Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Clin Genet ; 99(5): 673-683, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33463715

RESUMEN

A young boy with multifocal epilepsy with infantile spasms and hypsarrhythmia with minimal organic lesions of brain structures underwent DNA diagnosis using whole-exome sequencing. A heterozygous amino-acid substitution p.L519R in a PHACTR1 gene was identified. PHACTR1 belongs to a protein family of G-actin binding protein phosphatase 1 (PP1) cofactors and was not previously associated with a human disease. The missense single nucleotide variant in the proband was shown to occur de novo in the paternal allele. The mutation was shown in vitro to reduce the affinity of PHACTR1 for G-actin, and to increase its propensity to form complexes with the catalytic subunit of PP1. These properties are associated with altered subcellular localization of PHACTR1 and increased ability to induce cytoskeletal rearrangements. Although the molecular role of the PHACTR1 in neuronal excitability and differentiation remains to be defined, PHACTR1 has been previously shown to be involved in Slack channelopathy pathogenesis, consistent with our findings. We conclude that this activating mutation in PHACTR1 causes a severe type of sporadic multifocal epilepsy in the patient.


Asunto(s)
Epilepsia/genética , Proteínas de Microfilamentos/genética , Mutación , Espasmos Infantiles/genética , Actinas/metabolismo , Animales , Preescolar , Humanos , Lactante , Masculino , Ratones , Células 3T3 NIH , Secuenciación del Exoma
2.
Elife ; 92020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32975518

RESUMEN

PPP-family phosphatases such as PP1 have little intrinsic specificity. Cofactors can target PP1 to substrates or subcellular locations, but it remains unclear how they might confer sequence-specificity on PP1. The cytoskeletal regulator Phactr1 is a neuronally enriched PP1 cofactor that is controlled by G-actin. Structural analysis showed that Phactr1 binding remodels PP1's hydrophobic groove, creating a new composite surface adjacent to the catalytic site. Using phosphoproteomics, we identified mouse fibroblast and neuronal Phactr1/PP1 substrates, which include cytoskeletal components and regulators. We determined high-resolution structures of Phactr1/PP1 bound to the dephosphorylated forms of its substrates IRSp53 and spectrin αII. Inversion of the phosphate in these holoenzyme-product complexes supports the proposed PPP-family catalytic mechanism. Substrate sequences C-terminal to the dephosphorylation site make intimate contacts with the composite Phactr1/PP1 surface, which are required for efficient dephosphorylation. Sequence specificity explains why Phactr1/PP1 exhibits orders-of-magnitude enhanced reactivity towards its substrates, compared to apo-PP1 or other PP1 holoenzymes.


Specific arrangements of atoms such as bulky phosphate groups can change the activity of a protein and how it interacts with other molecules. Enzymes called kinases are responsible for adding these groups onto a protein, while phosphatases remove them. Kinases are generally specific for a small number of proteins, adding phosphate groups only at sites embedded in a particular sequence in the target protein. Phosphatases, however, are generalists: only a few different types exist, which exhibit little target sequence specificity. Partner proteins can attach to phosphatases to bring the enzymes to specific locations in the cell, or to deliver target proteins to them; yet, it is unclear whether partner binding could also change the structure of the enzyme so the phosphatase can recognise only a restricted set of targets. To investigate this, Fedoryshchak, Prechová et al. studied a phosphatase called PP1 and its partner, Phactr1. First, the structure of the Phactr1/PP1 complex was examined using biochemistry approaches and X-ray crystallography. This showed that binding of Phactr1 to PP1 creates a new surface pocket, which comprised elements of both proteins. In particular, this composite pocket is located next to the part of the PP1 enzyme responsible for phosphate removal. Next, mass spectrometry and genetics methods were harnessed to identify and characterise the targets of the Phactr1/PP1 complex. Structural analysis of the proteins most susceptible to Phactr1/PP1 activity showed that they had particular sequences that could interact with Phactr1/PP1's composite pocket. Further experiments revealed that, compared to PP1 acting alone, the pocket increased the binding efficiency and reactivity of the complex 100-fold. This work demonstrates that a partner protein can make phosphatases more sequence-specific, suggesting that future studies could adopt a similar approach to examine how other enzymes in this family perform their role. In addition, the results suggest that it will be possible to design Phactr1/PP1-specific drugs that act on the composite pocket. This would represent an important proof of principle, since current phosphatase-specific drugs do not target particular phosphatase complexes.


Asunto(s)
Proteínas de Microfilamentos/metabolismo , Animales , Dominio Catalítico , Cristalización , Citoesqueleto/metabolismo , Holoenzimas/química , Holoenzimas/metabolismo , Ratones , Proteínas de Microfilamentos/química , Proteínas del Tejido Nervioso/metabolismo , Fosfatos/metabolismo , Conformación Proteica , Espectrina/metabolismo , Especificidad por Sustrato
3.
Nat Cell Biol ; 21(7): 845-855, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31209295

RESUMEN

RPEL proteins, which contain the G-actin-binding RPEL motif, coordinate cytoskeletal processes with actin dynamics. We show that the ArhGAP12- and ArhGAP32-family GTPase-activating proteins (GAPs) are RPEL proteins. We determine the structure of the ArhGAP12/G-actin complex, and show that G-actin contacts the RPEL motif and GAP domain sequences. G-actin inhibits ArhGAP12 GAP activity, and this requires the G-actin contacts identified in the structure. In B16 melanoma cells, ArhGAP12 suppresses basal Rac and Cdc42 activity, F-actin assembly, invadopodia formation and experimental metastasis. In this setting, ArhGAP12 mutants defective for G-actin binding exhibit more effective downregulation of Rac GTP loading following HGF stimulation and enhanced inhibition of Rac-dependent processes, including invadopodia formation. Potentiation or disruption of the G-actin/ArhGAP12 interaction, by treatment with the actin-binding drugs latrunculin B or cytochalasin D, has corresponding effects on Rac GTP loading. The interaction of G-actin with RPEL-family rhoGAPs thus provides a negative feedback loop that couples Rac activity to actin dynamics.


Asunto(s)
Actinas/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Actinas/efectos de los fármacos , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Citocalasina D/farmacología , Proteínas Activadoras de GTPasa/efectos de los fármacos , Proteínas Activadoras de GTPasa/genética , Guanosina Trifosfato/metabolismo , Humanos , Ratones , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Conejos , Tiazolidinas/farmacología , Proteína de Unión al GTP cdc42/efectos de los fármacos , Proteínas de Unión al GTP rac/metabolismo
4.
J Immunol ; 201(6): 1681-1691, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30068599

RESUMEN

In mouse thymocyte development, signaling by the TCR through the ERK pathway is required for positive selection of conventional naive T cells. The Ets transcription factor ELK4 (SAP-1), an ERK-regulated cofactor of the SRF transcription factor, plays an important role in positive selection by activating immediate-early genes such as the Egr transcription factor family. The role of ELK4-SRF signaling in development of other T cell types dependent on ERK signaling has been unclear. In this article, we show that ELK4, and its close relative ELK1, act cell autonomously in the thymus to control the generation of innate-like αß CD8+ T cells with memory-like characteristics. Mice lacking ELK4 and ELK1 develop increased numbers of innate-like αß CD8+ T cells, which populate the periphery. These cells develop cell autonomously rather than through expansion of PLZF+ thymocytes and concomitantly increased IL-4 signaling. Their development is associated with reduced TCR-mediated activation of ELK4-SRF target genes and can be partially suppressed by overexpression of the ELK4-SRF target gene EGR2. Consistent with this, partial inhibition of ERK signaling in peripheral CD8+T cells promotes the generation of cells with innate-like characteristics. These data establish that low-level ERK signaling through ELK4 (and ELK1) promotes innate-like αß CD8+ T cell differentiation, tuning conventional versus innate-like development.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Timo/inmunología , Proteína Elk-1 con Dominio ets/inmunología , Proteína Elk-4 del Dominio ets/inmunología , Animales , Linfocitos T CD8-positivos/citología , Diferenciación Celular/genética , Inmunidad Innata , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Timo/citología , Proteína Elk-1 con Dominio ets/genética , Proteína Elk-4 del Dominio ets/genética
5.
Sci Rep ; 7(1): 518, 2017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28364121

RESUMEN

The myocardin-related transcription factor/serum response factor (MRTF/SRF) pathway represents a promising therapeutic target to prevent fibrosis. We have tested the effects of new pharmacological inhibitors of MRTF/SRF signalling in a preclinical model of fibrosis. CCG-222740, a novel MRTF/SRF inhibitor, markedly decreased SRF reporter gene activity and showed a greater inhibitory effect on MRTF/SRF target genes than the previously described MRTF-A inhibitor CCG-203971. CCG-222740 was also five times more potent, with an IC50 of 5 µM, in a fibroblast-mediated collagen contraction assay, was less cytotoxic, and a more potent inhibitor of alpha-smooth muscle actin protein expression than CCG-203971. Local delivery of CCG-222740 and CCG-203971 in a validated and clinically relevant rabbit model of scar tissue formation after glaucoma filtration surgery increased the long-term success of the surgery by 67% (P < 0.0005) and 33% (P < 0.01), respectively, and significantly decreased fibrosis and scarring histologically. Unlike mitomycin-C, neither CCG-222740 nor CCG-203971 caused any detectable epithelial toxicity or systemic side effects with very low drug levels measured in the aqueous, vitreous, and serum. We conclude that inhibitors of MRTF/SRF-regulated gene transcription such as CCG-222740, potentially represent a new therapeutic strategy to prevent scar tissue formation in the eye and other tissues.


Asunto(s)
Cicatriz/metabolismo , Cicatriz/patología , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Respuesta Sérica/metabolismo , Transactivadores/antagonistas & inhibidores , Transactivadores/metabolismo , Animales , Células Cultivadas , Cicatriz/prevención & control , Colágeno/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Matriz Extracelular , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis , Humanos , Conejos , Transducción de Señal/efectos de los fármacos
6.
Mol Cell ; 65(6): 1081-1095.e5, 2017 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-28286024

RESUMEN

We investigated the relationship among ERK signaling, histone modifications, and transcription factor activity, focusing on the ERK-regulated ternary complex factor family of SRF partner proteins. In MEFs, activation of ERK by TPA stimulation induced a common pattern of H3K9acS10ph, H4K16ac, H3K27ac, H3K9acK14ac, and H3K4me3 at hundreds of transcription start site (TSS) regions and remote regulatory sites. The magnitude of the increase in histone modification correlated well with changes in transcription. H3K9acS10ph preceded the other modifications. Most induced changes were TCF dependent, but TCF-independent TSSs exhibited the same hierarchy, indicating that it reflects gene activation per se. Studies with TCF Elk-1 mutants showed that TCF-dependent ERK-induced histone modifications required Elk-1 to be phosphorylated and competent to activate transcription. Analysis of direct TCF-SRF target genes and chromatin modifiers confirmed this and showed that H3S10ph required only Elk-1 phosphorylation. Induction of histone modifications following ERK stimulation is thus directed by transcription factor activation and transcription.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/enzimología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Histonas/metabolismo , Factor de Respuesta Sérica/metabolismo , Factores de Transcripción TCF/metabolismo , Transcripción Genética , Animales , Línea Celular , Cromatina/efectos de los fármacos , Cromatina/genética , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Activación Enzimática , Ratones , Ratones Noqueados , Mutación , Fosforilación , Interferencia de ARN , Factor de Respuesta Sérica/genética , Transducción de Señal , Factores de Transcripción TCF/genética , Acetato de Tetradecanoilforbol/farmacología , Sitio de Iniciación de la Transcripción , Transcripción Genética/efectos de los fármacos , Transfección , Proteína Elk-1 con Dominio ets/genética , Proteína Elk-1 con Dominio ets/metabolismo
7.
Genes Dev ; 31(23-24): 2361-2375, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29317486

RESUMEN

Both the MRTF-SRF and the YAP-TEAD transcriptional regulatory networks respond to extracellular signals and mechanical stimuli. We show that the MRTF-SRF pathway is activated in cancer-associated fibroblasts (CAFs). The MRTFs are required in addition to the YAP pathway for CAF contractile and proinvasive properties. We compared MRTF-SRF and YAP-TEAD target gene sets and identified genes directly regulated by one pathway, the other, or both. Nevertheless, the two pathways exhibit mutual dependence. In CAFs, expression of direct MRTF-SRF genomic targets is also dependent on YAP-TEAD activity, and, conversely, YAP-TEAD target gene expression is also dependent on MRTF-SRF signaling. In normal fibroblasts, expression of activated MRTF derivatives activates YAP, while activated YAP derivatives activate MRTF. Cross-talk between the pathways requires recruitment of MRTF and YAP to DNA via their respective DNA-binding partners (SRF and TEAD) and is therefore indirect, arising as a consequence of activation of their target genes. In both CAFs and normal fibroblasts, we found that YAP-TEAD activity is sensitive to MRTF-SRF-induced contractility, while MRTF-SRF signaling responds to YAP-TEAD-dependent TGFß signaling. Thus, the MRF-SRF and YAP-TEAD pathways interact indirectly through their ability to control cytoskeletal dynamics.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Fibroblastos Asociados al Cáncer/fisiología , Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Neoplasias Mamarias Animales/fisiopatología , Fosfoproteínas/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Fosfoproteínas/genética , Transducción de Señal , Factores de Transcripción de Dominio TEA , Transactivadores/genética , Activación Transcripcional/genética , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Señalizadoras YAP
8.
Crystals (Basel) ; 7(8): 242, 2017 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-29456874

RESUMEN

The present article describes how to use the computer program BLEND to help assemble complete datasets for the solution of macromolecular structures, starting from partial or complete datasets, derived from data collection from multiple crystals. The program is demonstrated on more than two hundred X-ray diffraction datasets obtained from 50 crystals of a complex formed between the SRF transcription factor, its cognate DNA, and a peptide from the SRF cofactor MRTF-A. This structure is currently in the process of being fully solved. While full details of the structure are not yet available, the repeated application of BLEND on data from this structure, as they have become available, has made it possible to produce electron density maps clear enough to visualise the potential location of MRTF sequences.

9.
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
10.
Science ; 354(6309): 233-237, 2016 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-27738173

RESUMEN

Multisite phosphorylation regulates many transcription factors, including the serum response factor partner Elk-1. Phosphorylation of the transcriptional activation domain (TAD) of Elk-1 by the protein kinase ERK at multiple sites potentiates recruitment of the Mediator transcriptional coactivator complex and transcriptional activation, but the roles of individual phosphorylation events had remained unclear. Using time-resolved nuclear magnetic resonance spectroscopy, we found that ERK2 phosphorylation proceeds at markedly different rates at eight TAD sites in vitro, which we classified as fast, intermediate, and slow. Mutagenesis experiments showed that phosphorylation of fast and intermediate sites promoted Mediator interaction and transcriptional activation, whereas modification of slow sites counteracted both functions, thereby limiting Elk-1 output. Progressive Elk-1 phosphorylation thus ensures a self-limiting response to ERK activation, which occurs independently of antagonizing phosphatase activity.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Elk-1 con Dominio ets/metabolismo , Animales , Activación Enzimática , Regulación de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Mutagénesis , Resonancia Magnética Nuclear Biomolecular , Fosforilación , Dominios Proteicos , Factor de Respuesta Sérica/metabolismo , Proteína Elk-1 con Dominio ets/química , Proteína Elk-1 con Dominio ets/genética
11.
Elife ; 52016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27304076

RESUMEN

The myocardin-related transcription factors (MRTF-A and MRTF-B) regulate cytoskeletal genes through their partner transcription factor SRF. The MRTFs bind G-actin, and signal-regulated changes in cellular G-actin concentration control their nuclear accumulation. The MRTFs also undergo Rho- and ERK-dependent phosphorylation, but the function of MRTF phosphorylation, and the elements and signals involved in MRTF-A nuclear export are largely unexplored. We show that Rho-dependent MRTF-A phosphorylation reflects relief from an inhibitory function of nuclear actin. We map multiple sites of serum-induced phosphorylation, most of which are S/T-P motifs and show that S/T-P phosphorylation is required for transcriptional activation. ERK-mediated S98 phosphorylation inhibits assembly of G-actin complexes on the MRTF-A regulatory RPEL domain, promoting nuclear import. In contrast, S33 phosphorylation potentiates the activity of an autonomous Crm1-dependent N-terminal NES, which cooperates with five other NES elements to exclude MRTF-A from the nucleus. Phosphorylation thus plays positive and negative roles in the regulation of MRTF-A.


Asunto(s)
Procesamiento Proteico-Postraduccional , Transactivadores/metabolismo , Actinas/metabolismo , Animales , Línea Celular , Ratones , Fosforilación , Transcripción Genética , Proteínas de Unión al GTP rho/metabolismo
12.
EMBO J ; 34(11): 1554-71, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-25896507

RESUMEN

Oncogenic levels of Myc expression sensitize cells to multiple apoptotic stimuli, and this protects long-lived organisms from cancer development. How cells discriminate physiological from supraphysiological levels of Myc is largely unknown. Here, we show that induction of apoptosis by Myc in breast epithelial cells requires association of Myc with Miz1. Gene expression and ChIP-Sequencing experiments show that high levels of Myc invade target sites that lack consensus E-boxes in a complex with Miz1 and repress transcription. Myc/Miz1-repressed genes encode proteins involved in cell adhesion and migration and include several integrins. Promoters of repressed genes are enriched for binding sites of the serum-response factor (SRF). Restoring SRF activity antagonizes Myc repression of SRF target genes, attenuates Myc-induced apoptosis, and reverts a Myc-dependent decrease in Akt phosphorylation and activity, a well-characterized suppressor of Myc-induced apoptosis. We propose that high levels of Myc engage Miz1 in repressive DNA binding complexes and suppress an SRF-dependent transcriptional program that supports survival of epithelial cells.


Asunto(s)
Apoptosis/fisiología , Células Epiteliales/metabolismo , Glándulas Mamarias Humanas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factor de Respuesta Sérica/metabolismo , Transcripción Genética/fisiología , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Células Epiteliales/citología , Femenino , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Glándulas Mamarias Humanas/citología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Factor de Respuesta Sérica/genética
13.
Nat Commun ; 6: 5978, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25585691

RESUMEN

Controlled transport of macromolecules between the cytoplasm and nucleus is essential for homeostatic regulation of cellular functions. For instance, gene expression entails coordinated nuclear import of transcriptional regulators to activate transcription and nuclear export of the resulting messenger RNAs for cytoplasmic translation. Here we link these two processes by reporting a novel role for the mRNA export factor Ddx19/Dbp5 in nuclear import of MKL1, the signal-responsive transcriptional activator of SRF. We show that Ddx19 is not a general nuclear import factor, and that its specific effect on MKL1 nuclear import is separate from its role in mRNA export. Both helicase and nuclear pore-binding activities of Ddx19 are dispensable for MKL1 nuclear import, but RNA binding is required. Mechanistically, Ddx19 operates by modulating the conformation of MKL1, which affects its interaction with Importin-ß for efficient nuclear import. Thus, Ddx19 participates in mRNA export, translation and nuclear import of a key transcriptional regulator.


Asunto(s)
Transporte Activo de Núcleo Celular , ARN Helicasas DEAD-box/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Transactivadores/metabolismo , Animales , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células HeLa , Homeostasis , Humanos , Ratones , Microscopía Fluorescente , Células 3T3 NIH , Unión Proteica , Conformación Proteica , ARN/metabolismo , beta Carioferinas/metabolismo
14.
Blood ; 125(8): 1244-55, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25573994

RESUMEN

Chemokine signaling is important for the seeding of different sites by hematopoietic stem cells (HSCs) during development. Serum response factor (SRF) controls multiple genes governing adhesion and migration, mainly by recruiting members of the myocardin-related transcription factor (MRTF) family of G-actin-regulated cofactors. We used vav-iCre to inactivate MRTF-SRF signaling early during hematopoietic development. In both Srf- and Mrtf-deleted animals, hematopoiesis in fetal liver and spleen is intact but does not become established in fetal bone marrow. Srf-null HSC progenitor cells (HSC/Ps) fail to effectively engraft in transplantation experiments, exhibiting normal proximal signaling responses to SDF-1, but reduced adhesiveness, F-actin assembly, and reduced motility. Srf-null HSC/Ps fail to polarize in response to SDF-1 and cannot migrate through restrictive membrane pores to SDF-1 or Scf in vitro. Mrtf-null HSC/Ps were also defective in chemotactic responses to SDF-1. Srf-null HSC/Ps exhibit substantial deficits in cytoskeletal gene expression. MRTF-SRF signaling is thus critical for expression of genes required for the response to chemokine signaling during hematopoietic development.


Asunto(s)
Médula Ósea/embriología , Médula Ósea/fisiología , Células Madre Hematopoyéticas/fisiología , Factor de Respuesta Sérica/fisiología , Nicho de Células Madre , Transactivadores/fisiología , Factores de Transcripción/fisiología , Animales , Médula Ósea/crecimiento & desarrollo , Movimiento Celular/genética , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética , Nicho de Células Madre/genética
15.
Invest Ophthalmol Vis Sci ; 55(7): 4560-7, 2014 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-25056592

RESUMEN

Tissue contraction and fibrosis are major causes of morbidity in the human body. In the eye in particular, fibrosis and scarring are responsible for the pathogenesis or failure of treatment of all major blinding diseases, with postoperative wound healing responses posing a major problem for most ocular surgery on a worldwide scale. This is one of the largest areas of unmet need in ophthalmology, with currently no antifibrotic treatments available clinically. This review focuses on the ubiquitous myocardin-related transcription factor/serum response factor (MRTF-A/SRF) transcription pathway as a potential novel therapeutic target in fibrotic eye diseases. It describes how the MRTF-A/SRF pathway is intricately linked to all the key regulators and pathways in ocular fibrosis, and how it could potentially lead to a new avenue of antifibrotic therapies in the future.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Ojo/patología , Proteínas de Fusión Oncogénica/fisiología , Factor de Respuesta Sérica/fisiología , Ojo/enzimología , Fibrosis , Humanos , Metaloproteinasas de la Matriz/metabolismo , Transactivadores
16.
Genes Dev ; 28(9): 943-58, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24732378

RESUMEN

The transcription factor SRF (serum response factor) recruits two families of coactivators, the MRTFs (myocardin-related transcription factors) and the TCFs (ternary complex factors), to couple gene transcription to growth factor signaling. Here we investigated the role of the SRF network in the immediate transcriptional response of fibroblasts to serum stimulation. SRF recruited its cofactors in a gene-specific manner, and virtually all MRTF binding was directed by SRF. Much of SRF DNA binding was serum-inducible, reflecting a requirement for MRTF-SRF complex formation in nucleosome displacement. We identified 960 serum-responsive SRF target genes, which were mostly MRTF-controlled, as assessed by MRTF chromatin immunoprecipitation (ChIP) combined with deep sequencing (ChIP-seq) and/or sensitivity to MRTF-linked signals. MRTF activation facilitates RNA polymerase II (Pol II) recruitment or promoter escape according to gene context. MRTF targets encode regulators of the cytoskeleton, transcription, and cell growth, underpinning the role of SRF in cytoskeletal dynamics and mechanosensing. Finally, we show that specific activation of either MRTFs or TCFs can reset the circadian clock.


Asunto(s)
Actinas/metabolismo , Fibroblastos/fisiología , Suero/metabolismo , Transducción de Señal , Transcripción Genética/genética , Animales , Proteínas CLOCK/genética , Relojes Circadianos/genética , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Factor de Respuesta Sérica/metabolismo
18.
Trends Biochem Sci ; 38(8): 376-7, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23810602

RESUMEN

The functions of nuclear actin have been a mystery for many years. Recent papers demonstrate that the nuclear and cytoplasmic actin pools are in dynamic communication, but that not all nuclear actin freely exchanges. Extracellular signals can induce changes in nuclear actin dynamics, affecting activity of the myocardin-related transcription factor (MRTF) transcriptional coactivators, which reversibly bind G-actin. By contrast, actin is stably associated with the Ino80 chromatin remodelling complex, where it plays a role in the recognition of nucleosome linker DNA.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Núcleo Celular/metabolismo , Modelos Biológicos , Animales , Ensamble y Desensamble de Cromatina , Citoplasma/genética , Humanos , Transporte de Proteínas , Transducción de Señal
19.
Cell ; 152(3): 492-503, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23374345

RESUMEN

In peripheral tissues circadian gene expression can be driven either by local oscillators or by cyclic systemic cues controlled by the master clock in the brain's suprachiasmatic nucleus. In the latter case, systemic signals can activate immediate early transcription factors (IETFs) and thereby control rhythmic transcription. In order to identify IETFs induced by diurnal blood-borne signals, we developed an unbiased experimental strategy, dubbed Synthetic TAndem Repeat PROMoter (STAR-PROM) screening. This technique relies on the observation that most transcription factor binding sites exist at a relatively high frequency in random DNA sequences. Using STAR-PROM we identified serum response factor (SRF) as an IETF responding to oscillating signaling proteins present in human and rodent sera. Our data suggest that in mouse liver SRF is regulated via dramatic diurnal changes of actin dynamics, leading to the rhythmic translocation of the SRF coactivator Myocardin-related transcription factor-B (MRTF-B) into the nucleus.


Asunto(s)
Actinas/metabolismo , Ritmo Circadiano , Regulación de la Expresión Génica , Técnicas Genéticas , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Transporte Activo de Núcleo Celular , Animales , Proteínas Sanguíneas/análisis , Proteínas Sanguíneas/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Humanos , Masculino , Ratones , Proteínas Circadianas Period/metabolismo , Ratas , Factores de Transcripción/metabolismo
20.
Structure ; 20(11): 1960-70, 2012 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-23041370

RESUMEN

The Phactr family of PP1-binding proteins and the myocardin-related transcription factor family of transcriptional coactivators contain regulatory domains comprising three copies of the RPEL motif, a G-actin binding element. We report the structure of a Phactr1 G-actin⋅RPEL domain complex. Three G-actins surround the crank-shaped RPEL domain forming a closed helical assembly. Their spatial relationship is identical to the RPEL-actins within the pentavalent MRTF G-actin⋅RPEL domain complex, suggesting that conserved cooperative interactions between actin⋅RPEL units organize the assembly. In the trivalent Phactr1 complex, each G-actin⋅RPEL unit makes secondary contacts with its downstream actin involving distinct RPEL residues. Similar secondary contacts are seen in G-actin⋅RPEL peptide crystals. Loss-of-secondary-contact mutations destabilize the Phactr1 G-actin⋅RPEL assembly. Furthermore, actin-mediated inhibition of Phactr1 nuclear import requires secondary contact residues in the Phactr1 N-terminal RPEL-N motif, suggesting that it involves interaction of RPEL-N with the C-terminal assembly. Secondary actin contacts by actin-bound RPEL motifs thus govern formation of multivalent actin⋅RPEL assemblies.


Asunto(s)
Actinas/metabolismo , Proteínas de Microfilamentos/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas de Microfilamentos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...