Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(7)2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38612493

RESUMO

Adrenergic pathways represent the main channel of communication between the nervous system and the immune system. During inflammation, blood monocytes migrate within tissue and differentiate into macrophages, which polarize to M1 or M2 macrophages with tissue-damaging or -reparative properties, respectively. This study investigates whether the ß-adrenergic receptor (ß-AR)-blocking drug propranolol modulates the monocyte-to-macrophage differentiation process and further influences macrophages in their polarization toward M1- and M2-like phenotypes. Six-day-human monocytes were cultured with M-CSF in the presence or absence of propranolol and then activated toward an M1 pro-inflammatory state or an M2 anti-inflammatory state. The chronic exposure of monocytes to propranolol during their differentiation into macrophages promoted the increase in the M1 marker CD16 and in the M2 markers CD206 and CD163 and peroxisome proliferator-activated receptor É£ expression. It also increased endocytosis and the release of IL-10, whereas it reduced physiological reactive oxygen species. Exposure to the pro-inflammatory conditions of propranolol-differentiated macrophages resulted in an anti-inflammatory promoting effect. At the molecular level, propranolol upregulated the expression of the oxidative stress regulators NRF2, heme oxygenase-1 and NQO1. By contributing to regulating macrophage activities, propranolol may represent a novel anti-inflammatory and immunomodulating compound with relevant therapeutic potential in several inflammatory diseases.


Assuntos
Monócitos , Propranolol , Humanos , Propranolol/farmacologia , Antioxidantes/farmacologia , Fator 2 Relacionado a NF-E2 , Macrófagos , Anti-Inflamatórios/farmacologia
2.
Eur J Pharmacol ; 948: 175700, 2023 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-37001579

RESUMO

Downregulation of cell surface ß-adrenergic receptors (ß-AR) is an important adaptive response that prevents deleterious effects of receptor overstimulation. Various factors including reactive oxygen species cause ß-AR downregulation. In this study, we evaluated the effects of ligands of the peripheral benzodiazepine receptor (PBR), a key protein in regulating oxidative stress, on surface density of endogenous ß1-and ß2-ARs in highly differentiated cells such as human monocytes, which express both ß-AR subtypes. ß-AR expression in human monocytes was evaluated by flow cytometry, qPCR and western blotting. Monocyte treatment with ß-AR agonist isoproterenol did not change surface ß1-AR density while downregulating surface ß2-AR density. This effect was antagonized by the ß-blocker propranolol. An opposite response was observed with benzodiazepine diazepam that led to a time-dependent reduction in ß1-AR density. In particular, while no significant downregulation was observed after 3 h of treatment, only 63% of ß1-ARs were still present on the cell surface after 48 h of treatment with diazepam at 1 µM. Treatment with the PBR antagonist PK11195, but not with propranolol, antagonized the effects of diazepam. No change in ß1-AR-mRNA or protein levels was observed at any time after diazepam treatment. We also found that diazepam did not affect Gs-protein or ß-arrestin-2 recruitment for both ß-ARs in engineered fibroblasts, further suggesting that diazepam activity on ß1-AR density is mediated by PBR. Finally, no sex-related differences were found. Collectively, these results indicate that monocyte ß1-ARs are resistant to catecholamine-mediated downregulation and suggest that PBR plays an important role in regulating ß1-AR density.


Assuntos
Monócitos , Propranolol , Humanos , Monócitos/metabolismo , Propranolol/farmacologia , Benzodiazepinas , Diazepam/farmacologia , Receptores Adrenérgicos beta 2/metabolismo , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 1/metabolismo
3.
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
4.
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
5.
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
6.
Eur J Pharmacol ; 882: 173287, 2020 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-32585157

RESUMO

Expression of the ß-myosin heavy chain (ß-MHC), a major component of the cardiac contractile apparatus, is tightly regulated as even modest increases can be detrimental to heart under stress. In healthy hearts, continuous inhibition of ß-adrenergic tone upregulates ß-MHC expression. However, it is unknown whether the duration of the ß-adrenergic inhibition and ß-MHC expression are related. Here, we evaluated the effects of intermittent ß-blockade on cardiac ß-MHC expression. To this end, the ß-blocker propranolol, at the dose of 15mg/kg, was administered once a day in mice for 14 days. This dosing schedule caused daily drug-free periods of at least 6 h as evidenced by propranolol plasma concentrations and cardiac ß-adrenergic responsiveness. Under these conditions, ß-MHC expression decreased by about 75% compared to controls. This effect was abolished in mice lacking ß1- but not ß2-adrenergic receptors (ß-AR) indicating that ß-MHC expression is regulated in a ß1-AR-dependent manner. In ß1-AR knockout mice, the baseline ß-MHC expression was fourfold higher than in wild-type mice. Also, we evaluated the impact of intermittent ß-blockade on ß-MHC expression in mice with systolic dysfunction, in which an increased ß-MHC expression occurs. At 3 weeks after myocardial infarction, mice showed systolic dysfunction and upregulation of ß-MHC expression. Intermittent ß-blockade decreased ß-MHC expression while attenuating cardiac dysfunction. In vitro studies showed that propranolol does not affect ß-MHC expression on its own but antagonizes catecholamine effects on ß-MHC expression. In conclusion, a direct relationship occurs between the duration of the ß-adrenergic inhibition and ß-MHC expression through the ß1-AR.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Miocárdio/metabolismo , Cadeias Pesadas de Miosina/genética , Propranolol/farmacologia , Receptores Adrenérgicos beta/genética , Miosinas Ventriculares/genética , Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/sangue , Antagonistas Adrenérgicos beta/farmacocinética , Antagonistas Adrenérgicos beta/uso terapêutico , Animais , Regulação para Baixo/efeitos dos fármacos , Feminino , Isoproterenol/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Propranolol/sangue , Propranolol/farmacocinética , Propranolol/uso terapêutico
7.
Int J Mol Sci ; 21(2)2020 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-31947522

RESUMO

Altered ß-adrenergic receptor (ß-AR) density has been reported in cells, animals, and humans receiving ß-blocker treatment. In some cases, ß-AR density is upregulated, but in others, it is unaffected or even reduced. Collectively, these results would imply that changes in ß-AR density and ß-blockade are not related. However, it has still not been clarified whether the effects of ß-blockers on receptor density are related to their ability to activate different ß-AR signaling pathways. To this aim, five clinically relevant ß-blockers endowed with inverse, partial or biased agonism at the ß2-AR were evaluated for their effects on ß2-AR density in both human embryonic kidney 293 (HEK293) cells expressing exogenous FLAG-tagged human ß2-ARs and human lymphocytes expressing endogenous ß2-ARs. Cell surface ß2-AR density was measured by enzyme-linked immunosorbent assay (ELISA) and flow cytometry. Treatment with propranolol, carvedilol, pindolol, sotalol, or timolol did not induce any significant change in surface ß2-AR density in both HEK293 cells and human lymphocytes. On the contrary, treatment with the ß-AR agonist isoproterenol reduced the number of cell surface ß2-ARs in the tested cell types without affecting ß2-AR-mRNA levels. Isoproterenol-induced effects on receptor density were completely antagonized by ß-blocker treatment. In conclusion, the agonistic activity of ß-blockers does not exert an important effect on short-term regulation of ß2-AR density.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Antagonistas de Receptores Adrenérgicos beta 2/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Imunofluorescência , Humanos , Especificidade de Órgãos
8.
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
9.
Cell Death Dis ; 9(2): 108, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29371598

RESUMO

Cardiovascular diseases (CVDs) are a major burden on the healthcare system: indeed, over two million new cases are diagnosed every year worldwide. Unfortunately, important drawbacks for the treatment of these patients derive from our current inability to stop the structural alterations that lead to heart failure, the common endpoint of many CVDs. In this scenario, a better understanding of the role of epigenetics - hereditable changes of chromatin that do not alter the DNA sequence itself - is warranted. To date, hyperacetylation of histones has been reported in hypertension and myocardial infarction, but the use of inhibitors for treating CVDs remains limited. Here, we studied the effect of the histone deacetylase inhibitor Givinostat on a mouse model of acute myocardial infarction. We found that it contributes to decrease endothelial-to-mesenchymal transition and inflammation, reducing cardiac fibrosis and improving heart performance and protecting the blood vessels from apoptosis through the modulatory effect of cardiac fibroblasts on endothelial cells. Therefore, Givinostat may have potential for the treatment of CVDs.


Assuntos
Carbamatos/farmacologia , Fibroblastos/patologia , Remodelação Ventricular/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Endotélio/efeitos dos fármacos , Endotélio/patologia , Epitélio/efeitos dos fármacos , Epitélio/patologia , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose , Regulação da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Inflamação/patologia , Camundongos Endogâmicos C57BL , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia
10.
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
11.
Exp Cell Res ; 339(2): 163-73, 2015 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-26524508

RESUMO

Clenbuterol (CLB) is a beta2-adrenergic agonist commonly used in asthma therapy, but is also a non-steroidal anabolic drug often abused in sport doping practices. Here we evaluated the in vitro impact of CLB on the physiology and function of human monocytes and dendritic cells (DCs), instrumental in the development of immune responses. We demonstrate that CLB inhibits the differentiation of monocytes into DCs and this effect is specific and dependent on ß2-adrenergic receptor (AR) activation. We found that CLB treatment reduced the percentage of CD1a(+) immature DCs, while increasing the frequency of monocytes retaining CD14 surface expression. Moreover, CLB inhibited tumor necrosis factor-alpha (TNF-alpha) enhanced IL-(interleukin)-10 and IL-6 production. In contrast, CLB did not modulate the phenotypic and functional properties of monocytes and DCs, such as the surface expression of HLA-DR, CD83, CD80 and CD86 molecules, cytokine production, immunostimulatory activity and phagocytic activity. Moreover, we found that CLB did not modulate the activation of NF-kB in DCs. Moreover, we found that the differentiation of monocytes into DCs was associated with a significant decrease of ß2-ARs mRNA expression. These results provide new insights on the effect of CLB on monocyte differentiation into DCs. Considering the frequent illegal use of CLB in doping, our work suggests that this drug is potentially harmful to immune responses decreasing the supply of DCs, thus subverting immune surveillance.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Diferenciação Celular/efeitos dos fármacos , Clembuterol/farmacologia , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Monócitos/citologia , Monócitos/efeitos dos fármacos , Diferenciação Celular/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Relação Dose-Resposta a Droga , Humanos , Monócitos/imunologia , Relação Estrutura-Atividade
12.
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
13.
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
14.
Blood Transfus ; 11(1): 88-93, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22790263

RESUMO

BACKGROUND: Chronic transfusion therapy causes a progressive iron overload that damages many organs including the heart. Recent evidence suggests that L-type calcium channels play an important role in iron uptake by cardiomyocytes under conditions of iron overload. Given that beta-adrenergic stimulation significantly enhances L-type calcium current, we hypothesised that beta-adrenergic blocking drugs could reduce the deleterious effects of iron overload on the heart. METHODS: Iron overload was generated by intraperitoneal injections of iron dextran (1g/kg) administered once a week for 8 weeks in male C57bl/6 mice, while propranolol was administered in drinking water at the dose of 40 mg/kg/day. Cardiac function and ventricular remodelling were evaluated by echocardiography and histological methods. RESULTS: As compared to placebo, iron injection caused cardiac iron deposition. Surprisingly, despite iron overload, myocardial function and ventricular geometry in the iron-treated mice resulted unchanged as compared to those in the placebo-treated mice. Administration of propranolol increased cardiac performance in iron-overloaded mice. Specifically, as compared to the values in the iron-overloaded group, in iron-overloaded animals treated with propranolol left ventricular fractional shortening increased (from 31.6% to 44.2%, P =0.01) whereas left ventricular end-diastolic diameter decreased (from 4.1 ± 0.1 mm to 3.5 ± 0.1 mm, P =0.03). Propranolol did not alter cardiac systolic function or left ventricular sizes in the placebo group. CONCLUSIONS: These results demonstrate that C57bl/6 mice are resistant to iron overload-induced myocardial injury and that treatment with propranolol is able to increase cardiac performance in iron-overloaded mice. However, since C57bl/6 mice were resistant to iron-induced injury, it remains to be evaluated further whether propranolol could prevent iron-overload cardiomyopathy.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Canais de Cálcio Tipo L/metabolismo , Resistência à Doença/efeitos dos fármacos , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/fisiopatologia , Ferro/metabolismo , Miocárdio/metabolismo , Propranolol/metabolismo , Animais , Canais de Cálcio Tipo L/genética , Modelos Animais de Doenças , Resistência à Doença/genética , Hematínicos/efeitos adversos , Hematínicos/farmacologia , Sobrecarga de Ferro/induzido quimicamente , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/patologia , Complexo Ferro-Dextran/efeitos adversos , Complexo Ferro-Dextran/farmacologia , Masculino , Camundongos , Miocárdio/patologia , Especificidade da Espécie
15.
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
16.
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
17.
Clin Cancer Res ; 13(2 Pt 1): 644-53, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17255288

RESUMO

PURPOSE: Immunotherapy is a promising antitumor strategy, which can be successfully combined with current anticancer treatments, as suggested by recent studies showing the paradoxical chemotherapy-induced enhancement of the immune response. The purpose of the present work is to dissect the biological events induced by chemotherapy that cooperate with immunotherapy in the success of the combined treatment against cancer. In particular, we focused on the following: (a) cyclophosphamide-induced modulation of several cytokines, (b) homeostatic proliferation of adoptively transferred lymphocytes, and (c) homing of transferred lymphocytes to secondary lymphoid organs and tumor mass. EXPERIMENTAL DESIGN: Here, we used the adoptive transfer of tumor-immune cells after cyclophosphamide treatment of tumor-bearing mice as a model to elucidate the mechanisms by which cyclophosphamide can render the immune lymphocytes competent to induce tumor rejection. RESULTS: The transfer of antitumor immunity was found to be dependent on CD4(+) T cells and on the cooperation of adoptively transferred cells with the host immune system. Of note, tumor-immune lymphocytes migrated specifically to the tumor only in mice pretreated with cyclophosphamide. Cyclophosphamide treatment also promoted homeostatic proliferation/activation of transferred B and T lymphocytes. Optimal therapeutic responses to the transfer of immune cells were associated with the cyclophosphamide-mediated induction of a "cytokine storm" [including granulocyte macrophage colony-stimulating factor, interleukin (IL)-1beta, IL-7, IL-15, IL-2, IL-21, and IFN-gamma], occurring during the "rebound phase" after drug-induced lymphodepletion. CONCLUSIONS: The ensemble of these data provides a new rationale for combining immunotherapy and chemotherapy to induce an effective antitumor response in cancer patients.


Assuntos
Antineoplásicos/farmacologia , Linfócitos B/metabolismo , Ciclofosfamida/farmacologia , Citocinas/metabolismo , Linfócitos T/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Sistema Imunitário , Imunoterapia/métodos , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Transplante de Neoplasias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA