Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
Curr Opin Physiol ; 372024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38094036

RESUMEN

The lethality of heart failure (HF), particularly in the context of post-acute sequelae SARS-CoV-2 infection (PASC)-related myocarditis, necessitates the discovery of the cellular pathways implicated in cardiovascular disease (CVD). We summarize the signaling mechanisms of the catecholamine-binding ß-adrenergic receptors (ß-ARs), with an emphasis on the role of ß-arrestins. ß-ARs, a subset of G protein-coupled receptors (GPCRs), canonically propagate signals through heterotrimeric G proteins. However, since their discovery in the late 1980s, ß-arrestins have been shown to, both (i) quench G protein signaling and (ii) initiate their own independent signaling cascades, which is influenced by post-translational modifications. ß-arrestin-biased agonism by the beta-blocker carvedilol and its allosteric modulation can serve a cardioprotective role. The increasingly labyrinthine nature of GPCR signaling suggests that ligand-dependent ß-AR signaling, either stimulated by an agonist or blocked by an antagonist, is selectively enhanced or suppressed by allosteric modulations, which are orchestrated by novel drugs or endogenous post-translational modifications.

4.
J Biol Chem ; 299(8): 105045, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37451484

RESUMEN

Glucagon signaling is essential for maintaining normoglycemia in mammals. The arrestin fold superfamily of proteins controls the trafficking, turnover, and signaling of transmembrane receptors as well as other intracellular signaling functions. Further investigation is needed to understand the in vivo functions of the arrestin domain-containing 4 (ARRDC4) protein family member and whether it is involved in mammalian glucose metabolism. Here, we show that mice with a global deletion of the ARRDC4 protein have impaired glucagon responses and gluconeogenesis at a systemic and molecular level. Mice lacking ARRDC4 exhibited lower glucose levels after fasting and could not suppress gluconeogenesis at the refed state. We also show that ARRDC4 coimmunoprecipitates with the glucagon receptor, and ARRDC4 expression is suppressed by insulin. These results define ARRDC4 as a critical regulator of glucagon signaling and glucose homeostasis and reveal a novel intersection of insulin and glucagon pathways in the liver.


Asunto(s)
Glucagón , Insulina , Péptidos y Proteínas de Señalización Intracelular , Hígado , Animales , Ratones , Glucagón/metabolismo , Gluconeogénesis , Glucosa/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Ratones Endogámicos C57BL , Péptidos y Proteínas de Señalización Intracelular/metabolismo
5.
J Biol Chem ; 299(7): 104911, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37311534

RESUMEN

Reversible lysine-63 (K63) polyubiquitination regulates proinflammatory signaling in vascular smooth muscle cells (SMCs) and plays an integral role in atherosclerosis. Ubiquitin-specific peptidase 20 (USP20) reduces NFκB activation triggered by proinflammatory stimuli, and USP20 activity attenuates atherosclerosis in mice. The association of USP20 with its substrates triggers deubiquitinase activity; this association is regulated by phosphorylation of USP20 on Ser334 (mouse) or Ser333 (human). USP20 Ser333 phosphorylation was greater in SMCs of atherosclerotic segments of human arteries as compared with nonatherosclerotic segments. To determine whether USP20 Ser334 phosphorylation regulates proinflammatory signaling, we created USP20-S334A mice using CRISPR/Cas9-mediated gene editing. USP20-S334A mice developed ∼50% less neointimal hyperplasia than congenic WT mice after carotid endothelial denudation. WT carotid SMCs showed substantial phosphorylation of USP20 Ser334, and WT carotids demonstrated greater NFκB activation, VCAM-1 expression, and SMC proliferation than USP20-S334A carotids. Concordantly, USP20-S334A primary SMCs in vitro proliferated and migrated less than WT SMCs in response to IL-1ß. An active site ubiquitin probe bound to USP20-S334A and USP20-WT equivalently, but USP20-S334A associated more avidly with TRAF6 than USP20-WT. IL-1ß induced less K63-linked polyubiquitination of TRAF6 and less downstream NFκB activity in USP20-S334A than in WT SMCs. Using in vitro phosphorylation with purified IRAK1 and siRNA-mediated gene silencing of IRAK1 in SMCs, we identified IRAK1 as a novel kinase for IL-1ß-induced USP20 Ser334 phosphorylation. Our findings reveal novel mechanisms regulating IL-1ß-induced proinflammatory signaling: by phosphorylating USP20 Ser334, IRAK1 diminishes the association of USP20 with TRAF6 and thus augments NFκB activation, SMC inflammation, and neointimal hyperplasia.


Asunto(s)
Aterosclerosis , Inflamación , Quinasas Asociadas a Receptores de Interleucina-1 , Interleucina-1beta , Músculo Liso Vascular , Miocitos del Músculo Liso , Fosfoserina , Ubiquitina Tiolesterasa , Animales , Humanos , Ratones , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células Cultivadas , Hiperplasia/metabolismo , Hiperplasia/patología , Inflamación/metabolismo , Inflamación/patología , Quinasas Asociadas a Receptores de Interleucina-1/química , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fosforilación , Fosfoserina/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Ubiquitina Tiolesterasa/química , Ubiquitina Tiolesterasa/metabolismo , FN-kappa B/metabolismo , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Interleucina-1beta/metabolismo , Ubiquitinación
6.
J Biol Chem ; 299(5): 104690, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37037304

RESUMEN

The pancreatic hormone glucagon activates the glucagon receptor (GCGR), a class B seven-transmembrane G protein-coupled receptor that couples to the stimulatory heterotrimeric G protein and provokes PKA-dependent signaling cascades vital to hepatic glucose metabolism and islet insulin secretion. Glucagon-stimulation also initiates recruitment of the endocytic adaptors, ßarrestin1 and ßarrestin2, which regulate desensitization and internalization of the GCGR. Unlike many other G protein-coupled receptors, the GCGR expressed at the plasma membrane is constitutively ubiquitinated and upon agonist-activation, internalized GCGRs are deubiquitinated at early endosomes and recycled via Rab4-containing vesicles. Herein we report a novel link between the ubiquitination status and signal transduction mechanism of the GCGR. In the deubiquitinated state, coupling of the GCGR to Gs is diminished, while binding to ßarrestin is enhanced with signaling biased to a ßarrestin1-dependent p38 mitogen activated protein kinase (MAPK) pathway. This ubiquitin-dependent signaling bias arises through the modification of lysine333 (K333) on the cytoplasmic face of transmembrane helix V. Compared with the GCGR-WT, the mutant GCGR-K333R has impaired ubiquitination, diminished G protein coupling, and PKA signaling but unimpaired potentiation of glucose-stimulated-insulin secretion in response to agonist-stimulation, which involves p38 MAPK signaling. Both WT and GCGR-K333R promote the formation of glucagon-induced ßarrestin1-dependent p38 signaling scaffold that requires canonical upstream MAPK-Kinase3, but is independent of Gs, Gi, and ßarrestin2. Thus, ubiquitination/deubiquitination at K333 in the GCGR defines the activation of distinct transducers with the potential to influence various facets of glucagon signaling in health and disease.


Asunto(s)
Glucagón , Receptores de Glucagón , Ubiquitinación , Glucagón/metabolismo , Glucosa/metabolismo , Hígado/metabolismo , Receptores de Glucagón/genética , Receptores de Glucagón/metabolismo , Humanos , Células HEK293
7.
J Biol Chem ; 298(5): 101837, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35307348

RESUMEN

Arrestins and their yeast homologs, arrestin-related trafficking adaptors (ARTs), share a stretch of 29 amino acids called the ART motif. However, the functionality of that motif is unknown. We now report that deleting this motif prevents agonist-induced ubiquitination of ß-arrestin2 (ß-arr2) and blocks its association with activated G protein-coupled receptors (GPCRs). Within the ART motif, we have identified a conserved phenylalanine residue, Phe116, that is critical for the formation of ß-arr2-GPCR complexes. ß-arr2 Phe116Ala mutant has negligible effect on blunting ß2-adrenergic receptor-induced cAMP generation unlike ß-arr2, which promotes rapid desensitization. Furthermore, available structures for inactive and inositol hexakisphosphate 6-activated forms of bovine ß-arr2 revealed that Phe116 is ensconced in a hydrophobic pocket, whereas the adjacent Phe117 and Phe118 residues are not. Mutagenesis of Phe117 and Phe118, but not Phe116, preserves GPCR interaction of ß-arr2. Surprisingly, Phe116 is dispensable for the association of ß-arr2 with its non-GPCR partners. ß-arr2 Phe116Ala mutant presents a significantly reduced protein half-life compared with ß-arr2 and undergoes constitutive Lys-48-linked polyubiquitination, which tags proteins for proteasomal degradation. We also found that Phe116 is critical for agonist-dependent ß-arr2 ubiquitination with Lys-63-polyubiquitin linkages that are known mediators of protein scaffolding and signal transduction. Finally, we have shown that ß-arr2 Phe116Ala interaction with activated ß2-adrenergic receptor can be rescued with an in-frame fusion of ubiquitin. Taken together, we conclude that Phe116 preserves structural stability of ß-arr2, regulates the formation of ß-arr2-GPCR complexes that inhibit G protein signaling, and promotes subsequent ubiquitin-dependent ß-arr2 localization and trafficking.


Asunto(s)
Fenilalanina , Receptores Acoplados a Proteínas G/metabolismo , Arrestina beta 2 , Animales , Bovinos , Ubiquitina/metabolismo , Arrestina beta 2/química , Arrestina beta 2/genética , Arrestina beta 2/metabolismo
8.
J Biol Chem ; 295(49): 16630-16642, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-32967969

RESUMEN

The glucagon receptor (GCGR) activated by the peptide hormone glucagon is a seven-transmembrane G protein-coupled receptor (GPCR) that regulates blood glucose levels. Ubiquitination influences trafficking and signaling of many GPCRs, but its characterization for the GCGR is lacking. Using endocytic colocalization and ubiquitination assays, we have identified a correlation between the ubiquitination profile and recycling of the GCGR. Our experiments revealed that GCGRs are constitutively ubiquitinated at the cell surface. Glucagon stimulation not only promoted GCGR endocytic trafficking through Rab5a early endosomes and Rab4a recycling endosomes, but also induced rapid deubiquitination of GCGRs. Inhibiting GCGR internalization or disrupting endocytic trafficking prevented agonist-induced deubiquitination of the GCGR. Furthermore, a Rab4a dominant negative (DN) that blocks trafficking at recycling endosomes enabled GCGR deubiquitination, whereas a Rab5a DN that blocks trafficking at early endosomes eliminated agonist-induced GCGR deubiquitination. By down-regulating candidate deubiquitinases that are either linked with GPCR trafficking or localized on endosomes, we identified signal-transducing adaptor molecule-binding protein (STAMBP) and ubiquitin-specific protease 33 (USP33) as cognate deubiquitinases for the GCGR. Our data suggest that USP33 constitutively deubiquitinates the GCGR, whereas both STAMBP and USP33 deubiquitinate agonist-activated GCGRs at early endosomes. A mutant GCGR with all five intracellular lysines altered to arginines remains deubiquitinated and shows augmented trafficking to Rab4a recycling endosomes compared with the WT, thus affirming the role of deubiquitination in GCGR recycling. We conclude that the GCGRs are rapidly deubiquitinated after agonist-activation to facilitate Rab4a-dependent recycling and that USP33 and STAMBP activities are critical for the endocytic recycling of the GCGR.


Asunto(s)
Enzimas Desubicuitinizantes/metabolismo , Endosomas/metabolismo , Receptores de Glucagón/metabolismo , Proteínas de Unión al GTP rab4/metabolismo , Línea Celular , Regulación hacia Abajo , Complejos de Clasificación Endosomal Requeridos para el Transporte/antagonistas & inhibidores , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Glucagón/farmacología , Humanos , Monensina/farmacología , Mutagénesis , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Glucagón/agonistas , Receptores de Glucagón/genética , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación/efectos de los fármacos , Proteínas de Unión al GTP rab4/genética , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo
9.
Cell ; 182(5): 1362-1362.e1, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32888497

RESUMEN

The arrestins are ubiquitously expressed adaptor proteins that orchestrate transmembrane signaling cascades triggered by the 7-transmembrane G protein-coupled receptors. While originally discovered as proteins that block receptor-G protein coupling, arrestins are now appreciated for their expanding repertoire of dynamic protein interactions and cellular functions.


Asunto(s)
Arrestinas/metabolismo , Membrana Celular/metabolismo , Mapas de Interacción de Proteínas/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología
10.
Cell Signal ; 75: 109759, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32860951

RESUMEN

Small ubiquitin like modifier (SUMO) conjugation or SUMOylation of ßarrestin2 promotes its association with the clathrin adaptor protein AP2 and facilitates rapid ß2 adrenergic receptor (ß2AR) internalization. However, disruption of the consensus SUMOylation site in ßarrestin2, did not prevent ßarrestin2's association with activated ß2ARs, dopamine D2 receptors (D2Rs), angiotensin type 1a receptors (AT1aRs) and V2 vasopressin receptors (V2Rs). To address the role of SUMOylation in the trafficking of ßarrestin and GPCR complexes, we generated and characterized a yellow fluorescent protein (YFP) tagged ßarrestin2-SUMO1 chimeric protein, which is resistant to de-SUMOylation. In HEK-293 cells, YFP-SUMO1 predominantly localized in the nucleus, whereas YFP-ßarrestin2 is cytoplasmic. YFP-ßarrestin2-SUMO1 in addition to being cytoplasmic, is localized at the nuclear membrane. Nonetheless, ßarrestin2-SUMO1 associated robustly with agonist-activated ß2ARs as evaluated by co-immunoprecipitation, confocal microscopy and bioluminescence resonance energy transfer (BRET). ßarrestin2-SUMO1 associated strongly with the D2R, which forms transient complexes with ßarrestin2. But, ßarrestin2-SUMO1 and ßarrestin2 showed equivalent binding with the V2R, which forms stable complexes with ßarrestin2. ßarrestin2 expression level directly correlated with the steady state levels of the unmodified form of RanGAP1, which upon SUMOylation associates with nuclear membrane. On the other hand, ßarrestin2-SUMO1 not only localized at the nuclear membrane, but also formed a macromolecular complex with RanGAP1. Taken together, our data suggest that SUMOylation of ßarrestin2 promotes its protein interactions at both cell and nuclear membranes. Furthermore, ßarrestin2-SUMO1 presents as a useful tool to characterize ßarrestin2 recruitment to GPCRs, which form transient and unstable complex with ßarrestin2.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Poro Nuclear/metabolismo , Proteína SUMO-1/metabolismo , Arrestina beta 2/metabolismo , Células HEK293 , Humanos , Unión Proteica , Transporte de Proteínas , Sumoilación
11.
Methods Mol Biol ; 1957: 93-104, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30919349

RESUMEN

Ubiquitination of G protein-coupled receptors (GPCRs) is an important dynamic posttranslational modification that has been linked to the intracellular trafficking of internalized GPCRs to lysosomes. Ubiquitination of GPCRs is mediated by specific E3 ubiquitin ligases that are scaffolded by the adaptor proteins called ß-arrestins. Traditionally, detection of GPCR ubiquitination is achieved by using ubiquitin antibodies to Western blot immunoprecipitates of detergent-solubilized GPCRs expressed in heterologous cells. However, studies have also shown that bioluminescence resonance energy transfer (BRET)-based techniques can reveal ubiquitination of GPCRs in intact cells and in real time. This chapter describes a step-by-step protocol to evaluate ubiquitination of GPCRs using the BRET methodology.


Asunto(s)
Transferencia de Energía por Resonancia de Bioluminiscencia/métodos , Receptores Acoplados a Proteínas G/metabolismo , Ubiquitinación , beta-Arrestinas/metabolismo , Análisis de Datos , Células HEK293 , Humanos , Unión Proteica , Ubiquitina/metabolismo
12.
J Biol Chem ; 294(7): 2500-2518, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30538132

RESUMEN

Reversible ubiquitination of G protein-coupled receptors regulates their trafficking and signaling; whether deubiquitinases regulate myocardial ß1-adrenergic receptors (ß1ARs) is unknown. We report that ubiquitin-specific protease 20 (USP20) deubiquitinates and attenuates lysosomal trafficking of the ß1AR. ß1AR-induced phosphorylation of USP20 Ser-333 by protein kinase A-α (PKAα) was required for optimal USP20-mediated regulation of ß1AR lysosomal trafficking. Both phosphomimetic (S333D) and phosphorylation-impaired (S333A) USP20 possess intrinsic deubiquitinase activity equivalent to WT activity. However, unlike USP20 WT and S333D, the S333A mutant associated poorly with the ß1AR and failed to deubiquitinate the ß1AR. USP20-KO mice showed normal baseline systolic function but impaired ß1AR-induced contractility and relaxation. Dobutamine stimulation did not increase cAMP in USP20-KO left ventricles (LVs), whereas NKH477-induced adenylyl cyclase activity was equivalent to WT. The USP20 homolog USP33, which shares redundant roles with USP20, had no effect on ß1AR ubiquitination, but USP33 was up-regulated in USP20-KO hearts suggesting compensatory regulation. Myocardial ß1AR expression in USP20-KO was drastically reduced, whereas ß2AR expression was maintained as determined by radioligand binding in LV sarcolemmal membranes. Phospho-USP20 was significantly increased in LVs of wildtype (WT) mice after a 1-week catecholamine infusion and a 2-week chronic pressure overload induced by transverse aortic constriction (TAC). Phospho-USP20 was undetectable in ß1AR KO mice subjected to TAC, suggesting a role for USP20 phosphorylation in cardiac response to pressure overload. We conclude that USP20 regulates ß1AR signaling in vitro and in vivo Additionally, ß1AR-induced USP20 phosphorylation may serve as a feed-forward mechanism to stabilize ß1AR expression and signaling during pathological insults to the myocardium.


Asunto(s)
Endopeptidasas/biosíntesis , Regulación Enzimológica de la Expresión Génica , Activación del Canal Iónico , Miocardio/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Sustitución de Aminoácidos , Animales , Endopeptidasas/genética , Ventrículos Cardíacos , Ratones , Ratones Noqueados , Mutación Missense , Fosforilación , Receptores Adrenérgicos beta 1/genética , Ubiquitina Tiolesterasa , Ubiquitinación
13.
ACS Pharmacol Transl Sci ; 2(4): 230-246, 2019 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-32259059

RESUMEN

G-protein-coupled receptors (GPCRs) can bias signaling through distinct biochemical pathways that originate from G-protein/receptor and ß-arrestin/receptor complexes. Receptor conformations supporting ß-arrestin engagement depend on multiple receptor determinants. Using ghrelin receptor GHR1a, we demonstrate by bioluminescence resonance energy transfer and fluorescence microscopy a critical role for its second intracellular loop 2 (ICL2) domain in stabilizing ß-arrestin/GHSR1a core interactions and determining receptor trafficking fate. We validate our findings in ICL2 gain- and loss-of-function experiments assessing ß-arrestin and ubiquitin-dependent internalization of the CC chemokine receptor, CCR1. Like all CC and CXC subfamily chemokine receptors, CCR1 lacks a critical proline residue found in the ICL2 consensus domain of rhodopsin-family GPCRs. Our study indicates that ICL2, C-tail determinants, and the orthosteric binding pocket that regulates ß-arrestin/receptor complex stability are sufficient to encode a broad repertoire of the trafficking fates observed for rhodopsin-family GPCRs, suggesting they provide the essential elements for regulating a large fraction of ß-arrestin signaling bias.

14.
Arterioscler Thromb Vasc Biol ; 38(10): 2295-2305, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354204

RESUMEN

Objective- Signaling that activates NFκB (nuclear factor κB) in smooth muscle cells (SMCs) is integral to atherosclerosis and involves reversible ubiquitination that activates proteins downstream of proatherogenic receptors. Deubiquitination of these proteins is mediated by USP20 (ubiquitin-specific protease 20), among other deubiquitinases. We sought to determine whether USP20 activity in SMCs decreases atherosclerosis. Approach and Results- To address this question, we used male Ldlr-/- mice without (control) or with SMC-specific expression of murine USP20 (SMC-USP20-transgenic) or its dominant-negative (DN; C154S/H643Q) mutant (SMC-DN-USP20-transgenic). Before the appearance of intimal macrophages, NFκB activation in aortic medial SMCs was greater in SMC-DN-USP20-transgenic than in control mice. After 16 weeks on a Western diet, SMC-DN-USP20-transgenic mice had 46% greater brachiocephalic artery atheroma area than control mice. Congruently, aortic atherosclerosis assessed en face was 21% greater than control in SMC-DN-USP20-transgenic mice and 13% less than control in SMC-USP20-transgenic mice. In response to TNF (tumor necrosis factor), SMCs from SMC-DN-USP20-transgenic mice showed ≈3-fold greater NFκB activation than control SMCs. Silencing USP20 in SMCs with siRNA (small interfering RNA) augmented NFκB activation by ≈50% in response to either TNF or IL-1ß (interleukin-1ß). Coimmunoprecipitation experiments revealed that USP20 associates with several components of the TNFR1 (TNF receptor-1) signaling pathway, including RIPK1 (receptor-interacting protein kinase 1), a critical checkpoint in TNF-induced NFκB activation and inflammation. TNF evoked ≈2-fold more RIPK1 ubiquitination in SMC-DN-USP20-transgenic than in control SMCs, and RIPK1 was deubiquitinated by purified USP20 in vitro. Conclusions- USP20 attenuates TNF- and IL-1ß-evoked atherogenic signaling in SMCs, by deubiquitinating RIPK1, among other signaling intermediates.


Asunto(s)
Aortitis/prevención & control , Aterosclerosis/prevención & control , Endopeptidasas/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Aorta/patología , Aortitis/enzimología , Aortitis/genética , Aortitis/patología , Aterosclerosis/enzimología , Aterosclerosis/genética , Aterosclerosis/patología , Células Cultivadas , Modelos Animales de Enfermedad , Endopeptidasas/genética , Femenino , Hiperplasia , Interleucina-1beta/farmacología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , FN-kappa B/metabolismo , Neointima , Placa Aterosclerótica , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de LDL , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Ubiquitina Tiolesterasa , Ubiquitinación
15.
Sci Signal ; 11(549)2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30254056

RESUMEN

G protein-coupled receptors (GPCRs) use diverse mechanisms to regulate the mitogen-activated protein kinases ERK1/2. ß-Arrestins (ßArr1/2) are ubiquitous inhibitors of G protein signaling, promoting GPCR desensitization and internalization and serving as scaffolds for ERK1/2 activation. Studies using CRISPR/Cas9 to delete ßArr1/2 and G proteins have cast doubt on the role of ß-arrestins in activating specific pools of ERK1/2. We compared the effects of siRNA-mediated knockdown of ßArr1/2 and reconstitution with ßArr1/2 in three different parental and CRISPR-derived ßArr1/2 knockout HEK293 cell pairs to assess the effect of ßArr1/2 deletion on ERK1/2 activation by four Gs-coupled GPCRs. In all parental lines with all receptors, ERK1/2 stimulation was reduced by siRNAs specific for ßArr2 or ßArr1/2. In contrast, variable effects were observed with CRISPR-derived cell lines both between different lines and with activation of different receptors. For ß2 adrenergic receptors (ß2ARs) and ß1ARs, ßArr1/2 deletion increased, decreased, or had no effect on isoproterenol-stimulated ERK1/2 activation in different CRISPR clones. ERK1/2 activation by the vasopressin V2 and follicle-stimulating hormone receptors was reduced in these cells but was enhanced by reconstitution with ßArr1/2. Loss of desensitization and receptor internalization in CRISPR ßArr1/2 knockout cells caused ß2AR-mediated stimulation of ERK1/2 to become more dependent on G proteins, which was reversed by reintroducing ßArr1/2. These data suggest that ßArr1/2 function as a regulatory hub, determining the balance between mechanistically different pathways that result in activation of ERK1/2, and caution against extrapolating results obtained from ßArr1/2- or G protein-deleted cells to GPCR behavior in native systems.


Asunto(s)
Sistemas CRISPR-Cas , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , beta-Arrestinas/metabolismo , Membrana Celular/metabolismo , Activación Enzimática , Eliminación de Gen , Edición Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Fosforilación , Receptores Adrenérgicos beta 2/metabolismo
16.
Cardiovasc Res ; 114(13): 1806-1815, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29931051

RESUMEN

Aims: The actin-binding protein Drebrin is up-regulated in response to arterial injury and reduces smooth muscle cell (SMC) migration and proliferation through its interaction with the actin cytoskeleton. We, therefore, tested the hypothesis that SMC Drebrin inhibits angiotensin II-induced remodelling of the proximal aorta. Methods and results: Angiotensin II was administered via osmotic minipumps at 1000 ng/kg/min continuously for 28 days in SM22-Cre+/Dbnflox/flox (SMC-Dbn-/-) and control mice. Blood pressure responses to angiotensin II were assessed by telemetry. After angiotensin II infusion, we assessed remodelling in the proximal ascending aorta by echocardiography and planimetry of histological cross sections. Although the degree of hypertension was equivalent in SMC-Dbn-/- and control mice, SMC-Dbn-/- mice nonetheless exhibited 60% more proximal aortic medial thickening and two-fold more outward aortic remodelling than control mice in response to angiotensin II. Proximal aortas demonstrated greater cellular proliferation and matrix deposition in SMC-Dbn-/- mice than in control mice, as evidenced by a higher prevalence of proliferating cell nuclear antigen-positive nuclei and higher levels of collagen I. Compared with control mouse aortas, SMC-Dbn-/- aortas demonstrated greater angiotensin II-induced NADPH oxidase activation and inflammation, evidenced by higher levels of Ser-536-phosphorylated NFκB p65 subunits and higher levels of vascular cell adhesion molecule-1, matrix metalloproteinase-9, and adventitial macrophages. Conclusions: We conclude that SMC Drebrin deficiency augments angiotensin II-induced inflammation and adverse aortic remodelling.


Asunto(s)
Angiotensina II , Enfermedades de la Aorta/metabolismo , Hipertensión/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neuropéptidos/metabolismo , Remodelación Vascular , Animales , Aorta/metabolismo , Aorta/patología , Aorta/fisiopatología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/fisiopatología , Presión Arterial , Proliferación Celular , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Células HEK293 , Humanos , Hipertensión/genética , Hipertensión/patología , Hipertensión/fisiopatología , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/patología , NADPH Oxidasas/metabolismo , Neuropéptidos/deficiencia , Neuropéptidos/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
17.
Cell Signal ; 41: 41-45, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28189586

RESUMEN

The ubiquitously expressed, multifunctional scaffolding proteins ß-arrestin1 and ß-arrestin2 each affect inflammatory signaling in a variety of cell lines. In addition to binding the carboxyl-terminal tails of innumerable 7-transmembrane receptors, ß-arrestins scaffold untold numbers of other plasma membrane and cytoplasmic proteins. Consequently, the effects of ß-arrestins on inflammatory signaling are diverse, and context-specific. This review highlights the roles of ß-arrestins in regulating canonical activation of the pro-inflammatory transcription factor NFκB.


Asunto(s)
Inflamación/metabolismo , FN-kappa B/metabolismo , beta-Arrestinas/metabolismo , Animales , Línea Celular , Humanos , Ratones , Ratones Noqueados , FN-kappa B/genética , Transfección , Ubiquitinación , beta-Arrestinas/genética
18.
Cell Signal ; 41: 9-16, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28137506

RESUMEN

G protein-coupled receptors (GPCRs) transduce a wide array of extracellular signals and regulate virtually every aspect of physiology. While GPCR signaling is essential, overstimulation can be deleterious, resulting in cellular toxicity or uncontrolled cellular growth. Accordingly, nature has developed a number of mechanisms for limiting GPCR signaling, which are broadly referred to as desensitization, and refer to a decrease in response to repeated or continuous stimulation. Short-term desensitization occurs over minutes, and is primarily associated with ß-arrestins preventing G protein interaction with a GPCR. Longer-term desensitization, referred to as downregulation, occurs over hours to days, and involves receptor internalization into vesicles, degradation in lysosomes and decreased receptor mRNA levels through unclear mechanisms. Phosphorylation of the receptor by GPCR kinases (GRKs) and the recruitment of ß-arrestins is critical to both these short- and long-term desensitization mechanisms. In addition to phosphorylation, both the GPCR and ß-arrestins are modified post-translationally in several ways, including by ubiquitination. For many GPCRs, receptor ubiquitination promotes degradation of agonist-activated receptors in the lysosomes. Other proteins also play important roles in desensitization, including phosphodiesterases, RGS family proteins and A-kinase-anchoring proteins. Together, this intricate network of kinases, ubiquitin ligases, and adaptor proteins orchestrate the acute and prolonged desensitization of GPCRs.


Asunto(s)
Endocitosis , Proteolisis , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Lisosomas/metabolismo , Fosforilación , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Ubiquitinación , beta-Arrestinas/metabolismo
19.
Cardiovasc Res ; 113(13): 1551-1559, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29048463

RESUMEN

AIMS: Chronic kidney disease (CKD) is a powerful independent risk factor for cardiovascular events, including vein graft failure. Because CKD impairs the clearance of small proteins, we tested the hypothesis that CKD exacerbates vein graft disease by elevating serum levels of critical cytokines that promote vein graft neointimal hyperplasia. METHODS AND RESULTS: We modelled CKD in C57BL/6 mice with 5/6ths nephrectomy, which reduced glomerular filtration rate by 60%, and we modelled vein grafting with inferior-vena-cava-to-carotid interposition grafting. CKD increased vein graft neointimal hyperplasia four-fold, decreased vein graft re-endothelialization two-fold, and increased serum levels of interleukin-9 (IL-9) five-fold. By quantitative immunofluorescence and histochemical staining, vein grafts from CKD mice demonstrated a ∼two-fold higher prevalence of mast cells, and a six-fold higher prevalence of activated mast cells. Concordantly, vein grafts from CKD mice showed higher levels of TNF and NFκB activation, as judged by phosphorylation of NFκB p65 on Ser536 and by expression of VCAM-1. Arteriovenous fistula veins from humans with CKD also showed up-regulation of mast cells and IL-9. Treating CKD mice with IL-9-neutralizing IgG reduced vein graft neointimal area four-fold, increased vein graft re-endothelialization ∼two-fold, and reduced vein graft total and activated mast cell levels two- and four-fold, respectively. Treating CKD mice with the mast cell stabilizer cromolyn reduced neointimal hyperplasia and increased re-endothelialization in vein grafts. In vitro, IL-9 promoted endothelial cell apoptosis but had no effect on smooth muscle cell proliferation. CONCLUSION: CKD aggravates vein graft disease through mechanisms involving IL-9 and mast cell activation.


Asunto(s)
Derivación Arteriovenosa Quirúrgica , Arteria Carótida Común/cirugía , Interleucina-9/metabolismo , Mastocitos/metabolismo , Insuficiencia Renal Crónica/complicaciones , Enfermedades Vasculares/complicaciones , Vena Cava Inferior/trasplante , Animales , Apoptosis , Arteria Carótida Común/inmunología , Arteria Carótida Común/metabolismo , Arteria Carótida Común/patología , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Hiperplasia , Interleucina-9/inmunología , Mastocitos/inmunología , Ratones Endogámicos C57BL , Neointima , Fosforilación , Insuficiencia Renal Crónica/inmunología , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal , Factores de Tiempo , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología , Vena Cava Inferior/inmunología , Vena Cava Inferior/metabolismo , Vena Cava Inferior/patología
20.
JCI Insight ; 2(17)2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28878120

RESUMEN

The oncoprotein Mdm2 is a RING domain-containing E3 ubiquitin ligase that ubiquitinates G protein-coupled receptor kinase 2 (GRK2) and ß-arrestin2, thereby regulating ß-adrenergic receptor (ßAR) signaling and endocytosis. Previous studies showed that cardiac Mdm2 expression is critical for controlling p53-dependent apoptosis during early embryonic development, but the role of Mdm2 in the developed adult heart is unknown. We aimed to identify if Mdm2 affects ßAR signaling and cardiac function in adult mice. Using Mdm2/p53-KO mice, which survive for 9-12 months, we identified a critical and potentially novel role for Mdm2 in the adult mouse heart through its regulation of cardiac ß1AR signaling. While baseline cardiac function was mostly similar in both Mdm2/p53-KO and wild-type (WT) mice, isoproterenol-induced cardiac contractility in Mdm2/p53-KO was significantly blunted compared with WT mice. Isoproterenol increased cAMP in left ventricles of WT but not of Mdm2/p53-KO mice. Additionally, while basal and forskolin-induced calcium handling in isolated Mdm2/p53-KO and WT cardiomyocytes were equivalent, isoproterenol-induced calcium handling in Mdm2/p53-KO was impaired. Mdm2/p53-KO hearts expressed 2-fold more GRK2 than WT. GRK2 polyubiquitination via lysine-48 linkages was significantly reduced in Mdm2/p53-KO hearts. Tamoxifen-inducible cardiomyocyte-specific deletion of Mdm2 in adult mice also led to a significant increase in GRK2, and resulted in severely impaired cardiac function, high mortality, and no detectable ßAR responsiveness. Gene delivery of either Mdm2 or GRK2-CT in vivo using adeno-associated virus 9 (AAV9) effectively rescued ß1AR-induced cardiac contractility in Mdm2/p53-KO. These findings reveal a critical p53-independent physiological role of Mdm2 in adult hearts, namely, regulation of GRK2-mediated desensitization of ßAR signaling.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Contracción Miocárdica/fisiología , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal , Agonistas Adrenérgicos beta/farmacología , Animales , Ecocardiografía , Quinasa 2 del Receptor Acoplado a Proteína-G/antagonistas & inhibidores , Corazón/diagnóstico por imagen , Corazón/fisiología , Hemodinámica/efectos de los fármacos , Isoproterenol/farmacología , Ratones , Ratones Noqueados , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA