Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Cell Biol ; 26(2): 278-293, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38302721

ABSTRACT

Lipids are indispensable for energy storage, membrane structure and cell signalling. However, dynamic changes in various categories of endogenous lipids in mammalian early embryonic development have not been systematically characterized. Here we comprehensively investigated the dynamic lipid landscape during mouse and human early embryo development. Lipid signatures of different developmental stages are distinct, particularly for the phospholipid classes. We highlight that the high degree of phospholipid unsaturation is a conserved feature as embryos develop to the blastocyst stage. Moreover, we show that lipid desaturases such as SCD1 are required for in vitro blastocyst development and blastocyst implantation. One of the mechanisms is through the regulation of unsaturated fatty-acid-mediated fluidity of the plasma membrane and apical proteins and the establishment of apical-basal polarity during development of the eight-cell embryo to the blastocyst. Overall, our study provides an invaluable resource about the remodelling of the endogenous lipidome in mammalian preimplantation embryo development and mechanistic insights into the regulation of embryogenesis and implantation by lipid unsaturation.


Subject(s)
Lipid Metabolism , Lipidomics , Pregnancy , Humans , Female , Mice , Animals , Embryo, Mammalian/metabolism , Embryonic Development/physiology , Blastocyst/metabolism , Phospholipids/metabolism , Mammals
2.
Nature ; 627(8004): 628-635, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38383790

ABSTRACT

Interleukin-10 (IL-10) is a key anti-inflammatory cytokine that can limit immune cell activation and cytokine production in innate immune cell types1. Loss of IL-10 signalling results in life-threatening inflammatory bowel disease in humans and mice-however, the exact mechanism by which IL-10 signalling subdues inflammation remains unclear2-5. Here we find that increased saturated very long chain (VLC) ceramides are critical for the heightened inflammatory gene expression that is a hallmark of IL-10 deficiency. Accordingly, genetic deletion of ceramide synthase 2 (encoded by Cers2), the enzyme responsible for VLC ceramide production, limited the exacerbated inflammatory gene expression programme associated with IL-10 deficiency both in vitro and in vivo. The accumulation of saturated VLC ceramides was regulated by a decrease in metabolic flux through the de novo mono-unsaturated fatty acid synthesis pathway. Restoring mono-unsaturated fatty acid availability to cells deficient in IL-10 signalling limited saturated VLC ceramide production and the associated inflammation. Mechanistically, we find that persistent inflammation mediated by VLC ceramides is largely dependent on sustained activity of REL, an immuno-modulatory transcription factor. Together, these data indicate that an IL-10-driven fatty acid desaturation programme rewires VLC ceramide accumulation and aberrant activation of REL. These studies support the idea that fatty acid homeostasis in innate immune cells serves as a key regulatory node to control pathologic inflammation and suggests that 'metabolic correction' of VLC homeostasis could be an important strategy to normalize dysregulated inflammation caused by the absence of IL-10.


Subject(s)
Inflammation , Interleukin-10 , Sphingolipids , Animals , Humans , Mice , Ceramides/chemistry , Ceramides/metabolism , Fatty Acids, Unsaturated/biosynthesis , Fatty Acids, Unsaturated/metabolism , Homeostasis , Immunity, Innate , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-10/metabolism , Proto-Oncogene Proteins c-rel , Sphingolipids/metabolism
3.
iScience ; 26(9): 107558, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37664623

ABSTRACT

LINC00116 encodes a microprotein first identified as Mitoregulin (MTLN), where it was reported to localize to the inner membrane of mitochondria to regulate fatty acid oxidation and oxidative phosphorylation. These initial discoveries were followed by reports with differing findings about its molecular functions and submitochondrial localization. To clarify the apparent discrepancies, we constructed multiple orthogonal methods of determining the localization of MTLN, including split GFP-based reporters that enable efficient and reliable topology analyses for microproteins. These methods unequivocally demonstrate MTLN primarily localizes to the outer membrane of mitochondria, where it interacts with enzymes of fatty acid metabolism including CPT1B and CYB5B. Loss of MTLN causes the accumulation of very long-chain fatty acids (VLCFAs), especially docosahexaenoic acid (DHA). Intriguingly, loss of MTLN protects mice against western diet/fructose-induced insulin-resistance, suggests a protective effect of VLCFAs in this context. MTLN thus serves as an attractive target to control the catabolism of VLCFAs.

4.
Nat Immunol ; 21(7): 746-755, 2020 07.
Article in English | MEDLINE | ID: mdl-32514064

ABSTRACT

Plasma membranes of animal cells are enriched for cholesterol. Cholesterol-dependent cytolysins (CDCs) are pore-forming toxins secreted by bacteria that target membrane cholesterol for their effector function. Phagocytes are essential for clearance of CDC-producing bacteria; however, the mechanisms by which these cells evade the deleterious effects of CDCs are largely unknown. Here, we report that interferon (IFN) signals convey resistance to CDC-induced pores on macrophages and neutrophils. We traced IFN-mediated resistance to CDCs to the rapid modulation of a specific pool of cholesterol in the plasma membrane of macrophages without changes to total cholesterol levels. Resistance to CDC-induced pore formation requires the production of the oxysterol 25-hydroxycholesterol (25HC), inhibition of cholesterol synthesis and redistribution of cholesterol to an esterified cholesterol pool. Accordingly, blocking the ability of IFN to reprogram cholesterol metabolism abrogates cellular protection and renders mice more susceptible to CDC-induced tissue damage. These studies illuminate targeted regulation of membrane cholesterol content as a host defense strategy.


Subject(s)
Bacterial Infections/immunology , Bacterial Toxins/immunology , Hydroxycholesterols/metabolism , Interferons/isolation & purification , Phagocytes/immunology , Streptolysins/immunology , Animals , Bacteria/immunology , Bacteria/metabolism , Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Cell Membrane/metabolism , Cell Membrane Permeability/immunology , Cells, Cultured , Disease Models, Animal , Disease Susceptibility/immunology , Female , Host Microbial Interactions/immunology , Humans , Intravital Microscopy , Male , Mice , Mice, Transgenic , Phagocytes/cytology , Phagocytes/metabolism , Primary Cell Culture , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Streptolysins/administration & dosage , Streptolysins/metabolism
5.
Cell Metab ; 32(1): 128-143.e5, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32516576

ABSTRACT

Macrophages reprogram their lipid metabolism in response to activation signals. However, a systems-level understanding of how different pro-inflammatory stimuli reshape the macrophage lipidome is lacking. Here, we use complementary "shotgun" and isotope tracer mass spectrometry approaches to define the changes in lipid biosynthesis, import, and composition of macrophages induced by various Toll-like receptors (TLRs) and inflammatory cytokines. "Shotgun" lipidomics data revealed that different TLRs and cytokines induce macrophages to acquire distinct lipidomes, indicating their specificity in reshaping lipid composition. Mechanistic studies showed that differential reprogramming of lipid composition is mediated by the opposing effects of MyD88- and TRIF-interferon-signaling pathways. Finally, we applied these insights to show that perturbing reprogramming of lipid composition can enhance inflammation and promote host defense to bacterial challenge. These studies provide a framework for understanding how inflammatory stimuli reprogram lipid composition of macrophages while providing a knowledge platform to exploit differential lipidomics to influence immunity.


Subject(s)
Lipidomics , Macrophages/metabolism , Toll-Like Receptors/metabolism , Animals , Cell Line , Male , Mice , Mice, Knockout , Mice, Transgenic , Signal Transduction
6.
Cell Rep ; 25(10): 2919-2934.e8, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30517876

ABSTRACT

It is well understood that fatty acids can be synthesized, imported, and modified to meet requisite demands in cells. However, following the movement of fatty acids through the multiplicity of these metabolic steps has remained difficult. To better address this problem, we developed Fatty Acid Source Analysis (FASA), a model that defines the contribution of synthesis, import, and elongation pathways to fatty acid homeostasis in saturated, monounsaturated, and polyunsaturated fatty acid pools. Application of FASA demonstrated that elongation can be a major contributor to cellular fatty acid content and showed that distinct pro-inflammatory stimuli (e.g., Toll-like receptors 2, 3, or 4) specifically reprogram homeostasis of fatty acids by differential utilization of synthetic and elongation pathways in macrophages. In sum, this modeling approach significantly advances our ability to interrogate cellular fatty acid metabolism and provides insight into how cells dynamically reshape their lipidomes in response to metabolic or inflammatory signals.


Subject(s)
Fatty Acids/metabolism , Isotope Labeling/methods , Models, Biological , Animals , Carbon/metabolism , Cell Line , Fatty Acids, Unsaturated/metabolism , Homeostasis , Humans , Inflammation/pathology , Macrophages/metabolism , Male , Mice, Inbred C57BL
7.
Cell ; 163(7): 1716-29, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26686653

ABSTRACT

Cellular lipid requirements are achieved through a combination of biosynthesis and import programs. Using isotope tracer analysis, we show that type I interferon (IFN) signaling shifts the balance of these programs by decreasing synthesis and increasing import of cholesterol and long chain fatty acids. Genetically enforcing this metabolic shift in macrophages is sufficient to render mice resistant to viral challenge, demonstrating the importance of reprogramming the balance of these two metabolic pathways in vivo. Unexpectedly, mechanistic studies reveal that limiting flux through the cholesterol biosynthetic pathway spontaneously engages a type I IFN response in a STING-dependent manner. The upregulation of type I IFNs was traced to a decrease in the pool size of synthesized cholesterol and could be inhibited by replenishing cells with free cholesterol. Taken together, these studies delineate a metabolic-inflammatory circuit that links perturbations in cholesterol biosynthesis with activation of innate immunity.


Subject(s)
Cholesterol/metabolism , Immunity, Innate , Interferon-gamma/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Humans , Interferon beta-1b , Membrane Proteins/metabolism , Mevalonic Acid/metabolism , Mice , Mice, Inbred C57BL , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...