Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
Int J Mol Sci ; 24(15)2023 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-37569407

RESUMO

Macrophage polarization is influenced by lipids, which also exert significant control over macrophage functions. Lipids and their metabolites are players in intricate signaling pathways that modulate macrophages' responses to pathogens, phagocytosis, ferroptosis, and inflammation. This review focuses on lipid metabolism and macrophage functions and addresses potential molecular targets for the treatment of macrophage-related diseases. While lipogenesis is crucial for lipid accumulation and phagocytosis in M1 macrophages, M2 macrophages likely rely on fatty acid ß-oxidation to utilize fatty acids as their primary energy source. Cholesterol metabolism, regulated by factors such as SREBPs, PPARs, and LXRs, is associated with the cholesterol efflux capacity and the formation of foam cells (M2-like macrophages). Foam cells, which are targets for atherosclerosis, are associated with an increase in inflammatory cytokines. Lipolysis and fatty acid uptake markers, such as CD36, also contribute to the production of cytokines. Enhancing the immune system through the inhibition of lipid-metabolism-related factors can potentially serve as a targeted approach against tumor cells. Cyclooxygenase inhibitors, which block the conversion of arachidonic acid into various inflammatory mediators, influence macrophage polarization and have generated attention in cancer research.


Assuntos
Polaridade Celular , Inflamação , Metabolismo dos Lipídeos , Macrófagos , Neoplasias , Metabolismo dos Lipídeos/imunologia , Polaridade Celular/imunologia , Inflamação/imunologia , Neoplasias/imunologia , Macrófagos/imunologia , Colesterol/metabolismo , Ácidos Graxos/metabolismo , Ferroptose , Humanos
2.
Int J Mol Sci ; 24(3)2023 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-36768336

RESUMO

Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a disabling multisystem illness in which individuals are plagued with fatigue, inflammatory symptoms, cognitive dysfunction, and the hallmark symptom, post-exertional malaise. While the cause of this disease remains unknown, there is evidence of a potential infectious component that, along with patient symptoms and common onsets of the disease, implicates immune system dysfunction. To further our understanding of the state of ME/CFS lymphocytes, we characterized the role of fatty acids in isolated Natural Killer cells, CD4+ T cells, and CD8+ T cells in circulation and after overnight stimulation, through implicit perturbations to fatty acid oxidation. We examined samples obtained from at least 8 and as many as 20 subjects for immune cell fatty acid characterization in a variety of experiments and found that all three isolated cell types increased their utilization of lipids and levels of pertinent proteins involved in this metabolic pathway in ME/CFS samples, particularly during higher energy demands and activation. In T cells, we characterized the cell populations contributing to these metabolic shifts, which included CD4+ memory cells, CD4+ effector cells, CD8+ naïve cells, and CD8+ memory cells. We also discovered that patients with ME/CFS and healthy control samples had significant correlations between measurements of CD4+ T cell fatty acid metabolism and demographic data. These findings provide support for metabolic dysfunction in ME/CFS immune cells. We further hypothesize about the consequences that these altered fuel dependencies may have on T and NK cell effector function, which may shed light on the illness's mechanism of action.


Assuntos
Síndrome de Fadiga Crônica , Ácidos Graxos , Linfócitos , Humanos , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Síndrome de Fadiga Crônica/imunologia , Células Matadoras Naturais , Ácidos Graxos/imunologia , Oxirredução , Metabolismo dos Lipídeos/imunologia , Linfócitos/imunologia , Subpopulações de Linfócitos/imunologia
3.
J Exp Clin Cancer Res ; 41(1): 16, 2022 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-34998392

RESUMO

BACKGROUND: KRAS is the predominant oncogene mutated in pancreatic ductal adenocarcinoma (PDAC), the fourth cause of cancer-related deaths worldwide. Mutant KRAS-driven tumors are metabolically programmed to support their growth and survival, which can be used to identify metabolic vulnerabilities. In the present study, we aimed to understand the role of extracellularly derived fatty acids in KRAS-driven pancreatic cancer. METHODS: To assess the dependence of PDAC cells on extracellular fatty acids we employed delipidated serum or RNAi-mediated suppression of ACSL3 (to inhibit the activation and cellular retention of extracellular fatty acids) followed by cell proliferation assays, qPCR, apoptosis assays, immunoblots and fluorescence microscopy experiments. To assess autophagy in vivo, we employed the KrasG12D/+;p53flox/flox;Pdx1-CreERT2 (KPC) mice crossed with Acsl3 knockout mice, and to assess the efficacy of the combination therapy of ACSL3 and autophagy inhibition we used xenografted human cancer cell-derived tumors in immunocompromised mice. RESULTS: Here we show that depletion of extracellularly derived lipids either by serum lipid restriction or suppression of ACSL3, triggers autophagy, a process that protects PDAC cells from the reduction of bioenergetic intermediates. Combined extracellular lipid deprivation and autophagy inhibition exhibits anti-proliferative and pro-apoptotic effects against PDAC cell lines in vitro and promotes suppression of xenografted human pancreatic cancer cell-derived tumors in mice. Therefore, we propose lipid deprivation and autophagy blockade as a potential co-targeting strategy for PDAC treatment. CONCLUSIONS: Our work unravels a central role of extracellular lipid supply in ensuring fatty acid provision in cancer cells, unmasking a previously unappreciated metabolic vulnerability of PDAC cells.


Assuntos
Autofagia/imunologia , Metabolismo dos Lipídeos/imunologia , Neoplasias Pancreáticas/terapia , Animais , Proliferação de Células , Modelos Animais de Doenças , Humanos , Camundongos , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas
4.
J Hepatol ; 76(2): 283-293, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34627976

RESUMO

BACKGROUND & AIMS: Recent experimental models and epidemiological studies suggest that specific environmental contaminants (ECs) contribute to the initiation and pathology of non-alcoholic fatty liver disease (NAFLD). However, the underlying mechanisms linking EC exposure with NAFLD remain poorly understood and there is no data on their impact on the human liver metabolome. Herein, we hypothesized that exposure to ECs, particularly perfluorinated alkyl substances (PFAS), impacts liver metabolism, specifically bile acid metabolism. METHODS: In a well-characterized human NAFLD cohort of 105 individuals, we investigated the effects of EC exposure on liver metabolism. We characterized the liver (via biopsy) and circulating metabolomes using 4 mass spectrometry-based analytical platforms, and measured PFAS and other ECs in serum. We subsequently compared these results with an exposure study in a PPARa-humanized mouse model. RESULTS: PFAS exposure appears associated with perturbation of key hepatic metabolic pathways previously found altered in NAFLD, particularly those related to bile acid and lipid metabolism. We identified stronger associations between the liver metabolome, chemical exposure and NAFLD-associated clinical variables (liver fat content, HOMA-IR), in females than males. Specifically, we observed PFAS-associated upregulation of bile acids, triacylglycerols and ceramides, and association between chemical exposure and dysregulated glucose metabolism in females. The murine exposure study further corroborated our findings, vis-à-vis a sex-specific association between PFAS exposure and NAFLD-associated lipid changes. CONCLUSIONS: Females may be more sensitive to the harmful impacts of PFAS. Lipid-related changes subsequent to PFAS exposure may be secondary to the interplay between PFAS and bile acid metabolism. LAY SUMMARY: There is increasing evidence that specific environmental contaminants, such as perfluorinated alkyl substances (PFAS), contribute to the progression of non-alcoholic fatty liver disease (NAFLD). However, it is poorly understood how these chemicals impact human liver metabolism. Here we show that human exposure to PFAS impacts metabolic processes associated with NAFLD, and that the effect is different in females and males.


Assuntos
Exposição Ambiental/efeitos adversos , Metabolismo dos Lipídeos/fisiologia , Hepatopatia Gordurosa não Alcoólica/complicações , Adulto , Aminoácidos/análise , Aminoácidos/sangue , Animais , Estudos de Coortes , Modelos Animais de Doenças , Exposição Ambiental/estatística & dados numéricos , Ácidos Graxos não Esterificados/análise , Ácidos Graxos não Esterificados/sangue , Feminino , Humanos , Metabolismo dos Lipídeos/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/metabolismo
5.
Cancer Res ; 82(2): 187-194, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34764204

RESUMO

Immunosuppressive myeloid cells play a major role in cancer by negatively regulating immune responses, promoting tumor progression, and limiting the efficacy of cancer immunotherapy. Immunosuppression is mediated by various mechanisms dependent upon the type of myeloid cell involved. In recent years, a more universal mechanism of immunosuppressive activity of myeloid cells has emerged: Generation of oxidized lipids. Oxidized lipids accumulate in all types of myeloid cells and are often transferred between cells. In this review, we discuss mechanisms involved in the generation and biological role of myeloid cell-derived oxidized lipids in cancer.


Assuntos
Tolerância Imunológica , Metabolismo dos Lipídeos/imunologia , Células Mieloides/imunologia , Células Mieloides/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Microambiente Tumoral/imunologia , Animais , Humanos , Neoplasias/patologia , Oxirredução
6.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-34884754

RESUMO

Chronic inflammation plays a critical role in the pathogenesis of atherosclerosis. Currently, the mechanism(s) by which inflammation contributes to this disease are not entirely understood. Inflammation is known to induce oxidative stress, which can lead to lipid peroxidation. Lipid peroxidation can result in the production of reactive by-products that can oxidatively modify macromolecules including DNA, proteins, and lipoproteins. A major reactive by-product of lipid peroxidation is malondialdehyde (MDA). MDA can subsequently break down to form acetaldehyde (AA). These two aldehydes can covalently interact with the epsilon (ε)-amino group of lysines within proteins and lipoproteins leading to the formation of extremely stable, highly immunogenic malondialdehyde/acetaldehyde adducts (MAA-adducts). The aim of this study was to investigate the inflammatory response to MAA-modified human serum albumin (HSA-MAA) and low-density lipoprotein (LDL-MAA). We found that animals injected with LDL-MAA generate antibodies specific to MAA-adducts. The level of anti-MAA antibodies were further increased in an animal model of atherosclerosis fed a Western diet. An animal model that combined both high fat diet and immunization of MAA-modified protein resulted in a dramatic increase in antibodies to MAA-adducts and vascular fat accumulation compared with controls. In vitro exposure of endothelial cells and macrophages to MAA-modified proteins resulted in increased fat accumulation as well as increased expression of adhesion molecules and pro-inflammatory cytokines. The expression of cytokines varied between the different cell lines and was unique to the individual modified proteins. The results of these studies demonstrate that different MAA-modified proteins elicit unique responses in different cell types. Additionally, the presence of MAA-modified proteins appears to modulate cellular metabolism leading to increased accumulation of triglycerides and further progression of the inflammatory response.


Assuntos
Inflamação/metabolismo , Lipoproteínas LDL/imunologia , Lipoproteínas LDL/metabolismo , Processamento de Proteína Pós-Traducional , Albumina Sérica Humana/imunologia , Albumina Sérica Humana/metabolismo , Acetaldeído/metabolismo , Animais , Aterosclerose/etiologia , Aterosclerose/imunologia , Aterosclerose/metabolismo , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Feminino , Humanos , Inflamação/etiologia , Inflamação/imunologia , Metabolismo dos Lipídeos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Malondialdeído/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Sprague-Dawley
7.
J Immunol Res ; 2021: 4084566, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34734090

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is closely related to glycolipid metabolism and liver inflammation. And there is no effective drug approved for its clinical therapy. In this study, we focused on mangiferin (Man) and explored its effects and mechanisms on NAFLD treatment based on the regulation of glycolipid metabolism and anti-inflammatory in vivo and in vitro. The results exhibited that Man can significantly attenuate liver injury, insulin resistance, and glucose tolerance in high-fat diet- (HFD-) induced NAFLD mice and significantly reduce fat accumulation and inflammation in hepatic tissue of NAFLD mice. The transcriptome level RNA-seq analysis showed that the significantly different expression genes between the Man treatment group and the HFD-induced NAFLD model group were mainly related to regulation of energy, metabolism, and inflammation in liver tissue. Furthermore, western blots, real-time PCR, and immunohistochemistry experiments confirmed that Man significantly activated the AMPK signal pathway and inhibited NLRP3 inflammasome activation and pyroptosis in NAFLD mice. In in vitro cell experiments, we further confirmed that Man can promote glucose consumption and reduce intracellular triglyceride (TG) accumulation induced by free fatty acids in HepG2 cells and further that it can be blocked by AMPK-specific inhibitors. Western blot results showed that Man upregulated p-AMPKα levels and exhibited a significant AMPK activation effect, which was blocked by compound C. At the same time, Man downregulated the expression of NLRP3 inflammasome-related proteins and inhibited the activation of NLRP3 inflammasome, alleviating cell pyroptosis and inflammation effects. These results indicate that Man anti-NAFLD activity is mediated through its regulation of glucolipid metabolism by AMPK activation and its anti-inflammatory effects by NLRP3 inflammasome inhibition. Our study indicates that Man is a promising prodrug for the therapy of NAFLD patients.


Assuntos
Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Xantonas/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Dieta Hiperlipídica , Modelos Animais de Doenças , Células Hep G2 , Humanos , Inflamassomos/antagonistas & inibidores , Inflamassomos/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/imunologia , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/patologia , Masculino , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Hepatopatia Gordurosa não Alcoólica/imunologia , Hepatopatia Gordurosa não Alcoólica/patologia , Piroptose/efeitos dos fármacos , Piroptose/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Xantonas/uso terapêutico
8.
Cell Rep ; 37(4): 109883, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34706232

RESUMO

Myeloid-derived suppressor cells (MDSCs) impede antitumor immunity; however, the precise mechanisms that regulate their suppressive function remain unresolved. Identifying these mechanisms could lead to therapeutic interventions to boost cancer immunotherapy efficacy. Here, we reveal that ß2 adrenergic receptor (ß2-AR) expression on MDSCs increases with tumor growth and that the ß2-AR stress pathway drives the immune suppressive activity of MDSCs by altering their metabolism. We show that ß2-AR signaling decreases glycolysis and increases oxidative phosphorylation and fatty acid oxidation (FAO). It also increases expression of the fatty acid transporter CPT1A, which is necessary for the FAO-mediated immunosuppressive function of MDSCs. Moreover, we show that ß2-AR signaling increases autophagy and activates the arachidonic acid cycle, both required for increasing the release of the immunosuppressive mediator, PGE2. Our data reveal that ß2-AR signaling triggered by stress is an important physiological regulator of key metabolic pathways in MDSCs, driving their immunosuppressive function.


Assuntos
Células Supressoras Mieloides/metabolismo , Proteínas de Neoplasias/imunologia , Neoplasias/imunologia , Receptores Adrenérgicos beta 2/imunologia , Transdução de Sinais/imunologia , Microambiente Tumoral/imunologia , Animais , Metabolismo dos Lipídeos/genética , Metabolismo dos Lipídeos/imunologia , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Neoplasias/genética , Fosforilação Oxidativa , Receptores Adrenérgicos beta 2/genética , Microambiente Tumoral/genética
9.
Int J Mol Sci ; 22(18)2021 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-34576053

RESUMO

Interleukin (IL)-6 has been studied since its discovery for its role in health and diseases. It is one of the most important pro-inflammatory cytokines. IL-6 was reported as an exacerbating factor in coronavirus disease. In recent years, it has become clear that the function of muscle-derived IL-6 is different from what has been reported so far. Exercise is accompanied by skeletal muscle contraction, during which, several bioactive substances, collectively named myokines, are secreted from the muscles. Many reports have shown that IL-6 is the most abundant myokine. Interestingly, it was indicated that IL-6 plays opposing roles as a myokine and as a pro-inflammatory cytokine. In this review, we discuss why IL-6 has different functions, the signaling mode of hyper-IL-6 via soluble IL-6 receptor (sIL-6R), and the involvement of soluble glycoprotein 130 in the suppressive effect of hyper-IL-6. Furthermore, the involvement of a disintegrin and metalloprotease family molecules in the secretion of sIL-6R is described. One of the functions of muscle-derived IL-6 is lipid metabolism in the liver. However, the differences between the functions of IL-6 as a pro-inflammatory cytokine and the functions of muscle-derived IL-6 are unclear. Although the involvement of myokines in lipid metabolism in adipocytes was previously discussed, little is known about the direct relationship between nonalcoholic fatty liver disease and muscle-derived IL-6. This review is the first to discuss the relationship between the function of IL-6 in diseases and the function of muscle-derived IL-6, focusing on IL-6 signaling and lipid metabolism in the liver.


Assuntos
Interleucina-6/metabolismo , Metabolismo dos Lipídeos/imunologia , Fígado/metabolismo , Músculos/metabolismo , Receptores de Interleucina-6/metabolismo , Adipócitos/imunologia , Adipócitos/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Fígado/imunologia , Camundongos , Músculos/imunologia , Transdução de Sinais/imunologia
10.
Clin Sci (Lond) ; 135(11): 1389-1408, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-34086048

RESUMO

The immune system protects the body against harm by inducing inflammation. During the immune response, cells of the immune system get activated, divided and differentiated in order to eliminate the danger signal. This process relies on the metabolic reprogramming of both catabolic and anabolic pathways not only to produce energy in the form of ATP but also to generate metabolites that exert key functions in controlling the response. Equally important to mounting an appropriate effector response is the process of immune resolution, as uncontrolled inflammation is implicated in the pathogenesis of many human diseases, including allergy, chronic inflammation and cancer. In this review, we aim to introduce the reader to the field of cholesterol immunometabolism and discuss how both metabolites arising from the pathway and cholesterol homeostasis are able to impact innate and adaptive immune cells, staging cholesterol homeostasis at the centre of an adequate immune response. We also review evidence that demonstrates the clear impact that cholesterol metabolism has in both the induction and the resolution of the inflammatory response. Finally, we propose that emerging data in this field not only increase our understanding of immunometabolism but also provide new tools for monitoring and intervening in human diseases, where controlling and/or modifying inflammation is desirable.


Assuntos
Colesterol/metabolismo , Sistema Imunitário/imunologia , Inflamação/metabolismo , Metabolismo dos Lipídeos/imunologia , Animais , Colesterol/imunologia , Metabolismo Energético/imunologia , Metabolismo Energético/fisiologia , Humanos , Inflamação/imunologia , Transdução de Sinais/imunologia , Transdução de Sinais/fisiologia
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1866(10): 158981, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34119681

RESUMO

Lipid droplets (LDs) perform several important functions like inflammatory responses, membrane trafficking, acts as secondary messengers, etc. rather than simply working as an energy reservoir. LDs have been implicated as a controlling factor in the progression of atherosclerosis followed by foam cell formation that derives from macrophages during the differentiation process. However, the role of LDs in monocyte differentiation or its further immunological function is still an area that mandates in-depth investigation. We report that LD dynamics is important for differentiation of monocytes and is absolutely required for sustained and prolonged functional activity of differentiated macrophages. In THP-1 cell line model system, we elucidated that increase in total LD content in monocyte by external lipid supplements, can induce monocyte differentiation independent of classical stimuli, PMA. Differential expression of PLIN2 and ATGL during the event, together with abrogation of de novo lipogenesis further confirmed the fact. Besides, an increase in LD content by free fatty acid supplement was able to exert a synergistic effect with PMA on differentiation and phagocytic activity compared to when they are used alone. Additionally, we have shown Rab5a to play a vital role in LDs biosynthesis/maturation in monocytes and thereby directly affecting differentiation of monocytes into macrophages via AKT pathway. Thus our study reveals the multi-faceted function of LDs during the process of monocyte to macrophage differentiation and thereby helping to maintain the functional activity.


Assuntos
Gotículas Lipídicas/metabolismo , Metabolismo dos Lipídeos/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Endocitose/efeitos dos fármacos , Endocitose/imunologia , Humanos , Microscopia Intravital , Gotículas Lipídicas/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Reguladores do Metabolismo de Lipídeos/farmacologia , Macrófagos/citologia , Macrófagos/metabolismo , Monócitos/citologia , Monócitos/metabolismo , Células THP-1 , Proteínas rab5 de Ligação ao GTP/metabolismo
12.
Int Immunopharmacol ; 97: 107609, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33887577

RESUMO

Metabolic-associated fatty liver disease (MAFLD) is a common type of chronic liver disease characterized by excessive lipid accumulation in hepatocytes, but the pathogenesis is still unclear. Neutrophils, the most abundant immune cells in the human body, defend against pathogens and regulate the inflammatory response. Recent studies have indicated that excessive liver infiltration of neutrophils is a significant histological hallmark of MAFLD, and neutrophils and their derived granule proteins participate in different stages of MAFLD, including hepatic steatosis, inflammation, fibrosis, cirrhosis and hepatocellular carcinoma. Hence, in this review, we summarize the role of neutrophils in the occurrence and progression of MAFLD and provide a perspective for the clinical application of neutrophils in MAFLD diagnosis and treatment.


Assuntos
Carcinoma Hepatocelular/imunologia , Cirrose Hepática/imunologia , Neoplasias Hepáticas/imunologia , Neutrófilos/imunologia , Hepatopatia Gordurosa não Alcoólica/imunologia , Animais , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Modelos Animais de Doenças , Progressão da Doença , Hepatócitos/imunologia , Hepatócitos/metabolismo , Humanos , Metabolismo dos Lipídeos/imunologia , Fígado/citologia , Fígado/imunologia , Fígado/patologia , Cirrose Hepática/diagnóstico , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Hepatopatia Gordurosa não Alcoólica/diagnóstico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia
13.
Cell Immunol ; 364: 104341, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33798909

RESUMO

Asthma is a chronic inflammatory disease of the lungs that poses a considerable health and socioeconomic burden. Several risk factors work synergistically to affect the progression of asthma. Lipid metabolism, especially in distinct cells such as T cells, macrophages, granulocytes, and non-immune cells, plays an essential role in the pathogenesis of asthma, as lipids are potent signaling molecules that regulate a multitude of cellular response. In this review, we focused on the metabolic pathways of lipid molecules, especially fatty acids and their derivatives, and summarized their roles in various cells during the pathogenesis of asthma along with the current pharmacological agents targeting lipid metabolism.


Assuntos
Asma/metabolismo , Granulócitos/imunologia , Metabolismo dos Lipídeos/imunologia , Macrófagos/imunologia , Linfócitos T/imunologia , Animais , Asma/tratamento farmacológico , Asma/epidemiologia , Ácidos Graxos/metabolismo , Humanos , Imunidade Celular , Terapia de Alvo Molecular , Fatores de Risco , Transdução de Sinais
14.
Front Immunol ; 12: 641240, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33833761

RESUMO

Metabolic Associated Fatty liver disease (MAFLD) is a global health problem and represents the most common cause of chronic liver disease in the world. MAFLD spectrum goes from simple steatosis to cirrhosis, in between metabolic steatohepatitis with progressive fibrosis, which pathogenesis is not completely understood. Hence, the role of the immune system has become an important fact in the trigger of inflammatory cascades in metabolic steatohepatitis and in the activation of hepatic stellate cells (HSCs). Among, the more studied immune cells in the pathogenesis of MAFLD are macrophages, T cells, natural killer and dendritic cells. In particular, hepatic dendritic cells had recently attracted a special attention, with a dual role in the pathogenesis of MAFLD. These cells have the capacity to switch from a tolerant state to active state inducing an inflammatory cascade. Furthermore, these cells play a role in the lipid storage within the liver, having, thus providing a crucial nexus between inflammation and lipid metabolism. In this review, we will discuss the current knowledge on the dual role of dendritic cells in lipid accumulation, as wells as in the triggering of hepatic inflammation and hepatocytes cell death in metabolic steatohepatitis.


Assuntos
Células Dendríticas/imunologia , Fígado Gorduroso/imunologia , Células Estreladas do Fígado/imunologia , Metabolismo dos Lipídeos/imunologia , Fígado/imunologia , Macrófagos/imunologia , Animais , Células Dendríticas/patologia , Fígado Gorduroso/patologia , Células Estreladas do Fígado/patologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Fígado/patologia , Macrófagos/patologia
15.
Eur J Immunol ; 51(7): 1628-1640, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33788250

RESUMO

In recent years there have been major advances in our understanding of the role of free fatty acids (FAs) and their metabolism in shaping the functional properties of macrophages and DCs. This review presents the most recent insights into how cell intrinsic FA metabolism controls DC and macrophage function, as well as the current evidence of the importance of various exogenous FAs (such as polyunsaturated FAs and their oxidation products-prostaglandins, leukotrienes, and proresolving lipid mediators) in affecting DC and macrophage biology, by modulating their metabolic properties. Finally, we explore whether targeted modulation of FA metabolism of myeloid cells to steer their function could hold promise in therapeutic settings.


Assuntos
Células Dendríticas/imunologia , Ácidos Graxos/imunologia , Macrófagos/imunologia , Animais , Humanos , Metabolismo dos Lipídeos/imunologia , Células Mieloides/imunologia
16.
Sci Rep ; 11(1): 3047, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33542362

RESUMO

Arachidonic acid (AA), docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA) derived lipids play key roles in initiating and resolving inflammation. Neuro-inflammation is thought to play a causal role in perioperative neurocognitive disorders, yet the role of these lipids in the human central nervous system in such disorders is unclear. Here we used liquid chromatography-mass spectrometry to quantify AA, DHA, and EPA derived lipid levels in non-centrifuged cerebrospinal fluid (CSF), centrifuged CSF pellets, and centrifuged CSF supernatants of older adults obtained before, 24 h and 6 weeks after surgery. GAGE analysis was used to determine AA, DHA and EPA metabolite pathway changes over time. Lipid mediators derived from AA, DHA and EPA were detected in all sample types. Postoperative lipid mediator changes were not significant in non-centrifuged CSF (p > 0.05 for all three pathways). The AA metabolite pathway showed significant changes in centrifuged CSF pellets and supernatants from before to 24 h after surgery (p = 0.0000247, p = 0.0155 respectively), from before to 6 weeks after surgery (p = 0.0000497, p = 0.0155, respectively), and from 24 h to 6 weeks after surgery (p = 0.0000499, p = 0.00363, respectively). These findings indicate that AA, DHA, and EPA derived lipids are detectable in human CSF, and the AA metabolite pathway shows postoperative changes in centrifuged CSF pellets and supernatants.


Assuntos
Fatores Imunológicos/líquido cefalorraquidiano , Metabolismo dos Lipídeos/imunologia , Lipídeos/imunologia , Transtornos Neurocognitivos/genética , Idoso , Idoso de 80 Anos ou mais , Ácido Araquidônico/líquido cefalorraquidiano , Ácido Araquidônico/imunologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Cromatografia Líquida , Ácidos Docosa-Hexaenoicos/líquido cefalorraquidiano , Ácidos Docosa-Hexaenoicos/imunologia , Ácido Eicosapentaenoico/líquido cefalorraquidiano , Ácido Eicosapentaenoico/imunologia , Feminino , Humanos , Fatores Imunológicos/imunologia , Inflamação/líquido cefalorraquidiano , Inflamação/imunologia , Lipídeos/líquido cefalorraquidiano , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Transtornos Neurocognitivos/líquido cefalorraquidiano , Transtornos Neurocognitivos/imunologia , Transtornos Neurocognitivos/patologia , Medicina Perioperatória
17.
J Exp Med ; 218(1)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33601415

RESUMO

Dysregulation in lipid metabolism is among the most prominent metabolic alterations in cancer. Cancer cells harness lipid metabolism to obtain energy, components for biological membranes, and signaling molecules needed for proliferation, survival, invasion, metastasis, and response to the tumor microenvironment impact and cancer therapy. Here, we summarize and discuss current knowledge about the advances made in understanding the regulation of lipid metabolism in cancer cells and introduce different approaches that have been clinically used to disrupt lipid metabolism in cancer therapy.


Assuntos
Proliferação de Células , Imunoterapia , Metabolismo dos Lipídeos/imunologia , Neoplasias , Microambiente Tumoral/imunologia , Animais , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia
18.
Aging (Albany NY) ; 13(3): 3428-3442, 2021 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-33428590

RESUMO

Atherosclerosis is a lipid-driven chronic inflammatory disease in which lipid-laden macrophage foam cells lead to inflamed lesions in arteries. Previous studies have proven that sulfotransferase 2B1b (SULT2B1b) has several roles in the regulation of lipid metabolism and the inflammatory response. However, little is known about the functions of SULT2B1b in ox-LDL-induced inflammation in macrophages. In this study, after treatment with either ox-LDL alone or combined with transfection of siRNAs targeting SULT2B1b, IL-6, TNF-α, NF-κB, IKKß and IκB mRNA and protein expression were determined in Raw264.7 cells by real-time PCR and Western blot, respectively. The proliferative capacity was determined by EdU staining and Cell Counting Kit-8. Our data demonstrated that SULT2B1b knockdown could reduce phosphorylated NF-κB levels and downregulate IKKß protein levels. Additionally, IκB levels were increased and the proliferation of ox-LDL stimulated cells was inhibited after SULT2B1b silencing. Downregulation of SULT2B1b expression was found to upregulate miR-148a-3p expression by microarray assay, while IKKß was a miR-148a-3p target gene. Our study suggests that SULT2B1b knockdown could promote miR148a-3p expression and inhibit activation of the IKKß/NF-κB signalling pathway, which suppressed the inflammatory response in macrophages. Therefore, targeting the SULT2B1b gene might be potentially beneficial for atherosclerosis prevention by decreasing the inflammatory response.


Assuntos
Quinase I-kappa B/genética , Inflamação/genética , Lipoproteínas LDL/imunologia , Macrófagos/metabolismo , MicroRNAs/genética , NF-kappa B/genética , Sulfotransferases/genética , Animais , Aterosclerose/imunologia , Proliferação de Células , Técnicas de Silenciamento de Genes , Quinase I-kappa B/imunologia , Inflamação/imunologia , Metabolismo dos Lipídeos/genética , Metabolismo dos Lipídeos/imunologia , Macrófagos/imunologia , Camundongos , NF-kappa B/imunologia , Células RAW 264.7 , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Sulfotransferases/imunologia
19.
Artigo em Inglês | MEDLINE | ID: mdl-33157277

RESUMO

Inflammatory bowel diseases (IBDs), such as Crohn's disease and ulcerative colitis, are lifelong diseases that remain challenging to treat. IBDs are characterized by alterations in intestinal barrier function and dysregulation of the innate and adaptive immunity. An increasing number of lipids are found to be important regulators of inflammation and immunity as well as gut physiology. Therefore, the study of lipid mediators in IBDs is expected to improve our understanding of disease pathogenesis and lead to novel therapeutic opportunities. Here, through selected examples - such as fatty acids, specialized proresolving mediators, lysophospholipids, endocannabinoids, and oxysterols - we discuss how lipid signaling is involved in IBD physiopathology and how modulating lipid signaling pathways could affect IBDs.


Assuntos
Colite Ulcerativa/imunologia , Doença de Crohn/imunologia , Fármacos Gastrointestinais/farmacologia , Mucosa Intestinal/patologia , Metabolismo dos Lipídeos/imunologia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/metabolismo , Colite Ulcerativa/patologia , Doença de Crohn/tratamento farmacológico , Doença de Crohn/metabolismo , Doença de Crohn/patologia , Modelos Animais de Doenças , Quimioterapia Combinada/métodos , Endocanabinoides/imunologia , Endocanabinoides/metabolismo , Ácidos Graxos/imunologia , Ácidos Graxos/metabolismo , Fármacos Gastrointestinais/uso terapêutico , Humanos , Imunidade Inata/efeitos dos fármacos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Mucosa Intestinal/imunologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Lisofosfolipídeos/imunologia , Lisofosfolipídeos/metabolismo , Oxisteróis/imunologia , Oxisteróis/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
20.
Front Immunol ; 11: 584303, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33224146

RESUMO

The interplay between cellular stress and immune response can be variable and sometimes contradictory. The mechanisms by which stress-activated pathways regulate the inflammatory response to a pathogen, in autoimmunity or during cancer progression remain unclear in many aspects, despite our recent knowledge of the signalling and transcriptional pathways involved in these diseases. In this context, over the last decade many studies demonstrated that cholesterol metabolism is an important checkpoint for immune homeostasis and cancer progression. Indeed, cholesterol is actively metabolized and can regulate, through its mobilization and/or production of active derivatives, many aspects of immunity and inflammation. Moreover, accumulation of cholesterol has been described in cancer cells, indicating metabolic addiction. The nuclear receptors liver-X-receptors (LXRs) are important regulators of intracellular cholesterol and lipids homeostasis. They have also key regulatory roles in immune response, as they can regulate inflammation, innate and adaptive immunity. Moreover, activation of LXRs has been reported to affect the proliferation and survival of different cancer cell types that show altered metabolic pathways and accumulation of cholesterol. In this minireview we will give an overview of the recent understandings about the mechanisms through which LXRs regulate inflammation, autoimmunity, and cancer, and the therapeutic potential for future treatment of these diseases through modulation of cholesterol metabolism.


Assuntos
Autoimunidade/imunologia , Colesterol/metabolismo , Inflamação/metabolismo , Receptores X do Fígado/metabolismo , Neoplasias/metabolismo , Imunidade Adaptativa/imunologia , Animais , Colesterol/imunologia , Humanos , Imunidade Inata/imunologia , Inflamação/imunologia , Metabolismo dos Lipídeos/imunologia , Redes e Vias Metabólicas/imunologia , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA