Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 36(6): 1047-59, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22749354

RESUMO

Dendritic cells (DCs), monocytes, and/or macrophages initiate host-protective immune responses to intracellular pathogens in part through interleukin-12 (IL-12) production, although the relative contribution of tissue resident versus recruited cells has been unclear. Here, we showed that after intraperitoneal infection with Toxoplasma gondii cysts, resident mononuclear phagocytes are replaced by circulating monocytes that differentiate in situ into inflammatory DCs (moDCs) and F4/80(+) macrophages. Importantly, NK cell-derived interferon-γ (IFN-γ) was required for both the loss of resident mononuclear phagocytes and the local differentiation of monocytes into macrophages and moDCs. This newly generated moDC population and not the resident DCs (or macrophages) served as the major source of IL-12 at the site of infection. Thus, NK cell-derived IFN-γ is important in both regulating inflammatory cell dynamics and in driving the local differentiation of monocytes into the cells required for initiating the immune response to an important intracellular pathogen.


Assuntos
Células Dendríticas/imunologia , Interferon gama/fisiologia , Células Matadoras Naturais/imunologia , Monócitos/imunologia , Transferência Adotiva , Animais , Antígenos Ly/análise , Diferenciação Celular , Quimiotaxia de Leucócito , Células Dendríticas/patologia , Células Dendríticas/transplante , Genes Reporter , Subunidade p40 da Interleucina-12/biossíntese , Subunidade p40 da Interleucina-12/genética , Células Matadoras Naturais/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/transplante , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/química , Monócitos/patologia , Monócitos/transplante , Fator 88 de Diferenciação Mieloide/fisiologia , Neutrófilos/imunologia , Peritonite/imunologia , Peritonite/parasitologia , Fagócitos/classificação , Fagócitos/imunologia , Fagócitos/patologia , Receptores de Interferon/deficiência , Receptores de Interferon/fisiologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Organismos Livres de Patógenos Específicos , Subpopulações de Linfócitos T/imunologia , Toxoplasmose Animal/imunologia , Receptor de Interferon gama
2.
J Cell Mol Med ; 24(6): 3314-3327, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32004417

RESUMO

Acute kidney injury (AKI) is a clinical condition that is associated with high morbidity and mortality. Inflammation is reported to play a key role in AKI. Although the M2 macrophages exhibit antimicrobial and anti-inflammatory activities, their therapeutic potential has not been evaluated for AKI. This study aimed to investigate the protective effect of peritoneal M2 macrophage transplantation on AKI in mice. The macrophages were isolated from peritoneal dialysates of mice. The macrophages were induced to undergo M2 polarization using interleukin (IL)-4/IL-13. AKI was induced in mice by restoring the blood supply after bilateral renal artery occlusion for 30 minutes. The macrophages were injected into the renal cortex of mice. The changes in renal function, inflammation and tubular proliferation were measured. The M2 macrophages were co-cultured with the mouse primary proximal tubular epithelial cells (PTECs) under hypoxia/reoxygenation conditions in vitro. The PTEC apoptosis and proliferation were analysed. The peritoneal M2 macrophages effectively alleviated the renal injury and inflammatory response in mice with ischaemia-reperfusion injury (IRI) and promoted the PTEC proliferation in vivo and in vitro. These results indicated that the peritoneal M2 macrophages ameliorated AKI by decreasing inflammatory response and promoting PTEC proliferation. Hence, the peritoneal M2 macrophage transplantation can serve as a potential cell therapy for renal diseases.


Assuntos
Injúria Renal Aguda/terapia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Macrófagos Peritoneais/transplante , Traumatismo por Reperfusão/terapia , Animais , Apoptose/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Inflamação/patologia , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obstrução da Artéria Renal , Cicatrização/fisiologia
3.
Biochem Biophys Res Commun ; 529(3): 540-547, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32736671

RESUMO

BACKGROUND: Liver X receptor alpha (Lxrα) is a sterol-regulated transcription factor that limits atherogenesis by regulating cholesterol homeostasis and inflammation in macrophages. Transcriptional profiling identified the reverse cholesterol transport protein Arf-like 7 (Arl7, Arl4c) as a Lxrα target gene. We hypothesized that the LXR response element (LXRE) sequence on the murine macrophage Arl7 promoter may play a critical role in Lxrα's atherosuppressive effects. METHODS: Employing low density lipoprotein receptor-deficient mice with macrophage-specific Lxrα overexpression (Ldlr-/- MΦ-Lxrα), we constructed a novel in vivo Ldlr-/- MΦ-Lxrα Arl7MutLXRE model possessing macrophage-specific mutations within the Arl7 promoter LXRE sequences (Arl7MutLXRE) using the CRISPR/spCas9 genome editing technique. In vitro and in vivo transplantation studies were conducted using bone marrow-derived macrophages (BMDMs) and peritoneal macrophages (PMs). RESULTS: Ldlr-/-, Ldlr-/- MΦ-Lxrα, and Ldlr-/- MΦ-Lxrα Arl7MutLXRE mice on a 60% high-fat diet displayed no significant differences in body weight, fat mass, glucose homeostasis, or lipid metabolism. Macrophage Lxrα promoted Arl7 expression, enhanced cholesterol efflux, and reduced foam cell formation in an Arl7 LXRE-dependent manner. In contrast, Lxrα reduced macrophage activation, inflammatory cytokine expression, and efferocytosis independent of Arl7 LXRE. Western diet-fed Ldlr-/- mice reconstituted with transgenic BMDMs revealed that macrophage Lxrα reduced atherosclerotic plaque formation independent of Arl7 LXRE. CONCLUSION: Lxrα's anti-atherosclerotic effects in Ldlr-/- mice are not primarily attributable to Lxrα's influence on Arl7 expression. This evidence suggests that Lxrα's effects on plaque inflammation may be more critical to in vivo atherogenesis than its effects on macrophage cholesterol efflux and foam cell development.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Aterosclerose/metabolismo , Receptores X do Fígado/metabolismo , Macrófagos Peritoneais/metabolismo , Macrófagos/metabolismo , Receptores de LDL/metabolismo , Fatores de Ribosilação do ADP/genética , Animais , Aterosclerose/genética , Aterosclerose/terapia , Colesterol/metabolismo , Células Espumosas/metabolismo , Metabolismo dos Lipídeos/genética , Macrófagos/transplante , Macrófagos Peritoneais/transplante , Masculino , Camundongos Knockout , Camundongos Transgênicos , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/terapia , Receptores de LDL/genética , Ativação Transcricional
4.
Kidney Int ; 95(3): 563-576, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30670317

RESUMO

The cholinergic anti-inflammatory pathway (CAP) links the nervous and immune systems and modulates innate and adaptive immunity. Activation of the CAP by vagus nerve stimulation exerts protective effects in a wide variety of clinical disorders including rheumatoid arthritis and Crohn's disease, and in murine models of acute kidney injury including ischemia/reperfusion injury (IRI). The canonical CAP pathway involves activation of splenic alpha7-nicotinic acetylcholine receptor (α7nAChR)-positive macrophages by splenic ß2-adrenergic receptor-positive CD4+ T cells. Here we demonstrate that ultrasound or vagus nerve stimulation also activated α7nAChR-positive peritoneal macrophages, and that adoptive transfer of these activated peritoneal macrophages reduced IRI in recipient mice. The protective effect required α7nAChR, and did not occur in splenectomized mice or in mice lacking T and B cells, suggesting a bidirectional interaction between α7nAChR-positive peritoneal macrophages and other immune cells including ß2-adrenergic receptor-positive CD4+ T cells. We also found that expression of hairy and enhancer of split-1 (Hes1), a basic helix-loop-helix DNA-binding protein, is induced in peritoneal macrophages by ultrasound or vagus nerve stimulation. Adoptive transfer of Hes1-overexpressing peritoneal macrophages reduced kidney IRI. Our data suggest that Hes1 is downstream of α7nAChR and is important to fully activate the CAP. Taken together, these results suggest that peritoneal macrophages play a previously unrecognized role in mediating the protective effect of CAP activation in kidney injury, and that Hes1 is a new candidate pharmacological target to activate the CAP.


Assuntos
Injúria Renal Aguda/imunologia , Macrófagos Peritoneais/imunologia , Traumatismo por Reperfusão/imunologia , Fatores de Transcrição HES-1/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/terapia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/efeitos da radiação , Linfócitos T CD4-Positivos/transplante , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Ativação de Macrófagos , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/transplante , Masculino , Camundongos , Neuroimunomodulação/efeitos da radiação , Células RAW 264.7 , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/terapia , Fatores de Transcrição HES-1/genética , Fatores de Transcrição HES-1/imunologia , Terapia por Ultrassom , Regulação para Cima/efeitos da radiação , Estimulação do Nervo Vago , Receptor Nicotínico de Acetilcolina alfa7/imunologia
5.
Arterioscler Thromb Vasc Biol ; 38(11): 2590-2600, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30354246

RESUMO

Objective- Inhibition of HMGCR (3-hydroxy-3-methylglutaryl-coenzyme A reductase) is atheroprotective primarily by decreasing plasma LDL (low-density lipoprotein)-cholesterol. However, it is unknown whether inhibition of HMGCR in myeloid cells contributes to this atheroprotection. We sought to determine the role of myeloid HMGCR in the development of atherosclerosis. Approach and Results- We generated mice with genetically reduced Hmgcr in myeloid cells ( Hmgcr m-/m-) using LysM (Cre) and compared various functions of their macrophages to those of Hmgcr fl/fl control mice. We further compared the extent of atherosclerosis in Hmgcr m-/ m- and Hmgcr fl/fl mice in the absence of Ldlr (LDL receptor). Hmgcr m-/ m- macrophages and granulocytes had significantly lower Hmgcr mRNA expression and cholesterol biosynthesis than Hmgcr fl/fl cells. In vitro, Hmgcr m-/ m- monocytes/macrophages had reduced ability to migrate, proliferate, and survive compared with Hmgcr fl/fl monocytes/macrophages. However, there was no difference in ability to adhere, phagocytose, store lipids, or polarize to M1 macrophages between the 2 types of macrophages. The amounts of plasma membrane-associated small GTPase proteins, such as RhoA (RAS homolog family member A), were increased in Hmgcr m-/ m- macrophages. In the setting of Ldlr deficiency, Hmgcr m-/ m- mice developed significantly smaller atherosclerotic lesions than Hmgcr fl/fl mice. However, there were no differences between the 2 types of mice either in plasma lipoprotein profiles or in the numbers of proliferating or apoptotic cells in the lesions in vivo. The in vivo migration of Hmgcr m-/ m- macrophages to the lesions was reduced compared with Hmgcr fl/fl macrophages. Conclusions- Genetic reduction of HMGCR in myeloid cells may exert atheroprotective effects primarily by decreasing the migratory activity of monocytes/macrophages to the lesions.


Assuntos
Aorta/enzimologia , Doenças da Aorta/enzimologia , Aterosclerose/enzimologia , Movimento Celular , Hidroximetilglutaril-CoA Redutases/metabolismo , Macrófagos Peritoneais/enzimologia , Monócitos/enzimologia , Transferência Adotiva , Animais , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Doenças da Aorta/prevenção & controle , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença , Hidroximetilglutaril-CoA Redutases/genética , Lipídeos/sangue , Macrófagos Peritoneais/patologia , Macrófagos Peritoneais/transplante , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/patologia , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Fenótipo , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais
6.
Am J Physiol Renal Physiol ; 314(4): F561-F571, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29357438

RESUMO

Cell therapy using macrophages requires large amounts of cells, which are difficult to collect from patients. Patients undergoing peritoneal dialysis (PD) discard huge numbers of peritoneal macrophages in dialysate daily. Macrophages can be modulated to become regulatory macrophages, which have shown great promise as a therapeutic strategy in experimental kidney disease and human kidney transplantation. This study aimed to examine the potential of using peritoneal macrophages (PMs) from peritoneal dialysate to treat kidney disease. Monocytes/macrophages accounted for >40% of total peritoneal leukocytes in both patients and mice undergoing PD. PMs from patients and mice undergoing PD were more mature than peripheral monocytes/macrophages, as shown by low expression of C-C motif chemokine receptor 2 (CCR2) and morphological changes during in vitro culture. PMs from patients and mice undergoing PD displayed normal macrophage function and could be modulated into a regulatory (M2) phenotype. In vivo, adoptive transfer of peritoneal M2 macrophages derived from PD mice effectively protected against kidney injury in mice with adriamycin nephropathy (AN). Importantly, the transfused peritoneal M2 macrophages maintained their M2 phenotype in kidney of AN mice. In conclusion, PMs derived from patients and mice undergoing PD exhibited conventional macrophage features. Peritoneal M2 macrophages derived from PD mice are able to reduce kidney injury in AN, suggesting that peritoneal macrophages from patients undergoing PD may have the potential for clinical therapeutic application.


Assuntos
Transferência Adotiva , Soluções para Diálise , Doxorrubicina , Nefropatias/prevenção & controle , Rim , Macrófagos Peritoneais/transplante , Diálise Peritoneal , Animais , Plasticidade Celular , Separação Celular/métodos , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação/metabolismo , Rim/imunologia , Rim/metabolismo , Rim/patologia , Nefropatias/induzido quimicamente , Nefropatias/imunologia , Nefropatias/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Fenótipo , Fatores de Tempo
7.
Arterioscler Thromb Vasc Biol ; 37(8): 1470-1481, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28642237

RESUMO

OBJECTIVE: MED1 (mediator 1) interacts with transcription factors to regulate transcriptional machinery. The role of MED1 in macrophage biology and the relevant disease state remains to be investigated. APPROACH AND RESULTS: To study the molecular mechanism by which MED1 regulates the M1/M2 phenotype switch of macrophage and the effect on atherosclerosis, we generated MED1/apolipoprotein E (ApoE) double-deficient (MED1ΔMac/ApoE-/-) mice and found that atherosclerosis was greater in MED1ΔMac/ApoE-/- mice than in MED1fl/fl/ApoE-/- littermates. The gene expression of M1 markers was increased and that of M2 markers decreased in both aortic wall and peritoneal macrophages from MED1ΔMac/ApoE-/- mice, whereas MED1 overexpression rectified the changes in M1/M2 expression. Moreover, LDLR (low-density lipoprotein receptor)-deficient mice received bone marrow from MED1ΔMac mice showed greater atherosclerosis. Mechanistically, MED1 ablation decreased the binding of PPARγ (peroxisome proliferator-activated receptor γ) and enrichment of H3K4me1 and H3K27ac to upstream region of M2 marker genes. Furthermore, interleukin 4 induction of PPARγ and MED1 increased the binding of PPARγ or MED1 to the PPAR response elements of M2 marker genes. CONCLUSIONS: Our data suggest that MED1 is required for the PPARγ-mediated M2 phenotype switch, with M2 marker genes induced but M1 marker genes suppressed. MED1 in macrophages has an antiatherosclerotic role via PPARγ-regulated transactivation.


Assuntos
Aorta/metabolismo , Doenças da Aorta/prevenção & controle , Aterosclerose/prevenção & controle , Plasticidade Celular , Macrófagos Peritoneais/metabolismo , Subunidade 1 do Complexo Mediador/metabolismo , Acetilação , Animais , Aorta/imunologia , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Sítios de Ligação , Transplante de Medula Óssea , Modelos Animais de Doenças , Epigênese Genética , Regulação da Expressão Gênica , Predisposição Genética para Doença , Histonas/metabolismo , Imunidade Inata , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/patologia , Macrófagos Peritoneais/transplante , Masculino , Subunidade 1 do Complexo Mediador/deficiência , Subunidade 1 do Complexo Mediador/genética , Metilação , Camundongos , Camundongos Knockout , PPAR gama/metabolismo , Fenótipo , Placa Aterosclerótica , Células RAW 264.7 , Interferência de RNA , Receptores de LDL/deficiência , Receptores de LDL/genética , Elementos de Resposta , Transdução de Sinais , Transcrição Gênica , Ativação Transcricional , Transfecção
8.
Cell Immunol ; 320: 20-28, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28942945

RESUMO

Non-steroidal anti-inflammatory drugs often cause ulcers in the human small intestine, but few effective agents exist to treat such injury. Ganoderma lucidum Karst, also known as "Reishi" or "Lingzhi", is a mushroom. We previously reported that a water-soluble extract from G. lucidum fungus mycelia (MAK) has anti-inflammatory effects in murine colitis induced by trinitrobenzene sulfonic acid, and induction of granulocyte macrophage colony-stimulating factor (GM-CSF) by MAK may provide anti-inflammatory effects. However, its effects on indomethacin-induced small intestinal injuries are unknown. The present study investigated the preventative effects of MAK via immunological function and the polysaccharides from MAK on indomethacin-induced ileitis in mice. Peritoneal macrophages (PMs) were stimulated in vitro with MAK and adoptively transferred to C57BL/6 mice intraperitoneally, which were then given indomethacin. Intestinal inflammation was evaluated after 24h. We performed in vivo antibody blockade to investigate the preventive role of GM-CSF, which derived from PMs stimulated with MAK. We then used PMs stimulated with MAK pre-treated by pectinase in an adoptive transfer assay to determine the preventive role of polysaccharides. Indomethacin-induced small intestinal injury was inhibited by adoptive transfer of PMs stimulated in vitro with MAK. In this transfer model, pre-treatment with anti-GM-CSF antibody but not with control antibody reversed the improvement of small intestinal inflammation by indomethacin. Pectinase pretreatment impaired the anti-inflammatory effect of MAK. PMs stimulated by MAK appear to contribute to the anti-inflammatory response through GM-CSF in small intestinal injury induced by indomethacin. The polysaccharides may be the components that elicit the anti-inflammatory effect.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Úlcera Duodenal/terapia , Polissacarídeos Fúngicos/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Indometacina/efeitos adversos , Intestino Delgado/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Reishi/química , Transferência Adotiva , Animais , Células Cultivadas , Misturas Complexas/química , Misturas Complexas/uso terapêutico , Úlcera Duodenal/induzido quimicamente , Úlcera Duodenal/imunologia , Polissacarídeos Fúngicos/isolamento & purificação , Intestino Delgado/imunologia , Macrófagos Peritoneais/transplante , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Micélio/química , Poligalacturonase/química
9.
Arterioscler Thromb Vasc Biol ; 36(1): 37-48, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26603156

RESUMO

OBJECTIVES: Monocyte/macrophage recruitment and activation at vascular predilection sites plays a central role in the pathogenesis of atherosclerosis. Heterotrimeric G proteins of the G12/13 family have been implicated in the control of migration and inflammatory gene expression, but their function in myeloid cells, especially during atherogenesis, is unknown. APPROACH AND RESULTS: Mice with myeloid-specific deficiency for G12/13 show reduced atherosclerosis with a clear shift to anti-inflammatory gene expression in aortal macrophages. These changes are because of neither altered monocyte/macrophage migration nor reduced activation of inflammatory gene expression; on the contrary, G12/13-deficient macrophages show an increased nuclear factor-κB-dependent gene expression in the resting state. Chronically increased inflammatory gene expression in resident peritoneal macrophages results in myeloid-specific G12/13-deficient mice in an altered peritoneal micromilieu with secondary expansion of peritoneal B1 cells. Titers of B1-derived atheroprotective antibodies are increased, and adoptive transfer of peritoneal cells from mutant mice conveys atheroprotection to wild-type mice. With respect to the mechanism of G12/13-mediated transcriptional control, we identify an autocrine feedback loop that suppresses nuclear factor-κB-dependent gene expression through a signaling cascade involving sphingosine 1-phosphate receptor subtype 2, G12/13, and RhoA. CONCLUSIONS: Together, these data show that selective inhibition of G12/13 signaling in macrophages can augment atheroprotective B-cell populations and ameliorate atherosclerosis.


Assuntos
Aorta/metabolismo , Doenças da Aorta/prevenção & controle , Aterosclerose/prevenção & controle , Subpopulações de Linfócitos B/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Ativação de Macrófagos , Macrófagos Peritoneais/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Transferência Adotiva , Animais , Aorta/imunologia , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/imunologia , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Comunicação Autócrina , Subpopulações de Linfócitos B/imunologia , Células Cultivadas , Modelos Animais de Doenças , Retroalimentação Fisiológica , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica , Mediadores da Inflamação/metabolismo , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/transplante , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Receptores de Lisoesfingolipídeo/deficiência , Receptores de Lisoesfingolipídeo/genética , Transdução de Sinais , Receptores de Esfingosina-1-Fosfato , Transcrição Gênica , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP
10.
J Immunol ; 188(11): 5734-40, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22547702

RESUMO

Reactive oxygen species (ROS) generated by NADPH oxidase are generally known to be proinflammatory, and it seems to be counterintuitive that ROS play a critical role in regulating the resolution of the inflammatory response. However, we observed that deficiency of the p47(phox) component of NADPH oxidase in macrophages was associated with a paradoxical accentuation of inflammation in a whole animal model of noninfectious sepsis induced by endotoxin. We have confirmed this observation by interrogating four separate in vivo models that use complementary methodology including the use of p47(phox-/-) mice, p47(phox-/-) bone marrow chimera mice, adoptive transfer of macrophages from p47(phox-/-) mice, and an isolated perfused lung edema model that all point to a relationship between excessive acute inflammation and p47(phox) deficiency in macrophages. Mechanistic data indicate that ROS deficiency in both cells and mice results in decreased production of IL-10 in response to treatment with LPS, at least in part, through attenuation of the Akt-GSK3-ß signal pathway and that it can be reversed by the administration of rIL-10. Our data support the innovative concept that generation of ROS is essential for counterregulation of acute lung inflammation.


Assuntos
Endotoxinas/toxicidade , Regulação da Expressão Gênica/imunologia , Interleucina-10/biossíntese , Pulmão/imunologia , Pulmão/patologia , Pneumonia/imunologia , Pneumonia/patologia , Espécies Reativas de Oxigênio/uso terapêutico , Transferência Adotiva , Animais , Modelos Animais de Doenças , Endotoxinas/antagonistas & inibidores , Humanos , Interleucina-10/antagonistas & inibidores , Luciferases/biossíntese , Luciferases/genética , Pulmão/metabolismo , Macrófagos Peritoneais/transplante , Camundongos , Camundongos Knockout , Camundongos Transgênicos , NADPH Oxidases/deficiência , NADPH Oxidases/genética , Pneumonia/terapia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/administração & dosagem
11.
Am J Physiol Lung Cell Mol Physiol ; 305(11): L866-77, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24077949

RESUMO

We sought to investigate the effects of cockroach allergen (CRA) exposure on the lung macrophage population to determine how different macrophage phenotypes influence exacerbation of disease. CRA exposure caused significantly reduced expression of CD86 on lung macrophages. These effects were not systemic, as peritoneal macrophage CD86 expression was not altered. To investigate whether naïve macrophages could reduce asthma-like pulmonary inflammation, autologous peritoneal macrophages were instilled into the airways 24 h before the final CRA challenge. Pulmonary inflammation was assessed by measurement of airway hyperresponsiveness, mucin production, inflammatory cell recruitment, and cytokine production. Cell transfer did not have significant effects in control mice, nor did it affect pulmonary mucin production or airway hyperresponsiveness in control or CRA-exposed mice. However, there was significant reduction in the number of eosinophils recovered in the bronchoalveolar lavage (BAL) (5.8 × 105 vs. 0.88 × 105), and total cell recruitment to the airways of CRA-exposed mice was markedly reduced (1.1 × 106 vs. 0.57 × 106). The reduced eosinophil recruitment was reflected by substantially lower levels of eosinophil peroxidase in the lung and significantly lower concentrations of eotaxins in BAL (eotaxin 1: 3 pg/ml vs. undetectable; eotaxin 2: 2,383 vs. 131 pg/ml) and lung homogenate (eotaxin 1: 1,043 vs. 218 pg/ml; eotaxin 2: 10 vs. 1.5 ng/ml). We conclude that CRA decreases lung macrophage CD86 expression. Furthermore, supplementation of the lung cell population with peritoneal macrophages inhibits eosinophil recruitment, achieved through reduction of eotaxin production. These data demonstrate that transfer of naïve macrophages will reduce some aspects of asthma-like pulmonary inflammation in response to CRA.


Assuntos
Asma/imunologia , Quimiocina CCL11/biossíntese , Quimiocina CCL24/biossíntese , Baratas/imunologia , Eosinófilos/imunologia , Macrófagos Peritoneais/imunologia , Alérgenos/imunologia , Animais , Animais não Endogâmicos , Líquido da Lavagem Broncoalveolar , Células Cultivadas , Técnicas de Cocultura , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Macrófagos Alveolares/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/transplante , Camundongos , Camundongos Endogâmicos ICR , Mucinas/metabolismo , Neutrófilos/imunologia
12.
Cell Immunol ; 271(2): 335-41, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21875704

RESUMO

In this study, we investigate the potential of peritoneal macrophages to differentiate into dendritic cell (DCs) in response to preferential uptake of oligomannose-coated liposomes (OMLs). About 30% of peritoneal cells (PECs) preferentially took up OMLs that were administered into the peritoneal cavity. The OML-ingesting cells expressed CD11b and F4/80, but lacked CD11c expression, indicating that the OML-ingesting PECs with a CD11b(high)CD11c(-) phenotype are resident peritoneal macrophages. During in vitro cultivation, CD11c(+) cells arose among the PECs with ingested OMLs. CD11c(+) cells also developed among enriched peritoneal CD11b(high)CD11(-) cells from OML-treated mice, and the resulting CD11c(+) cells expressed co-stimulatory molecules and MHC class II. In addition, OML-ingesting CD11b(high)CD11c(+) cells were found in spleen after the enriched peritoneal macrophages with ingested OMLs were transplanted in the peritoneal cavity of mice. These results show that a fraction of peritoneal macrophages can differentiate into mature DCs following uptake of OMLs.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/imunologia , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/imunologia , Oligossacarídeos/imunologia , Animais , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Feminino , Lipossomos , Macrófagos Peritoneais/transplante , Camundongos , Camundongos Endogâmicos C57BL , Oligossacarídeos/administração & dosagem
13.
JCI Insight ; 5(16)2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32814707

RESUMO

Type 1 diabetes (T1D) is a consequence of autoimmune ß cell destruction, but the role of lipids in this process is unknown. We previously reported that activation of Ca2+-independent phospholipase A2ß (iPLA2ß) modulates polarization of macrophages (MΦ). Hydrolysis of the sn-2 substituent of glycerophospholipids by iPLA2ß can lead to the generation of oxidized lipids (eicosanoids), pro- and antiinflammatory, which can initiate and amplify immune responses triggering ß cell death. As MΦ are early triggers of immune responses in islets, we examined the impact of iPLA2ß-derived lipids (iDLs) in spontaneous-T1D prone nonobese diabetic mice (NOD), in the context of MΦ production and plasma abundances of eicosanoids and sphingolipids. We find that (a) MΦNOD exhibit a proinflammatory lipid landscape during the prediabetic phase; (b) early inhibition or genetic reduction of iPLA2ß reduces production of select proinflammatory lipids, promotes antiinflammatory MΦ phenotype, and reduces T1D incidence; (c) such lipid changes are reflected in NOD plasma during the prediabetic phase and at T1D onset; and (d) importantly, similar lipid signatures are evidenced in plasma of human subjects at high risk for developing T1D. These findings suggest that iDLs contribute to T1D onset and identify select lipids that could be targeted for therapeutics and, in conjunction with autoantibodies, serve as early biomarkers of pre-T1D.


Assuntos
Biomarcadores/metabolismo , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/etiologia , Metabolismo dos Lipídeos , Macrófagos Peritoneais/metabolismo , Adolescente , Animais , Criança , Diabetes Mellitus Tipo 1/terapia , Eicosanoides/metabolismo , Ácidos Graxos/metabolismo , Feminino , Fosfolipases A2 do Grupo IV/antagonistas & inibidores , Fosfolipases A2 do Grupo IV/metabolismo , Humanos , Cetonas/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipídeos/sangue , Macrófagos Peritoneais/patologia , Macrófagos Peritoneais/transplante , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Naftalenos/farmacologia
14.
PLoS One ; 15(12): e0242488, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33301448

RESUMO

Macrophages play an indispensable role in both innate and acquired immunity, while the persistence of activated macrophages can sometimes be harmful to the host, resulting in multi-organ damage. Macrophages develop from monocytes in the circulation. However, little is known about the organ affinity of macrophages in the normal state. Using in vivo imaging with XenoLight DiR®, we observed that macrophages showed strong affinity for the liver, spleen and lung, and weak affinity for the gut and bone marrow, but little or no affinity for the kidney and skin. We also found that administered macrophages were still alive 168 hours after injection. On the other hand, treatment with clodronate liposomes, which are readily taken up by macrophages via phagocytosis, strongly reduced the number of macrophages in the liver, spleen and lung.


Assuntos
Rastreamento de Células/métodos , Lipossomos/farmacologia , Fígado/efeitos dos fármacos , Macrófagos Peritoneais/fisiologia , Coloração e Rotulagem/métodos , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Carbocianinas/química , Ácido Clodrônico/química , Ácido Clodrônico/farmacologia , Corantes Fluorescentes/química , Intestinos/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/metabolismo , Lipossomos/química , Fígado/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Macrófagos Peritoneais/transplante , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose/efeitos dos fármacos , Cultura Primária de Células , Pele/efeitos dos fármacos , Pele/metabolismo , Baço/efeitos dos fármacos , Baço/metabolismo
15.
Front Immunol ; 11: 1934, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33013847

RESUMO

Long non-coding RNA (lncRNA) is pivotal for multiple sclerosis (MS), but the potential mechanism of lncRNA PVT1 in MS animal model, experimental autoimmune encephalomyelitis (EAE) still remains unclear. In this study, macrophages were firstly isolated and induced to polarize into M2 macrophages. M2 macrophage-derived exosomes (M2-exos) were extracted and identified, and EAE mouse model was established and treated with M2-exos. The effect of M2-exos on EAE mice was evaluated by clinical scores. The proportion of Treg and Th17 cells in spinal cord cells and splenocytes, and levels of inflammatory factors were measured. The targeting relationships among PVT1, miR-21-5p, and SOCS5 were verified. The expression of JAKs/STAT3 pathway-related proteins was measured. After M2-exo treatment, the clinical score of EAE mice decreased, and demyelination and inflammatory infiltration improved; Th17 cells decreased, Treg cells increased, and the levels of inflammatory factors decreased significantly. SOCS5 and PVT1 were downregulated and miR-21-5p was upregulated in EAE mice. PVT1 could sponge miR-21-5p to regulate SOCS5. SOCS5 alleviated EAE symptoms by repressing the JAKs/STAT3 pathway. Together, M2-exos-carried lncRNA PVT1 sponged miR-21-5p to upregulate SOCS5 and inactivate the JAKs/STAT3 pathway, thus reducing inflammation and protecting EAE mice. This study may offer novel treatments for MS.


Assuntos
Encefalomielite Autoimune Experimental/terapia , Exossomos/transplante , Macrófagos Peritoneais/transplante , RNA Longo não Codificante/metabolismo , Medula Espinal/metabolismo , Células Th17/metabolismo , Animais , Células Cultivadas , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Exossomos/genética , Exossomos/metabolismo , Janus Quinases/metabolismo , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Fenótipo , RNA Longo não Codificante/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Medula Espinal/imunologia , Baço/imunologia , Baço/metabolismo , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th17/imunologia
16.
Biomed Pharmacother ; 117: 109172, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31261028

RESUMO

Macrophages in the kidney play different roles in renal interstitial fibrosis (RIF) depending on their phenotypes. M2 phenotype macrophages are believed to protect the kidney against RIF. Free fatty acid receptor GPR120 is expressed in macrophages, and its activation induces macrophage transition to M2 phenotype. In this study, the effects of GPR120 agonist-programmed macrophages on RIF were investigated. The peritoneal macrophages collected from rats were incubated with GPR120 agonist TUG891 in vitro for 24 h, and then they were transplanted autologously to the kidney with ureteral obstruction by intrarenal injection for 7 days on the same day following unilateral ureteral obstruction (UUO) operation. RIF was identified by Masson trichrome histological staining, and the expression of RIF-related proteins was analyzed by immunohistochemistry and western blot. It was observed that TUG891-programmed macrophages up-regulated the expression of CD206 and arginase-1 while the expression of interleukin-6 and tumor necrosis factor-α were down-regulated. RIF in rats was significantly increased following UUO, which was markedly alleviated by TUG891-programmed macrophages but not untreated macrophages. TUG891-programmed macrophages inhibited the abnormal expression of TGF-ß1 and SMAD2. The abnormal expression of epithelial-mesenchymal transition (EMT)-related proteins including vimentin, α-SMA and ß-catenin was also significantly decreased in rats with transplantation of TUG891-programmed macrophages as compared to UUO rats. This study suggests that autologous administration of peritoneal macrophages programmed in vitro by GPR120 agonist to kidney has a protective effect against RIF following UUO.


Assuntos
Nefropatias/patologia , Macrófagos Peritoneais/metabolismo , Substâncias Protetoras/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Obstrução Ureteral/complicações , Animais , Compostos de Bifenilo/farmacologia , Citocinas/metabolismo , Fibrose , Regulação da Expressão Gênica/efeitos dos fármacos , Nefropatias/genética , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/transplante , Masculino , Modelos Biológicos , Fenótipo , Fenilpropionatos/farmacologia , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/genética , Vimentina/metabolismo , beta Catenina/metabolismo
17.
Cell Transplant ; 17(1-2): 211-22, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18468252

RESUMO

Macrophages play a pivotal role in the development of newly formed vascular networks, in addition to their normal immunological functions. This research focuses on peritoneal macrophages as a novel source in cell implantation therapy for ischemic diseases. In this study, production of angiogenic growth factors by peritoneal macrophages and its in vivo effect of neovascularization were evaluated. Mononuclear cells from the peritoneal cavity (P-MNCs) enriched with macrophages were isolated and stimulated with hypoxia and interleukin-1beta (IL-1beta) to mimic an ischemic tissue environment in vitro. Expression of basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) of mRNA in P-MNCs was apparently enhanced by hypoxic stimulation, and the production of VEGF protein was also augmented by hypoxia and IL-1beta. A rat ischemic hind limb model was created and P-MNCs (8 x 10(6)/limb) were injected into the ischemic muscles. The blood flow, which was assessed using the colored microsphere method, showed that the percentage blood flow was significantly increased by P-MNCs injection 4 weeks after surgery (48.3 +/- 16.8% in noninjected ischemic limb vs. 84.3 +/- 13.0% in the P-MNCs-injected limb). A histological analysis revealed that the number of capillaries detected by alkaline phosphatase staining was increased in the P-MNCs group 4 weeks after injection. Furthermore, the number of alpha-smooth muscle actin-positive vessels also showed a significant increase following P-MNC injection. The injected P-MNCs labeled with fluorescence were detected in the interstitial space of ischemic muscles, and VEGF protein expression of the implanted cells was confirmed by immunohistochemistry. These results indicate that peritoneal macrophages stimulate capillary formation and arteriogenesis in the ischemic limbs, possibly through the production of angiogenic growth factors. These findings suggest that the physiological angiogenic property of peritoneal macrophages could therefore be utilized for neovascularization in cell implantation therapy.


Assuntos
Membro Posterior/irrigação sanguínea , Isquemia/terapia , Macrófagos Peritoneais/transplante , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Animais , Hipóxia Celular , Membro Posterior/fisiopatologia , Isquemia/fisiopatologia , Ativação de Macrófagos , Masculino , Músculo Esquelético/fisiopatologia , Ratos , Ratos Endogâmicos Lew
18.
Diabetes ; 67(2): 235-247, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29133512

RESUMO

Adipose-derived stem cells (ADSCs) play critical roles in controlling obesity-associated inflammation and metabolic disorders. Exosomes from ADSCs exert protective effects in several diseases, but their roles in obesity and related pathological conditions remain unclear. In this study, we showed that treatment of obese mice with ADSC-derived exosomes facilitated their metabolic homeostasis, including improved insulin sensitivity (27.8% improvement), reduced obesity, and alleviated hepatic steatosis. ADSC-derived exosomes drove alternatively activated M2 macrophage polarization, inflammation reduction, and beiging in white adipose tissue (WAT) of diet-induced obese mice. Mechanistically, exosomes from ADSCs transferred into macrophages to induce anti-inflammatory M2 phenotypes through the transactivation of arginase-1 by exosome-carried active STAT3. Moreover, M2 macrophages induced by ADSC-derived exosomes not only expressed high levels of tyrosine hydroxylase responsible for catecholamine release, but also promoted ADSC proliferation and lactate production, thereby favoring WAT beiging and homeostasis in response to high-fat challenge. These findings delineate a novel exosome-mediated mechanism for ADSC-macrophage cross talk that facilitates immune and metabolic homeostasis in WAT, thus providing potential therapy for obesity and diabetes.


Assuntos
Adipócitos Bege/patologia , Adipócitos Brancos/patologia , Adipogenia , Células-Tronco Adultas/patologia , Exossomos/transplante , Macrófagos Peritoneais/transplante , Obesidade/terapia , Adipócitos Bege/imunologia , Adipócitos Bege/metabolismo , Adipócitos Brancos/imunologia , Adipócitos Brancos/metabolismo , Células-Tronco Adultas/imunologia , Células-Tronco Adultas/metabolismo , Animais , Biomarcadores/metabolismo , Comunicação Celular , Polaridade Celular , Proliferação de Células , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Exossomos/imunologia , Exossomos/metabolismo , Exossomos/patologia , Resistência à Insulina , Ativação de Macrófagos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Masculino , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Obesidade/imunologia , Obesidade/patologia , Obesidade/fisiopatologia , Fagocitose
19.
Diabetes ; 67(8): 1589-1603, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29871859

RESUMO

Transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI) is a receptor for the TNF superfamily cytokines, B cell-activating factor (BAFF), and A proliferation-inducing ligand (APRIL). Here, we demonstrate that TACI-deficient mice subjected to high-fat diet (HFD) are protected from weight gain and dysregulated glucose homeostasis. Resistance to HFD-induced metabolic changes in TACI-deficient mice does not involve TACI-mediated adipogenesis. Instead, accumulation of M2 macrophages (Mϕs), eosinophils, and type 2 innate lymphoid cells in visceral adipose tissue (VAT) is implicated in the protection from obesity-induced assaults. In support of this hypothesis, adoptively transferred TACI-deficient peritoneal or adipose tissue Mϕs, but not B cells, can improve glucose metabolism in the obese host. Interestingly, the transferred TACI-deficient Mϕs not only home to host VAT but also trigger the accumulation of host M2 Mϕs and eosinophils in VAT. The increase in host M2 Mϕs in VAT is likely a result of eosinophil recruitment in response to eotaxin-2 produced by TACI-deficient Mϕs. Insulin signaling experiments revealed that IL-10 secreted by TACI-deficient Mϕs is responsible for maintaining adipocyte insulin sensitivity. Thus, the adoptive transfer experiments offer a model where TACI-deficient Mϕs accumulate in VAT and protect against metaflammation and obesity-associated dysregulation of glucose metabolism.


Assuntos
Adiposidade , Intolerância à Glucose/prevenção & controle , Imunoterapia Adotiva , Gordura Intra-Abdominal/imunologia , Macrófagos/transplante , Obesidade/terapia , Proteína Transmembrana Ativadora e Interagente do CAML/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Feminino , Regulação da Expressão Gênica , Intolerância à Glucose/etiologia , Intolerância à Glucose/imunologia , Mediadores da Inflamação/sangue , Mediadores da Inflamação/metabolismo , Resistência à Insulina , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Macrófagos Peritoneais/transplante , Camundongos , Camundongos Knockout , Obesidade/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia , Interferência de RNA , Proteína Transmembrana Ativadora e Interagente do CAML/antagonistas & inibidores , Proteína Transmembrana Ativadora e Interagente do CAML/química , Proteína Transmembrana Ativadora e Interagente do CAML/genética , Aumento de Peso
20.
Atherosclerosis ; 274: 120-127, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29772480

RESUMO

BACKGROUND AND AIMS: Proteoglycan 4 (Prg4) has a high structural similarity with the established atherosclerosis-modulating proteoglycan versican, but its role in atherogenesis is still unknown. Therefore, the impact of Prg4 deficiency on macrophage function in vitro and atherosclerosis susceptibility in vivo was investigated. METHODS: The presence and localization of Prg4 was studied in atherosclerotic lesions. Furthermore, the effect of Prg4 deficiency on macrophage foam cell formation, cholesterol efflux and lipopolysaccharide (LPS) response was determined. Finally, susceptibility for atherosclerotic lesion formation was investigated in bone marrow-specific Prg4 knockout (KO) mice. RESULTS: Prg4 mRNA expression was induced 91-fold (p<0.001) in murine initial atherosclerotic lesions and Prg4 protein co-localized with human lesional macrophages. Murine Prg4 KO macrophages showed increased foam cell formation (+2.1-fold, p<0.01). In parallel, the expression of the cholesterol efflux genes ATP-binding cassette transporter A1 and scavenger receptor type B1 was lower (-35%, p<0.05;-40%, p<0.05) in Prg4 KO macrophages. This translated into an impaired cholesterol efflux to high-density lipoprotein (-13%, p<0.001) and apolipoprotein A1 (-8%, p<0.05). Furthermore, Prg4 KO macrophages showed an impaired LPS-induced rise in TNFα secretion as compared to wild-type controls (-31%, p<0.001), indicating a reduced inflammatory response. Combined, these pro- and anti-atherogenic effects did not translate into a significant difference in atherosclerotic lesion formation upon bone marrow-specific deletion of Prg4 in low-density lipoprotein receptor KO mice. CONCLUSIONS: Prg4 is present in macrophages in both murine and human atherosclerotic lesions and critically influences macrophage function, but deletion of Prg4 in bone marrow-derived cells does not affect atherosclerotic lesion development.


Assuntos
Aterosclerose/metabolismo , Células da Medula Óssea/metabolismo , Artérias Carótidas/metabolismo , Doenças das Artérias Carótidas/metabolismo , Células Espumosas/metabolismo , Macrófagos Peritoneais/metabolismo , Placa Aterosclerótica , Proteoglicanas/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/patologia , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/patologia , Células Cultivadas , Colesterol/metabolismo , Modelos Animais de Doenças , Células Espumosas/efeitos dos fármacos , Células Espumosas/patologia , Células Espumosas/transplante , Humanos , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/patologia , Macrófagos Peritoneais/transplante , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Fenótipo , Proteoglicanas/deficiência , Proteoglicanas/genética , Receptores Depuradores Classe B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA