Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Small GTPases ; 12(4): 273-281, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-32043900

RESUMEN

Malignant melanoma is characterized by mutations in a number of driver genes, most notably BRAF and NRAS. Recent genomic analyses revealed that 4-9% of sun-exposed melanomas bear activating mutations in RAC1, which encodes a small GTPase that is known to play key roles in cell proliferation, survival, and migration. The RAC1 protein activates several effector pathways, including Group A p21-activated kinases (PAKs), phosphoinositol-3-kinases (PI3Ks), in particular the beta isoform, and the serum-response factor/myocardin-related transcription factor (SRF/MRTF). Having previously shown that inhibition of Group A PAKs impedes oncogenic signalling from RAC1P29S, we here extend this analysis to examine the roles of PI3Ks and SRF/MRTF in melanocytes and/or in a zebrafish model. We demonstrate that a selective Group A PAK inhibitor (Frax-1036), a pan-PI3K (BKM120), and two PI3Kß inhibitors (TGX221, GSK2636771) impede the growth of melanoma cells driven by mutant RAC1 but not by mutant BRAF, while other PI3K selective inhibitors, including PI3Kα, δ and γ, are less effective. Using these compounds as well as an SRF/MRTF inhibitor (CCG-203,971), we observed similar results in vivo, using embryonic zebrafish development as a readout. These results suggest that targeting Group A PAKs, PI3Kß, and/or SRF/MRTF represent a promising approach to suppress RAC1 signalling in malignant melanoma.


Asunto(s)
Embrión no Mamífero/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Melanoma/tratamiento farmacológico , Mutación , Proteína de Unión al GTP rac1/genética , Animales , Apoptosis , Proliferación Celular , Embrión no Mamífero/metabolismo , Embrión no Mamífero/patología , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Fosfatidilinositol 3-Quinasas/química , Factor de Respuesta Sérica/antagonistas & inhibidores , Transducción de Señal , Transactivadores/antagonistas & inhibidores , Células Tumorales Cultivadas , Pez Cebra , Quinasas p21 Activadas/antagonistas & inhibidores
2.
Trends Cancer ; 6(6): 478-488, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32460002

RESUMEN

Small GTPases of the RAS and RHO families are related signaling proteins that, when activated by growth factors or by mutation, drive oncogenic processes. While activating mutations in KRAS, NRAS, and HRAS genes have long been recognized and occur in many types of cancer, similar mutations in RHO family genes, such as RAC1 and RHOA, have only recently been detected as the result of extensive cancer genome-sequencing efforts and are linked to a restricted set of malignancies. In this review, we focus on the role of RAC1 signaling in malignant melanoma, emphasizing recent advances that describe how this oncoprotein alters melanocyte proliferation and motility and how these findings might lead to new therapeutics in RAC1-mutant tumors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Melanoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Mutación con Ganancia de Función , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Melanoma/genética , Melanoma/patología , Fosfatidilinositol 3-Quinasa , Inhibidores de Proteínas Quinasas/uso terapéutico , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Respuesta Sérica/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Transactivadores/antagonistas & inhibidores , Transactivadores/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
3.
Cells ; 9(2)2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-32075310

RESUMEN

Although thousands of long noncoding RNAs (lncRNAs) have been identified in porcine growth and development, the regulation mechanisms of functional lncRNAs have not been well explored. In this study, using 5'- and 3'-rapid amplification of cDNA ends (RACE) assays, we obtained two different variants of lncRNA maternally expressed gene 3 (MEG3), namely, MEG3 v1 and MEG3 v2, that were both highly expressed in porcine skeletal muscle and in the early stage of the differentiation of porcine satellite cells. Moreover, we identified the core transcript MEG3 v2. Functional analyses showed that MEG3 overexpression could effectively arrest myoblasts in the G1 phase, inhibit DNA replication, and promote myoblast differentiation, whereas MEG3 knockdown resulted in the opposite effects. Interestingly, the expression of serum response factor (SRF), a crucial transcription factor for myogenesis process, remarkably increased and decreased in mRNA and protein levels with the respective overexpression and knockdown of MEG3. Dual luciferase reporter assay showed that MEG3 could attenuate the decrease of luciferase activity of SRF induced by miR-423-5p in a dose-dependent manner. MEG3 overexpression could relieve the inhibitory effect on SRF and myoblast differentiation induced by miR-423-5p. In addition, results of RNA immunoprecipitation analysis suggested that MEG3 could act as a ceRNA for miR-423-5p. Our findings initially established a novel connection among MEG3, miR-423-5p, and SRF in porcine satellite cell differentiation. This novel role of MEG3 may shed new light on understanding of molecular regulation of lncRNA in porcine myogenesis.


Asunto(s)
MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Factor de Respuesta Sérica/metabolismo , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Masculino , MicroARNs/genética , Desarrollo de Músculos , ARN Largo no Codificante/genética , Células Satélite del Músculo Esquelético/citología , Factor de Respuesta Sérica/antagonistas & inhibidores , Porcinos , Transfección
4.
Cell Death Dis ; 10(6): 453, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31186405

RESUMEN

Hepatocellular carcinoma (HCC) has a high mortality rate due to the lack of effective treatments and drugs. Arsenic trioxide (ATO), which has been proved to successfully treat acute promyelocytic leukemia (APL), was recently reported to show therapeutic potential in solid tumors including HCC. However, its anticancer mechanisms in HCC still need further investigation. In this study, we demonstrated that ATO inhibits tumorigenesis and distant metastasis in mouse models, corresponding with a prolonged mice survival time. Also, ATO was found to significantly decrease the cancer stem cell (CSC)-associated traits. Minichromosome maintenance protein (MCM) 7 was further identified to be a potential target suppressed dramatically by ATO, of which protein expression is increased in patients and significantly correlated with tumor size, cellular differentiation, portal venous emboli, and poor patient survival. Moreover, MCM7 knockdown recapitulates the effects of ATO on CSCs and metastasis, while ectopic expression of MCM7 abolishes them. Mechanistically, our results suggested that ATO suppresses MCM7 transcription by targeting serum response factor (SRF)/MCM7 complex, which functions as an important transcriptional regulator modulating MCM7 expression. Taken together, our findings highlight the importance of ATO in the treatment of solid tumors. The identification of SRF/MCM7 complex as a target of ATO provides new insights into ATO's mechanism, which may benefit the appropriate use of this agent in the treatment of HCC.


Asunto(s)
Antineoplásicos/farmacología , Trióxido de Arsénico/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Componente 7 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Células Madre Neoplásicas/metabolismo , Factor de Respuesta Sérica/metabolismo , Animales , Antineoplásicos/uso terapéutico , Trióxido de Arsénico/uso terapéutico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/secundario , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/genética , Ontología de Genes , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/mortalidad , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Componente 7 del Complejo de Mantenimiento de Minicromosoma/genética , Células Madre Neoplásicas/efectos de los fármacos , Pronóstico , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Respuesta Sérica/genética , Trasplante Heterólogo
5.
J Med Chem ; 62(9): 4350-4369, 2019 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-30951312

RESUMEN

Through a phenotypic high-throughput screen using a serum response element luciferase promoter, we identified a novel 5-aryl-1,3,4-oxadiazol-2-ylthiopropionic acid lead inhibitor of Rho/myocardin-related transcription factor (MRTF)/serum response factor (SRF)-mediated gene transcription with good potency (IC50 = 180 nM). We were able to rapidly improve the cellular potency by 5 orders of magnitude guided by sharply defined and synergistic SAR. The remarkable potency and depth of the SAR, as well as the relatively low molecular weight of the series, suggests, but does not prove, that binding to the unknown molecular target may be occurring through a covalent mechanism. The series nevertheless has no observable cytotoxicity up to 100 µM. Ensuing pharmacokinetic optimization resulted in the development of two potent and orally bioavailable anti-fibrotic agents that were capable of dose-dependently reducing connective tissue growth factor gene expression in vitro as well as significantly reducing the development of bleomycin-induced dermal fibrosis in mice in vivo.


Asunto(s)
Ácidos Carboxílicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Fibrosis/tratamiento farmacológico , Oxadiazoles/uso terapéutico , Factor de Respuesta Sérica/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores , Animales , Ácidos Carboxílicos/síntesis química , Ácidos Carboxílicos/farmacocinética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacocinética , Femenino , Fibrosis/patología , Ratones Endogámicos C57BL , Microsomas Hepáticos/efectos de los fármacos , Estructura Molecular , Oxadiazoles/síntesis química , Oxadiazoles/farmacocinética , Esclerodermia Sistémica/tratamiento farmacológico , Esclerodermia Sistémica/patología , Transducción de Señal/efectos de los fármacos , Piel/patología , Relación Estructura-Actividad , Transcripción Genética/efectos de los fármacos , Proteínas de Unión al GTP rho/antagonistas & inhibidores
6.
JCI Insight ; 4(6)2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30762586

RESUMEN

About one-third of dilated cardiomyopathy (DCM) cases are caused by mutations in sarcomere or cytoskeletal proteins. However, treating the cytoskeleton directly is not possible because drugs that bind to actin are not well tolerated. Mutations in the actin binding protein CAP2 can cause DCM and KO mice, either whole body (CAP2-KO) or cardiomyocyte-specific KOs (CAP2-CKO) develop DCM with cardiac conduction disease. RNA sequencing analysis of CAP2-KO hearts and isolated cardiomyocytes revealed overactivation of fetal genes, including serum response factor-regulated (SRF-regulated) genes such as Myl9 and Acta2 prior to the emergence of cardiac disease. To test if we could treat CAP2-KO mice, we synthesized and tested the SRF inhibitor CCG-1423-8u. CCG-1423-8u reduced expression of the SRF targets Myl9 and Acta2, as well as the biomarker of heart failure, Nppa. The median survival of CAP2-CKO mice was 98 days, while CCG-1423-8u-treated CKO mice survived for 116 days and also maintained normal cardiac function longer. These results suggest that some forms of sudden cardiac death and cardiac conduction disease are under cytoskeletal stress and that inhibiting signaling through SRF may benefit DCM by reducing cytoskeletal stress.


Asunto(s)
Anilidas/administración & dosificación , Benzamidas/administración & dosificación , Cardiomiopatía Dilatada/tratamiento farmacológico , Proteínas Portadoras/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Factor de Respuesta Sérica/antagonistas & inhibidores , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/mortalidad , Cardiomiopatía Dilatada/patología , Proteínas Portadoras/metabolismo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/patología , Modelos Animales de Enfermedad , Femenino , Feto , Corazón/efectos de los fármacos , Corazón/embriología , Humanos , Longevidad/efectos de los fármacos , Masculino , Ratones , Miocardio/citología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , RNA-Seq , Factor de Respuesta Sérica/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Factores de Transcripción/metabolismo
7.
Cell Physiol Biochem ; 51(2): 763-777, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30463073

RESUMEN

BACKGROUND/AIMS: Vascular muscularity is a key event in vessel remodeling during pulmonary artery hypertension (PAH). Endothelial-mesenchymal transdifferentiation (EndMT) has been increasingly reported to play a role in disease occurrence. Galectin-3, a carbohydrate-binding protein regulates cell proliferation, differentiation, migration and neovascularization. However, whether galectin-3 controls endothelial cell transdifferentiation during the development of PAH is unknown. METHODS: Rats were exposed to normoxic or hypoxic conditions (fraction of inspired O2 0.10) for 21 d to establish PAH models. Hemodynamic changes were evaluated through surgery of the right jugular vein and ultrasound biomicroscopy inviVue. And vessel pathological alterations were detected by H&E staining. Galectin-3 (Gal-3)-induced pulmonary artery endothelium cell (PAEC) dynamic alterations were measured by MTT assays, Cell immunofluorescence, Flow cytometry, Real-time PCR and Western blot. RESULTS: Our study demonstrated that Gal-3 was expressed in hypoxic pulmonary vascular adventitia and intima. The increased Gal-3 expression was responsible for hypoxic vessel remodeling and PAH development in vivo. Gal-3 was found to inhibit cell proliferation and apoptosis in cultured endothelial cells. Meanwhile endothelial cell morphology was altered and exhibited smooth muscle-like cell features as demonstrated by the expression of α-SMA after Gal-3 treatment. Gal-3 activated Jagged1/Notch1 pathways and induced MyoD and SRF. When MyoD or SRF were silenced with siRNAs, Gal-3-initiated transdifferentiation in endothelial cells was blocked as indicated by a lack of α-SMA. CONCLUSION: These results suggest that Gal-3 induces PAECs to acquire an α-SMA phenotype via a transdifferentiation process which depends on the activation of Jagged1/Notch1 pathways that mediate MyoD and SRF expression.


Asunto(s)
Transdiferenciación Celular , Galectina 3/metabolismo , Remodelación Vascular , Animales , Proteínas de Ciclo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/metabolismo , Galectina 3/antagonistas & inhibidores , Galectina 3/genética , Humanos , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Pulmón/metabolismo , Masculino , Proteína MioD/antagonistas & inhibidores , Proteína MioD/genética , Proteína MioD/metabolismo , Arteria Pulmonar/citología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Receptor Notch1/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/farmacología , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Remodelación Vascular/efectos de los fármacos
8.
J Nanobiotechnology ; 16(1): 97, 2018 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-30482196

RESUMEN

BACKGROUND: Sustained drug delivery is a large unmet clinical need in glaucoma. Here, we incorporated a Myocardin-Related Transcription Factor/Serum Response Factor inhibitor, CCG-222740, into slow release large unilamellar vesicles derived from the liposomes DOTMA (1,2-di-O-octadecenyl-3-trimethylammonium propane) and DOPC (1,2-dioleoyl-sn-glycero-3-phosphocholine), and tested their effects in vitro and in vivo. RESULTS: The vesicles were spherical particles of around 130 nm and were strongly cationic. A large amount of inhibitor could be incorporated into the vesicles. We showed that the nanocarrier CCG-222740 formulation gradually released the inhibitor over 14 days using high performance liquid chromatography. Nanocarrier CCG-222740 significantly decreased ACTA2 gene expression and was not cytotoxic in human conjunctival fibroblasts. In vivo, nanocarrier CCG-222740 doubled the bleb survival from 11.0 ± 0.6 days to 22.0 ± 1.3 days (p = 0.001), decreased conjunctival scarring and did not have any local or systemic adverse effects in a rabbit model of glaucoma filtration surgery. CONCLUSIONS: Our study demonstrates proof-of-concept that a nanocarrier-based formulation efficiently achieves a sustained release of a Myocardin-Related Transcription Factor/Serum Response Factor inhibitor and prevents conjunctival fibrosis in an established rabbit model of glaucoma filtration surgery.


Asunto(s)
Preparaciones de Acción Retardada/química , Sistemas de Liberación de Medicamentos , Factor de Respuesta Sérica/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Enfermedades de la Conjuntiva/tratamiento farmacológico , Femenino , Fibroblastos/efectos de los fármacos , Fibrosis/tratamiento farmacológico , Humanos , Liposomas/química , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Conejos , Distribución Tisular , Transactivadores/antagonistas & inhibidores , Transactivadores/química
9.
Prostate ; 78(5): 343-352, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29341215

RESUMEN

BACKGROUND: The ERK signaling pathway is frequently deregulated in tumorigenesis, mostly by classical mechanisms such as gene mutation of its components (eg, RAS and RAF). However, whether and how multiple key components of ERK pathway are regulated by microRNAs are not clear. METHODS: We firstly predicted post-transcriptional regulation of multiple key components of the ERK signaling pathway by miR181c through bioinformatics analysis, and then confirmed the post-transcriptional regulation by dual luciferase reporter gene assays and Western blot analysis. The biological effects of miR181c on prostate cancer cell proliferation, apoptosis, migration, and invasion were measured by CCK-8 assay, flow cytometry, wound scratch assay, transwell cell migration, and invasion assays. RESULTS: miR181c post-transcriptionally regulated multiple key members of the ERK signaling pathway, including extracellular signal-regulated kinase 2 (ERK2), ribosomal S6 kinase 2 (RSK2), serum response factor (SRF), and FBJ murine osteosarcoma viral oncogene homolog (c-Fos). Ectopic expression of miR181c mimics effectively suppressed prostate cancer cell proliferation, migration, and invasion, but promoted cell apoptosis. Furthermore, miR181c treatment combined with the multi-kinase inhibitor sorafenib significantly enhanced these anti-tumor effects. CONCLUSIONS: Downregulation of miR181c results in deregulated ERK signaling and promotes prostate cancer cell growth and metastasis.


Asunto(s)
Sistema de Señalización de MAP Quinasas , MicroARNs/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Masculino , MicroARNs/genética , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Invasividad Neoplásica , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Proteínas Quinasas S6 Ribosómicas 90-kDa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Respuesta Sérica/metabolismo , Sorafenib/farmacología
10.
Cell Physiol Biochem ; 44(2): 701-715, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29169155

RESUMEN

BACKGROUND/AIMS: Our previous studies demonstrated that intrinsic aortic smooth muscle cell (VSMC) stiffening plays a pivotal role in aortic stiffening in aging and hypertension. However, the underlying molecular mechanisms remain largely unknown. We here hypothesized that Rho kinase (ROCK) acts as a novel mediator that regulates intrinsic VSMC mechanical properties through the serum response factor (SRF) /myocardin pathway and consequently regulates aortic stiffness and blood pressure in hypertension. METHODS: Four-month old male spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats were studied. Aortic stiffness was measured by echography. Intrinsic mechanical properties of VSMCs were measured by atomic force microscopy (AFM) in vitro. RESULTS: Compared to WKY rats, SHR showed a significant increase in aortic stiffness and blood pressure, which is accompanied by a remarkable cell stiffening and ROCK activation in thoracic aortic (TA) VSMCs. Theses alterations in SHR were abolished by Y-27632, a specific inhibitor of ROCK. Additionally, boosted filamentous/globular actin ratio was detected in TA VSMCs from SHR versus WKY rats, resulting in an up-regulation of SRF and myocardin expression and its downstream stiffness-associated genes including α-smooth muscle actin, SM22, smoothelin and myosin heavy chain 11. Reciprocally, these alterations in SHR TA VSMCs were also suppressed by Y-27632. Furthermore, a specific inhibitor of SRF/myocardin, CCG-100602, showed a similar effect to Y-27632 in SHR in both TA VSMCs stiffness in vitro and aorta wall stiffness in vivo. CONCLUSION: ROCK is a novel mediator modulating aortic VSMC stiffness through SRF/myocardin signaling which offers a therapeutic target to reduce aortic stiffening in hypertension.


Asunto(s)
Actinas/metabolismo , Hipertensión/fisiopatología , Músculo Liso Vascular/fisiología , Proteínas Nucleares/metabolismo , Factor de Respuesta Sérica/metabolismo , Transactivadores/metabolismo , Quinasas Asociadas a rho/metabolismo , Amidas/farmacología , Animales , Aorta Torácica/citología , Aorta Torácica/fisiología , Presión Sanguínea , Proteínas del Citoesqueleto/metabolismo , Ecocardiografía , Hipertensión/veterinaria , Masculino , Microscopía de Fuerza Atómica , Proteínas Musculares/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Cadenas Pesadas de Miosina/metabolismo , Ácidos Nipecóticos/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Piridinas/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Factor de Respuesta Sérica/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores , Ultrasonografía , Regulación hacia Arriba , Rigidez Vascular/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores
11.
J Biol Chem ; 292(24): 10180-10196, 2017 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-28465353

RESUMEN

We have previously shown that dysbindin is a potent inducer of cardiomyocyte hypertrophy via activation of Rho-dependent serum-response factor (SRF) signaling. We have now performed a yeast two-hybrid screen using dysbindin as bait against a cardiac cDNA library to identify the cardiac dysbindin interactome. Among several putative binding proteins, we identified tripartite motif-containing protein 24 (TRIM24) and confirmed this interaction by co-immunoprecipitation and co-immunostaining. Another tripartite motif (TRIM) family protein, TRIM32, has been reported earlier as an E3 ubiquitin ligase for dysbindin in skeletal muscle. Consistently, we found that TRIM32 also degraded dysbindin in neonatal rat ventricular cardiomyocytes as well. Surprisingly, however, TRIM24 did not promote dysbindin decay but rather protected dysbindin against degradation by TRIM32. Correspondingly, TRIM32 attenuated the activation of SRF signaling and hypertrophy due to dysbindin, whereas TRIM24 promoted these effects in neonatal rat ventricular cardiomyocytes. This study also implies that TRIM32 is a key regulator of cell viability and apoptosis in cardiomyocytes via simultaneous activation of p53 and caspase-3/-7 and inhibition of X-linked inhibitor of apoptosis. In conclusion, we provide here a novel mechanism of post-translational regulation of dysbindin and hypertrophy via TRIM24 and TRIM32 and show the importance of TRIM32 in cardiomyocyte apoptosis in vitro.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Hipertrófica/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Asociadas a la Distrofina/metabolismo , Miocitos Cardíacos/metabolismo , Factor de Respuesta Sérica/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Cardiomiopatía Dilatada/patología , Cardiomiopatía Hipertrófica/patología , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Células Cultivadas , Disbindina , Proteínas Asociadas a la Distrofina/química , Proteínas Asociadas a la Distrofina/genética , Células HEK293 , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/patología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Estabilidad Proteica , Proteolisis , Interferencia de ARN , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Factor de Respuesta Sérica/agonistas , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Respuesta Sérica/genética , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/antagonistas & inhibidores , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética
12.
J Biol Chem ; 292(28): 11777-11791, 2017 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-28546428

RESUMEN

Megakaryoblastic leukemia (MKL)/serum-response factor (SRF)-mediated gene transcription is a highly conserved mechanism that connects dynamic reorganization of the actin cytoskeleton to regulation of expression of a wide range of genes, including SRF itself and many important structural and regulatory components of the actin cytoskeleton. In this study, we examined the possible role of MKL/SRF in the context of regulation of profilin (Pfn), a major controller of actin dynamics and actin cytoskeletal remodeling in cells. We demonstrated that despite being located on different genomic loci, two major isoforms of Pfn (Pfn1 and Pfn2) are co-regulated by a common mechanism involving the action of MKL that is independent of its SRF-related activity. We found that MKL co-regulates the expression of Pfn isoforms indirectly by modulating signal transducer and activator of transcription 1 (STAT1) and utilizing its SAP-domain function. Unexpectedly, our studies revealed that cellular externalization, rather than transcription of Pfn1, is affected by the perturbations of MKL. We further demonstrated that MKL can influence cell migration by modulating Pfn1 expression, indicating a functional connection between MKL and Pfn1 in actin-dependent cellular processes. Finally, we provide initial evidence supporting the ability of Pfn to influence MKL and SRF expression. Collectively, these findings suggest that Pfn may play a role in a possible feedback loop of the actin/MKL/SRF signaling circuit.


Asunto(s)
Regulación de la Expresión Génica , Profilinas/metabolismo , Factor de Transcripción STAT1/metabolismo , Transactivadores/metabolismo , Línea Celular Tumoral , Movimiento Celular , Células HEK293 , Humanos , Oligopéptidos/genética , Oligopéptidos/metabolismo , Profilinas/genética , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Interferencia de ARN , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/genética , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Transactivadores/antagonistas & inhibidores , Transactivadores/genética
13.
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
14.
J Mol Cell Cardiol ; 74: 127-38, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24839911

RESUMEN

Valvular interstitial cells (VICs), the fibroblast-like cellular constituents of aortic heart valves, maintain structural integrity of valve tissue. Activation into contractile myofibroblasts occurs under pathological situations and under standard cell culture conditions of isolated VICs. Reversal of this phenotype switch would be of major importance in respect to fibrotic valve diseases. In this investigation, we found that exogenous polyunsaturated fatty acids (PUFAs) decreased contractility and expression of myofibroblastic markers like α-smooth muscle actin (αSMA) in cultured VICs from porcine aortic valves. The most active PUFAs, docosahexaenoic acid (DHA) and arachidonic acid (AA) reduced the level of active RhoA and increased the G/F-actin ratio. The G-actin-regulated nuclear translocation of myocardin-related transcription factors (MRTFs), co-activators of serum response factor, was also reduced by DHA and AA. The same effects were observed after blocking RhoA directly with C3 transferase. In addition, increased contractility after induction of actin polymerisation with jasplakinolide and concomitant expression of αSMA were ameliorated by active PUFAs. Furthermore, reduced αSMA expression under PUFA exposure was observed in valve tissue explants demonstrating physiological relevance. In conclusion, RhoA/G-actin/MRTF signalling is operative in VICs, and this pathway can be partially blocked by certain PUFAs whereby the activation into the myofibroblastic phenotype is reversed.


Asunto(s)
Válvula Aórtica/efectos de los fármacos , Ácido Araquidónico/farmacología , Ácidos Docosahexaenoicos/farmacología , Miofibroblastos/efectos de los fármacos , ADP Ribosa Transferasas/farmacología , Actinas/genética , Actinas/metabolismo , Animales , Válvula Aórtica/citología , Válvula Aórtica/metabolismo , Toxinas Botulínicas/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Depsipéptidos/farmacología , Regulación de la Expresión Génica , Miofibroblastos/citología , Miofibroblastos/metabolismo , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Porcinos , Técnicas de Cultivo de Tejidos , Transactivadores/antagonistas & inhibidores , Transactivadores/genética , Transactivadores/metabolismo , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
15.
Inflamm Bowel Dis ; 20(1): 154-65, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24280883

RESUMEN

BACKGROUND: Ras homolog gene family, member A (RhoA)/Rho-associated coiled-coil forming protein kinase signaling is a key pathway in multiple types of solid organ fibrosis, including intestinal fibrosis. However, the pleiotropic effects of RhoA/Rho-associated coiled-coil forming protein kinase signaling have frustrated targeted drug discovery efforts. Recent recognition of the role of Rho-regulated gene transcription by serum response factor (SRF) and its transcriptional cofactor myocardin-related transcription factor A (MRTF-A) suggest a novel locus for pharmacological intervention. METHODS: Because RhoA signaling is mediated by both physical and biochemical stimuli, we examined whether pharmacological inhibition of RhoA or the downstream transcription pathway of MRTF-A/SRF could block intestinal fibrogenesis in 2 in vitro models. RESULTS: In this study, we demonstrate that inhibition of RhoA signaling blocks both matrix-stiffness and transforming growth factor beta-induced fibrogenesis in human colonic myofibroblasts. Repression of alpha-smooth muscle actin and collagen expression was associated with the inhibition of MRTF-A nuclear localization. CCG-1423, a first-generation Rho/MRTF/SRF pathway inhibitor, repressed fibrogenesis in both models, yet has unacceptable cytotoxicity. Novel second-generation inhibitors (CCG-100602 and CCG-203971) repressed both matrix-stiffness and transforming growth factor beta-mediated fibrogenesis as determined by protein and gene expression in a dose-dependent manner. CONCLUSIONS: Targeting the Rho/MRTF/SRF mechanism with second-generation Rho/MRTF/SRF inhibitors may represent a novel approach to antifibrotic therapeutics.


Asunto(s)
Anilidas/farmacología , Benzamidas/farmacología , Colon/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Fibrosis/prevención & control , Miofibroblastos/efectos de los fármacos , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Western Blotting , Adhesión Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Colon/metabolismo , Colon/patología , Proteínas de Unión al ADN/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Proteínas de Fusión Oncogénica/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Respuesta Sérica/metabolismo , Transducción de Señal/efectos de los fármacos , Transactivadores , Factor de Crecimiento Transformador beta/genética , Proteínas de Unión al GTP rho/metabolismo
16.
J Basic Clin Physiol Pharmacol ; 24(4): 287-97, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23893683

RESUMEN

BACKGROUND: Modulation of inflammatory signaling has been elucidated in several disease models. Acrolein, an environmental pollutant, has been linked to diseases such as atherosclerosis and to the inflammatory process involving nuclear factor κB (NFκB). Serum response factor (SRF), a transcription factor, regulates cell development, differentiation and proliferation through signaling molecules such as extracellular signal-regulated kinase 1/2 (ERK1/2) and CD36. We hypothesized that acrolein toxicity involves SRF in the process of activating NFκB and may involve CD36/ERK1/2. METHODS: Vascular smooth muscle cells (VSMCs) were exposed to acrolein (0.5 µg/mL) in the presence or absence of 10 nM QNZ (NFκB inhibitor), 300 nM CCG1423 (SRF inhibitor) and 50 µM PD98059 (ERK1/2 inhibitor). Protein and RNA were isolated. Changes in expression were determined by Western blot and polymerase chain reaction (PCR) array. RESULTS: Subtoxic doses of acrolein increased ERK1/2, SRF and NFκB protein expression, whereas CD36 expression was unchanged. Increase in NFκB expression was accompanied by an increase in activity. ERK1/2 inhibition only blunted SRF expression. SRF inhibition blunted NFκB expression but not that of ERK1/2. CD36 expression was unchanged in the presence of either inhibitor. PCR array analysis indicated up-regulation of nine genes (>4- to 50-fold) and down-regulation of six genes (>4- to 12-fold) involved in inflammatory signaling. CONCLUSIONS: We propose that SRF is required in acrolein activation of NFκB and is ERK1/2 dependent.


Asunto(s)
Acroleína/toxicidad , Contaminantes Ambientales/toxicidad , Inflamación/inducido químicamente , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Factor de Respuesta Sérica/inmunología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Inflamación/genética , Inflamación/inmunología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Músculo Liso Vascular/inmunología , Miocitos del Músculo Liso/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Ratas , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Respuesta Sérica/genética
17.
Bioorg Med Chem Lett ; 23(13): 3826-32, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23707258

RESUMEN

CCG-1423 (1) is a novel inhibitor of Rho/MKL1/SRF-mediated gene transcription that inhibits invasion of PC-3 prostate cancer cells in a Matrigel model of metastasis. We recently reported the design and synthesis of conformationally restricted analogs (e.g., 2) with improved selectivity for inhibiting invasion versus acute cytotoxicity. In this study we conducted a survey of aromatic substitution with the goal of improving physicochemical parameters (e.g., ClogP, MW) for future efficacy studies in vivo. Two new compounds were identified that attenuated cytotoxicity even further, and were fourfold more potent than 2 at inhibiting PC-3 cell migration in a scratch wound assay. One of these (8a, CCG-203971, IC50=4.2 µM) was well tolerated in mice for 5 days at 100mg/kg/day i.p., and was able to achieve plasma levels exceeding the migration IC50 for up to 3 h.


Asunto(s)
Amidas/farmacología , Antineoplásicos/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Ácidos Nipecóticos/farmacología , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Neoplasias de la Próstata/tratamiento farmacológico , Factor de Respuesta Sérica/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Amidas/síntesis química , Amidas/química , Anilidas/síntesis química , Anilidas/química , Anilidas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Benzamidas/síntesis química , Benzamidas/química , Benzamidas/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Masculino , Estructura Molecular , Metástasis de la Neoplasia/tratamiento farmacológico , Ácidos Nipecóticos/síntesis química , Ácidos Nipecóticos/química , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Relación Estructura-Actividad , Transactivadores , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
18.
Nucleic Acid Ther ; 23(1): 62-70, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23308381

RESUMEN

To treat urethral strictures of the lower urinary tract, urethrotomy is the method of choice. But this minimally invasive method suffers from poor outcome rates and leads often to restenosis of the urinary tract because of hyper-proliferating fibroblasts. Our aim is to minimize the proliferation of excessive tissue due to a new minimal invasive therapeutic approach. As an appropriate model, we isolated fibroblasts from different benign prostatic hyperplasia patients and transfected them with small interfering RNA (siRNA) against the transcription factor serum response factor (SRF), a key factor for cell cycle regulation and apoptosis. The resulting knockdown of SRF was examined on the messenger RNA level by quantitative real-time polymerase chain reaction and on the protein level by western blot. The correlation of SRF silencing and impact on cell proliferation was examined by xCELLigence, 5-bromo-2'-deoxiuridine proliferation assay, total cell counts, and senescence assay. The transfection of primary prostatic fibroblasts with SRFsiRNA revealed specific and significant knockdown of SRF, leading to significant inhibition of proliferation after the second transfection, which was revealed by proliferation assay and total cell number. The results of this study indicate a substantial role of SRF in prostatic fibroblasts and we suggest that SRF silencing might be used for the treatment of urethral strictures to achieve a durably patent urethra.


Asunto(s)
Hiperplasia Prostática/terapia , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Factor de Respuesta Sérica/antagonistas & inhibidores , Proliferación Celular , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Masculino , Hiperplasia Prostática/genética , Hiperplasia Prostática/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Respuesta Sérica/genética , Transfección
19.
Cancer Biother Radiopharm ; 28(2): 146-52, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23134219

RESUMEN

Serum response factor (SRF) is a transcription factor of the MADS box family. To date, DNA binding sites for SRF [serum response elements (SREs)] have been found in the promoters of approximately 50 different genes known to be involved in the regulation cell proliferation, differentiation, and apoptosis. Recent studies have indicated that SRF plays a role in the development of some tumors, including hepatocellular, thyroid, esophageal, and lung carcinomas. However, expression of SRF and its roles in gastric carcinoma are unclear. We found SRF to be highly expressed in human gastric carcinoma as well as ectopic or reduced expression for E-cadherin and ß-catenin. Blockage of SRF expression was found to inhibit proliferation, invasion, and migration. We also found that an inhibitor (Y-27632) of Rho-associated coiled kinase (ROCK1), a regulator of actin cytoskeleton that regulates cell adhesion, migration, and motility, suppressed SRF expression as well. These results demonstrate that SRF is involved in the aggressive behavior of gastric carcinoma cells. We also found that the inhibition of ROCK1 by Y-27632 can inhibit the invasion and migration of gastric cells done at least, in part, by attenuating SRF expression.


Asunto(s)
Apoptosis , Movimiento Celular , Factor de Respuesta Sérica/metabolismo , Neoplasias Gástricas/patología , Western Blotting , Cadherinas/metabolismo , Adhesión Celular , Diferenciación Celular , Proliferación Celular , Femenino , Citometría de Flujo , Mucosa Gástrica/metabolismo , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Estudios Retrospectivos , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Respuesta Sérica/genética , Estómago/patología , Neoplasias Gástricas/metabolismo , Células Tumorales Cultivadas , Cicatrización de Heridas , beta Catenina/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
20.
Nat Neurosci ; 13(9): 1082-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20694003

RESUMEN

It has been suggested that gene expression and protein synthesis are required for both long-term memory consolidation and late phases of long-term potentiation and long-term depression (LTD). The necessary genes and the specific transcription factor binding sites in their promoters remain unknown. We found that inhibition of the transcription factor SRF or its cofactor MAL blocked the late phase of LTD in mouse cultured cerebellar Purkinje cells, as did deletion of the immediate early gene Arc. Using neuronal bacterial artificial chromosome (BAC) transfection, we found that, in Arc-/- cells transfected with a wild-type Arc BAC, late-phase LTD was rescued. However, mutation of one SRF-binding site in the Arc promoter (SRE 6.9) blocked this rescue. Co-transfection of wild-type Arc and SRF engineered to bind mutated SRE 6.9 restored late-phase LTD in Arc-/-, SRE 6.9 mutant BAC cells. Thus, SRF binding to SRE 6.9 in the Arc promoter is required for the late phase of cerebellar LTD.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Depresión Sináptica a Largo Plazo/fisiología , Proteínas del Tejido Nervioso/metabolismo , Células de Purkinje/fisiología , Factor de Respuesta Sérica/metabolismo , Animales , Células Cultivadas , Proteínas del Citoesqueleto/genética , Depresión Sináptica a Largo Plazo/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/genética , Regiones Promotoras Genéticas , Factor de Respuesta Sérica/antagonistas & inhibidores , Factores de Tiempo , Transactivadores/antagonistas & inhibidores , Transactivadores/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...