Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-37292897

RESUMEN

The sarco(endo)plasmic reticulum Ca 2+ ATPase (SERCA) is a membrane transporter that creates and maintains intracellular Ca 2+ stores. In the heart, SERCA is regulated by an inhibitory interaction with the monomeric form of the transmembrane micropeptide phospholamban (PLB). PLB also forms avid homo-pentamers, and dynamic exchange of PLB between pentamers and the regulatory complex with SERCA is an important determinant of cardiac responsiveness to exercise. Here, we investigated two naturally occurring pathogenic mutations of PLB, a cysteine substitution of arginine 9 (R9C) and an in-frame deletion of arginine 14 (R14del). Both mutations are associated with dilated cardiomyopathy. We previously showed that the R9C mutation causes disulfide crosslinking and hyperstabilization of pentamers. While the pathogenic mechanism of R14del is unclear, we hypothesized that this mutation may also alter PLB homo-oligomerization and disrupt the PLB-SERCA regulatory interaction. SDS-PAGE revealed a significantly increased pentamer:monomer ratio for R14del-PLB when compared to WT-PLB. In addition, we quantified homo-oligomerization and SERCA-binding in live cells using fluorescence resonance energy transfer (FRET) microscopy. R14del-PLB showed an increased affinity for homo-oligomerization and decreased binding affinity for SERCA compared to WT, suggesting that, like R9C, the R14del mutation stabilizes PLB in its pentameric form, decreasing its ability to regulate SERCA. Moreover, the R14del mutation reduces the rate of PLB unbinding from the pentamer after a transient Ca 2+ elevation, limiting the rate of re-binding to SERCA. A computational model predicted that hyperstabilization of PLB pentamers by R14del impairs the ability of cardiac Ca 2+ handling to respond to changing heart rates between rest and exercise. We postulate that impaired responsiveness to physiological stress contributes to arrhythmogenesis in human carriers of the R14del mutation.

2.
Proc Natl Acad Sci U S A ; 120(19): e2212118120, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37126683

RESUMEN

The prognosis and treatment outcomes of heart failure (HF) patients rely heavily on disease etiology, yet the majority of underlying signaling mechanisms are complex and not fully elucidated. Phosphorylation is a major point of protein regulation with rapid and profound effects on the function and activity of protein networks. Currently, there is a lack of comprehensive proteomic and phosphoproteomic studies examining cardiac tissue from HF patients with either dilated dilated cardiomyopathy (DCM) or ischemic cardiomyopathy (ICM). Here, we used a combined proteomic and phosphoproteomic approach to identify and quantify more than 5,000 total proteins with greater than 13,000 corresponding phosphorylation sites across explanted left ventricle (LV) tissue samples, including HF patients with DCM vs. nonfailing controls (NFC), and left ventricular infarct vs. noninfarct, and periinfarct vs. noninfarct regions of HF patients with ICM. Each pair-wise comparison revealed unique global proteomic and phosphoproteomic profiles with both shared and etiology-specific perturbations. With this approach, we identified a DCM-associated hyperphosphorylation cluster in the cardiomyocyte intercalated disc (ICD) protein, αT-catenin (CTNNA3). We demonstrate using both ex vivo isolated cardiomyocytes and in vivo using an AAV9-mediated overexpression mouse model, that CTNNA3 phosphorylation at these residues plays a key role in maintaining protein localization at the cardiomyocyte ICD to regulate conductance and cell-cell adhesion. Collectively, this integrative proteomic/phosphoproteomic approach identifies region- and etiology-associated signaling pathways in human HF and describes a role for CTNNA3 phosphorylation in the pathophysiology of DCM.


Asunto(s)
Cardiomiopatía Dilatada , Insuficiencia Cardíaca , Animales , Ratones , Humanos , Cardiomiopatía Dilatada/metabolismo , Ventrículos Cardíacos/metabolismo , Fosforilación , Proteómica , Miocardio/metabolismo , Insuficiencia Cardíaca/metabolismo , alfa Catenina/metabolismo
3.
STAR Protoc ; 4(1): 101933, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36574341

RESUMEN

Here, we describe a protocol for purifying functional clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) from Staphylococcus aureus within 24 h and over 90% purity. SaCas9 purification begins with immobilized metal affinity chromatography, followed by cation exchange chromatography, and ended with centrifugal concentrators. The simplicity, cost-effectiveness, and reproducibility of such protocols will enable general labs to produce a sizable amount of Cas9 proteins, further accelerating CRISPR research.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Staphylococcus aureus/genética , Análisis Costo-Beneficio , Reproducibilidad de los Resultados
4.
Nat Commun ; 13(1): 6166, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36257954

RESUMEN

The intercalated disc (ICD) is a unique membrane structure that is indispensable to normal heart function, yet its structural organization is not completely understood. Previously, we showed that the ICD-bound transmembrane protein 65 (Tmem65) was required for connexin43 (Cx43) localization and function in cultured mouse neonatal cardiomyocytes. Here, we investigate the functional and cellular effects of Tmem65 reductions on the myocardium in a mouse model by injecting CD1 mouse pups (3-7 days after birth) with recombinant adeno-associated virus 9 (rAAV9) harboring Tmem65 shRNA, which reduces Tmem65 expression by 90% in mouse ventricles compared to scrambled shRNA injection. Tmem65 knockdown (KD) results in increased mortality which is accompanied by eccentric hypertrophic cardiomyopathy within 3 weeks of injection and progression to dilated cardiomyopathy with severe cardiac fibrosis by 7 weeks post-injection. Tmem65 KD hearts display depressed hemodynamics as measured echocardiographically as well as slowed conduction in optical recording accompanied by prolonged PR intervals and QRS duration in electrocardiograms. Immunoprecipitation and super-resolution microscopy demonstrate a physical interaction between Tmem65 and sodium channel ß subunit (ß1) in mouse hearts and this interaction appears to be required for both the establishment of perinexal nanodomain structure and the localization of both voltage-gated sodium channel 1.5 (NaV1.5) and Cx43 to ICDs. Despite the loss of NaV1.5 at ICDs, whole-cell patch clamp electrophysiology did not reveal reductions in Na+ currents but did show reduced Ca2+ and K+ currents in Tmem65 KD cardiomyocytes in comparison to control cells. We conclude that disrupting Tmem65 function results in impaired ICD structure, abnormal cardiac electrophysiology, and ultimately cardiomyopathy.


Asunto(s)
Conexina 43 , Canal de Sodio Activado por Voltaje NAV1.5 , Ratones , Animales , Conexina 43/genética , Conexina 43/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , ARN Interferente Pequeño/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Transcripción/metabolismo
5.
ACS Appl Mater Interfaces ; 13(49): 58352-58368, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34873903

RESUMEN

Developing safe and effective strategies to deliver biomolecules such as oligonucleotides and proteins into cells has grown in importance over recent years, with an increasing demand for non-viral methods that enable clinical translation. Here, we investigate uniquely configured oligo-urethane nanoparticles based on synthetic chemistries that minimize the release of pro-inflammatory biomarkers from immune cells, show low cytotoxicity in a broad range of cells, and efficiently deliver oligonucleotides and proteins into mammalian cells. The mechanism of cell uptake for the self-assembled oligo-urethane nanoparticles was shown to be directed by caveolae-dependent endocytosis in murine myoblasts (C2C12) cells. Inhibiting caveolae functions with genistein and methyl-ß-cyclodextrin limited nanoparticle internalization. The nanoparticles showed a very high delivery efficiency for the genetic material (a 47-base oligonucleotide) (∼80% incorporation into cells) as well as the purified protein (full length firefly luciferase, 67 kDa) into human embryonic kidney (HEK293T) cells. Luciferase enzyme activity in HEK293T cells demonstrated that intact and functional proteins could be delivered and showed a significant extension of activity retention up to 24 h, well beyond the 2 h half-life of the free enzyme. This study introduces a novel self-assembled oligo-urethane nanoparticle delivery platform with very low associated production costs, enabled by their scalable chemistry (the benchwork cost is $ 0.152/mg vs $ 974.6/mg for typical lipid carriers) that has potential to deliver both oligonucleotides and proteins for biomedical purposes.


Asunto(s)
Materiales Biocompatibles/química , Sistemas de Liberación de Medicamentos , Nanopartículas/química , Oligonucleótidos/química , Animales , Materiales Biocompatibles/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células HEK293 , Humanos , Luciferasas/metabolismo , Ensayo de Materiales , Ratones , Estructura Molecular , Oligonucleótidos/genética , Oligonucleótidos/farmacología
6.
FEBS Lett ; 595(22): 2756-2767, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34693525

RESUMEN

Neuronatin (NNAT) is a transmembrane protein in the endoplasmic reticulum involved in metabolic regulation. It shares sequence homology with sarcolipin (SLN), which negatively regulates the sarco(endo)plasmic reticulum Ca2+ -ATPase (SERCA) that maintains energy homeostasis in muscles. Here, we examined whether NNAT could uncouple the Ca2+ transport activity of SERCA from ATP hydrolysis, similarly to SLN. NNAT significantly reduced Ca2+ uptake without altering SERCA activity, ultimately lowering the apparent coupling ratio of SERCA. This effect of NNAT was reversed by the adenylyl cyclase activator forskolin. Furthermore, soleus muscles from high fat diet (HFD)-fed mice showed a significant downregulation in NNAT content compared with chow-fed mice, whereas an upregulation in NNAT content was observed in fast-twitch muscles from HFD- versus chow- fed mice. Therefore, NNAT is a SERCA uncoupler in cells and may function in adaptive thermogenesis.


Asunto(s)
Acoplamiento Excitación-Contracción , Proteínas de la Membrana/metabolismo , Músculo Esquelético/metabolismo , Proteínas del Tejido Nervioso/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Animales , Señalización del Calcio , Colforsina/farmacología , Dieta Alta en Grasa , Células HEK293 , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/fisiología , Proteínas del Tejido Nervioso/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética
7.
Nat Commun ; 11(1): 965, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-32075961

RESUMEN

The sarco-endoplasmic reticulum (SR/ER) plays an important role in the development and progression of many heart diseases. However, many aspects of its structural organization remain largely unknown, particularly in cells with a highly differentiated SR/ER network. Here, we report a cardiac enriched, SR/ER membrane protein, REEP5 that is centrally involved in regulating SR/ER organization and cellular stress responses in cardiac myocytes. In vitro REEP5 depletion in mouse cardiac myocytes results in SR/ER membrane destabilization and luminal vacuolization along with decreased myocyte contractility and disrupted Ca2+ cycling. Further, in vivo CRISPR/Cas9-mediated REEP5 loss-of-function zebrafish mutants show sensitized cardiac dysfunction upon short-term verapamil treatment. Additionally, in vivo adeno-associated viral (AAV9)-induced REEP5 depletion in the mouse demonstrates cardiac dysfunction. These results demonstrate the critical role of REEP5 in SR/ER organization and function as well as normal heart function and development.


Asunto(s)
Corazón/fisiopatología , Proteínas de la Membrana/deficiencia , Retículo Sarcoplasmático/patología , Animales , Calcio/metabolismo , Células Cultivadas , Estrés del Retículo Endoplásmico , Técnicas de Inactivación de Genes , Silenciador del Gen , Corazón/crecimiento & desarrollo , Cardiopatías/metabolismo , Cardiopatías/patología , Cardiopatías/fisiopatología , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/metabolismo , Pez Cebra
8.
Physiol Rep ; 7(14): e14144, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31325238

RESUMEN

Phospholamban (PLN) is an important Ca2+ modulator at the sarcoplasmic reticulum (SR) of striated muscles. It physically interacts and inhibits sarcoplasmic reticulum Ca2+ ATPase (SERCA2) function, whereas a protein kinase A (PKA)-dependent phosphorylation at its serine 16 reverses the inhibition. The underlying mechanism of this post-translational modification, however, remains not fully understood. Using publicly available databases, we identified A-kinase anchoring protein 6 (AKAP6) as a candidate that might play some roles in PLN phosphorylation. Immunofluorescence showed colocalization between GFP-AKAP6 and PLN in transfected HEK-293T cells and cultured mouse neonatal cardiomyocytes (CMNCs). Co-immunoprecipitation confirmed the functional interaction between AKAP6 and PLN in HEK-293T and isolated adult rat cardiomyocytes in response to isoproterenol stimulation. Functionally, AKAP6 promoted Ca2+ uptake activity of SERCA1 in cotransfected HEK-293T cells despite the presence of PLN. These results were further confirmed in adult rat cardiomyocytes. Immunofluorescence showed colocalization of both proteins around the perinuclear region, while protein-protein interaction was corroborated by immunoprecipitation of the nucleus-enriched fraction of rat hearts. Our findings suggest AKAP6 as a novel interacting partner to PLN in HEK-293T and murine cardiomyocytes.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas de Unión al Calcio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Señalización del Calcio , Células Cultivadas , Células HEK293 , Humanos , Ratones , Unión Proteica , Ratas , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
9.
Nat Commun ; 6: 8391, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26403541

RESUMEN

Membrane proteins are crucial to heart function and development. Here we combine cationic silica-bead coating with shotgun proteomics to enrich for and identify plasma membrane-associated proteins from primary mouse neonatal and human fetal ventricular cardiomyocytes. We identify Tmem65 as a cardiac-enriched, intercalated disc protein that increases during development in both mouse and human hearts. Functional analysis of Tmem65 both in vitro using lentiviral shRNA-mediated knockdown in mouse cardiomyocytes and in vivo using morpholino-based knockdown in zebrafish show marked alterations in gap junction function and cardiac morphology. Molecular analyses suggest that Tmem65 interaction with connexin 43 (Cx43) is required for correct localization of Cx43 to the intercalated disc, since Tmem65 deletion results in marked internalization of Cx43, a shorter half-life through increased degradation, and loss of Cx43 function. Our data demonstrate that the membrane protein Tmem65 is an intercalated disc protein that interacts with and functionally regulates ventricular Cx43.


Asunto(s)
Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Ventrículos Cardíacos/metabolismo , Proteínas de la Membrana/genética , Miocitos Cardíacos/metabolismo , Proteínas de Pez Cebra/genética , Animales , Western Blotting , Cromatografía Líquida de Alta Presión , Técnica del Anticuerpo Fluorescente , Uniones Comunicantes/ultraestructura , Técnicas de Silenciamiento del Gen , Sistema de Conducción Cardíaco/fisiología , Técnicas In Vitro , Proteínas de la Membrana/metabolismo , Ratones , Microscopía Electrónica , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/ultraestructura , Proteómica , Dióxido de Silicio , Pez Cebra , Proteínas de Pez Cebra/metabolismo
10.
Proc Natl Acad Sci U S A ; 112(23): 7165-70, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26040000

RESUMEN

Phospholamban (PLN) is an effective inhibitor of the sarco(endo)plasmic reticulum Ca(2+) ATPase (SERCA). Here, we examined PLN stability and degradation in primary cultured mouse neonatal cardiomyocytes (CMNCs) and mouse hearts using immunoblotting, molecular imaging, and [(35)S]methionine pulse-chase experiments, together with lysosome (chloroquine and bafilomycin A1) and autophagic (3-methyladenine and Atg5 siRNA) antagonists. Inhibiting lysosomal and autophagic activities promoted endogenous PLN accumulation, whereas accelerating autophagy with metformin enhanced PLN degradation in CMNCs. This reduction in PLN levels was functionally correlated with an increased rate of SERCA2a activity, accounting for an inotropic effect of metformin. Metabolic labeling reaffirmed that metformin promoted wild-type and R9C PLN degradation. Immunofluorescence showed that PLN and the autophagy marker, microtubule light chain 3, became increasingly colocalized in response to chloroquine and bafilomycin treatments. Mechanistically, pentameric PLN was polyubiquitinylated at the K3 residue and this modification was required for p62-mediated selective autophagy trafficking. Consistently, attenuated autophagic flux in HECT domain and ankyrin repeat-containing E3 ubiquitin protein ligase 1-null mouse hearts was associated with increased PLN levels determined by immunoblots and immunofluorescence. Our study identifies a biological mechanism that traffics PLN to the lysosomes for degradation in mouse hearts.


Asunto(s)
Autofagia , Proteínas de Unión al Calcio/metabolismo , Metformina/farmacología , Miocitos Cardíacos/efectos de los fármacos , Animales , Células HEK293 , Humanos , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Proteolisis , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/fisiología , Ubiquitinación
11.
Cell Rep ; 7(3): 834-47, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24767997

RESUMEN

Mitochondrial production of reactive oxygen species (ROS) affects many processes in health and disease. SPG7 assembles with AFG3L2 into the mAAA protease at the inner membrane of mitochondria, degrades damaged proteins, and regulates the synthesis of mitochondrial ribosomes. SPG7 is cleaved and activated by AFG3L2 upon assembly. A variant in SPG7 that replaces arginine 688 with glutamine (Q688) is associated with several phenotypes, including toxicity of chemotherapeutic agents, type 2 diabetes mellitus, and (as reported here) coronary artery disease. We demonstrate that SPG7 processing is regulated by tyrosine phosphorylation of AFG3L2. Carriers of Q688 bypass this regulation and constitutively process and activate SPG7 mAAA protease. Cells expressing Q688 produce higher ATP levels and ROS, promoting cell proliferation. Our results thus reveal an unexpected link between the phosphorylation-dependent regulation of the mitochondria mAAA protease affecting ROS production and several clinical phenotypes.


Asunto(s)
Proteasas ATP-Dependientes/metabolismo , Metaloendopeptidasas/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteasas ATP-Dependientes/antagonistas & inhibidores , Proteasas ATP-Dependientes/genética , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Proliferación Celular , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Metaloendopeptidasas/genética , Mitocondrias/enzimología , Datos de Secuencia Molecular , Péptido Hidrolasas/metabolismo , Fenotipo , Fosforilación , Polimorfismo de Nucleótido Simple , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
12.
Expert Rev Proteomics ; 9(1): 13-5, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22292819

RESUMEN

Calcineurin (CN) is a calcium- and calmodulin-dependent protein phosphatase that consists of a catalytic subunit (calcineurin A [CnA]) and a calcium-binding, regulatory subunit (calcineurin B [CnB]). Calcineurin has been shown to be involved in a number of cellular processes, and aberrant signaling has been linked to multiple human diseases, such as cardiac hypertrophy and diabetes. Recent studies demonstrated that CN was involved in the survival of Cryptococcus neoformans, a fungal pathogen that infects humans, especially patients who are immunocompromised. CN appears to be essential for the survival and virulence of C. neoformans; however, the underlying mechanisms remain largely unknown. The Heitman laboratory recently identified a group of potential CnA-interacting proteins in C. neoformans during heat stress, and demonstrated an interaction of CnA with Sec28 and Sec13, which represent COPI and COPII protein complex members, respectively. The COP protein complexes are key proteins involved in intracellular endoplasmic reticulum and golgi protein trafficking. The results from the Heitman group suggest that CN interacts with components of the endoplasmic reticulum and the golgi during heat stress in C. neoformans and could highlight potential mechanisms by which these microbes could be targeted.

13.
PLoS One ; 6(8): e24100, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21887377

RESUMEN

BACKGROUND: Interferon regulatory factor 2 binding protein 2 (IRF2BP2) is a muscle-enriched transcription factor required to activate vascular endothelial growth factor-A (VEGFA) expression in muscle. IRF2BP2 is found in the nucleus of cardiac and skeletal muscle cells. During the process of skeletal muscle differentiation, some IRF2BP2 becomes relocated to the cytoplasm, although the functional significance of this relocation and the mechanisms that control nucleocytoplasmic localization of IRF2BP2 are not yet known. METHODOLOGY/PRINCIPAL FINDINGS: Here, by fusing IRF2BP2 to green fluorescent protein and testing a series of deletion and site-directed mutagenesis constructs, we mapped the nuclear localization signal (NLS) to an evolutionarily conserved sequence (354)ARKRKPSP(361) in IRF2BP2. This sequence corresponds to a classical nuclear localization motif bearing positively charged arginine and lysine residues. Substitution of arginine and lysine with negatively charged aspartic acid residues blocked nuclear localization. However, these residues were not sufficient because nuclear targeting of IRF2BP2 also required phosphorylation of serine 360 (S360). Many large-scale phosphopeptide proteomic studies had reported previously that serine 360 of IRF2BP2 is phosphorylated in numerous human cell types. Alanine substitution at this site abolished IRF2BP2 nuclear localization in C(2)C(12) myoblasts and CV1 cells. In contrast, substituting serine 360 with aspartic acid forced nuclear retention and prevented cytoplasmic redistribution in differentiated C(2)C(12) muscle cells. As for the effects of these mutations on VEGFA promoter activity, the S360A mutation interfered with VEGFA activation, as expected. Surprisingly, the S360D mutation also interfered with VEGFA activation, suggesting that this mutation, while enforcing nuclear entry, may disrupt an essential activation function of IRF2BP2. CONCLUSIONS/SIGNIFICANCE: Nuclear localization of IRF2BP2 depends on phosphorylation near a conserved NLS. Changes in phosphorylation status likely control nucleocytoplasmic localization of IRF2BP2 during muscle differentiation.


Asunto(s)
Transporte Activo de Núcleo Celular , Proteínas Portadoras/química , Señales de Localización Nuclear/análisis , Proteínas Nucleares/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Portadoras/metabolismo , Secuencia Conservada , Citoplasma/química , Proteínas de Unión al ADN , Humanos , Músculo Esquelético , Mutación , Proteínas Nucleares/metabolismo , Fosforilación , Factores de Transcripción , Factor A de Crecimiento Endotelial Vascular/genética
14.
FASEB J ; 24(12): 4825-34, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20702774

RESUMEN

We sought to identify an essential component of the TEAD4/VGLL4 transcription factor complex that controls vascular endothelial growth factor A (VEGFA) expression in muscle. A yeast 2-hybrid screen was used to clone a novel component of the TEAD4 complex from a human heart cDNA library. We identified interferon response factor 2 binding protein 2 (IRF2BP2) and confirmed its presence in the TEAD4/VGLL4 complex in vivo by coimmunoprecipitation and mammalian 2-hybrid assays. Coexpression of IRF2BP2 with TEAD4/VGLL4 or TEAD1 alone potently activated, whereas knockdown of IRF2BP2 reduced, VEGFA expression in C(2)C(12) muscle cells. Thus, IRF2BP2 is required to activate VEGFA expression. In mouse embryos, IRF2BP2 was ubiquitously expressed but became progressively enriched in the fetal heart, skeletal muscles, and lung. Northern blot analysis revealed high levels of IRF2BP2 mRNA in adult human heart and skeletal muscles, but immunoblot analysis showed low levels of IRF2BP2 protein in skeletal muscle, indicating post-transcriptional regulation of IRF2BP2 expression. IRF2BP2 protein levels are markedly increased by ischemia in skeletal and cardiac muscle compared to normoxic controls. IRF2BP2 is a novel ischemia-induced coactivator of VEGFA expression that may contribute to revascularization of ischemic cardiac and skeletal muscles.


Asunto(s)
Proteínas Portadoras/metabolismo , Isquemia/fisiopatología , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Northern Blotting , Proteínas Portadoras/genética , Línea Celular , Proteínas de Unión al ADN , Femenino , Haplorrinos , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Técnicas In Vitro , Ratones , Músculo Esquelético/patología , Miocardio/patología , Proteínas Nucleares/genética , Filogenia , Unión Proteica , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factor A de Crecimiento Endotelial Vascular/genética
15.
FASEB J ; 23(6): 1705-9, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19218499

RESUMEN

Diet-induced weight loss is affected by a wide range of factors, including genetic variation. Identifying functional polymorphisms will help to elucidate mechanisms that account for variation in dietary metabolism. Previously, we reported a strong association between a common single nucleotide polymorphism (SNP) rs2419621 (C>T) in the promoter of acyl-CoA synthetase long chain 5 (ACSL5), rapid weight loss in obese Caucasian females, and elevated ACSL5 mRNA levels in skeletal muscle biopsies. Here, we showed by electrophoretic mobility shift assay (EMSA) that the T allele creates a functional cis-regulatory E-box element (CANNTG) that is recognized by the myogenic regulatory factor MyoD. The T allele promoted MyoD-dependent activation of a 1089-base pair ACSL5 promoter fragment in nonmuscle CV1 cells. Differentiation of skeletal myoblasts significantly elevated expression of the ACSL5 promoter. The T allele sustained promoter activity 48 h after differentiation, whereas the C allele showed a significant decline. These results reveal a mechanism for elevated transcription of ACSL5 in skeletal muscle of carriers of the rs2419621(T) allele, associated with more rapid diet-induced weight loss. Natural selection favoring promoter polymorphisms that reduced expression of catabolic genes in skeletal muscle likely accounts for the resistance of obese individuals to dietary intervention.


Asunto(s)
Coenzima A Ligasas/genética , Dieta Reductora , Músculo Esquelético/fisiología , Polimorfismo Genético , Regiones Promotoras Genéticas , Pérdida de Peso/genética , Adulto , Alelos , Animales , Secuencia de Bases , Coenzima A Ligasas/metabolismo , Femenino , Humanos , Ratones , Datos de Secuencia Molecular , Proteína MioD/genética , Proteína MioD/metabolismo , Obesidad/genética , Obesidad/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Población Blanca/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...