Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Biochem Pharmacol ; 208: 115406, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36596415

RESUMEN

SAP97 is a member of the MAGUK family of proteins, but unlike other MAGUK proteins that are selectively expressed in the CNS, SAP97 is also expressed in peripheral organs, like the heart and kidneys. SAP97 has several protein binding cassettes, and this review will describe their involvement in creating SAP97-anchored multiprotein networks. SAP97-anchored networks localized at the inner leaflet of the cell membrane play a major role in trafficking and targeting of membrane G protein-coupled receptors (GPCR), channels, and structural proteins. SAP97 plays a major role in compartmentalizing voltage gated sodium and potassium channels to specific cellular compartments of heart cells. SAP97 undergoes extensive alternative splicing. These splice variants give rise to different SAP97 isoforms that alter its cellular localization, networking, signaling and trafficking effects. Regarding GPCR, SAP97 binds to the ß1-adrenergic receptor and recruits AKAP5/PKA and PDE4D8 to create a multiprotein complex that regulates trafficking and signaling of cardiac ß1-AR. In the kidneys, SAP97 anchored networks played a role in trafficking of aquaporin-2 water channels. Cardiac specific ablation of SAP97 (SAP97-cKO) resulted in cardiac hypertrophy and failure in aging mice. Similarly, instituting transverse aortic constriction (TAC) in young SAP97 c-KO mice exacerbated TAC-induced cardiac remodeling and dysfunction. These findings highlight a critical role for SAP97 in the pathophysiology of a number of cardiac and renal diseases, suggesting that SAP97 is a relevant target for drug discovery.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas de la Membrana , Ratones , Animales , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Membrana/metabolismo , Transporte de Proteínas/fisiología , Transducción de Señal/fisiología , Complejos Multiproteicos/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Proteínas de Anclaje a la Quinasa A/metabolismo
2.
Am J Physiol Renal Physiol ; 317(2): F375-F387, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31141395

RESUMEN

Arginine-vasopressin (AVP)-mediated translocation of aquaporin-2 (AQP2) protein-forming water channels from storage vesicles to the membrane of renal collecting ducts is critical for the renal conservation of water. The type-1 PDZ-binding motif (PBM) in AQP2, "GTKA," is a critical barcode for its translocation, but its precise role and that of its interacting protein partners in this process remain obscure. We determined that synapse-associated protein-97 (SAP97), a membrane-associated guanylate kinase protein involved in establishing epithelial cell polarity, was an avid binding partner to the PBM of AQP2. The role of PBM and SAP97 on AQP2 redistribution in response to AVP was assessed in LLC-PK1 renal collecting cells by confocal microscopy and cell surface biotinylation techniques. These experiments indicated that distribution of AQP2 and SAP97 overlapped in the kidneys and LLC-PK1 cells and that knockdown of SAP97 inhibited the translocation of AQP2 in response to AVP. Binding between AQP2 and SAP97 was mediated by specific interactions between the second PDZ of SAP97 and PBM of AQP2. Mechanistically, inactivation of the PBM of AQP2, global delocalization of PKA, or knockdown of SAP97 inhibited AQP2 translocation as well as AVP- and forskolin-mediated phosphorylation of Ser256 in AQP2, which serves as the major translocation barcode of AQP2. These results suggest that the targeting of PKA to the microdomain of AQP2 via SAP97-AQP2 interactions in association with cross-talk between two barcodes in AQP2, namely, the PBM and phospho-Ser256, plays an important role in the translocation of AQP2 in the kidney.


Asunto(s)
Acuaporina 2/metabolismo , Arginina Vasopresina/farmacología , Homólogo 1 de la Proteína Discs Large/metabolismo , Células Epiteliales/efectos de los fármacos , Túbulos Renales Proximales/efectos de los fármacos , Dominios PDZ , Animales , Acuaporina 2/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Homólogo 1 de la Proteína Discs Large/genética , Células Epiteliales/metabolismo , Túbulos Renales Proximales/metabolismo , Células LLC-PK1 , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Serina , Porcinos
3.
Mol Pharmacol ; 95(5): 563-572, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30894404

RESUMEN

G protein-coupled receptor family C group 6 member A (GPRC6A) is activated by testosterone and modulates prostate cancer progression. Most humans have a GPRC6A variant that contains a recently evolved KGKY insertion/deletion in the third intracellular loop (ICL3) (designated as GPRC6AICL3_KGKY) that replaces the ancestral KGRKLP sequence (GPRC6AICL3_RKLP) present in all other species. In vitro assays purport that human GPRC6AICL3_KGKY is retained intracellularly and lacks function. These findings contrast with ligand-dependent activation and coupling to mammalian target of rapamycin complex 1 (mTORC1) signaling of endogenous human GPRC6AICL3_KGKY in PC-3 cells. To understand these discrepant results, we expressed mouse (mGPRC6AICL3_KGRKLP), human (hGPRC6AICL3_KGKY), and humanized mouse (mGPRC6AICL3_KGKY) GPRC6A into human embryonic kidney 293 cells. Our results demonstrate that mGPRC6AICL3_KGRKLP acts as a classic G protein-coupled receptor, which is expressed at the cell membrane and internalizes in response to ligand activation by testosterone. In contrast, hGPRC6AICL3_KGKY and humanized mouse mGPRC6AICL3_KGKY are retained intracellularly in ligand naive cells, yet exhibit ß-arrestin-dependent signaling responses, mitogen-activated protein kinase [i.e., extracellular signal-regulated kinase (ERK)], and p70S6 kinase phosphorylation in response to testosterone, indicating that hGPRC6AICL3_KGKY is functional. Indeed, testosterone stimulates time- and dose-dependent activation of ERK, protein kinase B, and mTORC1 signaling in wild-type PC-3 cells that express endogenous GPRC6AICL3_KGKY In addition, testosterone stimulates GPRC6A-dependent cell proliferation in wild-type PC-3 cells and inhibits autophagy by activating mTORC1 effectors eukaryotic translation initiation factor 4E binding protein 1 and Unc-51 like autophagy activating kinase 1. Testosterone activation of GPRC6A has the obligate requirement for calcium in the incubation media. In contrast, in GPRC6A-deficient cells, the effect of testosterone to activate downstream signaling is abolished, indicating that human GPRC6A is required for mediating the effects of testosterone on cell proliferation and autophagy.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Testosterona/farmacología , Animales , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Humanos , Ligandos , Masculino , Ratones , Células PC-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo
4.
Mol Pharmacol ; 94(2): 862-875, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29848777

RESUMEN

The ß1-adrenergic receptor (ß1-AR) is a major cardiac G protein-coupled receptor, which mediates cardiac actions of catecholamines and is involved in genesis and treatment of numerous cardiovascular disorders. In mammalian cells, catecholamines induce the internalization of the ß1-AR into endosomes and their removal promotes the recycling of the endosomal ß1-AR back to the plasma membrane; however, whether these redistributive processes occur in terminally differentiated cells is unknown. Compartmentalization of the ß1-AR in response to ß-agonists and antagonists was determined by confocal microscopy in primary adult rat ventricular myocytes (ARVMs), which are terminally differentiated myocytes with unique structures such as transverse tubules (T-tubules) and contractile sarcomeres. In unstimulated ARVMs, the fluorescently labeled ß1-AR was expressed on the external membrane (the sarcolemma) of cardiomyocytes. Exposing ARVMs to isoproterenol redistributed surface ß1-ARs into small (∼225-250 nm) regularly spaced internal punctate structures that overlapped with puncta stained by Di-8 ANEPPS, a membrane-impermeant T-tubule-specific dye. Replacing the ß-agonist with the ß-blocker alprenolol, induced the translocation of the wild-type ß1-AR from these punctate structures back to the plasma membrane. This step was dependent on two barcodes, namely, the type-1 PDZ binding motif and serine at position 312 of the ß1-AR, which is phosphorylated by a pool of cAMP-dependent protein kinases anchored at the type-1 PDZ of the ß1-AR. These data show that redistribution of the ß1-AR in ARVMs from internal structures back to the plasma membrane was mediated by a novel sorting mechanism, which might explain unique aspects of cardiac ß1-AR signaling under normal or pathologic conditions.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 1/farmacología , Antagonistas de Receptores Adrenérgicos beta 1/farmacología , Miocitos Cardíacos/citología , Receptores Adrenérgicos beta 1/metabolismo , Alprenolol/farmacología , Animales , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Isoproterenol/farmacología , Miocitos Cardíacos/metabolismo , Transporte de Proteínas/efectos de los fármacos , Ratas , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/genética
5.
Methods Cell Biol ; 142: 67-78, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28964341

RESUMEN

G protein-coupled receptors (GPCRs) are recognized as one of the most fruitful group of therapeutic targets, accounting for more than 40% of all approved pharmaceuticals on the market. Therefore, the search for selective agents that affect GPCR function is of major interest to the pharmaceutical industry. This chapter describes methods for measuring agonist-promoted GPCR trafficking, which involves the internalization of the GPCR and its subsequent recycling back to the plasma membrane or retention and eventual degradation. These pathways will be analyzed by confocal cellular imaging, using the ß1-adrenergic receptor (ß1-AR) as a primary model. A major problem encountered in studying GPCR trafficking is the unavailability of antibodies that would recognize the native receptor in cells or tissues. Therefore, wild-type, point mutants, and ß1-AR chimeras are generated as epitope-tagged proteins, which are stably- or transiently expressed in mammalian cells. GPCR are labeled with a fluorophore-conjugated antibody directed against the N-terminal epitope tag. The trafficking of the fluorophore-tagged GPCR between divergent trafficking pathways that result in retention and eventual degradation or recycling and reinsertion into the plasma membrane can be followed by confocal immunofluorescence microscopy techniques outlined in this review.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 1/farmacología , Membrana Celular/metabolismo , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Transporte de Proteínas/efectos de los fármacos , Receptores Adrenérgicos beta 1/metabolismo , Animales , Endosomas/metabolismo , Colorantes Fluorescentes/química , Células HEK293 , Humanos , Ratones , MicroARNs/análisis , MicroARNs/química , Mutagénesis , Miocitos Cardíacos , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos beta 1/genética , Coloración y Etiquetado/métodos
6.
Cell Signal ; 36: 42-55, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28449947

RESUMEN

Proper signaling by G protein coupled receptors (GPCR) is dependent on the specific repertoire of transducing, enzymatic and regulatory kinases and phosphatases that shape its signaling output. Activation and signaling of the GPCR through its cognate G protein is impacted by G protein-coupled receptor kinase (GRK)-imprinted "barcodes" that recruit ß-arrestins to regulate subsequent desensitization, biased signaling and endocytosis of the GPCR. The outcome of agonist-internalized GPCR in endosomes is also regulated by sequence motifs or "barcodes" within the GPCR that mediate its recycling to the plasma membrane or retention and eventual degradation as well as its subsequent signaling in endosomes. Given the vast number of diverse sequences in GPCR, several trafficking mechanisms for endosomal GPCR have been described. The majority of recycling GPCR, are sorted out of endosomes in a "sequence-dependent pathway" anchored around a type-1 PDZ-binding module found in their C-tails. For a subset of these GPCR, a second "barcode" imprinted onto specific GPCR serine/threonine residues by compartmentalized kinase networks was required for their efficient recycling through the "sequence-dependent pathway". Mutating the serine/threonine residues involved, produced dramatic effects on GPCR trafficking, indicating that they played a major role in setting the trafficking itinerary of these GPCR. While endosomal SNX27, retromer/WASH complexes and actin were required for efficient sorting and budding of all these GPCR, additional proteins were required for GPCR sorting via the second "barcode". Here we will review recent developments in GPCR trafficking in general and the human ß1-adrenergic receptor in particular across the various trafficking roadmaps. In addition, we will discuss the role of GPCR trafficking in regulating endosomal GPCR signaling, which promote biochemical and physiological effects that are distinct from those generated by the GPCR signal transduction pathway in membranes.


Asunto(s)
Transporte de Proteínas , Receptores Acoplados a Proteínas G/metabolismo , Animales , Endosomas/metabolismo , Retroalimentación Fisiológica , Humanos , Procesamiento Proteico-Postraduccional , Transducción de Señal
7.
Cell Signal ; 29: 192-208, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27816670

RESUMEN

Recycling of the majority of agonist-internalized GPCR is dependent on a type I-PDZ "barcode" in their C-tail. The recycling of wild-type (WT) ß1-AR is also dependent on its default "type-1 PDZ barcode", but trafficking of the ß1-AR is inhibited when PKA or its substrate serine at position 312 (Ser312) are inactivated. We tested the hypothesis that phospho-Ser312 provided a second barcode for ß1-AR sorting from endosomes to the plasma membrane by determining the role of retromer/WASH complexes in ß1-AR trafficking. Recycling of WT ß1-AR or WT ß2-AR was dependent on targeting the retromer to endosomal membranes via SNX3 and rab7a, and on complexing the retromer to the WASH pentamer via the C-tail of FAM21 (FAM21C). These maneuvers however, did not inhibit the recycling of a phospho-Ser312 ß1-AR mimic ((S312D) ß1-AR). Knockdown of the trans-acting PDZ protein sorting nexin27 (SNX27) inhibited the recycling of WT ß1-AR and WT ß2-AR, but had no effect on (S312D) ß1-AR∆PDZ or on phosphorylation of WT ß1-AR by PKA at Ser312. However, depletion of FKBP15, a FAM21C-binding endosomal protein, selectively inhibited WT ß1-AR but not ß2-AR recycling, suggesting divergence might exist in GPCR trafficking roadmaps. These results indicate that two barcodes are involved in sorting WT ß1-AR out of early endosomes. The first and antecedent "barcode" was the "type-1 PDZ", followed by a second reversible "phospho-Ser312" verification "barcode". This organization allows tight regulation of ß1-AR density to signaling intensity in conditions associated with aberrant ß1-AR signaling such as in hypertension and heart failure.


Asunto(s)
Membrana Celular/metabolismo , Endosomas/metabolismo , Complejos Multiproteicos/metabolismo , Dominios PDZ , Fosfoserina/metabolismo , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/metabolismo , Membrana Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Endocitosis/efectos de los fármacos , Endosomas/efectos de los fármacos , Células HEK293 , Humanos , Isoproterenol/farmacología , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Complejos Multiproteicos/química , Proteínas de Unión a Fosfato , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Proteínas/metabolismo , ARN Interferente Pequeño/metabolismo , Nexinas de Clasificación/metabolismo , Relación Estructura-Actividad , Proteínas de Unión a Tacrolimus , Familia de Proteínas del Síndrome de Wiskott-Aldrich , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión a GTP rab7
8.
Biochem Pharmacol ; 120: 22-32, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27645110

RESUMEN

ß1-Adrenergic receptor (ß1-AR) agonists and antagonists are widely used in the treatment of major cardiovascular diseases such as heart failure and hypertension. The ß1-AR like other G protein-coupled receptors (GPCRs) are endocytosed in response to intense agonist activation. Recycling of the agonist-internalized ß1-AR is dependent on its carboxy-terminal type-1 PSD-95/DLG/ZO1 (PDZ) and on phospho-serine312 in the third intracellular loop of the ß1-AR. Progressive elongation of the ß1-AR at its C-tail inactivated the PDZ-biding domain and inhibited the recycling of the ß1-AR. However, fusing a twenty amino acid peptide derived from the multiple cloning region of the mammalian expression vector pCDNA3 to the C-tail of the ß1-AR (ß1-AR[+20]) produced a chimeric ß1-AR that recycled rapidly and efficiently. The ß1-AR[+20] recycled in a type-1 PDZ and phospho-Ser312-independent manner, indicating that this peptide provided a general GPCR recycling signal. Fusing the enhanced yellow fluorescent protein (EYFP) down-stream of ß1-AR[+20] generated a ß1-AR-EYFP chimera that was expressed on the membrane and recycled efficiently after agonist-induced internalization. This construct trafficked in a PDZ-SNX27/retromer-independent manner. We also fused EYFP to the N-terminus of the ß1-AR to created EYFP-WT ß1-AR. This construct recycled in PDZ and SNX27/retromer dependent manner. These ß1-AR-EYFP constructs would be useful for high throughput screening (HTS) programs to identify new entities that would interfere with the recycling of agonist internalized GPCR that traffic in PDZ-dependent vs. PDZ-independent roadmaps.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 1/farmacología , Descubrimiento de Drogas/métodos , Endocitosis/efectos de los fármacos , Modelos Moleculares , Receptores Adrenérgicos beta 1/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Inmunoprecipitación , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Oligopéptidos/genética , Oligopéptidos/metabolismo , Dominios PDZ , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosfoserina/metabolismo , Plásmidos/química , Plásmidos/genética , Plásmidos/metabolismo , Interferencia de ARN , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/genética , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Nexinas de Clasificación/antagonistas & inhibidores , Nexinas de Clasificación/química , Nexinas de Clasificación/genética , Nexinas de Clasificación/metabolismo
9.
Clin Radiol ; 71(1): e35-40, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26602930

RESUMEN

AIM: To compare readers' performance in detecting architectural distortion (AD) compared with other breast cancer types using digital mammography. MATERIALS AND METHODS: Forty-one experienced breast screen readers (20 US and 21 Australian) were asked to read a single test set of 30 digitally acquired mammographic cases. Twenty cases had abnormal findings (10 with AD, 10 non-AD) and 10 cases were normal. Each reader was asked to locate and rate any abnormalities. Lesion and case-based performance was assessed. For each collection of readers (US; Australian; combined), jackknife free-response receiver operating characteristic (JAFROC), figure of merit (FOM), and inferred receiver operating characteristic (ROC), area under curve (Az) were calculated using JAFROC v.4.1 software. Readers' sensitivity, location sensitivity, JAFROC, FOM, ROC, Az scores were compared between cases groups using Wilcoxon's signed ranked test statistics. RESULTS: For lesion-based analysis, significantly lower location sensitivity (p=0.001) was shown on AD cases compared with non-AD cases for all reader collections. The case-based analysis demonstrated significantly lower ROC Az values (p=0.02) for the first collection of readers, and lower sensitivity for the second collection of readers (p=0.04) and all-readers collection (p=0.008), for AD compared with non-AD cases. CONCLUSIONS: The current work demonstrates that AD remains a challenging task for readers, even in the digital era.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Competencia Clínica , Anciano , Australia , Femenino , Humanos , Mamografía , Persona de Mediana Edad , Variaciones Dependientes del Observador , Intensificación de Imagen Radiográfica , Sensibilidad y Especificidad , Estados Unidos
10.
Endocrinology ; 156(4): 1283-91, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25607895

RESUMEN

Neural precursor cell expressed developmentally down-regulated protein 4 (Nedd4) is the prototypical protein in the Nedd4 ubiquitin ligase (E3) family, which governs ubiquitin-dependent endocytosis and/or degradation of plasma membrane proteins. Loss of Nedd4 results in embryonic or neonatal lethality in mice and reduced insulin/IGF-1 signaling in embryonic fibroblasts. To delineate the roles of Nedd4 in vivo, we examined the phenotypes of heterozygous knockout mice using a high-fat diet-induced obesity (HFDIO) model. We observed that Nedd4+/- mice are moderately insulin resistant but paradoxically protected against HFDIO. After high-fat diet feeding, Nedd4+/- mice showed less body weight gain, less fat mass, and smaller adipocytes vs the wild type. Despite ameliorated HFDIO, Nedd4+/- mice did not manifest improvement in glucose tolerance vs the wild type in both genders. Nedd4+/- male, but not female, mice displayed significantly lower fasting blood glucose levels and serum insulin levels. Under obesogenic conditions, Nedd4+/- mice displayed elevated stimulated lipolytic activity, primarily through a ß2-adrenergic receptor. Combined, these data support novel complex roles for Nedd4 in metabolic regulation involving altered insulin and ß-adrenergic signaling pathways.


Asunto(s)
Peso Corporal/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Resistencia a la Insulina/genética , Lipólisis/genética , Obesidad/genética , Ubiquitina-Proteína Ligasas/genética , Adipocitos/metabolismo , Animales , Glucemia/metabolismo , Dieta Alta en Grasa , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Femenino , Haploinsuficiencia , Insulina/metabolismo , Masculino , Ratones , Ratones Noqueados , Ubiquitina-Proteína Ligasas Nedd4 , Obesidad/metabolismo , Caracteres Sexuales , Transducción de Señal/genética , Ubiquitina-Proteína Ligasas/metabolismo
11.
Cardiovasc Res ; 104(2): 270-9, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25225170

RESUMEN

AIMS: Cardiac ß-adrenergic receptors (ß-AR) are key regulators of cardiac haemodynamics and size. The scaffolding protein A-kinase anchoring protein 79/150 (AKAP5) is a key regulator of myocardial signalling by ß-ARs. We examined the function of AKAP5 in regulating cardiac haemodynamics and size, and the role of ß-ARs and Ca(2+)-regulated intracellular signalling pathways in this phenomenon. METHODS AND RESULTS: We used echocardiographic, histological, genetic, and biochemical methods to examine the effect of ablation of AKAP5 on cardiac haemodynamics, size, and signalling in mice. AKAP5(-/-) mice exhibited enhanced signs of cardiac dilatation and dysfunction that progressed with age. Infusions of isoprenaline worsened cardiac haemodynamics in wild-type (WT) mice only, but increased the ratio of heart-to-body weight equally in WT and in AKAP5(-/-) mice. Mechanistically, loss of AKAP5 was associated with enhanced activity of cardiac calmodulin kinase II (CaMKII) and calcineurin (CaN) as indexed by nuclear factor of activated T-cell-luciferase activity. Loss of AKAP5 interfered with the recycling of cardiac ß1-ARs, which was mediated in part by CaN binding to AKAP5. Carvedilol reversed cardiac hypertrophy and haemodynamic deficiencies in AKAP5(-/-) mice by normalizing the activities of cardiac CaN and CaMKII. CONCLUSIONS: These findings identify a novel cardioprotective role for AKAP5 that is mediated by regulating the activities of cardiac CaN and CaMKII and highlight a significant role for cardiac ß-ARs in this phenomenon.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/deficiencia , Antagonistas Adrenérgicos/farmacología , Calcineurina/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Carbazoles/farmacología , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Propanolaminas/farmacología , Disfunción Ventricular Izquierda/tratamiento farmacológico , Función Ventricular Izquierda/efectos de los fármacos , Proteínas de Anclaje a la Quinasa A/genética , Proteínas de Anclaje a la Quinasa A/metabolismo , Animales , Señalización del Calcio/efectos de los fármacos , Carvedilol , Células Cultivadas , Hemodinámica/efectos de los fármacos , Hipertrofia Ventricular Izquierda/enzimología , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Factores de Transcripción NFATC/metabolismo , Ratas Sprague-Dawley , Receptores Adrenérgicos beta 1/efectos de los fármacos , Receptores Adrenérgicos beta 1/metabolismo , Factores de Tiempo , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/fisiopatología
12.
J Biol Chem ; 289(4): 2277-94, 2014 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-24324269

RESUMEN

The ß1-adrenergic receptor (ß1-AR) is a target for treatment of major cardiovascular diseases, such as heart failure and hypertension. Recycling of agonist-internalized ß1-AR is dependent on type I PSD-95/DLG/ZO1 (PDZ) in the C-tail of the ß1-AR and on protein kinase A (PKA) activity (Gardner, L. A., Naren, A. P., and Bahouth, S. W. (2007) J. Biol. Chem. 282, 5085-5099). We explored the effects of point mutations in the PDZ and in the activity of PKA on recycling of the ß1-AR and its binding to the PDZ-binding protein SAP97. These studies indicated that ß1-AR recycling was inhibited by PKA inhibitors and by mutations in the PDZ that interfered with SAP97 binding. The trafficking effects of short sequences differing in PDZ and SAP97 binding were examined using chimeric mutant ß1-AR. ß1-AR chimera containing the type I PDZ of the ß2-adrenergic receptor that does not bind to SAP97 failed to recycle except when serine 312 was mutated to aspartic acid. ß1-AR chimera with type I PDZ sequences from the C-tails of aquaporin-2 or GluR1 recycled in a SAP97- and PKA-dependent manner. Non-PDZ ß1-AR chimera derived from µ-opioid, dopamine 1, or GluR2 receptors promoted rapid recycling of chimeric ß1-AR in a SAP97- and PKA-independent manner. Moreover, the nature of the residue at position -3 in the PDZ regulated whether the ß1-AR was internalized alone or in complex with SAP97. These results indicate that divergent pathways were involved in trafficking the ß1-AR and provide a roadmap for its trafficking via type I PDZs versus non-PDZs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Homólogo 1 de la Proteína Discs Large , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Estructura Terciaria de Proteína , Transporte de Proteínas/fisiología , Receptores AMPA/genética , Receptores AMPA/metabolismo , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
13.
J Biol Chem ; 288(47): 33797-33812, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24121510

RESUMEN

Protein kinase A-anchoring proteins (AKAPs) participate in the formation of macromolecular signaling complexes that include protein kinases, ion channels, effector enzymes, and G-protein-coupled receptors. We examined the role of AKAP79/150 (AKAP5) in trafficking and signaling of the ß1-adrenergic receptor (ß1-AR). shRNA-mediated down-regulation of AKAP5 in HEK-293 cells inhibited the recycling of the ß1-AR. Recycling of the ß1-AR in AKAP5 knockdown cells was rescued by shRNA-resistant AKAP5. However, truncated mutants of AKAP5 with deletions in the domains involved in membrane targeting or in binding to calcineurin or PKA failed to restore the recycling of the ß1-AR, indicating that full-length AKAP5 was required. Furthermore, recycling of the ß1-AR in rat neonatal cardiac myocytes was dependent on targeting the AKAP5-PKA complex to the C-terminal tail of the ß1-AR. To analyze the role of AKAP5 more directly, recycling of the ß1-AR was determined in ventricular myocytes from AKAP5(-/-) mice. In AKAP5(-/-) myocytes, the agonist-internalized ß1-AR did not recycle, except when full-length AKAP5 was reintroduced. These data indicate that AKAP5 exerted specific and profound effects on ß1-AR recycling in mammalian cells. Biochemical or real time FRET-based imaging of cyclic AMP revealed that deletion of AKAP5 sensitized the cardiac ß1-AR signaling pathway to isoproterenol. Moreover, isoproterenol-mediated increase in contraction rate, surface area, or expression of ß-myosin heavy chains was significantly greater in AKAP5(-/-) myocytes than in AKAP5(+/+) myocytes. These results indicate a significant role for the AKAP5 scaffold in signaling and trafficking of the ß1-AR in cardiac myocytes and mammalian cells.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Miocitos Cardíacos/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Transducción de Señal/fisiología , Proteínas de Anclaje a la Quinasa A/genética , Agonistas de Receptores Adrenérgicos beta 1/farmacología , Secuencia de Aminoácidos , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células HEK293 , Humanos , Isoproterenol/farmacología , Ratones , Ratones Noqueados , Miocitos Cardíacos/citología , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Ratas , Receptores Adrenérgicos beta 1/genética , Eliminación de Secuencia , Transducción de Señal/efectos de los fármacos
14.
J Clin Endocrinol Metab ; 98(9): E1448-55, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23824424

RESUMEN

CONTEXT: Human epicardial fat has been designated previously as brown-like fat. The supraclavicular fat depot in man has been defined as beige coexistent with classical brown based on its gene expression profile. OBJECTIVE: The aim of the study was to establish the gene expression profile and morphology of human epicardial and visceral paracardial fat compared with sc fat. SETTING: The study was conducted at a tertiary care hospital cardiac center. PATIENTS: Epicardial, visceral paracardial, and sc fat samples had been taken from middle-aged patients with severe coronary atherosclerosis or valvular heart disease. INTERVENTIONS: Gene expression was determined by reverse transcription-quantitative PCR and relative abundance of the mitochondrial uncoupling protein-1 (UCP-1) by Western blotting. Epicardial tissue sections from patients were examined by light microscopy, UCP-1 immunohistochemistry, and cell morphometry. MAIN OUTCOME MEASURES: We hypothesized that epicardial fat has a mixed phenotype with a gene expression profile similar to that described for beige cell lineage. RESULTS: Immunoreactive UCP-1 was clearly measurable in each epicardial sample analyzed but was undetectable in each of the 4 other visceral and sc depots. Epicardial fat exhibited high expression of genes for UCP-1, PRDM16, PGC-1α, PPARγ, and the beige adipocyte-specific marker CD137, which were also expressed in visceral paracardial fat but only weakly in sternal, upper abdominal, and lower extremity sc fat. Histology of epicardial fat showed small unilocular adipocytes without UCP-1 immunostaining. CONCLUSION: UCP-1 is relatively abundant in epicardial fat, and this depot possesses molecular features characteristic of those found in vitro in beige lineage adipocytes.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Grasa Intraabdominal/metabolismo , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Pericardio/metabolismo , Anciano , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Expresión Génica , Humanos , Canales Iónicos/genética , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/genética , PPAR gamma/genética , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Proteína Desacopladora 1
15.
Metabolism ; 62(10): 1503-11, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23831442

RESUMEN

BACKGROUND: Exercise training elevates circulating irisin and induces the expression of the FNDC5 gene in skeletal muscles of mice. Our objective was to determine whether exercise training also increases FNDC5 protein or mRNA expression in the skeletal muscles of pigs as well as plasma irisin. METHODS: Castrated male pigs of the Rapacz familial hypercholesterolemic (FHM) strain and normal (Yucatan miniature) pigs were sacrificed after 16-20 weeks of exercise training. Samples of cardiac muscle, deltoid and triceps brachii muscle, subcutaneous and epicardial fat were obtained and FNDC5 mRNA, along with that of 6 other genes, was measured in all tissues of FHM pigs by reverse transcription polymerase chain reaction. FNDC protein in deltoid and triceps brachii was determined by Western blotting in both FHM and normal pigs. Citrate synthase activity was measured in the muscle samples of all pigs as an index of exercise training. Irisin was measured by an ELISA assay. RESULTS: There was no statistically significant effect of exercise training on FNDC5 gene expression in epicardial or subcutaneous fat, deltoid muscle, triceps brachii muscle or heart muscle. Exercise-training elevated circulating levels of irisin in the FHM pigs and citrate synthase activity in deltoid and triceps brachii muscle. A similar increase in citrate synthase activity was seen in muscle extracts of exercise-trained normal pigs but there was no alteration in circulating irisin. CONCLUSION: Exercise training in pigs does not increase FNDC5 mRNA or protein in the deltoid or triceps brachii of FHM or normal pigs while increasing circulating irisin only in the FHM pigs. These data indicate that the response to exercise training in normal pigs is not comparable to that seen in mice.


Asunto(s)
Fibronectinas/genética , Fibronectinas/metabolismo , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Condicionamiento Físico Animal , ARN Mensajero/genética , Porcinos/fisiología , Animales , Citrato (si)-Sintasa/metabolismo , Expresión Génica/genética , Masculino , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Grasa Subcutánea/metabolismo , Porcinos/genética , Porcinos/metabolismo
16.
PLoS One ; 8(5): e63379, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23696820

RESUMEN

Previous studies have determined that the type-1 PDZ sequence at the extreme carboxy-terminus of the ß1-adrenergic receptor (ß1-AR) binds SAP97 and AKAP79 to organize a scaffold involved in trafficking of the ß1-AR. In this study we focused on characterizing the domains in SAP97 that were involved in recycling and resensitization of the ß1-AR in HEK-293 cells. Using a SAP97 knockdown and rescue strategy, we determined that PDZ-deletion mutants of SAP97 containing PDZ2 rescued the recycling and resensitization of the ß1-AR. Among the three PDZs of SAP97, PDZ2 displayed the highest affinity in binding to the ß1-AR. Expression of isolated PDZ2, but not the other PDZs, inhibited the recycling of the ß1-AR by destabilizing the macromolecular complex involved in trafficking and functional resensitization of the ß1-AR. In addition to its PDZs, SAP97 contains other protein interacting domains, such as the I3 sequence in the SRC homology-3 (SH3) domain, which binds to AKAP79. Deletion of I3 from SAP97 (ΔI3-SAP97) did not affect the binding of SAP97 to the ß1-AR. However, ΔI3-SAP97 could not rescue the recycling of the ß1-AR because it failed to incorporate AKAP79/PKA into the SAP97-ß1-AR complex. Therefore, bipartite binding of SAP97 to the ß1-AR and to AKAP79 is necessary for SAP97-mediated effects on recycling, externalization and functional resensitization of the ß1-AR. These data establish a prominent role for PDZ2 and I3 domains of SAP97 in organizing the ß1-adrenergic receptosome involved in connecting the ß1-AR to trafficking and signaling networks.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular , Humanos , Inmunoprecipitación , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Receptores Adrenérgicos beta 1/genética , Transducción de Señal
17.
J Recept Signal Transduct Res ; 33(2): 79-88, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23351074

RESUMEN

Abstract A cluster of hydrophobic amino acids at the cytoplasmic end of trans-membranal helix III (TM-III) is a common feature among class-A of G protein-coupled receptors (GPCR). We mutagenized alanine 159(3.53) to glutamic acid and isoleucine160(3.54) to arginine (A159E/I160R) in TM-III of the human ß(1)-adrenergic receptor (ß(1)-AR) to disrupt the function of the hydrophobic cluster. Structurally, the combined mutations of A159E/I160R caused an almost 90° tilt in the rotation of Arg156(3.50) in the E/DRY motif of TM-III and displaced Tyr166(3.60) in intracellular loop 2. The A159E/I160R ß(1)-AR was uncoupled from G(s) as determined by cyclic AMP/adenylyl cyclase assays and by FRET-based proximity measurements between the ß(1)-AR and G(s)α. Isoproterenol induced ß-arrestin trafficking in cells expressing both the wild-type ß(1)-AR and the A159E/I160R ß(1)-AR. Isoproterenol markedly increased the phosphorylation of ERK1/2 in cells expressing the WT ß(1)-AR and this effect was dependent on the activation of the G(s)-cyclic AMP-dependent protein kinase → Rap → B-raf axis. However, in cells bearing the A159E/I160R ß(1)-AR, isoproterenol failed to increase the phosphorylation of ERK(1/2). These results indicate that mutations in the G(s)α-binding pocket of the GPCR interfered with receptor coupling to G(s) and with its downstream signaling cascades.


Asunto(s)
Aminoácidos/química , Citoplasma/metabolismo , Proteínas de Unión al GTP/química , Receptores Adrenérgicos beta 1/química , Aminoácidos/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Isoproterenol/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/química , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Modelos Moleculares , Mutación , Fosforilación/efectos de los fármacos , Conformación Proteica , Receptores Adrenérgicos beta 1/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
18.
Metab Syndr Relat Disord ; 9(6): 433-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21679057

RESUMEN

BACKGROUND: Pro- and antiinflammatory genes are expressed in epicardial adipose tissue (EAT). Our objectives were to characterize genes in EAT that may contribute specifically to coronary atherogenesis and to measure circulating adipokines matched to their messenger RNAs (mRNAs) in EAT. We hypothesized that severe coronary atherosclerosis (CAD) would preferentially affect gene expression in EAT as compared to substernal fat or subcutaneous thoracic adipose tissue (SAT), as well as circulating levels of adipokines. METHODS: Fat mRNA was quantified using reverse transcription polymerase chain reaction (RT-PCR), and circulating adipokines were measured by enzyme-linked immunosorbent assays (ELISAs) in patients with severe stable CAD and controls without severe CAD undergoing open heart surgery. RESULTS: A total of 39 of 70 mRNAs in EAT were significantly increased in CAD. Only 4 and 3 of these mRNAs were increased in substernal fat and SAT, respectively. Of the mRNAs increased in EAT, 17 were either inflammatory adipokines or proteins known to be involved in inflammatory processes, 7 were involved in oxidative stress and or oxygen species regulation, whereas 15 were proteins involved in metabolism and regulation of gene transcription or proteins unique to fat cells. The largest increases, over three-fold, were seen in GPX3, gp91 phox, p47phox, heme oxygenase, and interleukin-8 (IL-8). Tpl2 mRNA was uniquely elevated in all three fat depots from CAD patients, and its expression in SAT, but not in EAT or substernal fat, was directly correlated with homeostasis model assessment of insulin resistance (HOMA-IR) values. Compared to controls, there were no associations between circulating levels of IL-8, lipocalin-2, nerve growth factor (NGF), RANTES, CD-163, GPX-3, monocyte chemotactic protein-1 (MCP-1)/CCL2, leptin, soluble vascular endothelial growth factor receptor-1 (sFLT1), fatty acid binding protein-4 (FABP-4), and plasminogen activator inhibitor-1 (PAI-1) and increases in their gene expression in EAT adjacent to CAD. CONCLUSIONS: Expression of proinflammatory, redox, endothelial cell, and angiogenic genes in EAT is depot specific and supports the hypothesis that pathophysiologically EAT contributes locally to CAD. CAD links with these fat depots might involve Tpl2 as a primary response indicator.


Asunto(s)
Tejido Adiposo/metabolismo , Enfermedad de la Arteria Coronaria/genética , Células Endoteliales/metabolismo , Inflamación/genética , Neovascularización Fisiológica/genética , Pericardio/metabolismo , Tejido Adiposo/patología , Anciano , Inductores de la Angiogénesis/metabolismo , Estudios de Casos y Controles , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Progresión de la Enfermedad , Células Endoteliales/patología , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Oxidación-Reducción , Índice de Severidad de la Enfermedad , Regulación hacia Arriba/genética
19.
Diabetes Care ; 34(3): 730-3, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21289232

RESUMEN

OBJECTIVE: To determine changes in gene expression in epicardial adipose tissue (EAT) associated with coronary atherosclerosis (CAD) and effects of pioglitazone therapy. RESEARCH DESIGN AND METHODS: Genes were quantified by RT-PCR in EAT and thoracic subcutaneous adipose tissue (SAT) obtained during surgery in CAD patients with metabolic syndrome (MS) or type 2 diabetes and control subjects with minimal or no CAD and no MS or type 2 diabetes. RESULTS: Increased expression of interleukin-1 receptor antagonist (IL-1Ra) and IL-10, a trend for higher IL-1ß, and no change in peroxisome proliferator-activated receptor-γ (PPARγ) was found in EAT from MS or type 2 diabetes. Only PPARγ mRNA was reduced in SAT. Pioglitazone therapy in type 2 diabetes was associated with decreased expression of IL-1ß, IL-1Ra, and IL-10 in EAT; decreased IL-10 in SAT; and increased PPARγ in SAT. CONCLUSIONS: In MS and type 2 diabetes with CAD, proinflammatory and anti-inflammatory genes were differentially increased in EAT and selectively reduced in association with pioglitazone treatment.


Asunto(s)
Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Síndrome Metabólico/tratamiento farmacológico , Pericardio/metabolismo , Tiazolidinedionas/uso terapéutico , Enfermedad de la Arteria Coronaria/genética , Diabetes Mellitus Tipo 2/genética , Humanos , Proteína Antagonista del Receptor de Interleucina 1/genética , Interleucina-10/genética , Interleucina-1beta/genética , Síndrome Metabólico/genética , Pioglitazona , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Cell Signal ; 23(1): 46-57, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20727405

RESUMEN

ß1-adrenergic receptors (ß1-AR) are internalized in response to agonists and then recycle back for another round of signaling. The serine 312 to alanine mutant of the ß1-AR (S312A) is internalized but does not recycle. We determined that WT ß1-AR and S312A were internalized initially to an early sorting compartment because they colocalized by >70% with the early endosomal markers rab5a and early endosomal antigen-1 (EEA1). Subsequently, the WT ß1-AR trafficked via rab4a-expressing sorting endosomes to recycling endosomes. In recycling endosomes WT ß1-AR were colocalized by >70% with the rab11 GTPase. S312A did not colocalize with either rab4a or rab11, instead they exited from early endosomes to late endosomes/lysosomes in which they were degraded. Rab11a played a prominent role in recycling of the WT ß1-AR because dominant negative rab11a inhibited, while constitutively active rab11a accelerated the recycling of the ß1-AR. Next, we determined the effect of each of the rab11-interacting proteins on trafficking of the WT ß1-AR. The recycling of the ß1-AR was markedly inhibited when myosin Vb, FIP2, FIP3 and rabphillin were knocked down. These data indicate that rab11a and a select group of its binding partners play a prominent role in recycling of the human ß1-AR.


Asunto(s)
Receptores Adrenérgicos beta 1/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Agonistas Adrenérgicos beta/farmacología , Sustitución de Aminoácidos , Línea Celular , Endosomas/metabolismo , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Isoproterenol/farmacología , Lisosomas/metabolismo , Mutagénesis Sitio-Dirigida , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo V/genética , Miosina Tipo V/metabolismo , Interferencia de ARN , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al GTP rab/fisiología , Proteínas de Unión al GTP rab4/metabolismo , Proteínas de Unión al GTP rab4/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA