Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Proc Natl Acad Sci U S A ; 120(23): e2300453120, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37252960

ABSTRACT

MuSK is a receptor tyrosine kinase (RTK) that plays essential roles in the formation and maintenance of the neuromuscular junction. Distinct from most members of RTK family, MuSK activation requires not only its cognate ligand agrin but also its coreceptors LRP4. However, how agrin and LRP4 coactivate MuSK remains unclear. Here, we report the cryo-EM structure of the extracellular ternary complex of agrin/LRP4/MuSK in a stoichiometry of 1:1:1. This structure reveals that arc-shaped LRP4 simultaneously recruits both agrin and MuSK to its central cavity, thereby promoting a direct interaction between agrin and MuSK. Our cryo-EM analyses therefore uncover the assembly mechanism of agrin/LRP4/MuSK signaling complex and reveal how MuSK receptor is activated by concurrent binding of agrin and LRP4.


Subject(s)
Agrin , Receptors, Cholinergic , Receptors, Cholinergic/metabolism , Agrin/chemistry , Agrin/metabolism , LDL-Receptor Related Proteins/chemistry , Signal Transduction , Neuromuscular Junction/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
2.
Cell Mol Life Sci ; 79(2): 113, 2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35099616

ABSTRACT

Induction of bone formation by Wnt ligands is inhibited when sclerostin (Scl), an osteocyte-produced antagonist, binds to its receptors, the low-density lipoprotein receptor-related proteins 5 or 6 (LRP5/6). Recently, it was shown that enhanced inhibition is achieved by Scl binding to the co-receptor LRP4. However, it is not clear if the binding of Scl to LRP4 facilitates Scl binding to LRP5/6 or inhibits the Wnt pathway in an LRP5/6-independent manner. Here, using the yeast display system, we demonstrate that Scl exhibits a stronger binding affinity for LRP4 than for LRP6. Moreover, we found stronger Scl binding to LRP6 in the presence of LRP4. We further show that a Scl mutant (SclN93A), which tightly binds LRP4 but not LRP6, does not inhibit the Wnt pathway on its own. We demonstrate that SclN93A competes with Scl for a common binding site on LRP4 and antagonizes Scl inhibition of the Wnt signaling pathway in osteoblasts in vitro. Finally, we demonstrate that 2 weeks of bi-weekly subcutaneous injections of SclN93A fused to the fragment crystallizable (Fc) domain of immunoglobulin (SclN93AFc), which retains the antagonistic activity of the mutant, significantly increases bone formation rate and enhances trabecular volumetric bone fraction, trabecular number, and bone length in developing mice. Our data show that LRP4 serves as an anchor that facilitates Scl-LRP6 binding and that inhibition of the Wnt pathway by Scl depends on its prior binding to LRP4. We further provide evidence that compounds that inhibit Scl-LRP4 interactions offer a potential strategy to promote anabolic bone functions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , LDL-Receptor Related Proteins/metabolism , Osteogenesis/drug effects , Recombinant Proteins/pharmacology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding, Competitive/drug effects , Binding, Competitive/genetics , Cells, Cultured , Female , HEK293 Cells , Humans , LDL-Receptor Related Proteins/antagonists & inhibitors , LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/genetics , Mice , Mice, Inbred C57BL , Mutant Proteins/chemistry , Mutant Proteins/pharmacology , Osteoblasts/drug effects , Osteoblasts/physiology , Osteogenesis/genetics , Protein Binding/drug effects , Protein Binding/genetics , Protein Interaction Domains and Motifs/drug effects , Protein Interaction Domains and Motifs/genetics , RNA, Small Interfering/pharmacology , Recombinant Proteins/chemistry
3.
J Clin Neurosci ; 89: 243-248, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34119275

ABSTRACT

The sortilin-related receptor 1 (SORL1) gene has been the subject of many studies focusing on frequent polymorphisms, which is associated with increased risk for Alzheimer's Disease (AD). By whole-exome sequencing (WES), we identified two pathogenic missense mutations c.579C > G (p.F193L) and c.1397A > G (p.N466S) in SORL1. The two mutations were located in the same protein domain, and the two unrelated probands both had an onset of memory problems at less than 65 years of age, but their clinical manifestations and cranial imaging are different. The protein structure and function affected by these mutations were predicted using bioinformatics analysis, which suggested they were pathogenic. 3D protein structural analysis revealed that these amino acid substitutions might result in instability of protein structure and adverse intramolecular interactions. These findings suggest that both F193L and N466S should be thought as potential causative mutations in early-onset Alzheimer's disease (EOAD) patients. Further functional studies are warranted to evaluate their roles in the pathogenesis of AD.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/genetics , Mutation/genetics , Adult , Amino Acid Sequence , Female , Genetic Predisposition to Disease/genetics , Humans , LDL-Receptor Related Proteins/chemistry , Male , Membrane Transport Proteins/chemistry , Middle Aged , Pedigree , Protein Structure, Secondary , Risk Factors , Exome Sequencing/methods
4.
Mol Neurobiol ; 58(3): 1212-1216, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33118139

ABSTRACT

LDL receptor-related protein (LRP) 10 was recently identified as a Parkinson's disease gene through genome-wide linkage and sequencing analysis, but its role in Parkinson's disease in various populations is still unclear. The aim of this study was to determine the frequency and spectrum of LRP10 mutations in a cohort of Parkinson's disease patients from mainland China. All LRP10 exons and their flanking intron regions were screened by direct sequencing in 567 unrelated Parkinson's disease patients and 600 unrelated controls. We detected 29 exonic or splicing variants in 79 patients with Parkinson's disease. Five variants (c.A181C:p.I61L, c.C652T:p.Q218X, c.C833T:p.T278I, c.T1592G:p.I531S, c.T1697C:p.L566P) were predicted to be disease-causing or damaging by multiple in silico tools. Our study provides genetic evidence that LRP10 defects may correlate with sporadic Parkinson's disease.


Subject(s)
Genetic Predisposition to Disease , LDL-Receptor Related Proteins/genetics , Mutation/genetics , Parkinson Disease/genetics , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Asian People/genetics , China , Ethnicity/genetics , Female , Humans , LDL-Receptor Related Proteins/chemistry , Male , Middle Aged , Protein Domains , Young Adult
5.
Mol Brain ; 13(1): 166, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33302985

ABSTRACT

The low-density lipoprotein receptor-related protein 4 (LRP4) is essential for inducing the neuromuscular junction (NMJ) formation in muscle fibers, and LRP4 plays a critical role in dendritic development and synaptogenesis in the central nervous system (CNS). As a single transmembrane protein, LRP4 contains an enormously sizeable extracellular domain (ECD), containing multiple LDLα repeats in the N-terminal of ECD. LRP4 only with extracellular domain acts as a similar mechanism of full-length LRP4 in muscles to stimulate acetylcholine receptor clustering. In this study, we elucidated that LDLα repeats of LRP4 maintained the body weight and survival rate. Dendritic branches of the pyramidal neurons in Lrp4-null mice with LRP4 LDLα repeats residue were more than in Lrp4-null mice without residual LRP4 domain. Supplement with conditioned medium from LRP4 LDLα overexpression cells, the primary culture pyramidal neurons achieved strong dendritic arborization ability. Besides, astrocytes with LRP4 LDLα repeats residue could promote pyramidal neuronal dendrite arborization in the primary co-cultured system. These observations signify that LRP4 LDLα repeats play a prominent underlying role in dendrite arborization.


Subject(s)
Astrocytes/metabolism , Dendrites/metabolism , LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/metabolism , Repetitive Sequences, Amino Acid , Animals , Body Weight , Cells, Cultured , Cerebral Cortex/pathology , HEK293 Cells , Humans , Mice, Knockout , Protein Domains , Pyramidal Cells/metabolism , Survival Analysis
6.
J Magn Reson ; 310: 106639, 2020 01.
Article in English | MEDLINE | ID: mdl-31734618

ABSTRACT

Membrane proteins perform many important cellular functions. Historically, structural studies of these proteins have been conducted in detergent preparations and synthetic lipid bilayers. More recently, magic-angle-spinning (MAS) solid-state NMR has been employed to analyze membrane proteins in native membrane environments, but resonance assignments with this technique remain challenging due to limited spectral resolution and high resonance degeneracy. To tackle this issue, we combine reverse labeling of amino acids, frequency-selective dipolar dephasing, and NMR difference spectroscopy. These methods have resulted in nearly complete resonance assignments of the transmembrane domain of human LR11 (SorLA) protein in E. coli membranes. To reduce background signals from E. coli lipids and proteins and improve spectral sensitivity, we effectively utilize amylose affinity chromatography to prepare membrane vesicles when MBP is included as a fusion partner in the expression construct.


Subject(s)
Cell Membrane/chemistry , Escherichia coli/chemistry , LDL-Receptor Related Proteins/chemistry , Membrane Proteins/chemistry , Membrane Transport Proteins/chemistry , Amino Acid Sequence , Amino Acids/chemistry , Magnetic Resonance Spectroscopy
7.
Hum Mol Genet ; 28(16): 2648-2658, 2019 08 15.
Article in English | MEDLINE | ID: mdl-30994901

ABSTRACT

Agrin is a large extracellular matrix protein whose isoforms differ in their tissue distribution and function. Motoneuron-derived y+z+ agrin regulates the formation of the neuromuscular junction (NMJ), while y-z- agrin is widely expressed and has diverse functions. Previously we identified a missense mutation (V1727F) in the second laminin globular (LG2) domain of agrin that causes severe congenital myasthenic syndrome. Here, we define pathogenic effects of the agrin V1727F mutation that account for the profound dysfunction of the NMJ. First, by expressing agrin variants in heterologous cells, we show that the V1727F mutation reduces the secretion of y+z+ agrin compared to wild type, whereas it has no effect on the secretion of y-z- agrin. Second, we find that the V1727F mutation significantly impairs binding of y+z+ agrin to both heparin and the low-density lipoprotein receptor-related protein 4 (LRP4) coreceptor. Third, molecular modeling of the LG2 domain suggests that the V1727F mutation primarily disrupts the y splice insert, and consistent with this we find that it partially occludes the contribution of the y splice insert to agrin binding to heparin and LRP4. Together, these findings identify several pathogenic effects of the V1727F mutation that reduce its expression and ability to bind heparan sulfate proteoglycan and LRP4 coreceptors involved in the muscle-specific kinase signaling pathway. These defects primarily impair the function of neural y+z+ agrin and combine to cause a severe CMS phenotype, whereas y-z- agrin function in other tissues appears preserved.


Subject(s)
Agrin/genetics , Agrin/metabolism , Amino Acid Substitution , Gene Expression Regulation , Heparan Sulfate Proteoglycans/metabolism , LDL-Receptor Related Proteins/metabolism , Mutation , Agrin/chemistry , Alleles , Alternative Splicing , Cell Line , Heparan Sulfate Proteoglycans/chemistry , Humans , Immunohistochemistry , LDL-Receptor Related Proteins/chemistry , Models, Biological , Models, Molecular , Protein Binding , Protein Conformation , Protein Isoforms , Structure-Activity Relationship
8.
Sci Rep ; 9(1): 611, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679749

ABSTRACT

SorLA and Sortilin are multifunctional receptors involved in endocytosis and intracellular sorting of different and unrelated ligands. SorLA has recently attracted much attention as a novel strong risk gene for Alzheimer's disease, and much effort is currently being put into understanding the underlying molecular mechanism. Trafficking of SorLA and Sortilin are mediated by interacting with AP-1, AP-2, GGA 1-3 and the retromer complex. Although these cytosolic adaptor proteins all bind to both SorLA and Sortilin, a large fraction of intracellular Sortilin and SorLA are located in different subcellular vesicles. This indicates that unknown specialised adaptor proteins targeting SorLA for trafficking are yet to be discovered. We have identified HSPA12A as a new adaptor protein that, among Vps10p-D receptors, selectively binds to SorLA in an ADP/ATP dependent manner. This is the first described substrate of HSPA12A, and we demonstrate that the binding, which affects both endocytic speed and subcellular localisation of SorLA, is mediated by specific acidic residues in the cytosolic domain of SorLA. The identification of the relatively unknown HSPA12A as a SorLA specific interaction partner could lead to novel insight into the molecular mechanism of SorLA, and re-emphasises the role of heat shock proteins in neurodegenerative diseases.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , LDL-Receptor Related Proteins/metabolism , Membrane Transport Proteins/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Amino Acid Sequence , Animals , Astrocytes/cytology , Astrocytes/metabolism , HEK293 Cells , HSP70 Heat-Shock Proteins/chemistry , Humans , LDL-Receptor Related Proteins/chemistry , Membrane Transport Proteins/chemistry , Mice , Mice, Inbred C57BL , Protein Binding , Protein Domains , Protein Transport , Substrate Specificity , Two-Hybrid System Techniques
9.
Clin Chim Acta ; 489: 29-34, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30448281

ABSTRACT

BACKGROUND: LR11 is a member of the low-density lipoprotein (LDL) receptor family with high expression in neurons. Some cell surface LR11 is cleaved and secreted into the cerebrospinal fluid (CSF) as soluble LR11 (sLR11). Patients with Alzheimer's disease (AD), particularly apolipoprotein E4 carriers, have high CSF-sLR11 and low CSF-amyloid ß (Aß) concentrations. Therefore, we assessed whether sLR11 is bound to CSF-high-density lipoprotein (HDL) and whether sLR11 competes with Aß in binding to apoE in CSF-HDL. METHODS: We measured CSF-sLR11 concentrations (50 controls and 16 patients with AD) using enzyme immunoassay. sLR11 and apoE distribution in the CSF was evaluated using non-denaturing two-dimensional gel electrophoresis (N-2DGE). ApoE bound to sLR11 or Aß was identified using co-immunoprecipitation assay. RESULTS: CSF-sLR11 concentrations were higher in patients with AD than controls (adjusted for sLR11 using phospholipid). N-2DGE analysis showed that sLR11 and Aß comigrated with a large apoE-containing CSF-HDL. Moreover, fewer apoE was bound to Aß when a higher amount of apoE was bound to sLR11 in patients with AD who presented with ε4/4. CONCLUSION: sLR11 binds to CSF-HDL and competes with Aß in binding to apoE in CSF-HDL, indicating that sLR11 affects Aß clearance via CSF-HDL.


Subject(s)
Amyloid beta-Peptides/metabolism , LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/metabolism , Lipoproteins/cerebrospinal fluid , Lipoproteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/metabolism , Apolipoproteins E/metabolism , Binding, Competitive , Female , Humans , Lipoproteins/chemistry , Male , Solubility
10.
Clin Chim Acta ; 490: 69-76, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30550937

ABSTRACT

BACKGROUND: The levels of plasma sLR11, released from intimal SMCs, are positively associated with intima-media thickness (IMT) in asymptomatic subjects. We have evaluated the yet unknown pathological significance of sLR11 for plaque conditions in patients with carotid artery stenosis. METHODS: The presence of LR11 in carotid plaques was investigated using autopsy specimens. A clinical ultrasonography study for elucidating relationships between sLR11 and plaque condition was performed in 46 patients. RESULTS: Immunohistochemistry showed high levels of LR11 in SMCs within thickened intima and at the media-intima border of atherosclerotic carotid plaques. The levels of sLR11 in patients were clearly elevated compared to healthy controls. Univariate analysis of sLR11 revealed significant positive correlation with plaque score and a tendency to correlate with the stenotic fraction. Univariate and multiple regression analyses of plaque scores showed that sLR11, maximum IMT, and HDL-cholesterol independently determined plaque score. Finally, univariate analysis of initial sLR11 levels for changes in imaging markers after one-year follow-up showed that initial sLR11 levels significantly correlated with stenotic fraction progression. CONCLUSIONS: The levels of sLR11, abundantly expressed in carotid atherosclerotic plaques, are highly associated with increased plaque score. sLR11 levels may be predictive of plaque conditions in patients with advanced carotid atherosclerosis.


Subject(s)
Carotid Stenosis/complications , Cell Movement , LDL-Receptor Related Proteins/blood , LDL-Receptor Related Proteins/chemistry , Membrane Transport Proteins/blood , Membrane Transport Proteins/chemistry , Myocytes, Smooth Muscle/pathology , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/complications , Aged, 80 and over , Cell Differentiation , Female , Humans , Male , Plaque, Atherosclerotic/pathology
11.
Int J Mol Sci ; 19(10)2018 Oct 09.
Article in English | MEDLINE | ID: mdl-30304853

ABSTRACT

Apolipoprotein E receptor 2 (ApoER2) and VLDL receptor belong to the low density lipoprotein receptor family and bind apolipoprotein E. These receptors interact with the clathrin machinery to mediate endocytosis of macromolecules but also interact with other adapter proteins to perform as signal transduction receptors. The best characterized signaling pathway in which ApoER2 and VLDL receptor (VLDLR) are involved is the Reelin pathway. This pathway plays a pivotal role in the development of laminated structures of the brain and in synaptic plasticity of the adult brain. Since Reelin and apolipoprotein E, are ligands of ApoER2 and VLDLR, these receptors are of interest with respect to Alzheimer's disease. We will focus this review on the complex structure of ApoER2 and VLDLR and a recently characterized ligand, namely clusterin.


Subject(s)
LDL-Receptor Related Proteins/metabolism , Receptors, LDL/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Clusterin/genetics , Clusterin/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression , Humans , LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Binding , Receptors, LDL/chemistry , Receptors, LDL/genetics , Reelin Protein , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Structure-Activity Relationship
12.
FASEB J ; 32(7): 3536-3546, 2018 07.
Article in English | MEDLINE | ID: mdl-29442527

ABSTRACT

Increasing evidence indicates that altered reelin signaling could contribute to synaptic dysfunction in Alzheimer's disease (AD). We found that reelin protein and mRNA levels were increased in the AD brain (particularly at advanced Braak stages in apolipoprotein E4 noncarriers), compared with that of control subjects. The ß-amyloid (Aß) protein impairs reelin activity and increases reelin expression through a mechanism that is not yet understood. To explore that mechanism, we examined the effect of Aß aa 1-42 (Aß42) on DNA methylation of the RELN promoter and the processing of reelin receptor apolipoprotein E receptor 2 (ApoER2) in differentiated SH-SY5Y cells because ApoER2 C-terminal fragments (CTFs), generated after reelin binding, regulate reelin expression. We found that Aß42 decreased nuclear levels of DNA-methyltransferase 1. However, RELN promoter methylation did not change in Aß42-treated cells or in AD brain extracts. Instead, the levels of ApoER2-CTF appeared significantly lower in Aß42-treated cells and in AD extracts from advanced Braak stages of apolipoprotein E4 noncarriers. Our data show that ApoER2-CTF levels are decreased, whereas reelin expression is increased in AD brain at advanced Braak stages and after Aß treatment, supporting the view that ApoER2-CTF exerts a modulatory role on reelin expression.-Mata-Balaguer, T., Cuchillo-Ibañez, I., Calero, M., Ferrer, I., Sáez-Valero, J. Decreased generation of C-terminal fragments of ApoER2 and increased reelin expression in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Nerve Tissue Proteins/genetics , Serine Endopeptidases/genetics , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line, Tumor , DNA Methylation , Extracellular Matrix Proteins/metabolism , Female , Humans , LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/genetics , Male , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/toxicity , Promoter Regions, Genetic , Reelin Protein , Serine Endopeptidases/metabolism
13.
EMBO Rep ; 18(6): 982-999, 2017 06.
Article in English | MEDLINE | ID: mdl-28446613

ABSTRACT

Apolipoprotein E receptor 2 (ApoER2) is a close homologue of low-density lipoprotein receptor (LDLR) that mediates the endocytosis of ligands, including LDL particles. LDLR family members have been presumed to explore a large conformational space to capture ligands in the extended conformation at the cell surface. Ligands are subsequently released through a pH-titrated structural transition to a self-docked, contracted-closed conformation. In addition to lipoprotein uptake, ApoER2 is implicated in signal transduction during brain development through capture of the extracellular protein reelin. From crystallographic analysis, we determine that the full-length ApoER2 ectodomain adopts an intermediate contracted-open conformation when complexed with the signaling-competent reelin fragment, and we identify a previously unappreciated auxiliary low-affinity binding interface. Based on mutational analyses, we propose that the pH shift during endocytosis weakens the affinity of the auxiliary interface and destabilizes the ligand-receptor complex. Furthermore, this study elucidates that the contracted-open conformation of ligand-bound ApoER2 at neutral pH resembles the contracted-closed conformation of ligand-unbound LDLR at acidic pH in a manner suggestive of being primed for ligand release even prior to internalization.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/metabolism , Nerve Tissue Proteins/physiology , Serine Endopeptidases/physiology , Animals , CHO Cells , Cell Adhesion Molecules, Neuronal/chemistry , Cricetulus , Crystallography , Endocytosis , Endosomes/physiology , Extracellular Matrix Proteins/chemistry , Humans , Hydrogen-Ion Concentration , LDL-Receptor Related Proteins/genetics , Ligands , Lipoproteins, LDL/metabolism , Nerve Tissue Proteins/chemistry , Neurons/physiology , Protein Conformation , Receptors, LDL/metabolism , Reelin Protein , Serine Endopeptidases/chemistry , Signal Transduction , Surface Plasmon Resonance
14.
Proc Natl Acad Sci U S A ; 114(5): 1159-1164, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28100490

ABSTRACT

The kainate receptor (KAR), a subtype of glutamate receptor, mediates excitatory synaptic responses at a subset of glutamatergic synapses. However, the molecular mechanisms underlying the trafficking of its different subunits are poorly understood. Here we use the CA1 hippocampal pyramidal cell, which lacks KAR-mediated synaptic currents, as a null background to determine the minimal requirements for the extrasynaptic and synaptic expression of the GluK2 subunit. We find that the GluK2 receptor itself, in contrast to GluK1, traffics to the neuronal surface and synapse efficiently and the auxiliary subunits Neto1 and Neto2 caused no further enhancement of these two trafficking processes. However, the regulation of GluK2 biophysical properties by Neto proteins is the same as that of GluK1. We further determine that it is the amino-terminal domains (ATDs) of GluK1 and GluK2 that control the strikingly different trafficking properties between these two receptors. Moreover, the ATDs are critical for synaptic expression of heteromeric receptors at mossy fiber-CA3 synapses and also mediate the differential dependence on Neto proteins for surface and synaptic trafficking of GluK1 and GluK2. These results highlight the fundamental differences between the two major KAR subunits and their interplay with Neto auxiliary proteins.


Subject(s)
CA1 Region, Hippocampal/physiology , LDL-Receptor Related Proteins/physiology , Membrane Proteins/physiology , Protein Transport/physiology , Pyramidal Cells/physiology , Receptors, Kainic Acid/physiology , Amino Acid Substitution , Animals , Dimerization , LDL-Receptor Related Proteins/chemistry , Membrane Proteins/chemistry , Mice , Miniature Postsynaptic Potentials/physiology , Organ Culture Techniques , Patch-Clamp Techniques , Protein Domains , Protein Interaction Maps , Rats , Receptors, Kainic Acid/chemistry , Receptors, Kainic Acid/genetics , Receptors, N-Methyl-D-Aspartate , Recombinant Proteins/metabolism , Structure-Activity Relationship , Synaptic Transmission/physiology , GluK2 Kainate Receptor
15.
Clin Chim Acta ; 463: 47-52, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27725222

ABSTRACT

BACKGROUND: LR11 (also called SorLA or SORL1) is a type I membrane protein, originally identified as a biomarker for atherosclerosis and Alzheimer's disease. We recently found that LR11 was specifically expressed in Diffuse Large B-cell lymphoma (DLBCL) cells, and high serum sLR11 concentrations in retrospective cohort indicated inferior survival. In this study, we prospectively validated the clinical impact of serum sLR11 in 97 patients with newly-diagnosed, untreated DLBCL. RESULTS: Serum sLR11 concentrations were increased in DLBCL patients compared to normal controls (mean±SD: 21.2±27.6 vs. 8.8±1.8ng/ml, P<0.0001), and significantly reduced at remission (mean±SD: 17.4±16.4 vs. 10.9±4.5ng/ml, P=0.02). Increased serum sLR11 concentrations were affected by tumor burden and bone marrow invasion. The 2-y OS and PFS were significantly lower in patients with high sLR11 concentrations (≤18.1ng/ml vs. >18.1ng/ml; 2-y OS: 89.0% vs. 56.4%, P<0.0001; 2-y PFS: 85.8% vs. 56.9%, P<0.0001). CONCLUSIONS: Serum sLR11 is a tumor-derived biomarker for predicting the survival of newly diagnosed patients with DLBCL.


Subject(s)
LDL-Receptor Related Proteins/blood , LDL-Receptor Related Proteins/chemistry , Lymphoma, Large B-Cell, Diffuse/blood , Lymphoma, Large B-Cell, Diffuse/diagnosis , Membrane Transport Proteins/blood , Membrane Transport Proteins/chemistry , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Biomarkers, Tumor/chemistry , Bone Marrow/pathology , Female , Humans , Male , Middle Aged , Reproducibility of Results , Solubility , Survival Analysis , Tumor Burden , Young Adult
16.
FEBS J ; 283(13): 2476-93, 2016 07.
Article in English | MEDLINE | ID: mdl-27192064

ABSTRACT

The sorting receptor SorLA is highly expressed in neurons and is also found in other polarized cells. The receptor has been reported to participate in the trafficking of several ligands, some of which are linked to human diseases, including the amyloid precursor protein, TrkB, and Lipoprotein Lipase (LpL). Despite this, only the trafficking in nonpolarized cells has been described so far. Due to the many differences between polarized and nonpolarized cells, we examined the localization and trafficking of SorLA in epithelial Madin-Darby canine kidney (MDCK) cells and rat hippocampal neurons. We show that SorLA is mainly found in sorting endosomes and on the basolateral surface of MDCK cells and in the somatodendritic domain of neurons. This polarized distribution of SorLA respectively depends on an acidic cluster and an extended version of this cluster and involves the cellular adaptor complex AP-1. Furthermore, we show that SorLA can mediate transcytosis across a tight cell layer.


Subject(s)
Cell Polarity/physiology , LDL-Receptor Related Proteins/metabolism , Neurons/metabolism , Animals , Cell Polarity/genetics , Dogs , Endosomes/metabolism , HEK293 Cells , Hippocampus/cytology , Humans , LDL-Receptor Related Proteins/chemistry , Madin Darby Canine Kidney Cells , Mice , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Transcytosis/genetics , Transcytosis/physiology
17.
Mol Ther ; 23(10): 1622-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26156244

ABSTRACT

Target identification is highly instructive in defining the biological roles of microRNAs. However, little is known about other small noncoding RNAs; for example, tRNA-derived RNA Fragments (tRFs). Some tRFs exhibit a gene-silencing mechanism distinctly different from that of typical microRNAs. We recently demonstrated that a respiratory syncytial virus (RSV)-induced tRF, called tRF5-GluCTC, promotes RSV replication. RSV is the single most important cause of lower respiratory tract infection in children. By using biochemical screening and bioinformatics analyses, we have identified apolipoprotein E receptor 2 (APOER2) as a target of tRF5-GluCTC. The 3'-portion of tRF5-GluCTC recognizes a target site in the 3'-untranslated region of APOER2 and suppresses its expression. We have also discovered that APOER2 is an anti-RSV protein whose suppression by tRF5-GluCTC promotes RSV replication. Our report represents the first identification of a natural target of a tRF and illustrates how a virus utilizes a host tRF to control a host gene to favor its replication.


Subject(s)
Gene Silencing , RNA Interference , Base Sequence , Binding Sites , Cell Line , Gene Expression Regulation , Humans , LDL-Receptor Related Proteins/chemistry , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Protein Binding , RNA, Small Untranslated/chemistry , RNA, Small Untranslated/genetics , RNA, Transfer/chemistry , RNA, Transfer/genetics , Respiratory Syncytial Virus, Human/physiology , Transfection , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism , Virus Replication/genetics
18.
Biochemistry ; 54(15): 2490-9, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25835329

ABSTRACT

The amyloid precursor protein (APP) and its neurotoxic cleavage product Aß are key players in the development of Alzheimer's disease (AD) and appear to be essential for neuronal development and cell homeostasis. Proteolytic processing of APP and its physiological function depend on its interaction with heparin and are influenced by the binding of metal ions and sorLA. We created various mutations of metal binding site M1 residing within the extracellular E2 domain of APP. Using isothermal titration calorimetry and circular dichroism spectroscopy, we analyzed the binding of Cu(2+) and Zn(2+) to APP E2 and identified two mutations that are most suited for functional studies to dissect ion specific effects of metal binding. The H313A mutation abrogates only copper-based effects, whereas the H382A mutation weakens any metal binding at M1 of APP E2. Subsequently, we tested the effect of Cu(2+) and Zn(2+) on the binding of heparin and sorLA to APP E2 using a chromatographic technique and surface plasmon resonance. We show that Zn(2+) and to a larger degree also Cu(2+) enhance the binding of heparin to APP E2, consistent with an extracellular regulation of the function of APP by both metal ions. In contrast, neither ion seemed to affect the interaction between APP E2 and sorLA. This supports an intracellular interaction between the latter two partners that would not sense extracellular variations of metal ions upon synaptic activity.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Copper/chemistry , Heparin/chemistry , LDL-Receptor Related Proteins/chemistry , Membrane Transport Proteins/chemistry , Mutation, Missense , Zinc/chemistry , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amino Acid Substitution , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Binding Sites , Copper/metabolism , Heparin/metabolism , Humans , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Protein Binding , Surface Plasmon Resonance , Zinc/metabolism
19.
Nat Struct Mol Biol ; 22(3): 199-206, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25643321

ABSTRACT

SorLA is a neuronal sorting receptor considered to be a major risk factor for Alzheimer's disease. We have recently reported that it directs lysosomal targeting of nascent neurotoxic amyloid-ß (Aß) peptides by directly binding Aß. Here, we determined the crystal structure of the human sorLA domain responsible for Aß capture, Vps10p, in an unbound state and in complex with two ligands. Vps10p assumes a ten-bladed ß-propeller fold with a large tunnel at the center. An internal ligand derived from the sorLA propeptide bound inside the tunnel to extend the ß-sheet of one of the propeller blades. The structure of the sorLA Vps10p-Aß complex revealed that the same site is used. Peptides are recognized by sorLA Vps10p in redundant modes without strict dependence on a particular amino acid sequence, thus suggesting a broad specificity toward peptides with a propensity for ß-sheet formation.


Subject(s)
Amyloid beta-Peptides/metabolism , LDL-Receptor Related Proteins/chemistry , Membrane Transport Proteins/chemistry , Binding Sites , Crystallography, X-Ray , Humans , LDL-Receptor Related Proteins/metabolism , LDL-Receptor Related Proteins/physiology , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/physiology , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary
20.
PLoS One ; 10(2): e0116701, 2015.
Article in English | MEDLINE | ID: mdl-25688974

ABSTRACT

Apolipoprotein E (ApoE) genotype is the strongest predictor of Alzheimer's Disease (AD) risk. ApoE is a cholesterol transport protein that binds to members of the Low-Density Lipoprotein (LDL) Receptor family, which includes LDL Receptor Related Protein 4 (Lrp4). Lrp4, together with one of its ligands Agrin and its co-receptors Muscle Specific Kinase (MuSK) and Amyloid Precursor Protein (APP), regulates neuromuscular junction (NMJ) formation. All four proteins are also expressed in the adult brain, and APP, MuSK, and Agrin are required for normal synapse function in the CNS. Here, we show that Lrp4 is also required for normal hippocampal plasticity. In contrast to the closely related Lrp8/Apoer2, the intracellular domain of Lrp4 does not appear to be necessary for normal expression and maintenance of long-term potentiation at central synapses or for the formation and maintenance of peripheral NMJs. However, it does play a role in limb development.


Subject(s)
Brain/embryology , Extremities/embryology , LDL-Receptor Related Proteins/genetics , Neuronal Plasticity/genetics , Organogenesis/genetics , Protein Interaction Domains and Motifs/genetics , Animals , Humans , LDL-Receptor Related Proteins/chemistry , Limb Deformities, Congenital/genetics , Long-Term Potentiation/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...