Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.846
Filter
1.
Cell ; 187(2): 360-374.e19, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38176410

ABSTRACT

The very-low-density lipoprotein receptor (VLDLR) comprises eight LDLR type A (LA) domains and supports entry of distantly related alphaviruses, including Eastern equine encephalitis virus (EEEV) and Semliki Forest virus (SFV). Here, by resolving multiple cryo-electron microscopy structures of EEEV-VLDLR complexes and performing mutagenesis and functional studies, we show that EEEV uses multiple sites (E1/E2 cleft and E2 A domain) to engage more than one LA domain simultaneously. However, no single LA domain is necessary or sufficient to support efficient EEEV infection. Whereas all EEEV strains show conservation of two VLDLR-binding sites, the EEEV PE-6 strain and a few other EEE complex members feature a single amino acid substitution that enables binding of LA domains to an additional site on the E2 B domain. These structural and functional analyses informed the design of a minimal VLDLR decoy receptor that neutralizes EEEV infection and protects mice from lethal challenge.


Subject(s)
Cryoelectron Microscopy , Encephalitis Virus, Eastern Equine , Encephalomyelitis, Equine , Receptors, LDL , Animals , Mice , Alphavirus/physiology , Encephalitis Virus, Eastern Equine/physiology , Encephalitis Virus, Eastern Equine/ultrastructure , Encephalomyelitis, Equine/metabolism , Horses , Protein Binding , Receptors, LDL/ultrastructure
2.
Cell ; 187(20): 5665-5678.e18, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39208797

ABSTRACT

In mammalian cells, two phosphatidylserine (PS) synthases drive PS synthesis. Gain-of-function mutations in the Ptdss1 gene lead to heightened PS production, causing Lenz-Majewski syndrome (LMS). Recently, pharmacological inhibition of PSS1 has been shown to suppress tumorigenesis. Here, we report the cryo-EM structures of wild-type human PSS1 (PSS1WT), the LMS-causing Pro269Ser mutant (PSS1P269S), and PSS1WT in complex with its inhibitor DS55980254. PSS1 contains 10 transmembrane helices (TMs), with TMs 4-8 forming a catalytic core in the luminal leaflet. These structures revealed a working mechanism of PSS1 akin to the postulated mechanisms of the membrane-bound O-acyltransferase family. Additionally, we showed that both PS and DS55980254 can allosterically inhibit PSS1 and that inhibition by DS55980254 activates the SREBP pathways, thus enhancing the expression of LDL receptors and increasing cellular LDL uptake. This work uncovers a mechanism of mammalian PS synthesis and suggests that selective PSS1 inhibitors have the potential to lower blood cholesterol levels.


Subject(s)
Phosphatidylserines , Humans , Phosphatidylserines/metabolism , Cryoelectron Microscopy , Lipoproteins, LDL/metabolism , Receptors, LDL/metabolism , CDPdiacylglycerol-Serine O-Phosphatidyltransferase/metabolism , CDPdiacylglycerol-Serine O-Phosphatidyltransferase/genetics , Animals , HEK293 Cells
3.
Cell ; 186(10): 2208-2218.e15, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37098345

ABSTRACT

Semliki Forest virus (SFV) is an alphavirus that uses the very-low-density lipoprotein receptor (VLDLR) as a receptor during infection of its vertebrate hosts and insect vectors. Herein, we used cryoelectron microscopy to study the structure of SFV in complex with VLDLR. We found that VLDLR binds multiple E1-DIII sites of SFV through its membrane-distal LDLR class A (LA) repeats. Among the LA repeats of the VLDLR, LA3 has the best binding affinity to SFV. The high-resolution structure shows that LA3 binds SFV E1-DIII through a small surface area of 378 Å2, with the main interactions at the interface involving salt bridges. Compared with the binding of single LA3s, consecutive LA repeats around LA3 promote synergistic binding to SFV, during which the LAs undergo a rotation, allowing simultaneous key interactions at multiple E1-DIII sites on the virion and enabling the binding of VLDLRs from divergent host species to SFV.


Subject(s)
Receptors, LDL , Semliki forest virus , Alphavirus/metabolism , Cryoelectron Microscopy , Semliki forest virus/metabolism , Semliki forest virus/ultrastructure , Receptors, LDL/metabolism , Receptors, LDL/ultrastructure , Receptors, Virus/metabolism , Receptors, Virus/ultrastructure
4.
Cell ; 179(6): 1276-1288.e14, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31778654

ABSTRACT

Although human genetic studies have implicated many susceptible genes associated with plasma lipid levels, their physiological and molecular functions are not fully characterized. Here we demonstrate that orphan G protein-coupled receptor 146 (GPR146) promotes activity of hepatic sterol regulatory element binding protein 2 (SREBP2) through activation of the extracellular signal-regulated kinase (ERK) signaling pathway, thereby regulating hepatic very low-density lipoprotein (VLDL) secretion, and subsequently circulating low-density lipoprotein cholesterol (LDL-C) and triglycerides (TG) levels. Remarkably, GPR146 deficiency reduces plasma cholesterol levels substantially in both wild-type and LDL receptor (LDLR)-deficient mice. Finally, aortic atherosclerotic lesions are reduced by 90% and 70%, respectively, in male and female LDLR-deficient mice upon GPR146 depletion. Taken together, these findings outline a regulatory role for the GPR146/ERK axis in systemic cholesterol metabolism and suggest that GPR146 inhibition could be an effective strategy to reduce plasma cholesterol levels and atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Hypercholesterolemia/metabolism , Receptors, G-Protein-Coupled/deficiency , Animals , Atherosclerosis/blood , Base Sequence , Cholesterol/blood , Dependovirus/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fasting , Female , Hepatocytes/metabolism , Humans , Hypercholesterolemia/blood , Lipoproteins, VLDL/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, LDL/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 2/metabolism , Triglycerides/blood , Up-Regulation
5.
Annu Rev Biochem ; 87: 783-807, 2018 06 20.
Article in English | MEDLINE | ID: mdl-28841344

ABSTRACT

Scap is a polytopic membrane protein that functions as a molecular machine to control the cholesterol content of membranes in mammalian cells. In the 21 years since our laboratory discovered Scap, we have learned how it binds sterol regulatory element-binding proteins (SREBPs) and transports them from the endoplasmic reticulum (ER) to the Golgi for proteolytic processing. Proteolysis releases the SREBP transcription factor domains, which enter the nucleus to promote cholesterol synthesis and uptake. When cholesterol in ER membranes exceeds a threshold, the sterol binds to Scap, triggering several conformational changes that prevent the Scap-SREBP complex from leaving the ER. As a result, SREBPs are no longer processed, cholesterol synthesis and uptake are repressed, and cholesterol homeostasis is restored. This review focuses on the four domains of Scap that undergo concerted conformational changes in response to cholesterol binding. The data provide a molecular mechanism for the control of lipids in cell membranes.


Subject(s)
Cholesterol/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Animals , Homeostasis , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Biological , Models, Molecular , Protein Conformation , Protein Transport , Proteolysis , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Proteins/metabolism
6.
Cell ; 172(1-2): 162-175.e14, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29328911

ABSTRACT

Long-term epigenetic reprogramming of innate immune cells in response to microbes, also termed "trained immunity," causes prolonged altered cellular functionality to protect from secondary infections. Here, we investigated whether sterile triggers of inflammation induce trained immunity and thereby influence innate immune responses. Western diet (WD) feeding of Ldlr-/- mice induced systemic inflammation, which was undetectable in serum soon after mice were shifted back to a chow diet (CD). In contrast, myeloid cell responses toward innate stimuli remained broadly augmented. WD-induced transcriptomic and epigenomic reprogramming of myeloid progenitor cells led to increased proliferation and enhanced innate immune responses. Quantitative trait locus (QTL) analysis in human monocytes trained with oxidized low-density lipoprotein (oxLDL) and stimulated with lipopolysaccharide (LPS) suggested inflammasome-mediated trained immunity. Consistently, Nlrp3-/-/Ldlr-/- mice lacked WD-induced systemic inflammation, myeloid progenitor proliferation, and reprogramming. Hence, NLRP3 mediates trained immunity following WD and could thereby mediate the potentially deleterious effects of trained immunity in inflammatory diseases.


Subject(s)
Cellular Reprogramming , Diet, Western , Epigenesis, Genetic , Immunity, Innate , Immunologic Memory , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Adult , Aged , Animals , Cells, Cultured , Female , Humans , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myeloid Cells/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Quantitative Trait Loci , Receptors, LDL/genetics
7.
Immunity ; 55(7): 1185-1199.e8, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35738281

ABSTRACT

Lipoprotein disorder is a common feature of chronic pancreatitis (CP); however, the relationship between lipoprotein disorder and pancreatic fibrotic environment is unclear. Here, we investigated the occurrence and mechanism of pancreatic stellate cell (PSC) activation by lipoprotein metabolites and the subsequent regulation of type 2 immune responses, as well as the driving force of fibrotic aggressiveness in CP. Single-cell RNA sequencing revealed the heterogeneity of PSCs and identified very-low-density lipoprotein receptor (VLDLR)+ PSCs that were characterized by a higher lipid metabolism. VLDLR promoted intracellular lipid accumulation, followed by interleukin-33 (IL-33) expression and release in PSCs. PSC-derived IL-33 strongly induced pancreatic group 2 innate lymphoid cells (ILC2s) to trigger a type 2 immune response accompanied by the activation of PSCs, eventually leading to fibrosis during pancreatitis. Our findings indicate that VLDLR-enhanced lipoprotein metabolism in PSCs promotes pancreatic fibrosis and highlight a dominant role of IL-33 in this pro-fibrotic cascade.


Subject(s)
Pancreatic Stellate Cells , Pancreatitis, Chronic , Receptors, LDL/metabolism , Cells, Cultured , Fibrosis , Humans , Immunity, Innate , Interleukin-33/metabolism , Lipid Metabolism , Lipoproteins, VLDL/metabolism , Lymphocytes/metabolism , Pancreas/pathology , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/pathology
8.
Cell ; 161(1): 161-172, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25815993

ABSTRACT

One-fourth of all deaths in industrialized countries result from coronary heart disease. A century of research has revealed the essential causative agent: cholesterol-carrying low-density lipoprotein (LDL). LDL is controlled by specific receptors (LDLRs) in liver that remove it from blood. Mutations that eliminate LDLRs raise LDL and cause heart attacks in childhood, whereas mutations that raise LDLRs reduce LDL and diminish heart attacks. If we are to eliminate coronary disease, lowering LDL should be the primary goal. Effective means to achieve this goal are currently available. The key questions are: who to treat, when to treat, and how long to treat.


Subject(s)
Cholesterol/metabolism , Coronary Vessels/pathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Plaque, Atherosclerotic/drug therapy , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Coronary Vessels/metabolism , Dietary Fats/metabolism , Humans , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Receptors, LDL/metabolism
9.
Nature ; 634(8033): 447-456, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39232165

ABSTRACT

Systemic immune responses caused by chronic hypercholesterolaemia contribute to atherosclerosis initiation, progression and complications1. However, individuals often change their dietary habits over time2, and the effects of an alternating high-fat diet (HFD) on atherosclerosis remain unclear. Here, to address this relevant issue, we developed a protocol using atherosclerosis-prone mice to compare an alternating versus continuous HFD while maintaining similar overall exposure periods. We found that an alternating HFD accelerated atherosclerosis in Ldlr-/- and Apoe-/- mice compared with a continuous HFD. This pro-atherogenic effect of the alternating HFD was also observed in Apoe-/-Rag2-/- mice lacking T, B and natural killer T cells, ruling out the role of the adaptive immune system in the observed phenotype. Discontinuing the HFD in the alternating HFD group downregulated RUNX13, promoting inflammatory signalling in bone marrow myeloid progenitors. After re-exposure to an HFD, these cells produced IL-1ß, leading to emergency myelopoiesis and increased neutrophil levels in blood. Neutrophils infiltrated plaques and released neutrophil extracellular traps, exacerbating atherosclerosis. Specific depletion of neutrophils or inhibition of IL-1ß pathways abolished emergency myelopoiesis and reversed the pro-atherogenic effects of the alternating HFD. This study highlights the role of IL-1ß-dependent neutrophil progenitor reprogramming in accelerated atherosclerosis induced by alternating HFD.


Subject(s)
Apolipoproteins E , Atherosclerosis , Cellular Reprogramming , Diet, High-Fat , Interleukin-1beta , Myelopoiesis , Neutrophils , Receptors, LDL , Animals , Diet, High-Fat/adverse effects , Atherosclerosis/pathology , Atherosclerosis/metabolism , Atherosclerosis/immunology , Mice , Neutrophils/immunology , Neutrophils/metabolism , Interleukin-1beta/metabolism , Male , Receptors, LDL/deficiency , Receptors, LDL/metabolism , Receptors, LDL/genetics , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Female , Plaque, Atherosclerotic/pathology , Signal Transduction , Mice, Inbred C57BL , Disease Models, Animal , DNA-Binding Proteins
10.
Nature ; 632(8025): 614-621, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39048821

ABSTRACT

Western equine encephalitis virus (WEEV) is an arthropod-borne virus (arbovirus) that frequently caused major outbreaks of encephalitis in humans and horses in the early twentieth century, but the frequency of outbreaks has since decreased markedly, and strains of this alphavirus isolated in the past two decades are less virulent in mammals than strains isolated in the 1930s and 1940s1-3. The basis for this phenotypic change in WEEV strains and coincident decrease in epizootic activity (known as viral submergence3) is unclear, as is the possibility of re-emergence of highly virulent strains. Here we identify protocadherin 10 (PCDH10) as a cellular receptor for WEEV. We show that multiple highly virulent ancestral WEEV strains isolated in the 1930s and 1940s, in addition to binding human PCDH10, could also bind very low-density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2), which are recognized by another encephalitic alphavirus as receptors4. However, whereas most of the WEEV strains that we examined bind to PCDH10, a contemporary strain has lost the ability to recognize mammalian PCDH10 while retaining the ability to bind avian receptors, suggesting WEEV adaptation to a main reservoir host during enzootic circulation. PCDH10 supports WEEV E2-E1 glycoprotein-mediated infection of primary mouse cortical neurons, and administration of a soluble form of PCDH10 protects mice from lethal WEEV challenge. Our results have implications for the development of medical countermeasures and for risk assessment for re-emerging WEEV strains.


Subject(s)
Encephalitis Virus, Western Equine , Host Specificity , Protocadherins , Receptors, Virus , Animals , Female , Humans , Male , Mice , Birds/metabolism , Birds/virology , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/virology , Encephalitis Virus, Western Equine/classification , Encephalitis Virus, Western Equine/metabolism , Encephalitis Virus, Western Equine/pathogenicity , Encephalomyelitis, Equine/epidemiology , Encephalomyelitis, Equine/virology , LDL-Receptor Related Proteins/metabolism , Neurons/metabolism , Neurons/virology , Phenotype , Protocadherins/metabolism , Receptors, LDL/metabolism , Receptors, LDL/genetics , Receptors, Virus/metabolism , Viral Envelope Proteins/metabolism , Viral Zoonoses/epidemiology , Viral Zoonoses/virology
11.
Nature ; 602(7897): 475-480, 2022 02.
Article in English | MEDLINE | ID: mdl-34929721

ABSTRACT

Alphaviruses, like many other arthropod-borne viruses, infect vertebrate species and insect vectors separated by hundreds of millions of years of evolutionary history. Entry into evolutionarily divergent host cells can be accomplished by recognition of different cellular receptors in different species, or by binding to receptors that are highly conserved across species. Although multiple alphavirus receptors have been described1-3, most are not shared among vertebrate and invertebrate hosts. Here we identify the very low-density lipoprotein receptor (VLDLR) as a receptor for the prototypic alphavirus Semliki forest virus. We show that the E2 and E1 glycoproteins (E2-E1) of Semliki forest virus, eastern equine encephalitis virus and Sindbis virus interact with the ligand-binding domains (LBDs) of VLDLR and apolipoprotein E receptor 2 (ApoER2), two closely related receptors. Ectopic expression of either protein facilitates cellular attachment, and internalization of virus-like particles, a VLDLR LBD-Fc fusion protein or a ligand-binding antagonist block Semliki forest virus E2-E1-mediated infection of human and mouse neurons in culture. The administration of a VLDLR LBD-Fc fusion protein has protective activity against rapidly fatal Semliki forest virus infection in mouse neonates. We further show that invertebrate receptor orthologues from mosquitoes and worms can serve as functional alphavirus receptors. We propose that the ability of some alphaviruses to infect a wide range of hosts is a result of their engagement of evolutionarily conserved lipoprotein receptors and contributes to their pathogenesis.


Subject(s)
Mosquito Vectors , Semliki forest virus , Animals , LDL-Receptor Related Proteins , Ligands , Mice , Receptors, LDL , Semliki forest virus/metabolism , Sindbis Virus/physiology
12.
PLoS Biol ; 22(8): e3002739, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39137238

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) poses a significant threat due to its tendency to evade early detection, frequent metastasis, and the subsequent challenges in devising effective treatments. Processes that govern epithelial-mesenchymal transition (EMT) in PDAC hold promise for advancing novel therapeutic strategies. SAMD1 (SAM domain-containing protein 1) is a CpG island-binding protein that plays a pivotal role in the repression of its target genes. Here, we revealed that SAMD1 acts as a repressor of genes associated with EMT. Upon deletion of SAMD1 in PDAC cells, we observed significantly increased migration rates. SAMD1 exerts its effects by binding to specific genomic targets, including CDH2, encoding N-cadherin, which emerged as a driver of enhanced migration upon SAMD1 knockout. Furthermore, we discovered the FBXO11-containing E3 ubiquitin ligase complex as an interactor and negative regulator of SAMD1, which inhibits SAMD1 chromatin-binding genome-wide. High FBXO11 expression in PDAC is associated with poor prognosis and increased expression of EMT-related genes, underlining an antagonistic relationship between SAMD1 and FBXO11. In summary, our findings provide insights into the regulation of EMT-related genes in PDAC, shedding light on the intricate role of SAMD1 and its interplay with FBXO11 in this cancer type.


Subject(s)
Carcinoma, Pancreatic Ductal , Epithelial-Mesenchymal Transition , F-Box Proteins , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms , Receptors, LDL , Animals , Humans , Cadherins/metabolism , Cadherins/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , F-Box Proteins/metabolism , F-Box Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , Receptors, LDL/genetics , Receptors, LDL/metabolism
13.
Cell ; 151(1): 138-52, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-23021221

ABSTRACT

Inflammation and macrophage foam cells are characteristic features of atherosclerotic lesions, but the mechanisms linking cholesterol accumulation to inflammation and LXR-dependent response pathways are poorly understood. To investigate this relationship, we utilized lipidomic and transcriptomic methods to evaluate the effect of diet and LDL receptor genotype on macrophage foam cell formation within the peritoneal cavities of mice. Foam cell formation was associated with significant changes in hundreds of lipid species and unexpected suppression, rather than activation, of inflammatory gene expression. We provide evidence that regulated accumulation of desmosterol underlies many of the homeostatic responses, including activation of LXR target genes, inhibition of SREBP target genes, selective reprogramming of fatty acid metabolism, and suppression of inflammatory-response genes, observed in macrophage foam cells. These observations suggest that macrophage activation in atherosclerotic lesions results from extrinsic, proinflammatory signals generated within the artery wall that suppress homeostatic and anti-inflammatory functions of desmosterol.


Subject(s)
Atherosclerosis/immunology , Cholesterol/biosynthesis , Desmosterol/metabolism , Foam Cells/metabolism , Lipid Metabolism , Transcriptome , Animals , Atherosclerosis/metabolism , Cholesterol/analogs & derivatives , Cholesterol/metabolism , Fatty Acids/metabolism , Foam Cells/immunology , Gene Knockdown Techniques , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, LDL/genetics , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Proteins/metabolism
14.
Nature ; 598(7882): 677-681, 2021 10.
Article in English | MEDLINE | ID: mdl-34646021

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is an enveloped RNA virus that causes encephalitis and potentially mortality in infected humans and equines1. At present, no vaccines or drugs are available that prevent or cure diseases caused by VEEV. Low-density lipoprotein receptor class A domain-containing 3 (LDLRAD3) was recently identified as a receptor for the entry of VEEV into host cells2. Here we present the cryo-electron microscopy structure of the LDLRAD3 extracellular domain 1 (LDLRAD3-D1) in complex with VEEV virus-like particles at a resolution of 3.0 Å. LDLRAD3-D1 has a cork-like structure and is inserted into clefts formed between adjacent VEEV E2-E1 heterodimers in the viral-surface trimer spikes through hydrophobic and polar contacts. Mutagenesis studies of LDLRAD3-D1 identified residues that are involved in the key interactions with VEEV. Of note, some of the LDLRAD3-D1 mutants showed a significantly increased binding affinity for VEEV, suggesting that LDLRAD3-D1 may serve as a potential scaffold for the development of inhibitors of VEEV entry. Our structures provide insights into alphavirus assembly and the binding of receptors to alphaviruses, which may guide the development of therapeutic countermeasures against alphaviruses.


Subject(s)
Encephalitis Virus, Venezuelan Equine/chemistry , Receptors, LDL/chemistry , Receptors, Virus/chemistry , Cryoelectron Microscopy , Humans , Models, Molecular , Protein Structure, Secondary , Virus Internalization
15.
Nature ; 598(7882): 672-676, 2021 10.
Article in English | MEDLINE | ID: mdl-34646020

ABSTRACT

LDLRAD3 is a recently defined attachment and entry receptor for Venezuelan equine encephalitis virus (VEEV)1, a New World alphavirus that causes severe neurological disease in humans. Here we present near-atomic-resolution cryo-electron microscopy reconstructions of VEEV virus-like particles alone and in a complex with the ectodomains of LDLRAD3. Domain 1 of LDLRAD3 is a low-density lipoprotein receptor type-A module that binds to VEEV by wedging into a cleft created by two adjacent E2-E1 heterodimers in one trimeric spike, and engages domains A and B of E2 and the fusion loop in E1. Atomic modelling of this interface is supported by mutagenesis and anti-VEEV antibody binding competition assays. Notably, VEEV engages LDLRAD3 in a manner that is similar to the way that arthritogenic alphaviruses bind to the structurally unrelated MXRA8 receptor, but with a much smaller interface. These studies further elucidate the structural basis of alphavirus-receptor interactions, which could inform the development of therapies to mitigate infection and disease against multiple members of this family.


Subject(s)
Encephalitis Virus, Venezuelan Equine/chemistry , Receptors, LDL/chemistry , Receptors, Virus/chemistry , Amino Acid Sequence , Animals , Cell Line , Cryoelectron Microscopy , Humans , Mice , Models, Molecular , Protein Structure, Secondary , Sequence Alignment , Virus Internalization
16.
Nature ; 589(7841): 287-292, 2021 01.
Article in English | MEDLINE | ID: mdl-33268892

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of mortality in the world, with most CVD-related deaths resulting from myocardial infarction or stroke. The main underlying cause of thrombosis and cardiovascular events is atherosclerosis, an inflammatory disease that can remain asymptomatic for long periods. There is an urgent need for therapeutic and diagnostic options in this area. Atherosclerotic plaques contain autoantibodies1,2, and there is a connection between atherosclerosis and autoimmunity3. However, the immunogenic trigger and the effects of the autoantibody response during atherosclerosis are not well understood3-5. Here we performed high-throughput single-cell analysis of the atherosclerosis-associated antibody repertoire. Antibody gene sequencing of more than 1,700 B cells from atherogenic Ldlr-/- and control mice identified 56 antibodies expressed by in-vivo-expanded clones of B lymphocytes in the context of atherosclerosis. One-third of the expanded antibodies were reactive against atherosclerotic plaques, indicating that various antigens in the lesion can trigger antibody responses. Deep proteomics analysis identified ALDH4A1, a mitochondrial dehydrogenase involved in proline metabolism, as a target antigen of one of these autoantibodies, A12. ALDH4A1 distribution is altered during atherosclerosis, and circulating ALDH4A1 is increased in mice and humans with atherosclerosis, supporting the potential use of ALDH4A1 as a disease biomarker. Infusion of A12 antibodies into Ldlr-/- mice delayed plaque formation and reduced circulating free cholesterol and LDL, suggesting that anti-ALDH4A1 antibodies can protect against atherosclerosis progression and might have therapeutic potential in CVD.


Subject(s)
1-Pyrroline-5-Carboxylate Dehydrogenase/immunology , Atherosclerosis/immunology , Atherosclerosis/prevention & control , Autoantibodies/immunology , Autoantigens/immunology , 1-Pyrroline-5-Carboxylate Dehydrogenase/blood , Animals , Atherosclerosis/blood , Atherosclerosis/diagnosis , Autoantibodies/blood , Autoantibodies/genetics , Autoantigens/blood , Autoimmunity , B-Lymphocytes/immunology , Biomarkers/blood , Cholesterol/blood , Diet, High-Fat , Disease Models, Animal , Disease Progression , Humans , Lipoproteins, LDL/blood , Male , Mice , Mice, Inbred C57BL , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/prevention & control , Proteomics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Single-Cell Analysis
17.
Circ Res ; 135(2): 335-349, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38828596

ABSTRACT

BACKGROUND: Individuals with type 1 diabetes (T1D) generally have normal or even higher HDL (high-density lipoprotein)-cholesterol levels than people without diabetes yet are at increased risk for atherosclerotic cardiovascular disease (CVD). Human HDL is a complex mixture of particles that can vary in cholesterol content by >2-fold. To investigate if specific HDL subspecies contribute to the increased atherosclerosis associated with T1D, we created mouse models of T1D that exhibit human-like HDL subspecies. We also measured HDL subspecies and their association with incident CVD in a cohort of people with T1D. METHODS: We generated LDL receptor-deficient (Ldlr-/-) mouse models of T1D expressing human APOA1 (apolipoprotein A1). Ldlr-/-APOA1Tg mice exhibited the main human HDL subspecies. We also generated Ldlr-/-APOA1Tg T1D mice expressing CETP (cholesteryl ester transfer protein), which had lower concentrations of large HDL subspecies versus mice not expressing CETP. HDL particle concentrations and sizes and proteins involved in lipoprotein metabolism were measured by calibrated differential ion mobility analysis and targeted mass spectrometry in the mouse models of T1D and in a cohort of individuals with T1D. Endothelial transcytosis was analyzed by total internal reflection fluorescence microscopy. RESULTS: Diabetic Ldlr-/-APOA1Tg mice were severely hyperglycemic and hyperlipidemic and had markedly elevated plasma APOB levels versus nondiabetic littermates but were protected from the proatherogenic effects of diabetes. Diabetic Ldlr-/-APOA1Tg mice expressing CETP lost the atheroprotective effect and had increased lesion necrotic core areas and APOB accumulation, despite having lower plasma APOB levels. The detrimental effects of low concentrations of larger HDL particles in diabetic mice expressing CETP were not explained by reduced cholesterol efflux. Instead, large HDL was more effective than small HDL in preventing endothelial transcytosis of LDL mediated by scavenger receptor class B type 1. Finally, in humans with T1D, increased concentrations of larger HDL particles relative to APOB100 negatively predicted incident CVD independently of HDL-cholesterol levels. CONCLUSIONS: Our results suggest that the balance between APOB lipoproteins and the larger HDL subspecies contributes to atherosclerosis progression and incident CVD in the setting of T1D and that larger HDLs exert atheroprotective effects on endothelial cells rather than by promoting macrophage cholesterol efflux.


Subject(s)
Apolipoprotein A-I , Atherosclerosis , Diabetes Mellitus, Type 1 , Receptors, LDL , Adult , Animals , Female , Humans , Male , Mice , Middle Aged , Apolipoprotein A-I/blood , Apolipoprotein A-I/metabolism , Apolipoprotein B-100/metabolism , Apolipoprotein B-100/genetics , Apolipoprotein B-100/blood , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/blood , Atherosclerosis/pathology , Cholesterol Ester Transfer Proteins/genetics , Cholesterol Ester Transfer Proteins/metabolism , Cholesterol Ester Transfer Proteins/blood , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/blood , Disease Models, Animal , Lipoproteins, HDL/blood , Lipoproteins, HDL/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, LDL/genetics , Receptors, LDL/deficiency , Receptors, LDL/metabolism
18.
Circ Res ; 134(11): 1405-1423, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38639096

ABSTRACT

BACKGROUND: While our understanding of the single-cell gene expression patterns underlying the transformation of vascular cell types during the progression of atherosclerosis is rapidly improving, the clinical and pathophysiological relevance of these changes remains poorly understood. METHODS: Single-cell RNA sequencing data generated with SmartSeq2 (≈8000 genes/cell) in 16 588 single cells isolated during atherosclerosis progression in Ldlr-/-Apob100/100 mice with human-like plasma lipoproteins and from humans with asymptomatic and symptomatic carotid plaques was clustered into multiple subtypes. For clinical and pathophysiological context, the advanced-stage and symptomatic subtype clusters were integrated with 135 tissue-specific (atherosclerotic aortic wall, mammary artery, liver, skeletal muscle, and visceral and subcutaneous, fat) gene-regulatory networks (GRNs) inferred from 600 coronary artery disease patients in the STARNET (Stockholm-Tartu Atherosclerosis Reverse Network Engineering Task) study. RESULTS: Advanced stages of atherosclerosis progression and symptomatic carotid plaques were largely characterized by 3 smooth muscle cells (SMCs), and 3 macrophage subtype clusters with extracellular matrix organization/osteogenic (SMC), and M1-type proinflammatory/Trem2-high lipid-associated (macrophage) phenotypes. Integrative analysis of these 6 clusters with STARNET revealed significant enrichments of 3 arterial wall GRNs: GRN33 (macrophage), GRN39 (SMC), and GRN122 (macrophage) with major contributions to coronary artery disease heritability and strong associations with clinical scores of coronary atherosclerosis severity. The presence and pathophysiological relevance of GRN39 were verified in 5 independent RNAseq data sets obtained from the human coronary and aortic artery, and primary SMCs and by targeting its top-key drivers, FRZB and ALCAM in cultured human coronary artery SMCs. CONCLUSIONS: By identifying and integrating the most gene-rich single-cell subclusters of atherosclerosis to date with a coronary artery disease framework of GRNs, GRN39 was identified and independently validated as being critical for the transformation of contractile SMCs into an osteogenic phenotype promoting advanced, symptomatic atherosclerosis.


Subject(s)
Atherosclerosis , Gene Regulatory Networks , Single-Cell Analysis , Humans , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Mice , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Male , Plaque, Atherosclerotic , Disease Progression , Female , Macrophages/metabolism , Macrophages/pathology , Mice, Knockout , Receptors, LDL/genetics , Receptors, LDL/metabolism , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology
19.
EMBO Rep ; 25(3): 951-970, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38287192

ABSTRACT

The exquisite specificity of antibodies can be harnessed to effect targeted degradation of membrane proteins. Here, we demonstrate targeted protein removal utilising a protein degradation domain derived from the endogenous human protein Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9). Recombinant antibodies genetically fused to this domain drive the degradation of membrane proteins that undergo constitutive internalisation and recycling, including the transferrin receptor and the human cytomegalovirus latency-associated protein US28. We term this approach PACTAC (PCSK9-Antibody Clearance-Targeting Chimeras).


Subject(s)
Proprotein Convertase 9 , Serine Endopeptidases , Humans , Proprotein Convertase 9/metabolism , Proprotein Convertases/metabolism , Membrane Proteins , Receptors, LDL/metabolism
20.
Nature ; 588(7837): 308-314, 2020 12.
Article in English | MEDLINE | ID: mdl-33208938

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a neurotropic alphavirus transmitted by mosquitoes that causes encephalitis and death in humans1. VEEV is a biodefence concern because of its potential for aerosol spread and the current lack of sufficient countermeasures. The host factors that are required for VEEV entry and infection remain poorly characterized. Here, using a genome-wide CRISPR-Cas9-based screen, we identify low-density lipoprotein receptor class A domain-containing 3 (LDLRAD3)-a highly conserved yet poorly characterized member of the scavenger receptor superfamily-as a receptor for VEEV. Gene editing of mouse Ldlrad3 or human LDLRAD3 results in markedly reduced viral infection of neuronal cells, which is restored upon complementation with LDLRAD3. LDLRAD3 binds directly to VEEV particles and enhances virus attachment and internalization into host cells. Genetic studies indicate that domain 1 of LDLRAD3 (LDLRAD3(D1)) is necessary and sufficient to support infection by VEEV, and both anti-LDLRAD3 antibodies and an LDLRAD3(D1)-Fc fusion protein block VEEV infection in cell culture. The pathogenesis of VEEV infection is abrogated in mice with deletions in Ldlrad3, and administration of LDLRAD3(D1)-Fc abolishes disease caused by several subtypes of VEEV, including highly virulent strains. The development of a decoy-receptor fusion protein suggests a strategy for the prevention of severe VEEV infection and associated disease in humans.


Subject(s)
Encephalitis Virus, Venezuelan Equine/metabolism , Receptors, LDL/metabolism , Receptors, Virus/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Line , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/metabolism , Encephalomyelitis, Venezuelan Equine/prevention & control , Encephalomyelitis, Venezuelan Equine/virology , Female , Genetic Complementation Test , Humans , Male , Mice , Mice, Inbred C57BL , Protein Binding , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, Virus/genetics , Virus Attachment , Virus Internalization
SELECTION OF CITATIONS
SEARCH DETAIL