Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Biochim Biophys Acta ; 1852(9): 1719-28, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26004395

RESUMO

Adenomatous polyposis coli (APC) is a tumor suppressor involved in the Wnt signaling, the primary driving force of the intestinal epithelium homeostasis. Alterations of components of the Wnt pathway, and in most cases mutations of APC, have been reported to promote colorectal cancer (CRC). During differentiation the enterocytes migrate from the crypt to the tip of the villus where they undergo apoptosis thus ensuring the continual renewal of the intestinal mucosa. The differentiation process is characterized by an activation gradient of the Wnt signaling pathway accompanied by a metabolic switch from glycolysis to mitochondrial oxidative phosphorylation along the crypt-villus axis. In the present work, we study the relationship between the expression of wild type APC protein and mitochondrial oxidative metabolism in HT29 colorectal cancer cells, originally carrying endogenous inactive APC alleles. By generating mtDNA-depleted (rho0) APC-inducible HT29 cells, we demonstrate for the first time that the APC-dependent apoptosis requires the production of reactive oxygen species (ROS) by the mitochondrial respiratory chain. The possible role of mitochondria as putative target in the prevention and/or therapy of colorectal cancer is herein discussed.

2.
Arterioscler Thromb Vasc Biol ; 34(8): 1689-96, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24925970

RESUMO

OBJECTIVE: Platelet-neutrophil interactions play a key role in cardiovascular disease and inflammatory processes. Src family kinases mediate P-selectin glycoprotein ligand-1-Mac-1 cross talk necessary for firm platelet-neutrophil adhesion. Because Src family kinase activity can be regulated by cAMP-dependent pathways, in this work, we evaluated the role of phosphodiesterases in the signaling events that are required to sustain platelet-neutrophil interactions and neutrophil recruitment at the site of vascular injury. APPROACH AND RESULTS: In neutrophils exposed to P-selectin, selective phosphodiesterase 4 (PDE4) inhibition prevented Src family kinase-mediated phosphorylation of the proline-rich tyrosine kinase 2 on Tyr579/Tyr580. The effects of PDE4 inhibition required protein kinase A, likely through protein kinase A-mediated activation of COOH-terminal Src kinase, a major negative regulator of Src family kinases. PDE4, but not other phosphodiesterase inhibitors, reduced platelet-neutrophil conjugates as well as neutrophil firm adhesion on spread platelets under flow conditions. The effect of PDE4 inhibition on neutrophil adhesion was primarily mediated by downregulation of P-selectin-induced activation of Mac-1. In a murine model of endovascular injury, selective inhibition of PDE4 significantly reduced neutrophil recruitment at the site of vascular damage. CONCLUSIONS: This study identifies PDE4 as a central node in the signaling network that mediates platelet-neutrophil adhesion and suggests that pharmacological inhibition of PDE4 may represent a novel therapeutic avenue for the treatment of cardiovascular disease.


Assuntos
Plaquetas/efeitos dos fármacos , Artéria Femoral/efeitos dos fármacos , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Selectina-P/metabolismo , Inibidores da Fosfodiesterase 4/farmacologia , Adesividade Plaquetária/efeitos dos fármacos , Lesões do Sistema Vascular/tratamento farmacológico , 4-(3-Butoxi-4-metoxibenzil)-2-imidazolidinona/farmacologia , Animais , Plaquetas/enzimologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Artéria Femoral/enzimologia , Artéria Femoral/lesões , Quinase 2 de Adesão Focal/metabolismo , Humanos , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/metabolismo , Camundongos , Camundongos Knockout , Neutrófilos/enzimologia , Selectina-P/genética , Fosforilação , Rolipram/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Lesões do Sistema Vascular/sangue , Lesões do Sistema Vascular/enzimologia , Quinases da Família src/metabolismo
3.
Biochim Biophys Acta ; 1832(12): 2289-301, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24060638

RESUMO

BACKGROUND: Nuclear receptors are a class of 48 ligand-activated transcription factors identified as key players of metabolic and developmental processes. Most of these receptors are potential targets for pharmacological strategies in the Metabolic Syndrome. In the present study, we analyzed changes in the mRNA expression of nuclear receptors in the peripheral blood mononuclear cells of patients with Metabolic Syndrome, in order to identify novel biomarkers of disease and candidate targets for putative therapeutical approaches. METHODS AND RESULTS: We enrolled thirty healthy controls (14 M:16 F) and thirty naïve patients (16 M: 14 F; >3 criteria for Metabolic Syndrome upon Adult Treatment Panel III) without organ damage. Using quantitative real-time PCR, we assessed the expression patterns of nuclear receptors in peripheral blood mononuclear cells. 33/48 nuclear receptors were expressed in peripheral blood mononuclear cells. In patients with Metabolic Syndrome, we found a significant down-regulation of the entire PPAR, NR4A and RAR families, together with a repression of RXRα, VDR, and Rev-Erbα. Furthermore, we performed a novel statistical analysis with classification trees, which allowed us to depict a predictive core of nuclear receptor expression patterns characterizing subjects with Metabolic Syndrome. Random Forest Analysis identified NOR1 and PPARδ, which were both reduced in peripheral blood mononuclear cells and specifically in CD14(+) cells (mostly monocytes), as classifiers of Metabolic Syndrome, with high specificity and sensitivity. CONCLUSIONS: Our results point to the use of PPAR and NR4A mRNA levels in the overall peripheral blood mononuclear cells as biomarkers of Metabolic Syndrome and bona fide putative targets of pharmacological therapy.


Assuntos
Biomarcadores/metabolismo , Leucócitos Mononucleares/metabolismo , Síndrome Metabólica/diagnóstico , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , Adulto , Western Blotting , Estudos de Casos e Controles , Estudos de Coortes , Regulação para Baixo , Feminino , Citometria de Fluxo , Humanos , Masculino , Síndrome Metabólica/genética , Síndrome Metabólica/metabolismo , Pessoa de Meia-Idade , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores Citoplasmáticos e Nucleares/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo
4.
Gastroenterology ; 144(7): 1497-507, 1507.e1-13, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23419360

RESUMO

BACKGROUND & AIMS: Liver X receptors (LXRs) are transcriptional regulators of cholesterol metabolism, controlling cholesterol flow into cells, catabolism, and efflux. Cholesterol controls cell proliferation; disruptions in cholesterol metabolism have been associated with the development of colon cancer. We investigated whether expression of activated LXR protects against intestinal tumorigenesis in mice. METHODS: We analyzed the development of colon cancer in mice that express a constitutive active form of LXRα only in the intestinal epithelium, under the control of villin promoter (iVP16LXRα). These mice were crossed with adenomatous polyposis coli (Apc)(min/+) mice, or given azoxymethane followed by dextran sodium sulfate, to assess intestinal tumor formation. We also assessed proliferation and apoptosis of a human colorectal cancer cell line (HT29) transfected with an adenoviral vector that expressed Ad VP16hLXRα, compared with cells expressing AdVP16 (control), and their ability to form xenograft tumors in mice. HT29 cells also were incubated with the LXR ligand GW3965. RESULTS: In human colorectal cancer cells, ligand-induced activation of LXR or transfection with Ad VP16hLXRα blocked the G1 phase, increased caspase-dependent apoptosis, and slowed growth of xenograft tumors in mice. iVP16LXRα mice formed fewer, smaller tumors than VP16 (control) mice after administration of azoxymethane and dextran sodium sulfate. APC(min/+)/iVP16LXRα mice also developed fewer, smaller intestinal tumors than APC(min/+)/iVP16 mice. Gene expression analysis indicated that activation of LXRα affected lipid metabolic networks and increased cholesterol efflux in the intestine. CONCLUSIONS: Expression of activated LXRα blocks proliferation of human colorectal cancer cells and slows the growth of xenograft tumors in mice. It also reduces intestinal tumor formation after administration of chemical carcinogens, and in Apc(min/+) mice. LXR agonists therefore might be developed as therapeutic treatments for colorectal cancer.


Assuntos
Adenocarcinoma/metabolismo , Adenoma/fisiopatologia , Transformação Celular Neoplásica , Neoplasias Colorretais/metabolismo , Neoplasias Intestinais/metabolismo , Receptores Nucleares Órfãos/fisiologia , Adenocarcinoma/patologia , Animais , Proliferação de Células , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Genes APC , Células HT29 , Humanos , Neoplasias Intestinais/patologia , Receptores X do Fígado , Camundongos , Camundongos Transgênicos , Receptores Nucleares Órfãos/metabolismo , Transdução de Sinais , Transplante Heterólogo
5.
Proc Natl Acad Sci U S A ; 108(16): 6603-8, 2011 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-21467224

RESUMO

Peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC1α) is a transcriptional coactivator able to up-regulate mitochondrial biogenesis, respiratory capacity, oxidative phosphorylation, and fatty acid ß-oxidation with the final aim of providing a more efficient pathway for aerobic energy production. In the continuously renewed intestinal epithelium, proliferative cells in the crypts migrate along the villus axis and differentiate into mature enterocytes, increasing their respiratory capacity and finally undergoing apoptosis. Here we show that in the intestinal epithelial surface, PGC1α drives mitochondrial biogenesis and respiration in the presence of reduced antioxidant enzyme activities, thus determining the accumulation of reactive oxygen species and fostering the fate of enterocytes toward apoptosis. Combining gain- and loss-of-function genetic approaches in human cells and mouse models of intestinal cancer, we present an intriguing scenario whereby PGC1α regulates enterocyte cell fate and protects against tumorigenesis.


Assuntos
Antioxidantes/metabolismo , Enterócitos/metabolismo , Proteínas de Choque Térmico/metabolismo , Neoplasias Intestinais/metabolismo , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição/metabolismo , Animais , Transformação Celular Neoplásica , Enterócitos/patologia , Proteínas de Choque Térmico/genética , Humanos , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Consumo de Oxigênio/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/genética
6.
FASEB J ; 24(10): 3970-80, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20530751

RESUMO

Inflammatory lung disease is a primary cause of morbidity and mortality in cystic fibrosis (CF). Mechanisms of unresolved acute inflammation in CF are not completely known, although the involvement of cystic fibrosis transmembrane conductance regulator (CFTR) in nonrespiratory cells is emerging. Here we examined CFTR expression and function in human platelets (PLTs) and found that they express a biologically active CFTR. CFTR blockade gave an ∼50% reduction in lipoxin A(4) (LXA(4)) formation during PLT/polymorphonuclear leukocytes (PMN) coincubations by inhibiting the lipoxin synthase activity of PLT 12-lipoxygenase. PLTs from CF patients generated ∼40% less LXA(4) compared to healthy subject PLTs. CFTR inhibition increased PLT-dependent PMN viability (33.0±5.7 vs. 61.2±8.2%; P=0.033), suppressed nitric oxide generation (0.23±0.04 vs. 0.11±0.002 pmol/10(8) PLTs; P=0.004), while reducing AKT (1.02±0.12 vs. 0.71±0.007 U; P=0.04), and increasing p38 MAPK phosphorylation (0.650±0.09 vs. 1.04±0.24 U; P=0.03). Taken together, these findings indicate that PLTs from CF patients are affected by the molecular defect of CFTR. Moreover, this CF PLT abnormality may explain the failure of resolution in CF.


Assuntos
Plaquetas/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/sangue , Mediadores da Inflamação/fisiologia , Apoptose , Linhagem Celular , Feminino , Humanos , Imuno-Histoquímica , Masculino , Microscopia Eletrônica de Transmissão , Fosforilação , Proteínas Quinases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Front Pharmacol ; 12: 702677, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34566635

RESUMO

Neutrophilic inflammation is a key determinant of cystic fibrosis (CF) lung disease. Neutrophil-derived free DNA, released in the form of extracellular traps (NETs), significantly correlates with impaired lung function in patients with CF, underlying their pathogenetic role in CF lung disease. Thus, specific approaches to control NETosis of neutrophils migrated into the lungs may be clinically relevant in CF. We investigated the efficacy of phosphodiesterase (PDE) type-4 inhibitors, in vitro, on NET release by neutrophils from healthy volunteers and individuals with CF, and in vivo, on NET accumulation and lung inflammation in mice infected with Pseudomonas aeruginosa. PDE4 blockade curbed endotoxin-induced NET production and preserved cellular integrity and apoptosis in neutrophils, from healthy subjects and patients with CF, challenged with endotoxin, in vitro. The pharmacological effects of PDE4 inhibitors were significantly more evident on CF neutrophils. In a mouse model of Pseudomonas aeruginosa chronic infection, aerosol treatment with roflumilast, a selective PDE4 inhibitor, gave a significant reduction in free DNA in the BALF. This was accompanied by reduced citrullination of histone H3 in neutrophils migrated into the airways. Roflumilast-treated mice showed a significant improvement in weight recovery. Our study provides the first evidence that PDE4 blockade controls NETosis in vitro and in vivo, in CF-relevant models. Since selective PDE4 inhibitors have been recently approved for the treatment of COPD and psoriasis, our present results encourage clinical trials to test the efficacy of this class of drugs in CF.

8.
Biochem J ; 396(1): 89-98, 2006 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-16433632

RESUMO

In cell suspensions subjected to high-shear rotatory motion, human PMN (polymorphonuclear cells) adhered to E-selectin-expressing CHO (Chinese-hamster ovary) cells (CHO-E), and formed homotypic aggregates when challenged by E-selectin-IgG fusion protein, by a mechanism that involved beta2 integrins. Both heterotypic and homotypic PMN adhesion was accompanied by tyrosine phosphorylation of a 110 kDa protein (P110). This event was prevented by blocking anti-(beta2 integrin) antibodies and by inhibitors of Src-family kinases, suggesting that it was part of an 'outside-in' signalling that was initiated by integrin engagement. Interestingly, Src-family kinase inhibitors prevented beta2-integrin-mediated (i) homotypic PMN adhesion triggered by E-selectin-IgG, (ii) heterotypic CHO-E/PMN adhesion in mixed-cell suspensions, and (iii) firm adhesion of PMN to CHO-E monolayers under physiological flow. Similarly to PMN treated with Src-family kinase inhibitors, PMN from hck-/-fgr-/- and hck-/-fgr-/-lyn-/- mice showed significant impairment of beta2-integrin-mediated adhesion to CHO-E. Moreover, the expression of beta2 integrin activation epitopes at the sites of cell-cell contact in CHO-E/PMN conjugates was abolished by Src-family kinase inhibitors. One component of P110 was identified as the FAK (focal adhesion kinase) Pyk2 (proline-rich tyrosine kinase 2), which was phosphorylated in a beta2 integrin- and Src-family-kinase-dependent manner. Thus, Src-family kinases, and perhaps Pyk2, mediate a signal necessary for beta2 integrin function in PMN tethered by E-selectin.


Assuntos
Antígenos CD18/fisiologia , Adesão Celular/fisiologia , Selectina E/fisiologia , Neutrófilos/citologia , Transdução de Sinais/fisiologia , Quinases da Família src/fisiologia , Animais , Células CHO , Cricetinae , Cricetulus , Ácido Egtázico/farmacologia , Citometria de Fluxo , Quinase 2 de Adesão Focal/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Processamento de Proteína Pós-Traducional , Pirazóis/farmacologia , Pirimidinas/farmacologia , Proteínas Recombinantes de Fusão/fisiologia , Reologia , Estresse Mecânico , Transfecção , Quinases da Família src/antagonistas & inibidores
9.
Thromb Haemost ; 94(3): 568-77, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16268474

RESUMO

Clopidogrel is considered to be an important therapeutic advance in anti-platelet therapy. We investigated whether inhibition by clopidogrel results in a reduced capacity of platelets to adhere and stimulate pro-atherothrombotic and inflammatory functions in polymorphonuclear leukocytes (PMN) and in monocytes (MN). An eventual effect on these processes could further substantiate anti-atherothrombotic properties of this drug. The effects of clopidogrel or of its active metabolite were investigated on ADP or thrombin receptor-induced platelet activation and on platelet-leukocyte interactions ex vivo in the mouse or in vitro in isolated human cells or whole blood, respectively. Clopidogrel inhibited platelet aggregation, expression of P-selectin, platelet-PMN adhesion and platelet-dependent ROS production in mouse PMN. Similarly pretreatment of human platelets with the active metabolite of clopidogrel in vitro resulted in a profound inhibition of platelet P-selectin expression, platelet-PMN adhesion and production of ROS by PMN. Pretreatment with the active metabolite of clopidogrel significantly impaired the ability of platelets to up-regulate the expression of TF procoagulant activity in MN, in a washed cell system. Moreover, the active metabolite of clopidogrel inhibited rapidTF exposure on platelet as well as on leukocyte surfaces in whole blood. By reducing platelet-dependent up-regulation of inflammatory and pro-atherothrombotic functions in leukocytes, clopidogrel may reduce inflammation that underlies the chronic process of atherosclerosis and its acute complications.


Assuntos
Plaquetas/efeitos dos fármacos , Leucócitos/efeitos dos fármacos , Ativação Plaquetária/efeitos dos fármacos , Adesividade Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Ticlopidina/análogos & derivados , Animais , Plaquetas/metabolismo , Clopidogrel , Relação Dose-Resposta a Droga , Humanos , Leucócitos/metabolismo , Masculino , Camundongos , Selectina-P/metabolismo , Inibidores da Agregação Plaquetária/administração & dosagem , Inibidores da Agregação Plaquetária/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Estereoisomerismo , Tromboplastina/metabolismo , Ticlopidina/administração & dosagem , Ticlopidina/metabolismo , Ticlopidina/farmacologia
10.
Biochem J ; 380(Pt 1): 57-65, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-14969582

RESUMO

In human PMN (polymorphonuclear cells), challenged by P-selectin, the beta2-integrin Mac-1 (macrophage antigen-1) promoted the activation of the SRC (cellular homologue of Rous sarcoma virus oncogenic protein) family members HCK (haematopoietic cell kinase) and LYN (an SRC family protein tyrosine kinase) and phosphorylation of a P-110 (110 kDa protein). SRC kinase activity in turn was necessary for macrophage antigen-1-mediated adhesion [Piccardoni, Sideri, Manarini, Piccoli, Martelli, de Gaetano, Cerletti and Evangelista (2001) Blood 98, 108-116]. This suggested that an SRC-dependent outside-in signalling strengthens the beta2-integrin interaction with the ligand. To support this hypothesis further, in the present study, we used the monoclonal antibody KIM127 or manganese to lock beta2 integrins in a high-affinity state, and homotypic PMN adhesion was analysed to monitor beta2-integrin adhesive function. KIM127 or manganese induced PMN homotypic adhesion and P-110 phosphorylation. Both these processes were abolished by blocking antibodies against the common beta2 chain, by a combination of antibodies against alphaL and alphaM or by inhibitors of SRC activity. Confocal microscopy showed that activation epitopes were expressed by beta2 integrins co-localized with patches of F-actin at the adhesion sites. Blockade of SRC kinases or of actin polymerization prevented clustering of activated integrins as well as F-actin accumulation. FACS analysis showed that SRC inhibitors modified neither basal nor manganese-induced KIM127 binding. An SRC-dependent outside-in signalling initiated by beta2 integrins was also required for adhesion triggered by interleukin-8. These results confirm the hypothesis that an SRC-dependent outside-in signalling triggered by high affinity and ligand binding is necessary to stabilize beta2-integrin-mediated adhesion. Allowing clustering of activated integrins, SRC might link the high-affinity with the high-avidity state. Proline-rich tyrosine kinase-2 appears to be involved in this process.


Assuntos
Antígeno-1 Associado à Função Linfocitária/fisiologia , Antígeno de Macrófago 1/fisiologia , Neutrófilos/fisiologia , Selectina-P/fisiologia , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Quinases/fisiologia , Actinas/fisiologia , Adulto , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Células Cultivadas/enzimologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Quinase 2 de Adesão Focal , Humanos , Interleucina-8/farmacologia , Neutrófilos/enzimologia , Fosforilação , Fosfotirosina/fisiologia , Estresse Mecânico
11.
Transplantation ; 74(8): 1152-8, 2002 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-12438963

RESUMO

BACKGROUND: We have previously demonstrated that interleukin (IL)-10 synergizes with dexamethasone (Dex) in inhibiting proliferation of human T cells, stimulated in an antigen-presenting cell (APC)-dependent manner. Because IL-10 effectively inhibits APC accessory functions, the synergism could have been a result of its effect on APC. We then investigated the effects of Dex and IL-10 on T-cell subpopulations, stimulated in an APC-independent manner. METHODS: CD4 and CD8 T cells were stimulated with anti-CD3, with or without Dex and IL-10, alone or in combination. Proliferation, glucocorticoid (GC) receptor binding, anti-CD3-induced tyrosine phosphorylation, IL-2 production, and expression of IL-2 receptor alpha, beta, and gamma chains were evaluated. The pharmacologic interactions were analyzed using the isobole method. RESULTS: IL-10 synergized with Dex in inhibiting CD4 but not CD8 T-cell proliferation. The synergism was not associated with modifications of GC receptor number or affinity, nor with modifications of anti-CD3-induced tyrosine phosphorylation. IL-10 synergized with Dex in inhibiting IL-2 production and increased Dex inhibitory effect on the expression of the IL-2 receptor alpha chain, which is up-regulated by CD3 stimulation and IL-2. Only Dex inhibited the beta and gamma chain expression, which, interestingly, is not up-regulated by IL-2. IL-2, as well as IL-7 and IL-15, reversed the effects of IL-10 but not those of Dex. CONCLUSIONS: IL-10 synergizes with Dex in inhibiting CD4 T-cell proliferation. Its synergizing effect is mediated by the inhibition of IL-2 production. Dex exerts additional activities, such as the inhibition of beta and gamma chain expression. Therefore, IL-10 could be useful for the enhancement of GC-based immunosuppressive therapies.


Assuntos
Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Interleucina-10/farmacologia , Anticorpos Monoclonais/farmacologia , Complexo CD3/imunologia , Complexo CD3/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/imunologia , Células Cultivadas , Sinergismo Farmacológico , Humanos , Interleucina-15/farmacologia , Interleucina-2/biossíntese , Interleucina-2/metabolismo , Interleucina-2/farmacologia , Interleucina-7/farmacologia , Fosforilação , Receptores de Glucocorticoides/metabolismo , Receptores de Interleucina-2/metabolismo , Tirosina/metabolismo
12.
Int J Dev Neurosci ; 21(7): 379-89, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14599484

RESUMO

Primary cultures of cerebellar granule neurons (CGNs) were prepared from 8-day-old Wistar rats, and maintained in an appropriate medium containing a high (25 mM) concentration of KCl. To induce apoptosis, culture medium was replaced with serum-free medium (containing 5mM KCl) 8 days after plating. Apoptosis was measured by the terminal deoxynucleotidyl transferase-mediated dUTP-fluorescein nick end-labeling (TUNEL) method, and by flow cytometry. Since there is evidence that an increased formation of reactive oxygen species (ROS) is involved in the apoptosis induced by low K(+) (5mM) concentrations, the potential anti-apoptotic effect of caffeic acid phenethyl ester (CAPE), a potent flavonoid antioxidant, was tested in this experimental model. It was found that CAPE (10 microg/ml) promoted cell survival and was capable of blocking the apoptotic process as assayed by both TUNEL and flow cytometric methods. The same concentration of CAPE prevented the formation of ROS induced by low K(+). Since there is evidence that low K(+)-induced apoptosis in CGNs is associated with a drop in intracellular Ca(2+) concentration ([Ca(2+)](i)), activation of the cell death effector proteases caspase-3 and caspase-9, and of the transcription factor nuclear factor kappa B (NF-kappaB), the interference of CAPE with these purported mediators of apoptosis was also evaluated. It was found that CAPE did not interfere with the marked decrease in [Ca(2+)](i) induced by low K(+), whereas it completely blocked caspase-3, caspase-9, and NF-kappaB activation. It is concluded that CAPE could exert its anti-apoptotic effect in CGNs by blocking ROS formation and by inhibiting caspase activity.


Assuntos
Apoptose/efeitos dos fármacos , Ácidos Cafeicos/farmacologia , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Álcool Feniletílico/análogos & derivados , Álcool Feniletílico/farmacologia , Potássio/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/fisiologia , Caspase 3 , Caspase 9 , Inibidores de Caspase , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Meios de Cultura/farmacologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo
13.
Eur J Pharmacol ; 453(1): 131-9, 2002 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-12393068

RESUMO

Polymorphonuclear leukocytes are strongly implicated in the pathogenesis of inflammatory disease. Polymorphonuclear leukocyte recruitment at sites of inflammation, mainly sustained by the beta2-integrins, is followed by the synthesis and release of inflammatory mediators, such as leukotrienes, proteolytic enzymes and reactive oxygen species. Functional and metabolic interactions between polymorphonuclear leukocytes and platelets can contribute to and exacerbate the process. The effects of the dual 5-lipoxygenase and cyclooxygenase inhibitor licofelone ([2,2-dimethyl-6-(4-chlorophenyl)-7-phenyl-2,3-dihydro-1H-pyrrolizine-5-yl]-acetic acid) were studied on arachidonic acid transcellular metabolism occurring between polymorphonuclear leukocytes and platelets. The formation of leukotriene C(4), a leukotriene A(4)-derived metabolite, by mixed polymorphonuclear leukocyte/platelet suspensions stimulated with 10 microM A23187 was inhibited by licofelone with an IC(50) of 3.8 +/- 0.07 microM. The formation of 5,12-di-hydroxy-eicosatetraenoic acid (HETE) was abolished at concentrations > or = 10 microM. Licofelone also inhibited the generation of reactive oxygen species by polymorphonuclear leukocytes stimulated with 1 microM n-formyl-methionyl-leucyl-phenylalanine (fMLP), 10 nM complement fraction 5a (C5a) and 1 microM platelet activating factor (PAF) with IC(50)s of 24.4 +/- 0.6, 11.0 +/- 1.5 and 11.7 +/-1.2 microM; elastase release induced by the three agonists was inhibited with IC(50)s of 12.2 +/- 2.2, 23.5 +/- 8 and 2.6 +/- 1 microM, respectively. Homotypic polymorphonuclear leukocyte aggregation induced by fMLP, C5A and PAF was inhibited by licofelone with IC(50)s of 23.7 +/- 4.8, 15.6 +/- 3.4 and 15.4 +/- 4 microM, respectively. The present study extends the anti-lipoxygenase and anti-cyclooxygenase activities of licofelone to the production of arachidonic acid metabolites generated as a consequence of polymorphonuclear leukocyte-platelet transcellular metabolism and to polymorphonuclear leukocyte responses relevant to the pathogenesis of inflammation. The coexistence within the same molecule of a wide spectrum of anti-inflammatory properties is of interest.


Assuntos
Acetatos/farmacologia , Inibidores de Ciclo-Oxigenase/farmacologia , Regulação para Baixo/efeitos dos fármacos , Inibidores de Lipoxigenase/farmacologia , Neutrófilos/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Pirróis/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo/fisiologia , Humanos , Lipoxigenase/metabolismo , Neutrófilos/enzimologia , Testes de Função Plaquetária , Prostaglandina-Endoperóxido Sintases/metabolismo
14.
PLoS One ; 9(8): e104449, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25116592

RESUMO

BACKGROUND & AIMS: Liver regeneration (LR) is a valuable model for studying mechanisms modulating hepatocyte proliferation. Nuclear receptors (NRs) are key players in the control of cellular functions, being ideal modulators of hepatic proliferation and carcinogenesis. METHODS & RESULTS: We used a previously validated RT-qPCR platform to profile modifications in the expression of all 49 members of the NR superfamily in mouse liver during LR. Twenty-nine NR transcripts were significantly modified in their expression during LR, including fatty acid (peroxisome proliferator-activated receptors, PPARs) and oxysterol (liver X receptors, Lxrs) sensors, circadian masters RevErbα and RevErbß, glucocorticoid receptor (Gr) and constitutive androxane receptor (Car). In order to detect the NRs that better characterize proliferative status vs. proliferating liver, we used the novel Random Forest (RF) analysis to selected a trio of down-regulated NRs (thyroid receptor alpha, Trα; farsenoid X receptor beta, Fxrß; Pparδ) as best discriminators of the proliferating status. To validate our approach, we further studied PPARδ role in modulating hepatic proliferation. We first confirmed the suppression of PPARδ both in LR and human hepatocellular carcinoma at protein level, and then demonstrated that PPARδ agonist GW501516 reduces the proliferative potential of hepatoma cells. CONCLUSIONS: Our data suggest that NR transcriptome is modulated in proliferating liver and is a source of biomarkers and bona fide pharmacological targets for the management of liver disease affecting hepatocyte proliferation.


Assuntos
Carcinoma Hepatocelular/genética , Hepatócitos/metabolismo , Neoplasias Hepáticas/genética , Regeneração Hepática/genética , Receptores Citoplasmáticos e Nucleares/genética , Animais , Carcinoma Hepatocelular/metabolismo , Linhagem Celular , Proliferação de Células , Análise por Conglomerados , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Fígado/metabolismo , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Receptores Citoplasmáticos e Nucleares/metabolismo , Transcriptoma
15.
Thromb Haemost ; 107(6): 1130-40, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22436970

RESUMO

Prasugrel, through its active metabolite, reduces atherothrombosis and its clinical manifestations by inhibiting platelet activation and aggregation. Platelets also contribute to inflammation through interaction with different classes of leukocytes. We investigated whether the inhibitory effect of prasugrel on platelets also counteract inflammatory responses. The effect of prasugrel active metabolite, R-138727, was investigated on platelet P-selectin expression, platelet adhesion to polymorphonuclear leukocytes (PMN) and monocytes (MN) and Mac-1 expression in PMN and MN, in vitro, in human cells. The ex vivo effect of prasugrel administration on P-selectin, thromboxane (TXB)2 formation, platelet-PMN conjugates and Mac-1 expression in PMN triggered by PAR-4 agonist peptide was examined in whole blood from healthy mice as well as from mice in which an acute inflammatory reaction was induced by treatment with endotoxin. The effect of prasugrel on inflammatory markers in endotoxin-treated animals was also tested in vivo. R-138727 inhibited agonist-stimulated expression of platelet P-selectin, platelet-PMN and platelet-MN adhesion and platelet-dependent Mac-1 expression in leukocytes. Addition of aspirin did not modify the inhibitory effect elicited by R-138727. Treatment of mice with prasugrel resulted in a profound inhibition of platelet P-selectin expression, TXB2 production, platelet-PMN adhesion and Mac-1 expression in PMN induced by ex vivo stimulation with PAR-4 agonist peptide of whole blood from healthy or endotoxin-treated mice. Measurement of markers revealed that prasugrel reduced TXB2 and tumour necrosis factor-α synthesis and increased nitric oxide metabolites in endotoxin-treated mice in vivo. In conclusion, prasugrel reduces platelet interactions with PMN and MN. Through these effects prasugrel may curb platelet-mediated inflammatory responses.


Assuntos
Anti-Inflamatórios/farmacologia , Plaquetas/efeitos dos fármacos , Mediadores da Inflamação/sangue , Leucócitos/efeitos dos fármacos , Piperazinas/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Choque Séptico/tratamento farmacológico , Tiofenos/farmacologia , Animais , Aspirina/farmacologia , Biomarcadores/sangue , Plaquetas/imunologia , Modelos Animais de Doenças , Regulação para Baixo , Humanos , Leucócitos/imunologia , Antígeno de Macrófago 1/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/sangue , Adesividade Plaquetária/efeitos dos fármacos , Testes de Função Plaquetária , Cloridrato de Prasugrel , Selenoproteína P/sangue , Choque Séptico/sangue , Choque Séptico/imunologia , Tromboxano B2/sangue , Fator de Necrose Tumoral alfa/sangue
16.
Cancer Biol Ther ; 13(14): 1471-81, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22986232

RESUMO

In the search for new strategies to efficiently fight colorectal cancer, efforts are being increasingly focused on targeting regulatory signaling pathways involved in cancer-specific features. As a result, several studies have recently addressed the therapeutic potential of molecularly-targeted drugs capable of inhibiting the activity of protein kinases involved in relevant signaling cascades. Here we show that simultaneous inhibition of the DFG-in and DFG-out conformations of p38α by means of type-I and type-II inhibitors is beneficial to impair more efficiently its kinase activity. Moreover, we found that SB202190 (type-I) and sorafenib (type-II) synergize at the molecular and biological level, as co-treatment with these compounds enhances tumor growth inhibition and induction of apoptosis both in colorectal cancer cell lines and animal models. These results support the need to reconsider sorafenib as a therapeutic agent against colorectal cancer and provide new insights that underline the importance to elucidate the activity of protein kinase inhibitors for the treatment of colorectal carcinoma.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Imidazóis/farmacologia , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Niacinamida/análogos & derivados , Compostos de Fenilureia/farmacologia , Piridinas/farmacologia , Animais , Caspase 3/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Sinergismo Farmacológico , Feminino , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Niacinamida/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sorafenibe , Transplante Heterólogo
17.
Cell Cycle ; 10(17): 2937-45, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21862870

RESUMO

We have recently shown that the transcriptional coactivator PGC1α, a master regulator of mitochondrial biogenesis and function, is involved in the control of the intestinal epithelium cell fate. Furthermore, PGC1α protects against colon cancer formation by promoting ROS accumulation and, consequently, mitochondria-mediated apoptosis. Here we provide an additional mechanistic insight into the tumor suppressor activity of PGC1α showing that its pro-apoptotic effect is mediated by Bax. In fact, PGC1α overexpression in HCT116 Bax (-/-) colorectal cancer cells stimulates mitochondrial production and activity, but it fails to induce cell death as well as to oppose tumor growth in the xenograft model. The lack of ROS accumulation in the Bax (-/-) cells strengthens our view that the PGC1α-induced oxidative burst represents one of the main apoptosis-driving factors in colorectal cancer cells.


Assuntos
Apoptose , Neoplasias do Colo/patologia , Proteínas de Choque Térmico/metabolismo , Fatores de Transcrição/metabolismo , Proteína X Associada a bcl-2/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Neoplasias do Colo/metabolismo , DNA Mitocondrial/análise , Células HCT116 , Humanos , Camundongos , Camundongos Nus , Mitocôndrias/genética , Mitocôndrias/metabolismo , PPAR gama/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Espécies Reativas de Oxigênio/metabolismo , Explosão Respiratória , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Br J Nutr ; 99(2): 240-7, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17845735

RESUMO

Lower CVD incidence is reported in Asian populations consuming soya-containing food. As polymorphonuclear leukocytes (PMN) are involved in the risk of CVD, we investigated the modulatory effect of soya isoflavones on several PMN functions and their molecular mechanisms in vitro. PMN, isolated from blood from healthy subjects, were tested upon activation with 1 microm- n-formyl-methyl-leucyl-phenylalanine (fMLP) for superoxide anion production (ferric cytochrome c reduction) and released elastase (chromogenic test). PMN homotypic aggregates stimulated by fMLP or P-selectin in dynamic conditions were detected by optical microscopy. PMN, mixed with thrombin-activated, washed platelets, formed cell aggregates, measured by flow cytometry. Phosphorylation of Pyk2, a focal adhesion kinase, was studied by immunoprecipitation and immunoblotting with specific antibodies. Genistein, daidzein and equol inhibited superoxide anion production (IC50 0.25 (sem 0.1), 21.0 (sem 4.2) and 13.0 (sem 2.8) microm, respectively); the release of elastase was prevented by genistein (IC50 63 (sem 17) microm). PMN homotypic aggregates, stimulated by fMLP, were significantly reduced (24 (sem 12) and 51 (sem 14) % of control) by 100 microm genistein and equol. P-selectin-induced aggregates were reduced to 19 (sem 6), 44 (sem 10) and 28 (sem 9) % of control by 100 microm genistein, daidzein and equol, respectively. Genistein, daidzein and equol also significantly reduced mixed platelet-PMN aggregates (IC50 4.0 (sem 0.9), 57 (sem 6) and 66 (sem 23) microm, respectively). In PMN challenged by fMLP or P-selectin, activation of Pyk2 was prevented by isoflavones. The cardioprotective effect of soya-containing food might be linked to reduction of PMN activation and PMN-platelet interaction, novel targets for the biological effects of soya isoflavones.


Assuntos
Glycine max/química , Isoflavonas/farmacologia , Neutrófilos/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Equol , Quinase 2 de Adesão Focal/metabolismo , Genisteína/farmacologia , Humanos , Antígeno de Macrófago 1/metabolismo , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/fisiologia , Selectina-P/farmacologia , Elastase Pancreática/metabolismo , Fosforilação , Adesividade Plaquetária/efeitos dos fármacos , Superóxidos/metabolismo
19.
J Am Coll Cardiol ; 49(22): 2182-90, 2007 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-17543638

RESUMO

OBJECTIVES: This study evaluated the impact of hyperhomocysteinemia (HHcy) on the CD40/CD40 ligand (CD40L) dyad in vivo and in vitro. BACKGROUND: Hyperhomocysteinemia is associated with an increased incidence of atherothrombosis, although the molecular mechanisms of this association are incompletely defined. The CD40L pair triggers inflammatory signals in cells of the vascular wall, representing a major pathogenetic pathway of atherosclerosis. METHODS: We used a commercially available enzyme-linked immunosorbent assay kit to evaluate circulating levels of soluble (s) CD40L in 24 patients with HHcy and 24 healthy subjects. We also used real-time polymerase chain reaction and flow cytometry to determine expression levels of CD40 and vascular cell adhesion molecule (VCAM)-1 in human umbilical vein endothelial cells (HUVECs) and of CD40L in human platelets. RESULTS: The sCD40L levels were significantly increased in HHcy patients (median [interquartile range] 8.0 [0.7 to 10.5] ng/ml vs. 2.1 [1.9 to 2.3] ng/ml, p = 0.0001). Positive correlations were noted between log sCD40L and log homocysteine (Hcy) (R = 0.68, p < 0.0001) or log sVCAM-1 (R = 0.41, p < 0.005). Homocysteine significantly stimulated CD40 mRNA expression in HUVECs (p = 0.033). Consistently, 24-h exposure to Hcy increased the percentage of CD40-expressing cells (p = 0.00025). Homocysteine also significantly enhanced CD40L expression in platelets (p = 0.025) to a comparable extent as that of thrombin. Notably, Hcy increased VCAM-1 protein expression induced by CD40L in HUVECs (p = 0.0046). CONCLUSIONS: The present results uncover a potential molecular target of Hcy, namely the CD40/CD40L dyad. Collectively, they indicate that upregulation of CD40/CD40L signaling may represent a link between HHcy and an increased risk of cardiovascular disease.


Assuntos
Antígenos CD40/sangue , Ligante de CD40/sangue , Homocisteína/fisiologia , Adulto , Plaquetas/metabolismo , Antígenos CD40/genética , Antígenos CD40/fisiologia , Ligante de CD40/genética , Ligante de CD40/fisiologia , Células Endoteliais/metabolismo , Feminino , Homocisteína/sangue , Homocisteína/genética , Humanos , Masculino , Metilenotetra-Hidrofolato Redutase (NADPH2)/sangue , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Pessoa de Meia-Idade , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
20.
Blood ; 109(6): 2461-9, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17095622

RESUMO

Polymorphonuclear leukocyte (PMN)-platelet interactions at sites of vascular damage contribute to local and systemic inflammation. We sought to determine the role of "outside-in" signaling by Src-family tyrosine kinases (SFKs) in the regulation of alphaMbeta2-integrin-dependent PMN recruitment by activated platelets under (patho)physiologic conditions. Activation-dependent epitopes in beta2 integrin were exposed at the contact sites between PMNs and platelets and were abolished by SFK inhibitors. PMNs from alphaMbeta2(-/-), hck(-/-)fgr(-/-), and hck(-/-)fgr(-/-)lyn(-/-) mice had an impaired capacity to adhere with activated platelets in suspension. Phosphorylation of Pyk2 accompanied PMN adhesion to platelets and was blocked by inhibition as well as by genetic deletion of alphaMbeta2 integrin and SFKs. A Pyk2 inhibitor reduced platelet-PMN adhesion, indicating that Pyk2 may be a downstream effector of SFKs. Analysis of PMN-platelet interactions under flow revealed that SFK signaling was required for alphaMbeta2-mediated shear-resistant adhesion of PMNs to adherent platelets, but was dispensable for P-selectin-PSGL-1-mediated recruitment and rolling. Finally, SFK activity was required to support PMN accumulation along adherent platelets at the site of vascular injury, in vivo. These results definitely establish a role for SFKs in PMN recruitment by activated platelets and suggest novel targets to disrupt the pathophysiologic consequences of platelet-leukocyte interactions in vascular disease.


Assuntos
Plaquetas/citologia , Plaquetas/enzimologia , Neutrófilos/citologia , Neutrófilos/enzimologia , Ativação Plaquetária , Quinases da Família src/metabolismo , Animais , Antígeno CD11b/metabolismo , Antígenos CD18/metabolismo , Adesão Celular , Quinase 2 de Adesão Focal/metabolismo , Humanos , Antígeno de Macrófago 1/metabolismo , Camundongos , Camundongos Knockout , Selectina-P/metabolismo , Fosfotirosina/metabolismo , Quinases da Família src/deficiência , Quinases da Família src/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA