Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
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
2.
Ann Rheum Dis ; 81(4): 544-555, 2022 04.
Article in English | MEDLINE | ID: mdl-34853001

ABSTRACT

OBJECTIVES: Dysregulated chondrocyte metabolism is closely associated with the pathogenesis of osteoarthritis (OA). Suppressing chondrocyte catabolism to restore cartilage homeostasis has been extensively explored, whereas far less effort has been invested toward enhancing chondrocyte anabolism. This study aimed to repurpose clinically approved drugs as potential stimulators of chondrocyte anabolism in treating OA. METHODS: Screening of a Food and Drug Administration-approved drug library; Assays for examining the chondroprotective effects of digoxin in vitro; Assays for defining the therapeutic effects of digoxin using a surgically-induced OA model; A propensity-score matched cohort study using The Health Improvement Network to examine the relationship between digoxin use and the risk of joint OA-associated replacement among patients with atrial fibrillation; identification and characterisation of the binding of digoxin to low-density lipoprotein receptor-related protein 4 (LRP4); various assays, including use of CRISPR-Cas9 genome editing to delete LRP4 in human chondrocytes, for examining the dependence on LRP4 of digoxin regulation of chondrocytes. RESULTS: Serial screenings led to the identification of ouabain and digoxin as stimulators of chondrocyte differentiation and anabolism. Ouabain and digoxin protected against OA and relieved OA-associated pain. The cohort study of 56 794 patients revealed that digoxin use was associated with reduced risk of OA-associated joint replacement. LRP4 was isolated as a novel target of digoxin, and deletion of LRP4 abolished digoxin's regulations of chondrocytes. CONCLUSIONS: These findings not only provide new insights into the understanding of digoxin's chondroprotective action and underlying mechanisms, but also present new evidence for repurposing digoxin for OA.


Subject(s)
Cartilage, Articular , Digoxin , LDL-Receptor Related Proteins , Osteoarthritis , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Cohort Studies , Digoxin/pharmacology , Drug Repositioning , Humans , LDL-Receptor Related Proteins/antagonists & inhibitors , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Ouabain/pharmacology
3.
Cell Rep ; 31(9): 107719, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32492427

ABSTRACT

SORL1/SORLA is a sorting receptor involved in retromer-related endosomal traffic and an Alzheimer's disease (AD) risk gene. Using CRISPR-Cas9, we deplete SORL1 in hiPSCs to ask if loss of SORL1 contributes to AD pathogenesis by endosome dysfunction. SORL1-deficient hiPSC neurons show early endosome enlargement, a hallmark cytopathology of AD. There is no effect of SORL1 depletion on endosome size in hiPSC microglia, suggesting a selective effect on neuronal endosomal trafficking. We validate defects in neuronal endosomal traffic by showing altered localization of amyloid precursor protein (APP) in early endosomes, a site of APP cleavage by the ß-secretase (BACE). Inhibition of BACE does not rescue endosome enlargement in SORL1-deficient neurons, suggesting that this phenotype is independent of amyloidogenic APP processing. Our data, together with recent findings, underscore how sporadic AD pathways regulating endosomal trafficking and autosomal-dominant AD pathways regulating APP cleavage independently converge on the defining cytopathology of AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Endosomes/metabolism , LDL-Receptor Related Proteins/metabolism , Membrane Transport Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , CRISPR-Cas Systems/genetics , Cell Differentiation , Cell Line , Gene Editing , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , LDL-Receptor Related Proteins/antagonists & inhibitors , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/genetics , Neurons/cytology , Neurons/metabolism , Protein Transport , RNA Interference , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Interfering/metabolism
5.
Stem Cell Res ; 20: 94-104, 2017 04.
Article in English | MEDLINE | ID: mdl-28340487

ABSTRACT

Understanding the regulatory networks underlying lineage differentiation and fate determination of human bone marrow stromal cells (hBMSC) is a prerequisite for their therapeutic use. The goal of the current study was to unravel the novel role of the low-density lipoprotein receptor-related protein 3 (LRP3) in regulating the osteogenic and adipogenic differentiation of immortalized hBMSCs. Gene expression profiling revealed significantly higher LRP3 levels in the highly osteogenic hBMSC clone imCL1 than in the less osteogenic clone imCL2, as well as a significant upregulation of LRP3 during the osteogenic induction of the imCL1 clone. Data from functional and gene expression assays demonstrated the role of LRP3 as a molecular switch promoting hBMSC lineage differentiation into osteoblasts and inhibiting differentiation into adipocytes. Interestingly, microRNA (miRNA) expression profiling identified miR-4739 as the most under-represented miRNA (-36.11 fold) in imCL1 compared to imCL2. The TargetScan prediction algorithm, combined with functional and biochemical assays, identified LRP3 mRNA as a novel target of miR-4739, with a single potential binding site for miR-4739 located in the LRP3 3' UTR. Regulation of LRP3 expression by miR-4739 was subsequently confirmed by qRT-PCR, western blotting, and luciferase assays. Over-expression of miR-4739 mimicked the effects of LRP3 knockdown on promoting adipogenic and suppressing osteogenic differentiation of hBMSCs. Hence, we report for the first time a novel biological role for the LRP3/hsa-miR-4739 axis in balancing osteogenic and adipocytic differentiation of hBMSCs. Our data support the potential utilization of miRNA-based therapies in regenerative medicine.


Subject(s)
Adipocytes/metabolism , Bone Marrow Cells/cytology , LDL-Receptor Related Proteins/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Osteoblasts/metabolism , 3' Untranslated Regions , Adipocytes/cytology , Adipogenesis , Antagomirs/metabolism , Base Sequence , Cell Differentiation , Cell Survival , Cells, Cultured , Gene Expression Profiling , Humans , LDL-Receptor Related Proteins/antagonists & inhibitors , LDL-Receptor Related Proteins/genetics , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Osteoblasts/cytology , Osteogenesis , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Sequence Alignment
6.
J Autoimmun ; 48-49: 143-8, 2014.
Article in English | MEDLINE | ID: mdl-24530233

ABSTRACT

Myasthenia gravis is characterized by muscle weakness and abnormal fatigability. It is an autoimmune disease caused by the presence of antibodies against components of the muscle membrane localized at the neuromuscular junction. In most cases, the autoantibodies are against the acetylcholine receptor (AChR). Recently, other targets have been described such as the MuSK protein (muscle-specific kinase) or the LRP4 (lipoprotein related protein 4). Myasthenia gravis can be classified according to the profile of the autoantibodies, the location of the affected muscles (ocular versus generalized), the age of onset of symptoms and thymic abnormalities. The disease generally begins with ocular symptoms (ptosis and/or diplopia) and extends to other muscles in 80% of cases. Other features that characterize MG include the following: variability, effort induced worsening, successive periods of exacerbation during the course of the disease, severity dependent on respiratory and swallowing impairment (if rapid worsening occurs, a myasthenic crisis is suspected), and an association with thymoma in 20% of patients and with other autoimmune diseases such as hyperthyroidism and Hashimoto's disease. The diagnosis is based on the clinical features, the benefit of the cholinesterase inhibitors, the detection of specific autoantibodies (anti-AChR, anti-MuSK or anti-LRP4), and significant decrement evidenced by electrophysiological tests. In this review, we briefly describe the history and epidemiology of the disease and the diagnostic and clinical classification. The neonatal form of myasthenia is explained, and finally we discuss the main difficulties of diagnosis.


Subject(s)
Autoimmune Diseases of the Nervous System/classification , Autoimmune Diseases of the Nervous System/diagnosis , Myasthenia Gravis/classification , Myasthenia Gravis/diagnosis , Animals , Autoantibodies/biosynthesis , Autoantibodies/classification , Autoimmune Diseases of the Nervous System/epidemiology , Autoimmune Diseases of the Nervous System/immunology , Fetal Diseases/classification , Fetal Diseases/diagnosis , Fetal Diseases/immunology , Humans , Infant, Newborn , Infant, Newborn, Diseases/classification , Infant, Newborn, Diseases/diagnosis , Infant, Newborn, Diseases/immunology , LDL-Receptor Related Proteins/antagonists & inhibitors , LDL-Receptor Related Proteins/immunology , Myasthenia Gravis/epidemiology , Myasthenia Gravis/immunology , Neuromuscular Junction/immunology , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Cholinergic/immunology
7.
FASEB J ; 28(4): 1543-54, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24344333

ABSTRACT

The reelin signaling protein and its downstream components have been associated with synaptic plasticity and neurotransmission. The reelin signaling pathway begins with the binding of reelin to the transmembrane lipoprotein receptor apolipoprotein E receptor 2 (ApoER2), which in turns induces the sequential cleavage of ApoER2 by the sequential action of α- and γ-secretases. Using conditional-knockout mice of the catalytic component of the γ-secretase complex, presenilin 1 (PS1), we demonstrated increased brain ApoER2 and reelin protein and transcript levels, with no changes in the number of reelin-positive cells. Using the human SH-SY5Y neuroblastoma cell line, we showed that ApoER2 processing occurs in the presence of PS1, producing an intracellular ApoER2 C-terminal fragment. In addition, the pharmacologic inhibition of γ-secretase in SH-SY5Y cells led to increased reelin levels. Overexpression of ApoER2 decreased reelin mRNA levels in these cells. A luciferase reporter gene assay and nuclear fractionation confirmed that increased amounts of intracellular fragment of ApoER2 suppressed reelin expression at a transcriptional level. Chromatin immunoprecipitation experiments corroborated that the intracellular fragment of ApoER2 bound to the RELN promoter region. Our study suggests that PS1/γ-secretase-dependent processing of the reelin receptor ApoER2 inhibits reelin expression and may regulate its signaling.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Extracellular Matrix Proteins/metabolism , LDL-Receptor Related Proteins/metabolism , Nerve Tissue Proteins/metabolism , Presenilin-1/metabolism , Serine Endopeptidases/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Blotting, Western , Cell Adhesion Molecules, Neuronal/genetics , Cell Line, Tumor , Dipeptides/pharmacology , Extracellular Matrix Proteins/genetics , Female , Gene Expression Regulation , HEK293 Cells , Humans , LDL-Receptor Related Proteins/antagonists & inhibitors , LDL-Receptor Related Proteins/genetics , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Nerve Tissue Proteins/genetics , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Presenilin-1/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Reelin Protein , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases/genetics , Signal Transduction/genetics
8.
Proc Natl Acad Sci U S A ; 110(51): 20783-8, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24297891

ABSTRACT

Myasthenia gravis (MG) is a severely debilitating autoimmune disease that is due to a decrease in the efficiency of synaptic transmission at neuromuscular synapses. MG is caused by antibodies against postsynaptic proteins, including (i) acetylcholine receptors, the neurotransmitter receptor, (ii) muscle-specific kinase (MuSK), a receptor tyrosine kinase essential for the formation and maintenance of neuromuscular synapses, and (iii) low-density lipoprotein receptor-related protein 4 (Lrp4), which responds to neural Agrin by binding and stimulating MuSK. Passive transfer studies in mice have shown that IgG4 antibodies from MuSK MG patients cause disease without requiring complement or other immune components, suggesting that these MuSK antibodies cause disease by directly interfering with MuSK function. Here we show that pathogenic IgG4 antibodies to MuSK bind to a structural epitope in the first Ig-like domain of MuSK, prevent binding between MuSK and Lrp4, and inhibit Agrin-stimulated MuSK phosphorylation. In contrast, these IgG4 antibodies have no direct effect on MuSK dimerization or MuSK internalization. These results provide insight into the unique pathogenesis of MuSK MG and provide clues toward development of specific treatment options.


Subject(s)
Autoantibodies/immunology , Immunoglobulin G/immunology , LDL-Receptor Related Proteins/immunology , Myasthenia Gravis/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Cholinergic/immunology , Receptors, LDL/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Agrin/immunology , Animals , Autoantibodies/pharmacology , Cell Line , Child , Child, Preschool , Epitopes/immunology , Female , Humans , Immunization, Passive , Immunoglobulin G/pharmacology , LDL-Receptor Related Proteins/antagonists & inhibitors , Male , Mice , Middle Aged , Myasthenia Gravis/chemically induced , Myasthenia Gravis/pathology , Phosphorylation/drug effects , Phosphorylation/immunology , Protein Multimerization/drug effects , Protein Multimerization/immunology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, LDL/antagonists & inhibitors
9.
Mol Cell Neurosci ; 54: 22-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23267846

ABSTRACT

Human immunodeficiency virus (HIV) infection of the CNS produces dendritic damage that correlates with cognitive decline in patients with HIV-associated neurocognitive disorders (HAND). HIV-induced neurotoxicity results in part from viral proteins shed from infected cells, including the HIV transactivator of transcription (Tat). We previously showed that Tat binds to the low density lipoprotein receptor-related protein (LRP), resulting in overactivation of NMDA receptors, activation of the ubiquitin-proteasome pathway, and subsequent loss of postsynaptic densities. Here, we show that Tat also induces a loss of presynaptic terminals. The number of presynaptic terminals was quantified using confocal imaging of synaptophysin fused to green fluorescent protein (Syn-GFP). Tat-induced loss of presynaptic terminals was secondary to excitatory postsynaptic mechanisms because treatment with an LRP antagonist or an NMDA receptor antagonist inhibited this loss. Treatment with nutlin-3, an E3 ligase inhibitor, prevented Tat-induced loss of presynaptic terminals. These data suggest that Tat-induced loss of presynaptic terminals is a consequence of excitotoxic postsynaptic activity. We previously found that ifenprodil, an NR2B subunit-selective NMDA receptor antagonist, induced recovery of postsynaptic densities. Here we show that Tat-induced loss of presynaptic terminals was reversed by ifenprodil treatment. Thus, Tat-induced loss of presynaptic terminals is reversible, and this recovery can be initiated by inhibiting a subset of postsynaptic NMDA receptors. Understanding the dynamics of synaptic changes in response to HIV infection of the CNS may lead to the design of improved pharmacotherapies for HAND patients.


Subject(s)
Hippocampus/cytology , Neurons/drug effects , Presynaptic Terminals/drug effects , tat Gene Products, Human Immunodeficiency Virus/toxicity , Animals , Cells, Cultured , HIV-1 , Hippocampus/drug effects , Imidazoles/pharmacology , LDL-Receptor Related Proteins/antagonists & inhibitors , Neurons/metabolism , Piperazines/pharmacology , Piperidines/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synaptophysin/genetics , Synaptophysin/metabolism
10.
J Neuroinflammation ; 9: 196, 2012 Aug 13.
Article in English | MEDLINE | ID: mdl-22889139

ABSTRACT

BACKGROUND: Microglia are resident brain macrophages that can phagocytose dead, dying or viable neurons, which may be beneficial or detrimental in inflammatory, ischaemic and neurodegenerative brain pathologies. Cell death caused by phagocytosis of an otherwise viable cell is called 'primary phagocytosis' or 'phagoptosis'. Calreticulin (CRT) exposure on the surface of cancer cells can promote their phagocytosis via LRP (low-density lipoprotein receptor-related protein) on macrophages, but it is not known whether this occurs with neurons and microglia. METHODS: We used primary cultures of cerebellar neurons, astrocytes and microglia to investigate the potential role of CRT/LRP phagocytic signalling in the phagocytosis of viable neurons by microglia stimulated with lipopolysaccharide (LPS) or nanomolar concentrations of amyloid-ß peptide1-42 (Aß). Exposure of CRT on the neuronal surface was investigated using surface biotinylation and western blotting. A phagocytosis assay was also developed using BV2 and PC12 cell lines to investigate CRT/LRP signalling in microglial phagocytosis of apoptotic cells. RESULTS: We found that BV2 microglia readily phagocytosed apoptotic PC12 cells, but this was inhibited by a CRT-blocking antibody or LRP-blocking protein (receptor-associated protein: RAP). Activation of primary rat microglia with LPS or Aß resulted in loss of co-cultured cerebellar granule neurons, and this was blocked by RAP or antibodies against CRT or against LRP, preventing all neuronal loss and death. CRT was present on the surface of viable neurons, and this exposure did not change in inflammatory conditions. CRT antibodies prevented microglia-induced neuronal loss when added to neurons, while LRP antibodies prevented neuronal loss when added to the microglia. Pre-binding of CRT to neurons promoted neuronal loss if activated microglia were added, but pre-binding of CRT to microglia or both cell types prevented microglia-induced neuronal loss. CONCLUSIONS: CRT exposure on the surface of viable or apoptotic neurons appears to be required for their phagocytosis via LRP receptors on activated microglia, but free CRT can block microglial phagocytosis of neurons by acting on microglia. Phagocytosis of CRT-exposing neurons by microglia can be a direct cause of neuronal death during inflammation, and might therefore contribute to neurodegeneration and be prevented by blocking the CRT/LRP pathway.


Subject(s)
Amyloid beta-Peptides/toxicity , Calreticulin/physiology , LDL-Receptor Related Proteins/physiology , Lipopolysaccharides/toxicity , Microglia/physiology , Neurons/physiology , Peptide Fragments/toxicity , Phagocytosis/physiology , Signal Transduction/physiology , Animals , Cell Survival/physiology , Cells, Cultured , Coculture Techniques , LDL-Receptor Related Proteins/antagonists & inhibitors , PC12 Cells , Rats
11.
Biochem J ; 436(2): 263-9, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21375506

ABSTRACT

Mest (mesoderm-specific transcript)/Peg1 (paternally expressed gene 1) is an imprinted gene that plays important roles in embryo development, although its biochemical role has not been determined. Ectopic expression of Mest/Peg1 inhibited Wnt-mediated reporter activity by enhancing the ubiquitination of ß-catenin. The maturation and plasma membrane localization of the Wnt co-receptor LRP6 [LDLR (low-density lipoprotein receptor)-related protein 6], which are both necessary for Wnt signalling, were blocked by the expression of Mest/Peg1. Mest/Peg1 inhibited maturation of LRP6 by controlling the glycosylation of LRP6. Knockdown of Mest/Peg1, which might enhance Wnt signalling, blocked adipogenic differentiation of 3T3-L1 cells. Overall, our results suggest that Mest/Peg1 is a novel regulator of Wnt/ß-catenin signalling during adipogenic differentiation.


Subject(s)
LDL-Receptor Related Proteins/metabolism , Proteins/physiology , Signal Transduction/physiology , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/physiology , 3T3-L1 Cells , Adipogenesis/physiology , Animals , Glycosylation , HEK293 Cells , Humans , LDL-Receptor Related Proteins/antagonists & inhibitors , LDL-Receptor Related Proteins/biosynthesis , Low Density Lipoprotein Receptor-Related Protein-6 , Mice
12.
Sci Signal ; 4(164): ra15, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21406690

ABSTRACT

Ligand binding to certain heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) stimulates the rapid synthesis of cyclic adenosine monophosphate (cAMP) through the G protein α(s) subunit, which activates adenylyl cyclase (AC). We found that the transmembrane receptor low-density lipoprotein receptor-related protein 6 (LRP6), a co-receptor for Wnt proteins, bound to the Gα(s)ßγ heterotrimer and that knockdown of LRP6 attenuated cAMP production by various GPCRs, including parathyroid hormone receptor 1 (PTH1R). Knockdown of LRP6 disrupted the localization of Gα(s) to the plasma membrane, which led to a decrease in the extent of coupling of Gα(s) to PTH1R and inhibited the production of cAMP and the activation of cAMP-dependent protein kinase (PKA) in response to PTH. PKA phosphorylated LRP6, which enhanced the binding of Gα(s) to LRP6, its localization to the plasma membrane, and the production of cAMP in response to PTH. Decreased PTH-dependent cAMP production was observed in single cells in which LRP6 was knocked down or mutated at the PKA site by monitoring the cAMP kinetics. Thus, we suggest that the binding of Gα(s) to LRP6 is required to establish a functional GPCR-Gα(s)-AC signaling pathway for the production of cAMP, providing an additional regulatory component to the current GPCR-cAMP paradigm.


Subject(s)
Cyclic AMP/biosynthesis , GTP-Binding Protein alpha Subunits, Gs/metabolism , LDL-Receptor Related Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Knockdown Techniques , Humans , LDL-Receptor Related Proteins/antagonists & inhibitors , LDL-Receptor Related Proteins/genetics , Ligands , Low Density Lipoprotein Receptor-Related Protein-5 , Low Density Lipoprotein Receptor-Related Protein-6 , Mice , Models, Biological , Molecular Sequence Data , Phosphorylation , RNA, Small Interfering/genetics , Rats , Receptor, Parathyroid Hormone, Type 1/metabolism , Second Messenger Systems , Signal Transduction
13.
Eur J Haematol ; 86(6): 453-65, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21342268

ABSTRACT

Wnt signaling is crucial for cell proliferation and differentiation. It represents a complex network with mechanisms of self-regulation through positive and negative feedback. Recent increasing interest in this signaling pathway has led to the discovery of many new proteins that down-regulate Wnt activity. Here, we provide a short description of the most important and best-studied inhibitors, group them according to the target molecule within the Wnt cascade, and discuss their clinical potential. Although most of the inhibitors discussed here may also interact with proteins from other signaling pathways, we focus only on their ability to modulate Wnt signaling.


Subject(s)
Signal Transduction/physiology , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/physiology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/physiology , Animals , Dishevelled Proteins , Humans , LDL-Receptor Related Proteins/antagonists & inhibitors , LDL-Receptor Related Proteins/physiology , Ligands , Models, Biological , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/physiology , beta Catenin/antagonists & inhibitors , beta Catenin/physiology
14.
J Clin Invest ; 121(1): 120-31, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21123944

ABSTRACT

In antiphospholipid syndrome (APS), antiphospholipid antibodies (aPL) binding to ß2 glycoprotein I (ß2GPI) induce endothelial cell-leukocyte adhesion and thrombus formation via unknown mechanisms. Here we show that in mice both of these processes are caused by the inhibition of eNOS. In studies of cultured human, bovine, and mouse endothelial cells, the promotion of monocyte adhesion by aPL entailed decreased bioavailable NO, and aPL fully antagonized eNOS activation by diverse agonists. Similarly, NO-dependent, acetylcholine-induced increases in carotid vascular conductance were impaired in aPL-treated mice. The inhibition of eNOS was caused by antibody recognition of domain I of ß2GPI and ß2GPI dimerization, and it was due to attenuated eNOS S1179 phosphorylation mediated by protein phosphatase 2A (PP2A). Furthermore, LDL receptor family member antagonism with receptor-associated protein (RAP) prevented aPL inhibition of eNOS in cell culture, and ApoER2-/- mice were protected from aPL inhibition of eNOS in vivo. Moreover, both aPL-induced increases in leukocyte-endothelial cell adhesion and thrombus formation were absent in eNOS-/- and in ApoER2-/- mice. Thus, aPL-induced leukocyte-endothelial cell adhesion and thrombosis are caused by eNOS antagonism, which is due to impaired S1179 phosphorylation mediated by ß2GPI, apoER2, and PP2A. Our results suggest that novel therapies for APS can now be developed targeting these mechanisms.


Subject(s)
Antibodies, Antiphospholipid/metabolism , Endothelial Cells/immunology , Leukocytes/immunology , Thrombosis/immunology , Animals , Cattle , Cell Adhesion/immunology , Cell Adhesion/physiology , Dimerization , Endothelial Cells/metabolism , Humans , In Vitro Techniques , LDL-Receptor Related Proteins/antagonists & inhibitors , LDL-Receptor Related Proteins/deficiency , LDL-Receptor Related Proteins/immunology , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Nitric Oxide/physiology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/immunology , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Structure, Tertiary , RNA, Small Interfering/genetics , Thrombosis/etiology , beta 2-Glycoprotein I/antagonists & inhibitors , beta 2-Glycoprotein I/chemistry , beta 2-Glycoprotein I/immunology
15.
J Pharmacol Sci ; 113(3): 203-7, 2010.
Article in English | MEDLINE | ID: mdl-20595786

ABSTRACT

Tissue-type plasminogen activator (t-PA) administration has been approved for treating acute ischemic stroke, but delayed treatment is associated with increased risk of cerebral hemorrhage and brain injury. t-PA, a serine proteinase, converts plasminogen to plasmin. Plasmin participates not only in the degradation of fibrin, causing clot lysis, but also in the degradation of various extracellular matrix proteins, either directly or via the activation of matrix metalloproteinase (MMPs). We established an animal stroke model and observed a phenomenon of spontaneous rethrombosis and thrombolysis in the cerebral vessels after vessel damage. Endogenous t-PA protected brain damage by recanalization, but the protective effect deteriorated when the occluded vessels were not reopened. On studying intracranial hemorrhage (ICH) induced by t-PA treatment of ischemic stroke, we observed that MMP-3 is relatively important for the enhanced ICH induced by t-PA. MMP-3 was upregulated by t-PA in endothelial cells, but the upregulation was prevented by the inhibition of either low-density lipoprotein receptor-related protein (LRP) or nuclear factor kappa-B (NF-kappaB) activation. Thus, t-PA causes ICH via MMP-3 induction in endothelial cells, which is regulated through the LRP/NF-kappaB pathway, and could be targeted to improve the therapeutic efficacy of t-PA for acute ischemic stroke.


Subject(s)
Brain Ischemia/drug therapy , Fibrinolytic Agents/adverse effects , Fibrinolytic Agents/therapeutic use , Stroke/drug therapy , Tissue Plasminogen Activator/adverse effects , Tissue Plasminogen Activator/therapeutic use , Animals , Antifibrinolytic Agents/pharmacology , Brain Ischemia/metabolism , Drug Design , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibrinolytic Agents/pharmacology , Humans , Intracranial Hemorrhages/chemically induced , Intracranial Hemorrhages/drug therapy , Intracranial Hemorrhages/metabolism , LDL-Receptor Related Proteins/antagonists & inhibitors , Matrix Metalloproteinase 3/metabolism , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Stroke/metabolism , Tissue Plasminogen Activator/pharmacology , Up-Regulation/drug effects
16.
Biochemistry ; 49(22): 4635-43, 2010 Jun 08.
Article in English | MEDLINE | ID: mdl-20446724

ABSTRACT

Mesd is a specialized chaperone for low-density lipoprotein receptor-related protein 5 (LRP5) and LRP6. In our previous studies, we found that Mesd binds to mature LRP6 on the cell surface and blocks the binding of Wnt antagonist Dickkopf-1 (Dkk1) to LRP6. Herein, we demonstrate that Mesd also binds to LRP5 with a high affinity and is a universal inhibitor of LRP5 and LRP6 ligands. Mesd not only blocks binding of Wnt antagonists Dkk1 and Sclerostin to LRP5 and LRP6 but also inhibits Wnt3A and Rspondin1-induced Wnt/beta-catenin signaling in LRP5- and LRP6-expressing cells. We also found that Mesd, Dkk1, and Sclerostin compete with one another for binding to LRP5 and LRP6 at the cell surface. More importantly, we demonstrated that Mesd is able to suppress LRP6 phosphorylation and Wnt/beta-catenin signaling in prostate cancer PC-3 cells and inhibits PC-3 cell proliferation. Our results indicate that recombinant Mesd protein is a useful tool for studying Wnt/beta-catenin signaling on the cell surface and has a potential therapeutic role in Wnt-dependent cancers.


Subject(s)
Antineoplastic Agents , Growth Inhibitors/physiology , LDL-Receptor Related Proteins/antagonists & inhibitors , Molecular Chaperones/physiology , Signal Transduction/physiology , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents/pharmacology , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/metabolism , CHO Cells , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cricetinae , Cricetulus , Genetic Markers , Humans , Intercellular Signaling Peptides and Proteins/metabolism , LDL-Receptor Related Proteins/biosynthesis , LDL-Receptor Related Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5 , Low Density Lipoprotein Receptor-Related Protein-6 , Mice , Molecular Chaperones/metabolism , Protein Binding , Wnt Proteins/physiology , beta Catenin/physiology
17.
Biorheology ; 46(5): 389-99, 2009.
Article in English | MEDLINE | ID: mdl-19940355

ABSTRACT

The anabolic effect of dynamic mechanical loading on skeletal architecture has been repeatedly demonstrated, but the cellular and molecular events occurring between load and ultimate bone formation remain obscure. The discovery of sclerostin, an antagonist of Wnt/Lrp5 signaling, and the sclerosing bone dysplasias that result from its mutation suggest its pivotal role in modulating bone formation. We examined expression of Sost mRNA across a variety of clonal cell lines spanning the osteogenic phenotype from immature osteoblast to mature osteocyte. No sclerostin expression was detected in immature MC3T3-E1 osteoblasts and, surprisingly, mature MLO-Y4 osteocytes, whereas immature MLO-A5 osteocytic cells expressed very low levels of Sost. Highest expression was observed in mature UMR 106.01 osteoblasts. We examined the influence of bone morphogenetic proteins on Sost expression. Treatment with BMP-2, -4 or -6 was without effect on Sost in mature MLO-Y4 osteocytes but elicited a robust increase in Sost expression in immature MLO-A5 osteocytes. Oscillatory fluid flow applied to mature UMR 106.01 osteoblasts transiently decreased expression of sclerostin at both the mRNA and protein level. Overall, our results indicate that BMP treatment and in vitro mechanical loading demonstrate opposite effects upon sclerostin expression.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Osteoblasts/metabolism , Osteocytes/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Protein 6/pharmacology , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Line , Genetic Markers/genetics , Glycoproteins , Humans , Intercellular Signaling Peptides and Proteins , LDL-Receptor Related Proteins/antagonists & inhibitors , LDL-Receptor Related Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5 , Mice , Signal Transduction , Stress, Mechanical , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism
18.
Cell Microbiol ; 10(12): 2509-19, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18717822

ABSTRACT

To exert its activity, anthrax toxin must be endocytosed and its enzymatic toxic subunits delivered to the cytoplasm. It has been proposed that, in addition to the anthrax toxin receptors (ATRs), lipoprotein-receptor-related protein 6 (LRP6), known for its role in Wnt signalling, is also required for toxin endocytosis. These findings have however been challenged. We show that LRP6 can indeed form a complex with ATRs, and that this interaction plays a role both in Wnt signalling and in anthrax toxin endocytosis. We found that ATRs control the levels of LRP6 in cells, and thus the Wnt signalling capacity. RNAi against ATRs indeed led to a drastic decrease in LRP6 levels and a subsequent drop in Wnt signalling. Conversely, LRP6 plays a role in anthrax toxin endocytosis, but is not essential. We indeed found that toxin binding triggered tyrosine phosphorylation of LRP6, induced its redistribution into detergent-resistant domains, and its subsequent endocytosis. RNAis against LRP6 strongly delayed toxin endocytosis. As the physiological role of ATRs is probably to interact with the extracellular matrix, our findings raise the interesting possibility that, through the ATR-LRP6 interaction, adhesion to the extracellular matrix could locally control Wnt signalling.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Endocytosis , LDL-Receptor Related Proteins/metabolism , Receptors, Peptide/metabolism , Gene Silencing , HeLa Cells , Humans , LDL-Receptor Related Proteins/antagonists & inhibitors , Low Density Lipoprotein Receptor-Related Protein-6 , Protein Binding , Receptors, Peptide/antagonists & inhibitors
19.
Mol Cell Biol ; 28(7): 2368-79, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18212053

ABSTRACT

Recombinant Wnt-3a stimulated the rapid formation of elongated processes in Ewing sarcoma family tumor (ESFT) cells that were identified as neurites. The processes stained positively for polymerized actin and microtubules as well as synapsin I and growth-associated protein 43. Inhibition of the Wnt receptor, Frizzled3 (Fzd3), with antiserum or by short interfering RNA (siRNA) markedly reduced neurite extension. Knockdown of Dishevelled-2 (Dvl-2) and Dvl-3 also suppressed neurite outgrowth. Surprisingly, disruption of the Wnt/Fzd/lipoprotein receptor-related protein (LRP) complex and the associated beta-catenin signaling by treating cells either with the Wnt antagonist Dickkopf-1 (Dkk1) or LRP5/LRP6 siRNA enhanced neuritogenesis. Neurite outgrowth induced by Dkk1 or with LRP5/LRP6 siRNA was inhibited by secreted Fzd-related protein 1, a Wnt antagonist that binds directly to Wnt. Moreover, Dkk1 stimulation of neurite outgrowth was blocked by Fzd3 siRNA. These results suggested that Dkk1 shifted endogenous Wnt activity from the beta-catenin pathway to Fzd3-mediated, noncanonical signaling that is responsible for neurite formation. In particular, c-Jun amino-terminal kinase (JNK) was important for neurite outgrowth stimulated by both Wnt-3a and Dkk1. Our data demonstrate that Fzd3, Dvl, and JNK activity mediate Wnt-dependent neurite outgrowth and that ESFT cell lines will be useful experimental models for the study of Wnt-dependent neurite extension.


Subject(s)
Frizzled Receptors/physiology , Intercellular Signaling Peptides and Proteins/physiology , JNK Mitogen-Activated Protein Kinases/physiology , Neoplasm Proteins/physiology , Neurites/physiology , Receptors, G-Protein-Coupled/physiology , Sarcoma, Ewing/ultrastructure , Wnt Proteins/physiology , Adaptor Proteins, Signal Transducing/physiology , Cell Line, Tumor/drug effects , Cell Line, Tumor/ultrastructure , Dishevelled Proteins , Frizzled Receptors/analysis , Humans , Intercellular Signaling Peptides and Proteins/genetics , LDL-Receptor Related Proteins/antagonists & inhibitors , LDL-Receptor Related Proteins/physiology , Low Density Lipoprotein Receptor-Related Protein-5 , Low Density Lipoprotein Receptor-Related Protein-6 , Membrane Proteins/genetics , Membrane Proteins/physiology , Neoplasm Proteins/genetics , Neurites/drug effects , Phosphoproteins/physiology , RNA, Small Interfering/pharmacology , Receptors, G-Protein-Coupled/analysis , Recombinant Fusion Proteins/physiology , Wnt Proteins/genetics , Wnt Proteins/pharmacology , Wnt1 Protein/physiology , Wnt3 Protein , Wnt3A Protein
20.
Proc Natl Acad Sci U S A ; 104(37): 14700-5, 2007 Sep 11.
Article in English | MEDLINE | ID: mdl-17804805

ABSTRACT

The R-Spondin (RSpo) family of secreted proteins act as potent activators of the Wnt/beta-catenin signaling pathway. We have previously shown that RSpo proteins can induce proliferative effects on the gastrointestinal epithelium in mice. Here we provide a mechanism whereby RSpo1 regulates cellular responsiveness to Wnt ligands by modulating the cell-surface levels of the coreceptor LRP6. We show that RSpo1 activity critically depends on the presence of canonical Wnt ligands and LRP6. Although RSpo1 does not directly activate LRP6, it interferes with DKK1/Kremen-mediated internalization of LRP6 through an interaction with Kremen, resulting in increased LRP6 levels on the cell surface. Our results support a model in which RSpo1 relieves the inhibition DKK1 imposes on the Wnt pathway.


Subject(s)
LDL-Receptor Related Proteins/antagonists & inhibitors , Signal Transduction , Thrombospondins/metabolism , Wnt Proteins/metabolism , Animals , Cell Line , Drosophila/cytology , Drosophila/metabolism , Gene Expression Regulation , Genes, Reporter , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Kidney/cytology , LDL-Receptor Related Proteins/metabolism , Ligands , Low Density Lipoprotein Receptor-Related Protein-6 , Luciferases/metabolism , Membrane Proteins/metabolism , Models, Biological , Phosphorylation , Precipitin Tests , Protein Binding , RNA, Small Interfering/metabolism , Recombinant Proteins/metabolism , Thrombospondins/genetics , Transfection , beta Catenin/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...