Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 276
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 179(1): 236-250.e18, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31495571

ABSTRACT

Immunotherapy has revolutionized cancer treatment, yet most patients do not respond. Here, we investigated mechanisms of response by profiling the proteome of clinical samples from advanced stage melanoma patients undergoing either tumor infiltrating lymphocyte (TIL)-based or anti- programmed death 1 (PD1) immunotherapy. Using high-resolution mass spectrometry, we quantified over 10,300 proteins in total and ∼4,500 proteins across most samples in each dataset. Statistical analyses revealed higher oxidative phosphorylation and lipid metabolism in responders than in non-responders in both treatments. To elucidate the effects of the metabolic state on the immune response, we examined melanoma cells upon metabolic perturbations or CRISPR-Cas9 knockouts. These experiments indicated lipid metabolism as a regulatory mechanism that increases melanoma immunogenicity by elevating antigen presentation, thereby increasing sensitivity to T cell mediated killing both in vitro and in vivo. Altogether, our proteomic analyses revealed association between the melanoma metabolic state and the response to immunotherapy, which can be the basis for future improvement of therapeutic response.


Subject(s)
Immunotherapy/methods , Melanoma/metabolism , Melanoma/therapy , Mitochondria/metabolism , Proteomics/methods , Skin Neoplasms/metabolism , Skin Neoplasms/therapy , Adoptive Transfer/methods , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cohort Studies , Female , Humans , Lipid Metabolism/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/immunology , Treatment Outcome , Young Adult
2.
Nat Immunol ; 20(5): 626-636, 2019 05.
Article in English | MEDLINE | ID: mdl-30936495

ABSTRACT

Muscle damage elicits a sterile immune response that facilitates complete regeneration. Here, we used mass spectrometry-based lipidomics to map the mediator lipidome during the transition from inflammation to resolution and regeneration in skeletal muscle injury. We observed temporal regulation of glycerophospholipids and production of pro-inflammatory lipid mediators (for example, leukotrienes and prostaglandins) and specialized pro-resolving lipid mediators (for example, resolvins and lipoxins) that were modulated by ibuprofen. These time-dependent profiles were recapitulated in sorted neutrophils and Ly6Chi and Ly6Clo muscle-infiltrating macrophages, with a distinct pro-resolving signature observed in Ly6Clo macrophages. RNA sequencing of macrophages stimulated with resolvin D2 showed similarities to transcriptional changes found during the temporal transition from Ly6Chi macrophage to Ly6Clo macrophage. In vivo, resolvin D2 increased Ly6Clo macrophages and functional improvement of the regenerating muscle. These results reveal dynamic lipid mediator signatures of innate immune cells and provide a proof of concept for their exploitable effector roles in muscle regeneration.


Subject(s)
Inflammation Mediators/immunology , Lipids/immunology , Macrophages/immunology , Muscle, Skeletal/immunology , Regeneration/immunology , Animals , Docosahexaenoic Acids/immunology , Docosahexaenoic Acids/pharmacology , Gene Expression/drug effects , Gene Expression/immunology , Gene Expression Profiling , Gene Ontology , High-Throughput Nucleotide Sequencing , Lipid Metabolism/immunology , Lipids/analysis , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Muscle, Skeletal/injuries , Muscle, Skeletal/physiopathology , Regeneration/genetics
3.
Nat Immunol ; 19(6): 561-570, 2018 06.
Article in English | MEDLINE | ID: mdl-29777213

ABSTRACT

Polarization of macrophages into pro-inflammatory or anti-inflammatory states has distinct metabolic requirements, with mechanistic target of rapamycin (mTOR) kinase signaling playing a critical role. However, it remains unclear how mTOR regulates metabolic status to promote polarization of these cells. Here we show that an mTOR-Semaphorin 6D (Sema6D)-Peroxisome proliferator receptor γ (PPARγ) axis plays critical roles in macrophage polarization. Inhibition of mTOR or loss of Sema6D blocked anti-inflammatory macrophage polarization, concomitant with severe impairments in PPARγ expression, uptake of fatty acids, and lipid metabolic reprogramming. Macrophage expression of the receptor Plexin-A4 is responsible for Sema6D-mediated anti-inflammatory polarization. We found that a tyrosine kinase, c-Abl, which associates with the cytoplasmic region of Sema6D, is required for PPARγ expression. Furthermore, Sema6D is important for generation of intestinal resident CX3CR1hi macrophages and prevents development of colitis. Collectively, these findings highlight crucial roles for Sema6D reverse signaling in macrophage polarization, coupling immunity, and metabolism via PPARγ.


Subject(s)
Inflammation/metabolism , Lipid Metabolism/immunology , Macrophages/metabolism , PPAR gamma/metabolism , Semaphorins/metabolism , Animals , Cell Differentiation/immunology , Colitis/immunology , Inflammation/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , PPAR gamma/immunology , Semaphorins/immunology , Signal Transduction/immunology , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
4.
Immunity ; 52(2): 374-387.e6, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32075729

ABSTRACT

Animals require complex metabolic and physiological adaptations to maintain the function of vital organs in response to environmental stresses and infection. Here, we found that infection or injury in Drosophila induced the excretion of hemolymphatic lipids by Malpighian tubules, the insect kidney. This lipid purge was mediated by a stress-induced lipid-binding protein, Materazzi, which was enriched in Malpighian tubules. Flies lacking materazzi had higher hemolymph concentrations of reactive oxygen species (ROS) and increased lipid peroxidation. These flies also displayed Malpighian tubule dysfunction and were susceptible to infections and environmental stress. Feeding flies with antioxidants rescued the materazzi phenotype, indicating that the main role of Materazzi is to protect the organism from damage caused by stress-induced ROS. Our findings suggest that purging hemolymphatic lipids presents a physiological adaptation to protect host tissues from excessive ROS during immune and stress responses, a process that is likely to apply to other organisms.


Subject(s)
Drosophila melanogaster/immunology , Hemolymph/metabolism , Lipid Metabolism/immunology , Malpighian Tubules/immunology , Reactive Oxygen Species/immunology , Adaptive Immunity , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Diglycerides/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Feces/chemistry , Lipid Peroxidation/immunology , MAP Kinase Signaling System/immunology , Malpighian Tubules/metabolism , Protein Conformation , Reactive Oxygen Species/metabolism , Stress, Physiological/immunology
5.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34006637

ABSTRACT

The liver X receptor (LXR) is a key transcriptional regulator of cholesterol, fatty acid, and phospholipid metabolism. Dynamic remodeling of immunometabolic pathways, including lipid metabolism, is a crucial step in T cell activation. Here, we explored the role of LXR-regulated metabolic processes in primary human CD4+ T cells and their role in controlling plasma membrane lipids (glycosphingolipids and cholesterol), which strongly influence T cell immune signaling and function. Crucially, we identified the glycosphingolipid biosynthesis enzyme glucosylceramide synthase as a direct transcriptional LXR target. LXR activation by agonist GW3965 or endogenous oxysterol ligands significantly altered the glycosphingolipid:cholesterol balance in the plasma membrane by increasing glycosphingolipid levels and reducing cholesterol. Consequently, LXR activation lowered plasma membrane lipid order (stability), and an LXR antagonist could block this effect. LXR stimulation also reduced lipid order at the immune synapse and accelerated activation of proximal T cell signaling molecules. Ultimately, LXR activation dampened proinflammatory T cell function. Finally, compared with responder T cells, regulatory T cells had a distinct pattern of LXR target gene expression corresponding to reduced lipid order. This suggests LXR-driven lipid metabolism could contribute to functional specialization of these T cell subsets. Overall, we report a mode of action for LXR in T cells involving the regulation of glycosphingolipid and cholesterol metabolism and demonstrate its relevance in modulating T cell function.


Subject(s)
Cholesterol/genetics , Glycosphingolipids/genetics , Liver X Receptors/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Benzoates/pharmacology , Benzylamines/pharmacology , Cell Membrane , Cholesterol/immunology , Female , Glucosyltransferases/genetics , Glycosphingolipids/biosynthesis , Glycosphingolipids/immunology , Humans , Immunological Synapses/drug effects , Immunological Synapses/genetics , Ligands , Lipid Metabolism/genetics , Lipid Metabolism/immunology , Liver X Receptors/agonists , Liver X Receptors/antagonists & inhibitors , Liver X Receptors/genetics , Male , Metabolic Networks and Pathways/immunology , Middle Aged , Oxysterols/pharmacology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocytes/metabolism , Young Adult
6.
Semin Cell Dev Biol ; 112: 137-144, 2021 04.
Article in English | MEDLINE | ID: mdl-32807643

ABSTRACT

Microglia are universal sensors of alterations in CNS physiology. These cells integrate complex molecular signals and undergo comprehensive phenotypical remodeling to adapt inflammatory responses. In the last years, single-cell analyses have revealed that microglia exhibit diverse phenotypes during development, growth and disease. Emerging evidence suggests that such phenotype transitions are mediated by reprogramming of cell metabolism. Indeed, metabolic pathways are distinctively altered in activated microglia and are central nodes controlling microglial responses. Microglial lipid metabolism has been specifically involved in the control of microglial activation and effector functions, such as migration, phagocytosis and inflammatory signaling, and minor disturbances in microglial lipid handling associates with altered brain function in disorders featuring neuroinflammation. In this review, we explore new and relevant aspects of microglial metabolism in health and disease. We give special focus on how different branches of lipid metabolism, such as lipid sensing, synthesis and oxidation, integrate and control essential aspects of microglial biology, and how disturbances in these processes associate with aging and the pathogenesis of, for instance, multiple sclerosis and Alzheimer's disease. Finally, challenges and advances in microglial lipid research are discussed.


Subject(s)
Brain/immunology , Immunity, Innate/genetics , Lipid Metabolism/immunology , Neuroinflammatory Diseases/immunology , Brain/metabolism , Humans , Lipid Metabolism/genetics , Lipids/genetics , Lipids/immunology , Microglia/immunology , Microglia/metabolism , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/pathology , Phagocytosis/genetics
7.
PLoS Pathog ; 17(3): e1009365, 2021 03.
Article in English | MEDLINE | ID: mdl-33647060

ABSTRACT

Parasites alter host energy homeostasis for their own development, but the mechanisms underlying this phenomenon remain largely unknown. Here, we show that Cotesia vestalis, an endoparasitic wasp of Plutella xylostella larvae, stimulates a reduction of host lipid levels. This process requires excess secretion of P. xylostella tachykinin (PxTK) peptides from enteroendocrine cells (EEs) in the midgut of the parasitized host larvae. We found that parasitization upregulates PxTK signaling to suppress lipogenesis in midgut enterocytes (ECs) in a non-cell-autonomous manner, and the reduced host lipid level benefits the development of wasp offspring and their subsequent parasitic ability. We further found that a C. vestalis bracovirus (CvBV) gene, CvBV 9-2, is responsible for PxTK induction, which in turn reduces the systemic lipid level of the host. Taken together, these findings illustrate a novel mechanism for parasite manipulation of host energy homeostasis by a symbiotic bracovirus gene to promote the development and increase the parasitic efficiency of an agriculturally important wasp species.


Subject(s)
Host-Parasite Interactions/immunology , Lipid Metabolism/physiology , Parasites/virology , Polydnaviridae/genetics , Animals , Digestive System/metabolism , Host-Parasite Interactions/genetics , Larva/metabolism , Larva/virology , Lipid Metabolism/immunology , Parasites/pathogenicity , Polydnaviridae/pathogenicity , Signal Transduction/immunology , Signal Transduction/physiology , Wasps/physiology , Wasps/virology
8.
Nat Immunol ; 12(1): 5-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21169997

ABSTRACT

The fields of immunology, microbiology, nutrition and metabolism are rapidly converging. Here we expand on a diet-microbiota model as the basis for the greater incidence of asthma and autoimmunity in developed countries.


Subject(s)
Diet , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Lipid Metabolism/immunology , Metagenome/immunology , Animals , Clinical Trials as Topic , Genetic Predisposition to Disease , Humans , Immunity, Mucosal/genetics , Immunomodulation , Inflammation , Mice , Models, Immunological , Signal Transduction/genetics , Signal Transduction/immunology
9.
Lipids Health Dis ; 22(1): 146, 2023 Sep 07.
Article in English | MEDLINE | ID: mdl-37679775

ABSTRACT

BACKGROUND: Lipid metabolism affects type 2 immunity; however, the association between plasma lipids and eosinophilic inflammation in humans is uncertain. This study analysed the relationship between plasma lipids and peripheral eosinophils and whether patterns differ with different body mass indexes (BMI). METHODS: A cross-sectional survey including 62,441 healthy participants recruited from a regular health screening programme was conducted. Participants were divided into normal weight, overweight and obese subgroups according to BMI. RESULTS: Multiple linear regression analysis revealed that elevated logarithmic-transformed eosinophil counts (log(EOS)) significantly correlated with high total cholesterol(TC), triglyceride(TG), low-density lipoprotein-cholesterol (LDL-C), and low high-density lipoprotein-cholesterol (HDL-C)levels in the overall population, as well as in men and women, while certain associations between peripheral blood eosinophil percentage and serum lipids varied by gender. These correlations existed across almost all BMI subgroups, and standardised ß values decreased sequentially with increasing BMI. HDL-C had the most significant effect on eosinophils in obese women. Two-factor analysis of variance showed log(EOS) increased with higher BMI and hyperlipidemia whether in male or female and a synergistic effect exists of lipid levels (TG and LDL-C) and BMI in men. CONCLUSIONS: Blood eosinophil counts were correlated with blood lipid levels and modified by body mass index status. The effects of lipid levels and body mass index on blood eosinophil counts were synergistic. Therefore, lipid metabolism may be involved in systemic eosinophil inflammation.


Subject(s)
Body Mass Index , East Asian People , Eosinophils , Inflammation , Lipids , Female , Humans , Male , Cholesterol, LDL , Cross-Sectional Studies , Eosinophils/immunology , Inflammation/blood , Inflammation/immunology , Lipids/blood , Lipid Metabolism/immunology
10.
PLoS Genet ; 16(11): e1009192, 2020 11.
Article in English | MEDLINE | ID: mdl-33227003

ABSTRACT

During infection, cellular resources are allocated toward the metabolically-demanding processes of synthesizing and secreting effector proteins that neutralize and kill invading pathogens. In Drosophila, these effectors are antimicrobial peptides (AMPs) that are produced in the fat body, an organ that also serves as a major lipid storage depot. Here we asked how activation of Toll signaling in the larval fat body perturbs lipid homeostasis to understand how cells meet the metabolic demands of the immune response. We find that genetic or physiological activation of fat body Toll signaling leads to a tissue-autonomous reduction in triglyceride storage that is paralleled by decreased transcript levels of the DGAT homolog midway, which carries out the final step of triglyceride synthesis. In contrast, Kennedy pathway enzymes that synthesize membrane phospholipids are induced. Mass spectrometry analysis revealed elevated levels of major phosphatidylcholine and phosphatidylethanolamine species in fat bodies with active Toll signaling. The ER stress mediator Xbp1 contributed to the Toll-dependent induction of Kennedy pathway enzymes, which was blunted by deleting AMP genes, thereby reducing secretory demand elicited by Toll activation. Consistent with ER stress induction, ER volume is expanded in fat body cells with active Toll signaling, as determined by transmission electron microscopy. A major functional consequence of reduced Kennedy pathway induction is an impaired immune response to bacterial infection. Our results establish that Toll signaling induces a shift in anabolic lipid metabolism to favor phospholipid synthesis and ER expansion that may serve the immediate demand for AMP synthesis and secretion but with the long-term consequence of insufficient nutrient storage.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Gram-Positive Bacterial Infections/immunology , Immunity, Innate , Lipid Metabolism/immunology , Animals , Animals, Genetically Modified , Antimicrobial Cationic Peptides/genetics , Choline-Phosphate Cytidylyltransferase/genetics , Choline-Phosphate Cytidylyltransferase/metabolism , DNA-Binding Proteins/metabolism , Diacylglycerol O-Acyltransferase/metabolism , Disease Models, Animal , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/immunology , Enterococcus faecalis/immunology , Fat Body/enzymology , Fat Body/immunology , Female , Gram-Positive Bacterial Infections/microbiology , Humans , Larva/enzymology , Larva/immunology , Lipid Metabolism/genetics , Male , Phospholipids/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptors/metabolism , Triglycerides/metabolism
11.
Int J Mol Sci ; 24(15)2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37569407

ABSTRACT

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.


Subject(s)
Cell Polarity , Inflammation , Lipid Metabolism , Macrophages , Neoplasms , Lipid Metabolism/immunology , Cell Polarity/immunology , Inflammation/immunology , Neoplasms/immunology , Macrophages/immunology , Cholesterol/metabolism , Fatty Acids/metabolism , Ferroptosis , Humans
12.
Int J Mol Sci ; 24(3)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36768336

ABSTRACT

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.


Subject(s)
Fatigue Syndrome, Chronic , Fatty Acids , Lymphocytes , Humans , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Fatigue Syndrome, Chronic/immunology , Killer Cells, Natural , Fatty Acids/immunology , Oxidation-Reduction , Lipid Metabolism/immunology , Lymphocytes/immunology , Lymphocyte Subsets/immunology
13.
J Hepatol ; 76(2): 283-293, 2022 02.
Article in English | MEDLINE | ID: mdl-34627976

ABSTRACT

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.


Subject(s)
Environmental Exposure/adverse effects , Lipid Metabolism/physiology , Non-alcoholic Fatty Liver Disease/complications , Adult , Amino Acids/analysis , Amino Acids/blood , Animals , Cohort Studies , Disease Models, Animal , Environmental Exposure/statistics & numerical data , Fatty Acids, Nonesterified/analysis , Fatty Acids, Nonesterified/blood , Female , Humans , Lipid Metabolism/immunology , Male , Mice , Middle Aged , Non-alcoholic Fatty Liver Disease/metabolism
14.
Eur J Immunol ; 51(7): 1628-1640, 2021 07.
Article in English | MEDLINE | ID: mdl-33788250

ABSTRACT

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.


Subject(s)
Dendritic Cells/immunology , Fatty Acids/immunology , Macrophages/immunology , Animals , Humans , Lipid Metabolism/immunology , Myeloid Cells/immunology
15.
Hepatology ; 73(2): 586-605, 2021 02.
Article in English | MEDLINE | ID: mdl-32297339

ABSTRACT

BACKGROUND AND AIMS: Milk fat globule-epidermal growth factor-factor 8 (MFGE8) has been shown to be a critical extracellular molecule that mediates apoptotic signaling in the pathological process of nonalcoholic fatty liver disease (NAFLD). MFGE8 is abundantly expressed in hepatocytes, but its function in the pathogenesis of NAFLD has not been characterized. APPROACH AND RESULTS: In our current study, hepatic MFGE8 showed a protective role in the pathogenesis of NAFLD. Hepatic MFGE8 deletion largely exacerbated lipid accumulation and inflammatory responses in the liver in response to overnutrition. Mechanistically, intercellular MFGE8 was shown to directly bind to apoptosis signal-regulating kinase 1 (ASK1) and to inhibit its dimerization and phosphorylation under a normal diet. However, under metabolic challenges, decreased cytoplasmic MFGE8 facilitated the dimerization and phosphorylation of ASK1 and subsequent mitogen-activated protein kinase signaling in hepatocytes. CONCLUSIONS: Hepatic MFGE8 is an endogenous inhibitor that halts the progression of hepatic steatosis and inflammation. Metabolic challenge-induced loss of intracellular MFGE8 facilitates ASK1 dimerization and phosphorylation. Therefore, maintaining hepatic MFGE8 levels may serve as an alternative strategy for the treatment of NAFLD.


Subject(s)
Antigens, Surface/metabolism , Liver/pathology , Milk Proteins/metabolism , Non-alcoholic Fatty Liver Disease/immunology , Animals , Antigens, Surface/genetics , Diet, High-Fat/adverse effects , Disease Models, Animal , Hepatocytes , Humans , Lipid Metabolism/immunology , Liver/immunology , MAP Kinase Kinase Kinase 5/metabolism , MAP Kinase Signaling System/immunology , Male , Mice , Mice, Knockout , Milk Proteins/genetics , Non-alcoholic Fatty Liver Disease/pathology , Phosphorylation/immunology , Protein Multimerization/immunology
16.
Blood ; 136(26): 3004-3017, 2020 12 24.
Article in English | MEDLINE | ID: mdl-32818230

ABSTRACT

Natural killer (NK) cells play critical roles in protection against hematological malignancies but can acquire a dysfunctional state, which limits antitumor immunity. However, the underlying reasons for this impaired NK cell function remain to be uncovered. We found that NK cells in aggressive B-cell lymphoma underwent substantial transcriptional reprogramming associated with increased lipid metabolism, including elevated expression of the transcriptional regulator peroxisome activator receptor-γ (PPAR-γ). Exposure to fatty acids in the lymphoma environment potently suppressed NK cell effector response and cellular metabolism. NK cells from both diffuse large B-cell lymphoma patients and Eµ-myc B-cell lymphoma-bearing mice displayed reduced interferon-γ (IFN-γ) production. Activation of PPAR-γ partially restored mitochondrial membrane potential and IFN-γ production. Overall, our data indicate that increased lipid metabolism, while impairing their function, is a functional adaptation of NK cells to the fatty-acid rich lymphoma environment.


Subject(s)
Killer Cells, Natural/immunology , Lipid Metabolism/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Tumor Microenvironment/immunology , Animals , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Killer Cells, Natural/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Membrane Potential, Mitochondrial/genetics , Membrane Potential, Mitochondrial/immunology , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , PPAR gamma/genetics , PPAR gamma/immunology , Tumor Microenvironment/genetics
17.
J Immunol ; 205(1): 193-201, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32434939

ABSTRACT

Thermal burn injuries are an important environmental stressor that can result in considerable morbidity and mortality. The exact mechanism by which an environmental stimulus to skin results in local and systemic effects is an area of active research. One potential mechanism to allow skin keratinocytes to disperse bioactive substances is via microvesicle particles, which are subcellular bodies released directly from cellular membranes. Our previous studies have indicated that thermal burn injury of the skin keratinocyte in vitro results in the production of the lipid mediator platelet-activating factor (PAF). The present studies demonstrate that thermal burn injury to keratinocytes in vitro and human skin explants ex vivo, and mice in vivo generate microvesicle particles. Use of pharmacologic and genetic tools indicates that the optimal release of microvesicles is dependent upon the PAF receptor. Of note, burn injury-stimulated microvesicle particles do not carry appreciable protein cytokines yet contain high levels of PAF. These studies describe a novel mechanism involving microvesicle particles by which a metabolically labile bioactive lipid can travel from cells in response to environmental stimuli.


Subject(s)
Burns/immunology , Cell-Derived Microparticles/immunology , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Skin/pathology , Animals , Biopsy , Burns/pathology , Cell Line , Cell-Derived Microparticles/metabolism , Disease Models, Animal , Female , Humans , Keratinocytes/immunology , Keratinocytes/metabolism , Lipid Metabolism/immunology , Mice , Mice, Knockout , Platelet Membrane Glycoproteins/genetics , Primary Cell Culture , Receptors, G-Protein-Coupled/genetics , Skin/immunology
18.
Int J Mol Sci ; 23(3)2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35163232

ABSTRACT

Despite all the advances of modern medicine, atherosclerosis continues to be one of the most important medical and social problems. Atherosclerosis is the cause of several cardiovascular diseases, which are associated with high rates of disability and mortality. The development of atherosclerosis is associated with the accumulation of lipids in the arterial intima and the disruption of mechanisms that maintain the balance between the development and resolution of inflammation. Fatty acids are involved in many mechanisms of inflammation development and maintenance. Endothelial cells demonstrate multiple cross-linkages between lipid metabolism and innate immunity. In addition, these processes are linked to hemodynamics and the function of other cells in the vascular wall, highlighting the central role of the endothelium in vascular biology.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/metabolism , Fatty Acids/metabolism , Animals , Cardiovascular Diseases/metabolism , Eicosanoids/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Fatty Acids/immunology , Hemodynamics , Humans , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Lipid Metabolism/immunology , Lipid Metabolism/physiology , Lipids/physiology , Tunica Intima/metabolism
19.
Am J Respir Cell Mol Biol ; 65(4): 390-402, 2021 10.
Article in English | MEDLINE | ID: mdl-34003729

ABSTRACT

Obstructive sleep apnea is associated with insulin resistance, lipid dysregulation, and hepatic steatosis and fibrosis in nonalcoholic fatty liver disease (NAFLD). We have previously shown that hepatocyte HIF-1 (hypoxia-inducible factor-1) mediates the development of liver fibrosis in a mouse model of NAFLD. We hypothesized that intermittent hypoxia (IH) modeling obstructive sleep apnea would worsen hepatic steatosis and fibrosis in murine NAFLD, via HIF-1. Mice with hepatocyte-specific deletion of Hif1a (Hif1a-/-hep) and wild-type (Hif1aF/F) controls were fed a high trans-fat diet to induce NAFLD with steatohepatitis. Half from each group were exposed to IH, and the other half were exposed to intermittent air. A glucose tolerance test was performed just prior to the end of the experiment. Mitochondrial efficiency was assessed in fresh liver tissue at the time of death. The hepatic malondialdehyde concentration and proinflammatory cytokine levels were assessed, and genes of collagen and fatty acid metabolism were examined. Hif1a-/-hep mice gained less weight than wild-type Hif1a mice (-2.3 g, P = 0.029). There was also a genotype-independent effect of IH on body weight, with less weight gain in mice exposed to IH (P = 0.003). Fasting glucose, homeostatic model assessment for insulin resistance, and glucose tolerance test results were all improved in Hif1a-/-hep mice. Liver collagen was increased in mice exposed to IH (P = 0.033) and was reduced in Hif1a-/-hep mice (P < 0.001), without any significant exposure/genotype interaction being demonstrated. Liver TNF-α and IL-1ß were significantly increased in mice exposed to IH and were decreased in Hif1a-/-hep mice. We conclude that HIF-1 signaling worsens the metabolic profile and hastens NAFLD progression and that IH may worsen liver fibrosis. These effects are plausibly mediated by hepatic inflammatory stress.


Subject(s)
Hepatocytes/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia/complications , Liver Cirrhosis/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Hepatocytes/pathology , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin Resistance/physiology , Lipid Metabolism/immunology , Liver/metabolism , Liver Cirrhosis/complications , Liver Cirrhosis/pathology , Mice
20.
Cell Immunol ; 364: 104341, 2021 06.
Article in English | MEDLINE | ID: mdl-33798909

ABSTRACT

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.


Subject(s)
Asthma/metabolism , Granulocytes/immunology , Lipid Metabolism/immunology , Macrophages/immunology , T-Lymphocytes/immunology , Animals , Asthma/drug therapy , Asthma/epidemiology , Fatty Acids/metabolism , Humans , Immunity, Cellular , Molecular Targeted Therapy , Risk Factors , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL