Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters











Publication year range
1.
bioRxiv ; 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38559079

ABSTRACT

The intrinsic pathways that control membrane organization in immune cells and the impact of such pathways on cellular function are not well defined. Here we report that the non-vesicular cholesterol transporter Aster-A links plasma membrane (PM) cholesterol availability in T cells to immune signaling and systemic metabolism. Aster-A is recruited to the PM during T-cell receptor (TCR) activation, where it facilitates the removal of newly generated "accessible" membrane cholesterol. Loss of Aster-A leads to excess PM cholesterol accumulation, resulting in enhanced TCR nano-clustering and signaling, and Th17 cytokine production. Finally, we show that the mucosal Th17 response is restrained by PM cholesterol remodeling. Ablation of Aster-A in T cells leads to enhanced IL-22 production, reduced intestinal fatty acid absorption, and resistance to diet-induced obesity. These findings delineate a multi-tiered regulatory scheme linking immune cell lipid flux to nutrient absorption and systemic physiology.

2.
Science ; 382(6671): eadf0966, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37943936

ABSTRACT

Intestinal absorption is an important contributor to systemic cholesterol homeostasis. Niemann-Pick C1 Like 1 (NPC1L1) assists in the initial step of dietary cholesterol uptake, but how cholesterol moves downstream of NPC1L1 is unknown. We show that Aster-B and Aster-C are critical for nonvesicular cholesterol movement in enterocytes. Loss of NPC1L1 diminishes accessible plasma membrane (PM) cholesterol and abolishes Aster recruitment to the intestinal brush border. Enterocytes lacking Asters accumulate PM cholesterol and show endoplasmic reticulum cholesterol depletion. Aster-deficient mice have impaired cholesterol absorption and are protected against diet-induced hypercholesterolemia. Finally, the Aster pathway can be targeted with a small-molecule inhibitor to manipulate cholesterol uptake. These findings identify the Aster pathway as a physiologically important and pharmacologically tractable node in dietary lipid absorption.


Subject(s)
Cholesterol, Dietary , Enterocytes , Intestinal Absorption , Membrane Transport Proteins , Animals , Mice , Biological Transport , Cholesterol, Dietary/metabolism , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice, Inbred C57BL , Enterocytes/metabolism , Liver X Receptors/metabolism , Humans , Jejunum/metabolism , Mice, Knockout
3.
J Clin Invest ; 133(23)2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37824203

ABSTRACT

Why apolipoprotein AV (APOA5) deficiency causes hypertriglyceridemia has remained unclear, but we have suspected that the underlying cause is reduced amounts of lipoprotein lipase (LPL) in capillaries. By routine immunohistochemistry, we observed reduced LPL staining of heart and brown adipose tissue (BAT) capillaries in Apoa5-/- mice. Also, after an intravenous injection of LPL-, CD31-, and GPIHBP1-specific mAbs, the binding of LPL Abs to heart and BAT capillaries (relative to CD31 or GPIHBP1 Abs) was reduced in Apoa5-/- mice. LPL levels in the postheparin plasma were also lower in Apoa5-/- mice. We suspected that a recent biochemical observation - that APOA5 binds to the ANGPTL3/8 complex and suppresses its capacity to inhibit LPL catalytic activity - could be related to the low intracapillary LPL levels in Apoa5-/- mice. We showed that an ANGPTL3/8-specific mAb (IBA490) and APOA5 normalized plasma triglyceride (TG) levels and intracapillary LPL levels in Apoa5-/- mice. We also showed that ANGPTL3/8 detached LPL from heparan sulfate proteoglycans and GPIHBP1 on the surface of cells and that the LPL detachment was blocked by IBA490 and APOA5. Our studies explain the hypertriglyceridemia in Apoa5-/- mice and further illuminate the molecular mechanisms that regulate plasma TG metabolism.


Subject(s)
Apolipoprotein A-V , Hypertriglyceridemia , Receptors, Lipoprotein , Animals , Mice , Capillaries/metabolism , Hypertriglyceridemia/genetics , Hypertriglyceridemia/metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism , Triglycerides/blood , Apolipoprotein A-V/genetics
4.
Proc Natl Acad Sci U S A ; 120(44): e2313825120, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37871217

ABSTRACT

Lipoprotein lipase (LPL), the enzyme that carries out the lipolytic processing of triglyceride-rich lipoproteins (TRLs), is synthesized by adipocytes and myocytes and secreted into the interstitial spaces. The LPL is then bound by GPIHBP1, a GPI-anchored protein of endothelial cells (ECs), and transported across ECs to the capillary lumen. The assumption has been that the LPL that is moved into capillaries remains attached to GPIHBP1 and that GPIHBP1 serves as a platform for TRL processing. In the current studies, we examined the validity of that assumption. We found that an LPL-specific monoclonal antibody (mAb), 88B8, which lacks the ability to detect GPIHBP1-bound LPL, binds avidly to LPL within capillaries. We further demonstrated, by confocal microscopy, immunogold electron microscopy, and nanoscale secondary ion mass spectrometry analyses, that the LPL detected by mAb 88B8 is located within the EC glycocalyx, distant from the GPIHBP1 on the EC plasma membrane. The LPL within the glycocalyx mediates the margination of TRLs along capillaries and is active in TRL processing, resulting in the delivery of lipoprotein-derived lipids to immediately adjacent parenchymal cells. Thus, the LPL that GPIHBP1 transports into capillaries can detach and move into the EC glycocalyx, where it functions in the intravascular processing of TRLs.


Subject(s)
Lipoprotein Lipase , Receptors, Lipoprotein , Antibodies, Monoclonal/metabolism , Capillaries/metabolism , Endothelial Cells/metabolism , Glycocalyx/metabolism , Lipoprotein Lipase/metabolism , Lipoproteins/metabolism , Receptors, Lipoprotein/metabolism , Triglycerides/metabolism , Humans , Animals
5.
bioRxiv ; 2023 Jul 10.
Article in English | MEDLINE | ID: mdl-37503112

ABSTRACT

Intestinal cholesterol absorption is an important contributor to systemic cholesterol homeostasis. Niemann-Pick C1 Like 1 (NPC1L1), the target of the drug ezetimibe (EZ), assists in the initial step of dietary cholesterol uptake. However, how cholesterol moves downstream of NPC1L1 is unknown. Here we show that Aster-B and Aster-C are critical for non-vesicular cholesterol movement in enterocytes, bridging NPC1L1 at the plasma membrane (PM) and ACAT2 in the endoplasmic reticulum (ER). Loss of NPC1L1 diminishes accessible PM cholesterol in enterocytes and abolishes Aster recruitment to the intestinal brush border. Enterocytes lacking Asters accumulate cholesterol at the PM and display evidence of ER cholesterol depletion, including decreased cholesterol ester stores and activation of the SREBP-2 transcriptional pathway. Aster-deficient mice have impaired cholesterol absorption and are protected against diet-induced hypercholesterolemia. Finally, we show that the Aster pathway can be targeted with a small molecule inhibitor to manipulate dietary cholesterol uptake. These findings identify the Aster pathway as a physiologically important and pharmacologically tractable node in dietary lipid absorption. One-Sentence Summary: Identification of a targetable pathway for regulation of dietary cholesterol absorption.

6.
Proc Natl Acad Sci U S A ; 120(8): e2219833120, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36787365

ABSTRACT

Lipoprotein lipase (LPL) is secreted into the interstitial spaces by parenchymal cells and then transported into capillaries by GPIHBP1. LPL carries out the lipolytic processing of triglyceride (TG)-rich lipoproteins (TRLs), but the tissue-specific regulation of LPL is incompletely understood. Plasma levels of TG hydrolase activity after heparin injection are often used to draw inferences about intravascular LPL levels, but the validity of these inferences is unclear. Moreover, plasma TG hydrolase activity levels are not helpful for understanding LPL regulation in specific tissues. Here, we sought to elucidate LPL regulation under thermoneutral conditions (30 °C). To pursue this objective, we developed an antibody-based method to quantify (in a direct fashion) LPL levels inside capillaries. At 30 °C, intracapillary LPL levels fell sharply in brown adipose tissue (BAT) but not heart. The reduced intracapillary LPL levels were accompanied by reduced margination of TRLs along capillaries. ANGPTL4 expression in BAT increased fourfold at 30 °C, suggesting a potential explanation for the lower intracapillary LPL levels. Consistent with that idea, Angptl4 deficiency normalized both LPL levels and TRL margination in BAT at 30 °C. In Gpihbp1-/- mice housed at 30 °C, we observed an ANGPTL4-dependent decrease in LPL levels within the interstitial spaces of BAT, providing in vivo proof that ANGPTL4 regulates LPL levels before LPL transport into capillaries. In conclusion, our studies have illuminated intracapillary LPL regulation under thermoneutral conditions. Our approaches will be useful for defining the impact of genetic variation and metabolic disease on intracapillary LPL levels and TRL processing.


Subject(s)
Adipose Tissue, Brown , Receptors, Lipoprotein , Animals , Mice , Adipose Tissue/metabolism , Adipose Tissue, Brown/metabolism , Antibodies/metabolism , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/metabolism , Temperature , Triglycerides/metabolism
7.
Nat Metab ; 5(1): 165-181, 2023 01.
Article in English | MEDLINE | ID: mdl-36646756

ABSTRACT

In cell models, changes in the 'accessible' pool of plasma membrane (PM) cholesterol are linked with the regulation of endoplasmic reticulum sterol synthesis and metabolism by the Aster family of nonvesicular transporters; however, the relevance of such nonvesicular transport mechanisms for lipid homeostasis in vivo has not been defined. Here we reveal two physiological contexts that generate accessible PM cholesterol and engage the Aster pathway in the liver: fasting and reverse cholesterol transport. During fasting, adipose-tissue-derived fatty acids activate hepatocyte sphingomyelinase to liberate sequestered PM cholesterol. Aster-dependent cholesterol transport during fasting facilitates cholesteryl ester formation, cholesterol movement into bile and very low-density lipoprotein production. During reverse cholesterol transport, high-density lipoprotein delivers excess cholesterol to the hepatocyte PM through scavenger receptor class B member 1. Loss of hepatic Asters impairs cholesterol movement into feces, raises plasma cholesterol levels and causes cholesterol accumulation in peripheral tissues. These results reveal fundamental mechanisms by which Aster cholesterol flux contributes to hepatic and systemic lipid homeostasis.


Subject(s)
Cholesterol , Liver , Cholesterol/metabolism , Biological Transport/physiology , Liver/metabolism , Homeostasis , Fatty Acids/metabolism
8.
Nature ; 613(7942): 160-168, 2023 01.
Article in English | MEDLINE | ID: mdl-36477540

ABSTRACT

Multilocular adipocytes are a hallmark of thermogenic adipose tissue1,2, but the factors that enforce this cellular phenotype are largely unknown. Here, we show that an adipocyte-selective product of the Clstn3 locus (CLSTN3ß) present in only placental mammals facilitates the efficient use of stored triglyceride by limiting lipid droplet (LD) expansion. CLSTN3ß is an integral endoplasmic reticulum (ER) membrane protein that localizes to ER-LD contact sites through a conserved hairpin-like domain. Mice lacking CLSTN3ß have abnormal LD morphology and altered substrate use in brown adipose tissue, and are more susceptible to cold-induced hypothermia despite having no defect in adrenergic signalling. Conversely, forced expression of CLSTN3ß is sufficient to enforce a multilocular LD phenotype in cultured cells and adipose tissue. CLSTN3ß associates with cell death-inducing DFFA-like effector proteins and impairs their ability to transfer lipid between LDs, thereby restricting LD fusion and expansion. Functionally, increased LD surface area in CLSTN3ß-expressing adipocytes promotes engagement of the lipolytic machinery and facilitates fatty acid oxidation. In human fat, CLSTN3B is a selective marker of multilocular adipocytes. These findings define a molecular mechanism that regulates LD form and function to facilitate lipid utilization in thermogenic adipocytes.


Subject(s)
Adipocytes , Calcium-Binding Proteins , Lipid Metabolism , Membrane Proteins , Animals , Female , Humans , Mice , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/metabolism , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Placenta , Triglycerides/metabolism , Endoplasmic Reticulum/metabolism , Lipid Droplets/metabolism , Fatty Acids/metabolism , Hypothermia/metabolism , Thermogenesis
9.
JCI Insight ; 6(16)2021 08 23.
Article in English | MEDLINE | ID: mdl-34423791

ABSTRACT

The mutant nuclear lamin protein (progerin) produced in Hutchinson-Gilford progeria syndrome (HGPS) results in loss of arterial smooth muscle cells (SMCs), but the mechanism has been unclear. We found that progerin induces repetitive nuclear membrane (NM) ruptures, DNA damage, and cell death in cultured SMCs. Reducing lamin B1 expression and exposing cells to mechanical stress - to mirror conditions in the aorta - triggered more frequent NM ruptures. Increasing lamin B1 protein levels had the opposite effect, reducing NM ruptures and improving cell survival. Remarkably, raising lamin B1 levels increased nuclear compliance in cells and was able to offset the increased nuclear stiffness caused by progerin. In mice, lamin B1 expression in aortic SMCs is normally very low, and in mice with a targeted HGPS mutation (LmnaG609G), levels of lamin B1 decrease further with age while progerin levels increase. Those observations suggest that NM ruptures might occur in aortic SMCs in vivo. Indeed, studies in LmnaG609G mice identified NM ruptures in aortic SMCs, along with ultrastructural abnormalities in the cell nucleus that preceded SMC loss. Our studies identify NM ruptures in SMCs as likely causes of vascular pathology in HGPS.


Subject(s)
Aorta/pathology , Lamin Type A/genetics , Muscle, Smooth, Vascular/pathology , Nuclear Envelope/pathology , Progeria/pathology , Animals , Aorta/cytology , Disease Models, Animal , Humans , Lamin Type A/metabolism , Lamin Type B/genetics , Lamin Type B/metabolism , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/cytology , Mutation , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/pathology , Progeria/genetics
10.
Nucleic Acids Res ; 49(1): 1-14, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33275144

ABSTRACT

Nucleic acid therapeutics (NATs) have proven useful in promoting the degradation of specific transcripts, modifying gene expression, and regulating mRNA splicing. In each situation, efficient delivery of nucleic acids to cells, tissues and intracellular compartments is crucial-both for optimizing efficacy and reducing side effects. Despite successes in NATs, our understanding of their cellular uptake and distribution in tissues is limited. Current methods have yielded insights into distribution of NATs within cells and tissues, but the sensitivity and resolution of these approaches are limited. Here, we show that nanoscale secondary ion mass spectrometry (NanoSIMS) imaging can be used to define the distribution of 5-bromo-2'-deoxythymidine (5-BrdT) modified antisense oligonucleotides (ASO) in cells and tissues with high sensitivity and spatial resolution. This approach makes it possible to define ASO uptake and distribution in different subcellular compartments and to quantify the impact of targeting ligands designed to promote ASO uptake by cells. Our studies showed that phosphorothioate ASOs are associated with filopodia and the inner nuclear membrane in cultured cells, and also revealed substantial cellular and subcellular heterogeneity of ASO uptake in mouse tissues. NanoSIMS imaging represents a significant advance in visualizing uptake and distribution of NATs; this approach will be useful in optimizing efficacy and delivery of NATs for treating human disease.


Subject(s)
Oligonucleotides, Antisense/analysis , Phosphorothioate Oligonucleotides/analysis , Spectrometry, Mass, Secondary Ion/methods , 3T3-L1 Cells , Acetylgalactosamine/administration & dosage , Acetylgalactosamine/analysis , Animals , Asialoglycoprotein Receptor/analysis , Cesium , HEK293 Cells , HeLa Cells , Humans , Kidney/chemistry , Kidney/ultrastructure , Liver/chemistry , Liver/ultrastructure , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron , Myocardium/chemistry , Myocardium/ultrastructure , Oligonucleotides, Antisense/pharmacokinetics , Phosphorothioate Oligonucleotides/pharmacokinetics , Pseudopodia/chemistry , Pseudopodia/ultrastructure , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics , Subcellular Fractions/chemistry , Sulfur/analysis , Sulfur Isotopes/analysis , Tissue Distribution
11.
J Clin Invest ; 131(2)2021 01 19.
Article in English | MEDLINE | ID: mdl-33211671

ABSTRACT

TH17 cell subpopulations have been defined that contribute to inflammation and homeostasis, yet the characteristics of TH17 cells that contribute to host defense against infection are not clear. To elucidate the antimicrobial machinery of the TH17 subset, we studied the response to Cutibacterium acnes, a skin commensal that is resistant to IL-26, the only known TH17-secreted protein with direct antimicrobial activity. We generated C. acnes-specific antimicrobial TH17 clones (AMTH17) with varying antimicrobial activity against C. acnes, which we correlated by RNA sequencing to the expression of transcripts encoding proteins that contribute to antimicrobial activity. Additionally, we validated that AMTH17-mediated killing of C. acnes and bacterial pathogens was dependent on the secretion of granulysin, granzyme B, perforin, and histone H2B. We found that AMTH17 cells can release fibrous structures composed of DNA decorated with histone H2B that entangle C. acnes that we call T cell extracellular traps (TETs). Within acne lesions, H2B and IL-17 colocalized in CD4+ T cells, in proximity to TETs in the extracellular space composed of DNA decorated with H2B. This study identifies a functionally distinct subpopulation of TH17 cells with an ability to form TETs containing secreted antimicrobial proteins that capture and kill bacteria.


Subject(s)
Acne Vulgaris/immunology , Extracellular Traps/immunology , Propionibacteriaceae/immunology , Skin Diseases, Bacterial/immunology , Th17 Cells/immunology , Acne Vulgaris/microbiology , Humans , RNA-Seq , Skin Diseases, Bacterial/microbiology
12.
Proc Natl Acad Sci U S A ; 117(27): 15827-15836, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32571911

ABSTRACT

Bromine and peroxidasin (an extracellular peroxidase) are essential for generating sulfilimine cross-links between a methionine and a hydroxylysine within collagen IV, a basement membrane protein. The sulfilimine cross-links increase the structural integrity of basement membranes. The formation of sulfilimine cross-links depends on the ability of peroxidasin to use bromide and hydrogen peroxide substrates to produce hypobromous acid (HOBr). Once a sulfilimine cross-link is created, bromide is released into the extracellular space and becomes available for reutilization. Whether the HOBr generated by peroxidasin is used very selectively for creating sulfilimine cross-links or whether it also causes oxidative damage to bystander molecules (e.g., generating bromotyrosine residues in basement membrane proteins) is unclear. To examine this issue, we used nanoscale secondary ion mass spectrometry (NanoSIMS) imaging to define the distribution of bromine in mammalian tissues. We observed striking enrichment of bromine (79Br, 81Br) in basement membranes of normal human and mouse kidneys. In peroxidasin knockout mice, bromine enrichment of basement membranes of kidneys was reduced by ∼85%. Proteomic studies revealed bromination of tyrosine-1485 in the NC1 domain of α2 collagen IV from kidneys of wild-type mice; the same tyrosine was brominated in collagen IV from human kidney. Bromination of tyrosine-1485 was reduced by >90% in kidneys of peroxidasin knockout mice. Thus, in addition to promoting sulfilimine cross-links in collagen IV, peroxidasin can also brominate a bystander tyrosine. Also, the fact that bromine enrichment is largely confined to basement membranes implies that peroxidasin activity is largely restricted to basement membranes in mammalian tissues.


Subject(s)
Basement Membrane/metabolism , Bromine/metabolism , Extracellular Matrix Proteins/metabolism , Peroxidase/metabolism , Animals , Biopsy , Bromates/metabolism , Bromides , Cells, Cultured , Collagen Type IV/metabolism , Humans , Hydrogen Peroxide/metabolism , Imines/metabolism , Kidney/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteomics , Peroxidasin
13.
Proc Natl Acad Sci U S A ; 117(19): 10476-10483, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32354992

ABSTRACT

Cholesterol-laden macrophage foam cells are a hallmark of atherosclerosis. For that reason, cholesterol metabolism in macrophages has attracted considerable scrutiny, particularly the mechanisms by which macrophages unload surplus cholesterol (a process referred to as "cholesterol efflux"). Many studies of cholesterol efflux in macrophages have focused on the role of ABC transporters in moving cholesterol onto high-density lipoproteins (HDLs), but other mechanisms for cholesterol efflux likely exist. We hypothesized that macrophages have the capacity to unload cholesterol directly onto adjacent cells. To test this hypothesis, we used methyl-ß-cyclodextrin (MßCD) to load mouse peritoneal macrophages with [13C]cholesterol. We then plated the macrophages (in the absence of serum or HDL) onto smooth muscle cells (SMCs) that had been metabolically labeled with [15N]choline. After incubating the cells overnight in the absence of HDL or serum, we visualized 13C and 15N distribution by nanoscale secondary ion mass spectrometry (NanoSIMS). We observed substantial 13C enrichment in SMCs that were adjacent to [13C]cholesterol-loaded macrophages-including in cytosolic lipid droplets of SMCs. In follow-up studies, we depleted "accessible cholesterol" from the plasma membrane of [13C]cholesterol-loaded macrophages with MßCD before plating the macrophages onto the SMCs. After an overnight incubation, we again observed substantial 13C enrichment in the SMCs adjacent to macrophages. Thus, macrophages transfer cholesterol to adjacent cells in the absence of serum or HDL. We suspect that macrophages within tissues transfer cholesterol to adjacent cells, thereby contributing to the ability to unload surplus cholesterol.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Cholesterol/metabolism , Macrophages/metabolism , ATP Binding Cassette Transporter 1/physiology , ATP-Binding Cassette Transporters/metabolism , Animals , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Biological Transport , Foam Cells/metabolism , Lipid Metabolism , Lipoproteins, HDL/metabolism , Macrophages/physiology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Serum/metabolism , beta-Cyclodextrins/metabolism
14.
Proc Natl Acad Sci U S A ; 116(51): 25870-25879, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31796586

ABSTRACT

Deficiencies in either lamin B1 or lamin B2 cause both defective migration of cortical neurons in the developing brain and reduced neuronal survival. The neuronal migration abnormality is explained by a weakened nuclear lamina that interferes with nucleokinesis, a nuclear translocation process required for neuronal migration. In contrast, the explanation for impaired neuronal survival is poorly understood. We hypothesized that the forces imparted on the nucleus during neuronal migration result in nuclear membrane (NM) ruptures, causing interspersion of nuclear and cytoplasmic contents-and ultimately cell death. To test this hypothesis, we bred Lmnb1-deficient mice that express a nuclear-localized fluorescent Cre reporter. Migrating neurons within the cortical plate of E18.5 Lmnb1-deficient embryos exhibited NM ruptures, evident by the escape of the nuclear-localized reporter into the cytoplasm and NM discontinuities by electron microscopy. The NM ruptures were accompanied by DNA damage and cell death. The NM ruptures were not observed in nonmigrating cells within the ventricular zone. NM ruptures, DNA damage, and cell death were also observed in cultured Lmnb1-/- and Lmnb2-/- neurons as they migrated away from neurospheres. To test whether mechanical forces on the cell nucleus are relevant to NM ruptures in migrating neurons, we examined cultured Lmnb1-/- neurons when exposed to external constrictive forces (migration into a field of tightly spaced silicon pillars). As the cells entered the field of pillars, there were frequent NM ruptures, accompanied by DNA damage and cell death.


Subject(s)
Cell Death/physiology , Cell Movement/physiology , Lamin Type B/metabolism , Neurons/metabolism , Nuclear Envelope/metabolism , Nuclear Lamina/metabolism , Animals , Cell Line , Cell Survival , Cytoplasm/metabolism , DNA Damage , Gene Expression Regulation , Lamin Type B/genetics , Mice , Mice, Knockout , Neurons/cytology , Nuclear Lamina/genetics
15.
Elife ; 82019 09 05.
Article in English | MEDLINE | ID: mdl-31486771

ABSTRACT

Cultured mouse peritoneal macrophages release large numbers of ~30-nm cholesterol-rich particles. Here, we show that those particles represent fragments of the plasma membrane that are pulled away and left behind during the projection and retraction of filopodia and lamellipodia. Consistent with this finding, the particles are enriched in proteins found in focal adhesions, which attach macrophages to the substrate. The release of particles is abolished by blocking cell movement (either by depolymerizing actin with latrunculin A or by inhibiting myosin II with blebbistatin). Confocal microscopy and NanoSIMS imaging studies revealed that the plasma membrane-derived particles are enriched in 'accessible cholesterol' (a mobile pool of cholesterol detectable with the modified cytolysin ALO-D4) but not in sphingolipid-sequestered cholesterol [a pool detectable with ostreolysin A (OlyA)]. The discovery that macrophages release cholesterol-rich particles during cellular locomotion is likely relevant to cholesterol efflux and could contribute to extracellular cholesterol deposition in atherosclerotic plaques.


Subject(s)
Cell Membrane/metabolism , Cell Movement , Cell-Derived Microparticles/chemistry , Cell-Derived Microparticles/metabolism , Cholesterol/analysis , Macrophages, Peritoneal/metabolism , Pseudopodia/metabolism , Animals , Cells, Cultured , Mice , Proteins/analysis
16.
PLoS Pathog ; 15(6): e1007848, 2019 06.
Article in English | MEDLINE | ID: mdl-31181121

ABSTRACT

Streptococcus agalactiae (Group B Streptococcus, GBS) normally colonizes healthy adults but can cause invasive disease, such as meningitis, in the newborn. To gain access to the central nervous system, GBS must interact with and penetrate brain or meningeal blood vessels; however, the exact mechanisms are still being elucidated. Here, we investigate the contribution of BspC, an antigen I/II family adhesin, to the pathogenesis of GBS meningitis. Disruption of the bspC gene reduced GBS adherence to human cerebral microvascular endothelial cells (hCMEC), while heterologous expression of BspC in non-adherent Lactococcus lactis conferred bacterial attachment. In a murine model of hematogenous meningitis, mice infected with ΔbspC mutants exhibited lower mortality as well as decreased brain bacterial counts and inflammatory infiltrate compared to mice infected with WT GBS strains. Further, BspC was both necessary and sufficient to induce neutrophil chemokine expression. We determined that BspC interacts with the host cytoskeleton component vimentin and confirmed this interaction using a bacterial two-hybrid assay, microscale thermophoresis, immunofluorescent staining, and imaging flow cytometry. Vimentin null mice were protected from WT GBS infection and also exhibited less inflammatory cytokine production in brain tissue. These results suggest that BspC and the vimentin interaction is critical for the pathogenesis of GBS meningitis.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Brain/metabolism , Meningitis, Bacterial/metabolism , Streptococcal Infections/metabolism , Streptococcus agalactiae/metabolism , Vimentin/metabolism , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Brain/blood supply , Brain/microbiology , Brain/pathology , Endothelium, Vascular , HeLa Cells , Humans , Male , Meningitis, Bacterial/genetics , Meningitis, Bacterial/pathology , Mice , Mice, Mutant Strains , Sheep , Streptococcal Infections/genetics , Streptococcal Infections/pathology , Streptococcus agalactiae/genetics , Streptococcus agalactiae/pathogenicity , Vimentin/genetics
17.
Elife ; 82019 06 06.
Article in English | MEDLINE | ID: mdl-31169500

ABSTRACT

GPIHBP1, a GPI-anchored protein of capillary endothelial cells, binds lipoprotein lipase (LPL) within the subendothelial spaces and shuttles it to the capillary lumen. GPIHBP1-bound LPL is essential for the margination of triglyceride-rich lipoproteins (TRLs) along capillaries, allowing the lipolytic processing of TRLs to proceed. In peripheral tissues, the intravascular processing of TRLs by the GPIHBP1-LPL complex is crucial for the generation of lipid nutrients for adjacent parenchymal cells. GPIHBP1 is absent from the capillaries of the brain, which uses glucose for fuel; however, GPIHBP1 is expressed in the capillaries of mouse and human gliomas. Importantly, the GPIHBP1 in glioma capillaries captures locally produced LPL. We use NanoSIMS imaging to show that TRLs marginate along glioma capillaries and that there is uptake of TRL-derived lipid nutrients by surrounding glioma cells. Thus, GPIHBP1 expression in gliomas facilitates TRL processing and provides a source of lipid nutrients for glioma cells.


Subject(s)
Glioma/metabolism , Lipoproteins/metabolism , Receptors, Lipoprotein/metabolism , Animals , Brain/blood supply , Brain/pathology , Capillaries/metabolism , Carbon Isotopes/metabolism , Endothelial Cells/metabolism , Fatty Acids/metabolism , Glioma/blood supply , Glioma/pathology , Glioma/ultrastructure , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Humans , Lipoprotein Lipase/metabolism , Mice, Inbred C57BL , Triglycerides/metabolism
18.
Cell ; 175(2): 514-529.e20, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30220461

ABSTRACT

The mechanisms underlying sterol transport in mammalian cells are poorly understood. In particular, how cholesterol internalized from HDL is made available to the cell for storage or modification is unknown. Here, we describe three ER-resident proteins (Aster-A, -B, -C) that bind cholesterol and facilitate its removal from the plasma membrane. The crystal structure of the central domain of Aster-A broadly resembles the sterol-binding fold of mammalian StARD proteins, but sequence differences in the Aster pocket result in a distinct mode of ligand binding. The Aster N-terminal GRAM domain binds phosphatidylserine and mediates Aster recruitment to plasma membrane-ER contact sites in response to cholesterol accumulation in the plasma membrane. Mice lacking Aster-B are deficient in adrenal cholesterol ester storage and steroidogenesis because of an inability to transport cholesterol from SR-BI to the ER. These findings identify a nonvesicular pathway for plasma membrane to ER sterol trafficking in mammals.


Subject(s)
Cholesterol, HDL/metabolism , Membrane Proteins/physiology , Membrane Proteins/ultrastructure , 3T3 Cells , Animals , Biological Transport/physiology , CD36 Antigens/metabolism , CHO Cells , Carrier Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cell Membrane/physiology , Cholesterol/metabolism , Cricetulus , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/physiology , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Membranes/metabolism , Sequence Alignment , Sterols/metabolism
19.
Biochem Biophys Res Commun ; 504(4): 899-902, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30224066

ABSTRACT

Heterogeneity in the metabolic properties of adipocytes in white adipose tissue has been well documented. We sought to investigate metabolic heterogeneity in adipocytes of brown adipose tissue (BAT), focusing on heterogeneity in nutrient uptake. To explore the possibility of metabolic heterogeneity in brown adipocytes, we used nanoscale secondary ion mass spectrometry (NanoSIMS) to quantify uptake of lipids in adipocytes interscapular BAT and perivascular adipose tissue (PVAT) after an intravenous injection of triglyceride-rich lipoproteins (TRLs) containing [2H]triglycerides (2H-TRLs). The uptake of deuterated lipids into brown adipocytes was quantified by NanoSIMS. We also examined 13C enrichment in brown adipocytes after administering [13C]glucose or 13C-labeled mixed fatty acids by gastric gavage. The uptake of 2H-TRLs-derived lipids into brown adipocytes was heterogeneous, with 2H enrichment in adjacent adipocytes varying by more than fourfold. We also observed substantial heterogeneity in 13C enrichment in adjacent brown adipocytes after administering [13C]glucose or [13C]fatty acids by gastric gavage. The uptake of nutrients by adjacent brown adipocytes within a single depot is variable, suggesting that there is heterogeneity in the metabolic properties of brown adipocytes.


Subject(s)
Adipocytes, Brown/metabolism , Nutrients/pharmacokinetics , Spectrometry, Mass, Secondary Ion/methods , Animals , Carbon Isotopes/analysis , Fatty Acids/pharmacokinetics , Glucose/pharmacokinetics , Lipids/pharmacokinetics , Lipoproteins/administration & dosage , Lipoproteins/pharmacokinetics , Mice, Inbred C57BL , Mice, Mutant Strains , Receptors, Lipoprotein/genetics
20.
Proc Natl Acad Sci U S A ; 115(40): 10100-10105, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30224463

ABSTRACT

The nuclear lamina, an intermediate filament meshwork lining the inner nuclear membrane, is formed by the nuclear lamins (lamins A, C, B1, and B2). Defects or deficiencies in individual nuclear lamin proteins have been reported to elicit nuclear blebs (protrusions or outpouchings of the nuclear envelope) and increase susceptibility for nuclear membrane ruptures. It is unclear, however, how a complete absence of nuclear lamins would affect nuclear envelope morphology and nuclear membrane integrity (i.e., whether nuclear membrane blebs or protrusions would occur and, if not, whether cells would be susceptible to nuclear membrane ruptures). To address these issues, we generated mouse embryonic fibroblasts (MEFs) lacking all nuclear lamins. The nuclear lamin-deficient MEFs had irregular nuclear shapes but no nuclear blebs or protrusions. Despite a virtual absence of nuclear blebs, MEFs lacking nuclear lamins had frequent, prolonged, and occasionally nonhealing nuclear membrane ruptures. By transmission electron microscopy, the inner nuclear membrane in nuclear lamin-deficient MEFs have a "wavy" appearance, and there were discrete discontinuities in the inner and outer nuclear membranes. Nuclear membrane ruptures were accompanied by a large increase in DNA damage, as judged by γ-H2AX foci. Mechanical stress increased both nuclear membrane ruptures and DNA damage, whereas minimizing transmission of cytoskeletal forces to the nucleus had the opposite effects.


Subject(s)
DNA Damage , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Lamins/deficiency , Nuclear Envelope/metabolism , Stress, Mechanical , Animals , Embryo, Mammalian/ultrastructure , Fibroblasts/ultrastructure , Mice , Mice, Knockout , Nuclear Envelope/genetics , Nuclear Envelope/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL