Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.494
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Eur J Pharmacol ; 971: 176488, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38458410

RESUMO

OBJECTIVE: Pathological cardiac remodelling, including cardiac hypertrophy and fibrosis, is a key pathological process in the development of heart failure. However, effective therapeutic approaches are limited. The ß-adrenergic receptors are pivotal signalling molecules in regulating cardiac function. G-alpha interacting protein (GAIP)-interacting protein, C-terminus 1 (GIPC1) is a multifunctional scaffold protein that directly binds to the C-terminus of ß1-adrenergic receptor (ß1-adrenergic receptor). However, little is known about its roles in heart function. Therefore, we investigated the role of GIPC1 in cardiac remodelling and its underlying molecular mechanisms. METHODS: Pathological cardiac remodelling in mice was established via intraperitoneal injection of isoprenaline for 14 d or transverse aortic constriction surgery for 8 weeks. Myh6-driving cardiomyocyte-specific GIPC1 conditional knockout (GIPC1 cKO) mice and adeno-associated virus 9 (AAV9)-mediated GIPC1 overexpression mice were used. The effect of GIPC1 on cardiac remodelling was assessed using echocardiographic, histological, and biochemical analyses. RESULTS: GIPC1 expression was consistently reduced in the cardiac remodelling model. GIPC1 cKO mice exhibited spontaneous abnormalities, including cardiac hypertrophy, fibrosis, and systolic dysfunction. In contrast, AAV9-mediated GIPC1 overexpression in the heart attenuated isoproterenol-induced pathological cardiac remodelling in mice. Mechanistically, GIPC1 interacted with the ß1-adrenergic receptor and stabilised its expression by preventing its ubiquitination and degradation, maintaining the balance of ß1-adrenergic receptor/ß2-adrenergic receptor, and inhibiting hyperactivation of the mitogen-activated protein kinase signalling pathway. CONCLUSIONS: These results suggested that GIPC1 plays a cardioprotective role and is a promising therapeutic target for the treatment of cardiac remodelling and heart failure.


Assuntos
Insuficiência Cardíaca , Remodelação Ventricular , Animais , Camundongos , Cardiomegalia/patologia , Fibrose , Insuficiência Cardíaca/patologia , Isoproterenol/efeitos adversos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos , Receptores Adrenérgicos beta/metabolismo
2.
Acta Pharmacol Sin ; 45(3): 531-544, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37919475

RESUMO

Cardiac inflammation contributes to heart failure (HF) induced by isoproterenol (ISO) through activating ß-adrenergic receptors (ß-AR). Recent evidence shows that myeloid differentiation factor 2 (MD2), a key protein in endotoxin-induced inflammation, mediates inflammatory heart diseases. In this study, we investigated the role of MD2 in ISO-ß-AR-induced heart injuries and HF. Mice were infused with ISO (30 mg·kg-1·d-1) via osmotic mini-pumps for 2 weeks. We showed that MD2 in cardiomyocytes and cardiac macrophages was significantly increased and activated in the heart tissues of ISO-challenged mice. Either MD2 knockout or administration of MD2 inhibitor L6H21 (10 mg/kg every 2 days, i.g.) could prevent mouse hearts from ISO-induced inflammation, remodelling and dysfunction. Bone marrow transplantation study revealed that both cardiomyocyte MD2 and bone marrow-derived macrophage MD2 contributed to ISO-induced cardiac inflammation and injuries. In ISO-treated H9c2 cardiomyocyte-like cells, neonatal rat primary cardiomyocytes and primary mouse peritoneal macrophages, MD2 knockout or pre-treatment with L6H21 (10 µM) alleviated ISO-induced inflammatory responses, and the conditioned medium from ISO-challenged macrophages promoted the hypertrophy and fibrosis in cardiomyocytes and fibroblasts. We demonstrated that ISO induced MD2 activation in cardiomyocytes via ß1-AR-cAMP-PKA-ROS signalling axis, and induced inflammatory responses in macrophages via ß2-AR-cAMP-PKA-ROS axis. This study identifies MD2 as a key inflammatory mediator and a promising therapeutic target for ISO-induced heart failure.


Assuntos
Insuficiência Cardíaca , Miócitos Cardíacos , Ratos , Camundongos , Animais , Miócitos Cardíacos/metabolismo , Isoproterenol/toxicidade , Receptores Adrenérgicos beta/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Macrófagos/metabolismo
3.
Circ Res ; 133(12): 1040-1055, 2023 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-37961889

RESUMO

BACKGROUND: Nitric oxide (NO) has been identified as a signaling molecule generated during ß-adrenergic receptor stimulation in the heart. Furthermore, a role for NO in triggering spontaneous Ca2+ release via S-nitrosylation of CaMKIIδ (Ca2+/calmodulin kinase II delta) is emerging. NO donors are routinely used clinically for their cardioprotective effects on the heart, but it is unknown how NO donors modulate the proarrhythmic CaMKII to alter cardiac arrhythmia incidence. We test the role of S-nitrosylation of CaMKIIδ at the Cysteine-273 inhibitory site and cysteine-290 activating site in cardiac Ca2+ handling and arrhythmogenesis before and during ß-adrenergic receptor stimulation. METHODS: We measured Ca2+-handling in isolated cardiomyocytes from C57BL/6J wild-type (WT) mice and mice lacking CaMKIIδ expression (CaMKIIδ-KO) or with deletion of the S-nitrosylation site on CaMKIIδ at cysteine-273 or cysteine-290 (CaMKIIδ-C273S and -C290A knock-in mice). Cardiomyocytes were exposed to NO donors, S-nitrosoglutathione (GSNO; 150 µM), sodium nitroprusside (200 µM), and ß-adrenergic agonist isoproterenol (100 nmol/L). RESULTS: Both WT and CaMKIIδ-KO cardiomyocytes responded to isoproterenol with a full inotropic and lusitropic Ca2+ transient response as well as increased Ca2+ spark frequency. However, the increase in Ca2+ spark frequency was significantly attenuated in CaMKIIδ-KO cardiomyocytes. The protection from isoproterenol-induced Ca2+ sparks and waves was mimicked by GSNO pretreatment in WT cardiomyocytes but lost in CaMKIIδ-C273S cardiomyocytes. When GSNO was applied after isoproterenol, this protection was not observed in WT or CaMKIIδ-C273S but was apparent in CaMKIIδ-C290A. In Langendorff-perfused isolated hearts, GSNO pretreatment limited isoproterenol-induced arrhythmias in WT but not CaMKIIδ-C273S hearts, while GSNO exposure after isoproterenol sustained or exacerbated arrhythmic events. CONCLUSIONS: We conclude that prior S-nitrosylation of CaMKIIδ at cysteine-273 can limit subsequent ß-adrenergic receptor-induced arrhythmias, but that S-nitrosylation at cysteine-290 might worsen or sustain ß-adrenergic receptor-induced arrhythmias. This has important implications for the administration of NO donors in the clinical setting.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Óxido Nítrico , Camundongos , Animais , Isoproterenol/farmacologia , Óxido Nítrico/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cisteína/metabolismo , Camundongos Endogâmicos C57BL , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/metabolismo , Miócitos Cardíacos/metabolismo , Fosforilação , Receptores Adrenérgicos beta/metabolismo , Cálcio/metabolismo , Retículo Sarcoplasmático/metabolismo
4.
Mol Metab ; 74: 101753, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37321371

RESUMO

OBJECTIVE: Norepinephrine stimulates the adipose tissue thermogenic program through a ß-adrenergic receptor (ßAR)-cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) signaling cascade. We discovered that a noncanonical activation of the mechanistic target of rapamycin complex 1 (mTORC1) by PKA is required for the ßAR-stimulation of adipose tissue browning. However, the downstream events triggered by PKA-phosphorylated mTORC1 activation that drive this thermogenic response are not well understood. METHODS: We used a proteomic approach of Stable Isotope Labeling by/with Amino acids in Cell culture (SILAC) to characterize the global protein phosphorylation profile in brown adipocytes treated with the ßAR agonist. We identified salt-inducible kinase 3 (SIK3) as a candidate mTORC1 substrate and further tested the effect of SIK3 deficiency or SIK inhibition on the thermogenic gene expression program in brown adipocytes and in mouse adipose tissue. RESULTS: SIK3 interacts with RAPTOR, the defining component of the mTORC1 complex, and is phosphorylated at Ser884 in a rapamycin-sensitive manner. Pharmacological SIK inhibition by a pan-SIK inhibitor (HG-9-91-01) in brown adipocytes increases basal Ucp1 gene expression and restores its expression upon blockade of either mTORC1 or PKA. Short-hairpin RNA (shRNA) knockdown of Sik3 augments, while overexpression of SIK3 suppresses, Ucp1 gene expression in brown adipocytes. The regulatory PKA phosphorylation domain of SIK3 is essential for its inhibition. CRISPR-mediated Sik3 deletion in brown adipocytes increases type IIa histone deacetylase (HDAC) activity and enhances the expression of genes involved in thermogenesis such as Ucp1, Pgc1α, and mitochondrial OXPHOS complex protein. We further show that HDAC4 interacts with PGC1α after ßAR stimulation and reduces lysine acetylation in PGC1α. Finally, a SIK inhibitor well-tolerated in vivo (YKL-05-099) can stimulate the expression of thermogenesis-related genes and browning of mouse subcutaneous adipose tissue. CONCLUSIONS: Taken together, our data reveal that SIK3, with the possible contribution of other SIKs, functions as a phosphorylation switch for ß-adrenergic activation to drive the adipose tissue thermogenic program and indicates that more work to understand the role of the SIKs is warranted. Our findings also suggest that maneuvers targeting SIKs could be beneficial for obesity and related cardiometabolic disease.


Assuntos
Tecido Adiposo , Proteômica , Camundongos , Animais , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Tecido Adiposo/metabolismo , Adipócitos Marrons/metabolismo , Receptores Adrenérgicos beta/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Termogênese , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo
5.
Cell Rep ; 42(5): 112433, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37099421

RESUMO

Lipolysis of stored triglycerides is stimulated via ß-adrenergic receptor (ß-AR)/3',5'-cyclic adenosine monophosphate (cAMP) signaling and inhibited via phosphodiesterases (PDEs). In type 2 diabetes, a dysregulation in the storage/lipolysis of triglycerides leads to lipotoxicity. Here, we hypothesize that white adipocytes regulate their lipolytic responses via the formation of subcellular cAMP microdomains. To test this, we investigate real-time cAMP/PDE dynamics at the single-cell level in human white adipocytes with a highly sensitive florescent biosensor and uncover the presence of several receptor-associated cAMP microdomains where cAMP signals are compartmentalized to differentially regulate lipolysis. In insulin resistance, we also detect cAMP microdomain dysregulation mechanisms that promote lipotoxicity, but regulation can be restored by the anti-diabetic drug metformin. Therefore, we present a powerful live-cell imaging technique capable of resolving disease-driven alterations in cAMP/PDE signaling at the subcellular level and provide evidence to support the therapeutic potential of targeting these microdomains.


Assuntos
Diabetes Mellitus Tipo 2 , Lipólise , Humanos , Lipólise/fisiologia , Adipócitos Brancos/metabolismo , AMP Cíclico/metabolismo , Receptores Adrenérgicos beta/metabolismo
6.
Cell Signal ; 107: 110664, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37004833

RESUMO

Stimulating ß-adrenergic receptors (ß-AR) culminates in pathological hypertrophy - a condition underlying multiple cardiovascular diseases (CVDs). The ensuing signal transduction network appears to involve mutually communicating phosphorylation-cascades and redox signaling modules, although the regulators of redox signaling processes remain largely unknown. We previously showed that H2S-induced Glucose-6-phosphate dehydrogenase (G6PD) activity is critical for suppressing cardiac hypertrophy in response to adrenergic stimulation. Here, we extended our findings and identified novel H2S-dependent pathways constraining ß-AR-induced pathological hypertrophy. We demonstrated that H2S regulated early redox signal transduction processes - including suppression of cue-dependent production of reactive oxygen species (ROS) and oxidation of cysteine thiols (R-SOH) on critical signaling intermediates (including AKT1/2/3 & ERK1/2). Consistently, the maintenance of intracellular levels of H2S dampened the transcriptional signature associated with pathological hypertrophy upon ß-AR-stimulation, as demonstrated by RNA-seq analysis. We further prove that H2S remodels cell metabolism by promoting G6PD activity to enforce changes in the redox state that favor physiological cardiomyocyte growth over pathological hypertrophy. Thus, our data suggest that G6PD is an effector of H2S-mediated suppression of pathological hypertrophy and that the accumulation of ROS in the G6PD-deficient background can drive maladaptive remodeling. Our study reveals an adaptive role for H2S relevant to basic and translational studies. Identifying adaptive signaling mediators of the ß-AR-induced hypertrophy may reveal new therapeutic targets and routes for CVD therapy optimization.


Assuntos
Glucosefosfato Desidrogenase , Sulfeto de Hidrogênio , Receptores Adrenérgicos beta , Humanos , Cardiomegalia/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais
7.
Int J Mol Sci ; 24(5)2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-36902129

RESUMO

ß-adrenoreceptors (ARs) are members of the superfamily of G-protein-coupled receptors (GPCRs), and are activated by catecholamines, such as epinephrine and norepinephrine. Three subtypes of ß-ARs (ß1, ß2, and ß3) have been identified with different distributions among ocular tissues. Importantly, ß-ARs are an established target in the treatment of glaucoma. Moreover, ß-adrenergic signaling has been associated with the development and progression of various tumor types. Hence, ß-ARs are a potential therapeutic target for ocular neoplasms, such as ocular hemangioma and uveal melanoma. This review aims to discuss the expression and function of individual ß-AR subtypes in ocular structures, as well as their role in the treatment of ocular diseases, including ocular tumors.


Assuntos
Oftalmopatias , Melanoma , Humanos , Receptores Adrenérgicos beta/metabolismo , Melanoma/metabolismo , Norepinefrina , Epinefrina/uso terapêutico , Receptores Adrenérgicos beta 2/metabolismo
8.
Int J Mol Sci ; 24(6)2023 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-36982930

RESUMO

In the inflamed uterus, the production and secretion of prostaglandins (PGs) and noradrenergic innervation pattern are changed. Receptor-based control of prostaglandin E2 (PGE2) production and secretion by noradrenaline during uterine inflammation is unknown. The aim of this study was to determine the role of α1-, α2- and ß-adrenoreceptors (ARs) in noradrenaline-influenced PG-endoperoxidase synthase-2 (PTGS-2) and microsomal PTGE synthase-1 (mPTGES-1) protein levels in the inflamed pig endometrium, and in the secretion of PGE2 from this tissue. E. coli suspension (E. coli group) or saline (CON group) was injected into the uterine horns. Eight days later, severe acute endometritis developed in the E. coli group. Endometrial explants were incubated with noradrenaline and/or α1-, α2- and ß-AR antagonists. In the CON group, noradrenaline did not significantly change PTGS-2 and mPTGES-1 protein expression and increased PGE2 secretion compared to the control values (untreated tissue). In the E. coli group, both enzyme expression and PGE2 release were stimulated by noradrenaline, and these values were higher versus the CON group. The antagonists of α1- and α2-AR isoforms and ß-AR subtypes do not significantly alter the noradrenaline effect on PTGS-2 and mPTGES-1 protein levels in the CON group, compared to noradrenaline action alone. In this group, α1A-, α2B- and ß2-AR antagonists partly eliminated noradrenaline-stimulated PGE2 release. Compared to the noradrenaline effect alone, α1A-, α1B-, α2A-, α2B-, ß1-, ß2- and ß3-AR antagonists together with noradrenaline reduced PTGS-2 protein expression in the E. coli group. Such effects were also exerted in this group by α1A-, α1D-, α2A-, ß2- and ß3-AR antagonists with noradrenaline on mPTGES-1 protein levels. In the E. coli group, the antagonists of all isoforms of α1-ARs and subtypes of ß-ARs as well as α2A-ARs together with noradrenaline decreased PGE2 secretion versus noradrenaline action alone. Summarizing, in the inflamed pig endometrium, α1(A, B)-, α2(A, B)- and ß(1, 2, 3)-ARs mediate the noradrenaline stimulatory effect on PTGE-2 protein expression, while noradrenaline via α1(A, D)-, α2A- and ß(2, 3)-ARs increases mPTGES-1 protein expression and α1(A, B, D)-, α2A- and ß(1, 2, 3)-ARs are involved in PGE2 release. Data suggest that noradrenaline may indirectly affect the processes regulated by PGE2 by influencing its production. Pharmacological modulation of particular AR isoforms/subtypes can be used to change PGE2 synthesis/secretion to alleviate inflammation and improve uterine function.


Assuntos
Dinoprostona , Norepinefrina , Feminino , Suínos , Animais , Norepinefrina/farmacologia , Norepinefrina/metabolismo , Escherichia coli/metabolismo , Receptores Adrenérgicos beta/metabolismo , Receptores Adrenérgicos alfa 1 , Endométrio/metabolismo
9.
Circ Res ; 132(7): 867-881, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-36884028

RESUMO

BACKGROUND: Loss of brain-derived neurotrophic factor (BDNF)/TrkB (tropomyosin kinase receptor B) signaling accounts for brain and cardiac disorders. In neurons, ß-adrenergic receptor stimulation enhances local BDNF expression. It is unclear if this occurs in a pathophysiological relevant manner in the heart, especially in the ß-adrenergic receptor-desensitized postischemic myocardium. Nor is it fully understood whether and how TrkB agonists counter chronic postischemic left ventricle (LV) decompensation, a significant unmet clinical milestone. METHODS: We conducted in vitro studies using neonatal rat and adult murine cardiomyocytes, SH-SY5Y neuronal cells, and umbilical vein endothelial cells. We assessed myocardial ischemia (MI) impact in wild type, ß3AR knockout, or myocyte-selective BDNF knockout (myoBDNF KO) mice in vivo (via coronary ligation [MI]) or in isolated hearts with global ischemia-reperfusion (I/R). RESULTS: In wild type hearts, BDNF levels rose early after MI (<24 hours), plummeting at 4 weeks when LV dysfunction, adrenergic denervation, and impaired angiogenesis ensued. The TrkB agonist, LM22A-4, countered all these adverse effects. Compared with wild type, isolated myoBDNF KO hearts displayed worse infarct size/LV dysfunction after I/R injury and modest benefits from LM22A-4. In vitro, LM22A-4 promoted neurite outgrowth and neovascularization, boosting myocyte function, effects reproduced by 7,8-dihydroxyflavone, a chemically unrelated TrkB agonist. Superfusing myocytes with the ß3AR-agonist, BRL-37344, increased myocyte BDNF content, while ß3AR signaling underscored BDNF generation/protection in post-MI hearts. Accordingly, the ß1AR blocker, metoprolol, via upregulated ß3ARs, improved chronic post-MI LV dysfunction, enriching the myocardium with BDNF. Last, BRL-37344-imparted benefits were nearly abolished in isolated I/R injured myoBDNF KO hearts. CONCLUSIONS: BDNF loss underscores chronic postischemic heart failure. TrkB agonists can improve ischemic LV dysfunction via replenished myocardial BDNF content. Direct cardiac ß3AR stimulation, or ß-blockers (via upregulated ß3AR), is another BDNF-based means to fend off chronic postischemic heart failure.


Assuntos
Insuficiência Cardíaca , Isquemia Miocárdica , Neuroblastoma , Disfunção Ventricular Esquerda , Ratos , Camundongos , Humanos , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Endoteliais/metabolismo , Neuroblastoma/metabolismo , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Isquemia Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Disfunção Ventricular Esquerda/metabolismo , Receptores Adrenérgicos beta/metabolismo
10.
Int J Mol Sci ; 24(4)2023 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-36834875

RESUMO

Abnormalities in the peripheral immune system are involved in the pathophysiology of fibromyalgia, although their contribution to the painful symptoms remains unknown. Our previous study reported the ability of splenocytes to develop pain-like behavior and an association between the central nervous system (CNS) and splenocytes. Since the spleen is directly innervated by sympathetic nerves, this study aimed to examine whether adrenergic receptors are necessary for pain development or maintenance using an acid saline-induced generalized pain (AcGP) model (an experimental model of fibromyalgia) and whether the activation of these receptors is also essential for pain reproduction by the adoptive transfer of AcGP splenocytes. The administration of selective ß2-blockers, including one with only peripheral action, prevented the development but did not reverse the maintenance of pain-like behavior in acid saline-treated C57BL/6J mice. Neither a selective α1-blocker nor an anticholinergic drug affects the development of pain-like behavior. Furthermore, ß2-blockade in donor AcGP mice eliminated pain reproduction in recipient mice injected with AcGP splenocytes. These results suggest that peripheral ß2-adrenergic receptors play an important role in the efferent pathway from the CNS to splenocytes in pain development.


Assuntos
Fibromialgia , Receptores Adrenérgicos beta 2 , Camundongos , Animais , Receptores Adrenérgicos beta 2/metabolismo , Fibromialgia/metabolismo , Baço/metabolismo , Camundongos Endogâmicos C57BL , Receptores Adrenérgicos/metabolismo , Dor/metabolismo , Sistema Nervoso Central/metabolismo , Sistema Nervoso Simpático/metabolismo , Receptores Adrenérgicos beta/metabolismo , Antagonistas Adrenérgicos beta/farmacologia
11.
Handb Exp Pharmacol ; 279: 59-82, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36592229

RESUMO

Calcium influx through voltage-gated calcium channels, Cav1.2, in cardiomyocytes initiates excitation-contraction coupling in the heart. The force and rate of cardiac contraction are modulated by the sympathetic nervous system, mediated substantially by changes in intracellular calcium. Norepinephrine released from sympathetic neurons innervating the heart and epinephrine secreted by the adrenal chromaffin cells bind to ß-adrenergic receptors on the sarcolemma of cardiomyocytes initiating a signaling cascade that generates cAMP and activates protein kinase A, the targets of which control calcium influx. For decades, the mechanisms by which PKA regulated calcium channels in the heart were not known. Recently, these mechanisms have been elucidated. In this chapter, we will review the history of the field and the studies that led to the identification of the evolutionarily conserved process.


Assuntos
Canais de Cálcio , Cálcio , Humanos , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Miócitos Cardíacos/metabolismo , Sistema Nervoso Simpático/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Canais de Cálcio Tipo L/metabolismo , Fosforilação
12.
Eur J Med Chem ; 246: 114961, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36495629

RESUMO

Biomedical applications of molecules that are able to modulate ß-adrenergic signaling have become increasingly attractive over the last decade, revealing that ß-adrenergic receptors (ß-ARs) are key targets for a plethora of therapeutic interventions, including cancer. Despite successes in ß-AR drug discovery, identification of ß-AR ligands that are useful as selective chemical tools in pharmacological studies of the three ß-AR subtypes, or lead compounds for drug development is still a highly challenging task. This is mainly due to the intrinsic plasticity of ß-ARs as G protein-coupled receptors in conjunction with the requirement for functional receptor subtype selectivity, tissue specificity and minimal off-target effects. With the aim to provide insight into structure-activity relationships for the three ß-AR subtypes, we have synthesized and obtained the pharmacological profile of a series of structurally diverse compounds (named MC) that were designed based on the aryloxy-propanolamine scaffold of SR59230A. Comparative analysis of their predicted binding mode within the active and inactive states of the receptors in combination with their pharmacological profile revealed key structural elements that control their activity as agonists or antagonists, in addition to clues about substituents that mediate selectivity for one receptor subtype over the others. We anticipate that these results will facilitate selective ß-AR drug development efforts.


Assuntos
Receptores Adrenérgicos beta , Receptores Acoplados a Proteínas G , Humanos , Receptores Adrenérgicos beta/química , Receptores Adrenérgicos beta/metabolismo , Ligantes , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade
13.
Bull Exp Biol Med ; 173(3): 330-334, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35852683

RESUMO

We studied the role of adrenoceptors in the regulation of activity of mitochondrial and cytoplasmic enzymes in cardiomyocytes by catecholamines and their metabolites. Different types of adrenergic receptors (AR) agonists acting either on both α- and ß-AR or selectively on α- or ß-AR, as well as quinoid metabolites of catecholamines were used. It was found that the activating effect of ß-AR agonist isadrin (isoproterenol) on succinate dehydrogenase of the mitochondria in the heart is prevented by ß-adrenergic blockade. The activating effect of dopamine, epinephrine, and isoproterenol on cytochrome C-oxidase and the inhibitory effect of dopamine, norepinephrine, epinephrine, and isoproterenol on Mg-activated ATPase was not mediated by adrenoreceptors. Hormones of the sympathoadrenal system epinephrine, dopamine, norepinephrine, isoproterenol, and catecholamine metabolites (adrenochrome and adrenoxyl) modulating activity of the respiratory chain enzymes of mitochondria in the heart regulate the processes of tissue respiration by transferring mitochondria into a state of "loose" phosphorylation and respiration coupling. Epinephrine as a ß-AR agonist increased activity of cytosolic enzymes catalyzing metabolism of purine nucleotides (adenosine deaminase and AMP deaminase), enzymes of antioxidant defense (glutathione peroxidase and catalase), and the level of malondialdehyde and diene conjugates. ß-AR blockade with metoprolol abolished the activating effect of epinephrine on glutathione reductase, glutathione peroxidase, and catalase and reduced the level of malondialdehyde and diene conjugates.


Assuntos
Catecolaminas , Dopamina , Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Catalase/metabolismo , Catecolaminas/metabolismo , Dopamina/metabolismo , Epinefrina/metabolismo , Epinefrina/farmacologia , Glutationa Peroxidase/metabolismo , Isoproterenol/farmacologia , Malondialdeído/metabolismo , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Norepinefrina/metabolismo , Norepinefrina/farmacologia , Receptores Adrenérgicos/metabolismo , Receptores Adrenérgicos beta/metabolismo
14.
EBioMedicine ; 82: 104149, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35816899

RESUMO

BACKGROUND: Medulloblastoma is the most frequent brain malignancy of childhood. The current multimodal treatment comes at the expense of serious and often long-lasting side effects. Drug repurposing is a strategy to fast-track anti-cancer therapy with low toxicity. Here, we showed the ability of ß-blockers to potentiate radiotherapy in medulloblastoma with bad prognosis. METHODS: Medulloblastoma cell lines, patient-derived xenograft cells, 3D spheroids and an innovative cerebellar organotypic model were used to identify synergistic interactions between ß-blockers and ionising radiations. Gene expression profiles of ß-adrenergic receptors were analysed in medulloblastoma samples from 240 patients. Signaling pathways were explored by RT-qPCR, RNA interference, western blotting and RNA sequencing. Medulloblastoma cell bioenergetics were evaluated by measuring the oxygen consumption rate, the extracellular acidification rate and superoxide production. FINDINGS: Low concentrations of ß-blockers significantly potentiated clinically relevant radiation protocols. Although patient biopsies showed detectable expression of ß-adrenergic receptors, the ability of the repurposed drugs to potentiate ionising radiations did not result from the inhibition of the canonical signaling pathway. We highlighted that the efficacy of the combinatorial treatment relied on a metabolic catastrophe that deprives medulloblastoma cells of their adaptive bioenergetics capacities. This led to an overproduction of superoxide radicals and ultimately to an increase in ionising radiations-mediated DNA damages. INTERPRETATION: These data provide the evidence of the efficacy of ß-blockers as potentiators of radiotherapy in medulloblastoma, which may help improve the treatment and quality of life of children with high-risk brain tumours. FUNDING: This study was funded by institutional grants and charities.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Criança , Metabolismo Energético , Humanos , Meduloblastoma/tratamento farmacológico , Meduloblastoma/genética , Meduloblastoma/radioterapia , Qualidade de Vida , Receptores Adrenérgicos beta/metabolismo , Receptores Adrenérgicos beta/uso terapêutico , Superóxidos
15.
Proc Natl Acad Sci U S A ; 119(26): e2205626119, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35737830

RESUMO

ß-adrenergic receptor (ß-AR) signaling plays predominant roles in modulating energy expenditure by triggering lipolysis and thermogenesis in adipose tissue, thereby conferring obesity resistance. Obesity is associated with diminished ß3-adrenergic receptor (ß3-AR) expression and decreased ß-adrenergic responses, but the molecular mechanism coupling nutrient overload to catecholamine resistance remains poorly defined. Ten-eleven translocation (TET) proteins are dioxygenases that alter the methylation status of DNA by oxidizing 5-methylcytosine to 5-hydroxymethylcytosine and further oxidized derivatives. Here, we show that TET proteins are pivotal epigenetic suppressors of ß3-AR expression in adipocytes, thereby attenuating the responsiveness to ß-adrenergic stimulation. Deletion of all three Tet genes in adipocytes led to increased ß3-AR expression and thereby enhanced the downstream ß-adrenergic responses, including lipolysis, thermogenic gene induction, oxidative metabolism, and fat browning in vitro and in vivo. In mouse adipose tissues, Tet expression was elevated after mice ate a high-fat diet. Mice with adipose-specific ablation of all TET proteins maintained higher levels of ß3-AR in both white and brown adipose tissues and remained sensitive to ß-AR stimuli under high-fat diet challenge, leading to augmented energy expenditure and decreased fat accumulation. Consequently, they exhibited improved cold tolerance and were substantially protected from diet-induced obesity, inflammation, and metabolic complications, including insulin resistance and hyperlipidemia. Mechanistically, TET proteins directly repressed ß3-AR transcription, mainly in an enzymatic activity-independent manner, and involved the recruitment of histone deacetylases to increase deacetylation of its promoter. Thus, the TET-histone deacetylase-ß3-AR axis could be targeted to treat obesity and related metabolic diseases.


Assuntos
Epigênese Genética , Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas , Tecido Adiposo Marrom/metabolismo , Animais , Regulação da Expressão Gênica/genética , Camundongos , Obesidade/genética , Obesidade/metabolismo , Proteínas Proto-Oncogênicas/genética , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo , Receptores Adrenérgicos beta 3/genética , Receptores Adrenérgicos beta 3/metabolismo , Termogênese/genética
16.
Int J Mol Sci ; 23(9)2022 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-35563024

RESUMO

Adrenergic receptors (AR) belong to the G protein-coupled receptor superfamily and regulate migration and proliferation in various cell types. The objective of this study was to evaluate whether ß-AR stimulation affects the antiproliferative action of α2-AR agonists on B16F10 cells and, if so, to determine the relative contribution of ß-AR subtypes. Using pharmacological approaches, evaluation of Ki-67 expression by flow cytometry and luciferase-based cAMP assay, we found that treatment with isoproterenol, a ß-AR agonist, increased cAMP levels in B16F10 melanoma cells without affecting cell proliferation. Propranolol inhibited the cAMP response to isoproterenol. In addition, stimulation of α2-ARs with agonists such as clonidine, a well-known antihypertensive drug, decreased cancer cell proliferation. This effect on cell proliferation was suppressed by treatment with isoproterenol. In turn, the suppressive effects of isoproterenol were abolished by the treatment with either ICI 118,551, a ß2-AR antagonist, or propranolol, suggesting that isoproterenol effects are mainly mediated by the ß2-AR stimulation. We conclude that the crosstalk between the ß2-AR and α2-AR signaling pathways regulates the proliferative activity of B16F10 cells and may therefore represent a therapeutic target for melanoma therapy.


Assuntos
Melanoma , Receptores Adrenérgicos alfa 2 , Receptores Adrenérgicos beta 2 , Agonistas Adrenérgicos beta/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Isoproterenol/farmacologia , Isoproterenol/uso terapêutico , Melanoma/metabolismo , Propranolol/farmacologia , Propranolol/uso terapêutico , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos beta/metabolismo , Receptores Adrenérgicos beta 1 , Receptores Adrenérgicos beta 2/metabolismo
17.
Bull Exp Biol Med ; 172(6): 671-675, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35501645

RESUMO

Cardiac-specific microRNA miR-133a-3p modulates adrenergic signaling. Adrenergic receptors and their intracellular pathways are the key players in proarrhythmic ectopy derived from the myocardial sleeves of the pulmonary veins. We studied the effect of miR-133a-3p on ectopy induced by norepinephrine in myocardial tissue of rat pulmonary veins. Using microelectrode technique, we revealed facilitation of proarrhythmic pattern of spontaneous bursts of action potentials induced by norepinephrine in tissue preparations of the pulmonary veins isolated from rats in 24 h after injection of a transfection mixture containing miR-133a-3p (1 mg/kg) in vivo. According to ELISA data, the cAMP level in the pulmonary vein myocardium of rats receiving miR-133a-3p was 2-fold higher than in control animals. Bioinformatic analysis showed that mRNA of protein phosphatases and some phosphodiesterases are most probable targets of miR-133a-3p. The proarrhythmic effect of miR-133a-3p can be related to inhibition of the expression of phosphodiesterases accompanied by cAMP accumulation and increased intracellular ß-adrenergic signaling.


Assuntos
AMP Cíclico , MicroRNAs , Miocárdio , Veias Pulmonares , Animais , AMP Cíclico/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Miocárdio/metabolismo , Norepinefrina/farmacologia , Diester Fosfórico Hidrolases/metabolismo , Veias Pulmonares/metabolismo , Ratos , Receptores Adrenérgicos beta/metabolismo
18.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35210363

RESUMO

Cancer-associated cachexia (CAC) is a hypermetabolic syndrome characterized by unintended weight loss due to the atrophy of adipose tissue and skeletal muscle. A phenotypic switch from white to beige adipocytes, a phenomenon called browning, accelerates CAC by increasing the dissipation of energy as heat. Addressing the mechanisms of white adipose tissue (WAT) browning in CAC, we now show that cachexigenic tumors activate type 2 immunity in cachectic WAT, generating a neuroprotective environment that increases peripheral sympathetic activity. Increased sympathetic activation, in turn, results in increased neuronal catecholamine synthesis and secretion, ß-adrenergic activation of adipocytes, and induction of WAT browning. Two genetic mouse models validated this progression of events. 1) Interleukin-4 receptor deficiency impeded the alternative activation of macrophages, reduced sympathetic activity, and restrained WAT browning, and 2) reduced catecholamine synthesis in peripheral dopamine ß-hydroxylase (DBH)-deficient mice prevented cancer-induced WAT browning and adipose atrophy. Targeting the intraadipose macrophage-sympathetic neuron cross-talk represents a promising therapeutic approach to ameliorate cachexia in cancer patients.


Assuntos
Tecido Adiposo Marrom/patologia , Caquexia/patologia , Comunicação Celular , Neoplasias/complicações , Neurônios/patologia , Sistema Nervoso Simpático/patologia , Animais , Caquexia/etiologia , Caquexia/metabolismo , Expressão Gênica , Xenoenxertos , Humanos , Camundongos , Neoplasias/metabolismo , Receptores Adrenérgicos beta/metabolismo , Termogênese
19.
J Cardiovasc Pharmacol ; 80(3): 328-333, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35099166

RESUMO

ABSTRACT: ß 1 -adrenergic receptors (ß 1 ARs) are the principle mediators of catecholamine actions in cardiomyocytes. ß 1 ARs rapidly adjust cardiac output and provide short-term hemodynamic support for the failing heart by activating a Gs-adenylyl cyclase pathway that increases 3'-5'-cyclic adenosine monophosphate and leads to the activation of protein kinase A and the phosphorylation of substrates involved in excitation-contraction coupling. However, chronic persistent ß 1 AR activation in the setting of heart failure leads to a spectrum of maladaptive changes that contribute to the evolution of heart failure. The molecular basis for ß 1 AR-driven maladaptive responses remains uncertain because chronic persistent ß 1 AR activation has been linked to the activation of both proapoptotic and antiapoptotic signaling pathways. Of note, studies to date have been predicated on the assumption that ß 1 ARs signal exclusively as full-length receptor proteins. Our recent studies show that ß 1 ARs are detected as both full-length and N-terminally truncated species in cardiomyocytes, that N-terminal cleavage is regulated by O-glycan modifications at specific sites on the ß 1 AR N-terminus, and that N-terminally truncated ß 1 ARs remain signaling competent, but their signaling properties differ from those of the full-length ß 1 AR. The N-terminally truncated form of the ß 1 AR constitutively activates the protein kinase B signaling pathway and confers protection against doxorubicin-dependent apoptosis in cardiomyocytes. These studies identify a novel signaling paradigm for the ß 1 AR, implicating the N-terminus as a heretofore-unrecognized structural determinant of ß 1 AR responsiveness that could be pharmacologically targeted for therapeutic advantage.


Assuntos
Insuficiência Cardíaca , Miócitos Cardíacos , Adenilil Ciclases/metabolismo , AMP Cíclico/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Humanos , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais/fisiologia
20.
Med Res Rev ; 42(3): 1179-1201, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34967048

RESUMO

The role of the ß-adrenoceptors (ß-ARs) in hypoxia-driven diseases has gained visibility after the demonstration that propranolol promotes the regression of infantile hemangiomas and ameliorates the signs of retinopathy of prematurity (ROP). Besides the role of ß2-ARs, preclinical studies in ROP have also revealed that ß3-ARs are upregulated by hypoxia and that they are possibly involved in retinal angiogenesis. In a sort of figurative round trip, peculiarities typical of ROP, where hypoxia drives retinal neovascularization, have been then translated to cancer, a disease equally characterized by hypoxia-driven angiogenesis. In this step, investigating the role of ß3-ARs has taken advantage of the assumption that cancer growth uses a set of strategies in common with embryo development. The possibility that hypoxic induction of ß3-ARs may represent one of the mechanisms through which primarily embryo (and then cancer, as an astute imitator) adapts to grow in an otherwise hostile environment, has grown evidence. In both cancer and embryo, ß3-ARs exert similar functions by exploiting a metabolic shift known as the Warburg effect, by acquiring resistance against xenobiotics, and by inducing a local immune tolerance. An additional potential role of ß3-AR as a marker of stemness has been suggested by the finding that its antagonism induces cancer cell differentiation evoking that ß3-ARs may help cancer to grow in a nonhospital environment, a strategy also exploited by embryos. From cancer, the round trip goes back to neonatal diseases for which new possible interpretative keys and potential pharmacological perspectives have been suggested.


Assuntos
Doenças do Recém-Nascido , Neoplasias , Receptores Adrenérgicos/metabolismo , Humanos , Recém-Nascido , Propranolol/farmacologia , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA