Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Lipid Res ; 63(2): 100167, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35007562

RESUMO

Niemann-Pick type C1 (NPC1) disease is a progressive lysosomal storage disorder caused by mutations of the NPC1 gene. While neurodegeneration is the most severe symptom, a large proportion of NPC1 patients also present with splenomegaly, which has been attributed to cholesterol and glycosphingolipid accumulation in late endosomes and lysosomes. However, recent data also reveal an increase in the inflammatory monocyte subset in the Npc1nih mouse model expressing an Npc1 null allele. We evaluated the contribution of hematopoietic cells to splenomegaly in NPC1 disease under conditions of hypercholesterolemia. We transplanted Npc1nih (Npc1 null mutation) or Npc1wt bone marrow (BM) into Ldlr-/- mice and fed these mice a cholesterol-rich Western-type diet. At 9 weeks after BM transplant, on a chow diet, the Npc1 null mutation increased plasma granulocyte-colony stimulating factor (G-CSF) by 2-fold and caused mild neutrophilia. At 18 weeks after BM transplant, including 9 weeks of Western-type diet feeding, the Npc1 mutation increased G-csf mRNA levels by ∼5-fold in splenic monocytes/macrophages accompanied by a ∼4-fold increase in splenic neutrophils compared with controls. We also observed ∼5-fold increased long-term and short-term hematopoietic stem cells (HSCs) in the spleen, and a ∼30-75% decrease of these populations in BM, reflecting HSC mobilization, presumably downstream of elevated G-CSF. In line with these data, four patients with NPC1 disease showed higher plasma G-CSF compared with age-matched and gender-matched healthy controls. In conclusion, we show elevated G-CSF levels and HSC mobilization in the setting of an Npc1 null mutation and propose that this contributes to splenomegaly in patients with NPC1 disease.


Assuntos
Mobilização de Células-Tronco Hematopoéticas
2.
Cell Immunol ; 370: 104439, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34607646

RESUMO

Macrophage migration inhibitory factor (MIF-1) and its homologue d-dopachrome tautomerase (MIF-2) share the common CD74 receptor and function innately to enhance severity of multiple sclerosis (MS) as well as the experimental autoimmune encephalomyelitis (EAE) model for MS. We previously demonstrated that genetically high-MIF-expressing male subjects with relapsing MS had a significantly greater risk of conversion to progressive MS (PMS) than lower-MIF-expressing males. To expand on this observation, we utilized MIF-1, MIF-2, and MIF-1/2-DUAL-deficient male mice to discern if there would be a greater contribution of these inflammatory factors in EAE mice with severe vs. moderate clinical disease signs. As shown previously, mice deficient in either MIF-1 or MIF-2 each had a ∼25% reduction of moderate EAE compared to WT mice, with significant differences in disease onset and trajectory. However, EAE induction in mice deficient in both MIF-1 and MIF-2 genes did not result in a further reduction in EAE severity. This result suggests that the two MIF homologues were likely affecting the same pathogenic pathways such that each could partially compensate for the other but not in an additive or synergistic manner. However, MIF-1-KO, MIF-2-KO, and MIF-1/2-DUAL-KO mice with severe EAE did not exhibit a significant reduction in cumulative EAE scores compared with WT mice, but the MIF-1-KO and, to a lesser extent, MIF-1/2-DUAL-KO mice did show a significant reduction in daily EAE scores over the last 3 days of observation, and MIF-2-KO mice showed a more modest but still consistent reduction over the same span. Furthermore, deletion of MIF-1 resulted in a massive reduction in the expression of EAE- and Complete Freund's Adjuvant-associated inflammatory factors, suggesting delayed involvement of the MIF/CD74 axis in promoting disease expression. To further explore modulation of MIF-1 and MIF-2 effects on EAE, we treated WT mice with moderate EAE using DRα1-mMOG-35-55, an inhibitor of CD74 that blocks both MIF-1 and MIF-2 action. This treatment reduced ongoing moderate EAE severity in excess of 25%, suggesting efficient blockade of the MIF/CD74 axis in disease-enhancing pathways. Moreover, DRα1-mMOG-35-55 treatment of mice with severe EAE strongly reversed EAE- and CFA-associated expression of inflammatory cytokines and chemokines including Tnf, Ccr7, Ccr6, Ccl8, Cxcr3, and Ccl19 in MIF-deficient mouse genotypes, and also exceeded innate MIF-1 and MIF-2 EAE enhancing effects, especially in MIF-1-KO mice. These results illustrate the therapeutic potential of targeting the disease-enhancing MIF/CD74 pathway in male mice with moderate and severe EAE, with implications for treatment of high-MIF-expressing RRMS human males at risk of conversion to progressive MS as well as those that have already transitioned to PMS.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Oxirredutases Intramoleculares/imunologia , Fatores Inibidores da Migração de Macrófagos/imunologia , Fármacos Neuroprotetores/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
3.
Cell Immunol ; 359: 104242, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33190849

RESUMO

Experimental autoimmune encephalomyelitis (EAE) is commonly used as an animal model for evaluating clinical, histological and immunological processes potentially relevant to the human disease multiple sclerosis (MS), for which the mode of disease induction remains largely unknown. An important caveat for interpreting EAE processes in mice is the inflammatory effect of immunization with myelin peptides emulsified in Complete Freund's Adjuvant (CFA), often followed by additional injections of pertussis toxin (Ptx) in some strains to induce EAE. The current study evaluated clinical, histological, cellular (spleen), and chemokine-driven processes in spinal cords of male vs. female C57BL/6 mice that were immunized with mouse (m)MOG-35-55/CFA/Ptx to induce EAE; immunized with saline/CFA/Ptx only (CFA, no EAE); or were untreated (Naïve, no EAE). Analysis of response curves utilized a rigorous and sophisticated methodology to parse and characterize the effects of EAE and adjuvant alone vs. the Naive baseline responses. The results demonstrated stronger pro-inflammatory responses of immune cells and their associated cytokines, chemokines, and receptors in male vs. female CFA and EAE mice that appeared to be offset partially by increased percentages of male anti-inflammatory, regulatory and checkpoint T cell, B cell, and monocyte/macrophage subsets. These sex differences in peripheral immune responses may explain the reduced cellular infiltration and differing chemokine profiles in the Central Nervous System (CNS) of male vs. female CFA immunized mice and the reduced CNS infiltration and demyelination observed in male vs. female EAE groups of mice that ultimately resulted in the same clinical EAE disease severity in both sexes. Our findings suggest EAE disease severity is governed not only by the degree of CNS infiltration and demyelination, but also by the balance of pro-inflammatory vs. regulatory cell types and their secreted cytokines and chemokines.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Adjuvante de Freund/farmacologia , Adjuvantes Imunológicos/farmacologia , Transferência Adotiva , Animais , Linfócitos B/imunologia , Sistema Nervoso Central/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/fisiopatologia , Feminino , Imunização/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/metabolismo , Fragmentos de Peptídeos/imunologia , Toxina Pertussis/farmacologia , Caracteres Sexuais , Fatores Sexuais , Medula Espinal/imunologia , Linfócitos T/imunologia
4.
J Lipid Res ; 61(3): 365-375, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31843957

RESUMO

Atherosclerosis-related CVD causes nearly 20 million deaths annually. Most patients are treated after plaques develop, so therapies must regress existing lesions. Current therapies reduce plaque volume, but targeting all apoB-containing lipoproteins with intensive combinations that include alirocumab or evinacumab, monoclonal antibodies against cholesterol-regulating proprotein convertase subtilisin/kexin type 9 and angiopoietin-like protein 3, may provide more benefit. We investigated the effect of such lipid-lowering interventions on atherosclerosis in APOE*3-Leiden.CETP mice, a well-established model for hyperlipidemia. Mice were fed a Western-type diet for 13 weeks and thereafter matched into a baseline group (euthanized at 13 weeks) and five groups that received diet alone (control) or with treatment [atorvastatin; atorvastatin and alirocumab; atorvastatin and evinacumab; or atorvastatin, alirocumab, and evinacumab (triple therapy)] for 25 weeks. We measured effects on cholesterol levels, plaque composition and morphology, monocyte adherence, and macrophage proliferation. All interventions reduced plasma total cholesterol (37% with atorvastatin to 80% with triple treatment; all P < 0.001). Triple treatment decreased non-HDL-C to 1.0 mmol/l (91% difference from control; P < 0.001). Atorvastatin reduced atherosclerosis progression by 28% versus control (P < 0.001); double treatment completely blocked progression and diminished lesion severity. Triple treatment regressed lesion size versus baseline in the thoracic aorta by 50% and the aortic root by 36% (both P < 0.05 vs. baseline), decreased macrophage accumulation through reduced proliferation, and abated lesion severity. Thus, high-intensive cholesterol-lowering triple treatment targeting all apoB-containing lipoproteins regresses atherosclerotic lesion area and improves lesion composition in mice, making it a promising potential approach for treating atherosclerosis.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Anticolesterolemiantes/uso terapêutico , Atorvastatina/uso terapêutico , Placa Aterosclerótica/tratamento farmacológico , Administração Oral , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticolesterolemiantes/administração & dosagem , Atorvastatina/administração & dosagem , Quimioterapia Combinada , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placa Aterosclerótica/induzido quimicamente , Placa Aterosclerótica/patologia
5.
J Lipid Res ; 61(12): 1617-1628, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32848049

RESUMO

The rise of drug-resistant tuberculosis poses a major risk to public health. Statins, which inhibit both cholesterol biosynthesis and protein prenylation branches of the mevalonate pathway, increase anti-tubercular antibiotic efficacy in animal models. However, the underlying molecular mechanisms are unknown. In this study, we used an in vitro macrophage infection model to investigate simvastatin's anti-tubercular activity by systematically inhibiting each branch of the mevalonate pathway and evaluating the effects of the branch-specific inhibitors on mycobacterial growth. The anti-tubercular activity of simvastatin used at clinically relevant doses specifically targeted the cholesterol biosynthetic branch rather than the prenylation branches of the mevalonate pathway. Using Western blot analysis and AMP/ATP measurements, we found that simvastatin treatment blocked activation of mechanistic target of rapamycin complex 1 (mTORC1), activated AMP-activated protein kinase (AMPK) through increased intracellular AMP:ATP ratios, and favored nuclear translocation of transcription factor EB (TFEB). These mechanisms all induce autophagy, which is anti-mycobacterial. The biological effects of simvastatin on the AMPK-mTORC1-TFEB-autophagy axis were reversed by adding exogenous cholesterol to the cells. Our data demonstrate that the anti-tubercular activity of simvastatin requires inhibiting cholesterol biosynthesis, reveal novel links between cholesterol homeostasis, the AMPK-mTORC1-TFEB axis, and Mycobacterium tuberculosis infection control, and uncover new anti-tubercular therapy targets.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antituberculosos/farmacologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Colesterol/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Sinvastatina/farmacologia , Animais , Autofagia/efeitos dos fármacos , Humanos , Lisossomos/metabolismo , Macrófagos/metabolismo , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
6.
J Nutr ; 150(5): 1303-1312, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32040591

RESUMO

BACKGROUND: Metabolic endotoxemia is considered a cause for high-fat diet (HFD)-induced inflammation. However, convincing experimental evidence in humans is scant. OBJECTIVE: We determined whether a HFD or moderately HFD increases LPS and LPS-mediated cytokine production in the postprandial blood (PPB). METHODS: Ninety-eight volunteers (age: 37.3 ± 1.5 y) from the cross-sectional phenotyping study (PS) and 62 volunteers (age: 26.8 ± 1.2 y) from the intervention study (IS) consumed a breakfast containing 60% kcal fat (HF) and 36% kcal fat (moderately HF), respectively. For the IS, only the results from the placebo group are presented. Blood samples were probed for LPS-mediated cytokine production by incubating them with LPS inhibitor polymyxin B (PMB) for 24 h at 37°C besides the Limulus amebocyte lysate (LAL) assay. Repeated-measures ANOVA was used to compare the temporal changes of metabolic profiles and treatment outcomes. RESULTS: At least 87.5% of the plasma LPS measurements in 32 PS volunteers from each time point were below the LAL assay sensitivity (0.002 EU/mL). PMB suppressed IL-1ß (P = 0.035) and IL-6 (P = 0.0487) production in the 3 h PPB of the PS after 24 h incubation at 37°C compared to the vehicle control, suggesting the presence of LPS. However, the amount of LPS did not increase the cytokine concentrations in the 3 h PPB above the fasting concentrations. Such suppression was not detected in the PPB of the IS. Treating whole blood with lipoprotein lipase (LPL) significantly (P < 0.05) increased FFA and cytokine (IL-1ß, IL-6, TNF-α) concentrations in both studies. CONCLUSION: LPS may not be the major cause of postprandial inflammation in healthy adults consuming a moderately HF meal (36% kcal fat, similar to the typical American diet) or a HF meal (60% kcal fat). Plasma FFAs may modulate postprandial inflammation. The prevailing concept of HFD-induced metabolic endotoxemia requires careful re-evaluation. The PS was registered at clinicaltrials.gov as NCT02367287 and the IS as NCT02472171.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Inflamação/sangue , Inflamação/etiologia , Lipopolissacarídeos/sangue , Período Pós-Prandial/fisiologia , Adulto , Desjejum , Estudos Transversais , Citocinas/sangue , Método Duplo-Cego , Ácidos Graxos não Esterificados/sangue , Feminino , Humanos , Lipopolissacarídeos/antagonistas & inibidores , Lipase Lipoproteica/metabolismo , Masculino , Placebos , Polimixina B/farmacologia
7.
J Lipid Res ; 59(12): 2255-2261, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29678958

RESUMO

Eukaryotic cells employ distinct means to release specific signals and material. Research within the last decade has identified different types of membrane-enclosed structures collectively called extracellular vesicles (EVs) as one of them. EVs fall into two categories depending on their subcellular origin. Exosomes are generated within the endosomal system and reach the extracellular space upon fusion of multivesicular bodies. Microvesicles or microparticles are generated by shedding of the plasma membrane. Sterols are essential components of eukaryotic membranes and serve as precursors or cofactors of numerous signaling molecules; their content and subcellular distribution are tightly controlled. The prominent roles of sterols in cells raise the question of whether and how these components impact EVs. In this review, we compile evidence for cholesterol accumulation in EVs and discuss its possible contribution to their biogenesis, release, and uptake. We also consider potential implications of EVs in cellular sterol homeostasis and in cholesterol-related diseases.


Assuntos
Colesterol/metabolismo , Vesículas Extracelulares/metabolismo , Animais , Micropartículas Derivadas de Células/metabolismo , Humanos , Transporte Proteico/fisiologia , Transdução de Sinais/fisiologia
8.
J Cell Physiol ; 233(3): 2602-2612, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28782808

RESUMO

Profound alterations in the lipid profile of raft and non-raft plasma membrane microdomains were found when RAW264.7 macrophages were supplemented with polyunsaturated fatty acids (PUFAs) in physiologically relevant concentrations. For the first time lipids in the detergent-free isolated membrane domains of phagocytic immune cells were characterized by mass spectrometry. The extent of remodeling of the membrane lipids differed with different n3 and n6 PUFA supplements. The mildest effects were detected for α-linolenic acid (LNA) and linoleic acid (LA), the C18 precursors of the n3 and n6 families, respectively. When the effects of highly unsaturated PUFAs were compared, eicosapentaenoic acid (EPA) caused more extensive restructuring of membrane lipids than docosahexaenoic acid (DHA) or arachidonic acid (AA). The supplements altered the lipid species composition of both the raft and non-raft membrane fractions. The rafts containing elevated proportions of highly unsaturated lipid species may relocate sterically incompatible lipids and proteins originally belonging to this microdomain. Such effect was evident for sphingomyelin, which favored non-rafts instead of rafts after EPA supplementation. The current work suggests that the different functional consequences found previously when supplementing macrophages with either EPA or DHA have their origin in the different effects of these PUFAs on membrane architecture.


Assuntos
Ácidos Graxos Insaturados/farmacologia , Macrófagos/efeitos dos fármacos , Lipídeos de Membrana/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Animais , Cromatografia Líquida de Alta Pressão , Macrófagos/metabolismo , Microdomínios da Membrana/metabolismo , Camundongos , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Células RAW 264.7 , Espectrometria de Massas em Tandem
9.
J Vasc Res ; 55(2): 63-74, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29393228

RESUMO

Abdominal aortic aneurysm (AAA) is a vascular disease characterized by chronic inflammation in the infrarenal aorta. Most cases of AAA remain asymptomatic until rupture, and the mortality rate of patients with AAA rupture is very high. Currently, the relation between dietary habits and AAA development remains unknown. In this study, we evaluated the effects of a high-fat diet on the development of AAA in a vascular hypoperfusion-induced animal model. The risk of AAA rupture and AAA diameter in the high-fat group significantly increased compared with those in the control group. The number and size of adipocytes in the vascular wall in the high-fat group significantly increased as compared with those in the control group. Additionally, the collagen-positive sections in the areas with adipocytes significantly decreased as compared with those without adipocytes. The protein levels of matrix metalloproteinase (MMP)-2, MMP-9, and MMP-12, and macrophage-positive areas in the parts with adipocytes also significantly increased as compared with those without adipocytes. These data suggested that AAA rupture risk increased through accelerating chronic inflammation due to the accumulation of adipocytes in the vascular wall in the high-fat group.


Assuntos
Aorta Abdominal , Aneurisma da Aorta Abdominal/etiologia , Ruptura Aórtica/etiologia , Dieta Hiperlipídica/efeitos adversos , Adipócitos/metabolismo , Adipócitos/patologia , Animais , Antígenos de Diferenciação , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Aorta Abdominal/fisiopatologia , Aorta Abdominal/cirurgia , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/fisiopatologia , Ruptura Aórtica/metabolismo , Ruptura Aórtica/patologia , Ruptura Aórtica/fisiopatologia , Quimiocina CCL2/metabolismo , Colágeno/metabolismo , Modelos Animais de Doenças , Ligadura , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Metaloproteinase 12 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional , Fatores de Tempo
10.
J Lipid Res ; 58(1): 236-246, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27811230

RESUMO

G protein-coupled receptor (GPR)120/FFA receptor (FFAR)4 (GPR120/FFAR4) activation by n-3 PUFAs attenuates inflammation, but its impact on atherosclerosis is unknown. We determined whether in vivo activation of leukocyte GPR120/FFAR4 by n-3 versus n-6 PUFAs is atheroprotective. Leukocyte GPR120/FFAR4 WT or KO mice in the LDL receptor KO background were generated by bone marrow transplantation. Mice were fed one of the four atherogenic diets containing 0.2% cholesterol and 10% calories as palm oil (PO) + 10% calories as: 1) PO, 2) fish oil (FO; 20:5 n-3 and 22:6 n-3 enriched), 3) echium oil (EO; 18:4 n-3 enriched), or 4) borage oil (BO; 18:3 n-6 enriched) for 16 weeks. Compared with PO, mice fed BO, EO, and FO had significantly reduced plasma cholesterol, TG, VLDL cholesterol, hepatic neutral lipid, and atherosclerosis that were equivalent for WT and KO mice. In BO-, EO-, and FO-fed mice, but not PO-fed mice, lack of leukocyte GPR120/FFAR4 resulted in neutrophilia, pro-inflammatory Ly6Chi monocytosis, increased aortic root monocyte recruitment, and increased hepatic inflammatory gene expression. In conclusion, leukocyte GPR120 expression has minimal effects on dietary PUFA-induced plasma lipid/lipoprotein reduction and atheroprotection, and there is no distinction between n-3 versus n-6 PUFAs in activating anti-inflammatory effects of leukocyte GPR120/FFAR4 in vivo.


Assuntos
Aterosclerose/genética , Leucócitos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de LDL/genética , Animais , Aorta/metabolismo , Aorta/patologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Colesterol/metabolismo , Dieta Aterogênica , Ácidos Graxos Ômega-3/genética , Ácidos Graxos Ômega-3/metabolismo , Ácidos Graxos Ômega-6/genética , Ácidos Graxos Ômega-6/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Leucócitos/patologia , Camundongos , Camundongos Knockout , Receptores Acoplados a Proteínas G/metabolismo , Ativação Transcricional/genética
11.
J Lipid Res ; 58(7): 1325-1337, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28442497

RESUMO

We developed an in silico mathematical model of retinal cholesterol (Ch) dynamics (RCD) to quantify the physiological rate of Ch turnover in the rod outer segment (ROS), the lipoprotein transport mechanisms by which Ch enters and leaves the outer retina, and the rates of drusen growth and macrophage-mediated clearance in dry age-related macular degeneration. Based on existing experimental data and mechanistic hypotheses, we estimated the Ch turnover rate in the ROS to be 1-6 pg/mm2/min, dependent on the rate of Ch recycling in the outer retina, and found comparable rates for LDL receptor-mediated endocytosis of Ch by the retinal pigment epithelium (RPE), ABCA1-mediated Ch transport from the RPE to the outer retina, ABCA1-mediated Ch efflux from the RPE to the choroid, and the secretion of 70 nm ApoB-Ch particles from the RPE. The drusen growth rate is predicted to increase from 0.7 to 4.2 µm/year in proportion to the flux of ApoB-Ch particles. The rapid regression of drusen may be explained by macrophage-mediated clearance if the macrophage density reaches ∼3,500 cells/mm2 The RCD model quantifies retinal Ch dynamics and suggests that retinal Ch turnover and recycling, ApoB-Ch particle efflux, and macrophage-mediated clearance may explain the dynamics of drusen growth and regression.


Assuntos
Colesterol/metabolismo , Simulação por Computador , Degeneração Macular/metabolismo , Retina/metabolismo , Transporte Biológico , Humanos , Degeneração Macular/fisiopatologia , Epitélio Pigmentado da Retina/metabolismo , Segmento Externo da Célula Bastonete/metabolismo
12.
J Lipid Res ; 56(6): 1191-205, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25921305

RESUMO

Echium oil (EO), which is enriched in 18:4 n-3, the immediate product of fatty acid desaturase 2 (FADS2) desaturation of 18:3 n-3, is as atheroprotective as fish oil (FO). The objective of this study was to determine whether botanical oils enriched in the FADS2 products 18:3 n-6 versus 18:4 n-3 are equally atheroprotective. LDL receptor KO mice were fed one of four atherogenic diets containing 0.2% cholesterol and 10% calories as palm oil (PO) plus 10% calories as: 1) PO; 2) borage oil (BO; 18:3 n-6 enriched); 3) EO (18:4 n-3 enriched); or 4) FO for 16 weeks. Mice fed BO, EO, and FO versus PO had significantly lower plasma total and VLDL cholesterol concentrations; hepatic neutral lipid content and inflammation, aortic CE content, aortic root intimal area and macrophage content; and peritoneal macrophage inflammation, CE content, and ex vivo chemotaxis. Atheromas lacked oxidized CEs despite abundant generation of macrophage 12/15 lipooxygenase-derived metabolites. We conclude that botanical oils enriched in 18:3 n-6 and 18:4 n-3 PUFAs beyond the rate-limiting FADS2 enzyme are equally effective in preventing atherosclerosis and hepatosteatosis compared with saturated/monounsaturated fat due to cellular enrichment of ≥20 PUFAs, reduced plasma VLDL, and attenuated macrophage inflammation.


Assuntos
Aterosclerose/dietoterapia , Ácidos Graxos Dessaturases/metabolismo , Fígado/metabolismo , Óleos de Plantas/administração & dosagem , Receptores de LDL/genética , Animais , Aterosclerose/metabolismo , VLDL-Colesterol/sangue , Dieta Aterogênica , Echium/química , Ácidos Graxos Ômega-3/administração & dosagem , Ácidos Graxos Ômega-3/química , Ácidos Graxos Ômega-6/administração & dosagem , Ácidos Graxos Ômega-6/química , Fígado Gorduroso/dietoterapia , Óleos de Peixe/administração & dosagem , Óleos de Peixe/química , Humanos , Fígado/efeitos dos fármacos , Camundongos , Camundongos Knockout , Óleo de Palmeira , Óleos de Plantas/química , Receptores de LDL/metabolismo , Ácido gama-Linolênico/administração & dosagem , Ácido gama-Linolênico/química
13.
J Lipid Res ; 56(2): 203-14, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25424004

RESUMO

Atherosclerotic lesions are often hypoxic and exhibit elevated lactate concentrations and local acidification of the extracellular fluids. The acidification may be a consequence of the abundant accumulation of lipid-scavenging macrophages in the lesions. Activated macrophages have a very high energy demand and they preferentially use glycolysis for ATP synthesis even under normoxic conditions, resulting in enhanced local generation and secretion of lactate and protons. In this review, we summarize our current understanding of the effects of acidic extracellular pH on three key players in atherogenesis: macrophages, apoB-containing lipoproteins, and HDL particles. Acidic extracellular pH enhances receptor-mediated phagocytosis and antigen presentation by macrophages and, importantly, triggers the secretion of proinflammatory cytokines from macrophages through activation of the inflammasome pathway. Acidity enhances the proteolytic, lipolytic, and oxidative modifications of LDL and other apoB-containing lipoproteins, and strongly increases their affinity for proteoglycans, and may thus have major effects on their retention and the ensuing cellular responses in the arterial intima. Finally, the decrease in the expression of ABCA1 at acidic pH may compromise cholesterol clearance from atherosclerotic lesions. Taken together, acidic extracellular pH amplifies the proatherogenic and proinflammatory processes involved in atherogenesis.


Assuntos
Aterosclerose/metabolismo , Túnica Íntima/metabolismo , Animais , Apolipoproteínas/metabolismo , Aterosclerose/etiologia , Humanos , Lipoproteínas/metabolismo , Fosfolipases/metabolismo , Proteoglicanas/metabolismo
14.
J Lipid Res ; 56(9): 1795-807, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26156077

RESUMO

Palmitic acid (PA) is associated with higher blood concentrations of medium-chain acylcarnitines (MCACs), and we hypothesized that PA may inhibit progression of FA ß-oxidation. Using a cross-over design, 17 adults were fed high PA (HPA) and low PA/high oleic acid (HOA) diets, each for 3 weeks. The [1-(13)C]PA and [13-(13)C]PA tracers were administered with food in random order with each diet, and we assessed PA oxidation (PA OX) and serum AC concentration to determine whether a higher PA intake promoted incomplete PA OX. Dietary PA was completely oxidized during the HOA diet, but only about 40% was oxidized during the HPA diet. The [13-(13)C]PA/[1-(13)C]PA ratio of PA OX had an approximate value of 1.0 for either diet, but the ratio of the serum concentrations of MCACs to long-chain ACs (LCACs) was significantly higher during the HPA diet. Thus, direct measurement of PA OX did not confirm that the HPA diet caused incomplete PA OX, despite the modest, but statistically significant, increase in the ratio of MCACs to LCACs in blood.


Assuntos
Carnitina/análogos & derivados , Dieta , Ácidos Graxos/sangue , Palmitatos/administração & dosagem , Adolescente , Adulto , Composição Corporal/efeitos dos fármacos , Carnitina/sangue , Citocinas/metabolismo , Gorduras na Dieta/administração & dosagem , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Peroxidação de Lipídeos/genética , Masculino , Ácido Oleico/administração & dosagem , Palmitatos/sangue
15.
J Lipid Res ; 55(11): 2296-308, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25240046

RESUMO

Macrophages play crucial roles in the formation of atherosclerotic lesions. Akt, a serine/threonine protein kinase B, is vital for cell proliferation, migration, and survival. Macrophages express three Akt isoforms, Akt1, Akt2, and Akt3, but the roles of Akt1 and Akt2 in atherosclerosis in vivo remain unclear. To dissect the impact of macrophage Akt1 and Akt2 on early atherosclerosis, we generated mice with hematopoietic deficiency of Akt1 or Akt2. After 8 weeks on Western diet, Ldlr(-/-) mice reconstituted with Akt1(-/-) fetal liver cells (Akt1(-/-)→Ldlr(-/-)) had similar atherosclerotic lesion areas compared with control mice transplanted with WT cells (WT→Ldlr(-/-)). In contrast, Akt2(-/-)→Ldlr(-/-) mice had dramatically reduced atherosclerotic lesions compared with WT→Ldlr(-/-) mice of both genders. Similarly, in the setting of advanced atherosclerotic lesions, Akt2(-/-)→Ldlr(-/-) mice had smaller aortic lesions compared with WT→Ldlr(-/-) and Akt1(-/-)→Ldlr(-/-) mice. Importantly, Akt2(-/-)→Ldlr(-/-) mice had reduced numbers of proinflammatory blood monocytes expressing Ly-6C(hi) and chemokine C-C motif receptor 2. Peritoneal macrophages isolated from Akt2(-/-) mice were skewed toward an M2 phenotype and showed decreased expression of proinflammatory genes and reduced cell migration. Our data demonstrate that loss of Akt2 suppresses the ability of macrophages to undergo M1 polarization reducing both early and advanced atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Macrófagos/metabolismo , Proteínas Proto-Oncogênicas c-akt/deficiência , Proteínas Proto-Oncogênicas c-akt/genética , Receptores de LDL/deficiência , Animais , Antígenos Ly/genética , Aterosclerose/enzimologia , Aterosclerose/genética , Aterosclerose/imunologia , Movimento Celular , Feminino , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Hematopoese , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Fenótipo , Receptores CCR2/genética , Receptores CCR2/metabolismo , Receptores de LDL/genética
16.
J Lipid Res ; 55(1): 138-45, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24212238

RESUMO

Lysosomal integral membrane protein-2 (LIMP2) mediates trafficking of glucocerebrosidase (GBA) to lysosomes. Deficiency of LIMP2 causes action myoclonus-renal failure syndrome (AMRF). LIMP2-deficient fibroblasts virtually lack GBA like the cells of patients with Gaucher disease (GD), a lysosomal storage disorder caused by mutations in the GBA gene. While GD is characterized by the presence of glucosylceramide-laden macrophages, AMRF patients do not show these. We studied the fate of GBA in relation to LIMP2 deficiency by employing recently designed activity-based probes labeling active GBA molecules. We demonstrate that GBA is almost absent in lysosomes of AMRF fibroblasts. However, white blood cells contain considerable amounts of residual enzyme. Consequently, AMRF patients do not acquire lipid-laden macrophages and do not show increased plasma levels of macrophage markers, such as chitotriosidase, in contrast to GD patients. We next investigated the consequences of LIMP2 deficiency with respect to plasma glycosphingolipid levels. Plasma glucosylceramide concentration was normal in the AMRF patients investigated as well as in LIMP2-deficient mice. However, a marked increase in the sphingoid base, glucosylsphingosine, was observed in AMRF patients and LIMP2-deficient mice. Our results suggest that combined measurements of chitotriosidase and glucosylsphingosine can be used for convenient differential laboratory diagnosis of GD and AMRF.


Assuntos
Epilepsias Mioclônicas Progressivas/diagnóstico , Animais , Células Cultivadas , Ensaios Enzimáticos , Fibroblastos/enzimologia , Imunofluorescência , Corantes Fluorescentes/química , Glucosilceramidase/metabolismo , Glucosilceramidas/metabolismo , Humanos , Leucócitos/enzimologia , Proteínas de Membrana Lisossomal/deficiência , Macrófagos/enzimologia , Camundongos , Epilepsias Mioclônicas Progressivas/enzimologia , Psicosina/análogos & derivados , Psicosina/metabolismo , Receptores Depuradores/deficiência
17.
Immunology ; 141(3): 292-301, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23876085

RESUMO

Neuroinflammatory changes are a characteristic of several, if not all, neurodegenerative diseases including Alzheimer's disease and are typified by increased microglial activation. Microglia express several receptors making them highly reactive and plastic cells, and, at least in vitro, they adopt different phenotypes in a manner analogous to their peripheral counterparts, macrophages. Microglia also express numerous cell surface proteins enabling them to interact with cells and the evidence indicates that maintenance of microglia in a quiescent state relies, at least to some extent, on an interaction with neurons by means of specific ligand-receptor pairs, for example CD200-CD200R. It is clear that microglia also interact with T cells and recent evidence indicates that co-incubation of microglia with T helper type 1 cells markedly increases their activation. Under normal conditions, small numbers of activated T cells gain entry to the brain and are involved in immune surveillance but infiltration of significant numbers of T cells occurs in disease and following injury. The consequences of T cell infiltration appear to depend on the conditions, with descriptions of both neurodestructive and neuroprotective effects in animal models of different diseases. This review will discuss the modulatory effect of T cells on microglia and the impact of infiltration of T cells into the brain with a focus on Alzheimer's disease, and will propose that infiltration of interferon-γ-producing cells may be an important factor in triggering inflammation that is pathogenic and destructive.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/imunologia , Microglia/imunologia , Linfócitos T/imunologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Comunicação Celular , Progressão da Doença , Humanos , Interferon gama/metabolismo , Microglia/metabolismo , Microglia/patologia , Fenótipo , Prognóstico , Transdução de Sinais , Linfócitos T/metabolismo
18.
Immunology ; 141(2): 174-80, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24032597

RESUMO

The rapid differentiation of monocytes into macrophages (MΦ) and dendritic cells is a pivotal aspect of the innate immune response. Differentiation is triggered following recognition of microbial ligands that activate pattern recognition receptors or directly by pro-inflammatory cytokines. We demonstrate that interleukin-1ß (IL-1ß) induces the rapid differentiation of monocytes into CD209(+) MΦ, similar to activation via Toll-like receptor 2/1, but with distinct phenotypic and functional characteristics. The IL-1ß induced MΦ express higher levels of key markers of phagocytosis, including the Fc-receptors CD16 and CD64, as well as CD36, CD163 and CD206. In addition, IL-1ß-induced MΦ exert potent phagocytic activity towards inert particles, oxidized low-density lipoprotein and mycobacteria. Furthermore, IL-1ß-induced MΦ express higher levels of HLA-DR and effectively present mycobacterial antigens to T cells. Therefore, the ability of IL-1ß to induce monocyte differentiation into MΦ with both phagocytosis and antigen-presenting function is a distinct part of the innate immune response in host defence against microbial infection.


Assuntos
Apresentação de Antígeno , Antígenos de Bactérias/imunologia , Diferenciação Celular/efeitos dos fármacos , Interleucina-1beta/farmacologia , Macrófagos/efeitos dos fármacos , Mycobacterium tuberculosis/imunologia , Linfócitos T/imunologia , Moléculas de Adesão Celular/análise , Humanos , Lectinas Tipo C/análise , Macrófagos/citologia , Macrófagos/fisiologia , Monócitos/citologia , Fagocitose , Receptores de Superfície Celular/análise , Receptor 2 Toll-Like/fisiologia
19.
J Lipid Res ; 54(8): 2095-2108, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23709692

RESUMO

ETC-1002 is an investigational drug currently in Phase 2 development for treatment of dyslipidemia and other cardiometabolic risk factors. In dyslipidemic subjects, ETC-1002 not only reduces plasma LDL cholesterol but also significantly attenuates levels of hsCRP, a clinical biomarker of inflammation. Anti-inflammatory properties of ETC-1002 were further investigated in primary human monocyte-derived macrophages and in in vivo models of inflammation. In cells treated with ETC-1002, increased levels of AMP-activated protein kinase (AMPK) phosphorylation coincided with reduced activity of MAP kinases and decreased production of proinflammatory cytokines and chemokines. AMPK phosphorylation and inhibitory effects of ETC-1002 on soluble mediators of inflammation were significantly abrogated by siRNA-mediated silencing of macrophage liver kinase B1 (LKB1), indicating that ETC-1002 activates AMPK and exerts its anti-inflammatory effects via an LKB1-dependent mechanism. In vivo, ETC-1002 suppressed thioglycollate-induced homing of leukocytes into mouse peritoneal cavity. Similarly, in a mouse model of diet-induced obesity, ETC-1002 restored adipose AMPK activity, reduced JNK phosphorylation, and diminished expression of macrophage-specific marker 4F/80. These data were consistent with decreased epididymal fat-pad mass and interleukin (IL)-6 release by inflamed adipose tissue. Thus, ETC-1002 may provide further clinical benefits for patients with cardiometabolic risk factors by reducing systemic inflammation linked to insulin resistance and vascular complications of metabolic syndrome.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Tecido Adiposo/efeitos dos fármacos , Anti-Inflamatórios não Esteroides/farmacologia , Ácidos Dicarboxílicos/farmacologia , Ácidos Graxos/farmacologia , Leucócitos/efeitos dos fármacos , Macrófagos/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Tecido Adiposo/citologia , Tecido Adiposo/imunologia , Animais , Relação Dose-Resposta a Droga , Humanos , Inflamação , Leucócitos/citologia , Leucócitos/imunologia , Macrófagos/efeitos dos fármacos , Masculino , Fosforilação , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
20.
Immunology ; 140(2): 179-90, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23710834

RESUMO

Plasticity is a hallmark of macrophages, and in response to environmental signals these cells undergo different forms of polarized activation, the extremes of which are called classic (M1) and alternative (M2). Rapamycin (RAPA) is crucial for survival and functions of myeloid phagocytes, but its effects on macrophage polarization are not yet studied. To address this issue, human macrophages obtained from six normal blood donors were polarized to M1 or M2 in vitro by lipopolysaccharide plus interferon-γ or interleukin-4 (IL-4), respectively. The presence of RAPA (10 ng/ml) induced macrophage apoptosis in M2 but not in M1. Beyond the impact on survival in M2, RAPA reduced CXCR4, CD206 and CD209 expression and stem cell growth factor-ß, CCL18 and CCL13 release. In contrast, in M1 RAPA increased CD86 and CCR7 expression and IL-6, tumour necrosis factor-α and IL-1ß release but reduced CD206 and CD209 expression and IL-10, vascular endothelial growth factor and CCL18 release. In view of the in vitro data, we examined the in vivo effect of RAPA monotherapy (0·1 mg/kg/day) in 12 patients who were treated for at least 1 month before islet transplant. Cytokine release by Toll-like receptor 4-stimulated peripheral blood mononuclear cells showed a clear shift to an M1-like profile. Moreover, macrophage polarization 21 days after treatment showed a significant quantitative shift to M1. These results suggest a role of mammalian target of rapamycin (mTOR) into the molecular mechanisms of macrophage polarization and propose new therapeutic strategies for human M2-related diseases through mTOR inhibitor treatment.


Assuntos
Imunossupressores/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Sirolimo/farmacologia , Adulto , Apoptose/efeitos dos fármacos , Citocinas/imunologia , Diabetes Mellitus Tipo 1/cirurgia , Feminino , Citometria de Fluxo , Rejeição de Enxerto/prevenção & controle , Humanos , Transplante das Ilhotas Pancreáticas/imunologia , Masculino , Pessoa de Meia-Idade , Serina-Treonina Quinases TOR/imunologia , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA