Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(9)2022 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-35563201

RESUMO

Several contributions of circulating microvesicles (MVs) to the endothelial dysfunction have been reported in the past; a head-to-head comparison of platelet- and monocyte-derived MVs has however never been performed. To this aim, we assessed the involvement of these MVs in vessel damage related processes, i.e., oxidative stress, inflammation, and leukocyte-endothelial adhesion. Platelets and monocytes isolated from healthy subjects (HS, n = 15) were stimulated with TRAP-6 and LPS to release MVs that were added to human vascular endothelial cell (hECV) culture to evaluate superoxide anion production, inflammatory markers (IL-6, TNFα, NF-κB mRNA expression), and hECV adhesiveness. The effects of the MVs-induced from HS were compared to those induced by MVs spontaneously released from cells of patients with ST-segment elevation myocardial infarction (STEMI, n = 7). MVs released by HS-activated cells triggered a threefold increase in oxidative burst in a concentration-dependent manner. Only MVs released from monocytes doubled IL-6, TNFα, and NF-κB mRNA expression and monocyte-endothelial adhesion. Interestingly, the effects of the MVs isolated from STEMI-monocytes were not superimposable to previous ones except for adhesion to hECV. Conversely, MVs released from STEMI-platelets sustained both redox state and inflammatory phenotype. These data provide evidence that MVs released from activated and/or pathologic platelets and monocytes differently affect endothelial behavior, highlighting platelet-MVs as causative factors of impaired endothelial function in the acute phase of STEMI.


Assuntos
Micropartículas Derivadas de Células , Infarto do Miocárdio com Supradesnível do Segmento ST , Plaquetas/metabolismo , Micropartículas Derivadas de Células/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Monócitos , NF-kappa B/metabolismo , RNA Mensageiro/metabolismo , Infarto do Miocárdio com Supradesnível do Segmento ST/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
2.
Pharmacol Res ; 107: 308-314, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27045818

RESUMO

Circulating human monocytes, a functionally and phenotypically heterogeneous population, are emerging as fundamental cell types in rheumatoid arthritis (RA). The aim of this pilot study was to assess the correlation, if any, among anti-rheumatic drug therapy, circulating CD14(+)CD16(+) monocytes and validated clinical scales (e.g., DAS28 score and ultrasonography US7 score) of disease severity in RA. Thirty consecutive RA patients, either naïve or under disease-modifying anti-rheumatic drugs (DMARDs) or biological therapy, and 10 age-matched healthy volunteers, were enrolled. Monocytes were prepared from heparinized blood samples; surface expression of CD14 and CD16 was determined by flow cytometry. RA patients presented a significantly higher percentage of CD14(+)CD16(+) monocytes, as compared to healthy subjects. There was a good correlation between DAS28 clinical score and the ultrasound composite score US7 (r=0.66), as well as between both scores and the percentage of CD14(+)CD16(+) monocytes (r=0.43 and 0.47, respectively). Naïve RA patients had the highest expression (19.2±3.2%) of CD14(+)CD16(+) monocytes and elevated DAS28 score; patients on DMARDs presented a 7-fold increased expression of CD14(+)CD16(+) monocytes, relatively to healthy volunteers (2.1±1.4%), and an intermediate disease severity. The RA patients treated with biological therapy had a low percentage of CD14(+)CD16(+) monocytes (5.1±3.6%; p<0.01 vs naïve and DMARDs groups), similar to the one detected in healthy controls, and reduced US7 and DAS28 scores. Interestingly, for the same DAS28 score, monocytes isolated from RA patients on biological therapy had a lower CD16 expression than patients on DMARDs. Therefore, CD14(+)CD16(+) circulating blood monocytes may represent an appropriate biomarker to assess RA disease activity along with DAS28 and US7 scores. Together, these three parameters may represent a better indicator for evaluating therapy efficacy.


Assuntos
Antirreumáticos/farmacologia , Artrite Reumatoide/imunologia , Receptores de Lipopolissacarídeos/imunologia , Monócitos/efeitos dos fármacos , Receptores de IgG/imunologia , Adulto , Anticorpos/farmacologia , Anticorpos/uso terapêutico , Antirreumáticos/uso terapêutico , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/metabolismo , Biomarcadores/metabolismo , Feminino , Proteínas Ligadas por GPI/imunologia , Humanos , Masculino , Metotrexato/farmacologia , Metotrexato/uso terapêutico , Pessoa de Meia-Idade , Monócitos/imunologia , Projetos Piloto , Prednisona/farmacologia , Prednisona/uso terapêutico , Índice de Gravidade de Doença
3.
Haematologica ; 100(7): 881-92, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25911555

RESUMO

A large fraction of factor VIII in blood originates from liver sinusoidal endothelial cells although extrahepatic sources also contribute to plasma factor VIII levels. Identification of cell-types other than endothelial cells with the capacity to synthesize and release factor VIII will be helpful for therapeutic approaches in hemophilia A. Recent cell therapy and bone marrow transplantation studies indicated that Küpffer cells, monocytes and mesenchymal stromal cells could synthesize factor VIII in sufficient amount to ameliorate the bleeding phenotype in hemophilic mice. To further establish the role of blood cells in expressing factor VIII, we studied various types of mouse and human hematopoietic cells. We identified factor VIII in cells isolated from peripheral and cord blood, as well as bone marrow. Co-staining for cell type-specific markers verified that factor VIII was expressed in monocytes, macrophages and megakaryocytes. We additionally verified that factor VIII was expressed in liver sinusoidal endothelial cells and endothelial cells elsewhere, e.g., in the spleen, lungs and kidneys. Factor VIII was well expressed in sinusoidal endothelial cells and Küpffer cells isolated from human liver, whereas by comparison isolated human hepatocytes expressed factor VIII at very low levels. After transplantation of CD34(+) human cord blood cells into NOD/SCIDγNull-hemophilia A mice, fluorescence activated cell sorting of peripheral blood showed >40% donor cells engrafted in the majority of mice. In these animals, plasma factor VIII activity 12 weeks after cell transplantation was up to 5% and nine of 12 mice survived after a tail clip-assay. In conclusion, hematopoietic cells, in addition to endothelial cells, express and secrete factor VIII: this information should offer further opportunities for understanding mechanisms of factor VIII synthesis and replenishment.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical , Células Endoteliais/metabolismo , Fator VIII/biossíntese , Hemofilia A/terapia , Hemorragia/prevenção & controle , Células de Kupffer/metabolismo , Animais , Coagulação Sanguínea/genética , Modelos Animais de Doenças , Células Endoteliais/patologia , Fator VIII/genética , Fator VIII/metabolismo , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Regulação da Expressão Gênica , Sobrevivência de Enxerto , Hemofilia A/sangue , Hemofilia A/genética , Hemofilia A/patologia , Hemorragia/sangue , Hemorragia/genética , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Células de Kupffer/patologia , Fígado/metabolismo , Fígado/patologia , Megacariócitos/metabolismo , Megacariócitos/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Monócitos/metabolismo , Monócitos/patologia , Fenótipo , Transplante Heterólogo
4.
Pharmacol Res ; 68(1): 24-30, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23142211

RESUMO

Increasing evidence suggests that tachykinins are involved in the control of different pathological conditions, including psychiatric disorders. In this study we evaluated the expression of NK(1) and NK(2) receptors (NK-1R and NK-2R), as well as the effects of substance P (SP) and neurokinin A (NKA), in monocytes isolated from 15 healthy subjects and 15 patients with recurrent major depressive disorder (RMDD), under stable antidepressant therapy. NK-1R expression in monocytes from RMDD patients was significantly decreased as compared to healthy subjects, whereas NK-2R expression was markedly increased. Both NK-1R and NK-2R expression correlated with HAM-D, but not HAM-A, score. SP, NKA and selective NK-1R and NK-2R agonists stimulated TNF-α release in monocytes of both groups, with a significant higher effect observed in RMDD. Moreover they induced NF-κB activation, which was reversed by selective NK-1R and NK-2R antagonists, so demonstrating that it was receptor-mediated. The occurrence of a profound alteration in NK receptor expression in RMDD is a novel finding that suggests NK-1R and NK-2R pathways as possible relevant players in major depressive disorder, so improving our understanding of the complex pathogenesis of the disease.


Assuntos
Transtorno Depressivo Maior/metabolismo , Receptores da Neurocinina-1/metabolismo , Receptores da Neurocinina-2/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Monócitos/metabolismo , NF-kappa B/metabolismo , Neurocinina A/farmacologia , Neurotransmissores/farmacologia , Receptores da Neurocinina-1/agonistas , Receptores da Neurocinina-2/agonistas , Substância P/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
5.
Pharmacol Res ; 76: 149-56, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23954527

RESUMO

Microparticles (MPs) are membrane fragments that may play a role in the pathogenesis of chronic respiratory diseases. We aimed to investigate whether human monocytes/macrophage-derived MPs could induce a pro-inflammatory phenotype in human bronchial smooth muscle cells (BSMC) and the effect of montelukast in this setting. Experimental methods included isolation of human monocytes/macrophages and generation of monocyte-derived MPs, RT-PCR analysis of gene expression, immunoenzymatic determination of pro-inflammatory factor release, bioluminescent assay of intracellular cAMP levels and electromobility shift assay analysis of NF-κB nuclear translocation. Stimulation of human BSMC with monocyte-derived MPs induced a pro-inflammatory switch in human BSMC by inducing gene expression (COX-2 and IL-8), protein release in the supernatant (PGE2 and IL-8), and heterologous ß2-adrenoceptor desensitization. The latter effect was most likely related to autocrine PGE2 since pre-treatment with COX inhibitors restored the ability of salbutamol to induce cAMP synthesis in desensitized cells. Challenge with MPs induced nuclear translocation of NF-κB and selective NF-κB inhibition decreased MP-induced cytokine release in the supernatant. Montelukast treatment prevented IL-8 release and heterologous ß2-adrenoceptor desensitization in human BSMC exposed to monocyte-derived MPs by blocking NF-κB nuclear translocation. These findings provide evidence on the role of human monocyte-derived MPs in the airway smooth muscle phenotype switch as a novel potential mechanism in the progression of chronic respiratory diseases and on the protective effects by montelukast in this setting.


Assuntos
Acetatos/farmacologia , Antiasmáticos/farmacologia , Brônquios/citologia , Micropartículas Derivadas de Células/imunologia , Monócitos/imunologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/imunologia , Quinolinas/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Ciclopropanos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-8/genética , Interleucina-8/imunologia , Monócitos/citologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , NF-kappa B/análise , NF-kappa B/imunologia , NF-kappa B/metabolismo , Receptores Adrenérgicos beta/imunologia , Transdução de Sinais/efeitos dos fármacos , Sulfetos
6.
Rheumatology (Oxford) ; 51(11): 1942-52, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22829690

RESUMO

OBJECTIVES: Peroxisome proliferator-activated receptor-gamma (PPARγ) is expressed by different cell types in the joints and plays a relevant anti-inflammatory role in various diseases. This pilot study aimed to evaluate PPARγ expression in monocytes/macrophages isolated from RA patients as compared with healthy subjects, the relationships between PPARγ expression, MMP-9 activity and disease, and the influence of therapy with anti-rheumatic drugs on these parameters. METHODS: Thirty RA patients of both sexes (treated with CSs and MTX, mainly) and 15 healthy volunteers were enrolled in this study. Disease severity was evaluated by the 28-joint disease activity score (DAS-28). Monocytes and monocyte-derived macrophages (MDMs) were isolated by standard procedures. PPARγ protein and mRNA expression were assessed by immunoblotting and real-time PCR, respectively; MMP-9 activity was determined by gelatin zymography. Moreover, we checked the ability of 15-deoxy-Δ(12,14)-prostaglandin J(2) (15d-PGJ, a PPARγ agonist), MTX and methylprednisolone (MP) to affect PPARγ expression and lipopolysaccharide (LPS)-induced MMP-9 activity. RESULTS: Monocytes/MDMs from RA patients have significantly enhanced PPARγ expression (both protein and mRNA) and MMP-9 activity as compared with healthy donors. Interestingly, cells from patients with less active disease (DAS-28 <3.2) present higher PPARγ protein expression and lower MMP-9 activity than RA patients with DAS-28 >3.2. At therapeutic concentrations, MTX and MP increase in vitro PPARγ protein expression and inhibit LPS-induced MMP-9 activity. CONCLUSION: PPARγ expression in human monocytes/MDMs could represent an indicator of disease activity and therapy efficacy in RA because patients with a DAS-28 score <3.2 show the highest expression.


Assuntos
Antirreumáticos/uso terapêutico , Artrite Reumatoide/metabolismo , Macrófagos/metabolismo , Monócitos/metabolismo , PPAR gama/metabolismo , Artrite Reumatoide/tratamento farmacológico , Estudos de Casos e Controles , Quimioterapia Combinada , Feminino , Humanos , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Projetos Piloto , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Resultado do Tratamento , Adulto Jovem
7.
J Cell Mol Med ; 15(7): 1443-57, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21362130

RESUMO

Cytopathological features of cells from males and females, i.e. XX and XY isolated cells, have been demonstrated to represent a key variable in the mechanism underlying gender disparity in human diseases. Major insights came from the studies of gender differences in cell fate, e.g. in apoptotic susceptibility. We report here some novel insights recently emerged from literature that are referred as to a cytoprotection mechanism by which cells recycle cytoplasm and dispose of excess or defective organelles, i.e. autophagy. Autophagy and related genes have first been identified in yeast. Orthologue genes have subsequently been found in other organisms, including human beings. This stimulated the research in the field and, thanks to the use of molecular genetics and cell biology in different model systems, autophagy gained the attention of several research groups operating to analyse the pathogenetic mechanisms of human diseases. It remains unclear, however, whether autophagy can exert a protective effect or instead contribute to the pathogenesis of important human diseases. On the basis of the growing importance of sex/gender as key determinant of human pathology and of the known differences between males and females in the onset, progression, drug susceptibility and outcome of a plethora of diseases, the idea that autophagy could represent key and critical factor should be taken into account. In the review, we summarize our current knowledge about the role of autophagy in some paradigmatic human diseases (cancer, neurodegenerative, autoimmune, cardiovascular) and the role of 'cell sex' differences in this context.


Assuntos
Doenças Autoimunes/fisiopatologia , Autofagia/fisiologia , Doenças Cardiovasculares/fisiopatologia , Neoplasias/fisiopatologia , Doenças Neurodegenerativas/fisiopatologia , Caracteres Sexuais , Feminino , Humanos , Masculino , Modelos Biológicos
8.
Pharmacol Res ; 62(5): 391-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20670683

RESUMO

Previous studies have shown that NCX 6550 (NCX), a nitric oxide (NO)-donating pravastatin, induces anti-inflammatory effects in murine macrophage cell lines. Here, we have studied its activity in human monocyte/macrophages, by investigating cytokine release, NF-κB translocation and peroxisome proliferator-activated receptor γ (PPARγ) expression and function. For comparison, pravastatin, isosorbide-5-mononitrate (ISMN), sodium nitroprusside (SNP) and the PPARγ ligand 15-deoxy-Δ(12,14)-prostaglandin J(2) (PGJ) were also tested. Monocytes and macrophages (MDM: monocyte-derived macrophages) were isolated from healthy donors; cytokine release was measured by ELISA, NF-κB by electrophoretic mobility shift assay and PPARγ by Western blot and Real-Time PCR. NCX (1 nM-50 µM) dose-dependently inhibited phorbol 12-myristate 13-acetate (PMA)-induced TNF-α release from monocytes (IC(50)=240 nM) and MDM (IC(50)=52 nM). At 50 µM, it was more effective than pravastatin, ISMN and SNP (P<0.05), but less efficient than PGJ. Similar results were obtained for IL-6. Likewise, NCX was more effective than pravastatin and the other NO donors in inhibiting PMA-induced NF-κB translocation in both cell types, and, at the highest concentration, significantly (P<0.05) enhanced PPARγ protein expression in monocytes. We conclude that NCX 6550 exerts a significant anti-inflammatory activity in human monocyte/macrophages, that is also contributed by its NO donating properties, as the effects exerted by NCX are significantly higher than those evoked by pravastatin in many experimental assays. These data further indicate that the incorporation of a NO-donating moiety into a statin structure confers pharmacological properties which may translate into useful therapeutic benefits.


Assuntos
Citocinas/metabolismo , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , NF-kappa B/metabolismo , Nitratos/farmacologia , PPAR gama/metabolismo , Pravastatina/análogos & derivados , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Relação Dose-Resposta a Droga , Humanos , Interleucina-6/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Monócitos/metabolismo , NF-kappa B/antagonistas & inibidores , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Pravastatina/farmacologia , Superóxidos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
9.
J Pharmacol Exp Ther ; 331(2): 531-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19644041

RESUMO

Peroxisome proliferator-activated receptor (PPAR) activation reduces inflammation and atherosclerosis, but recent evidence raised concerns about its beneficial clinical effects. However, the effects of gender on PPAR expression and basal cytokine release have not been investigated. In the present study, we evaluated PPAR-gamma and -alpha expression, as well as cytokine release, in monocyte/macrophages from 15 male and 15 female patients with coronary artery disease (CAD) in comparison with healthy controls. Both expression and activation of PPAR-alpha and PPAR-gamma proteins were evaluated by Western blot and electrophoretic mobility shift assay. Gene expression was evaluated by real-time polymerase chain reaction; cytokine release was measured by enzyme-linked immunosorbent assay. Monocyte/macrophages of CAD patients yielded a constitutively enhanced (approximately 10-fold; p < 0.001) protein expression of PPAR-gamma, but not PPAR-alpha, compared with healthy controls. Evaluation of PPAR-gamma gene expression showed a 60-fold increase in monocytes from CAD patients, compared with healthy donors. Moreover, monocytes spontaneously released higher amounts of proinflammatory cytokines than macrophages. It is interesting that monocytes from CAD females expressed significantly higher levels of PPAR-gamma protein compared with male patients (p < 0.05) and showed the lowest basal release of tumor necrosis factor-alpha. These results indicate that the expression of PPAR-gamma is significantly higher in CAD patients than in healthy donors and that, together with cytokine release, it seems to be gender-related. In fact, CAD women demonstrated the highest PPAR-gamma expression and the lowest cytokine release. Such differences may, in part, modulate the response to PPAR-gamma activators.


Assuntos
Doença das Coronárias/metabolismo , Macrófagos/metabolismo , Monócitos/metabolismo , PPAR gama/biossíntese , Idoso , Western Blotting , Doença das Coronárias/genética , Citocinas/biossíntese , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Expressão Gênica/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , PPAR alfa/biossíntese , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Caracteres Sexuais
10.
Br J Pharmacol ; 175(1): 113-124, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29057467

RESUMO

BACKGROUND AND PURPOSE: A crosstalk between the immune system and depression has been postulated, with monocytes/macrophages and cytokines having a key role in this interaction. In this study, we examined whether vortioxetine, a multimodal anti-depressive drug, was endowed with anti-inflammatory and antioxidative activity, leading to immunomodulatory effects on human monocytes and macrophages. EXPERIMENTAL APPROACH: Human monocytes were isolated from buffy coats and used as such or differentiated into M1 and M2 macrophages. Cells were treated with vortioxetine before or after differentiation, and their responsiveness was evaluated. This included oxy-radical and TNFα production, TNFα and PPARγ gene expression and NF-κB translocation. KEY RESULTS: Vortioxetine significantly reduced the PMA-induced oxidative burst in monocytes and in macrophages (M1 and M2), causing a concomitant shift of macrophages from the M1 to the M2 phenotype, demonstrated by a significant decrease in the expression of the surface marker CD86 and an increase in CD206. Moreover, treatment of monocytes with vortioxetine rendered macrophages derived from this population less sensitive to PMA, as it reduced the oxidative burst, NF-kB translocation, TNFα release and expression while inducing PPARγ gene expression. FACS analysis showed a significant decrease in the CD14+ /CD16+ /CD86+ M1 population. CONCLUSIONS AND IMPLICATIONS: These results demonstrate that in human monocytes/macrophages, vortioxetine has antioxidant activity and anti-inflammatory effects driving the polarization of macrophages towards their alternative phenotype. These findings suggest that vortioxetine, alongside its antidepressive effect, may have immunomodulatory properties.


Assuntos
Anti-Inflamatórios/farmacologia , Fatores Imunológicos/farmacologia , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Piperazinas/farmacologia , Sulfetos/farmacologia , Antioxidantes/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Macrófagos/imunologia , Monócitos/imunologia , Vortioxetina
11.
Am J Cardiol ; 99(8): 1082-6, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17437731

RESUMO

This study assessed clinical and angiographic efficacies of oral treatment with prednisone at low-dose (LD) versus the previous high-dose (HD) immunosuppressive dosage used after percutaneous coronary interventions (PCIs) with bare metal stents in patients with multivessel coronary artery disease. Forty-three patients with multivessel disease successfully treated with multiple PCIs were matched to the previous HD IMPRESS-2/MVD study population. The 43 patients were treated for 103 coronary stenoses and received 30-day oral prednisone treatment (LD group 1 mg/kg for 5 days, 0.5 mg/kg for 10 days, 0.25 mg/kg for 15 days) and were compared retrospectively with the 43 patients in the HD IMPRESS-2/MVD study with 116 treated coronary lesions (HD group 1 mg/kg for 10 days, 0.5 mg/kg for 20 days, 0.25 mg/kg for 15 days). The primary clinical end point was 12-month event-free survival rate (defined as freedom from death, myocardial infarction, and need for target vessel revascularization). The secondary end point was angiographic restenosis at 8 months assessed by quantitative coronary angiography. Event-free survival rates were 74% and 93% in the LD and HD groups, respectively (relative risk 4.6, 95% confidence interval 1.18 to 17.8, p = 0.019). Restenosis was observed in 4 lesions (4%) in the HD group and in 20 lesions (22%) in the LD group (p <0.001). Mean late lumen loss was 0.61 +/- 0.35 mm, and the loss index was 31.3 +/- 21.6% in the HD group compared with 0.87 +/- 0.61 mm and 52.03 +/- 25.1% in the LD group (p = 0.03 and 0.02, respectively). In conclusion, antirestenotic efficacy of oral treatment with prednisone after conventional PCI is dose sensitive. A 50% dose decrease in oral prednisone, as tested in this study, is less effective than the previously tested HD IMPRESS therapeutic scheme.


Assuntos
Angioplastia Coronária com Balão , Anti-Inflamatórios/administração & dosagem , Glucocorticoides/administração & dosagem , Imunossupressores/administração & dosagem , Prednisona/administração & dosagem , Administração Oral , Estudos de Casos e Controles , Angiografia Coronária , Reestenose Coronária/etiologia , Estenose Coronária/terapia , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/etiologia , Estudos Prospectivos , Retratamento , Estudos Retrospectivos , Segurança , Stents , Taxa de Sobrevida , Resultado do Tratamento
12.
Drugs ; 67(9): 1243-55, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17547469

RESUMO

Vessel luminal narrowing after percutaneous coronary intervention (PCI) is characterised by platelet aggregation, release of growth factors, inflammatory cell infiltration, medial smooth muscle cell (SMC) proliferation, proteoglycan deposition and extracellular matrix remodelling. It is broadly accepted that the central mechanism at the basis of the whole pathophysiological process of restenosis is inflammation, triggered by vascular injury and activated through autocrine or paracrine mediators. Glucocorticosteroids exert beneficial effects on platelet function, on SMC proliferation and on collagen synthesis as well as inflammatory cell migration and activation, thus interfering with several steps of the cascade leading to neointima formation and subsequent late lumen loss. Initial experiences with systemic administration of glucocorticoids after PCI failed to confirm the expected benefits of this treatment, probably as a result of inadequate dosage and pharmacokinetic calculations. Recently a short-term, high-dose immunosuppressive treatment scheme with oral prednisone has demonstrated remarkable clinical and angiographic results when prednisone was given orally at a dose of 1 mg/kg for 10 days, 0.5 mg/kg for 20 days and 0.25 mg/kg for 15 days. This treatment has dramatically reduced the incidence of clinical vascular events at 1 year compared with controls (relative risk 0.34; 95% CI 0.12, 0.96; p = 0.006) and reduced the incidence of angiographic restenosis below 10% in different clinical and angiographic subsets. Secondary effects of a short course of glucocorticoids are generally minor, predictable and reversible: gastric pain, water and salt retention and worsened hypertension manifest in nearly 10% of patients. The addition of diuretics and acid suppressants before discharge, and the upgrading of antihypertensive medication thereafter, if needed, are useful preventive measures to control these temporary disorders. A routine blood cell count 4 weeks after PCI is advised in patients receiving thienopyridines (clopidogrel or ticlopidine) in addition to prednisone to rule out infrequent haematological dyscrasias. Emerging evidence supports this strategy as a convenient and well tolerated alternative to more expensive and complex revascularisation procedures such as drug eluting stent (DES) implantation or cardiac surgery, provided that the treatment is reserved for carefully selected candidates, i.e. after the exclusion of those with diabetes mellitus, a recent transmural myocardial infarction, or contraindications to the administration of a short-course of high-dose glucocorticosteroids. The recent concerns regarding the long-term safety of first-generation DES and the as yet undetermined duration of dual anti-platelet treatment, further supports the need for a simple pharmacological treatment that can be applied in a large percentage of patients currently treated with PCI. Multicentre randomised studies aimed at defining the efficacy and safety of oral prednisone treatment compared with metallic stents and DES are ongoing, and will become available in upcoming years.


Assuntos
Angioplastia/efeitos adversos , Reestenose Coronária/prevenção & controle , Glucocorticoides/uso terapêutico , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/efeitos adversos , Anti-Inflamatórios/uso terapêutico , Ensaios Clínicos como Assunto , Reestenose Coronária/etiologia , Reestenose Coronária/fisiopatologia , Glucocorticoides/administração & dosagem , Glucocorticoides/efeitos adversos , Humanos , Imunossupressores/administração & dosagem , Imunossupressores/efeitos adversos , Imunossupressores/uso terapêutico , Prednisolona/uso terapêutico , Resultado do Tratamento
13.
Life Sci ; 81(11): 906-15, 2007 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-17765929

RESUMO

Previous observations demonstrated that Peroxisome Proliferator-Activated Receptor-gamma (PPAR-gamma), a key regulator of adipocyte differentiation, is expressed in a large variety of cells, including cells of the monocyte/macrophage lineage. This study was aimed to quantify both the constitutive and ligand-induced PPAR-gamma expression in monocytes and monocyte-derived macrophages (MDM) isolated from healthy smokers and non-smokers, and to evaluate the possible direct effect of nicotine. PPAR-gamma protein was detected by Western blot and quantification was performed by calculating the ratio between PPAR-gamma and beta-actin protein expression. Cytokine release was measured with enzyme-linked immunoassay kits. Constitutive PPAR-gamma protein was detected in human monocytes and its expression was up-regulated along with differentiation to MDM. The endogenous ligand 15-deoxy-delta(12,14)-prostaglandin J(2) and the synthetic agonist ciglitazone enhanced PPAR-gamma expression, the former being effective also at low micromolar concentrations. Both agonists significantly inhibited the basal secretion of pro-inflammatory cytokines (e.g., TNF-alpha, IL-6), ciglitazone being more potent. Monocytes and MDM from healthy smokers presented a significantly enhanced (4-fold and 2.5-fold, respectively) constitutive PPAR-gamma expression, as compared to those from healthy non-smokers. However, ligand-induced PPAR-gamma expression and inhibition of cytokine secretion were similar in healthy smokers and non-smokers. Nicotine dose-dependently enhanced PPAR-gamma expression with a maximum at 10 muM, and inhibited release of pro-inflammatory cytokines; these effects were reversed by alpha-bungarotoxin. Nicotine and PPAR-gamma agonists did not exert synergistic effects. In conclusion, monocytes and MDM from healthy smokers present a constitutively enhanced PPAR-gamma expression; this effect is reproduced, to some extent, by nicotine in vitro.


Assuntos
Regulação Enzimológica da Expressão Gênica , Macrófagos/metabolismo , Monócitos/metabolismo , Nicotina/farmacologia , PPAR gama/metabolismo , Fumar , Actinas/metabolismo , Adulto , Feminino , Humanos , Interleucina-6/metabolismo , Macrófagos/efeitos dos fármacos , Masculino , Monócitos/efeitos dos fármacos , Nicotina/efeitos adversos , Receptores Nicotínicos/metabolismo , Tiazolidinedionas/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima , Receptor Nicotínico de Acetilcolina alfa7
14.
Cardiovasc Res ; 113(7): 711-724, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28472454

RESUMO

Women and men, female and male animals and cells are biologically different, and acknowledgement of this fact is critical to advancing medicine. However, incorporating concepts of sex-specific analysis in basic research is largely neglected, introducing bias into translational findings, clinical concepts and drug development. Research funding agencies recently approached these issues but implementation of policy changes in the scientific community is still limited, probably due to deficits in concepts, knowledge and proper methodology. This expert review is based on the EUGenMed project (www.eugenmed.eu) developing a roadmap for implementing sex and gender in biomedical and health research. For sake of clarity and conciseness, examples are mainly taken from the cardiovascular field that may serve as a paradigm for others, since a significant amount of knowledge how sex and oestrogen determine the manifestation of many cardiovascular diseases (CVD) has been accumulated. As main concepts for implementation of sex in basic research, the study of primary cell and animals of both sexes, the study of the influence of genetic vs. hormonal factors and the analysis of sex chromosomes and sex specific statistics in genome wide association studies (GWAS) are discussed. The review also discusses methodological issues, and analyses strength, weaknesses, opportunities and threats in implementing sex-sensitive aspects into basic research.


Assuntos
Pesquisa Biomédica/métodos , Doenças Cardiovasculares , Sistema Cardiovascular , Disparidades nos Níveis de Saúde , Disparidades em Assistência à Saúde , Projetos de Pesquisa , Animais , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/fisiopatologia , Doenças Cardiovasculares/terapia , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatologia , Cromossomos Humanos X , Cromossomos Humanos Y , Feminino , Predisposição Genética para Doença , Hormônios Esteroides Gonadais/metabolismo , Humanos , Masculino , Fenótipo , Gravidez , Prognóstico , Fatores de Risco , Caracteres Sexuais , Fatores Sexuais
15.
Br J Pharmacol ; 148(4): 478-89, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16633352

RESUMO

Macrophage activation is a key feature of inflammatory reactions occurring during bacterial infections, immune responses and tissue injury. We previously demonstrated that human macrophages of different origin express the tyrosine kinase receptor recepteur d'origine nantaise, the human receptor for MSP (RON) and produce superoxide anion (O(2)(-)) when challenged with macrophage-stimulating protein (MSP), the endogenous ligand for RON. This study was aimed to evaluate the role of MSP in alveolar macrophages (AM) isolated from healthy volunteers and patients with interstitial lung diseases (sarcoidosis, idiopathic pulmonary fibrosis), either smokers or non-smokers, by evaluating the respiratory burst, cytokine release and nuclear factor-kappa B (NF-kappaB) activation. MSP effects were compared with those induced by known AM stimuli, for example, phorbol myristate acetate, N-formyl-methionyl-leucyl-phenylalanine, lipopolysaccharide.MSP evokes O(2)(-) production, cytokine release and NF-kappaB activation in a concentration-dependent manner. By evaluating the respiratory burst, we demonstrate a significantly increased O(2)(-) production in AM from healthy smokers or smokers with pulmonary fibrosis, as compared to non-smokers, thus suggesting MSP as an enhancer of cigarette smoke toxicity. Besides inducing interleukin-1 beta (IL-1beta) and interleukin-10 (IL-10) production, MSP triggers an enhanced tumor necrosis factor-alpha release, especially in healthy and pulmonary fibrosis smokers. On the contrary, MSP-induced IL-10 release is higher in AM from healthy non-smokers. MSP activates the transcription factor NF-kappaB; this effect is more potent in healthy and fibrosis smokers (2.5-fold increase in p50 subunit translocation). This effect is receptor-mediated, as it is prevented by a monoclonal anti-human MSP antibody. The higher effectiveness of MSP in AM from healthy smokers and patients with pulmonary fibrosis is suggestive of its role in these clinical conditions.


Assuntos
Citocinas/biossíntese , Fator de Crescimento de Hepatócito/farmacologia , Macrófagos Alveolares/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas/farmacologia , Explosão Respiratória/efeitos dos fármacos , Fumar/imunologia , Idoso , Antígenos CD/análise , Antígenos de Diferenciação Mielomonocítica/análise , Relação Dose-Resposta a Droga , Feminino , Antígenos HLA-DR/análise , Humanos , Receptores de Lipopolissacarídeos/análise , Macrófagos Alveolares/metabolismo , Masculino , Pessoa de Meia-Idade , NF-kappa B/genética , Fibrose Pulmonar/imunologia , Sarcoidose/imunologia , Superóxidos/metabolismo
16.
Eur J Pharmacol ; 780: 33-7, 2016 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-26997366

RESUMO

Tocilizumab, etanercept and abatacept are biological drugs used in the therapy of Rheumatoid Arthritis (RA). Their mechanism of action is well documented but their direct effects on human monocytes/macrophages have not been fully investigated. The objective of this study was to evaluate in vitro the influence of these drugs on monocytes/macrophages from healthy volunteers. Human monocytes were isolated from healthy anonymous volunteers and cultured as such or differentiated to monocyte-derived macrophages (MDMs). The effect of tocilizumab, etanercept and abatacept (at concentrations similar to those in plasma of patients) on superoxide anion production, matrix metalloproteinase-9 (MMP-9) gene expression and activity, Peroxisome Proliferator-Activated Receptor (PPAR)γ expression and cell phenotype was evaluated. Exposure of monocytes/macrophages to tocilizumab, etanercept or abatacept resulted in a significant decrease of the PMA-induced superoxide anion production. Interestingly, the expression of PPARγ was significantly increased only by tocilizumab, while etanercept was the only one able to significantly reduce MMP-9 gene expression and inhibit the LPS-induced MMP-9 activity in monocytes. When etanercept and abatacept were added to the differentiating medium, both significantly reduced the amount of CD206(+)MDM. This study demonstrates that etanercept, abatacept and tocilizumab affect differently human monocytes/macrophages. In particular, the IL-6 antagonist tocilizumab seems to be more effective in inducing an anti-inflammatory phenotype of monocytes/macrophages compared to etanercept and abatacept, also in light of the up-regulation of PPARγ whose anti-inflammatory effects are well recognised.


Assuntos
Abatacepte/farmacologia , Anticorpos Monoclonais Humanizados/farmacologia , Etanercepte/farmacologia , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Biomarcadores/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/genética , Monócitos/citologia , Monócitos/metabolismo , PPAR gama/metabolismo , Fenótipo
17.
Br J Pharmacol ; 145(3): 385-96, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15778738

RESUMO

1 Substance P (SP) is deeply involved in lung pathophysiology and plays a key role in the modulation of inflammatory-immune processes. We previously demonstrated that SP activates guinea-pig alveolar macrophages (AMs) and human monocytes, but a careful examination of its effects on human AMs is still scarce. 2 This study was undertaken to establish the role of SP in human AM isolated from healthy smokers and non-smokers, by evaluating the presence of tachykinin NK(1) receptors (NK-1R) and SP's ability to induce superoxide anion (O(2)(-)) production and cytokine release, as well as activation of the nuclear factor-kappaB (NF-kappaB) pathway. 3 By Western blot analysis and immunofluorescence, we demonstrate that authentic NK-1R are present on human AMs, a three-fold enhanced expression being observed in healthy smokers. These NK-1R are functional, as SP and NK(1) agonists dose-dependently induce O(2)(-) production and cytokine release. In AMs from healthy smokers, SP evokes an enhanced respiratory burst and a significantly increased release of tumor necrosis factor-alpha as compared to healthy non-smokers, but has inconsistent effects on IL-10 release. The NK(1) selective antagonist CP 96,345 ((2S,3S)-cis-2-diphenylmethyl-N[(2-methoxyphenyl)-methyl]-1-azabicyclo-octan-3-amine)) competitively antagonized SP-induced effects. 4 SP activates the transcription factor NF-kappaB, a three-fold increased nuclear translocation being observed in AMs from healthy smokers. This effect is receptor-mediated, as it is reproduced by the NK(1) selective agonist [Sar(9)Met(O(2))(11)]SP and reverted by CP 96,345. 5 These results clearly indicate that human AMs possess functional NK-1R on their surface, which are upregulated in healthy smokers, providing new insights on the mechanisms involved in tobacco smoke toxicity.


Assuntos
Citocinas/metabolismo , Macrófagos Alveolares/metabolismo , NF-kappa B/metabolismo , Receptores da Neurocinina-1/biossíntese , Superóxidos/metabolismo , Adulto , Idoso , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Macrófagos Alveolares/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Receptores da Neurocinina-1/agonistas , Receptores da Neurocinina-1/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fumar/genética , Fumar/metabolismo , Substância P/farmacologia
18.
J Affect Disord ; 178: 188-92, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25841180

RESUMO

BACKGROUND: Neurokinin 1 receptors (NK-1R) have been involved in several psychiatric disorders including major depression, but less is known for bipolar disorder (BD). METHOD: We compared NK-1R expression and Substance P (SP) ability to induce NF-κB activation in monocytes from BD patients and healthy donors (HD), also looking for the effects of tobacco smoke. After informed written consent, 20 euthymic BD patients, either bipolar type 1 (BDI) or type 2 (BDII), and 14 age-matched healthy donors (HD) were enrolled. NK-1R expression in monocytes was evaluated by Western blot and expressed as the ratio between NK-1R and Na(+)/K(+)-ATPase protein expressions. NF-κB activation was assessed by measuring the nuclear content of the p50 subunit (ELISA kit). RESULTS: NK-1R expression was significantly reduced (P<0.001) in monocytes from BD patients as compared to HD, with no major differences between BDI and BDII patients. Tobacco smoke enhanced NK-1R expression in HD, but not in BD patients. Un-stimulated monocytes from BD patients presented a constitutively higher (P<0.05) content of nuclear p50 subunit as compared to HD. SP and an NK-1R agonist induced NF-κB activation, with a higher effect in HD: this effect was receptor-mediated as it was abrogated by an NK-1R antagonist. LIMITATIONS: As a pilot study enrolling 20 BD patients, an obvious limitation is the sample size. CONCLUSIONS: Our results show the existence of a relevant alteration in NK-1R expression in BD patients and further suggest SP involvement in BD, so improving our understanding of the underlying mechanisms of this disease.


Assuntos
Transtorno Bipolar/metabolismo , Monócitos/metabolismo , NF-kappa B/metabolismo , Receptores da Neurocinina-1/metabolismo , Substância P/metabolismo , Adulto , Western Blotting , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Transdução de Sinais , ATPase Trocadora de Sódio-Potássio/metabolismo
19.
Pigment Cell Melanoma Res ; 28(6): 718-29, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26358657

RESUMO

High plasma levels of nicotinamide phosphoribosyltransferase (NAMPT), traditionally considered an intracellular enzyme with a key role in NAD synthesis, have been reported in several oncological, inflammatory and metabolic diseases. We now show that eNAMPT can be actively released by melanoma cells in vitro. We analysed the mechanisms of its release, and we found both classical and non-classical pathway involvement. eNAMPT released by melanoma cells, in our hands, has paracrine and autocrine effects: it activates MAPK, AKT and NF-κB pathways and increases colony formation in anchorage-independent conditions. eNAMPT also induces M1 polarization in human monocytes. Last, we demonstrate, for the first time in any cancer type, that eNAMPT levels in plasma of tumour-bearing mice increase and that this increase can be reconducted to the tumour itself. This provides an important cue on previous observations that eNAMPT is increased in patients with cancer. Moreover, silencing NAMPT in melanoma cells leads to a reduction in the tumour growth rate. Our findings extend the basis to consider eNAMPT as a cytokine involved in tumour progression.


Assuntos
Citocinas/metabolismo , Melanoma/enzimologia , Nicotinamida Fosforribosiltransferase/metabolismo , Neoplasias Cutâneas/enzimologia , Animais , Comunicação Autócrina/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citocinas/sangue , Espaço Extracelular/enzimologia , Humanos , Peróxido de Hidrogênio/farmacologia , Melanoma/patologia , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Nicotinamida Fosforribosiltransferase/sangue , Comunicação Parácrina/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Neoplasias Cutâneas/patologia
20.
J Control Release ; 217: 284-92, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26385167

RESUMO

Synthetic microstructures can be engineered to deliver bioactive compounds impacting on their pharmacokinetics and pharmacodynamics. Herein, we applied dextran-based layer-by-layer (LbL) microcapsules to deliver alpha-2-macroglobulin (α2MG), a protein with modulatory properties in inflammation. Extending recent observations made with dextran-microcapsules loaded with α2MG in experimental sepsis, we focused on the physical and chemical characteristics of these microstructures and determined their biology on rodent and human cells. We report an efficient encapsulation of α2MG into microcapsules, which enhanced i) human leukocyte recruitment to inflamed endothelium and ii) human macrophage phagocytosis: in both settings microcapsules were more effective than soluble α2MG or empty microcapsules (devoid of active protein). Translation of these findings revealed that intravenous administration of α2MG-microcapsules (but not empty microcapsules) promoted neutrophil migration into peritoneal exudates and augmented macrophage phagocytic functions, the latter response being associated with alteration of bioactive lipid mediators as assessed by mass spectrometry. The present study indicates that microencapsulation can be an effective strategy to harness the complex biology of α2MG with enhancing outcomes on fundamental processes of the innate immune response paving the way to potential future development in the control of sepsis.


Assuntos
Leucócitos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , alfa-Macroglobulinas/administração & dosagem , Animais , Cápsulas , Células Cultivadas , Dextranos/química , Composição de Medicamentos , Células Endoteliais da Veia Umbilical Humana , Humanos , Imunidade Inata/efeitos dos fármacos , Leucócitos/imunologia , Macrófagos/imunologia , Masculino , Camundongos Endogâmicos C57BL , alfa-Macroglobulinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA