Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
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
2.
Br J Pharmacol ; 179(7): 1371-1383, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34766341

RESUMO

BACKGROUND AND PURPOSE: Recently, ß-adrenoceptor blockade has emerged as a potential strategy to inhibit melanoma growth. It remains to be ascertained whether ß-adrenoceptor stimulation by circulating catecholamines increases melanoma growth in mice. EXPERIMENTAL APPROACH: B16F10 melanoma-bearing mice were used to evaluate effects of adrenaline and specific adrenoceptor (AR) ligands on tumour volume. AR expression and effects of AR ligands on cell viability, production of mitochondrial reactive oxygen species (mROS), and proliferation activity in B16F10 cells, were determined by biochemical analyses. KEY RESULTS: Real-time polymerase chain reaction (qPCR) analyses revealed that B16F10 cells express α1B-, α2A-, α2B- and ß2-ARs. We found that treatment with the α- and ß-AR agonist adrenaline or with the synthetic catecholamine isoprenaline, which selectively stimulates ß-ARs, did not affect melanoma growth. Conversely, adrenaline reduced tumour growth in mice cotreated with propranolol, a ß1ß2-AR antagonist. Adrenaline had no effect in tumour-bearing ß1ß2-AR knockout mice, in which ß1- and ß2-ARs are lacking, but it reduced tumour growth when co-administered with propranolol suggesting that tumour ß2-ARs negatively regulate adrenaline antitumour activity. Additionally, we found that α1-AR stimulation with cirazoline yielded a decrease in B16F10 melanoma size. These effects on melanoma growth were paralleled by reduced cell viability and proliferation activity as well as increased mROS production in α1-AR-stimulated B16F10 cells. Decreased viability, proliferation and mitochondrial function in B16F10 cells also occurred after α2-AR stimulation by α2-AR agonist ST91. CONCLUSIONS AND IMPLICATIONS: In the B16F10 melanoma model, stimulation of α-AR subtypes yields in vivo and in vitro anticancer activity.


Assuntos
Melanoma , Receptores Adrenérgicos alfa 1 , Animais , Catecolaminas , Epinefrina/farmacologia , Ligantes , Melanoma/metabolismo , Camundongos , Camundongos Knockout , Propranolol/farmacologia , Receptores Adrenérgicos alfa 1/metabolismo
3.
Int J Mol Sci ; 22(16)2021 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-34445662

RESUMO

Acute myocardial infarction (MI) is associated with an intense inflammatory response that is critical for cardiac repair but is also involved in the pathogenesis of adverse cardiac remodeling, i.e., the set of size, geometry, and structure changes that represent the structural substrate for the development of post-MI heart failure. Deciphering the pathophysiological mechanisms underlying cardiac repair after MI is, therefore, critical to favorably regulate cardiac wound repair and to prevent development of heart failure. Catecholamines and estrogen play an active role in regulating the inflammatory response in the infarcted area. For example, stress-induced catecholamines alter recruitment and trafficking of leukocytes to the heart. Additionally, estrogen affects rate of cardiac rupture during the acute phase of MI, as well as infarct size and survival in animal models of MI. In this review, we will summarize the role of ß-adrenergic receptors and estrogen in cardiac repair after infarction in preclinical studies.


Assuntos
Estrogênios/metabolismo , Insuficiência Cardíaca/prevenção & controle , Infarto do Miocárdio/complicações , Receptores Adrenérgicos beta/metabolismo , Remodelação Ventricular , Animais , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Humanos
4.
J Cell Physiol ; 234(11): 20453-20468, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30982981

RESUMO

Cardiac dysfunction is often observed in patients with cancer also representing a serious problem limiting chemotherapeutic intervention and even patient survival. In view of the recently established role of the immune system in the control of cancer growth, the present work has been undertaken to investigate the effects of a panel of the most important inflammatory cytokines on the integrity and function of mitochondria, as well as of the cytoskeleton, two key elements in the functioning of cardiomyocytes. Either mitochondria features or actomyosin cytoskeleton organization of in vitro-cultured cardiomyocytes treated with different inflammatory cytokines were analyzed. In addition, to investigate the interplay between tumor growth and cardiac function in an in vivo system, immunocompetent female mice were inoculated with cancer cells and treated with the chemotherapeutic drug doxorubicin at a dosing schedule able to suppress tumor growth without inducing cardiac alterations. Analyses carried out in cardiomyocytes treated with the inflammatory cytokines, such as tumor necrosis factor α (TNF-α), interferon γ (IFN-γ), interleukin 6 (IL-6), IL-8, and IL-1ß revealed severe phenotypic changes, for example, of contractile cytoskeletal elements, mitochondrial membrane potential, mitochondrial reactive oxygen species production and mitochondria network organization. Accordingly, in immunocompetent mice, the tumor growth was accompanied by increased levels of the inflammatory cytokines TNF-α, IFN-γ, IL-6, and IL-8, either in serum or in the heart tissue, together with a significant reduction of ventricular systolic function. The alterations of mitochondria and of microfilament system of cardiomyocytes, due to the systemic inflammation associated with cancer growth, could be responsible for remote cardiac injury and impairment of systolic function observed in vivo.


Assuntos
Citocinas/farmacologia , Citoesqueleto/efeitos dos fármacos , Inflamação/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Animais , Células Cultivadas , Feminino , Interferon gama/farmacologia , Interleucina-6/farmacologia , Camundongos Endogâmicos BALB C , Neoplasias/tratamento farmacológico , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
5.
Br J Pharmacol ; 174(2): 139-149, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27792834

RESUMO

BACKGROUND AND PURPOSE: Propranolol is a vasoactive drug that shows antiangiogenic and antitumour activities in melanoma. However, it is unknown whether these activities are dose-dependent and whether there is a relationship between systemic vascular effects of propranolol and anti-melanoma activity. EXPERIMENTAL APPROACH: Effects of increasing doses of propranolol (10, 20, 30 and 40 mg·kg-1 ·day-1 ) on tumour growth were studied in B16F10 melanoma-bearing mice. Histological and biochemical analyses were used to assess propranolol effects on angiogenesis and cancer cell proliferation. Systemic vascular resistance (SVR) was evaluated by measuring cardiac output and arterial BP. KEY RESULTS: In vitro analyses revealed that B16F10 cells expressed ß-adrenoceptors, but neither isoprenaline, a ß-adrenoceptor agonist, nor the ß-blocker propranolol affected cancer cell proliferation. In vivo studies showed that the antitumour efficacy of propranolol follows a U-shaped biphasic dose-response curve. Low doses (10 and 20 mg·kg-1 ·day-1 ) significantly inhibit tumour growth, whereas higher doses are progressively less effective. We also found that high-dose propranolol stimulates tumour arteriogenesis whereas no effect on angiogenesis was observed at any dose. Based on these data and considering that propranolol is a vasoactive drug, we hypothesized that it causes systemic vasoconstriction or vasodilation depending on the dose and thus alters tumour perfusion and growth. Consistent with this hypothesis, we found that propranolol has a biphasic effect on SVR with low and high doses producing vasoconstriction and vasodilation respectively. CONCLUSIONS AND IMPLICATIONS: Propranolol inhibits melanoma growth in a U-shaped biphasic manner. A direct relationship exists between SVR and anti-melanoma activity.


Assuntos
Antineoplásicos/farmacologia , Melanoma/tratamento farmacológico , Melanoma/patologia , Propranolol/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Masculino , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Relação Estrutura-Atividade , Células Tumorais Cultivadas
6.
Blood Transfus ; 12(4): 485-90, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24960657

RESUMO

BACKGROUND: Iron homeostasis in humans is tightly regulated by mechanisms aimed to conserve iron for reutilisation, with a negligible role played by excretory mechanisms. In a previous study we found that mice have an astonishing ability to tolerate very high doses of parenterally administered iron dextran. Whether this ability is linked to the existence of an excretory pathway remains to be ascertained. MATERIALS AND METHODS: Iron overload was generated by intraperitoneal injections of iron dextran (1 g/kg) administered once a week for 8 weeks in two different mouse strains (C57bl/6 and B6D2F1). Urinary and faecal iron excretion was assessed by inductively coupling plasma-mass spectrometry, whereas cardiac and liver architecture was evaluated by echocardiography and histological methods. For both strains, 24-hour faeces and urine samples were collected and iron concentration was determined on days 0, 1 and 2 after iron administration. RESULTS: In iron-overloaded C57bl/6 mice, the faecal iron concentration increased by 218% and 157% on days 1 and 2, respectively (p<0.01). The iron excreted represented a loss of 14% of total iron administered. Similar but smaller changes was also found in B6D2F1 mice. Conversely, we found no significant changes in the concentration of iron in the urine in either of the strains of mice. In both strains, histological examination showed accumulation of iron in the liver and heart which tended to decrease over time. CONCLUSIONS: This study indicates that mice have a mechanism for removal of excess body iron and provides insights into the possible mechanisms of excretion.


Assuntos
Hematínicos/efeitos adversos , Sobrecarga de Ferro/induzido quimicamente , Sobrecarga de Ferro/urina , Complexo Ferro-Dextran/efeitos adversos , Ferro/urina , Animais , Fezes , Hematínicos/farmacologia , Humanos , Sobrecarga de Ferro/fisiopatologia , Complexo Ferro-Dextran/farmacologia , Masculino , Camundongos
7.
J Cardiovasc Pharmacol ; 64(1): 21-7, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24621648

RESUMO

Recent results indicate that the reduction of ß-adrenergic signaling impairs angiogenesis under ischemic conditions. Because angiogenesis may occur in the absence of ischemia, it remains to be determined whether and how ß-adrenergic signaling regulates angiogenesis, which develops under normoxic conditions. The effect of ß-adrenergic ligands on angiogenesis was investigated using 3-dimensional cultures of mouse aortic rings embedded in collagen type I, in which luminized microvessels develop in response to vascular endothelial growth factor (VEGF). Under normoxic conditions, both isoproterenol, a ß-adrenergic receptor (ß-AR) agonist, and forskolin, an adenylate cyclase activator, were unable to influence aortic microvessel sprouting. On the contrary, treatment with propranolol, a ß-AR antagonist, caused an approximately 70% increase in VEGF-mediated microvessel sprouting. This effect was abolished in rings from both double ß-AR and ß1-AR knockout mice, but not in rings from ß2-AR knockout mice. Significant increases in microvessel sprouting were also observed when mouse aortic rings from C57BL/6 mice were treated with the ß1-AR-selective antagonists metoprolol and bisoprolol or with the ß2-AR-selective antagonist ICI 118,551. Conversely, carvedilol, a nonselective ß-AR antagonist, was unable to affect aortic sprouting. These findings suggest that some ß-blockers display proangiogenic activity through a mechanism that is independent of their ability to antagonize catecholamine action. The present results also identify a new function for ß-AR signaling as a facilitator for VEGF-mediated angiogenesis and have implications for understanding the mechanisms that regulate angiogenic responses under normoxic conditions.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Aorta Torácica/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Agonistas Adrenérgicos beta/farmacologia , Animais , Aorta Torácica/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Adrenérgicos beta/efeitos dos fármacos , Receptores Adrenérgicos beta/metabolismo , Receptores Adrenérgicos beta 2/genética , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Ann Ist Super Sanita ; 47(3): 290-5, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21952155

RESUMO

The ß-blocker propranolol displays antihypertrophic and antifibrotic properties in the heart subjected to pressure overload. Yet the underlying mechanisms responsible for these important effects remain to be completely understood. The purpose of this study was to determine signaling pathway-focused gene expression profile associated with the antihypertrophic action of propranolol in pressure overloaded hearts. To address this question, a focused real-time PCR array was used to screen left ventricular RNA expression of 84 gene transcripts representative of 18 different signaling pathways in C57BL/6 mice subjected to transverse aortic constriction (TAC) or sham surgery. On the surgery day, mice received either propranolol (80 mg/kg/day) or vehicle for 14 days. TAC caused a 49% increase in the left ventricular weight-to-body weight (LVW/BW) ratio without changing gene expression. Propranolol blunted LVW/BW ratio increase by approximately 50% while causing about a 3-fold increase in the expression of two genes, namely Brca1 and Cdkn2a, belonging to the TGF-beta and estrogen pathways, respectively. In conclusion, after 2 weeks of pressure overload, TAC hearts show a gene expression profile superimposable to that of sham hearts. Conversely, propranolol treatment is associated with an increased expression of genes which negatively regulate cell cycle progression. It remains to be established whether a mechanistic link between gene expression changes and the antihypertrophic action of propranolol occurs.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Coração/fisiopatologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Estresse Fisiológico/genética , Estresse Fisiológico/fisiologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Aorta/patologia , Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/patologia , Estenose da Valva Aórtica/fisiopatologia , Cardiomegalia/etiologia , Cardiomegalia/prevenção & controle , Ecocardiografia , Hemodinâmica/efeitos dos fármacos , Hemodinâmica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pressão , Propranolol/farmacologia , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real
9.
Br J Pharmacol ; 164(8): 1917-28, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21615725

RESUMO

BACKGROUND AND PURPOSE: Cell cycle regulators are regarded as essential for cardiomyocyte hypertrophic growth. Given that the ß-adrenoceptor antagonist propranolol blunts cardiomyocyte hypertrophic growth, we determined whether propranolol alters the expression of cell cycle-related genes in mouse hearts subjected to pressure overload. EXPERIMENTAL APPROACH: Pressure overload was induced by transverse aortic constriction (TAC), whereas the expression levels of 84 cell cycle-related genes were assayed by real-time PCR. Propranolol (80 mg·kg(-1) ·day(-1) ) was administered in drinking water for 14 days. KEY RESULTS: Two weeks after surgery, TAC caused a 46% increase in the left ventricular weight-to-body weight (LVW/BW) ratio but no significant changes in cell cycle gene expression. Propranolol, at plasma concentrations ranging from 10 to 140 ng·mL(-1) , blunted the LVW/BW ratio increase in TAC mice, while significantly increasing expression of 10 cell cycle genes including mitotic cyclins and proliferative markers such as Ki67. This increase in cell cycle gene expression was paralleled by a significant increase in the number of Ki67-positive non-cardiomyocyte cells as revealed by immunohistochemistry and confocal microscopy. ß-Adrenoceptor signalling was critical for cell cycle gene expression changes, as genetic deletion of ß-adrenoceptors also caused a significant increase in cyclins and Ki67 in pressure overloaded hearts. Finally, we found that metoprolol, a ß(1) -adrenoceptor antagonist, failed to enhance cell cycle gene expression in TAC mice. CONCLUSIONS AND IMPLICATIONS: Propranolol treatment enhances cell cycle-related gene expression in pressure overloaded hearts by increasing the number of cycling non-cardiomyocyte cells. These changes seem to occur via ß(2) -adrenoceptor-mediated mechanisms.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Cardiomiopatias/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Genes cdc , Propranolol/farmacologia , Animais , Cardiomiopatias/diagnóstico por imagem , Modelos Animais de Doenças , Ecocardiografia , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Ensaio Radioligante , Reação em Cadeia da Polimerase em Tempo Real , Receptores Adrenérgicos beta/genética
10.
J Mol Cell Cardiol ; 45(6): 761-9, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18851973

RESUMO

The treatment with beta-blockers causes an enhancement of the norepinephrine-induced fetal gene response in cultured cardiomyocytes. Here, we tested whether the activation of cAMP-mediated beta-adrenergic signaling antagonizes alpha(1)-adrenergic receptor (AR)-mediated fetal gene response. To address this question, the fetal gene program, of which atrial natriuretic peptide (ANP) and the beta-isoform of myosin heavy chain are classical members, was induced by phenylephrine (PE), an alpha(1)-AR agonist. In cultured neonatal rat cardiomyocytes, we found that stimulation of beta-ARs with isoproterenol, a beta-AR agonist, inhibited the fetal gene expression induced by PE. Similar results were also observed when cardiomyocytes were treated with forskolin (FSK), a direct activator of adenylyl cyclase, or 8-CPT-6-Phe-cAMP, a selective activator of protein kinase A (PKA). Conversely, the PE-induced fetal gene expression was further upregulated by H89, a selective PKA inhibitor. To evaluate whether these results could be generalized to Gq-mediated signaling and not specifically to alpha(1)-ARs, cardiomyocytes were treated with prostaglandin F(2)alpha, another Gq-coupled receptor agonist, which is able to promote fetal gene expression. This treatment caused an increase of both ANP mRNA and protein levels, which was almost completely abolished by FSK treatment. The capability of beta-adrenergic signaling to regulate the fetal gene expression was also evaluated in vivo conditions by using beta1- and beta2-AR double knockout mice, in which the predominant cardiac beta-AR subtypes are lacking, or by administering isoproterenol (ISO), a beta-AR agonist, at a subpressor dose. A significant increase of the fetal gene expression was found in beta(1)- and beta(2)-AR gene deficient mice. Conversely, we found that ANP, beta-MHC and skACT mRNA levels were significantly decreased in ISO-treated hearts. Collectively, these data indicate that cAMP-mediated beta-adrenergic signaling negatively regulates Gq cascade activation-induced fetal gene expression in cultured cardiomyocytes and that this inhibitory regulation is already operative in the mouse heart under physiological conditions.


Assuntos
Fator Natriurético Atrial/biossíntese , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , AMP Cíclico/metabolismo , Feto/metabolismo , Regulação da Expressão Gênica/fisiologia , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais/fisiologia , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Fator Natriurético Atrial/genética , Colforsina/farmacologia , AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dinoprosta/farmacologia , Ativadores de Enzimas/farmacologia , Feto/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Isoproterenol/farmacologia , Isoquinolinas/farmacologia , Camundongos , Miócitos Cardíacos/citologia , Cadeias Pesadas de Miosina/biossíntese , Cadeias Pesadas de Miosina/genética , Miosina não Muscular Tipo IIB/biossíntese , Miosina não Muscular Tipo IIB/genética , Fenilefrina/farmacologia , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta/genética , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA