Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
J Am Soc Nephrol ; 24(5): 744-58, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23559583

RESUMO

In the principal cells of the renal collecting duct, arginine vasopressin (AVP) stimulates the synthesis of cAMP, leading to signaling events that culminate in the phosphorylation of aquaporin-2 water channels and their redistribution from intracellular domains to the plasma membrane via vesicular trafficking. The molecular mechanisms that control aquaporin-2 trafficking and the consequent water reabsorption, however, are not completely understood. Here, we used a cell-based assay and automated immunofluorescence microscopy to screen 17,700 small molecules for inhibitors of the cAMP-dependent redistribution of aquaporin-2. This approach identified 17 inhibitors, including 4-acetyldiphyllin, a selective blocker of vacuolar H(+)-ATPase that increases the pH of intracellular vesicles and causes accumulation of aquaporin-2 in the Golgi compartment. Although 4-acetyldiphyllin did not inhibit forskolin-induced increases in cAMP formation and downstream activation of protein kinase A (PKA), it did prevent cAMP/PKA-dependent phosphorylation at serine 256 of aquaporin-2, which triggers the redistribution to the plasma membrane. It did not, however, prevent cAMP-induced changes to the phosphorylation status at serines 261 or 269. Last, we identified the fungicide fluconazole as an inhibitor of cAMP-mediated redistribution of aquaporin-2, but its target in this pathway remains unknown. In conclusion, our screening approach provides a method to begin dissecting molecular mechanisms underlying AVP-mediated water reabsorption, evidenced by our identification of 4-acetyldiphyllin as a modulator of aquaporin-2 trafficking.


Assuntos
Aquaporina 2/metabolismo , Benzodioxóis/farmacologia , Benzofuranos/farmacologia , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , Animais , Células Cultivadas , Colforsina/farmacologia , AMP Cíclico/fisiologia , Fluconazol/farmacologia , Complexo de Golgi/metabolismo , Ensaios de Triagem em Larga Escala , Humanos , Camundongos , Microscopia de Fluorescência , Fosforilação , Transporte Proteico/efeitos dos fármacos , Ratos
3.
J Biol Chem ; 286(11): 9079-96, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21177871

RESUMO

A-kinase anchoring proteins (AKAPs) tether protein kinase A (PKA) and other signaling proteins to defined intracellular sites, thereby establishing compartmentalized cAMP signaling. AKAP-PKA interactions play key roles in various cellular processes, including the regulation of cardiac myocyte contractility. We discovered small molecules, 3,3'-diamino-4,4'-dihydroxydiphenylmethane (FMP-API-1) and its derivatives, which inhibit AKAP-PKA interactions in vitro and in cultured cardiac myocytes. The molecules bind to an allosteric site of regulatory subunits of PKA identifying a hitherto unrecognized region that controls AKAP-PKA interactions. FMP-API-1 also activates PKA. The net effect of FMP-API-1 is a selective interference with compartmentalized cAMP signaling. In cardiac myocytes, FMP-API-1 reveals a novel mechanism involved in terminating ß-adrenoreceptor-induced cAMP synthesis. In addition, FMP-API-1 leads to an increase in contractility of cultured rat cardiac myocytes and intact hearts. Thus, FMP-API-1 represents not only a novel means to study compartmentalized cAMP/PKA signaling but, due to its effects on cardiac myocytes and intact hearts, provides the basis for a new concept in the treatment of chronic heart failure.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Miócitos Cardíacos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Sistemas do Segundo Mensageiro/fisiologia , Proteínas de Ancoragem à Quinase A/genética , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/genética , Animais , Doença Crônica , AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Masculino , Contração Miocárdica/efeitos dos fármacos , Ratos , Ratos Endogâmicos WKY , Sistemas do Segundo Mensageiro/efeitos dos fármacos
4.
Int Rev Cell Mol Biol ; 283: 235-330, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20801421

RESUMO

The second messenger cyclic adenosine monophosphate (cAMP), which is produced by adenylyl cyclases following stimulation of G-protein-coupled receptors, exerts its effect mainly through the cAMP-dependent serine/threonine protein kinase A (PKA). Due to the ubiquitous nature of the cAMP/PKA system, PKA signaling pathways underlie strict spatial and temporal control to achieve specificity. A-kinase anchoring proteins (AKAPs) bind to the regulatory subunit dimer of the tetrameric PKA holoenzyme and thereby target PKA to defined cellular compartments in the vicinity of its substrates. AKAPs promote the termination of cAMP signals by recruiting phosphodiesterases and protein phosphatases, and the integration of signaling pathways by binding additional signaling proteins. AKAPs are a heterogeneous family of proteins that only display similarity within their PKA-binding domains, amphipathic helixes docking into a hydrophobic groove formed by the PKA regulatory subunit dimer. This review summarizes the current state of information on compartmentalized cAMP/PKA signaling with a major focus on structural aspects, evolution, diversity, and (patho)physiological functions of AKAPs and intends to outline newly emerging directions of the field, such as the elucidation of AKAP mutations and alterations of AKAP expression in human diseases, and the validation of AKAP-dependent protein-protein interactions as new drug targets. In addition, alternative PKA anchoring mechanisms employed by noncanonical AKAPs and PKA catalytic subunit-interacting proteins are illustrated.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Animais , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/química , Doença , Humanos , Transdução de Sinais
5.
J Am Soc Nephrol ; 21(10): 1645-56, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20724536

RESUMO

Arginine-vasopressin (AVP) modulates the water channel aquaporin-2 (AQP2) in the renal collecting duct to maintain homeostasis of body water. AVP binds to vasopressin V2 receptors (V2R), increasing cAMP, which promotes the redistribution of AQP2 from intracellular vesicles into the plasma membrane. cAMP also increases AQP2 transcription, but whether altered degradation also modulates AQP2 protein levels is not well understood. Here, elevation of cAMP increased AQP2 protein levels within 30 minutes in primary inner medullary collecting duct (IMCD) cells, in human embryonic kidney (HEK) 293 cells ectopically expressing AQP2, and in mouse kidneys. Accelerated transcription or translation did not explain this increase in AQP2 abundance. In IMCD cells, cAMP inhibited p38-mitogen-activated protein kinase (p38-MAPK) via activation of protein kinase A (PKA). Inhibition of p38-MAPK associated with decreased phosphorylation (serine 261) and polyubiquitination of AQP2, preventing proteasomal degradation. Our results demonstrate that AVP enhances AQP2 protein abundance by altering its proteasomal degradation through a PKA- and p38-MAPK-dependent pathway.


Assuntos
Aquaporina 2/metabolismo , Arginina Vasopressina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Medula Renal/metabolismo , Túbulos Renais Coletores/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , Colforsina , AMP Cíclico/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Biossíntese de Proteínas , Ratos , Transcrição Gênica
6.
J Biol Chem ; 285(8): 5507-21, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20007971

RESUMO

A-kinase anchoring proteins (AKAPs) include a family of scaffolding proteins that target protein kinase A (PKA) and other signaling proteins to cellular compartments and thereby confine the activities of the associated proteins to distinct regions within cells. AKAPs bind PKA directly. The interaction is mediated by the dimerization and docking domain of regulatory subunits of PKA and the PKA-binding domain of AKAPs. Analysis of the interactions between the dimerization and docking domain and various PKA-binding domains yielded a generalized motif allowing the identification of AKAPs. Our bioinformatics and peptide array screening approaches based on this signature motif identified GSKIP (glycogen synthase kinase 3beta interaction protein) as an AKAP. GSKIP directly interacts with PKA and GSK3beta (glycogen synthase kinase 3beta). It is widely expressed and facilitates phosphorylation and thus inactivation of GSK3beta by PKA. GSKIP contains the evolutionarily conserved domain of unknown function 727. We show here that this domain of GSKIP and its vertebrate orthologues binds both PKA and GSK3beta and thereby provides a mechanism for the integration of PKA and GSK3beta signaling pathways.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Ancoragem à Quinase A/genética , Motivos de Aminoácidos/fisiologia , Linhagem Celular Tumoral , Proteínas Quinases Dependentes de AMP Cíclico/genética , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Ligação Proteica/fisiologia , Multimerização Proteica/fisiologia , Estrutura Terciária de Proteína/fisiologia
7.
J Pept Sci ; 15(7): 479-91, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19466696

RESUMO

The molecular basis for recognition of peptide ligands endothelin-1, -2 and -3 in endothelin receptors is poorly understood. Especially the origin of ligand selectivity for ET(A) or ET(B) is not clearly resolved. We derived sequence-structure-function relationships of peptides and receptors from mutational data and homology modeling. Our major findings are the dissection of peptide ligands into four epitopes and the delineation of four complementary structural portions on receptor side explaining ligand recognition in both endothelin receptor subtypes. In addition, structural determinants for ligand selectivity could be described. As a result, we could improve the selectivity of BQ3020 about 10-fold by a single amino acid substitution, validating our hypothesis for ligand selectivity caused by different entrances to the receptors' transmembrane binding sites. A narrow tunnel shape in ET(A) is restrictive for a selected group of peptide ligands' N-termini, whereas a broad funnel-shaped entrance in ET(B) accepts a variety of different shapes and properties of ligands.


Assuntos
Peptídeos/química , Receptor de Endotelina A/agonistas , Receptor de Endotelina A/química , Receptor de Endotelina B/agonistas , Receptor de Endotelina B/química , Animais , Sítios de Ligação , Bovinos , Humanos , Ligantes , Espectroscopia de Ressonância Magnética , Ligação Proteica , Estrutura Secundária de Proteína , Relação Estrutura-Atividade
8.
Handb Exp Pharmacol ; (190): 133-57, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19096775

RESUMO

Principal cells lining renal collecting ducts control the fine-tuning of body water homeostasis by regulating water reabsorption through the water channels aquaporin-2 (AQP2), aquaporin-3 (AQP3), and aquaporin-4 (AQP4). While the localization of AQP2 is subject to regulation by arginine-vasopressin (AVP), AQP3 and AQP4 are constitutively expressed in the basolateral plasma membrane. AVP adjusts the amount of AQP2 in the plasma membrane by triggering its redistribution from intracellular vesicles into the plasma membrane. This permits water entry into the cells and water exit through AQP3 and AQP4. The translocation of AQP2 is initiated by an increase in cAMP following V2R activation through AVP. The AVP-induced rise in cAMP activates protein kinase A (PKA), which in turn phosphorylates AQP2, and thereby triggers the redistribution of AQP2. Several proteins participating in the control of cAMP-dependent AQP2 trafficking have been identified; for example, A kinase anchoring proteins (AKAPs) tethering PKA to cellular compartments; phosphodiesterases (PDEs) regulating the local cAMP level; cytoskeletal components such as F-actin and microtubules; small GTPases of the Rho family controlling cytoskeletal dynamics; motor proteins transporting AQP2-bearing vesicles to and from the plasma membrane for exocytic insertion and endocytic retrieval; SNAREs inducing membrane fusions, hsc70, a chaperone, important for endocytic retrieval. In addition, cAMP-independent mechanisms of translocation mainly involving the F-actin cytoskeleton have been uncovered. Defects of AQP2 trafficking cause diseases such as nephrogenic diabetes insipidus (NDI), a disorder characterized by a massive loss of hypoosmotic urine.This review summarizes recent data elucidating molecular mechanisms underlying the trafficking of AQP2. In particular, we focus on proteins involved in the regulation of trafficking, and physiological and pathophysiological stimuli determining the cellular localization of AQP2. The identification of proteins and protein-protein interactions may lead to the development of drugs targeting AQP2 trafficking. Such drugs may be suitable for the treatment of diseases associated with dysregulation of body water homeostasis, including NDI or cardiovascular diseases (e.g., chronic heart failure) where the AVP level is elevated, inducing excessive water retention.


Assuntos
Aquaporina 2/metabolismo , Membrana Celular/metabolismo , Túbulos Renais Coletores/metabolismo , Equilíbrio Hidroeletrolítico , Água/metabolismo , Proteínas de Ancoragem à Quinase A/metabolismo , Animais , Arginina Vasopressina/metabolismo , Cálcio/metabolismo , Compartimento Celular , AMP Cíclico/metabolismo , Citoesqueleto/metabolismo , Diabetes Insípido Nefrogênico/metabolismo , Diabetes Insípido Nefrogênico/fisiopatologia , Endocitose , Homeostase , Humanos , Túbulos Renais Coletores/enzimologia , Diester Fosfórico Hidrolases/metabolismo , Fosforilação , Transporte Proteico , Transdução de Sinais
10.
Mol Pharmacol ; 73(3): 697-708, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18048502

RESUMO

The endoplasmic reticulum-associated degradation (ERAD), the main quality control pathway of the cell, is crucial for the elimination of unfolded or misfolded proteins. Several diseases are associated with the retention of misfolded proteins in the early secretory pathway. Among them is X-linked nephrogenic diabetes insipidus, caused by mutations in the gene encoding the V2 vasopressin receptor (V2R). We studied the degradation pathways of three intracellularly retained V2R mutants with different misfolded domains in human embryonic kidney 293 cells. At steady state, the wild-type V2R and the complex-glycosylated mutant G201D were partially located in lysosomes, whereas core-glycosylated mutants L62P and V226E were excluded from this compartment. In pulse-chase experiments, proteasomal inhibition stabilized the nonglycosylated and core-glycosylated forms of all studied receptors. In addition, all mutants and the wild-type receptor were found to be polyubiquitinylated. Nonglycosylated and core-glycosylated receptor forms were located in cytosolic and membrane fractions, respectively, confirming the deglycosylation and retrotranslocation of ERAD substrates to the cytosol. Distinct Derlin-1-dependent and -independent ERAD pathways have been proposed for proteins with different misfolded domains (cytosolic, extracellular, and membrane) in yeast. Here, we show for the first time that V2R mutants with different misfolded domains are able to coprecipitate the ERAD components p97/valosin-containing protein, Derlin-1 and the 26S proteasome regulatory subunit 7. Our results demonstrate the presence of a Derlin-1-mediated ERAD pathway degrading wild-type and disease-causing V2R mutants with different misfolded domains in a mammalian system.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Vasopressinas/química , Receptores de Vasopressinas/metabolismo , Adenilil Ciclases/análise , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Sequência de Aminoácidos , Arginina Vasopressina/farmacologia , Linhagem Celular , Cloroquina/farmacologia , Proteínas de Fluorescência Verde/metabolismo , Humanos , Rim/citologia , Lisossomos/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Plasmídeos , Estrutura Terciária de Proteína , Receptores de Vasopressinas/genética , Frações Subcelulares/metabolismo , Transfecção , Ubiquitinação , Proteína com Valosina
11.
Cell Signal ; 20(4): 590-601, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18061403

RESUMO

Cyclic adenosine monophosphate (cAMP) is a central second messenger controlling a plethora of vital functions. Studies of cAMP dynamics in living cells have revealed markedly inhomogeneous concentrations of the second messenger in different compartments. Moreover, cAMP effectors such as cAMP-dependent protein kinase (PKA) and cAMP-activated GTP-exchange factors (Epacs) are tethered to specific cellular sites. Both the tailoring of cAMP concentrations, and the activities of cAMP-dependent signalling systems at specific cellular locations are prerequisites for most, if not all, cAMP-dependent processes. This review focuses on the role of compartmentalized cAMP signalling in exocytic processes in non-neuronal cells. Particularly, the insertion of aquaporin-2 into the plasma membrane of renal principal cells as an example for a cAMP-dependent exocytic process in a non-secretory cell type, renin secretion from juxtaglomerular cells as a cAMP-triggered exocytosis from an endocrine cell, insulin release from pancreatic beta-cells as a Ca2+-mediated and cAMP-potentiated exocytic processes in an endocrine cell, and cAMP- or Ca2+ -triggered H+ secretion from gastric parietal cells as an exocytic process in an exocrine cell are discussed. The selected examples of cAMP-regulated exocytic pathways are reviewed with regard to key proteins involved: adenylyl cyclases, phosphodiesterases, PKA, A kinase anchoring proteins (AKAPs) and Epacs.


Assuntos
Compartimento Celular , AMP Cíclico/metabolismo , Exocitose , Sistemas do Segundo Mensageiro , Animais , Aquaporina 2/metabolismo , Cálcio/metabolismo , Ácido Gástrico/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Sistema Justaglomerular/citologia , Sistema Justaglomerular/metabolismo , Túbulos Renais/citologia , Túbulos Renais/metabolismo , Biologia Molecular , Células Parietais Gástricas/metabolismo , Renina/metabolismo
12.
EMBO Rep ; 8(11): 1061-7, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17901878

RESUMO

The beta-adrenergic receptor/cyclic AMP/protein kinase A (PKA) signalling pathway regulates heart rate and contractility. Here, we identified a supramolecular complex consisting of the sarcoplasmic reticulum Ca(2+)-ATPase (SERCA2), its negative regulator phospholamban (PLN), the A-kinase anchoring protein AKAP18delta and PKA. We show that AKAP18delta acts as a scaffold that coordinates PKA phosphorylation of PLN and the adrenergic effect on Ca(2+) re-uptake. Inhibition of the compartmentalization of this cAMP signalling complex by specific molecular disruptors interferes with the phosphorylation of PLN. This prevents the subsequent release of PLN from SERCA2, thereby affecting the Ca(2+) re-uptake into the sarcoplasmic reticulum induced by adrenergic stimuli.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Membrana/metabolismo , Miócitos Cardíacos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Proteínas de Ligação ao Cálcio/genética , Proteínas de Transporte/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas de Membrana/genética , Dados de Sequência Molecular , Complexos Multiproteicos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Ratos , Retículo Sarcoplasmático/ultraestrutura , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Alinhamento de Sequência
13.
Am J Physiol Cell Physiol ; 293(3): C1129-38, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17626240

RESUMO

Water reabsorption in the renal collecting duct is regulated by arginine vasopressin (AVP). AVP induces the insertion of the water channel aquaporin-2 (AQP2) into the plasma membrane of principal cells, thereby increasing the osmotic water permeability. The redistribution of AQP2 to the plasma membrane is a cAMP-dependent process and thus a paradigm for cAMP-controlled exocytic processes. Using primary cultured rat inner medullary collecting duct cells, we show that the redistribution of AQP2 to the plasma membrane is accompanied by the reorganization of microtubules and the redistribution of the small GTPase Rab11. In resting cells, AQP2 is colocalized with Rab11 perinuclearly. AVP induced the redistribution of AQP2 to the plasma membrane and of Rab11 to the cell periphery. The redistribution of both proteins was increased when microtubules were depolymerized by nocodazole. In addition, the depolymerization of microtubules prevented the perinuclear positioning of AQP2 and Rab11 in resting cells, which was restored if nocodazole was washed out and microtubules repolymerized. After internalization of AQP2, induced by removal of AVP, forskolin triggered the AQP2 redistribution to the plasma membrane even if microtubules were depolymerized and without the previous positioning of AQP2 in the perinuclear recycling compartment. Collectively, the data indicate that microtubule-dependent transport of AQP2 is predominantly responsible for trafficking and localization of AQP2 inside the cell after its internalization but not for the exocytic transport of the water channel. We also demonstrate that cAMP-signaling regulates the localization of Rab11-positive recycling endosomes in renal principal cells.


Assuntos
Aquaporina 2/metabolismo , Endocitose/fisiologia , Túbulos Renais Coletores/citologia , Microtúbulos/metabolismo , Animais , Antineoplásicos/farmacologia , Membrana Celular/metabolismo , Núcleo Celular , Células Cultivadas , AMP Cíclico/metabolismo , Dineínas/metabolismo , Exocitose/fisiologia , Complexo de Golgi/metabolismo , Microtúbulos/efeitos dos fármacos , Nocodazol/farmacologia , Ratos , Ratos Wistar , Proteínas rab de Ligação ao GTP/metabolismo
14.
Traffic ; 8(2): 110-23, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17156409

RESUMO

Arginine-vasopressin (AVP) regulates water reabsorption in renal collecting duct principal cells. Its binding to Gs-coupled vasopressin V2 receptors increases cyclic AMP (cAMP) and subsequently elicits the redistribution of the water channel aquaporin-2 (AQP2) from intracellular vesicles into the plasma membrane (AQP2 shuttle), thereby facilitating water reabsorption from primary urine. The AQP2 shuttle is a paradigm for cAMP-dependent exocytic processes. Using sections of rat kidney, the AQP2-expressing cell line CD8, and primary principal cells, we studied the role of the motor protein myosin Vb, its vesicular receptor Rab11, and the myosin Vb- and Rab11-binding protein Rab11-FIP2 in the AQP2 shuttle. Myosin Vb colocalized with AQP2 intracellularly in resting and at the plasma membrane in AVP-treated cells. Rab11 was found on AQP2-bearing vesicles. A dominant-negative myosin Vb tail construct and Rab11-FIP2 lacking the C2 domain (Rab11-FIP2-DeltaC2), which disrupt recycling, caused condensation of AQP2 in a Rab11-positive compartment and abolished the AQP2 shuttle. This effect was dependent on binding of myosin Vb tail and Rab11-FIP2-DeltaC2 to Rab11. In summary, we identified myosin Vb as a motor protein involved in AQP2 recycling and show that myosin Vb- and Rab11-FIP2-dependent recycling of AQP2 is an integral part of the AQP2 shuttle.


Assuntos
Aquaporina 2/metabolismo , Proteínas de Transporte/fisiologia , Rim/metabolismo , Proteínas de Membrana/fisiologia , Cadeias Pesadas de Miosina/fisiologia , Miosina Tipo V/fisiologia , Miosinas/fisiologia , Animais , Aquaporina 2/biossíntese , Proteínas de Transporte/biossíntese , Linhagem Celular , Membrana Celular/metabolismo , Humanos , Imuno-Histoquímica , Imunoprecipitação , Rim/citologia , Proteínas de Membrana/biossíntese , Cadeias Pesadas de Miosina/biossíntese , Miosina Tipo V/biossíntese , Miosinas/biossíntese , Ligação Proteica , Transporte Proteico , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Proteínas rab de Ligação ao GTP
15.
J Am Soc Nephrol ; 18(1): 199-212, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17135396

RESUMO

The cAMP/protein kinase A (PKA)-dependent insertion of water channel aquaporin-2 (AQP2)-bearing vesicles into the plasma membrane in renal collecting duct principal cells (AQP2 shuttle) constitutes the molecular basis of arginine vasopressin (AVP)-regulated water reabsorption. cAMP/PKA signaling systems are compartmentalized by A kinase anchoring proteins (AKAP) that tether PKA to subcellular sites and by phosphodiesterases (PDE) that terminate PKA signaling through hydrolysis of localized cAMP. In primary cultured principal cells, AVP causes focal activation of PKA. PKA and cAMP-specific phosphodiesterase-4D (PDE4D) are located on AQP2-bearing vesicles. The selective PDE4 inhibitor rolipram increases AKAP-tethered PKA activity on AQP2-bearing vesicles and enhances the AQP2 shuttle and thereby the osmotic water permeability. AKAP18delta, which is located on AQP2-bearing vesicles, directly interacts with PDE4D and PKA. In response to AVP, PDE4D and AQP2 translocate to the plasma membrane. Here PDE4D is activated through PKA phosphorylation and reduces the osmotic water permeability. Taken together, a novel, compartmentalized, and physiologically relevant cAMP-dependent signal transduction module on AQP2-bearing vesicles, comprising anchored PDE4D, AKAP18delta, and PKA, has been identified.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Arginina Vasopressina/metabolismo , AMP Cíclico/metabolismo , Túbulos Renais Coletores/metabolismo , 3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , 3',5'-AMP Cíclico Fosfodiesterases/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Aquaporina 2/metabolismo , Arginina Vasopressina/farmacologia , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3 , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Humanos , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/efeitos dos fármacos , Modelos Biológicos , Dados de Sequência Molecular , Inibidores de Fosfodiesterase/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Rolipram/farmacologia , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Água/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 26(6): 1288-96, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16601236

RESUMO

OBJECTIVE: The extracellular N terminus of the endothelin B (ETB) receptor is cleaved by a metalloprotease in an agonist-dependent manner, but the physiological role of this N-terminal proteolysis is not known. In this study, we aimed to determine the functional role of the ETB receptor and of its N-terminal cleavage in vascular smooth muscle cells (VSMCs). METHODS AND RESULTS: VSMCs expressing either the full-length ETB receptor or an N-terminally truncated ETB receptor (corresponding to the N-terminally cleaved receptor) were analyzed for ligand-induced mitogen-activated protein kinase activation and expression of contractile proteins. In VSMCs expressing the full-length ETB receptor, IRL1620 (an ETB-selective agonist) induced a biphasic extracellular signal-regulated kinase 1/2 (ERK1/2) activation and increased expression of contractile proteins (smooth muscle myosin-1 [SM-1]/SM-2, SM22alpha, and alpha-actin). Interestingly, the second phase of ERK1/2 activation required metalloprotease activity, epidermal growth factor (EGF) receptor transactivation, and predominantly activation of Gi proteins. In contrast, in VSMCs expressing N-terminally truncated ETB receptors, IRL1620 did not elicit EGF transactivation and failed to increase contractile protein expression. CONCLUSIONS: This study is the first to show that stimulation of full-length ETB receptors promotes expression of contractile proteins and may thus participate in the differentiation of VSMCs.


Assuntos
Proteínas Contráteis/metabolismo , Receptores ErbB/genética , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Peptídeo Hidrolases/metabolismo , Receptor de Endotelina B/química , Receptor de Endotelina B/metabolismo , Ativação Transcricional , Animais , Células Cultivadas , Endotelinas/farmacologia , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Proteínas de Fluorescência Verde/genética , Humanos , Músculo Liso Vascular/citologia , Mutação , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Ratos , Receptor de Endotelina B/agonistas , Receptor de Endotelina B/genética , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia
17.
Eur J Cell Biol ; 85(7): 673-8, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16500722

RESUMO

A plethora of stimuli including hormones and neurotransmitters mediate a rise of the cellular level of cAMP and thereby activation of protein kinase A (PKA). PKA phosphorylates and thereby modulates the activity of a wide range of cellular targets. It is now appreciated that different stimuli induce the activation of PKA at specific sites where the kinase phosphorylates particular substrates in close proximity. The tethering of PKA to cellular compartments is facilitated by A kinase-anchoring proteins (AKAPs). The incorporation of phosphodiesterases (PDEs) into AKAP-based signalling complexes provides gradients of cAMP that regulate PKA activity locally. An example for a process depending on compartmentalised cAMP/PKA signalling is the arginine-vasopressin (AVP)-mediated water reabsorption in renal collecting duct principal cells. Upon activation through AVP, PKA phosphorylates the water channel aquaporin-2 (AQP-2) located on intracellular vesicles. The phosphorylation triggers the redistribution of AQP2 to the plasma membrane. AKAP-anchored PKA has been shown to be involved in AQP2 shuttling. Here, AKAP18 isoforms and members of the PDE4 family of PDEs are shown to be differentially localised in renal principal cells.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/metabolismo , Distribuição Tecidual , Proteínas de Ancoragem à Quinase A , Animais , Aquaporina 2/metabolismo , Arginina Vasopressina/metabolismo , Compartimento Celular , AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Isoenzimas/metabolismo , Túbulos Renais Coletores/ultraestrutura
18.
Biochem J ; 396(2): 297-306, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16483255

RESUMO

PKA (protein kinase A) is tethered to subcellular compartments by direct interaction of its regulatory subunits (RI or RII) with AKAPs (A kinase-anchoring proteins). AKAPs preferentially bind RII subunits via their RII-binding domains. RII-binding domains form structurally conserved amphipathic helices with unrelated sequences. Their binding affinities for RII subunits differ greatly within the AKAP family. Amongst the AKAPs that bind RIIalpha subunits with high affinity is AKAP7delta [AKAP18delta; K(d) (equilibrium dissociation constant) value of 31 nM]. An N-terminally truncated AKAP7delta mutant binds RIIalpha subunits with higher affinity than the full-length protein presumably due to loss of an inhibitory region [Henn, Edemir, Stefan, Wiesner, Lorenz, Theilig, Schmidtt, Vossebein, Tamma, Beyermann et al. (2004) J. Biol. Chem. 279, 26654-26665]. In the present study, we demonstrate that peptides (25 amino acid residues) derived from the RII-binding domain of AKAP7delta bind RIIalpha subunits with higher affinity (K(d)=0.4+/-0.3 nM) than either full-length or N-terminally truncated AKAP7delta, or peptides derived from other RII binding domains. The AKAP7delta-derived peptides and stearate-coupled membrane-permeable mutants effectively disrupt AKAP-RII subunit interactions in vitro and in cell-based assays. Thus they are valuable novel tools for studying anchored PKA signalling. Molecular modelling indicated that the high affinity binding of the amphipathic helix, which forms the RII-binding domain of AKAP7delta, with RII subunits involves both the hydrophobic and the hydrophilic faces of the helix. Alanine scanning (25 amino acid peptides, SPOT technology, combined with RII overlay assays) of the RII binding domain revealed that hydrophobic amino acid residues form the backbone of the interaction and that hydrogen bond- and salt-bridge-forming amino acid residues increase the affinity of the interaction.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Membrana/metabolismo , Peptídeos/química , Ligação Proteica , Proteínas de Ancoragem à Quinase A , Proteínas Adaptadoras de Transdução de Sinal/química , Sequência de Aminoácidos , Aminoácidos/química , Aminoácidos/metabolismo , Animais , Sítios de Ligação , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/química , Eletrofisiologia , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Proteínas de Membrana/química , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/metabolismo , Peptídeos/farmacologia , Estrutura Terciária de Proteína , Subunidades Proteicas , Ratos , Alinhamento de Sequência , Fatores de Tempo
19.
J Biol Chem ; 279(45): 47254-63, 2004 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-15319430

RESUMO

Some membrane-permeable antagonists restore cell surface expression of misfolded receptors retained in the endoplasmic reticulum (ER) and are therefore termed pharmacochaperones. Whether pharmacochaperones increase protein stability, thereby preventing rapid degradation, or assist folding via direct receptor interactions or interfere with quality control components remains elusive. We now show that the cell surface expression and function (binding of the agonist) of the mainly ER-retained wild-type murine vasopressin V2 receptor GFP fusion protein (mV2R.GFP) is restored by the vasopressin receptor antagonists SR49059 and SR121463B with EC50 values similar to their KD values. This effect was preserved when protein synthesis was abolished. In addition, SR121463B rescued eight mutant human V2Rs (hV2Rs, three are responsible for nephrogenic diabetes insipidus) characterized by amino acid exchanges at the C-terminal end of transmembrane helix TM I and TM VII. In contrast, mutants with amino acid exchanges at the interface of TM II and IV were not rescued by either antagonist. The mechanisms involved in successful rescue of cell surface delivery are explained in a three-dimensional homology model of the antagonist-bound hV2R.


Assuntos
Membrana Celular/metabolismo , Chaperonas Moleculares/química , Processamento de Proteína Pós-Traducional , Receptores de Vasopressinas/química , Sequência de Aminoácidos , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos , Transporte Biológico , Biotinilação , Linhagem Celular , Retículo Endoplasmático/metabolismo , Transferência Ressonante de Energia de Fluorescência , Proteínas de Fluorescência Verde/metabolismo , Humanos , Immunoblotting , Imuno-Histoquímica , Cinética , Lasers , Camundongos , Microscopia de Fluorescência , Modelos Moleculares , Dados de Sequência Molecular , Morfolinas/farmacologia , Mutação , Peptídeos/química , Plasmídeos/metabolismo , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Receptores de Vasopressinas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Compostos de Espiro/farmacologia , Fatores de Tempo , Transfecção
20.
J Biol Chem ; 279(25): 26654-65, 2004 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-15037626

RESUMO

Arginine vasopressin (AVP) increases the water permeability of renal collecting duct principal cells by inducing the fusion of vesicles containing the water channel aquaporin-2 (AQP2) with the plasma membrane (AQP2 shuttle). This event is initiated by activation of vasopressin V2 receptors, followed by an elevation of cAMP and the activation of protein kinase A (PKA). The tethering of PKA to subcellular compartments by protein kinase A anchoring proteins (AKAPs) is a prerequisite for the AQP2 shuttle. During the search for AKAP(s) involved in the shuttle, a new splice variant of AKAP18, AKAP18delta, was identified. AKAP18delta functions as an AKAP in vitro and in vivo. In the kidney, it is mainly expressed in principal cells of the inner medullary collecting duct, closely resembling the distribution of AQP2. It is present in both the soluble and particulate fractions derived from renal inner medullary tissue. Within the particulate fraction, AKAP18delta was identified on the same intracellular vesicles as AQP2 and PKA. AVP not only recruited AQP2, but also AKAP18delta to the plasma membrane. The elevation of cAMP caused the dissociation of AKAP18delta and PKA. The data suggest that AKAP18delta is involved in the AQP2 shuttle.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Aquaporinas/química , Proteínas de Transporte/química , Proteínas de Transporte/fisiologia , Rim/citologia , Proteínas de Membrana/química , Proteínas de Membrana/fisiologia , Vasopressinas/química , Proteínas de Ancoragem à Quinase A , Animais , Aquaporina 2 , Arginina Vasopressina/química , Northern Blotting , Western Blotting , Linhagem Celular , Membrana Celular/metabolismo , Células Cultivadas , Clonagem Molecular , AMP Cíclico/metabolismo , DNA Complementar/metabolismo , Ativação Enzimática , Transferência Ressonante de Energia de Fluorescência , Biblioteca Gênica , Glutationa Transferase/metabolismo , Humanos , Imuno-Histoquímica , Rim/metabolismo , Medula Renal/metabolismo , Cinética , Testes de Precipitina , Ligação Proteica , Conformação Proteica , Isoformas de Proteínas , Transporte Proteico , RNA/química , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Frações Subcelulares , Ressonância de Plasmônio de Superfície , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA