Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 461
Filter
1.
Int J Mol Sci ; 23(9)2022 May 09.
Article in English | MEDLINE | ID: mdl-35563651

ABSTRACT

The fibrinolytic system is composed of the protease plasmin, its precursor plasminogen and their respective activators, tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA), counteracted by their inhibitors, plasminogen activator inhibitor type 1 (PAI-1), plasminogen activator inhibitor type 2 (PAI-2), protein C inhibitor (PCI), thrombin activable fibrinolysis inhibitor (TAFI), protease nexin 1 (PN-1) and neuroserpin. The action of plasmin is counteracted by α2-antiplasmin, α2-macroglobulin, TAFI, and other serine protease inhibitors (antithrombin and α2-antitrypsin) and PN-1 (protease nexin 1). These components are essential regulators of many physiologic processes. They are also involved in the pathogenesis of many disorders. Recent advancements in our understanding of these processes enable the opportunity of drug development in treating many of these disorders.


Subject(s)
Fibrinolysin , Fibrinolysis , Fibrinolysin/metabolism , Fibrinolysis/physiology , Plasminogen/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Protease Nexins , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , alpha-2-Antiplasmin
2.
Semin Cell Dev Biol ; 62: 160-169, 2017 02.
Article in English | MEDLINE | ID: mdl-27545616

ABSTRACT

Cultures of glial cells and fibroblasts allowed and lead to the identification SERPINE2/Protease Nexin-1 (SERPINE2/PN-1). Cellular, biochemical, immunological and molecular characterization substantiated its variable expression in many organs as a function of development, adult stages, pathological situations or following injury. It is not a circulating serpin, but as other members of the family, its target specificity is influenced by components of the extracellular matrix. The challenges are to identify where and when SERPINE2/PN-1 modulatory action becomes crucial or even possibly specific in a mosaic of feasible in vivo impacts. Data providing correlations are not sufficient to satisfy this aim. Genetically modified mice, or tissue derived thereof, provide interesting in vivo models to identify and study the relevance of this serpin. This review will highlight sometimes-intriguing results indicating a crucial impact of SERPINE2/PN-1, especially in the vasculature, the nervous system or the behavior of cancer cells in vivo. Data presently available will be discussed in an attempt to define general trends in the diversity of SERPINE2/PN-1 modes of action in vivo.


Subject(s)
Protease Nexins/metabolism , Serpin E2/metabolism , Animals , Enzyme Inhibitors/metabolism , Glycosaminoglycans/metabolism , Humans , Ligands , Receptors, Cell Surface/metabolism
3.
Am J Hum Genet ; 97(4): 546-54, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26387594

ABSTRACT

Multiciliated epithelial cells protect the upper and lower airways from chronic bacterial infections by moving mucus and debris outward. Congenital disorders of ciliary beating, referred to as primary ciliary dyskinesia (PCD), are characterized by deficient mucociliary clearance and severe, recurrent respiratory infections. Numerous genetic defects, most of which can be detected by transmission electron microscopy (TEM), are so far known to cause different abnormalities of the ciliary axoneme. However, some defects are not regularly discernable by TEM because the ciliary architecture of the axoneme remains preserved. This applies in particular to isolated defects of the nexin links, also known as the nexin-dynein regulatory complex (N-DRC), connecting the peripheral outer microtubular doublets. Immunofluorescence analyses of respiratory cells from PCD-affected individuals detected a N-DRC defect. Genome-wide exome sequence analyses identified recessive loss-of-function mutations in GAS8 encoding DRC4 in three independent PCD-affected families.


Subject(s)
Cytoskeletal Proteins/genetics , Dyneins/antagonists & inhibitors , Kartagener Syndrome/etiology , Multiprotein Complexes/antagonists & inhibitors , Mutation/genetics , Neoplasm Proteins/genetics , Protease Nexins/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Adult , Animals , Blotting, Western , Child , Cilia/physiology , Dyneins/genetics , Exome/genetics , Female , Humans , Intracellular Signaling Peptides and Proteins/physiology , Kartagener Syndrome/pathology , Male , Membrane Proteins , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Multiprotein Complexes/genetics , Nasal Mucosa/cytology , Nasal Mucosa/metabolism , Nitric Oxide/analysis , Pedigree , Phenotype , Prognosis , Protease Nexins/genetics , Respiratory System , Young Adult
4.
Nat Genet ; 39(2): 168-77, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17220890

ABSTRACT

The recycling of the amyloid precursor protein (APP) from the cell surface via the endocytic pathways plays a key role in the generation of amyloid beta peptide (Abeta) in Alzheimer disease. We report here that inherited variants in the SORL1 neuronal sorting receptor are associated with late-onset Alzheimer disease. These variants, which occur in at least two different clusters of intronic sequences within the SORL1 gene (also known as LR11 or SORLA) may regulate tissue-specific expression of SORL1. We also show that SORL1 directs trafficking of APP into recycling pathways and that when SORL1 is underexpressed, APP is sorted into Abeta-generating compartments. These data suggest that inherited or acquired changes in SORL1 expression or function are mechanistically involved in causing Alzheimer disease.


Subject(s)
Alzheimer Disease/genetics , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/genetics , Age of Onset , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Cell Line , Endosomes/metabolism , Genetic Variation , Haplotypes , Humans , Introns , Models, Genetic , Organ Specificity , Polymorphism, Single Nucleotide , Protease Nexins , Receptors, Cell Surface/metabolism , Vesicular Transport Proteins/metabolism
5.
Trends Biochem Sci ; 33(4): 151-5, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18343669

ABSTRACT

Alzheimer's disease is a major neurodegenerative disease of the brain, the incidence of which increases dramatically in old age. Currently, no drugs are available to halt or slow the progression of this disease, which poses an ever-expanding burden on health services, families and society. The prion protein has become infamous owing to its role as the causative agent of the transmissible spongiform encephalopathies such as Creutzfeldt-Jakob disease in humans. However, our view of the prion protein as an unwanted, harmful entity has been challenged recently. New data indicate that the normal cellular form of the prion protein might have a crucial role in suppressing the production of the amyloid-beta peptide, the neurotoxic molecule involved in the pathogenesis of Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/prevention & control , Prion Diseases/metabolism , Prions/metabolism , Aged , Alzheimer Disease/etiology , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Creutzfeldt-Jakob Syndrome/genetics , Creutzfeldt-Jakob Syndrome/pathology , Genetic Predisposition to Disease , Humans , Membrane Microdomains , Models, Biological , Models, Theoretical , Prion Diseases/etiology , Prion Diseases/genetics , Prions/genetics , Protease Nexins , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
6.
Blood ; 115(1): 97-106, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-19855083

ABSTRACT

Protease nexin-1 (PN-1) is a serpin that inhibits plasminogen activators, plasmin, and thrombin. PN-1 is barely detectable in plasma but is expressed by platelets. Here, we studied platelet PN-1 in resting and activated conditions and its function in thrombosis. Studies on human platelets from healthy donors and from patients with a Gray platelet syndrome demonstrate that PN-1 is present both at the platelet surface and in alpha-granules. The role of PN-1 was investigated in vitro using human platelets incubated with a blocking antibody and using platelets from PN-1-deficient mice. Both approaches indicate that platelet PN-1 is active on thrombin and urokinase-type plasminogen activator. Blockade and deficiency of platelet PN-1 result in accelerated and increased tissue factor-induced thrombin generation as indicated by calibrated automated thrombography. Moreover, platelets from PN-1-deficient mice respond to subthreshold doses of thrombin, as assessed by P-selectin expression and platelet aggregation. Thrombus formation, induced ex vivo by collagen in blood flow conditions and in vivo by FeCl(3)-induced injury, is significantly increased in PN-1-deficient mice, demonstrating the antithrombotic properties of platelet PN-1. Platelet PN-1 is thus a key player in the thrombotic process, whose negative regulatory role has been, up to now, markedly underestimated.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Anticoagulants/metabolism , Antithrombins/metabolism , Blood Platelets/enzymology , Receptors, Cell Surface/metabolism , Adult , Animals , Blood Circulation/drug effects , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Vessels/drug effects , Blood Vessels/enzymology , Blood Vessels/pathology , Blood Vessels/physiopathology , Cell Membrane/drug effects , Cell Membrane/enzymology , Collagen/pharmacology , Glycosaminoglycans/metabolism , Humans , Mice , Platelet Aggregation/drug effects , Platelet-Rich Plasma/metabolism , Protease Nexins , Serpin E2 , Thrombin/antagonists & inhibitors , Thromboplastin/metabolism , Thrombosis/enzymology , Thrombosis/pathology , Thrombosis/physiopathology , Time Factors , Urokinase-Type Plasminogen Activator/antagonists & inhibitors
7.
Proc Natl Acad Sci U S A ; 106(13): 5093-8, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19282473

ABSTRACT

Fe65 is a binding partner of the Alzheimer's beta-amyloid precursor protein APP. The possible involvement of this protein in the cellular response to DNA damage was suggested by the observation that Fe65 null mice are more sensitive to genotoxic stress than WT counterpart. Fe65 associated with chromatin under basal conditions and its involvement in DNA damage repair requires this association. A known partner of Fe65 is the histone acetyltransferase Tip60. Considering the crucial role of Tip60 in DNA repair, we explored the hypothesis that the phenotype of Fe65 null cells depended on its interaction with Tip60. We demonstrated that Fe65 knockdown impaired recruitment of Tip60-TRRAP complex to DNA double strand breaks and decreased histone H4 acetylation. Accordingly, the efficiency of DNA repair was decreased upon Fe65 suppression. To explore whether APP has a role in this mechanism, we analyzed a Fe65 mutant unable to bind to APP. This mutant failed to rescue the phenotypes of Fe65 null cells; furthermore, APP/APLP2 suppression results in the impairment of recruitment of Tip60-TRRAP complex to DNA double strand breaks, decreased histone H4 acetylation and repair efficiency. On these bases, we propose that Fe65 and its interaction with APP play an important role in the response to DNA damage by assisting the recruitment of Tip60-TRRAP to DNA damage sites.


Subject(s)
DNA Breaks , Histone Acetyltransferases/metabolism , Histones/metabolism , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Acetylation , Adaptor Proteins, Signal Transducing/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , DNA Repair , Lysine Acetyltransferase 5 , Mice , Nuclear Proteins/metabolism , Protease Nexins , Protein Transport , Receptors, Cell Surface/metabolism , Trans-Activators
8.
J Neurosci ; 30(26): 8974-83, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20592218

ABSTRACT

The two proteases beta-secretase and gamma-secretase generate the amyloid beta peptide and are drug targets for Alzheimer's disease. Here we tested the possibility of targeting the cellular environment of beta-secretase cleavage instead of the beta-secretase enzyme itself. beta-Secretase has an acidic pH optimum and cleaves the amyloid precursor protein in the acidic endosomes. We identified two drugs, bepridil and amiodarone, that are weak bases and are in clinical use as calcium antagonists. Independently of their calcium-blocking activity, both compounds mildly raised the membrane-proximal, endosomal pH and inhibited beta-secretase cleavage at therapeutically achievable concentrations in cultured cells, in primary neurons, and in vivo in guinea pigs. This shows that an alkalinization of the cellular environment could be a novel therapeutic strategy to inhibit beta-secretase. Surprisingly, bepridil and amiodarone also modulated gamma-secretase cleavage independently of endosomal alkalinization. Thus, both compounds act as dual modulators that simultaneously target beta- and gamma-secretase through distinct molecular mechanisms. In addition to Alzheimer's disease, compounds with dual properties may also be useful for drug development targeting other membrane proteins.


Subject(s)
Amiodarone/pharmacology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Bepridil/pharmacology , Enzyme Inhibitors/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Amiodarone/chemistry , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Bepridil/chemistry , Brain/drug effects , Brain/enzymology , Brain/metabolism , Cell Line , Cells, Cultured , Enzyme Inhibitors/chemistry , Female , Guinea Pigs , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/enzymology , Neurons/metabolism , Protease Nexins , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
9.
J Neurosci ; 30(33): 11157-66, 2010 Aug 18.
Article in English | MEDLINE | ID: mdl-20720123

ABSTRACT

We discovered a nonpeptidic compound, TAK-070, that inhibited BACE1, a rate-limiting protease for the generation of Abeta peptides that are considered causative for Alzheimer's disease (AD), in a noncompetitive manner. TAK-070 bound to full-length BACE1, but not to truncated BACE1 lacking the transmembrane domain. Short-term oral administration of TAK-070 decreased the brain levels of soluble Abeta, increased that of neurotrophic sAPPalpha by approximately 20%, and normalized the behavioral impairments in cognitive tests in Tg2576 mice, an APP transgenic mouse model of AD. Six-month chronic treatment decreased cerebral Abeta deposition by approximately 60%, preserving the pharmacological efficacy on soluble Abeta and sAPPalpha levels. These results support the feasibility of BACE1 inhibition with a noncompetitive inhibitor as disease-modifying as well as symptomatic therapy for AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Biphenyl Compounds/pharmacology , Brain/drug effects , Cognition Disorders/drug therapy , Enzyme Inhibitors/pharmacology , Naphthalenes/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Biphenyl Compounds/chemistry , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Cognition Disorders/metabolism , Cognition Disorders/pathology , Disease Models, Animal , Enzyme Inhibitors/chemistry , Feasibility Studies , Female , Humans , Male , Maze Learning/drug effects , Mice , Mice, Transgenic , Naphthalenes/chemistry , Protease Nexins , Random Allocation , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recognition, Psychology/drug effects , Treatment Outcome
10.
J Biol Chem ; 285(3): 1939-49, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19920152

ABSTRACT

The amyloid precursor protein (APP) is a ubiquitously expressed transmembrane adhesion protein and the progenitor of amyloid-beta peptides. The major splice isoforms of APP expressed by most tissues contain a Kunitz protease inhibitor domain; secreted APP containing this domain is also known as protease nexin 2 and potently inhibits serine proteases, including trypsin and coagulation factors. The atypical human trypsin isoform mesotrypsin is resistant to inhibition by most protein protease inhibitors and cleaves some inhibitors at a substantially accelerated rate. Here, in a proteomic screen to identify potential physiological substrates of mesotrypsin, we find that APP/protease nexin 2 is selectively cleaved by mesotrypsin within the Kunitz protease inhibitor domain. In studies employing the recombinant Kunitz domain of APP (APPI), we show that mesotrypsin cleaves selectively at the Arg(15)-Ala(16) reactive site bond, with kinetic constants approaching those of other proteases toward highly specific protein substrates. Finally, we show that cleavage of APPI compromises its inhibition of other serine proteases, including cationic trypsin and factor XIa, by 2 orders of magnitude. Because APP/protease nexin 2 and mesotrypsin are coexpressed in a number of tissues, we suggest that processing by mesotrypsin may ablate the protease inhibitory function of APP/protease nexin 2 in vivo and may also modulate other activities of APP/protease nexin 2 that involve the Kunitz domain.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Trypsin/metabolism , Amino Acid Sequence , Amyloid beta-Protein Precursor/pharmacology , Animals , Cell Line, Tumor , Factor XIa/antagonists & inhibitors , Humans , Kinetics , Mice , Molecular Sequence Data , Protease Inhibitors/pharmacology , Protease Nexins , Protein Structure, Tertiary , Substrate Specificity
11.
J Biol Chem ; 285(41): 31217-32, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20558735

ABSTRACT

Iron influx increases the translation of the Alzheimer amyloid precursor protein (APP) via an iron-responsive element (IRE) RNA stem loop in its 5'-untranslated region. Equal modulated interaction of the iron regulatory proteins (IRP1 and IRP2) with canonical IREs controls iron-dependent translation of the ferritin subunits. However, our immunoprecipitation RT-PCR and RNA binding experiments demonstrated that IRP1, but not IRP2, selectively bound the APP IRE in human neural cells. This selective IRP1 interaction pattern was evident in human brain and blood tissue from normal and Alzheimer disease patients. We computer-predicted an optimal novel RNA stem loop structure for the human, rhesus monkey, and mouse APP IREs with reference to the canonical ferritin IREs but also the IREs encoded by erythroid heme biosynthetic aminolevulinate synthase and Hif-2α mRNAs, which preferentially bind IRP1. Selective 2'-hydroxyl acylation analyzed by primer extension analysis was consistent with a 13-base single-stranded terminal loop and a conserved GC-rich stem. Biotinylated RNA probes deleted of the conserved CAGA motif in the terminal loop did not bind to IRP1 relative to wild type probes and could no longer base pair to form a predicted AGA triloop. An AGU pseudo-triloop is key for IRP1 binding to the canonical ferritin IREs. RNA probes encoding the APP IRE stem loop exhibited the same high affinity binding to rhIRP1 as occurs for the H-ferritin IRE (35 pm). Intracellular iron chelation increased binding of IRP1 to the APP IRE, decreasing intracellular APP expression in SH-SY5Y cells. Functionally, shRNA knockdown of IRP1 caused increased expression of neural APP consistent with IRP1-APP IRE-driven translation.


Subject(s)
5' Untranslated Regions , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Iron Regulatory Protein 1/metabolism , Nucleic Acid Conformation , Protein Biosynthesis , Receptors, Cell Surface/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Cell Line, Tumor , Humans , Iron/metabolism , Iron Regulatory Protein 1/genetics , Macaca mulatta , Mice , Protease Nexins , Receptors, Cell Surface/genetics
12.
J Biol Chem ; 285(27): 20664-74, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20427278

ABSTRACT

Ectodomain shedding of the amyloid precursor protein (APP) by the two proteases alpha- and beta-secretase is a key regulatory event in the generation of the Alzheimer disease amyloid beta peptide (Abeta). At present, little is known about the cellular mechanisms that control APP shedding and Abeta generation. Here, we identified a novel protein, transmembrane protein 59 (TMEM59), as a new modulator of APP shedding. TMEM59 was found to be a ubiquitously expressed, Golgi-localized protein. TMEM59 transfection inhibited complex N- and O-glycosylation of APP in cultured cells. Additionally, TMEM59 induced APP retention in the Golgi and inhibited Abeta generation as well as APP cleavage by alpha- and beta-secretase cleavage, which occur at the plasma membrane and in the endosomes, respectively. Moreover, TMEM59 inhibited the complex N-glycosylation of the prion protein, suggesting a more general modulation of Golgi glycosylation reactions. Importantly, TMEM59 did not affect the secretion of soluble proteins or the alpha-secretase like shedding of tumor necrosis factor alpha, demonstrating that TMEM59 did not disturb the general Golgi function. The phenotype of TMEM59 transfection on APP glycosylation and shedding was similar to the one observed in cells lacking conserved oligomeric Golgi (COG) proteins COG1 and COG2. Both proteins are required for normal localization and activity of Golgi glycosylation enzymes. In summary, this study shows that TMEM59 expression modulates complex N- and O-glycosylation and suggests that TMEM59 affects APP shedding by reducing access of APP to the cellular compartments, where it is normally cleaved by alpha- and beta-secretase.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Blotting, Northern , CHO Cells , COS Cells , Cell Line , Chlorocebus aethiops , Cricetinae , Cricetulus , Gene Knockdown Techniques , Genes, Reporter , Humans , Kidney , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Polylysine , Protease Nexins , RNA, Small Interfering/genetics , Receptors, Cell Surface/metabolism
13.
J Biol Chem ; 285(40): 30598-605, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20693289

ABSTRACT

Proteolytic processing of the amyloid precursor protein (APP) generates large soluble APP derivatives, ß-amyloid (Aß) peptides, and APP intracellular domain. Expression of the extracellular sequences of APP or its Caenorhabditis elegans counterpart has been shown to be sufficient in partially rescuing the CNS phenotypes of the APP-deficient mice and the lethality of the apl-1 null C. elegans, respectively, leaving open the question as what is the role of the highly conserved APP intracellular domain? To address this question, we created an APP knock-in allele in which the mouse Aß sequence was replaced by the human Aß. A frameshift mutation was introduced that replaced the last 39 residues of the APP sequence. We demonstrate that the C-terminal mutation does not overtly affect APP processing and amyloid pathology. In contrast, crossing the mutant allele with APP-like protein 2 (APLP2)-null mice results in similar neuromuscular synapse defects and early postnatal lethality as compared with mice doubly deficient in APP and APLP2, demonstrating an indispensable role of the APP C-terminal domain in these development activities. Our results establish an essential function of the conserved APP intracellular domain in developmental regulation, and this activity can be genetically uncoupled from APP processing and Aß pathogenesis.


Subject(s)
Alleles , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Neuromuscular Junction/metabolism , Receptors, Cell Surface/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Disease Models, Animal , Frameshift Mutation , Gene Knock-In Techniques , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Neuromuscular Junction/genetics , Neuromuscular Junction/pathology , Protease Nexins , Protein Structure, Tertiary , Receptors, Cell Surface/genetics
14.
J Biol Chem ; 285(14): 10678-89, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20145244

ABSTRACT

The beta-amyloid precursor protein (APP) represents a type I transmembrane glycoprotein that is ubiquitously expressed. In the brain, it is a key player in the molecular pathogenesis of Alzheimer disease. Its physiological function is however less well understood. Previous studies showed that APP is up-regulated in prostate, colon, pancreatic tumor, and oral squamous cell carcinoma. In this study, we show that APP has an essential role in growth control of pancreatic and colon cancer. Abundant APP staining was found in human pancreatic adenocarcinoma and colon cancer tissue. Interestingly, treating pancreatic and colon cancer cells with valproic acid (VPA, 2-propylpentanoic acid), a known histone deacetylase (HDAC) inhibitor, leads to up-regulation of GRP78, an endoplasmic reticulum chaperone immunoglobulin-binding protein. GRP78 is involved in APP maturation and inhibition of tumor cell growth by down-regulation of APP and secreted soluble APPalpha. Trichostatin A, a pan-HDAC inhibitor, also lowered APP and increased GRP78 levels. In contrast, treating cells with valpromide, a VPA derivative lacking HDAC inhibitory properties, had no effect on APP levels. VPA did not modify the level of epidermal growth factor receptor, another type I transmembrane protein, and APLP2, a member of the APP family, demonstrating the specificity of the VPA effect on APP. Small interfering RNA-mediated knockdown of APP also resulted in significantly decreased cell growth. Based on these observations, the data suggest that APP down-regulation via HDAC inhibition provides a novel mechanism for pancreatic and colon cancer therapy.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Anticonvulsants/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/prevention & control , Pancreatic Neoplasms/prevention & control , Receptors, Cell Surface/metabolism , Valproic Acid/pharmacology , Amyloid beta-Protein Precursor/antagonists & inhibitors , Amyloid beta-Protein Precursor/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Down-Regulation , Endoplasmic Reticulum Chaperone BiP , Histone Deacetylase Inhibitors/pharmacology , Humans , Immunoenzyme Techniques , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protease Nexins , RNA, Small Interfering/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Tumor Cells, Cultured
15.
J Cell Sci ; 122(Pt 22): 4177-85, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19861499

ABSTRACT

Deficiency of caytaxin results in hereditary ataxia or dystonia in humans, mice and rats. Our yeast two-hybrid screen identified kinesin light chains (KLCs) as caytaxin-binding proteins. The tetratricopeptide-repeat region of KLC1 recognizes the ELEWED sequence (amino acids 115-120) of caytaxin. This motif is conserved among BNIP-2 family members and other KLC-interacting kinesin cargo proteins such as calsyntenins. Caytaxin associates with kinesin heavy chains (KHCs) indirectly by binding to KLCs, suggesting that caytaxin binds to the tetrameric kinesin molecule. In cultured hippocampal neurons, we found that caytaxin is distributed in both axons and dendrites in punctate patterns, and it colocalizes with microtubules and KHC. GFP-caytaxin expressed in hippocampal neurons is transported at a speed ( approximately 1 mum/second) compatible with kinesin movement. Inhibition of kinesin-1 by dominant-negative KHC decreases the accumulation of caytaxin in the growth cone. Caytaxin puncta do not coincide with vesicles containing known kinesin cargos such as APP or JIP-1. A part of caytaxin, however, colocalizes with mitochondria and suppression of caytaxin expression by RNAi redistributes mitochondria away from the distal ends of neurites. These data indicate that caytaxin binds to kinesin-1 and functions as an adaptor that mediates intracellular transport of specific cargos, one of which is the mitochondrion.


Subject(s)
Axonal Transport/physiology , Kinesins/physiology , Nerve Tissue Proteins/physiology , Neurons/metabolism , Adaptor Proteins, Signal Transducing/physiology , Amino Acid Motifs , Amyloid beta-Protein Precursor/physiology , Animals , Ataxia , Binding Sites , Cell Line , Hippocampus , Humans , Kinesins/chemistry , Mice , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/physiology , Microtubules/physiology , Mitochondria/physiology , Nerve Tissue Proteins/chemistry , Neurons/ultrastructure , Protease Nexins , Protein Subunits/chemistry , Protein Subunits/physiology , RNA Interference , Rats , Receptors, Cell Surface/physiology , Two-Hybrid System Techniques
16.
Nat Med ; 10(10): 1065-6, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15448688

ABSTRACT

Recent reports suggest that some commonly used nonsteroidal anti-inflammatory drugs (NSAIDs) unexpectedly shift the cleavage products of amyloid precursor protein (APP) to shorter, less fibrillogenic forms, although the underlying mechanism remains unknown. We now demonstrate, using a fluorescence resonance energy transfer method, that Abeta(42)-lowering NSAIDs specifically affect the proximity between APP and presenilin 1 and alter presenilin 1 conformation both in vitro and in vivo, suggesting a novel allosteric mechanism of action.


Subject(s)
Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carrier Proteins/metabolism , Endopeptidases/metabolism , Membrane Proteins/chemistry , Models, Molecular , Peptide Fragments/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor , Animals , Aspartic Acid Endopeptidases , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Fluorescence Resonance Energy Transfer , Mice , Mice, Transgenic , Presenilin-1 , Protease Nexins , Protein Conformation/drug effects , Receptors, Cell Surface
17.
Nat Med ; 10(9): 959-65, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15286784

ABSTRACT

Inhibiting the activity of the beta-amyloid converting enzyme 1 (BACE1) or reducing levels of BACE1 in vivo decreases the production of amyloid-beta. The reticulon family of proteins has four members, RTN1, RTN2, RTN3 and RTN4 (also known as Nogo), the last of which is well known for its role in inhibiting neuritic outgrowth after injury. Here we show that reticulon family members are binding partners of BACE1. In brain, BACE1 mainly colocalizes with RTN3 in neurons, whereas RTN4 is more enriched in oligodendrocytes. An increase in the expression of any reticulon protein substantially reduces the production of Abeta. Conversely, lowering the expression of RTN3 by RNA interference increases the secretion of Abeta, suggesting that reticulon proteins are negative modulators of BACE1 in cells. Our data support a mechanism by which reticulon proteins block access of BACE1 to amyloid precursor protein and reduce the cleavage of this protein. Thus, changes in the expression of reticulon proteins in the human brain are likely to affect cellular amyloid-beta and the formation of amyloid plaques.


Subject(s)
Amyloid beta-Peptides/biosynthesis , Aspartic Acid Endopeptidases/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor , Base Sequence , Blotting, Western , Carrier Proteins/genetics , DNA Primers , Endopeptidases/metabolism , Gene Library , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Microscopy, Fluorescence , Molecular Sequence Data , Myelin Proteins , Nerve Tissue Proteins/genetics , Neurons/metabolism , Nogo Proteins , Oligodendroglia/metabolism , Protease Nexins , RNA Interference , Receptors, Cell Surface , Sequence Analysis, DNA
18.
Am J Respir Crit Care Med ; 182(5): 605-13, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20463177

ABSTRACT

RATIONALE: Several family-based studies have identified genetic linkage for lung function and airflow obstruction to chromosome 2q. OBJECTIVES: We hypothesized that merging results of high-resolution single nucleotide polymorphism (SNP) mapping in four separate populations would lead to the identification of chronic obstructive pulmonary disease (COPD) susceptibility genes on chromosome 2q. METHODS: Within the chromosome 2q linkage region, 2,843 SNPs were genotyped in 806 COPD cases and 779 control subjects from Norway, and 2,484 SNPs were genotyped in 309 patients with severe COPD from the National Emphysema Treatment Trial and 330 community control subjects. Significant associations from the combined results across the two case-control studies were followed up in 1,839 individuals from 603 families from the International COPD Genetics Network (ICGN) and in 949 individuals from 127 families in the Boston Early-Onset COPD Study. MEASUREMENTS AND MAIN RESULTS: Merging the results of the two case-control analyses, 14 of the 790 overlapping SNPs had a combined P < 0.01. Two of these 14 SNPs were consistently associated with COPD in the ICGN families. The association with one SNP, located in the gene XRCC5, was replicated in the Boston Early-Onset COPD Study, with a combined P = 2.51 x 10(-5) across the four studies, which remains significant when adjusted for multiple testing (P = 0.02). Genotype imputation confirmed the association with SNPs in XRCC5. CONCLUSIONS: By combining data from COPD genetic association studies conducted in four independent patient samples, we have identified XRCC5, an ATP-dependent DNA helicase, as a potential COPD susceptibility gene.


Subject(s)
Chromosomes, Human, Pair 2 , DNA Helicases/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Age of Onset , Aged , Amyloid beta-Protein Precursor/genetics , Case-Control Studies , Chromosome Mapping , Female , Genetic Linkage , Genetic Predisposition to Disease , Humans , Ku Autoantigen , Male , Middle Aged , Polymorphism, Single Nucleotide , Protease Nexins , Receptors, Cell Surface/genetics , Smoking/adverse effects
19.
Biophys J ; 98(10): 2299-308, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20483339

ABSTRACT

The amphipathicity of the natively unstructured amyloid-beta (Abeta40) peptide may play an important role in its aggregation into beta-sheet rich fibrils, which is linked to the pathogenesis of Alzheimer's disease. Using the air/subphase interface as a model interface, we characterized Abeta's surface activity and its conformation, assembly, and morphology at the interface. Abeta readily adsorbed to the air/subphase interface to form a 20 A thick film and showed a critical micelle concentration of approximately 120 nM. Abeta adsorbed at the air/subphase exhibited in-plane ordering that gave rise to Bragg peaks in grazing-incidence x-ray diffraction measurements. Analysis of the peaks showed that the air/subphase interface induced Abeta to fold into a beta-sheet conformation and to self-assemble into approximately 100 A-sized ordered clusters. The formation of these clusters at the air/subphase interface was not affected by pH, salts, or the presence of sucrose or urea, which are known to stabilize or denature native proteins, suggesting that interface-driven Abeta misfolding and assembly are strongly favored. Furthermore, Abeta at the interface seeded the growth of fibrils in the bulk with a distinct morphology compared to those formed by homogeneous nucleation. Our results indicate that interface-induced Abeta misfolding may serve as a heterogeneous, nucleation-controlled aggregation mechanism for Abeta fibrillogenesis in vivo.


Subject(s)
Amyloid beta-Peptides/adverse effects , Amyloid/chemistry , Peptide Fragments/pharmacology , Peptides/adverse effects , Protein Folding/drug effects , Proteostasis Deficiencies/etiology , Amyloid/adverse effects , Amyloid beta-Peptides/chemistry , Amyloid beta-Protein Precursor , Binding Sites , Circular Dichroism/methods , Lipid Bilayers/pharmacology , Microscopy, Atomic Force/methods , Peptides/pharmacology , Protease Nexins , Protein Conformation , Receptors, Cell Surface , Surface Properties
20.
J Neurosci ; 29(29): 9321-9, 2009 Jul 22.
Article in English | MEDLINE | ID: mdl-19625522

ABSTRACT

Downregulation of brain-derived neurotrophic factor (BDNF) in the cortex occurs early in the progression of Alzheimer's disease (AD). Since BDNF plays a critical role in neuronal survival, synaptic plasticity, and memory, BDNF reduction may contribute to synaptic and cellular loss and memory deficits characteristic of AD. In vitro evidence suggests that amyloid-beta (A beta) contributes to BDNF downregulation in AD, but the specific A beta aggregation state responsible for this downregulation in vivo is unknown. In the present study, we examined cortical levels of BDNF mRNA in three different transgenic AD mouse models harboring mutations in APP resulting in A beta overproduction, and in a genetic mouse model of Down syndrome. Two of the three A beta transgenic strains (APP(NLh) and TgCRND8) exhibited significantly decreased cortical BDNF mRNA levels compared with wild-type mice, whereas neither the other strain (APP(swe)/PS-1) nor the Down syndrome mouse model (Ts65Dn) was affected. Only APP(NLh) and TgCRND8 mice expressed high A beta(42)/A beta(40) ratios and larger SDS-stable A beta oligomers (approximately 115 kDa). TgCRND8 mice exhibited downregulation of BDNF transcripts III and IV; transcript IV is also downregulated in AD. Furthermore, in all transgenic mouse strains, there was a correlation between levels of large oligomers, A beta(42)/A beta(40), and severity of BDNF decrease. These data show that the amount and species of A beta vary among transgenic mouse models of AD and are negatively correlated with BDNF levels. These findings also suggest that the effect of A beta on decreased BDNF expression is specific to the aggregation state of A beta and is dependent on large oligomers.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Blotting, Western , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/metabolism , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Mice, Transgenic , Polymerase Chain Reaction , Presenilin-1/genetics , Protease Nexins , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics
SELECTION OF CITATIONS
SEARCH DETAIL