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










Base de datos
Intervalo de año de publicación
1.
FEBS J ; 289(17): 5322-5340, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35285161

RESUMEN

Macroautophagy (hereafter autophagy) is a process that degrades cellular components to maintain homeostasis. The Ca2+ sensor calmodulin (CaM) regulates numerous cell functions but is a limiting factor due to its insufficient availability for all target proteins. However, evidence that CaM availability regulates basal autophagy is lacking. Here, we have tested this hypothesis. CaM antagonists W-7, trifluoperazine and CGS9343b cause autophagosome accumulation and inhibit basal autophagic flux in the same manner as does chloroquine. These reagents promote the activity of AMP-activated protein kinase (AMPK) but not that of the mechanistic target of rapamycin (mTOR). Competitive binding assays using CaM sensors with different Ca2+ dependencies showed that chloroquine directly binds CaM in a Ca2+ -dependent fashion. The CaM antagonists have disparate effects on cytoplasmic Ca2+ , triggering from none to robust signals, indicating that their consistent inhibition of autophagy is due to inhibition of CaM and not Ca2+ . Chelating intracellular Ca2+ reduces the effect of the CaM antagonists to accumulate LC3-II, indicating that they do so by inhibiting CaM-dependent activities at basal Ca2+ level. The CaM antagonists cause lysosomal alkalinisation. Consistently, buffering CaM with a high-affinity CaM-binding protein that binds CaM at resting Ca2+ level increases lysosomal pH. Enhanced CaM buffering using a chimeric protein that contains two high-affinity CaM-binding sites that can collectively bind CaM at a large range of Ca2+ further increases lysosomal pH and increases LC3-II accumulation and AMPK activity, but not that of mTOR. These data demonstrate that CaM availability is required for basal autophagy.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Calmodulina , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia/fisiología , Calcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Cloroquina/farmacología , Serina-Treonina Quinasas TOR/metabolismo
2.
Mol Cell Biochem ; 477(5): 1489-1498, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35171400

RESUMEN

Calmodulin (CaM) is a Ca2+ sensor protein that is required for numerous vascular smooth muscle cell (VSMC) functions. Since CaM is not expressed enough for its many target proteins, factors that modulate its expression and interactions with targets in VSMCs can have extensive effects on vascular functions. VSMCs receive many regulatory inputs from endothelial cells (ECs). However, it is unknown if ECs regulate vascular functions via controlling expression of CaM and its interactions in VSMCs. In this work, we tested the hypothesis that ECs also affect VSMC signaling via regulation of CaM expression and interactions with its target proteins in VSMCs. Using ECs and VSMCs isolated from the same vessels and grown in a co-culture system, we observed that the presence of proliferating ECs significantly upregulates total CaM expression in VSMCs. An imaging module was devised to concurrently measure free Ca2+ and CaM levels in VSMCs in co-culture with ECs. Using indo-1/AM and a CaM biosensor built from a modified CaM-binding sequence of endothelial nitric oxide synthase (eNOS), this system revealed that in response to a generic Ca2+ signal, free Ca2+-bound CaM level is enhanced ~ threefold in VSMCs in co-culture with proliferating ECs. Interestingly, VSMCs express eNOS and eNOS-CaM association in response to the same Ca2+ stimulus is also enhanced ~ threefold in VSMCs co-cultured with ECs. Mechanistically, the endothelium-dependent upregulation of CaM in VSMCs is not affected by inhibition of NO production or endothelin receptors but is prevented by inhibition of vascular endothelial growth factor receptors. Consistently, VEGF-A level is upregulated in VSMCs co-cultured with proliferating ECs. These data indicate a new role of the endothelium in regulating vascular functions via upregulating CaM and its interactions in VSMCs.


Asunto(s)
Músculo Liso Vascular , Óxido Nítrico Sintasa de Tipo III , Señalización del Calcio , Calmodulina/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Life Sci ; 290: 120247, 2022 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-34954214

RESUMEN

AIMS: The Ca2+-binding protein calmodulin (CaM) modulates numerous target proteins but is produced insufficiently to bind all of them, generating a limiting CaM equilibrium. Menopause increases cardiac morbidity; however, it is unknown if the cardiac CaM equilibrium is affected by estrogen. We devised an assay to assess the effects of ovariectomy and estrogen treatment on the cardiac CaM equilibrium. MATERIALS AND METHODS: Sprague-Dawley rats received sham surgery or ovariectomy, followed by 2-week treatment with vehicle or 17ß-estradiol. Ca2+-saturated left ventricular (LV) lysates were processed through CaM sepharose columns, which retained CaM-binding proteins unoccupied by endogenous CaM. Eluants therefrom were subjected to a competitive binding assay against purified CaM and a CaM biosensor to assess the amounts of unoccupied CaM-binding sites. LV cellular composition was assessed by immunohistochemistry. KEY FINDINGS: LV eluants processed from sham animals reduce biosensor response by ~32%, indicating baseline presence of unoccupied CaM-binding sites and a limiting CaM equilibrium. Ovariectomy exacerbates the limiting CaM equilibrium, reducing biosensor response by ~65%. 17ß-estradiol treatment equalizes the difference between sham and ovariectomized animals. These changes reflect whole tissue responses and are not mirrored by changes in total surface areas of cardiomyocytes and fibroblasts. Consistently, Ca2+-dependent, but not Ca2+-independent, interaction between CaM and the cardiac inositol trisphosphate receptor (IP3R) is reduced following ovariectomy and is restored by subsequent 17ß-estradiol treatment. SIGNIFICANCE: Our assay provides a new parameter to assess tissue CaM equilibrium. The exacerbated limiting CaM equilibrium following estrogen loss may contribute to cardiac morbidity and is prevented by estrogen treatment.


Asunto(s)
Calmodulina/metabolismo , Estradiol/farmacología , Miocitos Cardíacos/metabolismo , Animales , Sitios de Unión , Señalización del Calcio/efectos de los fármacos , Calmodulina/efectos de los fármacos , Estradiol/metabolismo , Estrógenos/metabolismo , Estrógenos/farmacología , Femenino , Ventrículos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Ovariectomía , Posmenopausia/fisiología , Unión Proteica , Ratas , Ratas Sprague-Dawley
4.
Front Endocrinol (Lausanne) ; 11: 568203, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33133016

RESUMEN

17ß-Estradiol (E2) is the main estrogenic hormone in the body and exerts many cardiovascular protective effects. Via three receptors known to date, including estrogen receptors α (ERα) and ß (ERß) and the G protein-coupled estrogen receptor 1 (GPER, aka GPR30), E2 regulates numerous calcium-dependent activities in cardiovascular tissues. Nevertheless, effects of E2 and its receptors on components of the calcium signaling machinery (CSM), the underlying mechanisms, and the linked functional impact are only beginning to be elucidated. A picture is emerging of the reciprocality between estrogen biology and Ca2+ signaling. Therein, E2 and GPER, via both E2-dependent and E2-independent actions, moderate Ca2+-dependent activities; in turn, ERα and GPER are regulated by Ca2+ at the receptor level and downstream signaling via a feedforward loop. This article reviews current understanding of the effects of E2 and its receptors on the cardiovascular CSM and vice versa with a focus on mechanisms and combined functional impact. An overview of the main CSM components in cardiovascular tissues will be first provided, followed by a brief review of estrogen receptors and their Ca2+-dependent regulation. The effects of estrogenic agonists to stimulate acute Ca2+ signals will then be reviewed. Subsequently, E2-dependent and E2-independent effects of GPER on components of the Ca2+ signals triggered by other stimuli will be discussed. Finally, a case study will illustrate how the many mechanisms are coordinated to moderate Ca2+-dependent activities in the cardiovascular system.


Asunto(s)
Señalización del Calcio/fisiología , Sistema Cardiovascular/metabolismo , Estrógenos/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Fenómenos Fisiológicos Cardiovasculares , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Vasodilatación/fisiología
5.
Biochem Pharmacol ; 171: 113727, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31759979

RESUMEN

The G protein-coupled estrogen receptor 1 (GPER) produces cardioprotective effects. However, the underlying mechanisms are not well understood. We aimed to investigate the role of GPER in ß adrenoceptor-mediated cardiac contraction and myocardial signaling. In anesthetized animals, intrajugular administration of isoproterenol produces a rapid and sustained rise in left ventricular pressure (LVP) and increases ectopic contractions. Administration of the GPER agonist G-1 during the plateau phase of isoproterenol-induced LVP increase rapidly restores LVP to baseline levels and reduces the frequency of ectopic contractions. In freshly isolated cardiomyocytes, isoproterenol potentiates electrically induced peak currents of L-type Ca2+ channels (LTCC) and increases the potential sensitivity of their inactivation. Coadministration of G-1 prevents isoproterenol-induced potentiation of peak LTCC currents and makes channels more sensitive to being inactivated compared to isoproterenol alone. Isoproterenol treatment of cardiomyocytes without electrical stimulation triggers slow-rising Ca2+ signals that are inhibited by the ß1AR antagonist metoprolol but not by ß2AR antagonist ICI-118551. G-1 pretreatment dose-dependently suppresses isoproterenol-induced total Ca2+ signals and the amplitude and frequency of the intrinsic Ca2+ oscillatory deflections. Pretreatment with the GPER antagonist G-36 produces opposite effects, dose-dependently increasing these signals. ISO promotes robust phosphorylation of Cav1.2 channels at Ser1928. G-1 pretreatment inhibits isoproterenol-stimulated phosphorylation of Cav1.2 at Ser1928, while G-36 pretreatment enhances this signal. Our data indicate that GPER functions as an intrinsic component of ß1AR signaling to moderate myocardial Ca2+ dynamics and contraction.


Asunto(s)
Calcio/metabolismo , Receptor alfa de Estrógeno/metabolismo , Contracción Miocárdica/fisiología , Miocitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Antagonistas Adrenérgicos beta/farmacología , Animales , Benzodioxoles/farmacología , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Ciclopentanos/farmacología , Isoproterenol/farmacología , Cinética , Masculino , Ratones , Contracción Miocárdica/efectos de los fármacos , Miocardio/citología , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Quinolinas/farmacología
6.
J Proteome Res ; 18(10): 3780-3791, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31483676

RESUMEN

This work addresses the question of how the Ca2+ sensor protein calmodulin shapes cellular responses to Ca2+ signals. Proteins interacting with affinity tagged calmodulin were captured by rapid (t1/2 ≈ 7 s) photoactivated cross-linking under basal conditions, after brief removal of extracellular Ca2+ and during a cytosolic [Ca2+] transient in cells metabolically labeled with a photoreactive methionine analog. Tagged adducts were stringently enriched, and captured proteins were identified and quantified by LC-MS/MS. A set of 489 proteins including 27 known calmodulin interactors was derived. A threshold for fractional capture was applied to define a high specificity group of 170 proteins, including 22 known interactors, and a low specificity group of 319 proteins. Capture of ∼60% of the high specificity group was affected by manipulations of Ca2+, compared with ∼20% of the low specificity group. This suggests that the former is likely to contain novel interactors of physiological significance. The capture of 29 proteins, nearly all high specificity, was decreased by the removal of extracellular Ca2+, although this does not affect cytosolic [Ca2+]. Capture of half of these was unaffected by the cytosolic [Ca2+] transient, consistent with high local [Ca2+]. These proteins are hypothesized to reside in or near microdomains of high [Ca2+] supported by the Ca2+ influx.


Asunto(s)
Calmodulina/metabolismo , Células/metabolismo , Reactivos de Enlaces Cruzados/efectos de la radiación , Metionina/metabolismo , Proteínas/metabolismo , Calcio/metabolismo , Señalización del Calcio , Células/química , Células Cultivadas , Cromatografía Liquida , Humanos , Unión Proteica , Espectrometría de Masas en Tándem
7.
Eur J Pharmacol ; 848: 70-79, 2019 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-30690001

RESUMEN

Cardiovascular functions are mediated by multiple 7-pass transmembrane receptors whose activation promotes contraction or relaxation of the tissues. The α1 adrenoceptor type 1A plays important roles in the control of vascular tone and myocardial contractility via Ca2+-dependent actions. Here, using novel FRET-based biosensors, we identified a novel Ca2+-dependent interaction between calmodulin (CaM) and the human α1A adrenoceptor at the juxtamembranous region of its 4th submembrane domain (SMD4JM, a.a. 333-361). SMD4JM houses the known nuclear localization signal of α1A adrenoceptor (NLS, a.a. 334-349). We found that NLS itself also interacts with CaM, but with lower affinity and Ca2+ sensitivity, indicating that full interaction between CaM and α1A receptor in this region requires segment a.a. 333-361. Combined K353Q/L356A substitutions in the non-NLS segment of SMD4JM cause a 3.5-fold reduction in the affinity of CaM-SMD4JM interaction. Overexpression of wild-type α1A adrenoceptor in cells enhances phosphorylation of the extracellular signal-regulated kinases 1/2 (ERK1/2) stimulated by A61603, while overexpression of the K353Q/L356A α1A receptor mutant significantly reduces this signal. Norepinephrine stimulates intracellular Ca2+ signals that are higher in cells overexpressing wild-type receptor but lower in cells overexpressing the K353Q/L356A receptor compared to non-transfected cells in the same microscopic environments. These data support a novel and important role for Ca2+-dependent CaM interaction at SMD4JM in α1A adrenoceptor-mediated signaling.


Asunto(s)
Calcio/metabolismo , Calmodulina/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Calcio/farmacología , Calmodulina/farmacología , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Unión Proteica/fisiología , Receptores Adrenérgicos alfa 1/genética , Transducción de Señal/efectos de los fármacos
8.
Artículo en Inglés | MEDLINE | ID: mdl-31993020

RESUMEN

The G Protein-Coupled Estrogen Receptor (GPER) is a novel membrane-bound receptor that mediates non-genomic actions of the primary female sex hormone 17ß-estradiol. Studies over the past two decades have elucidated the beneficial actions of this receptor in a number of cardiometabolic diseases. This review will focus specifically on the cardiac actions of GPER, since this receptor is expressed in cardiomyocytes as well as other cells within the heart and most likely contributes to estrogen-induced cardioprotection. Studies outlining the impact of GPER on diastolic function, mitochondrial function, left ventricular stiffness, calcium dynamics, cardiac inflammation, and aortic distensibility are discussed. In addition, recent data using genetic mouse models with global or cardiomyocyte-specific GPER gene deletion are highlighted. Since estrogen loss due to menopause in combination with chronological aging contributes to unique aspects of cardiac dysfunction in women, this receptor may provide novel therapeutic effects. While clinical studies are still required to fully understand the potential for pharmacological targeting of this receptor in postmenopausal women, this review will summarize the evidence gathered thus far on its likely beneficial effects.

9.
Biochem Pharmacol ; 152: 187-200, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29605626

RESUMEN

The angiotensin II receptor type 1 (AT1R) mediates many Ca2+-dependent actions of angiotensin II (AngII). Calmodulin (CaM) is a key transducer of Ca2+ signals in cells. Two locations on the receptor's submembrane domains (SMD) 3 and 4 are known to interact with CaM. However, the binding sites for CaM, biochemical properties of the interactions, and their functional impact are not fully understood. Using a FRET-based screening method, we identified a new binding site for CaM on SMD2 (a.a. 125-141), in addition to SMD3 and the juxtamembranous region of SMD4 (SMD4JM, a.a., 309-327). Simultaneous measurements of CaM binding and free Ca2+ show that the interactions are Ca2+-dependent, with disparate Kd and EC50(Ca2+) values within the physiological range of cytoplasmic Ca2+. Full interaction between CaM and SMD3 requires the entire domain (a.a. 215-242) and has an EC50(Ca2+) value in the range of resting cytoplasmic Ca2+, suggesting AT1R-CaM interaction can occur in resting conditions in cells. AngII induces robust ERK1/2 phosphorylation in primary vascular smooth muscle cells. This effect is suppressed by AT1R inhibitor losartan and virtually abolished by CaM antagonist W-7. AngII-induced ERK1/2 phosphorylation is suppressed in cells expressing mutant AT1R with reduced CaM binding at each identified binding domain. AngII triggers transient Ca2+ signals in cells expressing wild-type AT1R. These signals are reduced in cells expressing mutant AT1R with reduced CaM binding at SMD3 or SMD4JM, but are very slow-rising, low amplitude signal in cells expressing AT1R with reduced CaM binding at SMD2. The data indicate that CaM interactions with AT1R can occur at various domains, with different affinities, at different physiological Ca2+ levels, and are important for AT1R-mediated signaling.


Asunto(s)
Calmodulina/farmacología , Receptor de Angiotensina Tipo 1/metabolismo , Angiotensina II/metabolismo , Animales , Señalización del Calcio , Quinasas MAP Reguladas por Señal Extracelular , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Fosforilación , Unión Proteica , Dominios Proteicos , ARN Mensajero/genética , ARN Mensajero/metabolismo
10.
Biochem J ; 474(21): 3627-3642, 2017 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-28935720

RESUMEN

The G protein-coupled estrogen receptor 1 (GPER, formerly also known as GPR30) modulates many Ca2+-dependent activities in endothelial cells. However, the underlying mechanisms are poorly understood. We recently reported that GPER acts to prolong cytoplasmic Ca2+ signals by interacting with and promoting inhibitory phosphorylation of the plasma membrane Ca2+-ATPase. In the present study, we examined the role of GPER activation in modulating store-operated Ca2+ entry (SOCE) via effects on the stromal interaction molecule 1 (STIM1). GPER activation by agonist G-1 reduces the peak but prolongs the plateau of bradykinin-induced Ca2+ signals in primary endothelial cells. G-1 dose-dependently inhibits thapsigargin-induced SOCE measured by the Mn2+ quenching method. GPER heterologous expression reduces SOCE, which is further pronounced by G-1 treatment. Consistently, GPER gene silencing in endothelial cells is associated with an increase in SOCE. Treatment with G-1 reduces puncta formation by STIM1 triggered by the activation of SOCE. The effect of GPER activation to inhibit SOCE is not affected by combined nonphosphorylatable substitutions at serines 486 and 668 on STIM1, but is substantially reduced by similar substitutions at serines 575, 608 and 621. Taken together with our recently reported inhibitory actions of GPER on Ca2+ efflux, the current data contribute to a model in which GPER acts to clamp agonist-induced cytoplasmic Ca2+ signals. Kinetic modeling based on current and reported data is used to estimate the overall effect of GPER activation on point activity of endothelial nitric oxide synthase during the time course of agonist-induced total Ca2+ signals.


Asunto(s)
Bradiquinina/farmacología , Ciclopentanos/farmacología , Células Endoteliales/metabolismo , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Quinolinas/farmacología , Molécula de Interacción Estromal 1/metabolismo , Sustitución de Aminoácidos , Animales , Señalización del Calcio , Células Endoteliales/citología , Células HEK293 , Humanos , Mutación Missense , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Molécula de Interacción Estromal 1/antagonistas & inhibidores , Molécula de Interacción Estromal 1/genética , Porcinos
11.
J Biol Chem ; 291(20): 10805-23, 2016 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-26987903

RESUMEN

Estrogen exerts many effects on the vascular endothelium. Calmodulin (CaM) is the transducer of Ca(2+) signals and is a limiting factor in cardiovascular tissues. It is unknown whether and how estrogen modifies endothelial functions via the network of CaM-dependent proteins. Here we show that 17ß-estradiol (E2) up-regulates total CaM level in endothelial cells. Concurrent measurement of Ca(2+) and Ca(2+)-CaM indicated that E2 also increases free Ca(2+)-CaM. Pharmacological studies, gene silencing, and receptor expression-specific cell studies indicated that the G protein-coupled estrogen receptor 1 (GPER/GPR30) mediates these effects via transactivation of EGFR and subsequent MAPK activation. The outcomes were then examined on four distinct members of the intracellular CaM target network, including GPER/GPR30 itself and estrogen receptor α, the plasma membrane Ca(2+)-ATPase (PMCA), and endothelial nitric-oxide synthase (eNOS). E2 substantially increases CaM binding to estrogen receptor α and GPER/GPR30. Mutations that reduced CaM binding to GPER/GPR30 in separate binding domains do not affect GPER/GPR30-Gßγ preassociation but decrease GPER/GPR30-mediated ERK1/2 phosphorylation. E2 increases CaM-PMCA association, but the expected stimulation of Ca(2+) efflux is reversed by E2-stimulated tyrosine phosphorylation of PMCA. These effects sustain Ca(2+) signals and promote Ca(2+)-dependent CaM interactions with other CaM targets. Consequently, E2 doubles CaM-eNOS interaction and also promotes dual phosphorylation of eNOS at Ser-617 and Ser-1179. Calculations using in-cell and in vitro data revealed substantial individual and combined contribution of these effects to total eNOS activity. Taken together, E2 generates a feedforward loop via GPER/GPR30, which enhances Ca(2+)/CaM signals and functional linkage in the endothelial CaM target network.


Asunto(s)
Señalización del Calcio/fisiología , Calmodulina/metabolismo , Endotelio Vascular/metabolismo , Estradiol/metabolismo , Estrógenos/farmacología , Animales , Células Cultivadas , Endotelio Vascular/citología , Receptor alfa de Estrógeno , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación/fisiología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Porcinos
12.
FEBS Lett ; 589(11): 1173-8, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25871521

RESUMEN

Activation of endothelial nitric oxide synthase (eNOS) by calmodulin (CaM) facilitates formation of a sequence of conformational states that is not well understood. Fluorescence decays of fluorescently labeled CaM bound to eNOS reveal four distinct conformational states and single-molecule fluorescence trajectories show multiple fluorescence states with transitions between states occurring on time scales of milliseconds to seconds. A model is proposed relating fluorescence quenching states to enzyme conformations. Specifically, we propose that the most highly quenched state corresponds to CaM docked to an oxygenase domain of the enzyme. In single-molecule trajectories, this state occurs with time lags consistent with the oxygenase activity of the enzyme.


Asunto(s)
Calmodulina/química , Modelos Moleculares , Complejos Multiproteicos/química , Óxido Nítrico Sintasa de Tipo III/química , Animales , Bovinos , Recuperación de Fluorescencia tras Fotoblanqueo , Fluorometría , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína
13.
J Biol Chem ; 290(21): 13293-307, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25847233

RESUMEN

The new G protein-coupled estrogen receptor 1 (GPER/GPR30) plays important roles in many organ systems. The plasma membrane Ca(2+)-ATPase (PMCA) is essential for removal of cytoplasmic Ca(2+) and for shaping the time courses of Ca(2+)-dependent activities. Here, we show that PMCA and GPER/GPR30 physically interact and functionally influence each other. In primary endothelial cells, GPER/GPR30 agonist G-1 decreases PMCA-mediated Ca(2+) extrusion by promoting PMCA tyrosine phosphorylation. GPER/GPR30 overexpression decreases PMCA activity, and G-1 further potentiates this effect. GPER/GPR30 knockdown increases PMCA activity, whereas PMCA knockdown substantially reduces GPER/GPR30-mediated phosphorylation of the extracellular signal-related kinase (ERK1/2). GPER/GPR30 co-immunoprecipitates with PMCA with or without treatment with 17ß-estradiol, thapsigargin, or G-1. Heterologously expressed GPER/GPR30 in HEK 293 cells co-localizes with PMCA4b, the main endothelial PMCA isoform. Endothelial cells robustly express the PDZ post-synaptic density protein (PSD)-95, whose knockdown reduces the association between GPER/GPR30 and PMCA. Additionally, the association between PMCA4b and GPER/GPR30 is substantially reduced by truncation of either or both of their C-terminal PDZ-binding motifs. Functionally, inhibition of PMCA activity is significantly reduced by truncation of GPER/GPR30's C-terminal PDZ-binding motif. These data strongly indicate that GPER/GPR30 and PMCA4b form a hetero-oligomeric complex in part via the anchoring action of PSD-95, in which they constitutively affect each other's function. Activation of GPER/GPR30 further inhibits PMCA activity through tyrosine phosphorylation of the pump. These interactions represent cross-talk between Ca(2+) signaling and GPER/GPR30-mediated activities.


Asunto(s)
Aorta/metabolismo , Calcio/metabolismo , Membrana Celular/metabolismo , Endotelio Vascular/metabolismo , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Aorta/citología , Western Blotting , Células Cultivadas , Endotelio Vascular/citología , Células HEK293 , Humanos , Inmunoprecipitación , Fosforilación , ATPasas Transportadoras de Calcio de la Membrana Plasmática/antagonistas & inhibidores , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , Unión Proteica , ARN Interferente Pequeño/genética , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Porcinos
15.
PLoS One ; 9(2): e89669, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586950

RESUMEN

The G protein-coupled estrogen receptor 1 (GPER) has been demonstrated to participate in many cellular functions, but its regulatory inputs are not clearly understood. Here we describe a new approach that identifies GPER as a calmodulin-binding protein, locates interaction sites, and characterizes their binding properties. GPER coimmunoprecipitates with calmodulin in primary vascular smooth muscle cells under resting conditions, which is enhanced upon acute treatment with either specific ligands or a Ca(2+)-elevating agent. To confirm direct interaction and locate the calmodulin-binding domain(s), we designed a series of FRET biosensors that consist of enhanced cyan and yellow fluorescent proteins flanking each of GPER's submembrane domains (SMDs). Responses of these biosensors showed that all four submembrane domains directly bind calmodulin. Modifications of biosensor linker identified domains that display the strongest calmodulin-binding affinities and largest biosensor dynamics, including a.a. 83-93, 150-175, 242-259, 330-351, corresponding respectively to SMDs 1, 2, 3, and the juxta-membranous section of SMD4. These biosensors bind calmodulin in a strictly Ca(2+)-dependent fashion and with disparate affinities in the order SMD2>SMD4>SMD3>SMD1, apparent K d values being 0.44 ± 0.03, 1.40 ± 0.16, 8.01 ± 0.29, and 136.62 ± 6.56 µM, respectively. Interestingly, simultaneous determinations of biosensor responses and suitable Ca(2+) indicators identified separate Ca(2+) sensitivities for their interactions with calmodulin. SMD1-CaM complexes display a biphasic Ca(2+) response, representing two distinct species (SMD1 sp1 and SMD1 sp2) with drastically different Ca(2+) sensitivities. The Ca(2+) sensitivities of CaM-SMDs interactions follow the order SMD1sp1>SMD4>SMD2>SMD1sp2>SMD3, EC50(Ca(2+)) values being 0.13 ± 0.02, 0.75 ± 0.05, 2.38 ± 0.13, 3.71 ± 0.13, and 5.15 ± 0.25 µM, respectively. These data indicate that calmodulin may regulate GPER-dependent signaling at the receptor level through multiple interaction sites. FRET biosensors represent a simple method to identify unknown calmodulin-binding domains in G protein-coupled receptors and to quantitatively assess binding properties.


Asunto(s)
Receptores de Estrógenos/química , Receptores Acoplados a Proteínas G/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Técnicas Biosensibles , Calcio/química , Calmodulina/química , Proteínas de Unión a Calmodulina/química , Proteínas de Unión a Calmodulina/metabolismo , Células Cultivadas , Estradiol/fisiología , Transferencia Resonante de Energía de Fluorescencia , Humanos , Datos de Secuencia Molecular , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sus scrofa
17.
FEBS Lett ; 587(3): 297-301, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23266515

RESUMEN

We have derived structures of intact calmodulin (CaM)-free and CaM-bound endothelial nitric oxide synthase (eNOS) by reconstruction from cryo-electron micrographs. The CaM-free reconstruction is well fitted by the oxygenase domain dimer, but the reductase domains are not visible, suggesting they are mobile and thus delocalized. Additional protein is visible in the CaM-bound reconstruction, concentrated in volumes near two basic patches on each oxygenase domain. One of these corresponds with a presumptive docking site for the reductase domain FMN-binding module. The other is proposed to correspond with a docking site for CaM. A model is suggested in which CaM binding and docking position the reductase domains near the oxygenase domains and promote docking of the FMN-binding modules required for electron transfer.


Asunto(s)
Calmodulina/metabolismo , Calmodulina/farmacología , Óxido Nítrico Sintasa de Tipo III/química , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Calmodulina/química , Bovinos , Modelos Moleculares , Oxigenasas/química , Oxigenasas/metabolismo , Conformación Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Ratas
18.
J Biol Chem ; 284(18): 11892-9, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19251696

RESUMEN

We have investigated the possible biochemical basis for enhancements in NO production in endothelial cells that have been correlated with agonist- or shear stress-evoked phosphorylation at Ser-1179. We have found that a phosphomimetic substitution at Ser-1179 doubles maximal synthase activity, partially disinhibits cytochrome c reductase activity, and lowers the EC(50)(Ca(2+)) values for calmodulin binding and enzyme activation from the control values of 182 +/- 2 and 422 +/- 22 nm to 116 +/- 2 and 300 +/- 10 nm. These are similar to the effects of a phosphomimetic substitution at Ser-617 (Tran, Q. K., Leonard, J., Black, D. J., and Persechini, A. (2008) Biochemistry 47, 7557-7566). Although combining substitutions at Ser-617 and Ser-1179 has no additional effect on maximal synthase activity, cooperativity between the two substitutions completely disinhibits reductase activity and further reduces the EC(50)(Ca(2+)) values for calmodulin binding and enzyme activation to 77 +/- 2 and 130 +/- 5 nm. We have confirmed that specific Akt-catalyzed phosphorylation of Ser-617 and Ser-1179 and phosphomimetic substitutions at these positions have similar functional effects. Changes in the biochemical properties of eNOS produced by combined phosphorylation at Ser-617 and Ser-1179 are predicted to substantially increase synthase activity in cells at a typical basal free Ca(2+) concentration of 50-100 nm.


Asunto(s)
Calcio/química , Calmodulina/química , Óxido Nítrico Sintasa de Tipo III/química , Proteínas Proto-Oncogénicas c-akt/química , Sustitución de Aminoácidos , Animales , Calcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Bovinos , Reductasas del Citocromo/química , Reductasas del Citocromo/genética , Reductasas del Citocromo/metabolismo , Humanos , Mutación Missense , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación/fisiología , Unión Proteica/fisiología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estrés Fisiológico/fisiología
19.
Biochemistry ; 47(28): 7557-66, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18558722

RESUMEN

We have investigated the effects of phosphorylation at Ser-617 and Ser-635 within an autoinhibitory domain (residues 595-639) in bovine endothelial nitric oxide synthase on enzyme activity and the Ca (2+) dependencies for calmodulin binding and enzyme activation. A phosphomimetic S617D substitution doubles the maximum calmodulin-dependent enzyme activity and decreases the EC 50(Ca (2+)) values for calmodulin binding and enzyme activation from the wild-type values of 180 +/- 2 and 397 +/- 23 nM to values of 109 +/- 2 and 258 +/- 11 nM, respectively. Deletion of the autoinhibitory domain also doubles the maximum calmodulin-dependent enzyme activity and decreases the EC 50(Ca (2+)) values for calmodulin binding and calmodulin-dependent enzyme activation to 65 +/- 4 and 118 +/- 4 nM, respectively. An S635D substitution has little or no effect on enzyme activity or EC 50(Ca (2+)) values, either alone or when combined with the S617D substitution. These results suggest that phosphorylation at Ser-617 partially reverses suppression by the autoinhibitory domain. Associated effects on the EC 50(Ca (2+)) values and maximum calmodulin-dependent enzyme activity are predicted to contribute equally to phosphorylation-dependent enhancement of NO production during a typical agonist-evoked Ca (2+) transient, while the reduction in EC 50(Ca (2+)) values is predicted to be the major contributor to enhancement at resting free Ca (2+) concentrations.


Asunto(s)
Calcio/metabolismo , Calmodulina/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Sitios de Unión , Bovinos , Clonación Molecular , Cartilla de ADN , Cinética , Mutagénesis , Óxido Nítrico Sintasa de Tipo III/genética , Fosfoproteínas/metabolismo , Fosforilación , Unión Proteica , Proteínas Recombinantes/metabolismo
20.
Cell Calcium ; 37(6): 541-53, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15862345

RESUMEN

In endothelial cells nitric oxide synthase is a dominant affector in the calmodulin network by virtue of its ability to bind a significant fraction of limiting intracellular calmodulin. We have investigated how this affector function influences the kinetics of calmodulin-dependent signaling in cells co-expressing the synthase and a fluorescent calmodulin target analog similar in its interactions with calmodulin to myosin light chain kinase. The synthase binds (Ca(2+))(4)-calmodulin with a K(d) value of approximately 0.2 nM and an association rate constant of approximately 1.5 x 10(5) M(-1) s(-1). These values are, respectively, 10- and 100-fold smaller than the corresponding values for the analog. Thus, when Ca(2+) is added to a mixture of calmodulin, target analog and synthase in vitro a large fluorescence transient with a relaxation time of approximately 600 s is observed as (Ca(2+))(4)-calmodulin is rapidly bound to the analog and then slowly captured by the higher affinity synthase. A rapid increase in the free Ca(2+) concentration elicits similar transient analog responses in cells expressing the cytoplasmic target analog and either a wild-type membrane bound or mutant cytoplasmic synthase. Transient responses are not observed in cells co-expressing the fluorescent analog and a mutant T497D synthase unable to bind calmodulin. These results demonstrate that dominant affectors in the calmodulin network shape both the magnitudes and time courses of target responses in the cell.


Asunto(s)
Calmodulina/metabolismo , Óxido Nítrico Sintasa/metabolismo , Transducción de Señal , Animales , Bovinos , Línea Celular , Colorantes Fluorescentes , Cinética , Quinasa de Cadena Ligera de Miosina/metabolismo , Óxido Nítrico Sintasa de Tipo III , Unión Proteica , Proteínas Recombinantes de Fusión/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...