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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
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
2.
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
3.
J Biol Chem ; 289(13): 9158-71, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24532802

RESUMO

Macrophage dysfunction and inflammasome activation have been implicated in the pathogenesis of diabetes and its complications. Prolonged inflammation and impaired healing are hallmarks of the diabetic response to tissue injury, and excessive inflammasome activation has been associated in these phenotypes. However, the mechanisms that regulate the inflammasome in response to lipid metabolic and inflammatory stress are incompletely understood. We have shown previously that IL-1ß secretion is induced in primary macrophages exposed to the dietary saturated fatty acid palmitate in combination with LPS. In this study, we sought to unravel the mechanisms underlying the activation of this lipotoxic inflammasome. We demonstrate that palmitate-loaded primary macrophages challenged with LPS activate the NLRP3 inflammasome through a mechanism that involves the lysosome. Interestingly, the lysosome was involved in both the regulation of pro-IL-1ß levels and its subsequent cleavage/release. The lysosomal protease cathepsin B was required for IL-1ß release but not pro-IL-1ß production. In contrast, disrupting lysosomal calcium regulation decreased IL-1ß release by reducing pro-IL-1ß levels. The calcium pathway involved the calcium-activated phosphatase calcineurin, which stabilized IL-1ß mRNA. Our findings provide evidence that the lysosome plays a key role in both the priming and assembly phases of the lipostoxic inflammasome. These findings have potential relevance to the hyperinflammatory phenotypes observed in diabetics during tissue damage or infection and identify lysosomes and calcineurin as potential therapeutic targets.


Assuntos
Inflamassomos/metabolismo , Lisossomos/metabolismo , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/metabolismo , Animais , Calcineurina/metabolismo , Inibidores de Calcineurina , Cálcio/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular , Ácidos Graxos/farmacologia , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Palmitatos/farmacologia , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/química , RNA Mensageiro/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
J Biol Chem ; 288(5): 2923-32, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23250746

RESUMO

Macrophages play a key role in host defense and in tissue repair after injury. Emerging evidence suggests that macrophage dysfunction in states of lipid excess can contribute to the development of insulin resistance and may underlie inflammatory complications of diabetes. Ceramides are sphingolipids that modulate a variety of cellular responses including cell death, autophagy, insulin signaling, and inflammation. In this study we investigated the intersection between TLR4-mediated inflammatory signaling and saturated fatty acids with regard to ceramide generation. Primary macrophages treated with lipopolysaccharide (LPS) did not produce C16 ceramide, whereas palmitate exposure led to a modest increase in this sphingolipid. Strikingly, the combination of LPS and palmitate led to a synergistic increase in C16 ceramide. This response occurred via cross-talk at the level of de novo ceramide synthesis in the ER. The synergistic response required TLR4 signaling via MyD88 and TIR-domain-containing adaptor-inducing interferon beta (TRIF), whereas palmitate-induced ceramide production occurred independent of these inflammatory molecules. This ceramide response augmented IL-1ß and TNFα release, a process that may contribute to the enhanced inflammatory response in metabolic diseases characterized by dyslipidemia.


Assuntos
Ceramidas/biossíntese , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Palmitatos/farmacologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Vias Biossintéticas/efeitos dos fármacos , Extratos Celulares , Células Cultivadas , Interleucina-1beta/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Esfingolipídeos/metabolismo , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
5.
Kidney Int Rep ; 8(11): 2235-2242, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38025233

RESUMO

Introduction: Individuals with chronic kidney disease (CKD) are at increased risk of adverse pregnancy outcomes and are susceptible to disempowerment and decisional burden when receiving reproductive counseling and considering pregnancy. Nephrologists do not frequently counsel about reproductive health, and no tools exist to support patient-centered reproductive counseling for those with CKD. Methods: A total of 30 patients aged 18 to 45 years with CKD stages 1 to 5 who were assigned female sex at birth and 12 nephrologists from a single academic medical center participated in semistructured qualitative interviews. They were asked about information needs, decision support needs, and facilitators and barriers to reproductive health care and counseling. Thematic analysis was performed. Results: The following 4 main themes were identified: (i) assessing reproductive intentions; (ii) information about reproductive health and kidney disease; (iii) reproductive risk; and (iv) communication and decision-making needs. Patients' reproductive intentions varied over time and shaped the content of information needed from nephrologists. Patients and nephrologists both felt that risk communication could be improved but focused on different aspects to improve the quality of this counseling; nephrologists focused on providing individualized risk estimates and patients focused on balancing risks with benefits and management. Patients desired nephrologists to bring up the topic of reproductive health and counseling in kidney clinic, and this is not frequently or systematically done currently. Conclusion: This work highlights a critical need for more dialog about reproductive health in kidney care, identified differences in what patients and nephrologists think is important in communication and decision-making, and provides an important step in developing patient-centered reproductive counseling tools in nephrology.

6.
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
7.
Nutrients ; 8(4): 215, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27077881

RESUMO

Obesity and diabetes are associated with excessive inflammation and impaired wound healing. Increasing evidence suggests that macrophage dysfunction is responsible for these inflammatory defects. In the setting of excess nutrients, particularly dietary saturated fatty acids (SFAs), activated macrophages develop lysosome dysfunction, which triggers activation of the NLRP3 inflammasome and cell death. The molecular pathways that connect lipid stress to lysosome pathology are not well understood, but may represent a viable target for therapy. Glutamine uptake is increased in activated macrophages leading us to hypothesize that in the context of excess lipids glutamine metabolism could overwhelm the mitochondria and promote the accumulation of toxic metabolites. To investigate this question we assessed macrophage lipotoxicity in the absence of glutamine using LPS-activated peritoneal macrophages exposed to the SFA palmitate. We found that glutamine deficiency reduced lipid induced lysosome dysfunction, inflammasome activation, and cell death. Under glutamine deficient conditions mTOR activation was decreased and autophagy was enhanced; however, autophagy was dispensable for the rescue phenotype. Rather, glutamine deficiency prevented the suppressive effect of the SFA palmitate on mitochondrial respiration and this phenotype was associated with protection from macrophage cell death. Together, these findings reveal that crosstalk between activation-induced metabolic reprogramming and the nutrient microenvironment can dramatically alter macrophage responses to inflammatory stimuli.


Assuntos
Ácidos Graxos/efeitos adversos , Glutamina/metabolismo , Macrófagos Peritoneais/metabolismo , Animais , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Morte Celular , Células Cultivadas , Gorduras na Dieta , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Inflamassomos/metabolismo , Lipídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
8.
J Leukoc Biol ; 100(5): 1113-1124, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27312848

RESUMO

Macrophage dysfunction in obesity and diabetes is associated with persistent inflammation and poor wound healing responses. Relevant to these phenotypes, we have previously shown that macrophage activation in a high-fat environment results in cell death via a mechanism that involves lysosome damage. While searching for signaling pathways that were required for this response, we discovered that mTOR inhibitors, torin and rapamycin, were protective against lipotoxic cell death in primary peritoneal macrophages. The protective effect of mTOR inhibition was also confirmed by using genetic loss-of-function approaches. Given the importance of mTOR in regulation of autophagy we hypothesized that this pathway would be important in protection from cell death. We first demonstrated that autophagy was disrupted in response to palmitate and LPS as a consequence of impaired lysosome function. Conversely, the mTOR inhibitor, torin, increased macrophage autophagy and protected against lysosome damage; however, the beneficial effects of torin persisted in autophagy-deficient cells. Inhibition of mTOR also triggered nuclear localization of TFEB, a transcription factor that regulates lysosome biogenesis and function, but the rescue phenotype did not require the presence of TFEB. Instead, we demonstrated that mTOR inhibition reduces mitochondrial oxidative metabolism and attenuates the negative effects of palmitate on LPS-induced mitochondrial respiration. These results suggest that inhibition of mTOR is protective against lipotoxicity via an autophagy-independent mechanism that involves relieving mitochondrial substrate overload. On the basis of these findings, we suggest that therapies to reduce macrophage mTOR activation may protect against dysfunctional inflammation in states of overnutrition, such as diabetes.


Assuntos
Proteínas Sanguíneas/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Palmitatos/toxicidade , Sirolimo/farmacologia , Estearatos/toxicidade , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Autofagia/efeitos dos fármacos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/fisiologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Núcleo Celular/metabolismo , Avaliação Pré-Clínica de Medicamentos , Lipopolissacarídeos/toxicidade , Lisossomos/efeitos dos fármacos , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Palmitatos/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , Organismos Livres de Patógenos Específicos , Estearatos/farmacologia , Serina-Treonina Quinases TOR/genética
9.
Int J Inflam ; 2014: 154936, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25587484

RESUMO

Lysosomes play a critical role in the degradation of both extracellular and intracellular material. These dynamic organelles also contribute to nutrient sensing and cell signaling pathways. Macrophages represent a heterogeneous group of phagocytic cells that contribute to tissue homeostasis and inflammation. Recently, there has been a renewed interest in understanding the role of macrophage autophagy and lysosome function in health and disease. Thioglycollate-elicited peritoneal and bone marrow-derived macrophages are commonly used ex vivo systems to study primary macrophage function. In this study, we reveal dramatic baseline differences in the lysosome morphology and function between these macrophage populations and provide evidence that these differences can be functionally relevant. Our results provide important insights into the diversity of lysosomes in primary macrophages and illustrate the importance of accounting for this in data interpretation.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA