Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 665
Filter
1.
Proc Natl Acad Sci U S A ; 121(37): e2408262121, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39226352

ABSTRACT

Truncating genetic variants of SORL1, encoding the endosome recycling receptor SORLA, have been accepted as causal of Alzheimer's disease (AD). However, most genetic variants observed in SORL1 are missense variants, for which it is complicated to determine the pathogenicity level because carriers come from pedigrees too small to be informative for penetrance estimations. Here, we describe three unrelated families in which the SORL1 coding missense variant rs772677709, that leads to a p.Y1816C substitution, segregates with Alzheimer's disease. Further, we investigate the effect of SORLA p.Y1816C on receptor maturation, cellular localization, and trafficking in cell-based assays. Under physiological circumstances, SORLA dimerizes within the endosome, allowing retromer-dependent trafficking from the endosome to the cell surface, where the luminal part is shed into the extracellular space (sSORLA). Our results showed that the p.Y1816C mutant impairs SORLA homodimerization in the endosome, leading to decreased trafficking to the cell surface and less sSORLA shedding. These trafficking defects of the mutant receptor can be rescued by the expression of the SORLA 3Fn-minireceptor. Finally, we find that iPSC-derived neurons with the engineered p.Y1816C mutation have enlarged endosomes, a defining cytopathology of AD. Our studies provide genetic as well as functional evidence that the SORL1 p.Y1816C variant is causal for AD. The partial penetrance of the mutation suggests this mutation should be considered in clinical genetic screening of multiplex early-onset AD families.


Subject(s)
Alzheimer Disease , Endosomes , LDL-Receptor Related Proteins , Membrane Transport Proteins , Pedigree , Humans , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Endosomes/metabolism , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Female , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mutation, Missense , Protein Transport , Protein Multimerization , Aged , Middle Aged , HEK293 Cells
2.
Biochem Biophys Res Commun ; 731: 150400, 2024 Oct 30.
Article in English | MEDLINE | ID: mdl-39024975

ABSTRACT

Neuromuscular signal transmission is affected in various diseases including myasthenia gravis, congenital myasthenic syndromes, and sarcopenia. We used an ATF2-luciferase system to monitor the phosphorylation of MuSK in HEK293 cells introduced with MUSK and LRP4 cDNAs to find novel chemical compounds that enhanced agrin-mediated acetylcholine receptor (AChR) clustering. Four compounds with similar chemical structures carrying benzene rings and heterocyclic rings increased the luciferase activities 8- to 30-folds, and two of them showed continuously graded dose dependence. The effects were higher than that of disulfiram, a clinically available aldehyde dehydrogenase inhibitor, which we identified to be the most competent preapproved drug to enhance ATF2-luciferase activity in the same assay system. In C2C12 myotubes, all the compounds increased the area, intensity, length, and number of AChR clusters. Three of the four compounds increased the phosphorylation of MuSK, but not of Dok7, JNK. ERK, or p38. Monitoring cell toxicity using the neurite elongation of NSC34 neuronal cells as a surrogate marker showed that all the compounds had no effects on the neurite elongation up to 1 µM. Extensive docking simulation and binding structure prediction of the four compounds with all available human proteins using AutoDock Vina and DiffDock showed that the four compounds were unlikely to directly bind to MuSK or Dok7, and the exact target remained unknown. The identified compounds are expected to serve as a seed to develop a novel therapeutic agent to treat defective NMJ signal transmission.


Subject(s)
Muscle Fibers, Skeletal , Receptors, Nicotinic , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Animals , Mice , Cell Line , Humans , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Genes, Reporter , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Multigene Family , Signal Transduction/drug effects , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neurites , Bungarotoxins/pharmacology , Benzene/pharmacology , Heterocyclic Compounds/pharmacology , Molecular Docking Simulation
3.
EMBO Rep ; 25(8): 3601-3626, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38956225

ABSTRACT

Signals emanating from the T-cell receptor (TCR), co-stimulatory receptors, and cytokine receptors each influence CD8 T-cell fate. Understanding how these signals respond to homeostatic and microenvironmental cues can reveal new ways to therapeutically direct T-cell function. Through forward genetic screening in mice, we discover that loss-of-function mutations in LDL receptor-related protein 10 (Lrp10) cause naive and central memory CD8 T cells to accumulate in peripheral lymphoid organs. Lrp10 encodes a conserved cell surface protein of unknown immunological function. T-cell activation induces Lrp10 expression, which post-translationally suppresses IL7 receptor (IL7R) levels. Accordingly, Lrp10 deletion enhances T-cell homeostatic expansion through IL7R signaling. Lrp10-deficient mice are also intrinsically resistant to syngeneic tumors. This phenotype depends on dense tumor infiltration of CD8 T cells, which display increased memory cell characteristics, reduced terminal exhaustion, and augmented responses to immune checkpoint inhibition. Here, we present Lrp10 as a new negative regulator of CD8 T-cell homeostasis and a host factor that controls tumor resistance with implications for immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Homeostasis , Receptors, Interleukin-7 , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Receptors, Interleukin-7/metabolism , Receptors, Interleukin-7/genetics , LDL-Receptor Related Proteins/metabolism , LDL-Receptor Related Proteins/genetics , Signal Transduction , Lymphocyte Activation/immunology , Mice, Knockout , Mice, Inbred C57BL , Immunologic Memory , Neoplasms/immunology , Neoplasms/genetics , Humans
4.
Biomolecules ; 14(7)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39062513

ABSTRACT

Lowe Syndrome (LS) is a rare X-linked disorder characterized by renal dysfunction, cataracts, and several central nervous system (CNS) anomalies. The mechanisms underlying the neurological dysfunction in LS remain unclear, albeit they share some phenotypic characteristics similar to the deficiency or dysfunction of the Reelin signaling, a relevant pathway with roles in CNS development and neuronal functions. In this study, we investigated the role of OCRL1, an inositol polyphosphate 5-phosphatase encoded by the OCRL gene, mutated in LS, focusing on its impact on endosomal trafficking and receptor recycling in human neuronal cells. Specifically, we tested the effects of OCRL1 deficiency in the trafficking and signaling of ApoER2/LRP8, a receptor for the ligand Reelin. We found that loss of OCRL1 impairs ApoER2 intracellular trafficking, leading to reduced receptor expression and decreased levels at the plasma membrane. Additionally, human neurons deficient in OCRL1 showed impairments in ApoER2/Reelin-induced responses. Our findings highlight the critical role of OCRL1 in regulating ApoER2 endosomal recycling and its impact on the ApoER2/Reelin signaling pathway, providing insights into potential mechanisms underlying the neurological manifestations of LS.


Subject(s)
Cell Adhesion Molecules, Neuronal , Endosomes , Extracellular Matrix Proteins , LDL-Receptor Related Proteins , Nerve Tissue Proteins , Neurons , Phosphoric Monoester Hydrolases , Protein Transport , Reelin Protein , Serine Endopeptidases , Humans , Phosphoric Monoester Hydrolases/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/deficiency , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/deficiency , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/deficiency , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/deficiency , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/deficiency , Endosomes/metabolism , Neurons/metabolism , LDL-Receptor Related Proteins/metabolism , LDL-Receptor Related Proteins/genetics , Signal Transduction , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/metabolism
5.
PLoS Genet ; 20(7): e1011348, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39038048

ABSTRACT

Disrupted alternative splicing plays a determinative role in neurological diseases, either as a direct cause or as a driver in disease susceptibility. Transcriptomic profiling of aged human postmortem brain samples has uncovered hundreds of aberrant mRNA splicing events in Alzheimer's disease (AD) brains, associating dysregulated RNA splicing with disease. We previously identified a complex array of alternative splicing combinations across apolipoprotein E receptor 2 (APOER2), a transmembrane receptor that interacts with both the neuroprotective ligand Reelin and the AD-associated risk factor, APOE. Many of the human APOER2 isoforms, predominantly featuring cassette splicing events within functionally important domains, are critical for the receptor's function and ligand interaction. However, a comprehensive repertoire and the functional implications of APOER2 isoforms under both physiological and AD conditions are not fully understood. Here, we present an in-depth analysis of the splicing landscape of human APOER2 isoforms in normal and AD states. Using single-molecule, long-read sequencing, we profiled the entire APOER2 transcript from the parietal cortex and hippocampus of Braak stage IV AD brain tissues along with age-matched controls and investigated several functional properties of APOER2 isoforms. Our findings reveal diverse patterns of cassette exon skipping for APOER2 isoforms, with some showing region-specific expression and others unique to AD-affected brains. Notably, exon 15 of APOER2, which encodes the glycosylation domain, showed less inclusion in AD compared to control in the parietal cortex of females with an APOE ɛ3/ɛ3 genotype. Also, some of these APOER2 isoforms demonstrated changes in cell surface expression, APOE-mediated receptor processing, and synaptic number. These variations are likely critical in inducing synaptic alterations and may contribute to the neuronal dysfunction underlying AD pathogenesis.


Subject(s)
Alternative Splicing , Alzheimer Disease , LDL-Receptor Related Proteins , Reelin Protein , Humans , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Alternative Splicing/genetics , Protein Isoforms/genetics , Sequence Analysis, RNA , Female , Aged , Brain/metabolism , Brain/pathology , Apolipoproteins E/genetics , Male , Hippocampus/metabolism , Hippocampus/pathology , Aged, 80 and over , RNA Splicing/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
6.
Mol Ther ; 32(7): 2176-2189, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38734896

ABSTRACT

The disassembly of the neuromuscular junction (NMJ) is an early event in amyotrophic lateral sclerosis (ALS), ultimately leading to motor dysfunction and lethal respiratory paralysis. The hexanucleotide GGGGCC repeat expansion in the C9orf72 gene is the most common genetic mutation, and the dipeptide repeat (DPR) proteins have been shown to cause neurodegeneration. While no drugs can treat ALS patients efficiently, new treatment strategies are urgently needed. Here, we report that a MuSK agonist antibody alleviates poly-PR-induced NMJ deficits in C9orf72-ALS mice. The HB9-PRF/F mice, which express poly-PR proteins in motor neurons, exhibited impaired motor behavior and NMJ deficits. Mechanistically, poly-PR proteins interacted with Agrin to disrupt the interaction between Agrin and Lrp4, leading to attenuated activation of MuSK. Treatment with a MuSK agonist antibody rescued NMJ deficits, and extended the lifespan of C9orf72-ALS mice. Moreover, impaired NMJ transmission was observed in C9orf72-ALS patients. These findings identify the mechanism by which poly-PR proteins attenuate MuSK activation and NMJ transmission, highlighting the potential of promoting MuSK activation with an agonist antibody as a therapeutic strategy to protect NMJ function and prolong the lifespan of ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis , C9orf72 Protein , Disease Models, Animal , Neuromuscular Junction , Receptor Protein-Tyrosine Kinases , Animals , Neuromuscular Junction/metabolism , Neuromuscular Junction/drug effects , Mice , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Humans , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Longevity/drug effects , Motor Neurons/metabolism , Motor Neurons/drug effects , Agrin/metabolism , Agrin/genetics , Mice, Transgenic , Antibodies/pharmacology , Receptors, Cholinergic/metabolism , Receptors, Cholinergic/genetics , LDL-Receptor Related Proteins/metabolism , LDL-Receptor Related Proteins/genetics
7.
Article in English | MEDLINE | ID: mdl-38726482

ABSTRACT

In patients of Asian ancestry, a heterozygous CGG repeat expansion of >100 units in LRP12 is the cause of oculopharyngodistal myopathy type 1 (OPDM1). Repeat lengths of between 61 and 100 units have been associated with rare amyotrophic lateral sclerosis (ALS) cases of Asian ancestry, although with unusually long disease duration and without significant upper motor neuron involvement. This study sought to determine whether LRP12 CGG repeat expansions were also present in ALS patients of European ancestry. Whole-genome sequencing data from 608 sporadic ALS patients, 35 familial ALS probands, and 4703 neurologically normal controls were screened for LRP12 CGG expansions using ExpansionHunter v4. All individuals had LRP12 CGG repeat lengths within the normal range of 3-25 units. To date, LRP12 CGG repeat expansions have not been reported in ALS patients of European ancestry and may be limited to rare ALS patients of Asian ancestry and atypical clinical presentations.


Subject(s)
Amyotrophic Lateral Sclerosis , White People , Humans , Amyotrophic Lateral Sclerosis/genetics , Male , Female , White People/genetics , Middle Aged , Aged , Adult , LDL-Receptor Related Proteins/genetics , Cohort Studies , Trinucleotide Repeat Expansion/genetics
8.
JCI Insight ; 9(9)2024 May 08.
Article in English | MEDLINE | ID: mdl-38716730

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths in the world, and non-small cell lung cancer (NSCLC) is the most common subset. We previously found that infiltration of tumor inflammatory monocytes (TIMs) into lung squamous carcinoma (LUSC) tumors is associated with increased metastases and poor survival. To further understand how TIMs promote metastases, we compared RNA-Seq profiles of TIMs from several LUSC metastatic models with inflammatory monocytes (IMs) of non-tumor-bearing controls. We identified Spon1 as upregulated in TIMs and found that Spon1 expression in LUSC tumors corresponded with poor survival and enrichment of collagen extracellular matrix signatures. We observed SPON1+ TIMs mediate their effects directly through LRP8 on NSCLC cells, which resulted in TGF-ß1 activation and robust production of fibrillar collagens. Using several orthogonal approaches, we demonstrated that SPON1+ TIMs were sufficient to promote NSCLC metastases. Additionally, we found that Spon1 loss in the host, or Lrp8 loss in cancer cells, resulted in a significant decrease of both high-density collagen matrices and metastases. Finally, we confirmed the relevance of the SPON1/LRP8/TGF-ß1 axis with collagen production and survival in patients with NSCLC. Taken together, our study describes how SPON1+ TIMs promote collagen remodeling and NSCLC metastases through an LRP8/TGF-ß1 signaling axis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Monocytes , Signal Transduction , Animals , Humans , Mice , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/secondary , Cell Line, Tumor , Collagen/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , LDL-Receptor Related Proteins/metabolism , LDL-Receptor Related Proteins/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lung Neoplasms/genetics , Monocytes/metabolism , Monocytes/pathology , Neoplasm Metastasis , Transforming Growth Factor beta1/metabolism
9.
J Biol Chem ; 300(6): 107313, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657864

ABSTRACT

Sortilin-related receptor 1 (SORL1) is an intracellular sorting receptor genetically implicated in Alzheimer's disease (AD) that impacts amyloid precursor protein trafficking. The objective of these studies was to test the hypothesis that SORL1 binds tau, modulates its cellular trafficking and impacts the aggregation of cytoplasmic tau induced by pathological forms of tau. Using surface plasmon resonance measurements, we observed high-affinity binding of tau to SORL1 and the vacuolar protein sorting 10 domain of SORL1. Interestingly, unlike LDL receptor-related protein 1, SORL1 binds tau at both pH 7.4 and pH 5.5, revealing its ability to bind tau at endosomal pH. Immunofluorescence studies confirmed that exogenously added tau colocalized with SORL1 in H4 neuroglioma cells, while overexpression of SORL1 in LDL receptor-related protein 1-deficient Chinese hamster ovary (CHO) cells resulted in a marked increase in the internalization of tau, indicating that SORL1 can bind and mediate the internalization of monomeric forms of tau. We further demonstrated that SORL1 mediates tau seeding when tau RD P301S FRET biosensor cells expressing SORL1 were incubated with high molecular weight forms of tau isolated from the brains of patients with AD. Seeding in H4 neuroglioma cells is significantly reduced when SORL1 is knocked down with siRNA. Finally, we demonstrate that the N1358S mutant of SORL1 significantly increases tau seeding when compared to WT SORL1, identifying for the first time a potential mechanism that connects this specific SORL1 mutation to Alzheimer's disease. Together, these studies identify SORL1 as a receptor that contributes to trafficking and seeding of pathogenic tau.


Subject(s)
Cricetulus , LDL-Receptor Related Proteins , Membrane Transport Proteins , tau Proteins , Humans , tau Proteins/metabolism , tau Proteins/genetics , Animals , CHO Cells , LDL-Receptor Related Proteins/metabolism , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cell Line, Tumor , Protein Binding , Protein Transport
10.
Behav Neurosci ; 138(2): 125-141, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38661671

ABSTRACT

Selenium is an essential trace element that is delivered to the brain by the selenium transport protein selenoprotein P (SEPP1), primarily by binding to its receptor low-density lipoprotein receptor-related protein 8 (LRP8), also known as apolipoprotein E receptor 2 (ApoER2), at the blood-brain barrier. Selenium transport is required for several important brain functions, with transgenic deletion of either Sepp1 or Lrp8 resulting in severe neurological dysfunction and death in mice fed a selenium-deficient diet. Previous studies have reported that although feeding a standard chow diet can prevent these severe deficits, some motor coordination and cognitive dysfunction remain. Importantly, no single study has directly compared the motor and cognitive performance of the Sepp1 and Lrp8 knockout (KO) lines. Here, we report the results of a comprehensive parallel analysis of the motor and spatial learning and memory function of Sepp1 and Lrp8 knockout mice fed a standard mouse chow diet. Our results revealed that Sepp1 knockout mice raised on a selenium-replete diet displayed motor and cognitive function that was indistinguishable from their wild-type littermates. In contrast, we found that although Lrp8-knockout mice fed a selenium-replete diet had normal motor function, their spatial learning and memory showed subtle deficits. We also found that the deficit in baseline adult hippocampal neurogenesis exhibited by Lrp8-deficit mice could not be rescued by dietary selenium supplementation. Taken together, these findings further highlight the importance of selenium transport in maintaining healthy brain function. (PsycInfo Database Record (c) 2024 APA, all rights reserved).


Subject(s)
LDL-Receptor Related Proteins , Mice, Knockout , Selenium , Spatial Learning , Animals , Mice , Diet , Hippocampus/metabolism , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Maze Learning/physiology , Maze Learning/drug effects , Memory/physiology , Memory/drug effects , Selenium/administration & dosage , Selenium/deficiency , Selenium/pharmacology , Selenoprotein P/genetics , Selenoprotein P/metabolism , Spatial Learning/physiology , Spatial Learning/drug effects , Spatial Memory/physiology , Spatial Memory/drug effects
12.
EMBO Rep ; 25(5): 2278-2305, 2024 May.
Article in English | MEDLINE | ID: mdl-38499808

ABSTRACT

SorLA, encoded by the gene SORL1, is an intracellular sorting receptor of the VPS10P domain receptor gene family. Although SorLA is best recognized for its ability to shuttle target proteins between intracellular compartments in neurons, recent data suggest that also its microglial expression can be of high relevance for the pathogenesis of brain diseases, including glioblastoma (GBM). Here, we interrogated the impact of SorLA on the functional properties of glioma-associated microglia and macrophages (GAMs). In the GBM microenvironment, GAMs are re-programmed and lose the ability to elicit anti-tumor responses. Instead, they acquire a glioma-supporting phenotype, which is a key mechanism promoting glioma progression. Our re-analysis of published scRNA-seq data from GBM patients revealed that functional phenotypes of GAMs are linked to the level of SORL1 expression, which was further confirmed using in vitro models. Moreover, we demonstrate that SorLA restrains secretion of TNFα from microglia to restrict the inflammatory potential of these cells. Finally, we show that loss of SorLA exacerbates the pro-inflammatory response of microglia in the murine model of glioma and suppresses tumor growth.


Subject(s)
Brain Neoplasms , Glioma , LDL-Receptor Related Proteins , Membrane Transport Proteins , Microglia , Tumor Microenvironment , Tumor Necrosis Factor-alpha , Animals , Humans , Mice , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Cell Line, Tumor , Disease Models, Animal , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Glioma/metabolism , Glioma/pathology , Glioma/genetics , Macrophages/metabolism , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/genetics , Microglia/metabolism , Microglia/pathology , Tumor Necrosis Factor-alpha/metabolism , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism
13.
Stem Cell Res ; 77: 103359, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38460235

ABSTRACT

Parkinson's disease (PD) is a highly prevalent and severe neurodegenerative disease that affects more than 10 million individuals worldwide. Pathogenic mutations in LRP10 have been associated with autosomal dominant PD. Here, we report an induced pluripotent stem cell (iPSC) line generated from a PD patient harboring the LRP10 c.688C > T (p.Arg230Trp) variant. Skin fibroblasts from the PD patient were successfully reprogrammed into iPSCs that expressed pluripotency markers, a normal karyotype, and the capacity to differentiate into the three germ layers in vivo. This iPSC line is a potential resource for studying the pathogenic mechanisms of PD.


Subject(s)
Induced Pluripotent Stem Cells , Mutation , Parkinson Disease , Induced Pluripotent Stem Cells/metabolism , Humans , Parkinson Disease/genetics , Parkinson Disease/pathology , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Cell Line , Cell Differentiation , Male
14.
Philos Trans R Soc Lond B Biol Sci ; 379(1899): 20220377, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38368933

ABSTRACT

SORLA, the protein encoded by the SORL1 gene, has an important role in recycling cargo proteins to the cell surface. While SORLA loss-of-function variants occur almost exclusively in Alzheimer's disease cases, the majority of SORL1 variants are missense variants that are individually rare and can have individual mechanisms how they impair SORLA function as well as have individual effect size on disease risk. However, since carriers mostly come from small pedigrees, it is challenging to determine variant penetrance, leaving clinical significance associated with most missense variants unclear. In this article, we present functional approaches to evaluate the pathogenicity of a SORL1 variant, p.D1105H. First, we generated our mutant receptor by inserting the D1105H variant into the full-length SORLA-WT receptor. Then using western blot analysis we quantified the effect of the mutation on maturation and shedding of the receptor for transfected cells, and finally applied a flow cytometry approach to quantify SORLA expression at the cell surface. The results showed decreased maturation, decreased shedding, and decreased cell surface expression of D1105H compared with wild-type SORLA. We propose how these approaches can be used to functionally assess the pathogenicity of SORL1 variants in the future. This article is part of a discussion meeting issue 'Understanding the endo-lysosomal network in neurodegeneration'.


Subject(s)
Alzheimer Disease , Humans , Virulence , Mutation , Alzheimer Disease/genetics , Genetic Predisposition to Disease , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Membrane Transport Proteins/genetics
15.
Prog Neurobiol ; 234: 102575, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38281682

ABSTRACT

Adaptor protein complex 4 (AP-4) is a heterotetrameric complex that promotes export of selected cargo proteins from the trans-Golgi network. Mutations in each of the AP-4 subunits cause a complicated form of Hereditary Spastic Paraplegia (HSP). Herein, we report that ApoER2, a receptor in the Reelin signaling pathway, is a cargo of the AP-4 complex. We identify the motif ISSF/Y within the ApoER2 cytosolic domain as necessary for interaction with the canonical signal-binding pocket of the µ4 (AP4M1) subunit of AP-4. AP4E1- knock-out (KO) HeLa cells and hippocampal neurons from Ap4e1-KO mice display increased co-localization of ApoER2 with Golgi markers. Furthermore, hippocampal neurons from Ap4e1-KO mice and AP4M1-KO human iPSC-derived cortical i3Neurons exhibit reduced ApoER2 protein expression. Analyses of biosynthetic transport of ApoER2 reveal differential post-Golgi trafficking of the receptor, with lower axonal distribution in KO compared to wild-type neurons, indicating a role of AP-4 and the ISSF/Y motif in the axonal localization of ApoER2. Finally, analyses of Reelin signaling in mouse hippocampal and human cortical KO neurons show that AP4 deficiency causes no changes in Reelin-dependent activation of the AKT pathway and only mild changes in Reelin-induced dendritic arborization, but reduces Reelin-induced ERK phosphorylation, CREB activation, and Golgi deployment. This work thus establishes ApoER2 as a novel cargo of the AP-4 complex, suggesting that defects in the trafficking of this receptor and in the Reelin signaling pathway could contribute to the pathogenesis of HSP caused by mutations in AP-4 subunits.


Subject(s)
Adaptor Protein Complex 4 , LDL-Receptor Related Proteins , Spastic Paraplegia, Hereditary , Animals , Humans , Mice , Adaptor Protein Complex 4/genetics , Adaptor Protein Complex 4/metabolism , HeLa Cells , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Receptors, Cell Surface , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism
16.
Acta Neuropathol ; 147(1): 20, 2024 01 20.
Article in English | MEDLINE | ID: mdl-38244079

ABSTRACT

The SORL1 gene has recently emerged as a strong Alzheimer's Disease (AD) risk gene. Over 500 different variants have been identified in the gene and the contribution of individual variants to AD development and progression is still largely unknown. Here, we describe a family consisting of 2 parents and 5 offspring. Both parents were affected with dementia and one had confirmed AD pathology with an age of onset > 75 years. All offspring were affected with AD with ages at onset ranging from 53 years to 74 years. DNA was available from the parent with confirmed AD and 5 offspring. We identified a coding variant, p.(Arg953Cys), in SORL1 in 5 of 6 individuals affected by AD. Notably, variant carriers had severe AD pathology, and the SORL1 variant segregated with TDP-43 pathology (LATE-NC). We further characterized this variant and show that this Arginine substitution occurs at a critical position in the YWTD-domain of the SORL1 translation product, SORL1. Functional studies further show that the p.R953C variant leads to retention of the SORL1 protein in the endoplasmic reticulum which leads to decreased maturation and shedding of the receptor and prevents its normal endosomal trafficking. Together, our analysis suggests that p.R953C is a pathogenic variant of SORL1 and sheds light on mechanisms of how missense SORL1 variants may lead to AD.


Subject(s)
Alzheimer Disease , Humans , Aged , Alzheimer Disease/genetics , Gene Frequency , Genetic Predisposition to Disease , Membrane Transport Proteins/genetics , Mutation, Missense , LDL-Receptor Related Proteins/genetics , Polymorphism, Single Nucleotide
17.
Article in English | MEDLINE | ID: mdl-37849306

ABSTRACT

OBJECTIVE: In Norway, 89% of patients with Amyotrophic lateral sclerosis (ALS) lacks a genetic diagnose. ALS genes and genes that cause other neuromuscular or neurodegenerative disorders extensively overlap. This population-based study examined whether patients with ALS have a family history of neurological disorders and explored the occurrence of rare genetic variants associated with other neurodegenerative or neuromuscular disorders. METHODS: During a two-year period, blood samples and clinical data from patients with ALS were collected from all 17 neurological departments in Norway. Our genetic analysis involved exome sequencing and bioinformatics filtering of 510 genes associated with neurodegenerative and neuromuscular disorders. The variants were interpreted using genotype-phenotype correlations and bioinformatics tools. RESULTS: A total of 279 patients from a Norwegian population-based ALS cohort participated in this study. Thirty-one percent of the patients had first- or second-degree relatives with other neurodegenerative disorders, most commonly dementia and Parkinson's disease. The genetic analysis identified 20 possible pathogenic variants, in ATL3, AFG3L2, ATP7A, BICD2, HARS1, KIF1A, LRRK2, MSTO1, NEK1, NEFH, and SORL1, in 25 patients. NEK1 risk variants were present in 2.5% of this ALS cohort. Only four of the 25 patients reported relatives with other neurodegenerative or neuromuscular disorders. CONCLUSION: Gene variants known to cause other neurodegenerative or neuromuscular disorders, most frequently in NEK1, were identified in 9% of the patients with ALS. Most of these patients had no family history of other neurodegenerative or neuromuscular disorders. Our findings indicated that AFG3L2, ATP7A, BICD2, KIF1A, and MSTO1 should be further explored as potential ALS-causing genes.


Subject(s)
Amyotrophic Lateral Sclerosis , Cell Cycle Proteins , Neurodegenerative Diseases , Humans , Genetic Predisposition to Disease/genetics , Amyotrophic Lateral Sclerosis/epidemiology , Amyotrophic Lateral Sclerosis/genetics , Genetic Association Studies , Family , Neurodegenerative Diseases/epidemiology , Neurodegenerative Diseases/genetics , ATPases Associated with Diverse Cellular Activities/genetics , ATP-Dependent Proteases/genetics , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/genetics , Kinesins/genetics , Cytoskeletal Proteins/genetics
18.
Mol Genet Genomic Med ; 12(1): e2319, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38013226

ABSTRACT

BACKGROUND: Cenani-Lenzsyndactyly syndrome (CLSS; OMIM 212780) is a rare autosomal recessive acral deformity, which is mainly manifested in the fusion of fingers or toes, disordered phalangeal structure, shortening or fusion of the radius and ulna, and renal hypoplasia. CASE PRESENTATION: Our report described an individual with mild phenotypes from China. His parents were not consanguineous. The affected individual was non-dysmorphic. Standard X-ray showed that the both hands have only four metacarpal bones. The distal end of the first metacarpal bone on the right was relatively slender, and the distal phalanx was absent. Multiple phalanges and some soft tissues of both hands were fused. Exome sequencing revealed a novel biallelic c.282C⟩Avariant in low-density lipoprotein receptor-related protein 4 (LRP4; OMIM604270; NM_002334.4) causing p. (Asn94Lys) change in the encoded protein. This variant is predicted to be potentially pathogenic, affecting protein structure and function. CONCLUSION: We report a novel missense variant present in homozygosity in LRP4 to broaden the pathogenic spectrum of LRP4 in syndactyly, and exome sequencing technology is a powerful tool for genetic analysis in prenatal diagnosis and medical research, as a preferred method for the diagnosis of syndactyly and related phenotypes.


Subject(s)
LDL-Receptor Related Proteins , Syndactyly , Humans , Mutation , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Syndactyly/genetics , Syndactyly/diagnosis , Mutation, Missense
19.
Calcif Tissue Int ; 114(2): 171-181, 2024 02.
Article in English | MEDLINE | ID: mdl-38051321

ABSTRACT

Pathogenic variants disrupting the binding between sclerostin (encoded by SOST) and its receptor LRP4 have previously been described to cause sclerosteosis, a rare high bone mass disorder. The sclerostin-LRP4 complex inhibits canonical WNT signaling, a key pathway regulating osteoblastic bone formation and a promising therapeutic target for common bone disorders, such as osteoporosis. In the current study, we crossed mice deficient for Sost (Sost-/-) with our p.Arg1170Gln Lrp4 knock-in (Lrp4KI/KI) mouse model to create double mutant Sost-/-;Lrp4KI/KI mice. We compared the phenotype of Sost-/- mice with that of Sost-/-;Lrp4KI/KI mice, to investigate a possible synergistic effect of the disease-causing p.Arg1170Trp variant in Lrp4 on Sost deficiency. Interestingly, presence of Lrp4KI alleles partially mitigated the Sost-/- phenotype. Cellular and dynamic histomorphometry did not reveal mechanistic insights into the observed phenotypic differences. We therefore determined the molecular effect of the Lrp4KI allele by performing bulk RNA sequencing on Lrp4KI/KI primary osteoblasts. Unexpectedly, mostly genes related to bone resorption or remodeling (Acp5, Rankl, Mmp9) were upregulated in Lrp4KI/KI primary osteoblasts. Verification of these markers in Lrp4KI/KI, Sost-/- and Sost-/-;Lrp4KI/KI mice revealed that sclerostin deficiency counteracts this Lrp4KI/KI effect in Sost-/-;Lrp4KI/KI mice. We therefore hypothesize that models with two inactivating Lrp4KI alleles rather activate bone remodeling, with a net gain in bone mass, whereas sclerostin deficiency has more robust anabolic effects on bone formation. Moreover, these effects of sclerostin and Lrp4 are stronger in female mice, contributing to a more severe phenotype than in males and more detectable phenotypic differences among different genotypes.


Subject(s)
Adaptor Proteins, Signal Transducing , Bone Remodeling , Hyperostosis , Syndactyly , Male , Female , Animals , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Mice, Knockout , Phenotype , Mutation , Bone Remodeling/genetics , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism
20.
Eur J Med Genet ; 67: 104903, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38101565

ABSTRACT

LRP4 is expressed in many organs. It mediates SOST-dependent inhibition of bone formation and acts as an inhibitor of WNT signaling. It is also a postsynaptic end plate cell surface receptor at the neuromuscular junction and is central to its development, maintenance, and function. Pathogenic variants of LRP4 that specifically affect the canonical WNT signaling pathway are known to be associated with Cenani-Lenz syndactyly syndrome or the overlapping condition sclerosteosis. However, site-specific pathogenic variants of LRP4 have been associated with the congenital myasthenic syndrome (CMS) type 17 with no abnormal bone phenotype. Only two studies reported biallelic variants of LRP4 associated with CMS17 that presented during childhood. All three reported variants (NM_002334.4: p.Glu1233Ala, p.Glu1233Lys, or p.Arg1277His) are located within the 3'-edge of the third ß-propeller domain of LRP4. We report on a patient with a biallelic variant of the LRP4 gene presenting with a severe and neonatal lethal phenotype; we also provide a literature review of the previously reported patients. A female neonate, born to healthy consanguineous parents, presented with severe hypotonia, congenital diaphragmatic hernia, pulmonary hypertension, and progressive hypoxemia. Two of her siblings presented with a similar condition in the past, and all three died shortly after birth. Clinical exome sequencing revealed homozygosity for the pathogenic variant NM_002334.4:c.3698A > C (p.[Glu1233Ala]).


Subject(s)
Hyperostosis , Myasthenic Syndromes, Congenital , Syndactyly , Female , Humans , Infant, Newborn , LDL-Receptor Related Proteins/genetics , Myasthenic Syndromes, Congenital/genetics , Neuromuscular Junction , Syndactyly/genetics , Male
SELECTION OF CITATIONS
SEARCH DETAIL