Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
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
2.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Article in English | MEDLINE | ID: mdl-34161290

ABSTRACT

Defects or deficiencies in nuclear lamins cause pathology in many cell types, and recent studies have implicated nuclear membrane (NM) ruptures as a cause of cell toxicity. We previously observed NM ruptures and progressive cell death in the developing brain of lamin B1-deficient mouse embryos. We also observed frequent NM ruptures and DNA damage in nuclear lamin-deficient fibroblasts. Factors modulating susceptibility to NM ruptures remain unclear, but we noted low levels of LAP2ß, a chromatin-binding inner NM protein, in fibroblasts with NM ruptures. Here, we explored the apparent link between LAP2ß and NM ruptures in nuclear lamin-deficient neurons and fibroblasts, and we tested whether manipulating LAP2ß expression levels would alter NM rupture frequency. In cortical plate neurons of lamin B1-deficient embryos, we observed a strong correlation between low LAP2ß levels and NM ruptures. We also found low LAP2ß levels and frequent NM ruptures in neurons of cultured Lmnb1-/- neurospheres. Reducing LAP2ß expression in Lmnb1-/- neurons with an siRNA markedly increased the NM rupture frequency (without affecting NM rupture duration), whereas increased LAP2ß expression eliminated NM ruptures and reduced DNA damage. Consistent findings were observed in nuclear lamin-deficient fibroblasts. Reduced LAP2ß expression increased NM ruptures, whereas increased LAP2ß expression virtually abolished NM ruptures. Increased LAP2ß expression nearly abolished NM ruptures in cells subjected to mechanical stress (an intervention that increases NM ruptures). Our studies showed that increasing LAP2ß expression bolsters NM integrity in nuclear lamin-deficient cells and markedly reduces NM rupture frequency.


Subject(s)
DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , Lamin Type B/deficiency , Membrane Proteins/metabolism , Neurons/metabolism , Nuclear Envelope/metabolism , Animals , Cell Death , Cell Differentiation , Cerebral Cortex/pathology , DNA Damage , Embryo, Mammalian/metabolism , Lamin Type A/deficiency , Lamin Type A/metabolism , Lamin Type B/metabolism , Mice, Knockout , Organ Specificity
3.
J Lipid Res ; 61(3): 413-421, 2020 03.
Article in English | MEDLINE | ID: mdl-31941672

ABSTRACT

Zinc metallopeptidase STE24 (ZMPSTE24) is essential for the conversion of farnesyl-prelamin A to mature lamin A, a key component of the nuclear lamina. In the absence of ZMPSTE24, farnesyl-prelamin A accumulates in the nucleus and exerts toxicity, causing a variety of disease phenotypes. By ∼4 months of age, both male and female Zmpste24-/- mice manifest a near-complete loss of adipose tissue, but it has never been clear whether this phenotype is a direct consequence of farnesyl-prelamin A toxicity in adipocytes. To address this question, we generated a conditional knockout Zmpste24 allele and used it to create adipocyte-specific Zmpste24-knockout mice. To boost farnesyl-prelamin A levels, we bred in the "prelamin A-only" Lmna allele. Gene expression, immunoblotting, and immunohistochemistry experiments revealed that adipose tissue in these mice had decreased Zmpste24 expression along with strikingly increased accumulation of prelamin A. In male mice, Zmpste24 deficiency in adipocytes was accompanied by modest changes in adipose stores (an 11% decrease in body weight, a 23% decrease in body fat mass, and significantly smaller gonadal and inguinal white adipose depots). No changes in adipose stores were detected in female mice, likely because prelamin A expression in adipose tissue is lower in female mice. Zmpste24 deficiency in adipocytes did not alter the number of macrophages in adipose tissue, nor did it alter plasma levels of glucose, triglycerides, or fatty acids. We conclude that ZMPSTE24 deficiency in adipocytes, and the accompanying accumulation of farnesyl-prelamin A, reduces adipose tissue stores, but only modestly and only in male mice.


Subject(s)
Adipose Tissue/metabolism , Lamin Type A/metabolism , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Adipose Tissue/chemistry , Alleles , Animals , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Female , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Metalloendopeptidases/deficiency , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Mice, Transgenic
4.
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
5.
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
6.
Proc Natl Acad Sci U S A ; 116(13): 6319-6328, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30850549

ABSTRACT

Lipoprotein lipase (LPL), the enzyme that hydrolyzes triglycerides in plasma lipoproteins, is assumed to be active only as a homodimer. In support of this idea, several groups have reported that the size of LPL, as measured by density gradient ultracentrifugation, is ∼110 kDa, twice the size of LPL monomers (∼55 kDa). Of note, however, in those studies the LPL had been incubated with heparin, a polyanionic substance that binds and stabilizes LPL. Here we revisited the assumption that LPL is active only as a homodimer. When freshly secreted human LPL (or purified preparations of LPL) was subjected to density gradient ultracentrifugation (in the absence of heparin), LPL mass and activity peaks exhibited the size expected of monomers (near the 66-kDa albumin standard). GPIHBP1-bound LPL also exhibited the size expected for a monomer. In the presence of heparin, LPL size increased, overlapping with a 97.2-kDa standard. We also used density gradient ultracentrifugation to characterize the LPL within the high-salt and low-salt peaks from a heparin-Sepharose column. The catalytically active LPL within the high-salt peak exhibited the size of monomers, whereas most of the inactive LPL in the low-salt peak was at the bottom of the tube (in aggregates). Consistent with those findings, the LPL in the low-salt peak, but not that in the high-salt peak, was easily detectable with single mAb sandwich ELISAs, in which LPL is captured and detected with the same antibody. We conclude that catalytically active LPL can exist in a monomeric state.


Subject(s)
Lipoprotein Lipase/chemistry , Lipoprotein Lipase/isolation & purification , Animals , CHO Cells , Cattle , Centrifugation, Density Gradient/methods , Chromatography, Affinity , Chromatography, Agarose , Cricetulus , Epitopes , Heparin , Humans , Lipoprotein Lipase/blood , Receptors, Lipoprotein/blood , Receptors, Lipoprotein/chemistry , Receptors, Lipoprotein/isolation & purification , Sepharose/analogs & derivatives , Triglycerides/metabolism , Ultracentrifugation
7.
J Lipid Res ; 60(4): 869-879, 2019 04.
Article in English | MEDLINE | ID: mdl-30598475

ABSTRACT

Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1), the protein that shuttles LPL to the capillary lumen, is essential for plasma triglyceride metabolism. When GPIHBP1 is absent, LPL remains stranded within the interstitial spaces and plasma triglyceride hydrolysis is impaired, resulting in severe hypertriglyceridemia. While the functions of GPIHBP1 in intravascular lipolysis are reasonably well understood, no one has yet identified DNA sequences regulating GPIHBP1 expression. In the current studies, we identified an enhancer element located ∼3.6 kb upstream from exon 1 of mouse Gpihbp1. To examine the importance of the enhancer, we used CRISPR/Cas9 genome editing to create mice lacking the enhancer (Gpihbp1Enh/Enh). Removing the enhancer reduced Gpihbp1 expression by >90% in the liver and by ∼50% in heart and brown adipose tissue. The reduced expression of GPIHBP1 was insufficient to prevent LPL from reaching the capillary lumen, and it did not lead to hypertriglyceridemia-even when mice were fed a high-fat diet. Compound heterozygotes (Gpihbp1Enh/- mice) displayed further reductions in Gpihbp1 expression and exhibited partial mislocalization of LPL (increased amounts of LPL within the interstitial spaces of the heart), but the plasma triglyceride levels were not perturbed. The enhancer element that we identified represents the first insight into DNA sequences controlling Gpihbp1 expression.


Subject(s)
Adipose Tissue, Brown/metabolism , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/genetics , Animals , CRISPR-Cas Systems/genetics , Chromatin/genetics , Heart , Humans , Mice , Mice, Inbred Strains , Receptors, Lipoprotein/analysis , Receptors, Lipoprotein/metabolism , Sequence Analysis, DNA , Triglycerides/blood , Triglycerides/metabolism
8.
J Lipid Res ; 59(4): 706-713, 2018 04.
Article in English | MEDLINE | ID: mdl-29449313

ABSTRACT

Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1), an endothelial cell protein, binds LPL in the subendothelial spaces and transports it to the capillary lumen. In Gpihbp1-/- mice, LPL remains stranded in the subendothelial spaces, causing hypertriglyceridemia, but how Gpihbp1-/- mice respond to metabolic stress (e.g., cold exposure) has never been studied. In wild-type mice, cold exposure increases LPL-mediated processing of triglyceride-rich lipoproteins (TRLs) in brown adipose tissue (BAT), providing fuel for thermogenesis and leading to lower plasma triglyceride levels. We suspected that defective TRL processing in Gpihbp1-/- mice might impair thermogenesis and blunt the fall in plasma triglyceride levels. Indeed, Gpihbp1-/- mice exhibited cold intolerance, but the effects on plasma triglyceride levels were paradoxical. Rather than falling, the plasma triglyceride levels increased sharply (from ∼4,000 to ∼15,000 mg/dl), likely because fatty acid release by peripheral tissues drives hepatic production of TRLs that cannot be processed. We predicted that the sharp increase in plasma triglyceride levels would not occur in Gpihbp1-/-Angptl4-/- mice, where LPL activity is higher and baseline plasma triglyceride levels are lower. Indeed, the plasma triglyceride levels in Gpihbp1-/-Angptl4-/- mice fell during cold exposure. Metabolic studies revealed increased levels of TRL processing in the BAT of Gpihbp1-/-Angptl4-/- mice.


Subject(s)
Cold Temperature , Receptors, Lipoprotein/blood , Receptors, Lipoprotein/deficiency , Thermogenesis , Triglycerides/blood , Animals , Apolipoproteins B/blood , Mice , Mice, Knockout
10.
J Lipid Res ; 58(9): 1893-1902, 2017 09.
Article in English | MEDLINE | ID: mdl-28694296

ABSTRACT

apoC-III is often assumed to retard the intravascular processing of triglyceride-rich lipoproteins (TRLs) by inhibiting LPL, but that view is based largely on studies of free LPL. We now recognize that intravascular LPL is neither free nor loosely bound, but instead is tightly bound to glycosylphosphatidylinositol-anchored HDL-binding protein 1 (GPIHBP1) on endothelial cells. Here, we revisited the effects of apoC-III on LPL, focusing on apoC-III's capacity to affect the activity of GPIHBP1-bound LPL. We found that TRLs from APOC3 transgenic mice bound normally to GPIHBP1-bound LPL on cultured cells in vitro and to heart capillaries in vivo. However, the triglycerides in apoC-III-enriched TRLs were hydrolyzed more slowly by free LPL, and the inhibitory effect of apoC-III on triglyceride lipolysis was exaggerated when LPL was bound to GPIHBP1 on the surface of agarose beads. Also, recombinant apoC-III reduced triglyceride hydrolysis by free LPL only modestly, but the inhibitory effect was greater when the LPL was bound to GPIHBP1. A mutant apoC-III associated with low plasma triglyceride levels (p.A23T) displayed a reduced capacity to inhibit free and GPIHBP1-bound LPL. Our results show that apoC-III potently inhibits triglyceride hydrolysis when LPL is bound to GPIHBP1.


Subject(s)
Apolipoprotein C-III/metabolism , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/metabolism , Triglycerides/metabolism , Animals , CHO Cells , Cricetulus , Humans , Hydrolysis , Mice , Protein Binding
11.
J Lipid Res ; 58(7): 1453-1461, 2017 07.
Article in English | MEDLINE | ID: mdl-28476858

ABSTRACT

Mutation of conserved cysteines in proteins of the Ly6 family cause human disease-chylomicronemia in the case of glycosylphosphatidylinositol-anchored HDL binding protein 1 (GPIHBP1) and paroxysmal nocturnal hemoglobinuria in the case of CD59. A mutation in a conserved cysteine in CD59 prevented the protein from reaching the surface of blood cells. In contrast, mutation of conserved cysteines in human GPIHBP1 had little effect on GPIHBP1 trafficking to the surface of cultured CHO cells. The latter findings were somewhat surprising and raised questions about whether CHO cell studies accurately model the fate of mutant GPIHBP1 proteins in vivo. To explore this concern, we created mice harboring a GPIHBP1 cysteine mutation (p.C63Y). The p.C63Y mutation abolished the ability of mouse GPIHBP1 to bind LPL, resulting in severe chylomicronemia. The mutant GPIHBP1 was detectable by immunohistochemistry on the surface of endothelial cells, but the level of expression was ∼70% lower than in WT mice. The mutant GPIHBP1 protein in mouse tissues was predominantly monomeric. We conclude that mutation of a conserved cysteine in GPIHBP1 abolishes the ability of GPIHBP1 to bind LPL, resulting in mislocalization of LPL and severe chylomicronemia. The mutation reduced but did not eliminate GPIHBP1 on the surface of endothelial cells in vivo.


Subject(s)
Conserved Sequence , Cysteine , Lipoprotein Lipase/metabolism , Mutation , Receptors, Lipoprotein/chemistry , Receptors, Lipoprotein/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Female , Humans , Lipoprotein Lipase/genetics , Mice , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Lipoprotein/genetics , Triglycerides/blood
12.
Exp Dermatol ; 26(11): 1134-1136, 2017 11.
Article in English | MEDLINE | ID: mdl-28418591

ABSTRACT

Mutations in SLURP1, a secreted protein of keratinocytes, cause a palmoplantar keratoderma (PPK) known as mal de Meleda. Slurp1 deficiency in mice faithfully recapitulates the human disease, with increased keratinocyte proliferation and thickening of the epidermis on the volar surface of the paws. There has long been speculation that SLURP1 serves as a ligand for a receptor that regulates keratinocyte growth and differentiation. We were intrigued that mutations leading to increased signalling through the epidermal growth factor receptor (EGFR) cause PPK. Here, we sought to determine whether reducing EGFR signalling would ameliorate the PPK associated with SLURP1 deficiency. To address this issue, we bred Slurp1-deficient mice that were homozygous for a hypomorphic Egfr allele. The hypomorphic Egfr allele, which leads to reduced EGFR signalling in keratinocytes, did not ameliorate the PPK elicited by SLURP1 deficiency, suggesting that SLURP1 deficiency causes PPK independently (or downstream) from the EGFR pathway.


Subject(s)
Antigens, Ly/genetics , Antigens, Ly/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Keratoderma, Palmoplantar/genetics , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Alleles , Animals , Genotype , Keratoderma, Palmoplantar/pathology , Male , Mice, Knockout , Phenotype , Signal Transduction/genetics , Urokinase-Type Plasminogen Activator/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...