Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Cell ; 171(2): 331-345.e22, 2017 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-28942921

RESUMO

Clearance of apoptotic cells (ACs) by phagocytes (efferocytosis) prevents post-apoptotic necrosis and dampens inflammation. Defective efferocytosis drives important diseases, including atherosclerosis. For efficient efferocytosis, phagocytes must be able to internalize multiple ACs. We show here that uptake of multiple ACs by macrophages requires dynamin-related protein 1 (Drp1)-mediated mitochondrial fission, which is triggered by AC uptake. When mitochondrial fission is disabled, AC-induced increase in cytosolic calcium is blunted owing to mitochondrial calcium sequestration, and calcium-dependent phagosome formation around secondarily encountered ACs is impaired. These defects can be corrected by silencing the mitochondrial calcium uniporter (MCU). Mice lacking myeloid Drp1 showed defective efferocytosis and its pathologic consequences in the thymus after dexamethasone treatment and in advanced atherosclerotic lesions in fat-fed Ldlr-/- mice. Thus, mitochondrial fission in response to AC uptake is a critical process that enables macrophages to clear multiple ACs and to avoid the pathologic consequences of defective efferocytosis in vivo.


Assuntos
Macrófagos/citologia , Dinâmica Mitocondrial , Animais , Apoptose , Humanos , Macrófagos/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Células Mieloides/metabolismo , Fagócitos/metabolismo , Fagossomos/metabolismo
2.
Immunity ; 49(4): 666-677.e6, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30291029

RESUMO

Regulatory T (Treg) cell responses and apoptotic cell clearance (efferocytosis) represent critical arms of the inflammation resolution response. We sought to determine whether these processes might be linked through Treg-cell-mediated enhancement of efferocytosis. In zymosan-induced peritonitis and lipopolysaccharide-induced lung injury, Treg cells increased early in resolution, and Treg cell depletion decreased efferocytosis. In advanced atherosclerosis, where defective efferocytosis drives disease progression, Treg cell expansion improved efferocytosis. Mechanistic studies revealed the following sequence: (1) Treg cells secreted interleukin-13 (IL-13), which stimulated IL-10 production in macrophages; (2) autocrine-paracrine signaling by IL-10 induced Vav1 in macrophages; and (3) Vav1 activated Rac1 to promote apoptotic cell engulfment. In summary, Treg cells promote macrophage efferocytosis during inflammation resolution via a transcellular signaling pathway that enhances apoptotic cell internalization. These findings suggest an expanded role of Treg cells in inflammation resolution and provide a mechanistic basis for Treg-cell-enhancement strategies for non-resolving inflammatory diseases.


Assuntos
Apoptose/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Linfócitos T Reguladores/imunologia , Animais , Linhagem Celular , Células Cultivadas , Humanos , Inflamação/metabolismo , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-10/metabolismo , Interleucina-13/genética , Interleucina-13/imunologia , Interleucina-13/metabolismo , Células Jurkat , Lipopolissacarídeos , Pneumopatias/induzido quimicamente , Pneumopatias/imunologia , Pneumopatias/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peritonite/induzido quimicamente , Peritonite/imunologia , Peritonite/metabolismo , Linfócitos T Reguladores/metabolismo , Zimosan
3.
J Cell Mol Med ; 26(20): 5267-5276, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36098213

RESUMO

Hypercholesterolemia exacerbates autoimmune response and accelerates the progression of several autoimmune disorders, but the mechanistic basis is not well understood. We recently demonstrated that hypercholesterolemia is associated with increased serum extracellular DNA levels secondary to a defect in DNase-mediated clearance of DNA. In this study, we tested whether the impaired DNase response plays a causal role in enhancing anti-nuclear antibody levels and renal immune complex deposition in an Apoe-/- mouse model of hypercholesterolemia. We demonstrate that hypercholesterolemic mice have enhanced anti-ds-DNA and anti-nucleosome antibody levels which is associated with increased immune complex deposition in the renal glomerulus. Importantly, treatment with DNase1 led to a decrease in both the autoantibody levels as well as renal pathology. Additionally, we show that humans with hypercholesterolemia have decreased systemic DNase activity and increased anti-nuclear antibodies. In this context, our data suggest that recombinant DNase1 may be an attractive therapeutic strategy to lower autoimmune response and disease progression in patients with autoimmune disorders associated with concomitant hypercholesterolemia.


Assuntos
Doenças Autoimunes , Desoxirribonucleases , Hipercolesterolemia , Lúpus Eritematoso Sistêmico , Animais , Complexo Antígeno-Anticorpo , Autoanticorpos , DNA , Desoxirribonucleases/metabolismo , Humanos , Hipercolesterolemia/genética , Camundongos , Camundongos Knockout para ApoE
4.
Arterioscler Thromb Vasc Biol ; 41(10): 2598-2615, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34348488

RESUMO

Objective: Hypercholesterolemia-induced NETosis and accumulation of neutrophil extracellular traps (NETs) in the atherosclerotic lesion exacerbates inflammation and is causally implicated in plaque progression. We investigated whether hypercholesterolemia additionally impairs the clearance of NETs mediated by endonucleases such as DNase1 and DNase1L3 and its implication in advanced atherosclerotic plaque progression. Approach and Results: Using a mouse model, we demonstrate that an experimental increase in the systemic level of NETs leads to a rapid increase in serum DNase activity, which is critical for the prompt clearance of NETs and achieving inflammation resolution. Importantly, hypercholesterolemic mice demonstrate an impairment in this critical NET-induced DNase response with consequent delay in the clearance of NETs and defective inflammation resolution. Administration of tauroursodeoxycholic acid, a chemical chaperone that relieves endoplasmic reticulum stress, rescued the hypercholesterolemia-induced impairment in the NET-induced DNase response suggesting a causal role for endoplasmic reticulum stress in this phenomenon. Correction of the defective DNase response with exogenous supplementation of DNase1 in Apoe-/- mice with advanced atherosclerosis resulted in a decrease in plaque NET content and significant plaque remodeling with decreased area of plaque necrosis and increased collagen content. From a translational standpoint, we demonstrate that humans with hypercholesterolemia have elevated systemic extracellular DNA levels and decreased plasma DNase activity. Conclusions: These data suggest that hypercholesterolemia impairs the NET-induced DNase response resulting in defective clearance and accumulation of NETs in the atherosclerotic plaque. Therefore, strategies aimed at rescuing this defect could be of potential therapeutic benefit in promoting inflammation resolution and atherosclerotic plaque stabilization.


Assuntos
Doenças da Aorta/etiologia , Aterosclerose/etiologia , Armadilhas Extracelulares/metabolismo , Hipercolesterolemia/complicações , Mediadores da Inflamação/metabolismo , Inflamação/etiologia , Neutrófilos/metabolismo , Placa Aterosclerótica , Animais , Doenças da Aorta/imunologia , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Células CACO-2 , Desoxirribonuclease I/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Endodesoxirribonucleases/metabolismo , Estresse do Retículo Endoplasmático , Feminino , Células HL-60 , Células Hep G2 , Humanos , Hipercolesterolemia/imunologia , Hipercolesterolemia/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Necrose , Neutrófilos/imunologia , Transdução de Sinais , Células THP-1
7.
Pharmacol Res ; 170: 105699, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34087352

RESUMO

Phagocytic clearance of dead cells and debris is critical for inflammation resolution and maintenance of tissue homeostasis. Consequently, defective clearance of dead cells and debris is associated with initiation and exacerbation of several autoimmune disorders and chronic inflammatory diseases such as atherosclerosis. The progressive loss of dead cell clearance capacity within the atherosclerotic plaque leads to accumulation of necrotic cells, chronic non-resolving inflammation, and expansion of the necrotic core, which triggers atherosclerotic plaque rupture and clinical manifestation of acute thrombotic cardiovascular adverse events. In this review, we describe the fundamental molecular and cellular mechanisms of dead cell clearance and how it goes awry in atherosclerosis. Finally, we highlight novel therapeutic strategies that enhance dead cell and debris clearance within the atherosclerotic plaque to promote inflammation resolution and atherosclerotic plaque stabilization.


Assuntos
Aterosclerose/patologia , Inflamação/patologia , Macrófagos/patologia , Fagocitose , Placa Aterosclerótica , Animais , Anti-Inflamatórios/uso terapêutico , Aterosclerose/tratamento farmacológico , Aterosclerose/imunologia , Aterosclerose/metabolismo , Morte Celular , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Necrose , Fagocitose/efeitos dos fármacos
8.
Nature ; 506(7489): 503-6, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24463523

RESUMO

The tissue-resident macrophages of barrier organs constitute the first line of defence against pathogens at the systemic interface with the ambient environment. In the lung, resident alveolar macrophages (AMs) provide a sentinel function against inhaled pathogens. Bacterial constituents ligate Toll-like receptors (TLRs) on AMs, causing AMs to secrete proinflammatory cytokines that activate alveolar epithelial receptors, leading to recruitment of neutrophils that engulf pathogens. Because the AM-induced response could itself cause tissue injury, it is unclear how AMs modulate the response to prevent injury. Here, using real-time alveolar imaging in situ, we show that a subset of AMs attached to the alveolar wall form connexin 43 (Cx43)-containing gap junction channels with the epithelium. During lipopolysaccharide-induced inflammation, the AMs remained sessile and attached to the alveoli, and they established intercommunication through synchronized Ca(2+) waves, using the epithelium as the conducting pathway. The intercommunication was immunosuppressive, involving Ca(2+)-dependent activation of Akt, because AM-specific knockout of Cx43 enhanced alveolar neutrophil recruitment and secretion of proinflammatory cytokines in the bronchoalveolar lavage. A picture emerges of a novel immunomodulatory process in which a subset of alveolus-attached AMs intercommunicates immunosuppressive signals to reduce endotoxin-induced lung inflammation.


Assuntos
Comunicação Celular , Macrófagos Alveolares/citologia , Macrófagos Alveolares/imunologia , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/imunologia , Mucosa Respiratória/citologia , Mucosa Respiratória/imunologia , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Cálcio/metabolismo , Adesão Celular , Conexina 43/deficiência , Conexina 43/genética , Conexina 43/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Feminino , Junções Comunicantes/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Neutrófilos/imunologia , Pneumonia/induzido quimicamente , Pneumonia/imunologia , Pneumonia/patologia
9.
Cytokine ; 122: 154190, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-29074034

RESUMO

Colony stimulating factors (CSFs) play a central role in the development and functional maturation of immune cells besides having pleiotropic effects on cells of the vascular wall. The production of CSFs is induced by multiple atherogenic and inflammatory stimuli and their expression levels are often correlated positively with advanced atherosclerotic plaques and adverse cardiovascular events in humans suggesting that CSFs play a critical role in the pathophysiology of atherosclerosis progression. Interestingly, recombinant CSFs as well as anti-CSFs are being increasingly used for diverse clinical indications. However, the effect of these novel therapeutics on atherosclerotic plaque progression is not well understood. Herein, we summarize the currently available literature on the complex role of CSFs in various stages of atherosclerosis and emphasize the necessity for conducting further mechanistic studies in animal models of atherosclerosis as well as the need for evaluating the cardiovascular safety of CSF-based therapies in humans.


Assuntos
Aterosclerose/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Fator Estimulador de Colônias de Macrófagos/metabolismo , Animais , Aterosclerose/metabolismo , Diferenciação Celular/efeitos dos fármacos , Fatores Estimuladores de Colônias/metabolismo , Fatores Estimuladores de Colônias/farmacologia , Progressão da Doença , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-3/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Modelos Animais , Placa Aterosclerótica/tratamento farmacológico , Placa Aterosclerótica/metabolismo
10.
Proc Natl Acad Sci U S A ; 113(23): 6526-31, 2016 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-27199481

RESUMO

The acute inflammatory response requires a coordinated resolution program to prevent excessive inflammation, repair collateral damage, and restore tissue homeostasis, and failure of this response contributes to the pathology of numerous chronic inflammatory diseases. Resolution is mediated in part by long-chain fatty acid-derived lipid mediators called specialized proresolving mediators (SPMs). However, how SPMs are regulated during the inflammatory response, and how this process goes awry in inflammatory diseases, are poorly understood. We now show that signaling through the Mer proto-oncogene tyrosine kinase (MerTK) receptor in cultured macrophages and in sterile inflammation in vivo promotes SPM biosynthesis by a mechanism involving an increase in the cytoplasmic:nuclear ratio of a key SPM biosynthetic enzyme, 5-lipoxygenase. This action of MerTK is linked to the resolution of sterile peritonitis and, after ischemia-reperfusion (I/R) injury, to increased circulating SPMs and decreased remote organ inflammation. MerTK is susceptible to ADAM metallopeptidase domain 17 (ADAM17)-mediated cell-surface cleavage under inflammatory conditions, but the functional significance is not known. We show here that SPM biosynthesis is increased and inflammation resolution is improved in a new mouse model in which endogenous MerTK was replaced with a genetically engineered variant that is cleavage-resistant (Mertk(CR)). Mertk(CR) mice also have increased circulating levels of SPMs and less lung injury after I/R. Thus, MerTK cleavage during inflammation limits SPM biosynthesis and the resolution response. These findings contribute to our understanding of how SPM synthesis is regulated during the inflammatory response and suggest new therapeutic avenues to boost resolution in settings where defective resolution promotes disease progression.


Assuntos
Mediadores da Inflamação/metabolismo , Inflamação/etiologia , Inflamação/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Proteína ADAM17/metabolismo , Animais , Araquidonato 5-Lipoxigenase/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Inflamação/patologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peritonite/etiologia , Peritonite/metabolismo , Peritonite/patologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/deficiência , Receptores Proteína Tirosina Quinases/genética , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais , c-Mer Tirosina Quinase
11.
Circ Res ; 116(2): e13-24, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25348165

RESUMO

RATIONALE: Granulocyte macrophage colony-stimulating factor (GM-CSF, Csf2) is a growth factor for myeloid-lineage cells that has been implicated in the pathogenesis of atherosclerosis and other chronic inflammatory diseases. However, the role of GM-CSF in advanced atherosclerotic plaque progression, the process that gives rise to clinically dangerous plaques, is unknown. OBJECTIVE: To understand the role of GM-CSF in advanced atherosclerotic plaque progression. METHODS AND RESULTS: Ldlr(-/-) mice and Csf2(-/-)Ldlr(-/-) mice were fed a Western-type diet for 12 weeks, and then parameters of advanced plaque progression in the aortic root were quantified. Lesions from the GM-CSF-deficient mice showed a substantial decrease in 2 key hallmarks of advanced atherosclerosis, lesional macrophage apoptosis and plaque necrosis, which indicates that GM-CSF promotes plaque progression. Based on a combination of in vitro and in vivo studies, we show that the mechanism involves GM-CSF-mediated production of interleukin-23, which increases apoptosis susceptibility in macrophages by promoting proteasomal degradation of the cell survival protein Bcl-2 (B-cell lymphoma 2) and by increasing oxidative stress. CONCLUSIONS: In low-density lipoprotein-driven atherosclerosis in mice, GM-CSF promotes advanced plaque progression by increasing macrophage apoptosis susceptibility. This action of GM-CSF is mediated by its interleukin-23-inducing activity rather than its role as a growth factor.


Assuntos
Apoptose/fisiologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/deficiência , Interleucina-23/metabolismo , Macrófagos/metabolismo , Placa Aterosclerótica/metabolismo , Transdução de Sinais/fisiologia , Animais , Peptídeos e Proteínas de Sinalização Intercelular , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose/metabolismo , Necrose/patologia , Placa Aterosclerótica/patologia
12.
Proc Natl Acad Sci U S A ; 110(16): 6506-11, 2013 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-23533277

RESUMO

Excessive inflammation and failed resolution of the inflammatory response are underlying components of numerous conditions such as arthritis, cardiovascular disease, and cancer. Hence, therapeutics that dampen inflammation and enhance resolution are of considerable interest. In this study, we demonstrate the proresolving activity of sub-100-nm nanoparticles (NPs) containing the anti-inflammatory peptide Ac2-26, an annexin A1/lipocortin 1-mimetic peptide. These NPs were engineered using biodegradable diblock poly(lactic-co-glycolic acid)-b-polyethyleneglycol and poly(lactic-co-glycolic acid)-b-polyethyleneglycol collagen IV-targeted polymers. Using a self-limited zymosan-induced peritonitis model, we show that the Ac2-26 NPs (100 ng per mouse) were significantly more potent than Ac2-26 native peptide at limiting recruitment of polymononuclear neutrophils (56% vs. 30%) and at decreasing the resolution interval up to 4 h. Moreover, systemic administration of collagen IV targeted Ac2-26 NPs (in as low as 1 µg peptide per mouse) was shown to significantly block tissue damage in hind-limb ischemia-reperfusion injury by up to 30% in comparison with controls. Together, these findings demonstrate that Ac2-26 NPs are proresolving in vivo and raise the prospect of their use in chronic inflammatory diseases such as atherosclerosis.


Assuntos
Anexina A1/farmacologia , Anexina A1/uso terapêutico , Nanopartículas/uso terapêutico , Nanotecnologia/métodos , Neutrófilos/efeitos dos fármacos , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Peritonite/tratamento farmacológico , Traumatismo por Reperfusão/tratamento farmacológico , Análise de Variância , Animais , Colágeno Tipo IV/metabolismo , Feminino , Citometria de Fluxo , Membro Posterior/efeitos dos fármacos , Membro Posterior/patologia , Cinética , Camundongos , Camundongos Endogâmicos C57BL
13.
Arterioscler Thromb Vasc Biol ; 34(5): 976-84, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24651678

RESUMO

OBJECTIVE: Coronary heart disease is associated with monocytosis. Studies using animal models of monocytosis and atherosclerosis such as ApoE(-/-) mice have shown bone marrow (BM) hematopoietic stem and multipotential progenitor cell (HSPC) expansion, associated with increased cell surface expression of the common ß subunit of the granulocyte macrophage colony-stimulating factor/interleukin-3 receptor (CBS) on HSPCs. ApoE(-/-) mice also display increased granulocyte macrophage colony-stimulating factor-dependent monocyte production in the spleen. We investigated the role of the CBS in cholesterol-driven HSPC expansion, monocytosis, and atherosclerosis. APPROACH AND RESULTS: Ldlr(-/-) mice were transplanted with ApoE(-/-)Cbs(-/-) or ApoE(-/-) BM followed by Western-type diet feeding. Compared with ApoE(-/-) BM-transplanted controls, ApoE(-/-)Cbs(-/-) BM-transplanted mice had reduced BM and splenic HSPC proliferation, fewer blood monocytes and neutrophils, and reduced macrophage content and area of early atherosclerotic lesions. More advanced lesions showed diminished macrophage and collagen content; however, lesion size was unchanged, reflecting an increase in necrotic core area, associated with a marked decrease in Abcg1 expression and increased macrophage apoptosis. Compared with wild-type mice, Western-type diet-fed ApoE(-/-) mice showed increased CBS expression on granulocyte macrophage colony-stimulating factor-producing innate response activator B cells and expansion of this population. ApoE(-/-)Cbs(-/-) BM-transplanted Ldlr(-/-) mice showed a marked decrease in innate response activator B cells compared with ApoE(-/-) BM-transplanted Ldlr(-/-) controls. CONCLUSIONS: Increased levels of CBS on HSPCs and splenic innate response activator B cells lead to expansion of these populations in ApoE(-/-) BM-transplanted Ldlr(-/-) mice, contributing to monocytosis and increased lesional macrophage content. However, in more advanced lesions, the CBS also has a role in atherosclerotic plaque stabilization.


Assuntos
Doenças da Aorta/metabolismo , Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Diferenciação Celular , Proliferação de Células , Subunidade beta Comum dos Receptores de Citocinas/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Macrófagos/metabolismo , Monócitos/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Doenças da Aorta/genética , Doenças da Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Subunidade beta Comum dos Receptores de Citocinas/deficiência , Subunidade beta Comum dos Receptores de Citocinas/genética , Modelos Animais de Doenças , Progressão da Doença , Células-Tronco Hematopoéticas/patologia , Imunidade Inata , Lipoproteínas/metabolismo , Macrófagos/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/patologia , Necrose , Placa Aterosclerótica , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais , Baço/metabolismo , Baço/patologia , Fatores de Tempo
14.
Cell Rep ; 42(7): 112808, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37436891

RESUMO

Macrophages release soluble mediators following efferocytic clearance of apoptotic cells to facilitate intercellular communication and promote the resolution of inflammation. However, whether inflammation resolution is modulated by extracellular vesicles (EVs) and vesicular mediators released by efferocytes is not known. We report that efferocyte-derived EVs express prosaposin, which binds to macrophage GPR37 to increase expression of the efferocytosis receptor Tim4 via an ERK-AP1-dependent signaling axis, leading to increased macrophage efferocytosis efficiency and accelerated resolution of inflammation. Neutralization and knockdown of prosaposin or blocking GRP37 abrogates the pro-resolution effects of efferocyte-derived EVs in vivo. Administration of efferocyte-derived EVs in a murine model of atherosclerosis is associated with an increase in lesional macrophage efferocytosis efficiency and a decrease in plaque necrosis and lesional inflammation. Thus, we establish a critical role for efferocyte-derived vesicular mediators in increasing macrophage efferocytosis efficiency and accelerating the resolution of inflammation and tissue injury.


Assuntos
Vesículas Extracelulares , Saposinas , Animais , Camundongos , Apoptose , Vesículas Extracelulares/metabolismo , Inflamação/metabolismo , Macrófagos/metabolismo , Fagocitose , Saposinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
15.
J Biol Chem ; 286(38): 33335-44, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21828049

RESUMO

Mer tyrosine kinase (MerTK) is an integral membrane protein that is preferentially expressed by phagocytic cells, where it promotes efferocytosis and inhibits inflammatory signaling. Proteolytic cleavage of MerTK at an unidentified site leads to shedding of its soluble ectodomain (soluble MER; sMER), which can inhibit thrombosis in mice and efferocytosis in vitro. Herein, we show that MerTK is cleaved at proline 485 in murine macrophages. Site-directed deletion of 6 amino acids spanning proline 485 rendered MerTK resistant to proteolysis and suppression of efferocytosis by cleavage-inducing stimuli. LPS is a known inducer of MerTK cleavage, and the intracellular signaling pathways required for this action are unknown. LPS/TLR4-mediated generation of sMER required disintegrin and metalloproteinase ADAM17 and was independent of Myd88, instead requiring TRIF adaptor signaling. LPS-induced cleavage was suppressed by deficiency of NADPH oxidase 2 (Nox2) and PKCδ. The addition of the antioxidant N-acetyl cysteine inhibited PKCδ, and silencing of PKCδ inhibited MAPK p38, which was also required. In a mouse model of endotoxemia, we discovered that LPS induced plasma sMER, and this was suppressed by Adam17 deficiency. Thus, a TRIF-mediated pattern recognition receptor signaling cascade requires NADPH oxidase to activate PKCδ and then p38, culminating in ADAM17-mediated proteolysis of MerTK. These findings link innate pattern recognition receptor signaling to proteolytic inactivation of MerTK and generation of sMER and uncover targets to test how MerTK cleavage affects efferocytosis efficiency and inflammation resolution in vivo.


Assuntos
Proteínas ADAM/metabolismo , Proteína Quinase C-delta/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas ADAM/química , Proteína ADAM17 , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Sequência de Aminoácidos , Animais , Lipopolissacarídeos/farmacologia , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Fator 88 de Diferenciação Mieloide/metabolismo , NADP/deficiência , NADP/metabolismo , Prolina/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Deleção de Sequência , Transdução de Sinais/efeitos dos fármacos , Solubilidade/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , c-Mer Tirosina Quinase
16.
Eur J Immunol ; 41(9): 2515-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21952808

RESUMO

Accumulating evidence supports the notion that defective phagocytic clearance of dying cells, or defective "efferocytosis," is causally linked to the progression of advanced atherosclerosis. In advanced atherosclerotic lesions, defective efferocytosis leads to post-apoptotic necrosis, expansion of plaque necrotic cores, and susceptibility to atherothrombosis. Both macrophages and DC-like efferocytes are juxtaposed near expanding necrotic cores, where they engage apoptotic cells. In this Viewpoint, we discuss how reduced efferocytosis by macrophages and CD11c(HI) DC-like cells may combine to reduce overall plaque stability and therefore promote susceptibility to acute atherothrombosis.


Assuntos
Aterosclerose/imunologia , Células Dendríticas/imunologia , Macrófagos/imunologia , Necrose/imunologia , Animais , Apoptose/imunologia , Aterosclerose/fisiopatologia , Antígeno CD11c , Progressão da Doença , Suscetibilidade a Doenças , Humanos , Fagocitose/imunologia , Trombose
17.
Curr Opin Pharmacol ; 66: 102283, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36037627

RESUMO

Atherosclerosis is a lipid-driven disease of the artery characterized by chronic non-resolving inflammation. Despite availability of excellent lipid-lowering therapies, atherosclerosis remains the leading cause of disability and death globally. The demonstration that suppressing inflammation prevents the adverse clinical manifestations of atherosclerosis in recent clinical trials has led to heightened interest in anti-inflammatory therapies. In this review, we briefly highlight some key anti-inflammatory and pro-resolution pathways, which could be targeted to modulate pathogenesis and stall atherosclerosis progression. We also highlight key challenges that must be overcome to turn the concept of inflammation targeting therapies into clinical reality for atherosclerotic heart disease.


Assuntos
Aterosclerose , Doença da Artéria Coronariana , Anti-Inflamatórios/uso terapêutico , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Aterosclerose/patologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/patologia , Lipídeos
18.
Front Pharmacol ; 13: 888300, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35721222

RESUMO

Atherosclerosis is a chronic lipid-driven inflammatory condition of the arteries and is a leading cause of stroke, myocardial infarction, and other peripheral arterial diseases. Plant products rich in polyphenols such as pomegranate juice and peel extract are known to have beneficial effects in suppressing atherogenesis. However, the mechanism of action and its effect on advanced atherosclerosis progression which results in adverse clinical outcomes are not well understood. Herein, we use a standardized hydroethanolic extract of Punica granatum (pomegranate) peel in the Apoe -/- a murine model of advanced atherosclerosis. It was observed that the pomegranate peel extract fed mice have decreased plaque necrosis and elevated lesional collagen content which was associated with a favorable metabolic profile including lowering of blood glucose, cholesterol, and triglyceride. The decrease in plaque necrosis was linked with increased lesional macrophage efferocytosis efficiency which was associated with enhanced expression of the efferocytosis receptor Mertk. Using in vitro studies, we show that pomegranate peel extract blocks the shedding of Mertk and preserves macrophage efferocytosis efficiency. These data identify a novel mechanism by which pomegranate peel extract promotes the resolution of inflammation in atherosclerosis.

19.
Cell Metab ; 33(8): 1509-1511, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34348095

RESUMO

The tumor microenvironment is immunosuppressive. Here we preview two recent studies from Ma et al. (2021) in Cell Metabolism and Xu et al. (2021) in Immunity that describe a key role of T cell-expressed CD36 in enhancing lipid uptake and mediating lipid peroxidation that ultimately leads to CD8+ T cell dysfunction, ferroptosis, and reduced anti-tumor function.


Assuntos
Ferroptose , Neoplasias , Antígenos CD36 , Linfócitos T CD8-Positivos , Humanos , Microambiente Tumoral
20.
J Clin Invest ; 131(6)2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33529176

RESUMO

Tyro3, AXL, and MerTK (TAM) receptors are activated in macrophages in response to tissue injury and as such have been proposed as therapeutic targets to promote inflammation resolution during sterile wound healing, including myocardial infarction. Although the role of MerTK in cardioprotection is well characterized, the unique role of the other structurally similar TAMs, and particularly AXL, in clinically relevant models of myocardial ischemia/reperfusion infarction (IRI) is comparatively unknown. Utilizing complementary approaches, validated by flow cytometric analysis of human and murine macrophage subsets and conditional genetic loss and gain of function, we uncover a maladaptive role for myeloid AXL during IRI in the heart. Cross signaling between AXL and TLR4 in cardiac macrophages directed a switch to glycolytic metabolism and secretion of proinflammatory IL-1ß, leading to increased intramyocardial inflammation, adverse ventricular remodeling, and impaired contractile function. AXL functioned independently of cardioprotective MerTK to reduce the efficacy of cardiac repair, but like MerTK, was proteolytically cleaved. Administration of a selective small molecule AXL inhibitor alone improved cardiac healing, which was further enhanced in combination with blockade of MerTK cleavage. These data support further exploration of macrophage TAM receptors as therapeutic targets for myocardial infarction.


Assuntos
Macrófagos/metabolismo , Infarto do Miocárdio/complicações , Infarto do Miocárdio/metabolismo , Miocardite/etiologia , Miocardite/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Humanos , Inflamassomos/metabolismo , Ativação de Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica/complicações , Traumatismo por Reperfusão Miocárdica/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Receptor Cross-Talk , Receptores Proteína Tirosina Quinases/deficiência , Receptores Proteína Tirosina Quinases/genética , Infarto do Miocárdio com Supradesnível do Segmento ST/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , c-Mer Tirosina Quinase/deficiência , c-Mer Tirosina Quinase/genética , c-Mer Tirosina Quinase/metabolismo , Receptor Tirosina Quinase Axl
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa