Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Circ Res ; 121(1): 19-30, 2017 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-28438779

RESUMO

RATIONALE: Vascular calcification is a process similar to bone formation leading to an inappropriate deposition of calcium phosphate minerals in advanced atherosclerotic plaques. Monocyte-derived macrophages, located in atherosclerotic lesions and presenting heterogeneous phenotypes, from classical proinflammatory M1 to alternative anti-inflammatory M2 macrophages, could potentially display osteoclast-like functions. OBJECTIVE: To characterize the phenotype of macrophages located in areas surrounding the calcium deposits in human atherosclerotic plaques. METHODS AND RESULTS: Macrophages near calcium deposits display an alternative phenotype being both CD68 and mannose receptor-positive, expressing carbonic anhydrase type II, but relatively low levels of cathepsin K. In vitro interleukin-4-polarization of human primary monocytes into macrophages results in lower expression and activity of cathepsin K compared with resting unpolarized macrophages. Moreover, interleukin-4 polarization lowers expression levels of the osteoclast transcriptional activator nuclear factor of activated T cells type c-1, associated with increased gene promoter levels of the transcriptional repression mark H3K27me3 (histone 3 lysine 27 trimethylation). Despite higher expression of the receptor activator of nuclear factor κB receptor, receptor activator of nuclear factor κB ligand/macrophage colony-stimulating factor induction of nuclear factor of activated T cells type c-1 and cathepsin K expression is defective in these macrophages because of reduced Erk/c-fos-mediated downstream signaling resulting in impaired bone resorption capacity. CONCLUSIONS: These results indicate that macrophages surrounding calcium deposits in human atherosclerotic plaques are phenotypically defective being unable to resorb calcification.


Assuntos
Reabsorção Óssea/metabolismo , Macrófagos/metabolismo , Osteoclastos/metabolismo , Placa Aterosclerótica/metabolismo , Ligante RANK/metabolismo , Calcificação Vascular/metabolismo , Reabsorção Óssea/patologia , Células Cultivadas , Humanos , Microdissecção e Captura a Laser/métodos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Macrófagos/patologia , Osteoclastos/patologia , Placa Aterosclerótica/patologia , Calcificação Vascular/patologia
2.
Immunol Rev ; 262(1): 153-66, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25319333

RESUMO

Initiation and progression of atherosclerosis depend on local inflammation and accumulation of lipids in the vascular wall. Although many cells are involved in the development and progression of atherosclerosis, macrophages are fundamental contributors. For nearly a decade, the phenotypic heterogeneity and plasticity of macrophages has been studied. In atherosclerotic lesions, macrophages are submitted to a large variety of micro-environmental signals, such as oxidized lipids and cytokines, which influence the phenotypic polarization and activation of macrophages resulting in a dynamic plasticity. The macrophage phenotype spectrum is characterized, at the extremes, by the classical M1 macrophages induced by T-helper 1 (Th-1) cytokines and by the alternative M2 macrophages induced by Th-2 cytokines. M2 macrophages can be further classified into M2a, M2b, M2c, and M2d subtypes. More recently, additional plaque-specific macrophage phenotypes have been identified, termed as Mox, Mhem, and M4. Understanding the mechanisms and functional consequences of the phenotypic heterogeneity of macrophages will contribute to determine their potential role in lesion development and plaque stability. Furthermore, research on macrophage plasticity could lead to novel therapeutic approaches to counteract cardiovascular diseases such as atherosclerosis. The present review summarizes our current knowledge on macrophage subsets in atherosclerotic plaques and mechanism behind the modulation of the macrophage phenotype.


Assuntos
Aterosclerose/etiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Fenótipo , Animais , Aterosclerose/patologia , Movimento Celular , Humanos , Ativação de Macrófagos , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Fatores de Transcrição/metabolismo
3.
Biochim Biophys Acta ; 1842(4): 584-93, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24361460

RESUMO

BACKGROUND: Obesity, characterized by low grade inflammation, induces adipose tissue macrophage (ATM) infiltration in white adipose tissue (AT) in both humans and rodents, thus contributing to insulin resistance. Previous studies have shown altered prolactin secretion in obesity, however, studies linking ATM infiltration and prolactin (PRL) secretion to the pathogenesis of the metabolic syndrome, obesity and diabetes are lacking. METHODS/RESULTS: In vivo, qPCR and Western blot analysis demonstrated that prolactin expression was increased in AT of obese rats and also in human AT from obese, obese pre-diabetic and obese diabetic compared to lean counterparts. Immunohistochemistry of obese rat and human AT sections demonstrated a specific expression of prolactin in macrophages. In vitro, we demonstrated that hyperglycemia and inflammation stimulated macrophages (human THP-1 cell line and sorted rat ATM) to express PRL, when challenged with different glucose concentrations with or without IL1ß. In in vivo and in vitro experiments, we assessed the expression of Pit-1 (PRL-specific transcription factor) and found that its expression was parallel to PRL expression. CONCLUSIONS: In this study, we show that rodent and human macrophages synthesize prolactin in response to inflammation and high glucose concentrations. GENERAL SIGNIFICANCE: Our data shed new light on the potential role of macrophages in the physiopathology of diabesity via the PRL expression and on its expression mechanism and regulation.


Assuntos
Tecido Adiposo/fisiologia , Diabetes Mellitus/etiologia , Inflamação/metabolismo , Macrófagos/fisiologia , Obesidade/complicações , Prolactina/fisiologia , Animais , Células Cultivadas , Humanos , Obesidade/sangue , Prolactina/sangue , Ratos , Ratos Wistar , Fator de Transcrição Pit-1/análise
4.
Biochim Biophys Acta ; 1841(6): 827-35, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24603323

RESUMO

Liver X receptors (LXRα and LXRß) are key transcription factors in cholesterol metabolism that regulate cholesterol biosynthesis/efflux and bile acid metabolism/excretion in the liver and numerous organs. In macrophages, LXR signaling modulates cholesterol handling and the inflammatory response, pathways involved in atherosclerosis. Since regulatory pathways of LXR transcription control are well understood, in the present study we aimed at identifying post-transcriptional regulators of LXR activity. MicroRNAs (miRs) are such post-transcriptional regulators of genes that in the canonical pathway mediate mRNA inactivation. In silico analysis identified miR-206 as a putative regulator of LXRα but not LXRß. Indeed, as recently shown, we found that miR-206 represses LXRα activity and expression of LXRα and its target genes in hepatic cells. Interestingly, miR-206 regulates LXRα differently in macrophages. Stably overexpressing miR-206 in THP-1 human macrophages revealed an up-regulation and miR-206 knockdown led to a down-regulation of LXRα and its target genes. In support of these results, bone marrow-derived macrophages (BMDMs) from miR-206 KO mice also exhibited lower expression of LXRα target genes. The physiological relevance of these findings was proven by gain- and loss-of-function of miR-206; overexpression of miR-206 enhanced cholesterol efflux in human macrophages and knocking out miR-206 decreased cholesterol efflux from MPMs. Moreover, we show that miR-206 expression in macrophages is repressed by LXRα activation, while oxidized LDL and inflammatory stimuli profoundly induced miR-206 expression. We therefore propose a feed-back loop between miR-206 and LXRα that might be part of an LXR auto-regulatory mechanism to fine tune LXR activity.


Assuntos
Colesterol/metabolismo , Metabolismo dos Lipídeos/genética , MicroRNAs/genética , Receptores Nucleares Órfãos/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/patologia , Colesterol/genética , Regulação da Expressão Gênica , Células Hep G2 , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Receptores X do Fígado , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Receptores Nucleares Órfãos/genética , Transdução de Sinais
5.
Circ Res ; 113(11): 1196-205, 2013 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-24036496

RESUMO

RATIONALE: In atherosclerotic plaques, iron preferentially accumulates in macrophages where it can exert pro-oxidant activities. OBJECTIVE: The objective of this study was, first, to better characterize the iron distribution and metabolism in macrophage subpopulations in human atherosclerotic plaques and, second, to determine whether iron homeostasis is under the control of nuclear receptors, such as the liver X receptors (LXRs). METHODS AND RESULTS: Here we report that iron depots accumulate in human atherosclerotic plaque areas enriched in CD68 and mannose receptor (MR)-positive (CD68(+)MR(+)) alternative M2 macrophages. In vitro IL-4 polarization of human monocytes into M2 macrophages also resulted in a gene expression profile and phenotype favoring iron accumulation. However, M2 macrophages on iron exposure acquire a phenotype favoring iron release, through a strong increase in ferroportin expression, illustrated by a more avid oxidation of extracellular low-density lipoprotein by iron-loaded M2 macrophages. In line, in human atherosclerotic plaques, CD68(+)MR(+) macrophages accumulate oxidized lipids, which activate LXRα and LXRß, resulting in the induction of ABCA1, ABCG1, and apolipoprotein E expression. Moreover, in iron-loaded M2 macrophages, LXR activation induces nuclear factor erythroid 2-like 2 expression, thereby increasing ferroportin expression, which, together with a decrease of hepcidin mRNA levels, promotes iron export. CONCLUSIONS: These data identify a role for M2 macrophages in iron handling, a process regulated by LXR activation.


Assuntos
Ferro/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Receptores Nucleares Órfãos/fisiologia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Apolipoproteínas E/metabolismo , Transporte Biológico/fisiologia , Células Cultivadas , Homeostase/fisiologia , Humanos , Técnicas In Vitro , Lectinas Tipo C/metabolismo , Receptores X do Fígado , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Fenótipo , Receptores de Superfície Celular/metabolismo
6.
Handb Exp Pharmacol ; 224: 617-30, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25523004

RESUMO

Dyslipidaemia is a major risk factor for cardiovascular diseases. Pharmacological lowering of LDL-C levels using statins reduces cardiovascular risk. However, a substantial residual risk persists especially in patients with type 2 diabetes mellitus. Because of the inverse association observed in epidemiological studies of HDL-C with the risk for cardiovascular diseases, novel therapeutic strategies to raise HDL-C levels or improve HDL functionality are developed as complementary therapy for cardiovascular diseases. However, until now most therapies targeting HDL-C levels failed in clinical trials because of side effects or absence of clinical benefits. This chapter will highlight the emerging small molecules currently developed and tested in clinical trials to pharmacologically modulate HDL-C and functionality including new CETP inhibitors (anacetrapib, evacetrapib), novel PPAR agonists (K-877, CER-002, DSP-8658, INT131 and GFT505), LXR agonists (ATI-111, LXR-623, XL-652) and RVX-208.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Desenho de Fármacos , Dislipidemias/tratamento farmacológico , Hipolipemiantes/uso terapêutico , Animais , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Dislipidemias/complicações , Dislipidemias/metabolismo , Humanos , Hipolipemiantes/química , Receptores X do Fígado , Terapia de Alvo Molecular , Receptores Nucleares Órfãos/agonistas , Receptores Nucleares Órfãos/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Quinazolinas/uso terapêutico , Quinazolinonas , Transdução de Sinais/efeitos dos fármacos
7.
J Am Soc Nephrol ; 25(11): 2658-68, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24854267

RESUMO

Endothelial dysfunction begins in early CKD and contributes to cardiovascular mortality. HDL is considered antiatherogenic, but may have adverse vascular effects in cardiovascular disease, diabetes, and inflammatory conditions. The effect of renal failure on HDL properties is unknown. We studied the endothelial effects of HDL isolated from 82 children with CKD stages 2-5 (HDL(CKD)), who were free of underlying inflammatory diseases, diabetes, or active infections. Compared with HDL from healthy children, HDL(CKD) strongly inhibited nitric oxide production, promoted superoxide production, and increased vascular cell adhesion molecule-1 expression in human aortic endothelial cells, and reduced cholesterol efflux from macrophages. The effects on endothelial cells correlated with CKD grade, with the most profound changes induced by HDL from patients on dialysis, and partial recovery observed with HDL isolated after kidney transplantation. Furthermore, the in vitro effects on endothelial cells associated with increased aortic pulse wave velocity, carotid intima-media thickness, and circulating markers of endothelial dysfunction in patients. Symmetric dimethylarginine levels were increased in serum and fractions of HDL from children with CKD. In a longitudinal follow-up of eight children undergoing kidney transplantation, HDL-induced production of endothelial nitric oxide, superoxide, and vascular cell adhesion molecule-1 in vitro improved significantly at 3 months after transplantation, but did not reach normal levels. These results suggest that in children with CKD without concomitant disease affecting HDL function, HDL dysfunction begins in early CKD, progressing as renal function declines, and is partially reversed after kidney transplantation.


Assuntos
HDL-Colesterol/sangue , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/mortalidade , Doenças Vasculares/sangue , Doenças Vasculares/mortalidade , Adolescente , Arginina/análogos & derivados , Arginina/sangue , Biomarcadores/sangue , Criança , LDL-Colesterol/sangue , Endotélio Vascular/metabolismo , Feminino , Humanos , Transplante de Rim/mortalidade , Masculino , Óxido Nítrico/metabolismo , Diálise Renal/mortalidade , Insuficiência Renal Crônica/cirurgia , Triglicerídeos/sangue , Doenças Vasculares/cirurgia
8.
FASEB J ; 27(4): 1519-31, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23303209

RESUMO

11ß-Hydroxysteroid dehydrogenase type-1 (11ß-HSD1) converts inert cortisone into active cortisol, amplifying intracellular glucocorticoid action. 11ß-HSD1 deficiency improves cardiovascular risk factors in obesity but exacerbates acute inflammation. To determine the effects of 11ß-HSD1 deficiency on atherosclerosis and its inflammation, atherosclerosis-prone apolipoprotein E-knockout (ApoE-KO) mice were treated with a selective 11ß-HSD1 inhibitor or crossed with 11ß-HSD1-KO mice to generate double knockouts (DKOs) and challenged with an atherogenic Western diet. 11ß-HSD1 inhibition or deficiency attenuated atherosclerosis (74-76%) without deleterious effects on plaque structure. This occurred without affecting plasma lipids or glucose, suggesting independence from classical metabolic risk factors. KO plaques were not more inflamed and indeed had 36% less T-cell infiltration, associated with 38% reduced circulating monocyte chemoattractant protein-1 (MCP-1) and 36% lower lesional vascular cell adhesion molecule-1 (VCAM-1). Bone marrow (BM) cells are key to the atheroprotection, since transplantation of DKO BM to irradiated ApoE-KO mice reduced atherosclerosis by 51%. 11ß-HSD1-null macrophages show 76% enhanced cholesterol ester export. Thus, 11ß-HSD1 deficiency reduces atherosclerosis without exaggerated lesional inflammation independent of metabolic risk factors. Selective 11ß-HSD1 inhibitors promise novel antiatherosclerosis effects over and above their benefits for metabolic risk factors via effects on BM cells, plausibly macrophages.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 1/deficiência , Aterosclerose/metabolismo , Medula Óssea/metabolismo , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/antagonistas & inibidores , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/metabolismo , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Aterosclerose/genética , Medula Óssea/efeitos dos fármacos , Glucocorticoides/metabolismo , Camundongos , Camundongos Knockout , Fatores de Risco , Molécula 1 de Adesão de Célula Vascular/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 33(6): 1127-34, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23640494

RESUMO

Macrophages are plastic and versatile cells adapting their function/phenotype to the microenvironment. Distinct macrophage subpopulations with different functions, including classically (M1) and (M2) activated macrophages, have been described. Reciprocal skewing of macrophage polarization between the M1 and M2 state is a process modulated by transcription factors, such as the nuclear peroxisome proliferator-activated receptors. However, whether the estrogen/estrogen receptor pathways control the balance between M1/M2 macrophages is only partially understood. Estrogen-dependent effects on the macrophage system may be regarded as potential targets of pharmacological approaches to protect postmenopausal women from the elevated risk of cardiovascular disease.


Assuntos
Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/fisiopatologia , Estrogênios/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Comunicação Celular/imunologia , Comunicação Celular/fisiologia , Progressão da Doença , Estrogênios/genética , Feminino , Humanos , Imunidade Inata/fisiologia , Inflamação/imunologia , Inflamação/fisiopatologia , Fenótipo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 33(2): 431-8, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23241402

RESUMO

OBJECTIVE: Abdominal aortic aneurysms (AAAs), dilations of the infrarenal aorta, are characterized by inflammation and oxidative stress. We previously showed increased levels of peroxiredoxin-1 (PRDX-1) in macrophages cultured from AAA patients. The purpose of the study was to determine which subpopulation of macrophages is present in AAAs and is involved in upregulation of PRDX-1 in aneurysmal disease. METHODS AND RESULTS: This study used immunohistochemistry with antibodies against CD68 and mannose receptor (MR) to determine the subtype of macrophages in AAA tissue samples (n=33); laser capture microdissection to isolate each subtype; and quantitative-reverse transcriptase-polymerase chain reaction, Western blot, and ELISA to assess PRDX-1 mRNA and PRDX-1protein levels in both types. Proinflammatory CD68(+)MR(-) macrophages predominated in adventitial tissue, whereas the intraluminal thrombus contained CD68(+)MR(+) macrophages. The presence of lipids and iron-containing deposits confirmed their phagocytic phenotype. Laser capture microdissection-isolated CD68(+)MR(-) and CD68(+)MR(+) macrophages, characterized by quantitative-reverse transcriptase-polymerase chain reaction (TNF, IL1B, MRC1, and CCL18) and Western blot (stabilin and hemoglobin), validated the microdissected subtypes. PRDX-1 expression was colocalized with CD68(+)MR(-) macrophages. PRDX-1 mRNA and PRDX-1 protein were both more abundant in CD68(+)MR(-) than CD68(+)MR(+) macrophages in AAA. CONCLUSIONS: These findings suggest that the proteins or mRNAs expressed by the proinflammatory CD68(+)MR(-) macrophages may contribute to aneurysmal pathology.


Assuntos
Antígenos CD/análise , Antígenos de Diferenciação Mielomonocítica/análise , Aorta Abdominal/enzimologia , Aneurisma da Aorta Abdominal/enzimologia , Mediadores da Inflamação/análise , Macrófagos/enzimologia , Peroxirredoxinas/metabolismo , Aorta Abdominal/imunologia , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/imunologia , Aneurisma da Aorta Abdominal/patologia , Biomarcadores/análise , Western Blotting , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Regulação Enzimológica da Expressão Gênica , Humanos , Imuno-Histoquímica , Microdissecção e Captura a Laser , Macrófagos/imunologia , Macrófagos/patologia , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Peroxirredoxinas/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
11.
Circ J ; 78(8): 1775-81, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24998279

RESUMO

Atherosclerosis is the result of a chronic inflammatory response in the arterial wall related to uptake of low-density lipoprotein by macrophages and their subsequent transformation in foam cells. Monocyte-derived macrophages are the principal mediators of tissue homeostasis and repair, response to pathogens and inflammation. However, macrophages are a homogeneous cell population presenting a continuum phenotypic spectrum with, at the extremes, the classically Th-1 polarized M1 and alternatively Th-2 polarized M2 macrophage phenotypes, which have been well described. Moreover, M2 macrophages also present several subtypes often termed M2a, b, c and d, each of them expressing specific markers and exhibiting specialized properties. Macrophage plasticity is mirrored also in the atherosclerotic lesions, where different stimuli can influence the phenotype giving rise to a complex system of subpopulations, such as Mox, Mhem, M(Hb) and M4 macrophages. An abundant literature has described the potential modulators of the reciprocal skewing between pro-inflammatory M1 and anti-inflammatory M2 macrophages including lesion stage and localization, miRNA, transcription factors such as PPARγ, KLF4 and NR4A family members, high-density lipoproteins and plaque lipid content, pathways such as the rapamycin-mTOR1 pathway, molecules such as thioredoxin-1, infection by helminths and irradiation. We hope to provide an overview of the macrophage phenotype complexity in cardiovascular diseases, particularly atherosclerosis.


Assuntos
Aterosclerose/imunologia , Macrófagos/classificação , Macrófagos/imunologia , Animais , Aterosclerose/patologia , Humanos , Fator 4 Semelhante a Kruppel , Lipoproteínas HDL/imunologia , Macrófagos/patologia , MicroRNAs/imunologia , Serina-Treonina Quinases TOR/imunologia , Células Th1/imunologia , Células Th1/patologia , Células Th2/imunologia , Células Th2/patologia , Tiorredoxinas/imunologia , Fatores de Transcrição/imunologia
12.
J Biol Chem ; 287(26): 21904-13, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22511784

RESUMO

Obesity is associated with a significantly increased risk for cancer suggesting that adipose tissue dysfunctions might play a crucial role therein. Macrophages play important roles in adipose tissue as well as in cancers. Here, we studied whether human adipose tissue macrophages (ATM) modulate cancer cell function. Therefore, ATM were isolated and compared with monocyte-derived macrophages (MDM) from the same obese patients. ATM, but not MDM, were found to secrete factors inducing inflammation and lipid accumulation in human T47D and HT-29 cancer cells. Gene expression profile comparison of ATM and MDM revealed overexpression of functional clusters, such as cytokine-cytokine receptor interaction (especially CXC-chemokine) signaling as well as cancer-related pathways, in ATM. Comparison with gene expression profiles of human tumor-associated macrophages showed that ATM, but not MDM resemble tumor-associated macrophages. Indirect co-culture experiments demonstrated that factors secreted by preadipocytes, but not mature adipocytes, confer an ATM-like phenotype to MDM. Finally, the concentrations of ATM-secreted factors related to cancer are elevated in serum of obese subjects. In conclusion, ATM may thus modulate the cancer cell phenotype.


Assuntos
Adipócitos/citologia , Tecido Adiposo/metabolismo , Regulação Neoplásica da Expressão Gênica , Macrófagos/citologia , Neoplasias/metabolismo , Compostos Azo/farmacologia , Linhagem Celular Tumoral , Quimiocinas/metabolismo , Progressão da Doença , Humanos , Imuno-Histoquímica/métodos , Inflamação , Macrófagos/metabolismo , Obesidade/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo
13.
Blood ; 118(9): 2556-66, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21636855

RESUMO

The CDKN2A locus, which contains the tumor suppressor gene p16(INK4a), is associated with an increased risk of age-related inflammatory diseases, such as cardiovascular disease and type 2 diabetes, in which macrophages play a crucial role. Monocytes can polarize toward classically (CAMϕ) or alternatively (AAMϕ) activated macrophages. However, the molecular mechanisms underlying the acquisition of these phenotypes are not well defined. Here, we show that p16(INK4a) deficiency (p16(-/-)) modulates the macrophage phenotype. Transcriptome analysis revealed that p16(-/-) BM-derived macrophages (BMDMs) exhibit a phenotype resembling IL-4-induced macrophage polarization. In line with this observation, p16(-/-) BMDMs displayed a decreased response to classically polarizing IFNγ and LPS and an increased sensitivity to alternative polarization by IL-4. Furthermore, mice transplanted with p16(-/-) BM displayed higher hepatic AAMϕ marker expression levels on Schistosoma mansoni infection, an in vivo model of AAMϕ phenotype skewing. Surprisingly, p16(-/-) BMDMs did not display increased IL-4-induced STAT6 signaling, but decreased IFNγ-induced STAT1 and lipopolysaccharide (LPS)-induced IKKα,ß phosphorylation. This decrease correlated with decreased JAK2 phosphorylation and with higher levels of inhibitory acetylation of STAT1 and IKKα,ß. These findings identify p16(INK4a) as a modulator of macrophage activation and polarization via the JAK2-STAT1 pathway with possible roles in inflammatory diseases.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Genes p16 , Inflamação/genética , Janus Quinase 2/fisiologia , Ativação de Macrófagos , Fator de Transcrição STAT1/fisiologia , Animais , Transplante de Medula Óssea , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Citocinas/biossíntese , Quinase I-kappa B/fisiologia , Interferon gama/farmacologia , Interleucina-4/farmacologia , Lipopolissacarídeos/farmacologia , Fígado/metabolismo , Fígado/patologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Processamento de Proteína Pós-Traducional , Quimera por Radiação , Fator de Transcrição STAT6/fisiologia , Esquistossomose/imunologia , Transdução de Sinais
14.
Circ Res ; 108(8): 985-95, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21350215

RESUMO

RATIONALE: A crucial step in atherogenesis is the infiltration of the subendothelial space of large arteries by monocytes where they differentiate into macrophages and transform into lipid-loaded foam cells. Macrophages are heterogeneous cells that adapt their response to environmental cytokines. Th1 cytokines promote monocyte differentiation into M1 macrophages, whereas Th2 cytokines trigger an "alternative" M2 phenotype. OBJECTIVE: We previously reported the presence of CD68(+) mannose receptor (MR)(+) M2 macrophages in human atherosclerotic plaques. However, the function of these plaque CD68(+)MR(+) macrophages is still unknown. METHODS AND RESULTS: Histological analysis revealed that CD68(+)MR(+) macrophages locate far from the lipid core of the plaque and contain smaller lipid droplets compared to CD68(+)MR(-) macrophages. Interleukin (IL)-4-polarized CD68(+)MR(+) macrophages display a reduced capacity to handle and efflux cellular cholesterol because of low expression levels of the nuclear receptor liver x receptor (LXR)α and its target genes, ABCA1 and apolipoprotein E, attributable to the high 15-lipoxygenase activity in CD68(+)MR(+) macrophages. By contrast, CD68(+)MR(+) macrophages highly express opsonins and receptors involved in phagocytosis, resulting in high phagocytic activity. In M2 macrophages, peroxisome proliferator-activated receptor (PPAR)γ activation enhances the phagocytic but not the cholesterol trafficking pathways. CONCLUSIONS: These data identify a distinct macrophage subpopulation with a low susceptibility to become foam cells but high phagocytic activity resulting from different regulatory activities of the PPARγ-LXRα pathways.


Assuntos
Colesterol/metabolismo , Macrófagos/metabolismo , Receptores Nucleares Órfãos/metabolismo , PPAR gama/metabolismo , Fagocitose/fisiologia , Placa Aterosclerótica/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Predisposição Genética para Doença , Humanos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Receptores X do Fígado , Macrófagos/patologia , Receptores Nucleares Órfãos/fisiologia , Placa Aterosclerótica/patologia
15.
Arterioscler Thromb Vasc Biol ; 32(3): 677-85, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22207732

RESUMO

OBJECTIVE: 11ß-Hydroxysteroid dehydrogenase type 1 (11ß-HSD1) catalyzes the intracellular reduction of inactive cortisone to active cortisol, the natural ligand activating the glucocorticoid receptor (GR). Peroxisome proliferator- activated receptor-γ (PPARγ) is a nuclear receptor controlling inflammation, lipid metabolism, and the macrophage polarization state. In this study, we investigated the impact of macrophage polarization on the expression and activity of 11ß-HSD1 and the role of PPARγ therein. METHODS AND RESULTS: 11ß-HSD1 gene expression is higher in proinflammatory M1 and anti-inflammatory M2 macrophages than in resting macrophages, whereas its activity is highest in M2 macrophages. Interestingly, PPARγ activation induces 11ß-HSD1 enzyme activity in M2 macrophages but not in resting macrophages or M1 macrophages. Consequently, human M2 macrophages displayed enhanced responsiveness to the 11ß-HSD1 substrate cortisone, an effect amplified by PPARγ induction of 11ß-HSD1 activity, as illustrated by an increased expression of GR target genes. CONCLUSION: Our data identify a positive cross-talk between PPARγ and GR in human M2 macrophages via the induction of 11ß-HSD1 expression and activity.


Assuntos
Inflamação/enzimologia , Macrófagos/efeitos dos fármacos , PPAR gama/agonistas , Tiazolidinedionas/farmacologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/biossíntese , 11-beta-Hidroxiesteroide Desidrogenase Tipo 1/genética , Células Cultivadas , Cortisona/metabolismo , Indução Enzimática , Genes Reporter , Humanos , Hidrocortisona/metabolismo , Inflamação/genética , Inflamação/imunologia , Interleucina-4/metabolismo , Macrófagos/enzimologia , Macrófagos/imunologia , PPAR gama/genética , PPAR gama/metabolismo , Interferência de RNA , Receptores de Glucocorticoides/metabolismo , Rosiglitazona , Fatores de Tempo , Transfecção
16.
Cell Metab ; 6(2): 137-43, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17681149

RESUMO

Th1 cytokines promote monocyte differentiation into proatherogenic M1 macrophages, while Th2 cytokines lead to an "alternative" anti-inflammatory M2 macrophage phenotype. Here we show that in human atherosclerotic lesions, the expression of M2 markers and PPARgamma, a nuclear receptor controlling macrophage inflammation, correlate positively. Moreover, PPARgamma activation primes primary human monocytes into M2 differentiation, resulting in a more pronounced anti-inflammatory activity in M1 macrophages. However, PPARgamma activation does not influence M2 marker expression in resting or M1 macrophages, nor does PPARgamma agonist treatment influence the expression of M2 markers in atherosclerotic lesions, indicating that only native monocytes can be primed by PPARgamma activation to an enhanced M2 phenotype. Furthermore, PPARgamma activation significantly increases expression of the M2 marker MR in circulating peripheral blood mononuclear cells. These data demonstrate that PPARgamma activation skews human monocytes toward an anti-inflammatory M2 phenotype.


Assuntos
Inflamação/metabolismo , Inflamação/patologia , Macrófagos/citologia , Macrófagos/metabolismo , Monócitos/citologia , PPAR gama/metabolismo , Benzofenonas/farmacologia , Biomarcadores , Células Sanguíneas/efeitos dos fármacos , Doenças das Artérias Carótidas/patologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células Espumosas/efeitos dos fármacos , Células Espumosas/patologia , Humanos , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , PPAR gama/agonistas , Comunicação Parácrina/efeitos dos fármacos , Fenótipo , Células-Tronco/efeitos dos fármacos , Tirosina/análogos & derivados , Tirosina/farmacologia
17.
Curr Opin Lipidol ; 22(5): 365-72, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21825981

RESUMO

PURPOSE OF REVIEW: To discuss recent findings on the role and regulation of macrophage polarization in obesity and atherosclerosis. RECENT FINDINGS: Macrophages infiltrate the vascular wall during atherosclerosis and adipose tissue during obesity. At least two distinct subpopulations with different functions, the classically (M1) and the alternatively (M2) activated macrophages, have been found in these tissues. Reciprocal skewing of macrophage polarization between the M1 and M2 states is a process modulated by diet, humoral and transcription factors, such as the nuclear receptor peroxisome proliferator-activated receptor gamma. SUMMARY: Recent literature highlights the importance not only of the number of infiltrated macrophages, but also their activation in the maintenance of the inflammation state. Identifying mechanisms and molecules able to modify the balance between M1 and M2 represents a promising field of research.


Assuntos
Macrófagos/citologia , Macrófagos/metabolismo , Doenças Metabólicas/imunologia , Tecido Adiposo/imunologia , Animais , Humanos , Inflamação/imunologia
18.
Biochem Biophys Res Commun ; 404(1): 458-62, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21145308

RESUMO

Adipose tissue macrophages (ATM) are the major source of visfatin, a visceral fat adipokine upregulated during obesity. Also known to play a role in B cell differentiation (pre-B cell colony-enhancing factor (PBEF)) and NAD biosynthesis (nicotinamide phosphoribosyl transferase (NAMPT)), visfatin has been suggested to play a role in inflammation. Liver X Receptor (LXR) and Peroxisome Proliferator-Activated Receptor (PPAR)γ are nuclear receptors expressed in macrophages controlling the inflammatory response. Recently, we reported visfatin as a PPARγ target gene in human macrophages. In this study, we examined whether LXR regulates macrophage visfatin expression. Synthetic LXR ligands decreased visfatin gene expression in a LXR-dependent manner in human and murine macrophages. The decrease of visfatin mRNA was paralleled by a decrease of protein secretion. Consequently, a modest and transient decrease of NAD(+) concentration was observed. Interestingly, LXR activation decreased the PPARγ-induced visfatin gene and protein secretion in human macrophages. Our results identify visfatin as a gene oppositely regulated by the LXR and PPARγ pathways in human macrophages.


Assuntos
Citocinas/antagonistas & inibidores , Citocinas/genética , Regulação Enzimológica da Expressão Gênica , Macrófagos/enzimologia , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Nicotinamida Fosforribosiltransferase/genética , Receptores Nucleares Órfãos/metabolismo , Células Cultivadas , Citocinas/metabolismo , Citocinas/farmacologia , Regulação para Baixo , Expressão Gênica , Humanos , Receptores X do Fígado , NAD/metabolismo , Nicotinamida Fosforribosiltransferase/metabolismo , Receptores Nucleares Órfãos/agonistas , Receptores Nucleares Órfãos/genética , PPAR gama/metabolismo
19.
J Trace Elem Med Biol ; 67: 126786, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34022567

RESUMO

AIMS: Dysmetabolic iron overload syndrome (DIOS) is common but the clinical relevance of iron overload is not understood. Macrophages are central cells in iron homeostasis and inflammation. We hypothesized that iron overload in DIOS could affect the phenotype of monocytes and impair macrophage gene expression. METHODS: This study compared 20 subjects with DIOS to 20 subjects with metabolic syndrome (MetS) without iron overload, and 20 healthy controls. Monocytes were phenotyped by Fluorescence-Activated Cell Sorting (FACS) and differentiated into anti-inflammatory M2 macrophages in the presence of IL-4. The expression of 38 genes related to inflammation, iron metabolism and M2 phenotype was assessed by real-time PCR. RESULTS: FACS showed no difference between monocytes across the three groups. The macrophagic response to IL-4-driven differentiation was altered in four of the five genes of M2 phenotype (MRC1, F13A1, ABCA1, TGM2 but not FABP4), in DIOS vs Mets and controls demonstrating an impaired M2 polarization. The expression profile of inflammatory genes was not different in DIOS vs MetS. Several genes of iron metabolism presented a higher expression in DIOS vs MetS: SCL11A2 (a free iron transporter, +76 %, p = 0.04), SOD1 (an antioxidant enzyme, +27 %, p = 0.02), and TFRC (the receptor 1 of transferrin, +59 %, p = 0.003). CONCLUSIONS: In DIOS, macrophage polarization toward the M2 alternative phenotype is impaired but not associated with a pro-inflammatory profile. The up regulation of transferrin receptor 1 (TFRC) in DIOS macrophages suggests an adaptive role that may limit iron toxicity in DIOS.


Assuntos
Sobrecarga de Ferro , Síndrome Metabólica , Estudos de Casos e Controles , Humanos , Inflamação , Interleucina-4 , Ferro , Macrófagos
20.
Biochem Biophys Res Commun ; 386(3): 459-62, 2009 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-19527689

RESUMO

Macrophages adapt their response to micro-environmental signals. While Th1 cytokines promote pro-inflammatory M1 macrophages, Th2 cytokines promote an "alternative" anti-inflammatory M2 macrophage phenotype. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors expressed in macrophages where they control the inflammatory response. It has been shown that PPARgamma promotes the differentiation of monocytes into anti-inflammatory M2 macrophages in humans and mice, while a role for PPARbeta/delta in this process has been reported only in mice and no data are available for PPARalpha. Here, we show that in contrast to PPARgamma, expression of PPARalpha and PPARbeta/delta overall does not correlate with the expression of M2 markers in human atherosclerotic lesions, whereas a positive correlation with genes of lipid metabolism exists. Moreover, unlike PPARgamma, PPARalpha or PPARbeta/delta activation does not influence human monocyte differentiation into M2 macrophages in vitro. Thus, PPARalpha and PPARbeta/delta do not appear to modulate the alternative differentiation of human macrophages.


Assuntos
Aterosclerose/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , PPAR alfa/biossíntese , PPAR delta/biossíntese , PPAR beta/biossíntese , Diferenciação Celular , Células Cultivadas , Humanos , Macrófagos/metabolismo , Monócitos/imunologia , PPAR alfa/agonistas , PPAR alfa/genética , PPAR delta/agonistas , PPAR delta/genética , PPAR gama/agonistas , PPAR gama/biossíntese , PPAR gama/genética , PPAR beta/agonistas , PPAR beta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA