción Forkhead/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas 14-3-3/metabolismo , Transporte Activo de Núcleo Celular , Animales , Sitios de Unión , Proliferación Celular , Regulación hacia Abajo , Factores de Transcripción Forkhead/genética , Glucógeno Sintasa Quinasa 3/genética , Células HEK293 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Fosforilación , Unión Proteica , Transducción de Señal
3.
Mol Cell ; 67(3): 512-527.e4, 2017 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-28757207

RESUMEN

Aberrant signaling by the mammalian target of rapamycin (mTOR) contributes to the devastating features of cancer cells. Thus, mTOR is a critical therapeutic target and catalytic inhibitors are being investigated as anti-cancer drugs. Although mTOR inhibitors initially block cell proliferation, cell viability and migration in some cancer cells are quickly restored. Despite sustained inhibition of mTORC1/2 signaling, Akt, a kinase regulating cell survival and migration, regains phosphorylation at its regulatory sites. Mechanistically, mTORC1/2 inhibition promotes reorganization of integrin/focal adhesion kinase-mediated adhesomes, induction of IGFR/IR-dependent PI3K activation, and Akt phosphorylation via an integrin/FAK/IGFR-dependent process. This resistance mechanism contributes to xenograft tumor cell growth, which is prevented with mTOR plus IGFR inhibitors, supporting this combination as a therapeutic approach for cancers.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Quinasa 1 de Adhesión Focal/metabolismo , Melanoma/tratamiento farmacológico , Complejos Multiproteicos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Somatomedina/antagonistas & inhibidores , Neoplasias Cutáneas/tratamiento farmacológico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Quinasa 1 de Adhesión Focal/genética , Humanos , Integrina alfa2/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Melanoma/enzimología , Melanoma/patología , Ratones Desnudos , Complejos Multiproteicos/metabolismo , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Receptor IGF Tipo 1 , Receptores de Somatomedina/genética , Receptores de Somatomedina/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo , Transfección , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Liver Transpl ; 29(8): 804-812, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37029084

RESUMEN

Total plasma exchange (TPE) can play a role in cancer treatment by eliminating immune checkpoint inhibitors. This study investigated whether TPE improved oncological outcomes in patients with HCC who underwent ABO-incompatible living donor liver transplantation (LT). The study included 152 patients who underwent ABO-incompatible living donor LT for HCC between 2010 and 2021 at Samsung Medical Center. Overall survival was analyzed using the Kaplan-Meier curve, whereas HCC-specific recurrence-free survival (RFS) was analyzed using the cumulative incidence curve after propensity score matching. Cox regression and competing risks subdistribution hazard models were used to identify the risk factors associated with overall survival and HCC-specific RFS, respectively. The propensity score matching resulted in 54 matched pairs, grouped according to whether they underwent postoperative TPE [post-transplant TPE(+)] or not [post-transplant TPE(-)]. The 5-year HCC-specific RFS cumulative incidence was superior in the post-transplant TPE (+) group [12.5% (95% CI: 3.1%-21.9%)] compared with the post-transplant TPE(-) group [38.1% (95% CI: 24.4%-51.8%), p = 0.005]. In subgroup analysis for patients with microvascular invasion and those beyond the Milan criteria, the post-transplant TPE(+) group showed significantly superior HCC-specific survival. The multivariable analysis also showed that postoperative TPE had a protective effect on HCC-specific RFS (HR = 0.26, 95% CI: 0.10-0.64, p = 0.004) and that the more post-transplant TPE was performed, the better RFS was observed (HR = 0.71, 95% CI: 0.55-0.93, p = 0.012). Post-transplant TPE was found to improve RFS after ABO-incompatible living donor LT for HCC, particularly in advanced cases with microvascular invasion and beyond Milan criteria. These findings suggest that TPE may have a potential role in improving oncological outcomes in patients with HCC undergoing LT.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Trasplante de Hígado , Humanos , Carcinoma Hepatocelular/cirugía , Neoplasias Hepáticas/cirugía , Trasplante de Hígado/efectos adversos , Intercambio Plasmático , Donadores Vivos , Estudios Retrospectivos , Recurrencia Local de Neoplasia/epidemiología , Recurrencia Local de Neoplasia/etiología
6.
Cell ; 133(2): 303-13, 2008 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-18423201

RESUMEN

Different protein complexes form on newly spliced mRNA to ensure the accuracy and efficiency of eukaryotic gene expression. For example, the exon junction complex (EJC) plays an important role in mRNA surveillance. The EJC also influences the first, or pioneer round of protein synthesis through a mechanism that is poorly understood. We show that the nutrient-, stress-, and energy-sensing checkpoint kinase, mTOR, contributes to the observed enhanced translation efficiency of spliced over nonspliced mRNAs. We demonstrate that, when activated, S6K1 is recruited to the newly synthesized mRNA by SKAR, which is deposited at the EJC during splicing, and that SKAR and S6K1 increase the translation efficiency of spliced mRNA. Thus, SKAR-mediated recruitment of activated S6K1 to newly processed mRNPs serves as a conduit between mTOR checkpoint signaling and the pioneer round of translation when cells exist in conditions supportive of protein synthesis.


Asunto(s)
Proteínas Nucleares/metabolismo , Biosíntesis de Proteínas , Proteínas Quinasas/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Factor 4A Eucariótico de Iniciación/metabolismo , Exones , Humanos , Complejo Proteico Nuclear de Unión a la Caperuza/metabolismo , Empalme del ARN , Ribonucleoproteínas/metabolismo , Serina-Treonina Quinasas TOR
7.
Mol Cell ; 59(3): 382-98, 2015 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-26190261

RESUMEN

Insufficient nutrients disrupt physiological homeostasis, resulting in diseases and even death. Considering the physiological and pathological consequences of this metabolic stress, the adaptive responses that cells utilize under this condition are of great interest. We show that under low-glucose conditions, cells initiate adaptation followed by apoptosis responses using PERK/Akt and MEK1/ERK2 signaling, respectively. For adaptation, cells engage the ER stress-induced unfolded protein response, which results in PERK/Akt activation and cell survival. Sustained and extreme energetic stress promotes a switch to isoform-specific MEK1/ERK2 signaling, induction of GCN2/eIF2α phosphorylation, and ATF4 expression, which overrides PERK/Akt-mediated adaptation and induces apoptosis through ATF4-dependent expression of pro-apoptotic factors including Bid and Trb3. ERK2 activation during metabolic stress contributes to changes in TCA cycle and amino acid metabolism, and cell death, which is suppressed by glutamate and α-ketoglutarate supplementation. Taken together, our results reveal promising targets to protect cells or tissues from metabolic stress.


Asunto(s)
Glucosa/farmacología , Ácido Glutámico/farmacología , Ácidos Cetoglutáricos/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Transducción de Señal/efectos de los fármacos , Apoptosis , Supervivencia Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Estrés Fisiológico/efectos de los fármacos
8.
Proc Natl Acad Sci U S A ; 116(8): 2967-2976, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30728292

RESUMEN

ERK is a key coordinator of the epithelial-to-mesenchymal transition (EMT) in that a variety of EMT-inducing factors activate signaling pathways that converge on ERK to regulate EMT transcription programs. However, the mechanisms by which ERK controls the EMT program are not well understood. Through an analysis of the global changes of gene expression mediated by ERK2, we identified the transcription factor FoxO1 as a potential mediator of ERK2-induced EMT, and thus we investigated the mechanism by which ERK2 regulates FoxO1. Additionally, our analysis revealed that ERK2 induced the expression of Dock10, a Rac1/Cdc42 GEF, during EMT. We demonstrate that the activation of the Rac1/JNK signaling axis downstream of Dock10 leads to an increase in FoxO1 expression and EMT. Taken together, our study uncovers mechanisms by which epithelial cells acquire less proliferative but more migratory mesenchymal properties and reveals potential therapeutic targets for cancers evolving into a metastatic disease state.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Proteína Forkhead Box O1/genética , Factores de Intercambio de Guanina Nucleótido/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Línea Celular Tumoral , Regulación de la Expresión Génica/genética , Humanos , Sistema de Señalización de MAP Quinasas/genética , Activación Transcripcional/genética , Proteína de Unión al GTP rac1/genética
9.
Exp Cell Res ; 379(1): 55-64, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30922922

RESUMEN

Metabolic studies of human pluripotent stem cells (hPSCs) have focused on how the cells produce energy through the catabolic pathway. The less-studied anabolic pathway, by which hPSCs expend energy in the form of adenosine triphosphate (ATP), is not yet fully understood. Compared to fully differentiated somatic cells, hPSCs undergo significant changes not only in their gene expression but also in their production and/or expenditure of ATP. Here, we investigate how hPSCs tightly control their energy homeostasis by studying the main energy-consuming process, mRNA translation. In addition, change of subcellular organelles regarding energy homeostasis has been investigated. Lysosomes are organelles that play an important role in the elimination of unnecessary cellular materials by digestion and in the recycling system of the cell. We have found that hPSCs control their lysosome numbers in part by regulating lysosomal gene/protein expression. Thus, because the levels of mRNA translation rate are lower in hPSCs than in somatic cells, not only the global translational machinery but also the lysosomal recycling machinery is suppressed in hPSCs. Overall, the results of our study suggest that hPSCs reprogram gene expression and signaling to regulate energy-consuming processes and energy-controlling organelles.


Asunto(s)
Metabolismo Energético/fisiología , Orgánulos/metabolismo , Células Madre Pluripotentes/metabolismo , Adenosina Trifosfato/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Expresión Génica/fisiología , Homeostasis/fisiología , Humanos , Lisosomas/metabolismo , Biosíntesis de Proteínas/fisiología , ARN Mensajero/metabolismo , Transducción de Señal/fisiología
11.
Mol Cell ; 38(1): 114-27, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20385094

RESUMEN

Hyperactivation of Ras-ERK1/2 signaling is critical to the development of many human malignancies, but little is known regarding the specific contribution of ERK1 or ERK2 to oncogenic processes. We demonstrate that ERK2 but not ERK1 signaling is necessary for Ras-induced epithelial-to-mesenchymal transformation (EMT). Further, ERK2 but not ERK1 overexpression is sufficient to induce EMT. Many ERK1/2-interacting proteins contain amino acid motifs, e.g., DEF or D-motifs, which regulate docking with ERK1/2. Remarkably, ERK2 signaling to DEF motif-containing targets is required to induce EMT and correlates with increased migration, invasion, and survival. Importantly, the late-response gene product Fra1 is necessary for Ras- and ERK2-induced EMT through upregulation of ZEB1/2 proteins. Thus, an apparent critical role for ERK2 DEF motif signaling during tumorigenesis is the regulation of Fra1 and the subsequent induction of ZEB1/2, suggesting a potential therapeutic target for Ras-regulated tumorigenesis.


Asunto(s)
Secuencias de Aminoácidos , Diferenciación Celular/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Transducción de Señal/fisiología , Biomarcadores/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Células Epiteliales/citología , Células Epiteliales/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Mutación , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Interferencia de ARN , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Homeobox 1 de Unión a la E-Box con Dedos de Zinc , Proteínas ras/genética , Proteínas ras/metabolismo
12.
Mol Cell ; 38(4): 487-99, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20513425

RESUMEN

The mTORC1-signaling pathway integrates environmental conditions into distinct signals for cell growth by balancing anabolic and catabolic processes. Accordingly, energetic stress inhibits mTORC1 signaling predominantly through AMPK-dependent activation of TSC1/2. Thus, TSC1/2-/- cells are hypersensitive to glucose deprivation, and this has been linked to increased p53 translation and activation of apoptosis. Herein, we show that mTORC1 inhibition during glucose deprivation prevented not only the execution of death, but also induction of energetic stress. mTORC1 inhibition during glucose deprivation decreased AMPK activation and allowed ATP to remain high, which was both necessary and sufficient for protection. This effect was not due to increased catabolic activities such as autophagy, but rather exclusively due to decreased anabolic processes, reducing energy consumption. Specifically, TSC1/2-/- cells become highly dependent on glutamate dehydrogenase-dependent glutamine metabolism via the TCA cycle for survival. Therefore, mTORC1 inhibition during energetic stress is primarily to balance metabolic demand with supply.


Asunto(s)
Glucosa/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Células Cultivadas , Proteínas Quinasas/metabolismo , Ratas , Transducción de Señal , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/metabolismo
14.
Proc Natl Acad Sci U S A ; 108(47): E1204-13, 2011 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-22065737

RESUMEN

The p70 ribosomal protein S6 kinase 1 (S6K1) plays a key role in cell growth and proliferation by regulating insulin sensitivity, metabolism, protein synthesis, and cell cycle. Thus, deregulation of S6K contributes to the progression of type 2 diabetes, obesity, aging, and cancer. Considering the biological and clinical importance of S6K1, a complete understanding of its regulation is critical. One of the key motifs in the activation of S6K1 is a turn motif, but its regulation is not well understood. Here we provide evidence for two mechanisms of modulating turn motif phosphorylation and S6K1 activity. First, mammalian target of rapamycin regulates turn motif phosphorylation by inhibiting its dephosphorylation. Second, we unexpectedly found that glycogen synthase kinase (GSK)-3 promotes turn motif phosphorylation. Our studies show that mammalian target of rapamycin and GSK-3 cooperate to control the activity of S6K1, an important regulator of cell proliferation and growth. Our unexpected results provide a clear rationale for the development and use of drugs targeting GSK-3 to treat diseases such as diabetes, cancer, and age-related diseases that are linked to improper regulation of S6K1.


Asunto(s)
Proliferación Celular , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Línea Celular Tumoral , Cromatografía Liquida , Técnicas de Silenciamiento del Gen , Humanos , Immunoblotting , Inmunoprecipitación , Fosforilación , Espectrometría de Masas en Tándem , Transfección
15.
Cell Rep ; 42(8): 112868, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37494188

RESUMEN

Cells maintain and dynamically change their proteomes according to the environment and their needs. Mechanistic target of rapamycin (mTOR) is a key regulator of proteostasis, homeostasis of the proteome. Thus, dysregulation of mTOR leads to changes in proteostasis and the consequent progression of diseases, including cancer. Based on the physiological and clinical importance of mTOR signaling, we investigated mTOR feedback signaling, proteostasis, and cell fate. Here, we reveal that mTOR targeting inhibits eIF4E-mediated cap-dependent translation, but feedback signaling activates a translation initiation factor, eukaryotic translation initiation factor 3D (eIF3D), to sustain alternative non-canonical translation mechanisms. Importantly, eIF3D-mediated protein synthesis enables cell phenotype switching from proliferative to more migratory. eIF3D cooperates with mRNA-binding proteins such as heterogeneous nuclear ribonucleoprotein F (hnRNPF), heterogeneous nuclear ribonucleoprotein K (hnRNPK), and Sjogren syndrome antigen B (SSB) to support selective mRNA translation following mTOR inhibition, which upregulates and activates proteins involved in insulin receptor (INSR)/insulin-like growth factor 1 receptor (IGF1R)/insulin receptor substrate (IRS) and interleukin 6 signal transducer (IL-6ST)/Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling. Our study highlights the mechanisms by which cells establish the dynamic change of proteostasis and the resulting phenotype switch.


Asunto(s)
Proteostasis , Receptor de Insulina , ARN Mensajero/metabolismo , Receptor de Insulina/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Sirolimus , Biosíntesis de Proteínas
16.
Am J Physiol Cell Physiol ; 303(7): C743-56, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22855295

RESUMEN

Vascular morphogenesis is a key process for development, reproduction, and pathogenesis. Thus understanding the mechanisms of this process is of pathophysiological importance. Despite the fact that collagen I is the most abundant and potent promorphogenic molecule known, the molecular mechanisms by which this protein regulates endothelial cell tube morphogenesis are still unclear. Here we provide strong evidence that collagen I induces tube morphogenesis by inhibiting glycogen synthase kinase 3ß (GSK3ß). Further mechanistic studies revealed that GSK3ß activity is regulated by protein kinase D (PKD). PKD inhibited GSK3ß activity, which was required for collagen I-induced endothelial tube morphogenesis. We also found that GSK3ß regulated trafficking of integrin α(2)ß(1) in a Rab11-dependent manner. Taken together, our studies highlight the important role of PKD in the regulation of collagen I-induced vascular morphogenesis and show that it is mediated by the modulation of GSK3ß activity and integrin α(2)ß(1) trafficking.


Asunto(s)
Células Endoteliales/enzimología , Glucógeno Sintasa Quinasa 3/fisiología , Integrina alfa2beta1/fisiología , Morfogénesis/fisiología , Proteína Quinasa C/fisiología , Glucógeno Sintasa Quinasa 3 beta , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Transporte de Proteínas/fisiología , Transducción de Señal
17.
Sci Signal ; 15(715): eabm6211, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34982577

RESUMEN

DNA damage and subsequent cellular response are the basis for many cancer treatments. In this issue of Science Signaling, Liu et al. elucidate a mechanism by which cancer cells survive DNA damage induced by radiation and chemotherapy.


Asunto(s)
Daño del ADN , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
18.
Proc Natl Acad Sci U S A ; 105(45): 17414-9, 2008 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-18955708

RESUMEN

The mammalian translational initiation machinery is a tightly controlled system that is composed of eukaryotic initiation factors, and which controls the recruitment of ribosomes to mediate cap-dependent translation. Accordingly, the mTORC1 complex functionally controls this cap-dependent translation machinery through the phosphorylation of its downstream substrates 4E-BPs and S6Ks. It is generally accepted that rapamycin, a specific inhibitor of mTORC1, is a potent translational repressor. Here we report the unexpected discovery that rapamycin's ability to regulate cap-dependent translation varies significantly among cell types. We show that this effect is mechanistically caused by rapamycin's differential effect on 4E-BP1 versus S6Ks. While rapamycin potently inhibits S6K activity throughout the duration of treatment, 4E-BP1 recovers in phosphorylation within 6 h despite initial inhibition (1-3 h). This reemerged 4E-BP1 phosphorylation is rapamycin-resistant but still requires mTOR, Raptor, and mTORC1's activity. Therefore, these results explain how cap-dependent translation can be maintained in the presence of rapamycin. In addition, we have also defined the condition by which rapamycin can control cap-dependent translation in various cell types. Finally, we show that mTOR catalytic inhibitors are effective inhibitors of the rapamycin-resistant phenotype.


Asunto(s)
Proteínas Portadoras/antagonistas & inhibidores , Regulación de la Expresión Génica/efectos de los fármacos , Fosfoproteínas/antagonistas & inhibidores , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas/antagonistas & inhibidores , Sirolimus/farmacología , Factores de Transcripción/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Factores Eucarióticos de Iniciación , Ratones , Fosfoproteínas/metabolismo , Fosforilación , Biosíntesis de Proteínas/fisiología , Proteínas Quinasas S6 Ribosómicas/metabolismo , Factores de Transcripción/metabolismo
19.
J Cell Biol ; 168(1): 29-33, 2005 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-15631989

RESUMEN

We report that the activity of glycogen synthase kinase-3 (GSK-3) is necessary for the maintenance of the epithelial architecture. Pharmacological inhibition of its activity or reducing its expression using small interfering RNAs in normal breast and skin epithelial cells results in a reduction of E-cadherin expression and a more mesenchymal morphology, both of which are features associated with an epithelial-mesenchymal transition (EMT). Importantly, GSK-3 inhibition also stimulates the transcription of Snail, a repressor of E-cadherin and an inducer of the EMT. We identify NFkappaB as a transcription factor inhibited by GSK-3 in epithelial cells that is relevant for Snail expression. These findings indicate that epithelial cells must sustain activation of a specific kinase to impede a mesenchymal transition.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Mesodermo , Factores de Transcripción/metabolismo , Transcripción Genética , Mama/anatomía & histología , Cadherinas/metabolismo , Línea Celular , Proteínas de Unión al ADN/genética , Células Epiteliales/citología , Femenino , Genes Reporteros , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Humanos , Mesodermo/citología , Mesodermo/metabolismo , FN-kappa B/metabolismo , Fenotipo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
20.
Biochem Biophys Rep ; 20: 100656, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31467990

RESUMEN

Naïve pluripotent stem cells (PSCs) display a distinctive phenotype when compared to their "primed" counterparts, including, but not limited to, increased potency to differentiate and more robust mitochondrial respiration. The cultivation and maintenance of naïve PSCs have been notoriously challenging, requiring the use of complex cytokine cocktails. NME7AB is a newly discovered embryonic stem cell growth factor that is expressed exclusively in the first few days of human blastocyst development. It has been previously reported that growing primed induced PSCs (iPSCs) in bFGF-depleted medium with NME7AB as the only added growth factor facilitates the regression of these cells to their naïve state. Here, we confirm this regression by demonstrating the reactivation of mitochondrial function in the induced naïve-like PSCs and increased ATP production in these cells, as compared to that in primed iPSCs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA