Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 64(6): 1048-1061, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27867007

RESUMEN

The ERK-regulated ternary complex factors (TCFs) act with the transcription factor serum response factor (SRF) to activate mitogen-induced transcription. However, the extent of their involvement in the immediate-early transcriptional response, and their wider functional significance, has remained unclear. We show that, in MEFs, TCF inactivation significantly inhibits over 60% of TPA-inducible gene transcription and impairs cell proliferation. Using integrated SRF ChIP-seq and Hi-C data, we identified over 700 TCF-dependent SRF direct target genes involved in signaling, transcription, and proliferation. These also include a significant number of cytoskeletal gene targets for the Rho-regulated myocardin-related transcription factor (MRTF) SRF cofactor family. The TCFs act as general antagonists of MRTF-dependent SRF target gene expression, competing directly with the MRTFs for access to SRF. As a result, TCF-deficient MEFs exhibit hypercontractile and pro-invasive behavior. Thus, competition between TCFs and MRTFs for SRF determines the balance between antagonistic proliferative and contractile programs of gene expression.


Asunto(s)
Fibroblastos/metabolismo , Regulación de la Expresión Génica , Factor de Respuesta Sérica/genética , Factores Complejos Ternarios/genética , Animales , Secuencia de Bases , Línea Celular , Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Perfilación de la Expresión Génica , Prueba de Complementación Genética , Humanos , Ratones , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Factores Complejos Ternarios/antagonistas & inhibidores , Factores Complejos Ternarios/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Transactivadores/genética , Transactivadores/metabolismo , Transcripción Genética , Proteína Elk-1 con Dominio ets/genética , Proteína Elk-1 con Dominio ets/metabolismo
2.
Int J Mol Sci ; 23(19)2022 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-36232837

RESUMEN

CCG-1423 is a Rho A pathway inhibitor that has been reported to inhibit Rho/SRF-mediated transcriptional regulation. Serum response factor and its cofactors, which include ternary complex factors and myocardin-related transcription factors, regulate various cellular functions. In this study, we observed that CCG-1423 modulates the mitochondrial functions. The effect of this small molecule drug was determined by measuring mitochondrial function using an XFe96 Analyzer and an Oxygraph 2k (O2k) high-resolution respirometer. CCG-1423 treatment significantly reduced oxidative phosphorylation in a dose-dependent manner. However, CCG-1423 increased the glycolytic rate. We also observed that histone 4 at lysine-16 underwent hyperacetylation with the treatment of this drug. Immunolabeling with F-actin and MitoTracker revealed the alteration in the actin cytoskeleton and mitochondria. Taken together, our findings highlight a critical role of CCG-1423 in inhibiting the transcription of SRF/p49 and PGC-1α, ß, resulting in the downregulation of mitochondrial genes, leading to the repression of mitochondrial oxidative phosphorylation and overall ATP reduction. This study provides a better understanding of the effects of CCG-1423 on mitochondria, which may be useful for the assessment of the potential clinical application of CCG-1423 and its derivatives.


Asunto(s)
Actinas , Factor de Respuesta Sérica , Actinas/metabolismo , Adenosina Trifosfato , Anilidas , Benzamidas , Histonas , Lisina , Mitocondrias/metabolismo , Factores Complejos Ternarios/metabolismo , Factores de Transcripción/metabolismo
3.
Proc Natl Acad Sci U S A ; 115(11): E2653-E2662, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29487210

RESUMEN

There is currently an unmet need for versatile techniques to monitor the assembly and dynamics of ternary complexes in live cells. Here we describe bioluminescence resonance energy transfer with fluorescence enhancement by combined transfer (BRETFect), a high-throughput technique that enables robust spectrometric detection of ternary protein complexes based on increased energy transfer from a luciferase to a fluorescent acceptor in the presence of a fluorescent intermediate. Its unique donor-intermediate-acceptor relay system is designed so that the acceptor can receive energy either directly from the donor or indirectly via the intermediate in a combined transfer, taking advantage of the entire luciferase emission spectrum. BRETFect was used to study the ligand-dependent cofactor interaction properties of the estrogen receptors ERα and ERß, which form homo- or heterodimers whose distinctive regulatory properties are difficult to dissect using traditional methods. BRETFect uncovered the relative capacities of hetero- vs. homodimers to recruit receptor-specific cofactors and regulatory proteins, and to interact with common cofactors in the presence of receptor-specific ligands. BRETFect was also used to follow the assembly of ternary complexes between the V2R vasopressin receptor and two different intracellular effectors, illustrating its use for dissection of ternary protein-protein interactions engaged by G protein-coupled receptors. Our results indicate that BRETFect represents a powerful and versatile technique to monitor the dynamics of ternary interactions within multimeric complexes in live cells.


Asunto(s)
Técnicas Citológicas/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Factores Complejos Ternarios/metabolismo , Células HEK293 , Humanos , Proteínas Luminiscentes , Receptores Citoplasmáticos y Nucleares/análisis , Receptores Citoplasmáticos y Nucleares/química , Receptores Acoplados a Proteínas G/análisis , Receptores Acoplados a Proteínas G/química , Factores Complejos Ternarios/análisis , Factores Complejos Ternarios/química
4.
Dev Biol ; 416(1): 173-186, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27235147

RESUMEN

Elk proteins are Ets family transcription factors that regulate cell proliferation, survival, and differentiation in response to ERK (extracellular-signal regulated kinase)-mediated phosphorylation. Here we report the embryonic expression and function of Sp-Elk, the single Elk gene of the sea urchin Strongylocentrotus purpuratus. Sp-Elk is zygotically expressed throughout the embryo beginning at late cleavage stage, with peak expression occurring at blastula stage. Morpholino antisense-mediated knockdown of Sp-Elk causes blastula-stage developmental arrest and embryo disintegration due to apoptosis, a phenotype that is rescued by wild-type Elk mRNA. Development is also rescued by Elk mRNA encoding a serine to aspartic acid substitution (S402D) that mimics ERK-mediated phosphorylation of a conserved site that enhances DNA binding, but not by Elk mRNA encoding an alanine substitution at the same site (S402A). This demonstrates both that the apoptotic phenotype of the morphants is specifically caused by Elk depletion, and that phosphorylation of serine 402 of Sp-Elk is critical for its anti-apoptotic function. Knockdown of Sp-Elk results in under-expression of several regulatory genes involved in cell fate specification, cell cycle control, and survival signaling, including the transcriptional regulator Sp-Runt-1 and its target Sp-PKC1, both of which were shown previously to be required for cell survival during embryogenesis. Both Sp-Runt-1 and Sp-PKC1 have sequences upstream of their transcription start sites that specifically bind Sp-Elk. These results indicate that Sp-Elk is the signal-dependent activator of a feed-forward gene regulatory circuit, consisting also of Sp-Runt-1 and Sp-PKC1, which actively suppresses apoptosis in the early embryo.


Asunto(s)
Supervivencia Celular , Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Erizos de Mar/embriología , Transducción de Señal , Factores Complejos Ternarios/metabolismo , Animales , Apoptosis/genética , Blástula , Supervivencia Celular/genética , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Oligonucleótidos Antisentido , Fosforilación , Regiones Promotoras Genéticas , Erizos de Mar/genética , Erizos de Mar/metabolismo , Transducción de Señal/genética
5.
Mol Pharmacol ; 87(4): 617-28, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25576487

RESUMEN

The steroid pregnenolone sulfate activates the transcription factor activator protein-1 (AP-1) via stimulation of transient receptor potential melastatin-3 (TRPM3) channels. Here, we show that the signaling pathway requires an influx of Ca(2+) ions into the cells and a rise in the intracellular Ca(2+) levels. The upregulation of AP-1 was attenuated in cells that overexpressed mitogen activated protein kinase phosphatase-1, indicating that Ca(2+) ions prolong the signaling cascade via activation of mitogen activated protein kinases. On the transcriptional level, expression of a dominant-negative mutant of the basic region leucine zipper protein c-Jun, a major constituent of the AP-1 transcription factor complex, or expression of a c-Jun-specific short hairpin RNA attenuated pregnenolone sulfate-induced AP-1 activation. In addition, stimulation of TRPM3 channels increased the transcriptional activation potential of the basic region leucine zipper protein ATF2. Inhibition of ATF2 target gene expression via expression of a dominant-negative mutant of ATF2 or expression of an ATF2-specific short hairpin RNA interfered with TRPM3-mediated stimulation of AP-1. Moreover, we show that a dominant-negative mutant of the ternary complex factor (TCF) Elk-1 attenuated the upregulation of AP-1 following stimulation of TRPM3 channels. Thus, c-Jun, ATF2, and TCFs are required to connect the intracellular signaling cascade elicited by activation of TRPM3 channels with enhanced transcription of AP-1-regulated genes. We conclude that pregnenolone sulfate-induced TRPM3 channel activation changes the gene expression pattern of the cells by activating transcription of c-Jun-, ATF2-, and TCF-controlled genes.


Asunto(s)
Factor de Transcripción Activador 2/metabolismo , Calcio/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Canales Catiónicos TRPM/metabolismo , Factores Complejos Ternarios/metabolismo , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción Activador 2/genética , Cationes , Células HEK293 , Humanos , Pregnenolona/farmacología , Proteínas Proto-Oncogénicas c-jun/genética , Canales Catiónicos TRPM/genética , Factores Complejos Ternarios/genética , Transcripción Genética , Regulación hacia Arriba , Proteína Elk-1 con Dominio ets/genética , Proteína Elk-1 con Dominio ets/metabolismo
6.
Biochim Biophys Acta ; 1829(3-4): 306-17, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23092677

RESUMEN

The synthesis of ribosomal RNA (rRNA) precursor molecules by RNA polymerase I (Pol I) terminates with the dissociation of the protein-DNA-RNA ternary complex. Based on in vitro results the mechanism of Pol I termination appeared initially to be rather conserved and simple until this process was more thoroughly re-investigated in vivo. A picture emerged that Pol I termination seems to be connected to co-transcriptional processing, re-initiation of transcription and, possibly, other processes downstream of Pol I transcription units. In this article, our current understanding of the mechanism of Pol I termination and how this process might be implicated in other biological processes in yeast and mammals is summarized and discussed. This article is part of a Special Issue entitled: Transcription by Odd Pols.


Asunto(s)
ARN Polimerasa I/metabolismo , Terminación de la Transcripción Genética , Animales , Humanos , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , Precursores del ARN/biosíntesis , Precursores del ARN/genética , ARN Ribosómico/biosíntesis , ARN Ribosómico/genética , Factores Complejos Ternarios/metabolismo , Levaduras/genética , Levaduras/metabolismo
7.
Genes Cells ; 17(8): 688-97, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22734669

RESUMEN

Dock3, a new member of the guanine nucleotide exchange factor family, causes cellular morphological changes by activating the small GTPase Rac1. Overexpression of Dock3 in neural cells promotes neurite outgrowth through the formation of a protein complex with Fyn and WAVE downstream of brain-derived neurotrophic factor (BDNF) signaling. Here, we report a novel Dock3-mediated BDNF pathway for neurite outgrowth. We show that Dock3 forms a complex with Elmo and activated RhoG downstream of BDNF-TrkB signaling and induces neurite outgrowth via Rac1 activation in PC12 cells. We also show the importance of Dock3 phosphorylation in Rac1 activation and show two key events that are necessary for efficient Dock3 phosphorylation: membrane recruitment of Dock3 and interaction of Dock3 with Elmo. These results suggest that Dock3 plays important roles downstream of BDNF signaling in the central nervous system where it stimulates actin polymerization by multiple pathways.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuritas/metabolismo , Receptor trkB/metabolismo , Transducción de Señal , Factores Complejos Ternarios/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células COS , Proteínas Portadoras/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Chlorocebus aethiops , Activación Enzimática , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Factores de Intercambio de Guanina Nucleótido , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Neuritas/efectos de los fármacos , Neuritas/fisiología , Neuropéptidos/genética , Neuropéptidos/metabolismo , Células PC12 , Fosforilación , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Ratas , Receptor trkB/genética , Factores Complejos Ternarios/genética , Transfección , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1 , Proteínas de Unión al GTP rho
8.
J Pharmacol Exp Ther ; 342(1): 41-52, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22454534

RESUMEN

Morphine-induced signaling via opioid receptors (ORs) in dorsal root ganglia (DRG) neurons, the spinal cord, and various brain regions has been shown to modulate gene activity. Hitherto, little attention has been paid to extracellular signal-regulated kinases-1/2 (ERK-1/2)-mediated activation of the serum response factor (SRF) and ternary complex factors (TCFs) such as the E twenty six-like transcription factor-1 (ELK-1) in this context. Using TCF/SRF-dependent reporter gene constructs, a specific ERK-1/2 inhibitor and a dominant-negative ELK-1 mutant, we show herein that morphine activates ELK-1 via ERK-1/2 in DRG-derived F11 cells endogenously expressing µ and δ ORs. Previous studies with glioma cell lines such as NG108-15 cells attributed morphine-induced gene expression to the activation of the cAMP-responsive element binding protein (CREB). Thus, we also analyzed morphine-dependent activation of CREB in F11 and NG108-15 cells. In contrast to the CREB stimulation found in NG108-15 cells, we observed an inhibitory effect of morphine in F11 cells, indicating cell type-specific regulation of CREB by morphine. To obtain data about putative target genes of morphine-induced ELK-1/SRF activation, we analyzed mRNA levels of 15 ELK-1/SRF-dependent genes in cultured rat DRG neurons and F11 cells. We identified the early growth response protein-4 (EGR-4) as the strongest up-regulated gene in both cell types and observed ELK-1 activity-dependent activation of an EGR-4-driven reporter in F11 cells. Overall, we reveal an important role of ELK-1 for morphine-dependent gene induction in DRG-derived cells and propose that ELK-1 and EGR-4 contribute to the effects of morphine on neuronal plasticity.


Asunto(s)
Ganglios Espinales/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Morfina/farmacología , Neuronas/efectos de los fármacos , Factor de Respuesta Sérica/metabolismo , Proteína Elk-1 con Dominio ets/metabolismo , Animales , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Factores de Transcripción de la Respuesta de Crecimiento Precoz/genética , Factores de Transcripción de la Respuesta de Crecimiento Precoz/metabolismo , Ganglios Espinales/metabolismo , Ratones , Neuronas/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Factor de Respuesta Sérica/genética , Transducción de Señal/efectos de los fármacos , Factores Complejos Ternarios/genética , Factores Complejos Ternarios/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteína Elk-1 con Dominio ets/genética
9.
Elife ; 112022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-35044299

RESUMEN

Serum response factor (SRF) is an essential transcription factor that influences many cellular processes including cell proliferation, migration, and differentiation. SRF directly regulates and is required for immediate early gene (IEG) and actin cytoskeleton-related gene expression. SRF coordinates these competing transcription programs through discrete sets of cofactors, the ternary complex factors (TCFs) and myocardin-related transcription factors (MRTFs). The relative contribution of these two programs to in vivo SRF activity and mutant phenotypes is not fully understood. To study how SRF utilizes its cofactors during development, we generated a knock-in SrfaI allele in mice harboring point mutations that disrupt SRF-MRTF-DNA complex formation but leave SRF-TCF activity unaffected. Homozygous SrfaI/aI mutants die at E10.5 with notable cardiovascular phenotypes, and neural crest conditional mutants succumb at birth to defects of the cardiac outflow tract but display none of the craniofacial phenotypes associated with complete loss of SRF in that lineage. Our studies further support an important role for MRTF mediating SRF function in cardiac neural crest and suggest new mechanisms by which SRF regulates transcription during development.


Asunto(s)
Cresta Neural/embriología , Factor de Respuesta Sérica/genética , Factores Complejos Ternarios/genética , Factores de Transcripción/genética , Animales , Ratones , Factor de Respuesta Sérica/metabolismo , Factores Complejos Ternarios/metabolismo , Factores de Transcripción/metabolismo
10.
J Biol Chem ; 285(29): 22036-49, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20466732

RESUMEN

The induction of expression of many cellular immediate early genes (IEG) involves the transcription factor serum response factor (SRF). Two families of SRF coactivators have also been implicated in IEG induction, the ternary complex factors (TCFs), ELK1, Sap1, and Net, and the myocardin-related factors, MKL1 and MKL2. We found that serum induction of some SRF target genes is preferentially regulated by MKL1/2, whereas others are redundantly activated by both TCFs and MKL1/2. Yet ELK1 can also repress transcription. Binding of ELK1 and MKL1 to SRF has been found to be mutually exclusive in vitro, suggesting that ELK1 could repress expression of IEGs by blocking MKL1 binding. We characterized the in vivo binding of MKL1 and ELK1 to target genes and found an inverse relationship of serum-induced MKL1 binding and serum-decreased ELK1 binding. However, experiments with short hairpin RNA-mediated MKL1/2 depletion and expression of a nuclear MKL1 (N100) variant in stably transfected cells failed to alter ELK1 binding, suggesting that ELK1 binding to target genes is regulated independently of MKL1/2. Nevertheless, we found that short interfering RNA-mediated depletion of TCFs increased target gene expression in cells containing the N100 MKL1 activator, most notably in cells under continuous growth conditions. These results indicate that the TCFs can function both as activators and repressors of target gene expression depending upon the cellular growth conditions.


Asunto(s)
Regulación de la Expresión Génica , Genes Inmediatos-Precoces/genética , Proteínas Represoras/metabolismo , Factor de Respuesta Sérica/metabolismo , Animales , Núcleo Celular/metabolismo , Proteínas Co-Represoras/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Humanos , Ratones , Células 3T3 NIH , Regiones Promotoras Genéticas/genética , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Interferente Pequeño/metabolismo , Eliminación de Secuencia , Suero , Factores Complejos Ternarios/metabolismo , Transactivadores/química , Transactivadores/deficiencia , Activación Transcripcional/genética , Proteína Elk-1 con Dominio ets/metabolismo
11.
Genes Cells ; 15(3): 297-311, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20184660

RESUMEN

EphA4 belongs to a superfamily of receptor tyrosine kinases and interacts with several molecules including fibroblast growth factor receptors (FGFRs) as we reported earlier. Several receptor tyrosine kinases, FGFRs, Trks, Alk and Ret, are currently known to transduce a signal through a docking protein, fibroblast growth factor receptor substrate 2α (FRS2α). However, nothing has been reported about the interaction of FRS2α with EphA4. Using the yeast two-hybrid system and the in vitro binding and kinase assays, we found that the mid-kinase region of EphA4 directly interacts with the FRS2α PTB domain upon tyrosine phosphorylation of the EphA4 juxtamembrane (JM) domain and EphA4 directly phosphorylates FRS2α. We also found that the FRS2α PTB domain and the amino-terminal region of EphA4 bind to the amino- and carboxy-terminal regions of the FGFR JM domain, respectively, suggesting that FRS2α and EphA4 interact with FGFR simultaneously. Furthermore, a kinase-dead EphA4 mutant that constitutively binds to FGFR functions as a dominant-negative molecule for signaling through both EphA4 and FGFR, and so does the truncated FRS2α lacking multiple tyrosine phosphorylation sites. These dominant-negative mutants similarly inhibit the ligand-dependent proliferation of the mouse embryonic neural stem/progenitor cells. These results suggest the formation of a ternary complex comprising EphA4, FGFR and FRS2α. The signaling complex appears to integrate the input from FGFR and EphA4, and release the output signal through FRS2α.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proliferación Celular , Células Madre Embrionarias/metabolismo , Proteínas de la Membrana/metabolismo , Células-Madre Neurales/metabolismo , Receptor EphA4/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Factores Complejos Ternarios/metabolismo , Animales , Línea Celular , Células Madre Embrionarias/citología , Humanos , Ratones , Células-Madre Neurales/citología , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal
12.
Mol Cell Neurosci ; 44(2): 165-77, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20304071

RESUMEN

ELK transcription factors are known to be expressed in a number of regions in the nervous system. We show by RT-PCR that the previously described Elk1, Elk3/Elk3b/Elk3c and Elk4 mRNAs are expressed in adult dorsal root ganglia (DRG), together with the novel alternatively spliced isoforms Elk1b, Elk3d and Elk4c/Elk4d/Elk4e. These isoforms are also expressed in brain, heart, kidney and testis. In contrast to Elk3 protein, the novel Elk3d isoform is cytoplasmic, fails to bind ETS binding sites and yet can activate transcription by an indirect mechanism. The Elk3 and Elk4 genes are overlapped by co-expressed Pctk2 (Cdk17) and Mfsd4 genes, respectively, with the potential formation of Elk3/Pctaire2 and Elk4/Mfsd4 sense-antisense mRNA heteroduplexes. After peripheral nerve injury the Elk3 mRNA isoforms are each upregulated approximately 2.3-fold in DRG (P<0.005), whereas the natural antisense Pctaire2 isoforms show only a small increase (21%, P<0.01) and Elk1 and Elk4 mRNAs are unchanged.


Asunto(s)
Empalme Alternativo/genética , Ganglios Espinales/metabolismo , ARN sin Sentido/genética , ARN Mensajero/genética , Células Receptoras Sensoriales/metabolismo , Factores Complejos Ternarios/metabolismo , Animales , Axotomía/efectos adversos , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Modelos Animales de Enfermedad , Ganglios Espinales/citología , Regulación de la Expresión Génica/genética , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Regeneración Nerviosa/genética , Técnicas de Cultivo de Órganos , Traumatismos de los Nervios Periféricos , Nervios Periféricos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Células Receptoras Sensoriales/citología , Factores Complejos Ternarios/genética , Regulación hacia Arriba/genética , Proteína Elk-1 con Dominio ets/genética , Proteína Elk-1 con Dominio ets/metabolismo , Proteína Elk-4 del Dominio ets/genética , Proteína Elk-4 del Dominio ets/metabolismo
13.
Nat Commun ; 12(1): 6829, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34819513

RESUMEN

Signals are relayed from receptor tyrosine kinases (RTKs) at the cell surface to effector systems in the cytoplasm and nucleus, and coordination of this process is important for the execution of migratory phenotypes, such as cell scattering and invasion. The endosomal system influences how RTK signalling is coded, but the ways in which it transmits these signals to the nucleus to influence gene expression are not yet clear. Here we show that hepatocyte growth factor, an activator of MET (an RTK), promotes Rab17- and clathrin-dependent endocytosis of EphA2, another RTK, followed by centripetal transport of EphA2-positive endosomes. EphA2 then mediates physical capture of endosomes on the outer surface of the nucleus; a process involving interaction between the nuclear import machinery and a nuclear localisation sequence in EphA2's cytodomain. Nuclear capture of EphA2 promotes RhoG-dependent phosphorylation of the actin-binding protein, cofilin to oppose nuclear import of G-actin. The resulting depletion of nuclear G-actin drives transcription of Myocardin-related transcription factor (MRTF)/serum-response factor (SRF)-target genes to implement cell scattering and the invasive behaviour of cancer cells.


Asunto(s)
Núcleo Celular/metabolismo , Endosomas/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias/patología , Factores Complejos Ternarios/metabolismo , Actinas/metabolismo , Transporte Activo de Núcleo Celular/genética , Animales , Línea Celular Tumoral , Citoplasma/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Ratones , Ratones Noqueados , Invasividad Neoplásica/genética , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo
14.
FEBS J ; 288(10): 3120-3134, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32885587

RESUMEN

Serum response factor (SRF), a member of the Mcm1, Agamous, Deficiens, and SRF (MADS) box transcription factor, is widely expressed in all cell types and plays a crucial role in the physiological function and development of diseases. SRF regulates its downstream genes by binding to their CArG DNA box by interacting with various cofactors. However, the underlying mechanisms are not fully understood, therefore attracting increasing research attention due to the importance of this topic. This review's objective is to discuss the new progress in the studies of the molecular mechanisms involved in the activation of SRF and its impacts in physiological and pathological conditions. Notably, we summarized the recent studies on the interaction of SRF with its two main types of cofactors belonging to the myocardin families of transcription factors and the members of the ternary complex factors. The knowledge of these mechanisms will create new opportunities for understanding the dynamics of many traits and disease pathogenesis especially, cardiovascular diseases and cancer that could serve as targets for pharmacological control and treatment of these diseases.


Asunto(s)
Enfermedades Cardiovasculares/genética , Neoplasias/genética , Proteínas Nucleares/genética , Factor de Respuesta Sérica/genética , Factores Complejos Ternarios/genética , Transactivadores/genética , Transcripción Genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patología , Animales , Apoptosis/genética , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Proliferación Celular , ADN/genética , ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/metabolismo , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Estrés Fisiológico , Factores Complejos Ternarios/metabolismo , Transactivadores/metabolismo
15.
Mol Cell Biol ; 27(11): 4133-41, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17403894

RESUMEN

Hypoxia and the Net ternary complex factor (TCF) regulate similar processes (angiogenesis, wound healing, and cellular migration) and genes (PAI-1, c-fos, erg-1, NOS-2, HO-1, and vascular endothelial growth factor genes), suggesting that they are involved in related pathways. We show here that hypoxia regulates Net differently from the other TCFs and that Net plays a role in the hypoxic response in vivo in mice and in cells. Hypoxia induces Net depletion from target promoters, nuclear export, ubiquitylation, and proteasomal degradation. Key mediators of the hypoxic response, the prolyl-4-hydroxylases containing domain proteins (PHDs), regulate Net. PHD downregulation in normoxia leads to Net degradation, and PHD overexpression delays Net downregulation by hypoxia. Net inhibition by RNA interference or mutation leads to altered regulation by hypoxia of the Net targets PAI-1, c-fos, and egr-1. We propose that hypoxia stimulates transcription of target promoters through removal of the repressor function of Net. Interestingly, the hematocrit response to a chemical inducer of hypoxia-like responses (cobalt chloride) is strongly altered in Net mutant mice. Our results show that the Net TCF is part of the biological response to hypoxia, adding a new component to an important pathological and physiological process.


Asunto(s)
Regulación de la Expresión Génica , Hipoxia , Proteínas Proto-Oncogénicas c-ets/metabolismo , Factores Complejos Ternarios/metabolismo , Animales , Células Cultivadas , Regulación hacia Abajo , Humanos , Ratones , Mutación , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , ARN Interferente Pequeño/metabolismo , Factores Complejos Ternarios/genética , Ubiquitina/metabolismo
16.
Science ; 368(6489)2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32327570

RESUMEN

Protein quality control is essential for the proper function of cells and the organisms that they make up. The resulting loss of proteostasis, the processes by which the health of the cell's proteins is monitored and maintained at homeostasis, is associated with a wide range of age-related human diseases. Here, we highlight how the integrated stress response (ISR), a central signaling network that responds to proteostasis defects by tuning protein synthesis rates, impedes the formation of long-term memory. In addition, we address how dysregulated ISR signaling contributes to the pathogenesis of complex diseases, including cognitive disorders, neurodegeneration, cancer, diabetes, and metabolic disorders. The development of tools through which the ISR can be modulated promises to uncover new avenues to diminish pathologies resulting from it for clinical benefit.


Asunto(s)
Factor 2 Eucariótico de Iniciación/metabolismo , Proteostasis , Estrés Fisiológico , Factores Complejos Ternarios/metabolismo , Acetamidas/química , Acetamidas/farmacología , Animales , Ciclohexilaminas/química , Ciclohexilaminas/farmacología , Factor 2 Eucariótico de Iniciación/antagonistas & inhibidores , Humanos , Inmunidad , Enfermedades Metabólicas/metabolismo , Ratones , Neoplasias/metabolismo , Fosfotransferasas/metabolismo
17.
Dev Biol ; 316(2): 260-74, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18339368

RESUMEN

The Serum Response Factor (SRF) is an important regulator of cell proliferation and differentiation. Dictyostelium discoideum srfB gene codes for an SRF homologue and is expressed in vegetative cells and during development under the control of three alternative promoters, which show different cell-type specific patterns of expression. The two more proximal promoters directed gene transcription in prestalk AB, stalk and lower-cup cells. The generation of a strain where the srfB gene has been interrupted (srfB(-)) has shown that this gene is required for regulation of actin-cytoskeleton-related functions, such as cytokinesis and macropinocytosis. The mutant failed to develop well in suspension, but could be rescued by cAMP pulsing, suggesting a defect in cAMP signaling. srfB(-) cells showed impaired chemotaxis to cAMP and defective lateral pseudopodium inhibition. Nevertheless, srfB(-) cells aggregated on agar plates and nitrocellulose filters 2 h earlier than wild type cells, and completed development, showing an increased tendency to form slug structures. Analysis of wild type and srfB(-) strains detected significant differences in the regulation of gene expression upon starvation. Genes coding for lysosomal and ribosomal proteins, developmentally-regulated genes, and some genes coding for proteins involved in cytoskeleton regulation were deregulated during the first stages of development.


Asunto(s)
Dictyostelium/fisiología , Factores Complejos Ternarios/genética , Factores de Transcripción/genética , Actinas/metabolismo , Animales , Núcleo Celular/fisiología , Citocinesis/fisiología , Eliminación de Gen , Genes Reporteros , Pinocitosis/fisiología , Regiones Promotoras Genéticas , Factores Complejos Ternarios/metabolismo , Factores de Transcripción/metabolismo
18.
BMC Mol Biol ; 10: 40, 2009 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-19432968

RESUMEN

BACKGROUND: The serine protease thrombin catalyzes fibrin clot formation by converting fibrinogen into fibrin. Additionally, thrombin stimulation leads to an activation of stimulus-responsive transcription factors in different cell types, indicating that the gene expression pattern is changed in thrombin-stimulated cells. The objective of this study was to analyze the signaling cascade leading to the expression of the zinc finger transcription factor Egr-1 in thrombin-stimulated lung fibroblasts. RESULTS: Stimulation of 39M1-81 fibroblasts with thrombin induced a robust and transient biosynthesis of Egr-1. Reporter gene analysis revealed that the newly synthesized Egr-1 was biologically active. The signaling cascade connecting thrombin stimulation with Egr-1 gene expression required elevated levels of cytosolic Ca2+, the activation of diacylgycerol-dependent protein kinase C isoenzymes, and the activation of extracellular signal-regulated protein kinase (ERK). Stimulation of the cells with thrombin triggered the phosphorylation of the transcription factor Elk-1. Expression of a dominant-negative mutant of Elk-1 completely prevented Egr-1 expression in stimulated 39M1-81 cells, indicating that Elk-1 or related ternary complex factors connect the intracellular signaling cascade elicited by activation of protease-activated receptors with transcription of the Egr-1 gene. Lentiviral-mediated expression of MAP kinase phosphatase-1, a dual-specific phosphatase that dephosphorylates and inactivates ERK in the nucleus, prevented Elk-1 phosphorylation and Egr-1 biosynthesis in thrombin stimulated 39M1-81 cells, confirming the importance of nuclear ERK and Elk-1 for the upregulation of Egr-1 expression in thrombin-stimulated lung fibroblasts. 39M1-81 cells additionally express M1 muscarinic acetylcholine receptors. A comparison between the signaling cascades induced by thrombin or carbachol showed no differences, except that signal transduction via M1 muscarinic acetylcholine receptors required the transactivation of the EGF receptor, while thrombin signaling did not. CONCLUSION: This study shows that stimulus-transcription coupling in thrombin-treated lung fibroblasts relies on the elevation of the intracellular Ca2+-concentration and the activation of PKC and ERK. In the nucleus, ternary complex factors function as key proteins linking the intracellular signaling cascade with enhanced transcription of the Egr-1 gene. This study further shows that the dominant-negative Elk-1 mutant is a valuable tool to study Elk-1-mediated gene transcription.


Asunto(s)
Calcio/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibroblastos/metabolismo , Factores Complejos Ternarios/genética , Trombina/metabolismo , Activación Transcripcional , Regulación hacia Arriba , Animales , Cricetinae , Cricetulus , Citosol/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Expresión Génica , Humanos , Fosforilación , Transducción de Señal , Factores Complejos Ternarios/metabolismo , Trombina/genética
19.
Mol Cell Biol ; 26(11): 4134-48, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16705166

RESUMEN

The transcription factor serum response factor (SRF) interacts with its cofactor, MAL/MKL1, a member of the myocardin-related transcription factor (MRTF) family, through its DNA-binding domain. We define a seven-residue sequence within the conserved MAL B1 region essential and sufficient for complex formation. The neighboring Q-box sequence facilitates this interaction. The B1 and Q-box regions also have antagonistic effects on MAL nuclear import, but the residues involved are largely distinct. Both MAL and the ternary complex factor (TCF) family of SRF cofactors interact with a hydrophobic groove and pocket on the SRF DNA-binding domain. Unlike the TCFs, however, interaction of MAL with SRF is impaired by SRF alphaI-helix mutations that reduce DNA bending in the SRF-DNA complex. A clustered SRF alphaI-helix mutation strongly impairs MAL-SRF complex formation but does not affect DNA distortion in the MAL-SRF complex. MAL-SRF complex formation is facilitated by DNA binding. DNase I footprinting indicates that in the SRF-MAL complex MAL directly contacts DNA. These contacts, which flank the DNA sequences protected from DNase I by SRF, are required for effective MAL-SRF complex formation in gel mobility shift assays. We propose a model of MAL-SRF complex formation in which MAL interacts with SRF by the addition of a beta-strand to the SRF DNA-binding domain beta-sheet region, while SRF-induced DNA bending facilitates MAL-DNA contact.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factor de Respuesta Sérica/química , Factor de Respuesta Sérica/metabolismo , Factores Complejos Ternarios/metabolismo , Transactivadores/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Núcleo Celular/metabolismo , ADN/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Células 3T3 NIH , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
20.
Georgian Med News ; (168): 11-5, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19359711

RESUMEN

The stability of albumin-bilirubin complex was investigated depending on pH of solution. It was shown that the stability of complex increases in presence of Mn (II) ions. It was also investigated the paramagnetic composition of gallstones by the Electron Spin Resonance (ESR) method. It was found that all investigated gallstones contain a free bilirubin radical-the stable product of its radical oxidation. Accordingly the paramagnetic composition gallstones could be divided on three main types: cholesterol, brown pigment and black pigment stones. ESR spectra of cholesterol stones is singlet with g=2.003 and Delta H=1.0 mT. At the same time the brown gallstones, besides aforementioned signal contain the ESR spectrum which is characteristic for Mn(II) ion complexes with inorganic compounds and, finally, in the black pigment stones it was found out Fe(III) and Cu(II) complexes with organic compounds and a singlet of bilirubin free radical. It is supposed that crystallization centers of gallstones could be the polymer network of bilirubin radical polymerization in complex with different metal ions.


Asunto(s)
Albúminas/análisis , Bilirrubina/análisis , Espectroscopía de Resonancia por Spin del Electrón/métodos , Cálculos Biliares/metabolismo , Cálculos Biliares/patología , Manganeso/análisis , Factores Complejos Ternarios/metabolismo , Humanos , Concentración de Iones de Hidrógeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA