Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Leukoc Biol ; 106(4): 803-814, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31166619

RESUMO

Obesity and diabetes are associated with macrophage dysfunction and increased NLRP3 inflammasome activation. Saturated fatty acids (FAs) are abundant in these metabolic disorders and have been associated with lysosome dysfunction and inflammasome activation in macrophages. However, the interplay between cellular metabolic pathways and lipid-induced toxicity in macrophages remains poorly understood. In this study, we investigated the role of the lipid metabolic enzyme long chain acyl-CoA synthetase (ACSL1) in primary macrophages. ACSL1 is upregulated in TLR4-activated macrophages via a TIR (toll/IL-1R) domain-containing adapter inducing IFN-ß (TRIF)-dependent pathway, and knockout of this enzyme decreased NLRP3 inflammasome activation. The mechanism of this response was not related to inflammasome priming, lipid uptake, or endoplasmic reticulum (ER) stress generation. Rather, ACSL1 was associated with mitochondria where it modulated fatty acid metabolism. The development of lysosome damage with palmitate exposure likely occurs via the formation of intracellular crystals. Herein, we provide evidence that loss of ACSL1 in macrophages decreases FA crystal formation thereby reducing lysosome damage and IL-1ß release. These findings suggest that targeting lipid metabolic pathways in macrophages may be a strategy to reduce lipotoxity and to decrease pathologic inflammation in metabolic disease.


Assuntos
Coenzima A Ligases/metabolismo , Inflamassomos/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Palmitatos/toxicidade , Animais , Células Cultivadas , Coenzima A Ligases/deficiência , Humanos , Interleucina-1beta/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Lisossomos/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/ultraestrutura , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
J Immunol ; 201(7): 2054-2069, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30143592

RESUMO

Obesity and diabetes modulate macrophage activation, often leading to prolonged inflammation and dysfunctional tissue repair. Increasing evidence suggests that the NLRP3 inflammasome plays an important role in obesity-associated inflammation. We have previously shown that activation of the lipotoxic inflammasome by excess fatty acids in macrophages occurs via a lysosome-dependent pathway. However, the mechanisms that link cellular lipid metabolism to altered inflammation remain poorly understood. PPARγ is a nuclear receptor transcription factor expressed by macrophages that is known to alter lipid handling, mitochondrial function, and inflammatory cytokine expression. To undercover novel links between metabolic signaling and lipotoxic inflammasome activation, we investigated mouse primary macrophages deficient in PPARγ. Contrary to our expectation, PPARγ knockout (KO) macrophages released significantly less IL-1ß and IL-1α in response to lipotoxic stimulation. The suppression occurred at the transcriptional level and was apparent for multiple activators of the NLRP3 inflammasome. RNA sequencing revealed upregulation of IFN-ß in activated PPARγKO macrophages, and this was confirmed at the protein level. A blocking Ab against the type 1 IFNR restored the release of IL-1ß to wild type levels in PPARγKO cells, confirming the mechanistic link between these events. Conversely, PPARγ activation with rosiglitazone selectively suppressed IFN-ß expression in activated macrophages. Loss of PPARγ also resulted in diminished expression of genes involved in sterol biosynthesis, a pathway known to influence IFN production. Together, these findings demonstrate a cross-talk pathway that influences the interplay between metabolism and inflammation in macrophages.


Assuntos
Inflamassomos/metabolismo , Inflamação/imunologia , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/fisiologia , Obesidade/imunologia , PPAR gama/genética , Animais , Células Cultivadas , Interferon Tipo I/metabolismo , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Rosiglitazona/farmacologia , Análise de Sequência de RNA
3.
Proc Natl Acad Sci U S A ; 111(45): 16029-34, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25349429

RESUMO

The mechanistic basis for why inflammation is simultaneously both deleterious and essential for tissue repair is not fully understood. Recently, a new paradigm has emerged: Organs are replete with resident macrophages of embryonic origin distinct from monocyte-derived macrophages. This added complexity raises the question of whether distinct immune cells drive inflammatory and reparative activities after injury. Previous work has demonstrated that the neonatal heart has a remarkable capacity for tissue repair compared with the adult heart, offering an ideal context to examine these concepts. We hypothesized that unrecognized differences in macrophage composition is a key determinant of cardiac tissue repair. Using a genetic model of cardiomyocyte ablation, we demonstrated that neonatal mice expand a population of embryonic-derived resident cardiac macrophages, which generate minimal inflammation and promote cardiac recovery through cardiomyocyte proliferation and angiogenesis. During homeostasis, the adult heart contains embryonic-derived macrophages with similar properties. However, after injury, these cells were replaced by monocyte-derived macrophages that are proinflammatory and lacked reparative activities. Inhibition of monocyte recruitment to the adult heart preserved embryonic-derived macrophage subsets, reduced inflammation, and enhanced tissue repair. These findings indicate that embryonic-derived macrophages are key mediators of cardiac recovery and suggest that therapeutics targeting distinct macrophage lineages may serve as novel treatments for heart failure.


Assuntos
Embrião de Mamíferos/metabolismo , Macrófagos/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Recuperação de Função Fisiológica , Remodelação Ventricular , Animais , Inflamação/metabolismo , Inflamação/terapia , Camundongos , Miócitos Cardíacos/transplante , Regeneração
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA