Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Nat Struct Mol Biol ; 31(1): 82-91, 2024 Jan.
Article En | MEDLINE | ID: mdl-38177670

The NLR family caspase activation and recruitment domain-containing 4 (NLRC4) inflammasome is a critical cytosolic innate immune machine formed upon the direct sensing of bacterial infection and in response to cell stress during sterile chronic inflammation. Despite its major role in instigating the subsequent host immune response, a more complete understanding of the molecular events in the formation of the NLRC4 inflammasome in humans is lacking. Here we identify Bacillus thailandensis type III secretion system needle protein (Needle) as a potent trigger of the human NLR family apoptosis inhibitory protein (NAIP)/NLRC4 inflammasome complex formation and determine its structural features by cryogenic electron microscopy. We also provide a detailed understanding of how type III secretion system pathogen components are sensed by human NAIP to form a cascade of NLRC4 protomer through a critical lasso-like motif, a 'lock-key' activation model and large structural rearrangement, ultimately forming the full human NLRC4 inflammasome. These results shed light on key regulatory mechanisms specific to the NLRC4 inflammasome assembly, and the innate immune modalities of pathogen sensing in humans.


Inflammasomes , Type III Secretion Systems , Humans , Macrophages/metabolism , Macrophages/microbiology , Flagellin/metabolism , Calcium-Binding Proteins/metabolism , CARD Signaling Adaptor Proteins , Neuronal Apoptosis-Inhibitory Protein/metabolism
2.
Comput Struct Biotechnol J ; 20: 4952-4968, 2022.
Article En | MEDLINE | ID: mdl-36147680

Antibodies are fundamental effectors of humoral immunity, and have become a highly successful class of therapeutics. There is increasing evidence that antibodies utilize transient homotypic interactions to enhance function, and elucidation of such interactions can provide insights into their biology and new opportunities for their optimization as drugs. Yet the transitory nature of weak interactions makes them difficult to investigate. Capitalizing on their rich structural data and high conservation, we have characterized all the ways that antibody fragment antigen-binding (Fab) regions interact crystallographically. This approach led to the discovery of previously unrealized interfaces between antibodies. While diverse interactions exist, ß-sheet dimers and variable-constant elbow dimers are recurrent motifs. Disulfide engineering enabled interactions to be trapped and investigated structurally and functionally, providing experimental validation of the interfaces and illustrating their potential for optimization. This work provides first insight into previously undiscovered oligomeric interactions between antibodies, and enables new opportunities for their biotherapeutic optimization.

3.
Structure ; 29(6): 564-571.e3, 2021 06 03.
Article En | MEDLINE | ID: mdl-33513362

Immunoglobulins (Ig) A and M are the only human antibodies that form oligomers and undergo transcytosis to mucosal secretions via the polymeric Ig receptor (pIgR). When complexed with the J-chain (JC) and the secretory component (SC) of pIgR, secretory IgA and IgM (sIgA and sIgM) play critical roles in host-pathogen defense. Recently, we determined the structure of sIgA-Fc which elucidated the mechanism of polymeric IgA assembly and revealed an extensive binding interface between IgA-Fc, JC, and SC. Despite low sequence identity shared with IgA-Fc, IgM-Fc also undergoes JC-mediated assembly and binds pIgR. Here, we report the structure of sIgM-Fc and carryout a systematic comparison to sIgA-Fc. Our structural analysis reveals a remarkably conserved mechanism of JC-templated oligomerization and SC recognition of both IgM and IgA through a highly conserved network of interactions. These studies reveal the structurally conserved features of sIgM and sIgA required for function in mucosal immunity.


Immunoglobulin A, Secretory/chemistry , Immunoglobulin J-Chains/metabolism , Immunoglobulin M/chemistry , Secretory Component/metabolism , Cell Line , Humans , Immunoglobulin A, Secretory/metabolism , Immunoglobulin M/metabolism , Models, Molecular , Protein Conformation , Structural Homology, Protein , Transcytosis
4.
Mol Cell ; 80(4): 736-743.e4, 2020 11 19.
Article En | MEDLINE | ID: mdl-33098764

The phosphoinositide PI(3,5)P2, generated exclusively by the PIKfyve lipid kinase complex, is key for lysosomal biology. Here, we explore how PI(3,5)P2 levels within cells are regulated. We find the PIKfyve complex comprises five copies of the scaffolding protein Vac14 and one copy each of the lipid kinase PIKfyve, generating PI(3,5)P2 from PI3P and the lipid phosphatase Fig4, reversing the reaction. Fig4 is active as a lipid phosphatase in the ternary complex, whereas PIKfyve within the complex cannot access membrane-incorporated phosphoinositides due to steric constraints. We find further that the phosphoinositide-directed activities of both PIKfyve and Fig4 are regulated by protein-directed activities within the complex. PIKfyve autophosphorylation represses its lipid kinase activity and stimulates Fig4 lipid phosphatase activity. Further, Fig4 is also a protein phosphatase acting on PIKfyve to stimulate its lipid kinase activity, explaining why catalytically active Fig4 is required for maximal PI(3,5)P2 production by PIKfyve in vivo.


Cell Membrane/metabolism , Flavoproteins/metabolism , Homeostasis , Lysosomes/metabolism , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/metabolism , Flavoproteins/chemistry , Flavoproteins/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Protein Binding , Protein Conformation , Protein Transport
5.
Science ; 367(6481): 1008-1014, 2020 02 28.
Article En | MEDLINE | ID: mdl-32029686

Secretory immunoglobulin A (sIgA) represents the immune system's first line of defense against mucosal pathogens. IgAs are transported across the epithelium, as dimers and higher-order polymers, by the polymeric immunoglobulin receptor (pIgR). Upon reaching the luminal side, sIgAs mediate host protection and pathogen neutralization. In recent years, an increasing amount of attention has been given to IgA as a novel therapeutic antibody. However, despite extensive studies, sIgA structures have remained elusive. Here, we determine the atomic resolution structures of dimeric, tetrameric, and pentameric IgA-Fc linked by the joining chain (JC) and in complex with the secretory component of the pIgR. We suggest a mechanism in which the JC templates IgA oligomerization and imparts asymmetry for pIgR binding and transcytosis. This framework will inform the design of future IgA-based therapeutics.


Immunoglobulin A, Secretory/chemistry , Immunoglobulin Fc Fragments/chemistry , Protein Multimerization , Humans , Immunoglobulin J-Chains/chemistry , Receptors, Polymeric Immunoglobulin/chemistry , Transcytosis
6.
J Cell Biol ; 218(6): 1787-1798, 2019 06 03.
Article En | MEDLINE | ID: mdl-30952800

During macroautophagic stress, autophagosomes can be produced continuously and in high numbers. Many different organelles have been reported as potential donor membranes for this sustained autophagosome growth, but specific machinery to support the delivery of lipid to the growing autophagosome membrane has remained unknown. Here we show that the autophagy protein, ATG2, without a clear function since its discovery over 20 yr ago, is in fact a lipid-transfer protein likely operating at the ER-autophagosome interface. ATG2A can bind tens of glycerophospholipids at once and transfers lipids robustly in vitro. An N-terminal fragment of ATG2A that supports lipid transfer in vitro is both necessary and fully sufficient to rescue blocked autophagosome biogenesis in ATG2A/ATG2B KO cells, implying that regulation of lipid homeostasis is the major autophagy-dependent activity of this protein and, by extension, that protein-mediated lipid transfer across contact sites is a principal contributor to autophagosome formation.


Autophagosomes/physiology , Autophagy-Related Proteins/metabolism , Autophagy , Endoplasmic Reticulum/metabolism , Lipids/physiology , Vesicular Transport Proteins/metabolism , Autophagy-Related Proteins/antagonists & inhibitors , Autophagy-Related Proteins/genetics , Biological Transport , CRISPR-Cas Systems , HEK293 Cells , Humans , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics
7.
J Cell Biol ; 217(10): 3625-3639, 2018 10 01.
Article En | MEDLINE | ID: mdl-30093493

Mutations in the human VPS13 genes are responsible for neurodevelopmental and neurodegenerative disorders including chorea acanthocytosis (VPS13A) and Parkinson's disease (VPS13C). The mechanisms of these diseases are unknown. Genetic studies in yeast hinted that Vps13 may have a role in lipid exchange between organelles. In this study, we show that the N-terminal portion of VPS13 is tubular, with a hydrophobic cavity that can solubilize and transport glycerolipids between membranes. We also show that human VPS13A and VPS13C bind to the ER, tethering it to mitochondria (VPS13A), to late endosome/lysosomes (VPS13C), and to lipid droplets (both VPS13A and VPS13C). These findings identify VPS13 as a lipid transporter between the ER and other organelles, implicating defects in membrane lipid homeostasis in neurological disorders resulting from their mutations. Sequence and secondary structure similarity between the N-terminal portions of Vps13 and other proteins such as the autophagy protein ATG2 suggest lipid transport roles for these proteins as well.


Endoplasmic Reticulum/metabolism , Proteins/metabolism , Vesicular Transport Proteins/metabolism , Animals , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , COS Cells , Chlorocebus aethiops , Endoplasmic Reticulum/genetics , Endosomes/genetics , Endosomes/metabolism , HeLa Cells , Humans , Lipid Droplets/metabolism , Lysosomes/genetics , Lysosomes/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Protein Domains , Protein Structure, Secondary , Proteins/genetics , Saccharomyces cerevisiae , Vesicular Transport Proteins/genetics
8.
J Cell Sci ; 127(Pt 19): 4308-21, 2014 Oct 01.
Article En | MEDLINE | ID: mdl-25086068

Kindlins are essential FERM-domain-containing focal adhesion (FA) proteins required for proper integrin activation and signaling. Despite the widely accepted importance of each of the three mammalian kindlins in cell adhesion, the molecular basis for their function has yet to be fully elucidated, and the functional differences between isoforms have generally not been examined. Here, we report functional differences between kindlin-2 and -3 (also known as FERMT2 and FERMT3, respectively); GFP-tagged kindlin-2 localizes to FAs whereas kindlin-3 does not, and kindlin-2, but not kindlin-3, can rescue α5ß1 integrin activation defects in kindlin-2-knockdown fibroblasts. Using chimeric kindlins, we show that the relatively uncharacterized kindlin-2 F2 subdomain drives FA targeting and integrin activation. We find that the integrin-linked kinase (ILK)-PINCH-parvin complex binds strongly to the kindlin-2 F2 subdomain but poorly to that of kindlin-3. Using a point-mutated kindlin-2, we establish that efficient kindlin-2-mediated integrin activation and FA targeting require binding to the ILK complex. Thus, ILK-complex binding is crucial for normal kindlin-2 function and differential ILK binding contributes to kindlin isoform specificity.


Focal Adhesions/metabolism , Integrins/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetulus , HEK293 Cells , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , Protein Isoforms , Signal Transduction
9.
J Cardiovasc Pharmacol ; 63(2): 132-143, 2014 Feb.
Article En | MEDLINE | ID: mdl-24508946

FTY720, an analogue of sphingosine-1-phosphate, is cardioprotective during acute injury. Whether long-term FTY720 affords cardioprotection is unknown. Here, we report the effects of oral FTY720 on ischemia/reperfusion injury and in hypomorphic apoE mice deficient in SR-BI receptor expression (ApoeR61(h/h)/SRB1(-/- mice), a model of diet-induced coronary atherosclerosis and heart failure. We added FTY720 (0.3 mg·kg(-1)·d(-1)) to the drinking water of C57BL/6J mice. After ex vivo cardiac ischemia/reperfusion injury, these mice had significantly improved left ventricular (LV) developed pressure and reduced infarct size compared with controls. Subsequently, ApoeR61(h/h)/SRB1(-/-) mice fed a high-fat diet for 4 weeks were treated or not with oral FTY720 (0.05 mg·kg(-1)·d(-1)). This sharply reduced mortality (P < 0.02) and resulted in better LV function and less LV remodeling compared with controls without reducing hypercholesterolemia and atherosclerosis. Oral FTY720 reduced the number of blood lymphocytes and increased the percentage of CD4+Foxp3+ regulatory T cells (Tregs) in the circulation, spleen, and lymph nodes. FTY720-treated mice exhibited increased TGF-ß and reduced IFN-γ expression in the heart. Also, CD4 expression was increased and strongly correlated with molecules involved in natural Treg activity, such as TGF-ß and GITR. Our data suggest that long-term FTY720 treatment enhances LV function and increases longevity in mice with heart failure. These benefits resulted not from atheroprotection but from systemic immunosuppression and a moderate reduction of inflammation in the heart.


Apolipoproteins E/genetics , Coronary Artery Disease/drug therapy , Myocardial Infarction/drug therapy , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Animals , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/pharmacology , Coronary Artery Disease/physiopathology , Diet, High-Fat/adverse effects , Disease Models, Animal , Fingolimod Hydrochloride , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacology , Inflammation/drug therapy , Inflammation/etiology , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology , Propylene Glycols/administration & dosage , Sphingosine/administration & dosage , Sphingosine/pharmacology , Survival Rate , T-Lymphocytes, Regulatory/metabolism , Time Factors , Transforming Growth Factor beta/metabolism , Ventricular Function, Left/drug effects
10.
Am J Pathol ; 183(6): 1981-1992, 2013 Dec.
Article En | MEDLINE | ID: mdl-24113453

Diabetic patients are known to be more susceptible to atherosclerosis and its associated cardiovascular complications. However, the effects of hyperglycemia on atherosclerosis regression remain unclear. We hypothesized that hyperglycemia impairs atherosclerosis regression by modulating the biological function of lesional macrophages. HypoE (Apoe(h/h)Mx1-Cre) mice express low levels of apolipoprotein E (apoE) and develop atherosclerosis when fed a high-fat diet. Atherosclerosis regression occurs in these mice upon plasma lipid lowering induced by a change in diet and the restoration of apoE expression. We examined the morphological characteristics of regressed lesions and assessed the biological function of lesional macrophages isolated with laser-capture microdissection in euglycemic and hyperglycemic HypoE mice. Hyperglycemia induced by streptozotocin treatment impaired lesion size reduction (36% versus 14%) and lipid loss (38% versus 26%) after the reversal of hyperlipidemia. However, decreases in lesional macrophage content and remodeling in both groups of mice were similar. Gene expression analysis revealed that hyperglycemia impaired cholesterol transport by modulating ATP-binding cassette A1, ATP-binding cassette G1, scavenger receptor class B family member (CD36), scavenger receptor class B1, and wound healing pathways in lesional macrophages during atherosclerosis regression. Hyperglycemia impairs both reduction in size and loss of lipids from atherosclerotic lesions upon plasma lipid lowering without significantly affecting the remodeling of the vascular wall.


Apolipoproteins E , Atherosclerosis , Gene Expression Regulation/genetics , Hyperglycemia , Lipids/blood , Macrophages , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/blood , Atherosclerosis/complications , Atherosclerosis/genetics , Atherosclerosis/pathology , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Female , Hyperglycemia/blood , Hyperglycemia/complications , Hyperglycemia/genetics , Hyperglycemia/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Transgenic
11.
Arterioscler Thromb Vasc Biol ; 33(8): 1759-67, 2013 Aug.
Article En | MEDLINE | ID: mdl-23788760

OBJECTIVE: To study atherosclerosis regression in mice after plasma lipid reduction to moderately elevated apolipoprotein B (apoB)-lipoprotein levels. APPROACH AND RESULTS: Chow-fed hypomorphic Apoe mice deficient in low-density lipoprotein receptor expression (Apoe(h/h)Ldlr(-/-)Mx1-cre mice) develop hyperlipidemia and atherosclerosis. These mice were studied before and after inducible cre-mediated Apoe gene repair. By 1 week, induced mice displayed a 2-fold reduction in plasma cholesterol and triglyceride levels and a decrease in the non-high-density lipoprotein:high-density lipoprotein-cholesterol ratio from 87%:13% to 60%:40%. This halted atherosclerotic lesion growth and promoted macrophage loss and accumulation of thick collagen fibers for up to 8 weeks. Concomitantly, blood Ly-6C(high) monocytes were decreased by 2-fold but lesional macrophage apoptosis was unchanged. The expression of several genes involved in extracellular matrix remodeling and cell migration was changed in lesional macrophages 1 week after Apoe gene repair. However, mRNA levels of numerous genes involved in cholesterol efflux and inflammation were not significantly changed at this time point. CONCLUSIONS: Restoring apoE expression in Apoe(h/h)Ldlr(-/-)Mx1-cre mice resulted in lesion stabilization in the context of a human-like ratio of non-high-density lipoprotein:high-density lipoprotein-cholesterol. Our data suggest that macrophage loss derived in part from reduced blood Ly-6C(high) monocytes levels and genetic reprogramming of lesional macrophages.


Apolipoproteins E/genetics , Genetic Therapy/methods , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/therapy , Receptors, LDL/genetics , Animals , Apolipoprotein B-100 , Apolipoproteins B/blood , Apolipoproteins B/genetics , Apolipoproteins E/blood , Apolipoproteins E/deficiency , Apoptosis/physiology , Cholesterol/blood , Cholesterol, HDL/blood , Disease Models, Animal , Disease Progression , Gene Expression Regulation/physiology , Humans , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Hyperlipidemias/therapy , Macrophages/cytology , Mice , Mice, Knockout , Monocytes/cytology , Plaque, Atherosclerotic/metabolism , Receptors, LDL/deficiency , Triglycerides/blood
12.
PLoS One ; 7(5): e35816, 2012.
Article En | MEDLINE | ID: mdl-22606237

BACKGROUND: Apolipoprotein (apo) E is best known for its ability to lower plasma cholesterol and protect against atherosclerosis. Although the liver is the major source of plasma apoE, extra-hepatic sources of apoE, including from macrophages, account for up to 10% of plasma apoE levels. This study examined the contribution of macrophage-derived apoE expression levels in diet-induced hyperlipidemia and atherosclerosis. METHODOLOGY/PRINCIPAL FINDINGS: Hypomorphic apoE (Apoe(h/h)) mice expressing wildtype mouse apoE at ∼2-5% of physiological levels in all tissues were derived by gene targeting in embryonic stem cells. Cre-mediated gene repair of the Apoe(h/h) allele in Apoe(h/h)LysM-Cre mice raised apoE expression levels by 26 fold in freshly isolated peritoneal macrophages, restoring it to 37% of levels seen in wildtype mice. Chow-fed Apoe(h/h)LysM-Cre and Apoe(h/h) mice displayed similar plasma apoE and cholesterol levels (55.53±2.90 mg/dl versus 62.70±2.77 mg/dl, n = 12). When fed a high-cholesterol diet (HCD) for 16 weeks, Apoe(h/h)LysM-Cre mice displayed a 3-fold increase in plasma apoE and a concomitant 32% decrease in plasma cholesterol when compared to Apoe(h/h) mice (602.20±22.30 mg/dl versus 888.80±24.99 mg/dl, n = 7). On HCD, Apoe(h/h)LysM-Cre mice showed increased apoE immunoreactivity in lesional macrophages and liver-associated Kupffer cells but not hepatocytes. In addition, Apoe(h/h)LysM-Cre mice developed 35% less atherosclerotic lesions in the aortic root than Apoe(h/h) mice (167×10(3)±16×10(3) µm(2) versus 259×10(3)±56×10(3) µm(2), n = 7). This difference in atherosclerosis lesions size was proportional to the observed reduction in plasma cholesterol. CONCLUSIONS/SIGNIFICANCE: Macrophage-derived apoE raises plasma apoE levels in response to diet-induced hyperlipidemia and by such reduces atherosclerosis proportionally to the extent to which it lowers plasma cholesterol levels.


Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/prevention & control , Hyperlipidemias/prevention & control , Macrophages/metabolism , Animals , Apolipoproteins E/blood , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/therapy , Cholesterol/blood , Diet, Atherogenic , Gene Expression , Genetic Therapy , Hyperlipidemias/blood , Hyperlipidemias/etiology , Hyperlipidemias/therapy , Kupffer Cells/metabolism , Mice , Mice, Knockout , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Arterioscler Thromb Vasc Biol ; 32(5): 1116-23, 2012 May.
Article En | MEDLINE | ID: mdl-22441102

OBJECTIVE: Apolipoprotein (apo) E4 is an established risk factor for atherosclerosis, but the structural components underlying this association remain unclear. ApoE4 is characterized by 2 biophysical properties: domain interaction and molten globule state. Substituting Arg-61 for Thr-61 in mouse apoE introduces domain interaction without molten globule state, allowing us to delineate potential proatherogenic effects of domain interaction in vivo. METHODS AND RESULTS: We studied atherosclerosis susceptibility of hypomorphic Apoe mice expressing either Thr-61 or Arg-61 apoE (ApoeT(h/h) or ApoeR(h/h)mice). On a chow diet, both mouse models were normolipidemic with similar levels of plasma apoE and lipoproteins. However, on a high-cholesterol diet, ApoeR(h/h) mice displayed increased levels of total plasma cholesterol and very-low-density lipoprotein as well as larger atherosclerotic plaques in the aortic root, arch, and descending aorta compared with ApoeT(h/h) mice. In addition, evidence of cellular dysfunction was identified in peritoneal ApoeR(h/h) macrophages which released lower amounts of apoE in culture medium and displayed increased expression of major histocompatibility complex class II molecules. CONCLUSIONS: These data indicate that domain interaction mediates proatherogenic effects of apoE4 in part by modulating lipoprotein metabolism and macrophage biology. Pharmaceutical targeting of domain interaction could lead to new treatments for atherosclerosis in apoE4 individuals.


Apolipoprotein E4/genetics , Atherosclerosis/genetics , DNA/genetics , Gene Expression Regulation , Genetic Predisposition to Disease , Animals , Apolipoprotein E4/biosynthesis , Atherosclerosis/etiology , Atherosclerosis/metabolism , Diet, Atherogenic/adverse effects , Disease Models, Animal , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
Arterioscler Thromb Vasc Biol ; 32(2): 264-72, 2012 Feb.
Article En | MEDLINE | ID: mdl-22053073

OBJECTIVE: We investigated atheroprotective properties of apolipoprotein (apo) E beyond its ability to lower plasma cholesterol. We hypothesized that apoE reduces atherosclerosis by decreasing lipid accumulation in circulating monocytes and the inflammatory state of monocytes and the vascular endothelium. METHODS AND RESULTS: We developed mice with spontaneous hyperlipidemia with and without plasma apoE. Hypomorphic apoE mice deficient in low-density lipoprotein receptor (Apoe(h/h)Ldlr(-/-)) were compared to Apoe(-/-)Ldlr(-/-) mice. Despite 4-fold more plasma apoE than WT mice, Apoe(h/h)Ldlr(-/-) mice displayed similar plasma cholesterol as Apoe(-/-) Ldlr(-/-) mice but developed 4-fold less atherosclerotic lesions by 5 months of age. The aortic arch of Apoe(h/h)Ldlr(-/-) mice showed decreased endothelial expression of ICAM-1, PECAM-1, and JAM-A. In addition, Apoe(h/h)Ldlr(-/-) mice had less circulating leukocytes and proinflammatory Ly6C(high) monocytes. These monocytes had decreased neutral lipid content and reduced surface expression of ICAM-1, VLA-4, and L-Selectin. Apoe(h/h)Ldlr(-/-) mice displayed increased levels of apoA1-rich HDL that were potent in promoting cellular cholesterol efflux. CONCLUSIONS: Our findings suggest that apoE reduces atherosclerosis in the setting of hyperlipidemia by increasing plasma apoA1-HDL that likely contribute to reduce intracellular lipid accumulation and thereby the activation of circulating leukocytes and the vascular endothelium.


Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Endothelium, Vascular/metabolism , Inflammation Mediators/metabolism , Lipid Metabolism , Monocytes/metabolism , Animals , Apolipoproteins E/deficiency , Cell Adhesion Molecules/metabolism , Cholesterol/metabolism , Disease Models, Animal , Integrin alpha4beta1/metabolism , Intercellular Adhesion Molecule-1/metabolism , L-Selectin/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, Cell Surface/metabolism , Receptors, LDL/deficiency , Receptors, LDL/metabolism
...