Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Atherosclerosis ; 185(1): 32-8, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15993884

RESUMO

Lipid accumulation and inflammation are key hallmarks of the atherosclerotic plaque and macrophage uptake of oxidized low-density lipoprotein (oxLDL) is believed to drive these processes. Initial experiments show that supernatants from oxLDL treated macrophages could induce IL-1beta production in naïve macrophages. To search for potential paracrine mediators that could mediate this effect a DNA microarray scan of oxLDL treated human macrophages was performed. This analysis revealed that oxLDL induced activation of heat shock protein (HSP) expression. HSPs have been implicated in the development of atherosclerosis, but the exact mechanisms for this is unclear. Extracellular heat shock protein 70 (HSP70) has been shown to elicit a pro-inflammatory cytokine response in monocytes and could therefore be a potential paracrine pro-inflammatory mediator. After 24 h of oxLDL treatment there was a significant increase of HSP70 concentrations in supernatants from oxLDL treated macrophages (oxLDLsup) compared to untreated controls (P<0.05). OxLDLsup could induce both interleukin (IL)-1beta and IL-12 secretion in naïve macrophages. We also demonstrate that the effect of oxLDLsup on cytokine production and release could be blocked by inhibition of HSP70 transcription or secretion or by the use of HSP70 neutralizing antibodies. This suggests that extracellular HSP70 can mediate pro-inflammatory changes in macrophages in response to oxLDL.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Interleucina-1/biossíntese , Lipoproteínas LDL/farmacologia , Macrófagos/metabolismo , DNA/genética , Ensaio de Imunoadsorção Enzimática , Líquido Extracelular/metabolismo , Citometria de Fluxo , Proteínas de Choque Térmico HSP70/genética , Humanos , Técnicas In Vitro , Líquido Intracelular/metabolismo , Macrófagos/efeitos dos fármacos , Masculino , Mutação , Oxirredução , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
Atherosclerosis ; 185(2): 282-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16046214

RESUMO

Using DNA microarray analysis, we found that human macrophages respond to oxidized low-density lipoprotein (oxLDL) by activating the antioxidative glutathione and thioredoxin systems. Several genes of the glutathione and thioredoxin systems were expressed at high levels in macrophages when compared to 80 other human tissues and cell types, indicating that these systems may be of particular importance in macrophages. The up-regulation of three genes in these systems, thioredoxin (P < 0.005), thioredoxin reductase 1 (P < 0.001) and glutathione reductase (P < 0.001) was verified with real-time RT-PCR, using human macrophages from 10 healthy donors. To investigate the possible role of these antioxidative systems in the development of atherosclerosis, expression levels in macrophages from 15 subjects with atherosclerosis (12 men, 3 women) and 15 matched controls (12 men, 3 women) were analyzed using DNA microarrays. Two genes in the glutathione system Mn superoxide dismutase (P < 0.05) and catalase (P < 0.05) differed in expression between the groups. We conclude that macrophage uptake of oxidized LDL induces a coordinated up-regulation of genes of the glutathione and thioredoxin systems, suggesting that these systems may participate in the cellular defense against oxidized LDL and possibly modulate the development of atherosclerosis.


Assuntos
Glutationa/metabolismo , Lipoproteínas LDL/farmacologia , Macrófagos/metabolismo , Tiorredoxinas/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Feminino , Expressão Gênica , Glutationa/genética , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Oxirredução , Estresse Oxidativo , Via de Pentose Fosfato , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tiorredoxinas/genética , Regulação para Cima
3.
BMC Cardiovasc Disord ; 5: 25, 2005 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-16122381

RESUMO

BACKGROUND: The protective role of high-density lipoprotein (HDL) in the cardiovascular system is related to its role in the reverse transport of cholesterol from the arterial wall to the liver for subsequent excretion via the bile. Scavenger receptor class B type I (SR-BI) binds HDL and mediates selective uptake of cholesterol ester and cellular efflux of cholesterol to HDL. The role of SR-BI in atherosclerosis has been well established in murine models but it remains unclear whether SR-BI plays an equally important role in atherosclerosis in humans. The aim of this study was to investigate the expression of SR-BI and its isoforms in human macrophages and atherosclerotic plaques. METHODS: The effect of hypoxia and minimally modified low-density lipoprotein (mmLDL), two proatherogenic stimuli, on SR-BI expression was studied in human monocyte-derived macrophages from healthy subjects using real-time PCR. In addition, SR-BI expression was determined in macrophages obtained from subjects with atherosclerosis (n = 15) and healthy controls (n = 15). Expression of SR-BI isoforms was characterized in human atherosclerotic plaques and macrophages using RT-PCR and DNA sequencing. RESULTS: SR-BI expression was decreased in macrophages after hypoxia (p < 0.005). In contrast, SR-BI expression was increased by exposure to mmLDL (p < 0.05). There was no difference in SR-BI expression in macrophages from patients with atherosclerosis compared to controls. In both groups, SR-BI expression was increased by exposure to mmLDL (p < 0.05). Transcripts corresponding to SR-BI and SR-BII were detected in macrophages. In addition, a third isoform, referred to as SR-BIII, was discovered. All three isoforms were also expressed in human atherosclerotic plaque. Compared to the other isoforms, the novel SR-BIII isoform was predicted to have a unique intracellular C-terminal domain containing 53 amino acids. CONCLUSION: We conclude that SR-BI is regulated by proatherogenic stimuli in humans. However, we found no differences between subjects with atherosclerosis and healthy controls. This indicates that altered SR-BI expression is not a common cause of atherosclerosis. In addition, we identified SR-BIII as a novel isoform expressed in human macrophages and in human atherosclerotic plaques.


Assuntos
Macrófagos/metabolismo , Receptores Depuradores Classe B/metabolismo , Adulto , Processamento Alternativo , Motivos de Aminoácidos , Sequência de Aminoácidos , Aterosclerose/etiologia , Aterosclerose/metabolismo , Sequência de Bases , Hipóxia Celular , Células Cultivadas , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Lipoproteínas LDL/farmacologia , Proteínas de Membrana Lisossomal/química , Proteínas de Membrana Lisossomal/genética , Proteínas de Membrana Lisossomal/metabolismo , Macrófagos/efeitos dos fármacos , Masculino , Dados de Sequência Molecular , Isoformas de Proteínas , RNA Mensageiro/metabolismo , Receptores Depuradores , Receptores Depuradores Classe B/química , Receptores Depuradores Classe B/genética , Sialoglicoproteínas/química , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Domínios de Homologia de src
4.
Atherosclerosis ; 177(2): 287-90, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15530901

RESUMO

Identification of cell and tissue specific genes may provide novel insights to signaling systems and functions. Macrophages play a key role in many diseases including atherosclerosis. Using DNA microarrays we compared the expression of approximately 10,000 genes in 56 human tissues and identified 23 genes with predominant expression in macrophages. The identified genes include both genes known to be macrophage specific and genes previously not well described in this cell type. Tissue distribution of two genes, liver X receptor (LXR) alpha and interleukin-1 receptor antagonist (IL1RN), was verified by real-time RT-PCR. We conclude that comparison of expression profiles from a large number of tissues can be used to identify genes that are predominantly expressed in certain tissues. Identification of novel macrophage specific genes may increase our understanding of the role of this cell in different diseases.


Assuntos
Macrófagos/fisiologia , Receptores Citoplasmáticos e Nucleares/genética , Sialoglicoproteínas/genética , Proteínas de Ligação a DNA , Humanos , Proteína Antagonista do Receptor de Interleucina 1 , Receptores X do Fígado , Macrófagos/química , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Nucleares Órfãos , Distribuição Tecidual
5.
Arterioscler Thromb Vasc Biol ; 24(11): 2040-5, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15358603

RESUMO

OBJECTIVE: Macrophage-mediated oxidation of low-density lipoprotein (LDL) by enzymes, such as the lipoxygenases, is considered of major importance for the formation of oxidized LDL during atherogenesis. Macrophages have been identified in hypoxic areas in atherosclerotic plaques. METHODS AND RESULTS: To investigate the role of hypoxia in macrophage-mediated LDL oxidation, we incubated human monocyte-derived macrophages with LDL under normoxic (21% O2) or hypoxic (0% O2) conditions. The results showed that hypoxic macrophages oxidized LDL to a significantly higher extent than normoxic cells. Interestingly, the mRNA and protein expression of 15-lipoxygenase-2 (15-LOX-2) as well as the activity of this enzyme are elevated in macrophages incubated at hypoxia. Both the unspliced 15-LOX-2 and the spliced variant 15-LOX-2sv-a are found in macrophages. In addition, 15-LOX-2 was identified in carotid plaques in some macrophage-rich areas but was only expressed at low levels in nondiseased arteries. CONCLUSIONS: In summary, these observations show for the first time that 15-LOX-2 is expressed in hypoxic macrophages and in atherosclerotic plaques and suggest that 15-LOX-2 may be one of the factors involved in macrophage-mediated LDL oxidation at hypoxia.


Assuntos
Araquidonato 15-Lipoxigenase/biossíntese , Hipóxia/enzimologia , Hipóxia/patologia , Lipoproteínas LDL/metabolismo , Macrófagos/enzimologia , Processamento Alternativo/genética , Araquidonato 15-Lipoxigenase/genética , Araquidonato 15-Lipoxigenase/imunologia , Arteriosclerose/enzimologia , Arteriosclerose/metabolismo , Arteriosclerose/patologia , Artérias Carótidas/enzimologia , Artérias Carótidas/patologia , Células Cultivadas , Ativação Enzimática/genética , Éxons/genética , Variação Genética/genética , Humanos , Imuno-Histoquímica/métodos , Macrófagos/citologia , Artéria Torácica Interna/fisiologia , Oxirredução , Deleção de Sequência/genética
6.
Atherosclerosis ; 170(2): 245-52, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14612204

RESUMO

Interleukin-8 (IL-8) is a chemotactic factor for T-lymphocytes and smooth muscle cells and may therefore have an important effect in atherogenesis. It is secreted from oxysterol-containing foam cells which have been found in hypoxic zones in atherosclerotic plaques. The aim of this study was to investigate the effect of hypoxia on the secretion of IL-8 by oxysterol-stimulated macrophages. Hypoxia enhances 25-hydroxycholesterol (25-OH-chol)-induced IL-8 secretion in human monocyte-derived macrophages. The effect is most pronounced when macrophages are incubated with low concentrations of 25-OH-chol. Both 25-OH-chol and hypoxia increases the intracellular level of the signalling molecule hydrogen peroxide (H(2)O(2)). This event coincided with an enhanced binding of the transcription factor c-jun to the IL-8 gene promoter and an increased IL-8 mRNA expression in hypoxic macrophages. These observations suggest that similar intracellular signalling pathways are used for both 25-OH-chol-induced IL-8 expression and hypoxia-induced IL-8 expression. Thus, hypoxia in atherosclerotic plaques may increase the secretion of IL-8 from oxysterol-containing foam cells, which subsequently may accelerate the progression of atherosclerosis.


Assuntos
Hipóxia Celular , Hidroxicolesteróis/farmacologia , Interleucina-8/metabolismo , Macrófagos/metabolismo , Complexos de ATP Sintetase/antagonistas & inibidores , Arteriosclerose/metabolismo , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Inibidores Enzimáticos/farmacologia , Humanos , Peróxido de Hidrogênio/metabolismo , Interleucina-8/genética , Oligomicinas/farmacologia , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , Fator de Transcrição AP-1/metabolismo , Vitamina E/farmacologia
7.
Atherosclerosis ; 169(2): 259-67, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12921977

RESUMO

Monocyte infiltration followed by differentiation into macrophages and accumulation of oxidised LDL (oxLDL) comprise early stages of atherosclerosis. Vascular endothelial growth factor (VEGF), which is upregulated by oxLDL, may contribute to atherogenesis through monocyte recruitment, increased vascular permeability and promotion of intraplaque vessels. The VEGF receptor-1 (VEGFR-1/Flt-1) mediates monocyte migration towards VEGF and regulates the levels of available VEGF through ligand-entrapment. In this study we investigated the effect of oxLDL on VEGFR-1 expression in human monocyte-derived macrophages. mRNA expression was estimated using RT-PCR, protein secretion was measured with ELISA and the amount of membrane-bound VEGFR-1 was analysed using flow cytometry analysis. Binding of transcription factors to the promoter was studied with EMSA. Incubation with oxLDL decreased VEGFR-1 mRNA expression in a time- and dose-dependent manner, followed by a subsequent decrease in protein secretion of VEGFR-1 and a reduction of the amount of receptor expressed on the cell surface. Furthermore, the PPARgamma agonists 9-hydroxy-(S)-10,12-octadecadienoic acid (9-HODE) and darglitazone also decreased VEGFR-1 mRNA expression. Incubation of macrophages with oxLDL or 9-HODE decreased binding of the transcription factor AP-1 (c-jun/ATF-2) to the VEGFR-1 promoter. Together, these data suggest that oxLDL decreases VEGFR-1 expression in macrophages, probably through a PPARgamma-dependent reduction in the AP-1 transcriptional activity. This implies that oxLDL has effects on the biological availability of VEGF, besides its direct effect on VEGF expression.


Assuntos
Lipoproteínas LDL/farmacologia , Macrófagos/química , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/análise , Fatores Ativadores da Transcrição , Proteínas Sanguíneas/metabolismo , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico , Interpretação Estatística de Dados , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Ácidos Linoleicos Conjugados/farmacologia , Monócitos , Oxirredução , Proteínas/metabolismo , RNA Mensageiro/análise , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tiazolidinedionas/farmacologia , Fator de Transcrição AP-1/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia
8.
Atherosclerosis ; 169(1): 71-6, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12860252

RESUMO

Accumulation of lipids and cholesterol by macrophages and subsequent transformation into foam cells are key features in development of atherosclerosis. Serum copper concentrations have been shown to be associated with cardiovascular disease. However, the mechanism behind the proatherogenic effect of copper is not clear. We used DNA microarrays to define the changes in gene expression profile in response to copper exposure of human macrophages. Expression monitoring by DNA microarray revealed 91 genes that were regulated. Copper increased the expression of seven cholesterogenic genes (3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) synthase, IPP isomerase, squalene synthase, squalene epoxidase, methyl sterol oxidase, H105e3 mRNA and sterol-C5-desaturase) and low-density lipoprotein receptor (LDL-R), and decreased the expression of CD36 and lipid binding proteins. The expression of LDL-R and HMG CoA reductase was also investigated using real time PCR. The expression of both of these genes was increased after copper treatment of macrophages (P<0.01 and P<0.01, respectively). We conclude that copper activates cholesterogenic genes in macrophages, which may provide a mechanism for the association between copper and atherosclerosis. The effect of copper on cholesterogenic genes may also have implications for liver steatosis in early stages of Wilson's disease.


Assuntos
Colesterol/genética , Sulfato de Cobre/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/metabolismo , Perfilação da Expressão Gênica , Humanos , Hidroximetilglutaril-CoA Redutases/genética , Hidroximetilglutaril-CoA Redutases/metabolismo , Técnicas In Vitro , Macrófagos/efeitos dos fármacos , Reação em Cadeia da Polimerase , Receptores de LDL/genética , Receptores de LDL/metabolismo
9.
Cell Immunol ; 220(2): 83-95, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12657243

RESUMO

Lactoferrin, a glycoprotein present in milk, mucosal secretions and neutrophils contributes to host defense. We have previously shown that orally given milk lactoferrin (LF) mediates anti-infectious and anti-inflammatory activities in vivo. Moreover, we have shown that LF could inhibit the LPS-induced IL-6 secretion in a human monocytic cell line, THP-1. This observation was expanded in the present study investigating the capacity of LF to inhibit cytokine mRNA expression and the involvement of nuclear transcription factor kappa B (NF-kappa B). Cells (THP-1 and Mono Mac 6 monocytic cell lines) were stimulated with Escherichia coli LPS (5-10 ng/10(6) cells) and LF was added (50-500 microg/10(6) cells) 30 min before, or after the LPS addition. By a semiquantitative RT-PCR lower levels of TNF-alpha, IL-1 beta, IL-6, and IL-8 mRNA expression were detected at the peak of the expression in THP-1 cells treated with LF. The reduction in the cytokine expression was followed by a similar reduction in the secreted cytokines as analyzed by ELISA. LF down-regulated also the IL-10 secretion (detected only in LPS-stimulated Mono Mac 6 cells). A similar level of inhibition of these cytokines was detected regardless of the time at which LF was added to the cells in relation to LPS. In addition, LF was internalized into cells and detected in the nucleoli as determined by immunostaining and immunofluorescence. Moreover, by electrophoretic mobility shift assay (EMSA) analysis LF decreased the LPS-induced binding of NF-kappa B to the TNF-alpha promoter. The results show that LF down-regulates the LPS-induced cytokine production in monocytic cells. The inhibitory mechanism is suggested to involve the interference of LF with NF-kappa B activation.


Assuntos
Citocinas/biossíntese , Lactoferrina/imunologia , Monócitos/imunologia , NF-kappa B/imunologia , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Regulação para Baixo/imunologia , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Lactoferrina/metabolismo , Lipopolissacarídeos/imunologia , Microscopia Confocal , Monócitos/metabolismo , NF-kappa B/metabolismo , Regiões Promotoras Genéticas/imunologia , RNA Mensageiro/química , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...