Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 33(10): 1117-33, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24596247

RESUMEN

Macrophages contribute to tissue homeostasis and influence inflammatory responses by modulating their phenotype in response to the local environment. Understanding the molecular mechanisms governing this plasticity would open new avenues for the treatment for inflammatory disorders. We show that deletion of calcineurin (CN) or its inhibition with LxVP peptide in macrophages induces an anti-inflammatory population that confers resistance to arthritis and contact hypersensitivity. Transfer of CN-targeted macrophages or direct injection of LxVP-encoding lentivirus has anti-inflammatory effects in these models. Specific CN targeting in macrophages induces p38 MAPK activity by downregulating MKP-1 expression. However, pharmacological CN inhibition with cyclosporin A (CsA) or FK506 did not reproduce these effects and failed to induce p38 activity. The CN-inhibitory peptide VIVIT also failed to reproduce the effects of LxVP. p38 inhibition prevented the anti-inflammatory phenotype of CN-targeted macrophages, and mice with defective p38-activation were resistant to the anti-inflammatory effect of LxVP. Our results identify a key role for CN and p38 in the modulation of macrophage phenotype and suggest an alternative treatment for inflammation based on redirecting macrophages toward an anti-inflammatory status.


Asunto(s)
Calcineurina/metabolismo , Fosfatasa 1 de Especificidad Dual/metabolismo , Macrófagos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Calcineurina/genética , Diferenciación Celular , Línea Celular , Células Cultivadas , Fosfatasa 1 de Especificidad Dual/genética , Citometría de Flujo , Humanos , Immunoblotting , Inmunohistoquímica , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Osteoclastos/citología , Osteoclastos/metabolismo , Fagocitosis/genética , Fagocitosis/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Quinasas p38 Activadas por Mitógenos/genética
2.
Mol Cell Proteomics ; 15(5): 1740-60, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26893027

RESUMEN

The coordinated behavior of proteins is central to systems biology. However, the underlying mechanisms are poorly known and methods to analyze coordination by conventional quantitative proteomics are still lacking. We present the Systems Biology Triangle (SBT), a new algorithm that allows the study of protein coordination by pairwise quantitative proteomics. The Systems Biology Triangle detected statistically significant coordination in diverse biological models of very different nature and subjected to different kinds of perturbations. The Systems Biology Triangle also revealed with unprecedented molecular detail an array of coordinated, early protein responses in vascular smooth muscle cells treated at different times with angiotensin-II. These responses included activation of protein synthesis, folding, turnover, and muscle contraction - consistent with a differentiated phenotype-as well as the induction of migration and the repression of cell proliferation and secretion. Remarkably, the majority of the altered functional categories were protein complexes, interaction networks, or metabolic pathways. These changes could not be detected by other algorithms widely used by the proteomics community, and the vast majority of proteins involved have not been described before to be regulated by AngII. The unique capabilities of The Systems Biology Triangle to detect functional protein alterations produced by the coordinated action of proteins in pairwise quantitative proteomics experiments make this algorithm an attractive choice for the biological interpretation of results on a routine basis.


Asunto(s)
Proteoma/análisis , Proteómica/métodos , Biología de Sistemas/métodos , Algoritmos , Animales , Ensayos Analíticos de Alto Rendimiento , Humanos , Mapas de Interacción de Proteínas
3.
J Mol Cell Cardiol ; 109: 38-47, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28684310

RESUMEN

AIMS: Ischaemic cardiovascular disease is a major cause of morbidity and mortality worldwide. Despite promising results from pre-clinical animal models, VEGF-based strategies for therapeutic angiogenesis have yet to achieve successful reperfusion of ischaemic tissues in patients. Failure to restore efficient VEGF activity in the ischaemic organ remains a major problem in current pro-angiogenic therapeutic approaches. Plasma membrane calcium ATPase 4 (PMCA4) negatively regulates VEGF-activated angiogenesis via inhibition of the calcineurin/NFAT signalling pathway. PMCA4 activity is inhibited by the small molecule aurintricarboxylic acid (ATA). We hypothesize that inhibition of PMCA4 with ATA might enhance VEGF-induced angiogenesis. METHODS AND RESULTS: We show that inhibition of PMCA4 with ATA in endothelial cells triggers a marked increase in VEGF-activated calcineurin/NFAT signalling that translates into a strong increase in endothelial cell motility and blood vessel formation. ATA enhances VEGF-induced calcineurin signalling by disrupting the interaction between PMCA4 and calcineurin at the endothelial-cell membrane. ATA concentrations at the nanomolar range, that efficiently inhibit PMCA4, had no deleterious effect on endothelial-cell viability or zebrafish embryonic development. However, high ATA concentrations at the micromolar level impaired endothelial cell viability and tubular morphogenesis, and were associated with toxicity in zebrafish embryos. In mice undergoing experimentally-induced hindlimb ischaemia, ATA treatment significantly increased the reperfusion of post-ischaemic limbs. CONCLUSIONS: Our study provides evidence for the therapeutic potential of targeting PMCA4 to improve VEGF-based pro-angiogenic interventions. This goal will require the development of refined, highly selective versions of ATA, or the identification of novel PMCA4 inhibitors.


Asunto(s)
ATPasas Transportadoras de Calcio/metabolismo , Membrana Celular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Ácido Aurintricarboxílico/farmacología , ATPasas Transportadoras de Calcio/genética , Membrana Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Ratones , Ratones Noqueados , Neovascularización Fisiológica/efectos de los fármacos , Pez Cebra/genética , Proteínas de Pez Cebra/genética
4.
Mol Cell ; 33(5): 616-26, 2009 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-19285944

RESUMEN

The phosphatase calcineurin, a target of the immunosuppressants cyclosporin A and FK506, dephosphorylates NFAT transcription factors to promote immune activation and development of the vascular and nervous systems. NFAT interacts with calcineurin through distinct binding motifs: the PxIxIT and LxVP sites. Although many calcineurin substrates contain PxIxIT motifs, the generality of LxVP-mediated interactions is unclear. We define critical residues in the LxVP motif, and we demonstrate its binding to a hydrophobic pocket at the interface of the two calcineurin subunits. Mutations in this region disrupt binding of mammalian calcineurin to NFATC1 and the interaction of yeast calcineurin with substrates including Rcn1, which contains an LxVP motif. These mutations also interfere with calcineurin-immunosuppressant binding, and an LxVP-based peptide competes with immunosuppressant-immunophilin complexes for binding to calcineurin. These studies suggest that LxVP-type sites are a common feature of calcineurin substrates, and that immunosuppressant-immunophilin complexes inhibit calcineurin by interfering with this mode of substrate recognition.


Asunto(s)
Calcineurina/metabolismo , Inmunosupresores/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Calcineurina/química , Calcineurina/genética , Inhibidores de la Calcineurina , Clonación Molecular , Simulación por Computador , Secuencia Conservada , Genes Reporteros , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Inmunofilinas/metabolismo , Inmunosupresores/farmacología , Péptidos y Proteínas de Señalización Intracelular , Células Jurkat , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Factores de Transcripción NFATC/metabolismo , Péptidos/metabolismo , Conformación Proteica , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transducción de Señal , Propiedades de Superficie , Proteína 1A de Unión a Tacrolimus/metabolismo , Transcripción Genética , Transfección
5.
Carcinogenesis ; 36(7): 792-9, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25916653

RESUMEN

The members of the human regulators of calcineurin (RCAN) protein family are endogenous regulators of the calcineurin (CN)-cytosolic nuclear factor of activated T-cells (NFATc) pathway activation. This function is explained by the presence of a highly conserved calcipressin inhibitor of calcineurin (CIC) motif in RCAN proteins, which has been shown to compete with NFATc for the binding to CN and therefore are able to inhibit NFATc dephosphorylation and activation by CN. Very recently, emerging roles for NFATc proteins in transformation, tumor angiogenesis and metastasis have been described in different cancer cell types. In this work, we report that the overexpression of RCAN3 dramatically inhibits tumor growth and tumor angiogenesis in an orthotopic human breast cancer model. We suggest that RCAN3 exerts these effects in a CN-dependent manner, as mutation of the CIC motif in RCAN3 abolishes the tumor suppressor effect. Moreover, the expression of the EGFP-R3(178-210) peptide, spanning the CIC motif of RCAN3, is able to reproduce all the antitumor effects of RCAN3 full-length protein. Finally, we show that RCAN3 and the EGFP-R3(178-210) peptide inhibit the CN-NFATc signaling pathway and the induction of the NFATc-dependent gene cyclooxygenase-2. Our work suggests that the EGFP-R3(178-210) peptide possess potent tumor suppressor properties and therefore constitutes a novel lead for the development of potent and specific antitumoral agents. Moreover, we propose the targeting of the CN-NFATc pathway in the tumor cells constitutes an effective way to hamper tumor progression by impairing the paracrine network among tumor, endothelial and polymorphonucleated cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Fragmentos de Péptidos/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Calcineurina/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Factores de Transcripción NFATC , Neovascularización Patológica/genética , Fragmentos de Péptidos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Arterioscler Thromb Vasc Biol ; 34(10): 2310-20, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25147342

RESUMEN

OBJECTIVE: Vascular endothelial growth factor (VEGF) has been identified as a crucial regulator of physiological and pathological angiogenesis. Among the intracellular signaling pathways triggered by VEGF, activation of the calcineurin/nuclear factor of activated T cells (NFAT) signaling axis has emerged as a critical mediator of angiogenic processes. We and others previously reported a novel role for the plasma membrane calcium ATPase (PMCA) as an endogenous inhibitor of the calcineurin/NFAT pathway, via interaction with calcineurin, in cardiomyocytes and breast cancer cells. However, the functional significance of the PMCA/calcineurin interaction in endothelial pathophysiology has not been addressed thus far. APPROACH AND RESULTS: Using in vitro and in vivo assays, we here demonstrate that the interaction between PMCA4 and calcineurin in VEGF-stimulated endothelial cells leads to downregulation of the calcineurin/NFAT pathway and to a significant reduction in the subsequent expression of the NFAT-dependent, VEGF-activated, proangiogenic genes RCAN1.4 and Cox-2. PMCA4-dependent inhibition of calcineurin signaling translates into a reduction in endothelial cell motility and blood vessel formation that ultimately impairs in vivo angiogenesis by VEGF. CONCLUSIONS: Given the importance of the calcineurin/NFAT pathway in the regulation of pathological angiogenesis, targeted modulation of PMCA4 functionality might open novel therapeutic avenues to promote or attenuate new vessel formation in diseases that occur with angiogenesis.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Calcineurina/metabolismo , ATPasas Transportadoras de Calcio/metabolismo , Células Endoteliales/efectos de los fármacos , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Proteínas de Unión al Calcio , ATPasas Transportadoras de Calcio/deficiencia , ATPasas Transportadoras de Calcio/genética , Movimiento Celular , Proliferación Celular , Ciclooxigenasa 2/metabolismo , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Células HEK293 , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isquemia/enzimología , Isquemia/fisiopatología , Ratones , Ratones Noqueados , Proteínas Musculares/metabolismo , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección
7.
EMBO Mol Med ; 16(1): 132-157, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38177536

RESUMEN

Thoracic aortic aneurysm and dissection (TAAD) is a life-threatening condition associated with Marfan syndrome (MFS), a disease caused by fibrillin-1 gene mutations. While various conditions causing TAAD exhibit aortic accumulation of the proteoglycans versican (Vcan) and aggrecan (Acan), it is unclear whether these ECM proteins are involved in aortic disease. Here, we find that Vcan, but not Acan, accumulated in Fbn1C1041G/+ aortas, a mouse model of MFS. Vcan haploinsufficiency protected MFS mice against aortic dilation, and its silencing reverted aortic disease by reducing Nos2 protein expression. Our results suggest that Acan is not an essential contributor to MFS aortopathy. We further demonstrate that Vcan triggers Akt activation and that pharmacological Akt pathway inhibition rapidly regresses aortic dilation and Nos2 expression in MFS mice. Analysis of aortic tissue from MFS human patients revealed accumulation of VCAN and elevated pAKT-S473 staining. Together, these findings reveal that Vcan plays a causative role in MFS aortic disease in vivo by inducing Nos2 via Akt activation and identify Akt signaling pathway components as candidate therapeutic targets.


Asunto(s)
Aneurisma de la Aorta Torácica , Enfermedades de la Aorta , Disección Aórtica , Azidas , Desoxiglucosa , Síndrome de Marfan , Animales , Humanos , Ratones , Aneurisma de la Aorta Torácica/complicaciones , Aneurisma de la Aorta Torácica/genética , Aneurisma de la Aorta Torácica/metabolismo , Enfermedades de la Aorta/complicaciones , Desoxiglucosa/análogos & derivados , Síndrome de Marfan/complicaciones , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Versicanos/metabolismo
8.
Blood ; 118(3): 795-803, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21642596

RESUMEN

The nuclear factor of activated T cells (NFAT) family of transcription factors plays important roles in many biologic processes, including the development and function of the immune and vascular systems. Cells usually express more than one NFAT member, raising the question of whether NFATs play overlapping roles or if each member has selective functions. Using mRNA knock-down, we show that NFATc3 is specifically required for IL2 and cyclooxygenase-2 (COX2) gene expression in transformed and primary T cells and for T-cell proliferation. We also show that NFATc3 regulates COX2 in endothelial cells, where it is required for COX2, dependent migration and angiogenesis in vivo. These results indicate that individual NFAT members mediate specific functions through the differential regulation of the transcription of target genes. These effects, observed on short-term suppression by mRNA knock-down, are likely to have been masked by compensatory effects in gene-knockout studies.


Asunto(s)
Ciclooxigenasa 2/genética , Interleucina-2/genética , Activación de Linfocitos/fisiología , Factores de Transcripción NFATC , Neovascularización Fisiológica/fisiología , Linfocitos T/fisiología , División Celular/fisiología , Movimiento Celular/fisiología , Ciclooxigenasa 2/inmunología , Células Endoteliales/citología , Células Endoteliales/fisiología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Interleucina-2/inmunología , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/inmunología , Factores de Transcripción NFATC/metabolismo , Regiones Promotoras Genéticas/fisiología , Transcripción Genética/fisiología
9.
Mol Cell Proteomics ; 10(1): M110.003335, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20807836

RESUMEN

MS-based quantitative proteomics plays an increasingly important role in biological and medical research and the development of these techniques remains one of the most important challenges in mass spectrometry. Numerous stable isotope labeling approaches have been proposed. However, and particularly in the case of (18)O-labeling, a standard protocol of general applicability is still lacking, and statistical issues associated to these methods remain to be investigated. In this work we present an improved high-throughput quantitative proteomics method based on whole proteome concentration by SDS-PAGE, optimized in-gel digestion, peptide (18)O-labeling, and separation by off-gel isoelectric focusing followed by liquid chromatography-LIT-MS. We demonstrate that the off-gel technique is fully compatible with (18)O peptide labeling in any pH range. A recently developed statistical model indicated that partial digestions and methionine oxidation do not alter protein quantification and that variances at the scan, peptide, and protein levels are stable and reproducible in a variety of proteomes of different origin. We have also analyzed the dynamic range of quantification and demonstrated the practical utility of the method by detecting expression changes in a model of activation of Jurkat T-cells. Our protocol provides a general approach to perform quantitative proteomics by (18)O-labeling in high-throughput studies, with the added value that it has a validated statistical model for the null hypothesis. To the best of our knowledge, this is the first report where a general protocol for stable isotope labeling is tested in practice using a collection of samples and analyzed at this degree of statistical detail.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Marcaje Isotópico/métodos , Proteoma/análisis , Proteómica/métodos , Análisis de Varianza , Animales , Línea Celular Tumoral , Fraccionamiento Químico , Citoplasma/metabolismo , Electroforesis en Gel de Poliacrilamida , Humanos , Focalización Isoeléctrica , Metionina/metabolismo , Proteínas de Neoplasias/metabolismo , Oxidación-Reducción , Isótopos de Oxígeno , Péptidos/análisis , Ratas
10.
Proc Natl Acad Sci U S A ; 106(15): 6117-22, 2009 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-19332797

RESUMEN

Specificity of signaling kinases and phosphatases toward their targets is usually mediated by docking interactions with substrates and regulatory proteins. Here, we characterize the motifs involved in the physical and functional interaction of the phosphatase calcineurin with a group of modulators, the RCAN protein family. Mutation of key residues within the hydrophobic docking-cleft of the calcineurin catalytic domain impairs binding to all human RCAN proteins and to the calcineurin interacting proteins Cabin1 and AKAP79. A valine-rich region within the RCAN carboxyl region is essential for binding to the docking site in calcineurin. Although a peptide containing this sequence compromises NFAT signaling in living cells, it does not inhibit calcineurin catalytic activity directly. Instead, calcineurin catalytic activity is inhibited by a motif at the extreme C-terminal region of RCAN, which acts in cis with the docking motif. Our results therefore indicate that the inhibitory action of RCAN on calcineurin-NFAT signaling results not only from the inhibition of phosphatase activity but also from competition between NFAT and RCAN for binding to the same docking site in calcineurin. Thus, competition by substrates and modulators for a common docking site appears to be an essential mechanism in the regulation of Ca(2+)-calcineurin signaling.


Asunto(s)
Calcineurina/química , Calcineurina/metabolismo , Proteínas Musculares/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Calcineurina/genética , Línea Celular , Secuencia Conservada , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Datos de Secuencia Molecular , Proteínas Musculares/química , Proteínas Musculares/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Estructura Secundaria de Proteína , Alineación de Secuencia , Transducción de Señal , Especificidad por Sustrato
11.
Nat Metab ; 4(7): 901-917, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35879461

RESUMEN

Early-life determinants are thought to be a major factor in the rapid increase of obesity. However, while maternal nutrition has been extensively studied, the effects of breastfeeding by the infant on the reprogramming of energy balance in childhood and throughout adulthood remain largely unknown. Here we show that delayed weaning in rat pups protects them against diet-induced obesity in adulthood, through enhanced brown adipose tissue thermogenesis and energy expenditure. In-depth metabolic phenotyping in this rat model as well as in transgenic mice reveals that the effects of prolonged suckling are mediated by increased hepatic fibroblast growth factor 21 (FGF21) production and tanycyte-controlled access to the hypothalamus in adulthood. Specifically, FGF21 activates GABA-containing neurons expressing dopamine receptor 2 in the lateral hypothalamic area and zona incerta. Prolonged breastfeeding thus constitutes a protective mechanism against obesity by affecting long-lasting physiological changes in liver-to-hypothalamus communication and hypothalamic metabolic regulation.


Asunto(s)
Lactancia Materna , Obesidad , Animales , Femenino , Factores de Crecimiento de Fibroblastos , Humanos , Hipotálamo/metabolismo , Hígado/metabolismo , Ratones , Obesidad/metabolismo , Obesidad/prevención & control , Ratas
12.
Nat Commun ; 12(1): 2628, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976159

RESUMEN

Thoracic aortic aneurysm, as occurs in Marfan syndrome, is generally asymptomatic until dissection or rupture, requiring surgical intervention as the only available treatment. Here, we show that nitric oxide (NO) signaling dysregulates actin cytoskeleton dynamics in Marfan Syndrome smooth muscle cells and that NO-donors induce Marfan-like aortopathy in wild-type mice, indicating that a marked increase in NO suffices to induce aortopathy. Levels of nitrated proteins are higher in plasma from Marfan patients and mice and in aortic tissue from Marfan mice than in control samples, indicating elevated circulating and tissue NO. Soluble guanylate cyclase and cGMP-dependent protein kinase are both activated in Marfan patients and mice and in wild-type mice treated with NO-donors, as shown by increased plasma cGMP and pVASP-S239 staining in aortic tissue. Marfan aortopathy in mice is reverted by pharmacological inhibition of soluble guanylate cyclase and cGMP-dependent protein kinase and lentiviral-mediated Prkg1 silencing. These findings identify potential biomarkers for monitoring Marfan Syndrome in patients and urge evaluation of cGMP-dependent protein kinase and soluble guanylate cyclase as therapeutic targets.


Asunto(s)
Aneurisma de la Aorta Torácica/patología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Síndrome de Marfan/complicaciones , Guanilil Ciclasa Soluble/metabolismo , Animales , Aorta/citología , Aorta/diagnóstico por imagen , Aorta/efectos de los fármacos , Aorta/patología , Aneurisma de la Aorta Torácica/diagnóstico , Aneurisma de la Aorta Torácica/etiología , Aneurisma de la Aorta Torácica/prevención & control , Biomarcadores/sangre , Biomarcadores/metabolismo , Carbazoles/administración & dosificación , GMP Cíclico/sangre , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibrilina-1/genética , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Síndrome de Marfan/sangre , Síndrome de Marfan/genética , Síndrome de Marfan/patología , Ratones , Músculo Liso Vascular/citología , Mutación , Miocitos del Músculo Liso , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/administración & dosificación , Cultivo Primario de Células , Guanilil Ciclasa Soluble/antagonistas & inhibidores , Ultrasonografía
13.
FEBS J ; 286(1): 46-65, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30548183

RESUMEN

Previous studies have demonstrated that activation of calcineurin induces pathological cardiac hypertrophy (CH). In these studies, loss-of-function was mostly achieved by systemic administration of the calcineurin inhibitor cyclosporin A. The lack of conditional knockout models for calcineurin function has impeded progress toward defining the role of this protein during the onset and the development of CH in adults. Here, we exploited a mouse model of CH based on the infusion of a hypertensive dose of angiotensin II (AngII) to model the role of calcineurin in CH in adulthood. AngII-induced CH in adult mice was reduced by treatment with cyclosporin A, without affecting the associated increase in blood pressure, and also by induction of calcineurin deletion in adult mouse cardiomyocytes, indicating that cardiomyocyte calcineurin is required for AngII-induced CH. Surprisingly, cardiac-specific deletion of calcineurin, but not treatment of mice with cyclosporin A, significantly reduced AngII-induced cardiac fibrosis and apoptosis. Analysis of profibrotic genes revealed that AngII-induced expression of Tgfß family members and Lox was not inhibited by cyclosporin A but was markedly reduced by cardiac-specific calcineurin deletion. These results show that AngII induces a direct, calcineurin-dependent prohypertrophic effect in cardiomyocytes, as well as a systemic hypertensive effect that is independent of calcineurin activity.


Asunto(s)
Calcineurina/fisiología , Cardiomegalia/patología , Fibrosis/patología , Miocitos Cardíacos/patología , Angiotensina II/toxicidad , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/metabolismo , Progresión de la Enfermedad , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Transducción de Señal , Vasoconstrictores/toxicidad
14.
J Exp Med ; 215(11): 2901-2918, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30327417

RESUMEN

MHCII in antigen-presenting cells (APCs) is a key regulator of adaptive immune responses. Expression of MHCII genes is controlled by the transcription coactivator CIITA, itself regulated through cell type-specific promoters. Here we show that the transcription factor NFAT5 is needed for expression of Ciita and MHCII in macrophages, but not in dendritic cells and other APCs. NFAT5-deficient macrophages showed defective activation of MHCII-dependent responses in CD4+ T lymphocytes and attenuated capacity to elicit graft rejection in vivo. Ultrasequencing analysis of NFAT5-immunoprecipitated chromatin uncovered an NFAT5-regulated region distally upstream of Ciita This region was required for CIITA and hence MHCII expression, exhibited NFAT5-dependent characteristics of active enhancers such as H3K27 acetylation marks, and required NFAT5 to interact with Ciita myeloid promoter I. Our results uncover an NFAT5-regulated mechanism that maintains CIITA and MHCII expression in macrophages and thus modulates their T lymphocyte priming capacity.


Asunto(s)
Elementos de Facilitación Genéticos/inmunología , Regulación de la Expresión Génica/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Macrófagos/inmunología , Proteínas Nucleares/inmunología , Transactivadores/inmunología , Factores de Transcripción/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Reordenamiento Génico/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Macrófagos/citología , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Transactivadores/genética , Factores de Transcripción/genética
15.
Nat Med ; 23(8): 964-974, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28692064

RESUMEN

Polo-like kinase 1 (PLK1), an essential regulator of cell division, is currently undergoing clinical evaluation as a target for cancer therapy. We report an unexpected function of Plk1 in sustaining cardiovascular homeostasis. Plk1 haploinsufficiency in mice did not induce obvious cell proliferation defects but did result in arterial structural alterations, which frequently led to aortic rupture and death. Specific ablation of Plk1 in vascular smooth muscle cells (VSMCs) led to reduced arterial elasticity, hypotension, and an impaired arterial response to angiotensin II in vivo. Mechanistically, we found that Plk1 regulated angiotensin II-dependent activation of RhoA and actomyosin dynamics in VSMCs in a mitosis-independent manner. This regulation depended on Plk1 kinase activity, and the administration of small-molecule Plk1 inhibitors to angiotensin II-treated mice led to reduced arterial fitness and an elevated risk of aneurysm and aortic rupture. We thus conclude that a partial reduction of Plk1 activity that does not block cell division can nevertheless impair aortic homeostasis. Our findings have potentially important implications for current approaches aimed at PLK1 inhibition for cancer therapy.


Asunto(s)
Angiotensina II/metabolismo , Aneurisma de la Aorta/genética , Rotura de la Aorta/genética , Proteínas de Ciclo Celular/genética , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas de Unión al GTP rho/metabolismo , Animales , Aorta/metabolismo , Aorta/ultraestructura , Aneurisma de la Aorta/metabolismo , Rotura de la Aorta/metabolismo , Presión Sanguínea , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/genética , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Haploinsuficiencia , Homeostasis/genética , Hipotensión/genética , Immunoblotting , Ratones , Microscopía Electrónica de Transmisión , Mitosis , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Rigidez Vascular/genética , Proteína de Unión al GTP rhoA , Quinasa Tipo Polo 1
16.
Nat Commun ; 7: 10477, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26795633

RESUMEN

Disrupted organ growth leads to disease development. Hypertrophy underlies postnatal heart growth and is triggered after stress, but the molecular mechanisms involved in these processes are largely unknown. Here we show that cardiac activation of p38γ and p38δ increases during postnatal development and by hypertrophy-inducing stimuli. p38γ/δ promote cardiac hypertrophy by phosphorylating the mTORC1 and mTORC2 inhibitor DEPTOR, which leads to its degradation and mTOR activation. Hearts from mice lacking one or both kinases are below normal size, have high levels of DEPTOR, low activity of the mTOR pathway and reduced protein synthesis. The phenotype of p38γ/δ(-/-) mice is reverted by overactivation of mTOR with amino acids, shRNA-mediated knockdown of Deptor, or cardiomyocyte overexpression of active p38γ and p38δ. Moreover, in WT mice, heart weight is reduced by cardiac overexpression of DEPTOR. Our results demonstrate that p38γ/δ control heart growth by modulating mTOR pathway through DEPTOR phosphorylation and subsequent degradation.


Asunto(s)
Cardiomegalia/enzimología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Quinasa 12 Activada por Mitógenos/metabolismo , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Cardiomegalia/genética , Cardiomegalia/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 12 Activada por Mitógenos/genética , Proteína Quinasa 13 Activada por Mitógenos/genética , Complejos Multiproteicos/genética , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Fosforilación , Proteolisis , Serina-Treonina Quinasas TOR/genética
17.
J Am Heart Assoc ; 4(1): e001218, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25609414

RESUMEN

BACKGROUND: Biomarkers are frequently used to estimate infarct size (IS) as an endpoint in experimental and clinical studies. Here, we prospectively studied the impact of left ventricular (LV) hypertrophy (LVH) on biomarker release in clinical and experimental myocardial infarction (MI). METHODS AND RESULTS: ST-segment elevation myocardial infarction (STEMI) patients (n=140) were monitored for total creatine kinase (CK) and cardiac troponin I (cTnI) over 72 hours postinfarction and were examined by cardiac magnetic resonance (CMR) at 1 week and 6 months postinfarction. MI was generated in pigs with induced LVH (n=10) and in sham-operated pigs (n=8), and serial total CK and cTnI measurements were performed and CMR scans conducted at 7 days postinfarction. Regression analysis was used to study the influence of LVH on total CK and cTnI release and IS estimated by CMR (gold standard). Receiver operating characteristic (ROC) curve analysis was performed to study the discriminatory capacity of the area under the curve (AUC) of cTnI and total CK in predicting LV dysfunction. Cardiomyocyte cTnI expression was quantified in myocardial sections from LVH and sham-operated pigs. In both the clinical and experimental studies, LVH was associated with significantly higher peak and AUC of cTnI, but not with differences in total CK. ROC curves showed that the discriminatory capacity of AUC of cTnI to predict LV dysfunction was significantly worse for patients with LVH. LVH did not affect the capacity of total CK to estimate IS or LV dysfunction. Immunofluorescence analysis revealed significantly higher cTnI content in hypertrophic cardiomyocytes. CONCLUSIONS: Peak and AUC of cTnI both significantly overestimate IS in the presence of LVH, owing to the higher troponin content per cardiomyocyte. In the setting of LVH, cTnI release during STEMI poorly predicts postinfarction LV dysfunction. LV mass should be taken into consideration when IS or LV function are estimated by troponin release.


Asunto(s)
Forma MB de la Creatina-Quinasa/metabolismo , Hipertrofia Ventricular Izquierda/sangre , Infarto del Miocardio/sangre , Investigación Biomédica Traslacional/métodos , Troponina I/metabolismo , Anciano , Animales , Área Bajo la Curva , Biomarcadores/sangre , Forma MB de la Creatina-Quinasa/sangre , Modelos Animales de Enfermedad , Electrocardiografía/métodos , Femenino , Humanos , Hipertrofia Ventricular Izquierda/diagnóstico , Imagen por Resonancia Cinemagnética/métodos , Masculino , Persona de Mediana Edad , Monitoreo Fisiológico/métodos , Infarto del Miocardio/diagnóstico , Pronóstico , Estudios Prospectivos , Curva ROC , Análisis de Regresión , Índice de Severidad de la Enfermedad , Sus scrofa , Porcinos , Troponina I/sangre , Función Ventricular Izquierda
18.
Curr Med Chem ; 11(8): 997-1007, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15078162

RESUMEN

The well known calcium-sensitive phosphatase calcineurin is implicated in many eukaryotic activation and developmental programmes, including lymphocyte activation, heart-valve morphogenesis, angiogenesis, and neural and muscle development. The importance of this phosphatase is graphically illustrated by the observation that the immunosuppressive actions of the microbial drugs Cyclosporin A (CsA) and FK506 arise from their inhibition of calcineurin. As substrates of calcineurin, transcription factors of the NFAT family play an essential role in lymphocyte activation, and it follows that their function is also inhibited by CsA and FK506. Although the use of these drugs has been crucial for the success of organ transplantation, their therapeutic use is associated with severe side effects. There is, therefore a need to develop better, less toxic immunosuppressive agents. In recent years, a number of endogenous calcineurin inhibitor proteins have been identified that bind calcineurin and block its phosphatase activity. In some cases the calcineurin interaction domains of these proteins, or their corresponding docking sites on calcineurin, have been described. However, their mode of action and regulatory mechanisms are not completely known. In a more recent development, specific amino acidic sequences implicated in the interaction between calcineurin and NFAT have been identified. It is of special interest that specific disruption of this pathway has been obtained through the expression of peptides based on some of these sequences. A more profound analysis of these issues could open up new perspectives in immunosuppressive therapy; promising compounds with features of endogenous calcineurin inhibitors (and thus likely to have fewer toxic effects than CsA and FK506), or selective blockers of calcineurin-NFAT interactions that would not alter the functioning of other calcineurin substrates.


Asunto(s)
Inhibidores de la Calcineurina , Ciclosporina/farmacología , Proteínas de Unión al ADN/antagonistas & inhibidores , Inmunosupresores/farmacología , Proteínas Nucleares , Transducción de Señal/efectos de los fármacos , Tacrolimus/farmacología , Factores de Transcripción/antagonistas & inhibidores , Calcineurina/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Modelos Biológicos , Factores de Transcripción NFATC , Factores de Transcripción/metabolismo
19.
J Proteomics ; 75(2): 561-71, 2011 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21920478

RESUMEN

Proteins secreted by cells are of the highest biomedical relevance since they play a significant role in the progression of numerous diseases. However, characterization of the proteins specifically secreted in response to precise stimuli is challenging, since these proteins are contaminated by cellular byproducts. Here we present a method to characterize a dynamic secretome and demonstrate its utility by performing the deepest quantitative analysis to date of proteins secreted by lymphoid Jurkat T-cells upon activation. Cell-free supernatant proteins were analyzed by using an optimized protocol for differential (18)O/(16)O-labeling and LC-MS/MS, followed by statistical analysis using a random-effects model. More than 4000 unique peptides belonging to 1288 proteins were identified and a large proportion could be quantified. To determine the proteins whose secretion was up-regulated upon T-cell activation, protein variance of the null hypothesis was estimated after protein classification in terms of secretion and ontology using bioinformatic tools. 62 proteins showed a statistically significant change in abundance upon cell activation and most of them (49 proteins) were up-regulated. These proteins were functionally involved mainly in inflammatory response, signal transduction, cell growth and differentiation and cell redox homeostasis. Our approach provides a promising technology for the high-throughput quantitative study of dynamic secretomes.


Asunto(s)
Proteínas de Neoplasias/metabolismo , Proteoma/metabolismo , Linfocitos T/metabolismo , Proliferación Celular , Cromatografía Liquida/métodos , Humanos , Células Jurkat , Activación de Linfocitos , Transducción de Señal , Espectrometría de Masas en Tándem/métodos , Regulación hacia Arriba
20.
J Biol Chem ; 283(43): 29004-10, 2008 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-18676376

RESUMEN

Calcineurin/NFAT signaling is involved in multiple aspects of skeletal muscle development and disease. The myogenic basic helix-loop-helix transcription factors, MyoD, myogenin, Myf5, and MRF4 specify the myogenic lineage. Here we show that calcineurin/NFAT (nuclear factor of activated T cells) signaling is required for primary myogenesis by transcriptional cooperation with the basic helix-loop-helix transcription factor MyoD. Calcineurin/NFAT signaling is involved in myogenin expression in differentiating myoblasts, where the myogenic regulatory factor MyoD synergistically cooperates with NFATc2/c3 at the myogenin promoter. Using gel shift and chromatin immunoprecipitation assays, we identified two conserved NFAT binding sites in the myogenin promoter that were occupied by NFATc3 upon skeletal muscle differentiation. The transcriptional integration between NFATc3 and MyoD is crucial for primary myogenesis in vivo, as myogenin expression is weak in myod:nfatc3 double null embryos, whereas myogenin expression is unaffected in embryos with null mutations for either factor alone. Thus, the combined findings provide a novel transcriptional paradigm for the first steps of myogenesis, where a calcineurin/NFATc3 pathway regulates myogenin induction in cooperation with MyoD during myogenesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteína MioD/metabolismo , Miogenina/biosíntesis , Factores de Transcripción NFATC/metabolismo , Animales , Células COS , Calcineurina/metabolismo , Línea Celular , Chlorocebus aethiops , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Miogenina/metabolismo , Regiones Promotoras Genéticas , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA