Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Intervalo de año de publicación
1.
Int J Mol Sci ; 25(12)2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38928226

RESUMEN

Cell death-inducing p53-target protein 1 (CDIP1) is a proapoptotic protein that is normally expressed at low levels and is upregulated by genotoxic and endoplasmic reticulum stresses. CDIP1 has been reported to be localized to endosomes and to interact with several proteins, including B-cell receptor-associated protein 31 (BAP31) and apoptosis-linked gene 2 (ALG-2). However, the cellular and molecular mechanisms underlying CDIP1 expression-induced apoptosis remain unclear. In this study, we first demonstrated that CDIP1 was upregulated after treatment with the anticancer drug adriamycin in human breast cancer MCF-7 cells but was degraded rapidly in the lysosomal pathway. We also demonstrated that treatment with the cyclin-dependent kinase 5 (CDK5) inhibitor roscovitine led to an increase in the electrophoretic mobility of CDIP1. In addition, a phosphomimetic mutation at Ser-32 in CDIP1 resulted in an increase in CDIP1 expression-induced apoptosis. We also found that CDIP1 expression led to the induction of autophagy prior to apoptosis. Treatment of cells expressing CDIP1 with SAR405, an inhibitor of the class III phosphatidylinositol 3-kinase VPS34, caused a reduction in autophagy and promoted apoptosis. Therefore, autophagy is thought to be a defense mechanism against CDIP1 expression-induced apoptosis.


Asunto(s)
Apoptosis , Autofagia , Neoplasias de la Mama , Femenino , Humanos , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Autofagia/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Fosfatidilinositol 3-Quinasas Clase III/genética , Citoprotección/efectos de los fármacos , Doxorrubicina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células MCF-7
2.
Int J Mol Sci ; 24(4)2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36835331

RESUMEN

Environmental factors are important regulators of cell growth and proliferation. Mechanistic target of rapamycin (mTOR) is a central kinase that maintains cellular homeostasis in response to a variety of extracellular and intracellular inputs. Dysregulation of mTOR signaling is associated with many diseases, including diabetes and cancer. Calcium ion (Ca2+) is important as a second messenger in various biological processes, and its intracellular concentration is tightly regulated. Although the involvement of Ca2+ mobilization in mTOR signaling has been reported, the detailed molecular mechanisms by which mTOR signaling is regulated are not fully understood. The link between Ca2+ homeostasis and mTOR activation in pathological hypertrophy has heightened the importance in understanding Ca2+-regulated mTOR signaling as a key mechanism of mTOR regulation. In this review, we introduce recent findings on the molecular mechanisms of regulation of mTOR signaling by Ca2+-binding proteins, particularly calmodulin (CaM).


Asunto(s)
Serina-Treonina Quinasas TOR , Calcio/metabolismo , Calmodulina/metabolismo , Proteínas Portadoras/metabolismo , Transducción de Señal , Sirolimus , Serina-Treonina Quinasas TOR/metabolismo
3.
Int J Mol Sci ; 22(3)2021 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-33503978

RESUMEN

Apoptosis-linked gene 2 (ALG-2, also known as PDCD6) is a member of the penta-EF-hand (PEF) family of Ca2+-binding proteins. The murine gene encoding ALG-2 was originally reported to be an essential gene for apoptosis. However, the role of ALG-2 in cell death pathways has remained elusive. In the present study, we found that cell death-inducing p53 target protein 1 (CDIP1), a pro-apoptotic protein, interacts with ALG-2 in a Ca2+-dependent manner. Co-immunoprecipitation analysis of GFP-fused CDIP1 (GFP-CDIP1) revealed that GFP-CDIP1 associates with tumor susceptibility gene 101 (TSG101), a known target of ALG-2 and a subunit of endosomal sorting complex required for transport-I (ESCRT-I). ESCRT-I is a heterotetrameric complex composed of TSG101, VPS28, VPS37 and MVB12/UBAP1. Of diverse ESCRT-I species originating from four VPS37 isoforms (A, B, C, and D), CDIP1 preferentially associates with ESCRT-I containing VPS37B or VPS37C in part through the adaptor function of ALG-2. Overexpression of GFP-CDIP1 in HEK293 cells caused caspase-3/7-mediated cell death. In addition, the cell death was enhanced by co-expression of ALG-2 and ESCRT-I, indicating that ALG-2 likely promotes CDIP1-induced cell death by promoting the association between CDIP1 and ESCRT-I. We also found that CDIP1 binds to vesicle-associated membrane protein-associated protein (VAP)A and VAPB through the two phenylalanines in an acidic tract (FFAT)-like motif in the C-terminal region of CDIP1, mutations of which resulted in reduction of CDIP1-induced cell death. Therefore, our findings suggest that different expression levels of ALG-2, ESCRT-I subunits, VAPA and VAPB may have an impact on sensitivity of anticancer drugs associated with CDIP1 expression.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Reguladoras de la Apoptosis/genética , Sitios de Unión , Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Muerte Celular/genética , Línea Celular , Humanos , Unión Proteica , Relación Estructura-Actividad
4.
J Biomed Sci ; 27(1): 87, 2020 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-32799865

RESUMEN

The mechanistic target of rapamycin complex 1 (mTORC1) is an essential regulator of cell growth and metabolism through the modulation of protein and lipid synthesis, lysosome biogenesis, and autophagy. The activity of mTORC1 is dynamically regulated by several environmental cues, including amino acid availability, growth factors, energy levels, and stresses, to coordinate cellular status with environmental conditions. Dysregulation of mTORC1 activity is closely associated with various diseases, including diabetes, cancer, and neurodegenerative disorders. The discovery of Rag GTPases has greatly expanded our understanding of the regulation of mTORC1 activity by amino acids, especially leucine and arginine. In addition to Rag GTPases, other factors that also contribute to the modulation of mTORC1 activity have been identified. In this review, we discuss the mechanisms of regulation of mTORC1 activity by particular amino acids.


Asunto(s)
Aminoácidos/metabolismo , Regulación de la Expresión Génica , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Transducción de Señal , Animales , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones
5.
Int J Mol Sci ; 19(2)2018 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-29463029

RESUMEN

NFAT is a cytoplasm-localized hyper-phosphorylated transcription factor that is activated through dephosphorylation by calcineurin, a Ca2+/calmodulin-dependent phosphatase. A non-palindromic NFAT-response element (RE) found in the IL2 promoter region has been commonly used for a Ca2+-response reporter gene system, but requirement of concomitant activation of AP-1 (Fos/Jun) often complicates the interpretation of obtained results. A new nanoluciferase (NanoLuc) reporter gene containing nine-tandem repeats of a pseudo-palindromic NFAT-RE located upstream of the IL8 promoter was designed to monitor Ca2+-induced transactivation activity of NFAT in human embryonic kidney (HEK) 293 cells by measuring luciferase activities of NanoLuc and co-expressed firefly luciferase for normalization. Ionomycin treatment enhanced the relative luciferase activity (RLA), which was suppressed by calcineurin inhibitors. HEK293 cells that stably express human STIM1 and Orai1, components of the store-operated calcium entry (SOCE) machinery, gave a much higher RLA by stimulation with thapsigargin, an inhibitor of sarcoplasmic/endoplamic reticulum Ca2+-ATPase (SERCA). HEK293 cells deficient in a penta-EF-hand Ca2+-binding protein ALG-2 showed a higher RLA value than the parental cells by stimulation with an acetylcholine receptor agonist carbachol. The novel reporter gene system is found to be useful for applications to cell signaling research to monitor biological endpoint effects of cellular Ca2+ mobilization.


Asunto(s)
Calcio/metabolismo , Genes Reporteros , Luciferasas/metabolismo , Factores de Transcripción NFATC/genética , Nanopartículas/química , Elementos de Respuesta/genética , Secuencias Repetidas en Tándem/genética , Animales , Secuencia de Bases , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Carbacol/farmacología , AMP Cíclico/genética , Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Ratones , Factor de Transcripción AP-1/genética
6.
Biochem Biophys Res Commun ; 497(2): 492-498, 2018 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-29432744

RESUMEN

MAP1B (microtubule-associated protein 1B) binds to microtubules and regulates microtubule dynamics. Previously, we showed calcium-dependent interaction between MAP1B and a calcium-binding protein ALG-2 (apoptosis-linked gene 2), which is involved in regulation of the protein secretion pathway. Although ALG-2 generally binds to proteins through two consensus binding motifs such as ABM-1 and ABM-2, the absence of these motifs in MAP1B suggests a unique binding mode between MAP1B and ALG-2. Here, we identified the region of mouse MAP1B responsible for binding to ALG-2, and found point mutations that abrogated binding of MAP1B to ALG-2. Furthermore, interaction between MAP1B and ALG-2 selectively prevented ALG-2 from binding to proteins with ABM-2 such as Sec31A, suggesting competition between MAP1B and ABM-2-containing proteins for binding to ALG-2. Consistently, in MAP1B knockout cells, co-localization of ALG-2 with Sec31A was increased. Moreover, overexpression of wild-type MAP1B, but not the MAP1B mutant defective in ALG-2 binding, altered localizations of ALG-2 and Sec31A into dispersed distributions, suggesting that MAP1B regulates localizations of ALG-2 and Sec31A in the cells. Finally, we found two cancer-associated mutations of human MAP1B located near ALG-2 binding sites. The introduction of the corresponding mutations in mouse MAP1B dramatically reduced the binding ability to ALG-2. Thus, these results suggest that MAP1B plays a role in regulation of ALG-2 and Sec31A localizations, and that dysregulation of calcium-dependent binding of ALG-2 to MAP1B might influence pathological conditions such as cancers.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Secuencia de Aminoácidos , Proteínas Reguladoras de la Apoptosis/análisis , Sitios de Unión , Proteínas de Unión al Calcio/análisis , Células HEK293 , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/análisis , Proteínas Asociadas a Microtúbulos/genética , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Unión Proteica , Proteínas de Transporte Vesicular/análisis , Proteínas de Transporte Vesicular/metabolismo
7.
Int J Mol Sci ; 16(2): 3677-99, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25667979

RESUMEN

ALG-2, a 22-kDa penta-EF-hand protein, is involved in cell death, signal transduction, membrane trafficking, etc., by interacting with various proteins in mammalian cells in a Ca2+-dependent manner. Most known ALG-2-interacting proteins contain proline-rich regions in which either PPYPXnYP (type 1 motif) or PXPGF (type 2 motif) is commonly found. Previous X-ray crystal structural analysis of the complex between ALG-2 and an ALIX peptide revealed that the peptide binds to the two hydrophobic pockets. In the present study, we resolved the crystal structure of the complex between ALG-2 and a peptide of Sec31A (outer shell component of coat complex II, COPII; containing the type 2 motif) and found that the peptide binds to the third hydrophobic pocket (Pocket 3). While amino acid substitution of Phe85, a Pocket 3 residue, with Ala abrogated the interaction with Sec31A, it did not affect the interaction with ALIX. On the other hand, amino acid substitution of Tyr180, a Pocket 1 residue, with Ala caused loss of binding to ALIX, but maintained binding to Sec31A. We conclude that ALG-2 recognizes two types of motifs at different hydrophobic surfaces. Furthermore, based on the results of serial mutational analysis of the ALG-2-binding sites in Sec31A, the type 2 motif was newly defined.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/metabolismo , Péptidos/química , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/genética , Sustitución de Aminoácidos , Proteínas Reguladoras de la Apoptosis/genética , Sitios de Unión , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/metabolismo , Cristalografía por Rayos X , Análisis Mutacional de ADN , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Unión Proteica
8.
FEBS J ; 281(16): 3642-55, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24953135

RESUMEN

Calpain-7 (CAPN7) is a unique intracellular cysteine protease that has a tandem repeat of microtubule interacting and trafficking (MIT) domains and lacks a penta-EF-hand domain. Although the MIT domains of CAPN7 were previously shown to interact with a subset of endosomal sorting complex required for transport (ESCRT)-III and ESCRT-III-related proteins, including charged multivesicular body protein 1 and increased sodium tolerance (IST)1, knowledge of the involvement of the protease in membrane trafficking has been limited. In the present study, compared with control cells, we found that epidermal growth factor receptor (EGFR) degradation was mildly delayed in CAPN7-knockdown HeLa cells and mouse embryonic fibroblast cells established from CAPN7 knockout (Capn7(-/-) ) mice. Re-expression of wild-type CAPN7 but not a protease-inactive mutant of CAPN7 (CAPN7(C290S) ) resulted in a recovery of the rate of EGFR degradation. We found, by immunofluorescence microscopic analysis, that monomeric GFP fused with the protease-inactive mutant of CAPN7 [monomeric green fluorescent protein (mGFP)-CAPN7(C290S) ] was mobilized to EGFR-positive endosomes upon epidermal growth factor stimulation in HeLa cells. Although mGFP-CAPN7(C290S) exhibited dominant-negative effects on EGFR degradation, a deletion mutant of MIT domains in mGFP-CAPN7(C290S) did not have such properties, suggesting that the interaction between the MIT domains and ESCRT proteins is important for the function of CAPN7. Moreover, we found that epidermal growth factor stimulation induces translocation of IST1 from the cytosol to endosomes positive in both EGFR and mGFP-CAPN7(C290S) . When IST1 was knocked down, mGFP-CAPN7(C290S) lost its co-localization with EGFR. These results demonstrate for the first time that the proteolytic activity of CAPN7 is important for the acceleration of EGFR degradation via the endosomal sorting pathway utilizing a part of the ESCRT system. STRUCTURED DIGITAL ABSTRACT: EGFR and CAPN7 colocalize by fluorescence microscopy (View interaction) EGFR, CAPN7 and IST1 colocalize by fluorescence microscopy (View interaction) EEA1 and CAPN7 colocalize by fluorescence microscopy (View interaction) CAPN7 and LAMP1 colocalize by fluorescence microscopy (View interaction).


Asunto(s)
Calpaína/fisiología , Endosomas/enzimología , Receptores ErbB/metabolismo , Animales , Calpaína/química , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Proteínas Oncogénicas/metabolismo , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteolisis
9.
Biosci Biotechnol Biochem ; 77(7): 1608-11, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23832372

RESUMEN

The targets of rapamycin (Tor) kinases play central roles in the integrated regulation of cellular activities. Although the molecular mechanisms of Tor-mediated signaling pathways have been studied extensively in yeast, the relationship between kinase activity and the redox maintenance system remains obscure. In this study, we established a quantitative extraction and determination method for glutathione-related compounds in Saccharomyces cerevisiae utilizing liquid chromatography-tandem mass spectrometry (LC-MS/MS). We found decreases in the levels of glutathione and its precursors resulting from the introduction of a Tor1 hyper-active mutation. In line with this finding, the mutant was more sensitive to several heavy metal ions, indicating a physiological defect arising from a failure to regulate the kinase activity.


Asunto(s)
Glutatión/metabolismo , Espacio Intracelular/metabolismo , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/enzimología , Serina-Treonina Quinasas TOR/metabolismo , Cromatografía Liquida , Activación Enzimática , Mutación , Saccharomyces cerevisiae/genética , Serina-Treonina Quinasas TOR/genética , Espectrometría de Masas en Tándem
10.
J Biochem ; 154(1): 1-10, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23698095

RESUMEN

The target of rapamycin (TOR) is an evolutionarily conserved protein kinase that regulates cell growth in response to various environmental as well as intracellular cues through the formation of 2 distinct TOR complexes (TORC), TORC1 and TORC2. Dysregulation of TORC1 and TORC2 activity is closely associated with various diseases, including diabetes, cancer and neurodegenerative disorders. Over the past few years, new regulatory mechanisms of TORC1 and TORC2 activity have been elucidated. Furthermore, recent advances in the study of TOR inhibitors have revealed previously unrecognized cellular functions of TORC1. In this review, we briefly summarize the current understanding of the evolutionarily conserved TOR signalling from upstream regulators to downstream events.


Asunto(s)
Serina-Treonina Quinasas TOR/metabolismo , Animales , Autofagia , Secuencia Conservada , Evolución Molecular , Humanos , Lípidos/biosíntesis , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Modelos Biológicos , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribosomas/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/química , Serina-Treonina Quinasas TOR/genética
11.
J Biol Chem ; 283(46): 31861-70, 2008 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-18812319

RESUMEN

The mammalian target of rapamycin (mTOR) is a Ser/Thr kinase that plays essential roles in the regulation of a wide array of growth-related processes such as protein synthesis, cell sizing, and autophagy. mTOR forms two functionally distinct complexes, termed the mTOR complex 1 (mTORC1) and 2 (mTORC2); only the former of which is inhibited by rapamycin. Based on the similarity between the cellular responses caused by rapamycin treatment and by nutrient starvation, it has been widely accepted that modulation in the mTORC1 activity in response to nutrient status directs these cellular responses, although direct evidence has been scarce. Here we report isolation of hyperactive mutants of mTOR. The isolated mTOR mutants exhibited enhanced kinase activity in vitro and rendered cells refractory to the dephosphorylation of the mTORC1 substrates upon amino acid starvation. Cells expressing the hyperactive mTOR mutant displayed larger cell size in a normal growing condition and were resistant to cell size reduction and autophagy induction in an amino acid-starved condition. These results indicate that the activity of mTORC1 actually directs these cellular processes in response to nutrient status and confirm the biological functions of mTORC1, which had been proposed solely from loss-of-function analyses using rapamycin and (molecular)genetic techniques. Additionally, the hyperactive mTOR mutant did not induce cellular transformation of NIH/3T3 cells, suggesting that concomitant activation of additional pathways is required for tumorigenesis. This hyperactive mTOR mutant will be a valuable tool for establishing physiological consequences of mTOR activation in cells as well as in organisms.


Asunto(s)
Proteínas Quinasas/metabolismo , Animales , Autofagia , Línea Celular , Tamaño de la Célula , Activación Enzimática , Regulación Enzimológica de la Expresión Génica , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos , Mutación/genética , Proteínas Quinasas/genética , Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA