Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(16)2023 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-37629047

RESUMEN

Current drugs for treating heart failure (HF), for example, angiotensin II receptor blockers and ß-blockers, possess specific target molecules involved in the regulation of the cardiac circulatory system. However, most clinically approved drugs are effective in the treatment of HF with reduced ejection fraction (HFrEF). Novel drug classes, including angiotensin receptor blocker/neprilysin inhibitor (ARNI), sodium-glucose co-transporter-2 (SGLT2) inhibitor, hyperpolarization-activated cyclic nucleotide-gated (HCN) channel blocker, soluble guanylyl cyclase (sGC) stimulator/activator, and cardiac myosin activator, have recently been introduced for HF intervention based on their proposed novel mechanisms. SGLT2 inhibitors have been shown to be effective not only for HFrEF but also for HF with preserved ejection fraction (HFpEF). In the myocardium, excess cyclic adenosine monophosphate (cAMP) stimulation has detrimental effects on HFrEF, whereas cyclic guanosine monophosphate (cGMP) signaling inhibits cAMP-mediated responses. Thus, molecules participating in cGMP signaling are promising targets of novel drugs for HF. In this review, we summarize molecular pathways of cGMP signaling and clinical trials of emerging drug classes targeting cGMP signaling in the treatment of HF.


Asunto(s)
Insuficiencia Cardíaca , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Humanos , Insuficiencia Cardíaca/tratamiento farmacológico , Volumen Sistólico , Corazón , Miocardio , Antagonistas de Receptores de Angiotensina , Bloqueadores de los Canales de Calcio , AMP Cíclico , GMP Cíclico , Vasodilatadores
2.
Int J Mol Sci ; 24(4)2023 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-36835211

RESUMEN

Nonalcoholic steatohepatitis (NASH) is a disease that progresses from nonalcoholic fatty liver (NAFL) and which is characterized by inflammation and fibrosis. The purinergic P2Y6 receptor (P2Y6R) is a pro-inflammatory Gq/G12 family protein-coupled receptor and reportedly contributes to intestinal inflammation and cardiovascular fibrosis, but its role in liver pathogenesis is unknown. Human genomics data analysis revealed that the liver P2Y6R mRNA expression level is increased during the progression from NAFL to NASH, which positively correlates with inductions of C-C motif chemokine 2 (CCL2) and collagen type I α1 chain (Col1a1) mRNAs. Therefore, we examined the impact of P2Y6R functional deficiency in mice crossed with a NASH model using a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD). Feeding CDAHFD for 6 weeks markedly increased P2Y6R expression level in mouse liver, which was positively correlated with CCL2 mRNA induction. Unexpectedly, the CDAHFD treatment for 6 weeks increased liver weights with severe steatosis in both wild-type (WT) and P2Y6R knockout (KO) mice, while the disease marker levels such as serum AST and liver CCL2 mRNA in CDAHFD-treated P2Y6R KO mice were rather aggravated compared with those of CDAHFD-treated WT mice. Thus, P2Y6R may not contribute to the progression of liver injury, despite increased expression in NASH liver.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Receptores Purinérgicos P2 , Animales , Humanos , Ratones , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , ARN Mensajero/metabolismo , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo
3.
Biol Pharm Bull ; 45(6): 669-674, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35650094

RESUMEN

Agonists are defined as the ligands that activate intracellular signaling and evoke cellular responses. Synthetic and endogenous agonists should bind specific amino acids to activate G protein-coupled receptor (GPCR). Agonists that induce maximal responses are full agonists. Partial agonists cannot induce full responses unlike full agonists. In definition, antagonists inhibit agonist-stimulated responses by binding to orthosteric or allosteric sites. Antagonists modulate agonist-induced responses and are often related with inverse agonist activity. However, the relationship between antagonists and partial agonists is complex. An antagonist behaves as a partial agonist when the constitutive activity of the GPCR is high. In contrast, a partial agonist with very weak intrinsic activity may be classified as an antagonist. Thus, antagonisms of the compounds are influenced by constitutive activity of GPCRs, intrinsic activity and differences in the binding sites of GPCRs. Since "antagonism" has been revealed to have multiple aspects and more complex than previously thought, it may be difficult to classify each compound as simply "agonist" or "antagonist" as before. In this review, we discuss the recent findings and perspectives on the pharmacology of GPCR-binding antagonists, inverse agonists, and signaling.


Asunto(s)
Receptores Acoplados a Proteínas G , Transducción de Señal , Sitios de Unión , Ligandos , Receptores Acoplados a Proteínas G/metabolismo
4.
Development ; 145(14)2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-29986870

RESUMEN

Erk5 belongs to the mitogen-activated protein kinase (MAPK) family. Following its phosphorylation by Mek5, Erk5 modulates several signaling pathways in a number of cell types. In this study, we demonstrated that Erk5 inactivation in mesenchymal cells causes abnormalities in skeletal development by inducing Sox9, an important transcription factor of skeletogenesis. We further demonstrate that Erk5 directly phosphorylates and activates Smurf2 (a ubiquitin E3 ligase) at Thr249, which promotes the proteasomal degradation of Smad proteins and phosphorylates Smad1 at Ser206 in the linker region known to trigger its proteasomal degradation by Smurf1. Smads transcriptionally activated the expression of Sox9 in mesenchymal cells. Accordingly, removal of one Sox9 allele in mesenchymal cells from Erk5-deficient mice rescued some abnormalities of skeletogenesis. These findings highlight the importance of the Mek5-Erk5-Smurf-Smad-Sox9 axis in mammalian skeletogenesis.


Asunto(s)
Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Osteogénesis , Factor de Transcripción SOX9/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Diferenciación Celular , Condrogénesis , Humanos , Mesodermo/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteolisis , Cráneo/anomalías , Ubiquitina/metabolismo , Ubiquitinación
5.
FASEB J ; 34(6): 8749-8763, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32385915

RESUMEN

Leukotriene B4 receptor 1 (BLT1), a high-affinity G-protein-coupled receptor for leukotriene B4 (LTB4 ), is expressed on various inflammatory cells and plays critical roles in several inflammatory diseases. In myocardial infarction (MI), various inflammatory cells are known to be recruited to the infarcted area, but the function of BLT1 in MI is poorly understood. Here, we investigated the role of BLT1 in MI and the therapeutic effect of a BLT1 antagonist, ONO-4057, on MI. Mice with infarcted hearts showed increased BLT1 expression and LTB4 levels. BLT1-knockout mice with infarcted hearts exhibited attenuated leukocyte infiltration, proinflammatory cytokine production, and cell death, which led to reduced mortality and improved cardiac function after MI. Bone-marrow transplantation studies showed that BLT1 expressed on bone marrow-derived cells was responsible for the exacerbation of inflammation in infarcted hearts. Furthermore, ONO-4057 administration attenuated the inflammatory responses in hearts surgically treated for MI, which resulted in reduced mortality and improved cardiac function after MI. Our study demonstrated that BLT1 contributes to excessive inflammation after MI and could represent a new therapeutic target for MI.


Asunto(s)
Inflamación/metabolismo , Infarto del Miocardio/metabolismo , Receptores de Leucotrieno B4/metabolismo , Animales , Modelos Animales de Enfermedad , Leucotrieno B4/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología
6.
Biochem Biophys Res Commun ; 529(2): 224-230, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32703415

RESUMEN

Fibrosis is attributed to excess deposition of extracellular matrix (ECM) proteins including collagen and is associated with various organ dysfunction. This excessive ECM is produced by myofibroblasts, which are differentiated from various cells by a variety of stimuli, represented by TGF-ß. However, molecular mechanisms for the regulation of ECM production in myofibroblasts remain obscure. In this study, we demonstrate that the expression of drebrin, which binds to and increases the stability of actin filament in neurons, is increased in mouse hearts and lungs upon fibrosis. Drebrin is mainly expressed in myofibroblasts in the fibrotic hearts and lungs and promotes the expression of fibrosis-related genes, such as Acta2 and Col1a1. Taken together, our study identifies drebrin as a molecule that promotes the production of fibrosis-related genes in myofibroblasts.


Asunto(s)
Pulmón/patología , Miocardio/patología , Miofibroblastos/patología , Neuropéptidos/genética , Animales , Diferenciación Celular , Células Cultivadas , Fibrosis , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miofibroblastos/metabolismo , Células 3T3 NIH , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Regulación hacia Arriba
7.
J Biol Chem ; 292(25): 10586-10599, 2017 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-28487374

RESUMEN

Dioxin and related chemicals alter the expression of a number of genes by activating the aryl hydrocarbon receptors (AHR) to produce a variety of disorders including hepatotoxicity. However, it remains largely unknown how these changes in gene expression are linked to toxicity. To address this issue, we initially examined the effect of 2,3,7,8-tetrachrolodibenzo-p-dioxin (TCDD), a most toxic dioxin, on the hepatic and serum metabolome in male pubertal rats and found that TCDD causes many changes in the level of fatty acids, bile acids, amino acids, and their metabolites. Among these findings was the discovery that TCDD increases the content of leukotriene B4 (LTB4), an inducer of inflammation due to the activation of leukocytes, in the liver of rats and mice. Further analyses suggested that an increase in LTB4 comes from a dual mechanism consisting of an induction of arachidonate lipoxygenase-5, a rate-limiting enzyme in LTB4 synthesis, and the down-regulation of LTC4 synthase, an enzyme that converts LTA4 to LTC4. The above changes required AHR activation, because the same was not observed in AHR knock-out rats. In agreement with LTB4 accumulation, TCDD caused the marked infiltration of neutrophils into the liver. However, deleting LTB4 receptors (BLT1) blocked this effect. A TCDD-produced increase in the mRNA expression of inflammatory markers, including tumor-necrosis factor and hepatic damage, was also suppressed in BLT1-null mice. The above observations focusing on metabolomic changes provide novel evidence that TCDD accumulates LTB4 in the liver by an AHR-dependent induction of LTB4 biosynthesis to cause hepatotoxicity through neutrophil activation.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Dioxinas/toxicidad , Leucotrieno B4/biosíntesis , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Leucotrieno B4/genética , Activación Neutrófila/efectos de los fármacos , Infiltración Neutrófila/genética , Neutrófilos/patología , Ratas , Ratas Mutantes , Receptores de Hidrocarburo de Aril/genética , Receptores de Leucotrieno B4/genética , Receptores de Leucotrieno B4/metabolismo
8.
J Immunoassay Immunochem ; 37(3): 296-306, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26744787

RESUMEN

We propose a new approach of an indirect enzyme-linked immunosorbent assay (ELISA) for determination of D-glutamic acid (D-Glu) using a monoclonal antibody against D-glutamic acid (D-Glu-MAb), which recognizes D-Glu-glutaraldehyde (GA) molecule but not D-Glu molecule. Human serum albumin (HSA) was coated on an immunoplate and reacted with D-Glu via GA to produce D-Glu-GA-HSA conjugates in situ in the well to be recognized by D-Glu-MAb, which enabled the development of an indirect ELISA for the determination of free D-Glu. In this indirect ELISA, D-Glu can be specifically detected with limit of detection of 7.81 µ g/mL. Since anti-conjugate antibodies are often produced, even though anti-hapten antibodies are desired, this new approach could be very useful as an application of anti-conjugate antibodies to the development of quantitative analysis for detecting hapten.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Glutamatos/análisis , Glutamatos/química , Glutaral/química , Glutaral/inmunología , Reacciones Antígeno-Anticuerpo , Humanos , Estructura Molecular , Albúmina Sérica/química , Albúmina Sérica/inmunología
9.
Biochem Biophys Res Commun ; 461(2): 307-13, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25881508

RESUMEN

G protein-coupled receptor kinases (GRKs) comprise a family of seven serine/threonine kinases that phosphorylate agonist-activated G protein-coupled receptors (GPCRs). It has recently been reported that GRKs regulate GPCR-independent signaling through the phosphorylation of intracellular proteins. To date, several intracellular substrates for GRK2 and GRK5 have been reported. However, those for GRK6 are poorly understood. Here we identified IκBα, a negative regulator of NF-κB signaling, as a substrate for GRK6. GRK6 directly phosphorylated IκBα at Ser(32)/Ser(36), and the kinase activity of GRK6 was required for the promotion of NF-κB signaling after TNF-α stimulation. Knockout of GRK6 in peritoneal macrophages remarkably attenuated the transcription of inflammatory genes after TNF-α stimulation. In addition, we developed a bioluminescence resonance energy transfer (BRET) probe to monitor GRK6 activity. Using this probe, we revealed that the conformational change of GRK6 was induced by TNF-α. In summary, our study demonstrates that TNF-α induces GRK6 activation, and GRK6 promotes inflammatory responses through the phosphorylation of IκBα.


Asunto(s)
Quinasas de Receptores Acoplados a Proteína-G/inmunología , Proteínas I-kappa B/inmunología , Inflamación/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Células Cultivadas , Quinasas de Receptores Acoplados a Proteína-G/química , Quinasas de Receptores Acoplados a Proteína-G/genética , Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Proteínas I-kappa B/química , Proteínas I-kappa B/metabolismo , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa , FN-kappa B/inmunología , Células 3T3 NIH , Fosforilación , Conformación Proteica
10.
Circ Res ; 112(2): 327-34, 2013 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-23212582

RESUMEN

RATIONALE: The clinical problem of loss of ß-adrenergic receptor (ß-AR) response, both in the pathogenesis of heart failure and during therapeutic application of ß-agonists, is attributable, at least in part, to desensitization, internalization, and downregulation of the receptors. In the regulation of ß-AR signaling, G protein-coupled receptor kinase 2 (GRK2) primarily phosphorylates agonist-occupied ß-ARs, and this modification promotes desensitization, internalization, and downregulation of ß-ARs. It has been demonstrated that GRK2 is inhibited by its S-nitrosylation. However, compounds that induce S-nitrosylation, such as S-nitrosoglutathione, simultaneously generate NO, which has been demonstrated to operate for cardiovascular protection. OBJECTIVE: We examine whether S-nitrosylation without NO generation inhibits desensitization of ß(2)-AR by GRK2. We thus aim to synthesize compounds that specifically induce S-nitrosylation. METHODS AND RESULTS: We have developed water-soluble N-nitrosamines that have S-nitrosylating activity but lack NO-generating activity. These compounds, at least partly, rescue ß-AR from desensitization in HEK 293 cells expressing FLAG-tagged human ß(2)-AR and in rat cardiac myocytes. They inhibit isoproterenol-dependent phosphorylation and internalization of ß(2)-AR. Indeed, they nitrosylate GRK2 in vitro and in cells, and their S-nitrosylation of GRK2 likely underlies their inhibition of ß(2)-AR desensitization. CONCLUSIONS: Compounds that induce S-nitrosylation without NO release inhibit GRK2 and attenuate ß(2)-AR desensitization. Developing water-soluble drugs that specifically induce S-nitrosylation may be a promising therapeutic strategy for heart failure.


Asunto(s)
Óxido Nítrico , Nitrosaminas/metabolismo , Nitrosaminas/farmacología , Receptores Adrenérgicos beta 2/metabolismo , Agua/fisiología , Animales , Células HEK293 , Humanos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Óxido Nítrico/metabolismo , Nitrosaminas/química , Ratas , Solubilidad/efectos de los fármacos , Agua/química
11.
Genes Cells ; 18(12): 1095-106, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24134321

RESUMEN

Hyperactivation of Gq signaling causes cardiac hypertrophy, and ß-adrenergic receptor-mediated Gs signaling is attenuated in hypertrophic cardiomyocytes. Here, we found the increase in a global ubiquitination in hypertrophic mouse heart. The activation of Gq signaling resulted in the ubiquitination of Gαs in neonatal rat cardiomyocytes, reduced Gαs expression, and suppressed cAMP response to ß-adrenergic receptor stimulation. Ectopic expression of Gαq induced a similar suppression, which is due to the degradation of Gαs by a ubiquitin-proteasome pathway. Co-expression of Ric-8B, a positive regulator of Gαs, effectively canceled the Gαq-induced ubiquitination of Gαs and recovered the cAMP accumulation. In vitro, Gαq competes for the binding of Gαs to Ric-8B. These data show a new role of Ric-8B in the crosstalk of two distinct G protein signaling pathways, which are possibly involved in a part of mechanisms of chronic heart failure.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Miocitos Cardíacos/metabolismo , Ubiquitinación , Animales , Cardiomegalia/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Ratas , Ratas Wistar , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal
12.
Nat Chem Biol ; 8(8): 714-24, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22772154

RESUMEN

An emerging aspect of redox signaling is the pathway mediated by electrophilic byproducts, such as nitrated cyclic nucleotide (for example, 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP)) and nitro or keto derivatives of unsaturated fatty acids, generated via reactions of inflammation-related enzymes, reactive oxygen species, nitric oxide and secondary products. Here we report that enzymatically generated hydrogen sulfide anion (HS(-)) regulates the metabolism and signaling actions of various electrophiles. HS(-) reacts with electrophiles, best represented by 8-nitro-cGMP, via direct sulfhydration and modulates cellular redox signaling. The relevance of this reaction is reinforced by the significant 8-nitro-cGMP formation in mouse cardiac tissue after myocardial infarction that is modulated by alterations in HS(-) biosynthesis. Cardiac HS(-), in turn, suppresses electrophile-mediated H-Ras activation and cardiac cell senescence, contributing to the beneficial effects of HS(-) on myocardial infarction-associated heart failure. Thus, this study reveals HS(-)-induced electrophile sulfhydration as a unique mechanism for regulating electrophile-mediated redox signaling.


Asunto(s)
Sulfuro de Hidrógeno/química , Sulfuro de Hidrógeno/metabolismo , Transducción de Señal/fisiología , Animales , Aniones , Línea Celular , Membrana Celular , GMP Cíclico/análogos & derivados , GMP Cíclico/química , GMP Cíclico/metabolismo , Regulación de la Expresión Génica , Genes ras/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Interferencia de ARN , Ratas
13.
Proc Natl Acad Sci U S A ; 108(16): 6662-7, 2011 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21464294

RESUMEN

Cross-talk between G protein-coupled receptor (GPCR) signaling pathways serves to fine tune cellular responsiveness by neurohumoral factors. Accumulating evidence has implicated nitric oxide (NO)-based signaling downstream of GPCRs, but the molecular details are unknown. Here, we show that adenosine triphosphate (ATP) decreases angiotensin type 1 receptor (AT(1)R) density through NO-mediated S-nitrosylation of nuclear factor κB (NF-κB) in rat cardiac fibroblasts. Stimulation of purinergic P2Y(2) receptor by ATP increased expression of inducible NO synthase (iNOS) through activation of nuclear factor of activated T cells, NFATc1 and NFATc3. The ATP-induced iNOS interacted with p65 subunit of NF-κB in the cytosol through flavin-binding domain, which was indispensable for the locally generated NO-mediated S-nitrosylation of p65 at Cys38. ß-Arrestins anchored the formation of p65/IκBα/ß-arrestins/iNOS quaternary complex. The S-nitrosylated p65 resulted in decreases in NF-κB transcriptional activity and AT(1)R density. In pressure-overloaded mouse hearts, ATP released from cardiomyocytes led to decrease in AT(1)R density through iNOS-mediated S-nitrosylation of p65. These results show a unique regulatory mechanism of heterologous regulation of GPCRs in which cysteine modification of transcriptional factor rather than protein phosphorylation plays essential roles.


Asunto(s)
Regulación hacia Abajo , Miocardio/metabolismo , Óxido Nítrico/metabolismo , Receptor de Angiotensina Tipo 1/biosíntesis , Receptores Purinérgicos P2Y2/metabolismo , Factor de Transcripción ReIA/metabolismo , Adenosina Trifosfato/farmacología , Animales , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Óxido Nítrico/genética , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Ratas , Receptor de Angiotensina Tipo 1/genética , Receptores Purinérgicos P2Y2/genética , Factor de Transcripción ReIA/genética
14.
J Biol Chem ; 287(42): 35669-35677, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22888001

RESUMEN

G-protein coupled receptors (GPCRs) have long been known as receptors that activate G protein-dependent cellular signaling pathways. In addition to the G protein-dependent pathways, recent reports have revealed that several ligands called "biased ligands" elicit G protein-independent and ß-arrestin-dependent signaling through GPCRs (biased agonism). Several ß-blockers are known as biased ligands. All ß-blockers inhibit the binding of agonists to the ß-adrenergic receptors. In addition to ß-blocking action, some ß-blockers are reported to induce cellular responses through G protein-independent and ß-arrestin-dependent signaling pathways. However, the physiological significance induced by the ß-arrestin-dependent pathway remains much to be clarified in vivo. Here, we demonstrate that metoprolol, a ß(1)-adrenergic receptor-selective blocker, could induce cardiac fibrosis through a G protein-independent and ß-arrestin2-dependent pathway. Metoprolol, a ß-blocker, increased the expression of fibrotic genes responsible for cardiac fibrosis in cardiomyocytes. Furthermore, metoprolol induced the interaction between ß(1)-adrenergic receptor and ß-arrestin2, but not ß-arrestin1. The interaction between ß(1)-adrenergic receptor and ß-arrestin2 by metoprolol was impaired in the G protein-coupled receptor kinase 5 (GRK5)-knockdown cells. Metoprolol-induced cardiac fibrosis led to cardiac dysfunction. However, the metoprolol-induced fibrosis and cardiac dysfunction were not evoked in ß-arrestin2- or GRK5-knock-out mice. Thus, metoprolol is a biased ligand that selectively activates a G protein-independent and GRK5/ß-arrestin2-dependent pathway, and induces cardiac fibrosis. This study demonstrates the physiological importance of biased agonism, and suggests that G protein-independent and ß-arrestin-dependent signaling is a reason for the diversity of the effectiveness of ß-blockers.


Asunto(s)
Antagonistas de Receptores Adrenérgicos beta 1/efectos adversos , Arrestinas/metabolismo , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Cardiopatías/inducido químicamente , Cardiopatías/metabolismo , Metoprolol/efectos adversos , Proteínas Musculares/metabolismo , Transducción de Señal/efectos de los fármacos , Antagonistas de Receptores Adrenérgicos beta 1/farmacología , Animales , Arrestinas/genética , Fibrosis , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Células HEK293 , Cardiopatías/genética , Cardiopatías/patología , Humanos , Metoprolol/farmacología , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , beta-Arrestinas
15.
J Pharmacol Sci ; 122(1): 1-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23575451

RESUMEN

ß-Arrestin-biased agonists are a new class of drugs with promising therapeutic effects. The molecular mechanisms of ß-arrestin-biased agonists are still not completely identified. Here, we investigated the effect of angiotensin II (AngII) and [Sar1,Ile4,Ile8] AngII (SII), a ß-arrestin-biased agonist, on ezrin-radixin-moesin (ERM) phosphorylation in NIH3T3 cells (a fibroblast cell line) stably expressing AngII type 1A receptor. ERM proteins are cross-linkers between the plasma membrane and the actin cytoskeleton and control a number of signaling pathways. We also investigated the role of Gαq protein and ß-arrestins in mediating ERM phosphorylation. We found that AngII stimulates ERM phosphorylation by acting as a ß-arrestin-biased agonist and AngII-stimulated ERM phosphorylation is mediated by ß-arrestin2 not ß-arrestin1. We also found that SII inhibits ERM phosphorylation by acting as a Gαq protein-biased agonist. We concluded that ERM phosphorylation is a unique ß-arrestin-biased agonism signal. Both AngII and SII can activate either Gαq protein or ß-arrestin-mediated signaling as functional biased agonists according to the type of the cell on which they act.


Asunto(s)
Angiotensina II/farmacología , Proteínas del Citoesqueleto/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Animales , Arrestinas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Ratones , Células 3T3 NIH , Fosforilación , Receptor de Angiotensina Tipo 1/agonistas , Transducción de Señal , beta-Arrestinas
16.
EMBO J ; 27(23): 3104-15, 2008 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-19008857

RESUMEN

Cardiac fibrosis, characterized by excessive deposition of extracellular matrix proteins, is one of the causes of heart failure, and it contributes to the impairment of cardiac function. Fibrosis of various tissues, including the heart, is believed to be regulated by the signalling pathway of angiotensin II (Ang II) and transforming growth factor (TGF)-beta. Transgenic expression of inhibitory polypeptides of the heterotrimeric G12 family G protein (Galpha(12/13)) in cardiomyocytes suppressed pressure overload-induced fibrosis without affecting hypertrophy. The expression of fibrogenic genes (TGF-beta, connective tissue growth factor, and periostin) and Ang-converting enzyme (ACE) was suppressed by the functional inhibition of Galpha(12/13). The expression of these fibrogenic genes through Galpha(12/13) by mechanical stretch was initiated by ATP and UDP released from cardiac myocytes through pannexin hemichannels. Inhibition of G-protein-coupled P2Y6 receptors suppressed the expression of ACE, fibrogenic genes, and cardiac fibrosis. These results indicate that activation of Galpha(12/13) in cardiomyocytes by the extracellular nucleotides-stimulated P2Y(6) receptor triggers fibrosis in pressure overload-induced cardiac fibrosis, which works as an upstream mediator of the signalling pathway between Ang II and TGF-beta.


Asunto(s)
Presión Sanguínea , Fibrosis , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Miocardio/patología , Miocitos Cardíacos/fisiología , Receptores Purinérgicos P2/fisiología , Transducción de Señal , Adenosina Trifosfato/metabolismo , Animales , Moléculas de Adhesión Celular/biosíntesis , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Peptidil-Dipeptidasa A/biosíntesis , Antagonistas del Receptor Purinérgico P2 , Ratas , Factor de Crecimiento Transformador beta/biosíntesis , Uridina Difosfato/metabolismo
17.
Circ Res ; 106(12): 1849-60, 2010 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-20448219

RESUMEN

RATIONALE: Atrial and brain natriuretic peptides (ANP and BNP, respectively) exert antihypertrophic effects in the heart via their common receptor, guanylyl cyclase (GC)-A, which catalyzes the synthesis of cGMP, leading to activation of protein kinase (PK)G. Still, much of the network of molecular mediators via which ANP/BNP-GC-A signaling inhibit cardiac hypertrophy remains to be characterized. OBJECTIVE: We investigated the effect of ANP-GC-A signaling on transient receptor potential subfamily C (TRPC)6, a receptor-operated Ca(2+) channel known to positively regulate prohypertrophic calcineurin-nuclear factor of activated T cells (NFAT) signaling. METHODS AND RESULTS: In cardiac myocytes, ANP induced phosphorylation of TRPC6 at threonine 69, the PKG phosphorylation site, and significantly inhibited agonist-evoked NFAT activation and Ca(2+) influx, whereas in HEK293 cells, it dramatically inhibited agonist-evoked TRPC6 channel activity. These inhibitory effects of ANP were abolished in the presence of specific PKG inhibitors or by substituting an alanine for threonine 69 in TRPC6. In model mice lacking GC-A, the calcineurin-NFAT pathway is constitutively activated, and BTP2, a selective TRPC channel blocker, significantly attenuated the cardiac hypertrophy otherwise seen. Conversely, overexpression of TRPC6 in mice lacking GC-A exacerbated cardiac hypertrophy. BTP2 also significantly inhibited angiotensin II-induced cardiac hypertrophy in mice. CONCLUSIONS: Collectively, these findings suggest that TRPC6 is a critical target of antihypertrophic effects elicited via the cardiac ANP/BNP-GC-A pathway and suggest TRPC6 blockade could be an effective therapeutic strategy for preventing pathological cardiac remodeling.


Asunto(s)
Factor Natriurético Atrial/metabolismo , Miocardio/patología , Péptido Natriurético Encefálico/metabolismo , Receptores del Factor Natriurético Atrial/metabolismo , Transducción de Señal/fisiología , Canales Catiónicos TRPC/antagonistas & inhibidores , Anilidas/farmacología , Animales , Canales de Calcio/metabolismo , Células Cultivadas , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Humanos , Hipertrofia/metabolismo , Hipertrofia/patología , Hipertrofia/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Factores de Transcripción NFATC/metabolismo , Técnicas de Placa-Clamp , Ratas , Receptores del Factor Natriurético Atrial/genética , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6 , Tiadiazoles/farmacología
18.
Arterioscler Thromb Vasc Biol ; 31(10): 2278-86, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21799177

RESUMEN

OBJECTIVE: The goal of this study was to determine whether inhibition of transient receptor potential canonical (TRPC) channels underlies attenuation of angiotensin II (Ang II)-induced vasoconstriction by phosphodiesterase (PDE) 3 inhibition. METHODS AND RESULTS: Pretreatment of rat thoracic aorta with cilostazol, a selective PDE3 inhibitor, suppressed vasoconstriction induced by Ang II but not that induced by KCl. The Ang II-induced contraction was largely dependent on Ca(2+) influx via receptor-operated cation channels. Cilostazol specifically suppressed diacylglycerol-activated TRPC channels (TRPC3/TRPC6/TRPC7) through protein kinase A (PKA)-dependent phosphorylation of TRPC channels in HEK293 cells. In contrast, we found that phosphorylation of TRPC6 at Thr69 was essential for the suppression of Ang II-induced Ca(2+) influx by PDE3 inhibition in rat aortic smooth muscle cells (RAoSMCs). Cilostazol specifically induced phosphorylation of endogenous TRPC6 at Thr69. The endogenous TRPC6, but not TRPC3, formed a ternary complex with PDE3 and PKA in RAoSMCs, suggesting the specificity of TRPC6 phosphorylation by PDE3 inhibition. Furthermore, inhibition of PDE3 suppressed the Ang II-induced contraction of reconstituted ring with RAoSMCs, which were abolished by the expression of a phosphorylation-deficient mutant of TRPC6. CONCLUSIONS: PKA-mediated phosphorylation of TRPC6 at Thr69 is essential for the vasorelaxant effects of PDE3 inhibition against the vasoconstrictive actions of Ang II.


Asunto(s)
Angiotensina II/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Inhibidores de Fosfodiesterasa 3/farmacología , Canales Catiónicos TRPC/efectos de los fármacos , Tetrazoles/farmacología , Vasoconstricción/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/enzimología , Señalización del Calcio/efectos de los fármacos , Cilostazol , Diglicéridos/metabolismo , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Masculino , Ratones , Músculo Liso Vascular/enzimología , Mutación , Miocitos del Músculo Liso/enzimología , Fosforilación , Procesamiento Proteico-Postraduccional , Ratas , Ratas Sprague-Dawley , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6 , Transfección , Vasoconstrictores/farmacología , Proteínas de Unión al GTP rho/metabolismo
19.
J Pharmacol Sci ; 118(4): 408-12, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22447307

RESUMEN

ß-Arrestins (ß-arrestin-1 and ß-arrestin-2) were first identified as proteins that have the ability to desensitize G protein-coupled receptors (GPCRs). However, it has recently been found that ß-arrestins can activate signaling pathways independent of G protein activation. The diversity of these signaling pathways has also been recognized. This leads to an appreciation of ß-arrestin-biased agonists, which is a new class of drugs that selectively activate ß-arrestin-mediated signaling without G protein activation. In this review, we will discuss the recent advance of ß-arrestin-mediated signaling pathways, including a brief account of different biased agonists, their pharmacological applications, and novel ß-arrestin research.


Asunto(s)
Arrestinas/fisiología , Transducción de Señal/fisiología , Animales , Arrestinas/agonistas , Carbazoles/farmacología , Carbazoles/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/fisiopatología , Carvedilol , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Piperazinas/farmacología , Piperazinas/uso terapéutico , Propanolaminas/farmacología , Propanolaminas/uso terapéutico , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , beta-Arrestina 1 , Arrestina beta 2 , beta-Arrestinas
20.
J Immunol ; 185(8): 4863-72, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20861350

RESUMEN

Lysophosphatidic acid (LPA) is a phospholipid mediator that exerts a variety of biological responses through specific G-protein-coupled receptors (LPA(1)-LPA(5) and P2Y5). LPA is thought to be involved in airway inflammation by regulating the expression of anti-inflammatory and proinflammatory genes. Chemokines such as CCL5/RANTES are secreted from airway epithelium and play a key role in allergic airway inflammation. CCL5/RANTES is a chemoattractant for eosinophils, T lymphocytes, and monocytes and seems to exacerbate asthma. We stimulated CCL5/RANTES production in a human bronchial epithelial cell line, BEAS-2B, with IFN-γ and TNF-α. When LPA was added, CCL5/RANTES mRNA expression and protein secretion were inhibited, despite the presence of IFN-γ and TNF-α. The LPA effect was attenuated by Ki16425, a LPA(1)/LPA(3) antagonist, but not by dioctylglycerol pyrophosphate 8:0, an LPA(3) antagonist. Pertussis toxin, the inhibitors for PI3K and Akt also attenuated the inhibitory effect of LPA on CCL5/RANTES secretion. We also identify the transcription factor IFN regulatory factor-1 (IRF-1) as being essential for CCL5/RANTES production. Interestingly, LPA inhibited IFN-γ and TNF-α-induced IRF-1 activation by blocking the binding of IRF-1 to its DNA consensus sequence without changing IRF-1 induction and its nuclear translocation. Ki16425, pertussis toxin, and PI3K inhibitors attenuated the inhibitory effect of LPA on IRF-1 activation. Our results suggest that LPA inhibits IFN-γ- and TNF-α-induced CCL5/RANTES production in BEAS-2B cells by blocking the binding of IRF-1 to the CCL5/RANTES promoter. LPA(1) coupled to G(i) and activation of PI3K is required for this unique effect.


Asunto(s)
Quimiocina CCL5/metabolismo , Regulación de la Expresión Génica/inmunología , Factor 1 Regulador del Interferón/metabolismo , Lisofosfolípidos/metabolismo , Mucosa Respiratoria/metabolismo , Western Blotting , Bronquios/inmunología , Bronquios/metabolismo , Línea Celular , Quimiocina CCL5/inmunología , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Expresión Génica , Humanos , Factor 1 Regulador del Interferón/inmunología , Lisofosfolípidos/inmunología , Regiones Promotoras Genéticas , Mucosa Respiratoria/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA