Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
Cell Rep ; 27(5): 1345-1355.e6, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31042463

ABSTRACT

Amyloid precursor protein (APP) and its metabolites play key roles in Alzheimer's disease (AD) pathophysiology. Whereas short amyloid-ß (Aß) peptides derived from APP are pathogenic, the APP holoprotein serves multiple purposes in the nervous system through its cell adhesion and receptor-like properties. Our studies focused on the signaling mediated by the APP cytoplasmic tail. We investigated whether sustained APP signaling during brain development might favor neuronal plasticity and memory process through a direct interaction with the heterotrimeric G-protein subunit GαS (stimulatory G-protein alpha subunit). Our results reveal that APP possesses autonomous regulatory capacity within its intracellular domain that promotes APP cell surface residence, precludes Aß production, facilitates axodendritic development, and preserves cellular substrates of memory. Altogether, these events contribute to strengthening cognitive functions and are sufficient to modify the course of AD pathology.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Memory , Neurogenesis , Signal Transduction , Amyloid beta-Protein Precursor/chemistry , Animals , Brain/growth & development , Brain/metabolism , Cells, Cultured , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity , Protein Domains
3.
Proc Natl Acad Sci U S A ; 114(45): E9665-E9674, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29078331

ABSTRACT

Alzheimer's disease (AD) is a devastating neurodegenerative disorder characterized by pathological brain lesions and a decline in cognitive function. ß-Amyloid peptides (Aß), derived from proteolytic processing of amyloid precursor protein (APP), play a central role in AD pathogenesis. ß-Site APP cleaving enzyme 1 (BACE1), the transmembrane aspartyl protease which initiates Aß production, is axonally transported in neurons and accumulates in dystrophic neurites near cerebral amyloid deposits in AD. BACE1 is modified by S-palmitoylation at four juxtamembrane cysteine residues. S-palmitoylation is a dynamic posttranslational modification that is important for trafficking and function of several synaptic proteins. Here, we investigated the in vivo significance of BACE1 S-palmitoylation through the analysis of knock-in mice with cysteine-to-alanine substitution at the palmitoylated residues (4CA mice). BACE1 expression, as well as processing of APP and other neuronal substrates, was unaltered in 4CA mice despite the lack of BACE1 S-palmitoylation and reduced lipid raft association. Whereas steady-state Aß levels were similar, synaptic activity-induced endogenous Aß production was not observed in 4CA mice. Furthermore, we report a significant reduction of cerebral amyloid burden and BACE1 accumulation in dystrophic neurites in the absence of BACE1 S-palmitoylation in mouse models of AD amyloidosis. Studies in cultured neurons suggest that S-palmitoylation is required for dendritic spine localization and axonal targeting of BACE1. Finally, the lack of BACE1 S-palmitoylation mitigates cognitive deficits in 5XFAD mice. Using transgenic mouse models, these results demonstrate that intrinsic posttranslational S-palmitoylation of BACE1 has a significant impact on amyloid pathogenesis and the consequent cognitive decline.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid/metabolism , Aspartic Acid Endopeptidases/metabolism , Memory Disorders/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloidogenic Proteins/metabolism , Amyloidosis/metabolism , Animals , Axons/metabolism , Brain/metabolism , Disease Models, Animal , Female , Lipoylation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Protein Processing, Post-Translational/physiology
4.
Mol Neurodegener ; 11(1): 59, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27488240

ABSTRACT

BACKGROUND: Genome-wide association studies have identified BIN1 within the second most significant susceptibility locus in late-onset Alzheimer's disease (AD). BIN1 undergoes complex alternative splicing to generate multiple isoforms with diverse functions in multiple cellular processes including endocytosis and membrane remodeling. An increase in BIN1 expression in AD and an interaction between BIN1 and Tau have been reported. However, disparate descriptions of BIN1 expression and localization in the brain previously reported in the literature and the lack of clarity on brain BIN1 isoforms present formidable challenges to our understanding of how genetic variants in BIN1 increase the risk for AD. METHODS: In this study, we analyzed BIN1 mRNA and protein levels in human brain samples from individuals with or without AD. In addition, we characterized the BIN1 expression and isoform diversity in human and rodent tissue by immunohistochemistry and immunoblotting using a panel of BIN1 antibodies. RESULTS: Here, we report on BIN1 isoform diversity in the human brain and document alterations in the levels of select BIN1 isoforms in individuals with AD. In addition, we report striking BIN1 localization to white matter tracts in rodent and the human brain, and document that the large majority of BIN1 is expressed in mature oligodendrocytes whereas neuronal BIN1 represents a minor fraction. This predominant non-neuronal BIN1 localization contrasts with the strict neuronal expression and presynaptic localization of the BIN1 paralog, Amphiphysin 1. We also observe upregulation of BIN1 at the onset of postnatal myelination in the brain and during differentiation of cultured oligodendrocytes. Finally, we document that the loss of BIN1 significantly correlates with the extent of demyelination in multiple sclerosis lesions. CONCLUSION: Our study provides new insights into the brain distribution and cellular expression of an important risk factor associated with late-onset AD. We propose that efforts to define how genetic variants in BIN1 elevate the risk for AD would behoove to consider BIN1 function in the context of its main expression in mature oligodendrocytes and the potential for a role of BIN1 in the membrane remodeling that accompanies the process of myelination.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/metabolism , Nuclear Proteins/metabolism , Oligodendroglia/metabolism , Tumor Suppressor Proteins/metabolism , White Matter/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Aged, 80 and over , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , Neurogenesis/genetics , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , White Matter/pathology , tau Proteins/metabolism
5.
Elife ; 52016 05 19.
Article in English | MEDLINE | ID: mdl-27196744

ABSTRACT

Presenilin 1 (PS1) is an essential γ-secretase component, the enzyme responsible for amyloid precursor protein (APP) intramembraneous cleavage. Mutations in PS1 lead to dominant-inheritance of early-onset familial Alzheimer's disease (FAD). Although expression of FAD-linked PS1 mutations enhances toxic Aß production, the importance of other APP metabolites and γ-secretase substrates in the etiology of the disease has not been confirmed. We report that neurons expressing FAD-linked PS1 variants or functionally deficient PS1 exhibit enhanced axodendritic outgrowth due to increased levels of APP intracellular C-terminal fragment (APP-CTF). APP expression is required for exuberant neurite outgrowth and hippocampal axonal sprouting observed in knock-in mice expressing FAD-linked PS1 mutation. APP-CTF accumulation initiates CREB signaling cascade through an association of APP-CTF with Gαs protein. We demonstrate that pathological PS1 loss-of-function impinges on neurite formation through a selective APP gain-of-function that could impact on axodendritic connectivity and contribute to aberrant axonal sprouting observed in AD patients.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neurons/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Animals , Gene Expression , Gene Knock-In Techniques , Humans , Mice
6.
Trends Pharmacol Sci ; 37(5): 390-411, 2016 05.
Article in English | MEDLINE | ID: mdl-26837733

ABSTRACT

Amyloid precursor protein (APP) and its metabolites play a key role in Alzheimer's disease pathogenesis. The idea that APP may function as a receptor has gained momentum based on its structural similarities to type I transmembrane receptors and the identification of putative APP ligands. We review the recent experimental evidence in support of this notion and discuss how this concept is viewed in the field. Specifically, we focus on the structural and functional characteristics of APP as a cell surface receptor, and on its interaction with adaptors and signaling proteins. We also address the importance of APP function as a receptor in Alzheimer's disease etiology and discuss how this function might be potentially important for the development of novel therapeutic approaches.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Alzheimer Disease/metabolism , Animals , Humans
7.
Cell Rep ; 5(6): 1552-63, 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24373286

ABSTRACT

Abnormal accumulation of ß-secretase (BACE1) in dystrophic neurites and presynaptic ß-amyloid (Aß) production contribute to Alzheimer's disease pathogenesis. Little, however, is known about BACE1 sorting and dynamic transport in neurons. We investigated BACE1 trafficking in hippocampal neurons using live-cell imaging and selective labeling. We report that transport vesicles containing internalized BACE1 in dendrites undergo exclusive retrograde transport toward the soma, whereas they undergo bidirectional transport in axons. Unidirectional dendritic transport requires Eps15-homology-domain-containing (EHD) 1 and 3 protein function. Furthermore, loss of EHD function compromises dynamic axonal transport and overall BACE1 levels in axons. EHD1/3 colocalize with BACE1 and APP ß-C-terminal fragments in hippocampal mossy fiber terminals, and their depletion in neurons significantly attenuates Aß levels. These results demonstrate unidirectional endocytic transport of a dendritic cargo and reveal a role for EHD proteins in neuronal BACE1 transcytosis and Aß production, processes that are highly relevant for Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/metabolism , Axonal Transport , Carrier Proteins/metabolism , Dendrites/metabolism , Vesicular Transport Proteins/metabolism , Animals , Carrier Proteins/genetics , Cells, Cultured , HEK293 Cells , HeLa Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice , Protein Transport , Vesicular Transport Proteins/genetics
8.
J Neurosci ; 32(5): 1714-29, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22302812

ABSTRACT

Numerous physiological functions, including a role as a cell surface receptor, have been ascribed to Alzheimer's disease-associated amyloid precursor protein (APP). However, detailed analysis of intracellular signaling mediated by APP in neurons has been lacking. Here, we characterized intrinsic signaling associated with membrane-bound APP C-terminal fragments, which are generated following APP ectodomain release by α- or ß-secretase cleavage. We found that accumulation of APP C-terminal fragments or expression of membrane-tethered APP intracellular domain results in adenylate cyclase-dependent activation of PKA (protein kinase A) and inhibition of GSK3ß signaling cascades, and enhancement of axodendritic arborization in rat immortalized hippocampal neurons, mouse primary cortical neurons, and mouse neuroblastoma. We discovered an interaction between BBXXB motif of APP intracellular domain and the heterotrimeric G-protein subunit Gα(S), and demonstrate that Gα(S) coupling to adenylate cyclase mediates membrane-tethered APP intracellular domain-induced neurite outgrowth. Our study provides clear evidence that APP intracellular domain can have a nontranscriptional role in regulating neurite outgrowth through its membrane association. The novel functional coupling of membrane-bound APP C-terminal fragments with Gα(S) signaling identified in this study could impact several brain functions such as synaptic plasticity and memory formation.


Subject(s)
Amyloid beta-Protein Precursor/physiology , GTP-Binding Protein alpha Subunits, Gs/physiology , Intracellular Membranes/physiology , Signal Transduction/physiology , Adenylyl Cyclases/metabolism , Adenylyl Cyclases/physiology , Amino Acid Sequence , Amyloid beta-Protein Precursor/chemistry , Animals , COS Cells , Cell Line, Transformed , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/physiology , Cell Proliferation , Chlorocebus aethiops , Female , GTP-Binding Protein alpha Subunits, Gs/chemistry , Intracellular Membranes/chemistry , Male , Mice , Molecular Sequence Data , Neurites/physiology , Protein Structure, Tertiary , Rats
9.
Mol Neurodegener ; 6: 87, 2011 Dec 28.
Article in English | MEDLINE | ID: mdl-22204304

ABSTRACT

BACKGROUND: p23 belongs to the highly conserved p24 family of type I transmembrane proteins, which participate in the bidirectional protein transport between the endoplasmic reticulum and Golgi apparatus. Mammalian p23 has been shown to interact with γ-secretase complex, and modulate secretory trafficking as well as intramembranous processing of amyloid precursor protein in cultured cells. Negative modulation of ß-amyloid production by p23 in cultured cell lines suggested that elevation of p23 expression in neurons might mitigate cerebral amyloid burden. RESULTS: We generated several lines of transgenic mice expressing human p23 in neurons under the control of Thy-1.2 promoter. We found that even a 50% increase in p23 levels in the central nervous system of mice causes post-natal growth retardation, severe neurological problems characterized by tremors, seizure, ataxia, and uncoordinated movements, and premature death. The severity of the phenotype closely correlated with the level of p23 overexpression in multiple transgenic lines. While the number and general morphology of neurons in Hup23 mice appeared to be normal throughout the brain, abnormal non-Golgi p23 localization was observed in a subset of neurons with high transgene expression in brainstem. Moreover, detailed immunofluorescence analysis revealed marked proliferation of astrocytes, activation of microglia, and thinning of myelinated bundles in brainstem of Hup23 mice. CONCLUSIONS: These results demonstrate that proper level of p23 expression is critical for neuronal function, and perturbing p23 function by overexpression initiates a cascade of cellular reactions in brainstem that leads to severe motor deficits and other neurological problems, which culminate in premature death. The neurological phenotype observed in Hup23 mice highlights significant adverse effects associated with manipulating neuronal expression of p23, a previously described negative modulator of γ-secretase activity and ß-amyloid production. Moreover, our report has broader relevance to molecular mechanisms in several neurodegenerative diseases as it highlights the inherent vulnerability of the early secretory pathway mechanisms that ensure proteostasis in neurons.


Subject(s)
Membrane Proteins/metabolism , Motor Activity/physiology , Movement Disorders/physiopathology , Neurons/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/anatomy & histology , Brain/metabolism , Cells, Cultured , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Movement Disorders/pathology , Myelin Basic Protein/metabolism , Neurons/cytology , Neurons/pathology , Nucleocytoplasmic Transport Proteins
10.
J Neurosci ; 30(48): 16160-9, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21123562

ABSTRACT

Sequential cleavage of amyloid precursor protein by ß- and γ-secretases generates ß-amyloid peptides (Aß), which accumulate in the brains of patients with Alzheimer's disease. We recently identified S-palmitoylation of two γ-secretase subunits, APH1 and nicastrin. S-Palmitoylation is an essential posttranslational modification for the proper trafficking and function of many neuronal proteins. In cultured cell lines, lack of S-palmitoylation causes instability of nascent APH1 and nicastrin but does not affect γ-secretase processing of amyloid precursor protein. To determine the importance of γ-secretase S-palmitoylation for Aß deposition in the brain, we generated transgenic mice coexpressing human wild-type or S-palmitoylation-deficient APH1aL and nicastrin in neurons in the forebrain. We found that lack of S-palmitoylation did not impair the ability of APH1aL and nicastrin to form enzymatically active protein complexes with endogenous presenilin 1 and PEN2 or affect the localization of γ-secretase subunits in dendrites and axons of cortical neurons. When we crossed these mice with 85Dbo transgenic mice, which coexpress familial Alzheimer's disease-causing amyloid precursor protein and presenilin 1 variants, we found that coexpression of wild-type or mutant APH1aL and nicastrin led to marked stabilization of transgenic presenilin 1 in the brains of double-transgenic mice. Interestingly, we observed a moderate, but significant, reduction in amyloid deposits in the forebrain of mice expressing S-palmitoylation-deficient γ-secretase subunits compared with mice overexpressing wild-type subunits, as well as a reduction in the levels of insoluble Aß(40-42). These results indicate that γ-secretase S-palmitoylation modulates Aß deposition in the brain.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/deficiency , Amyloid beta-Peptides/metabolism , Endopeptidases/deficiency , Lipoylation/physiology , Membrane Glycoproteins/deficiency , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amino Acid Sequence , Amyloid Precursor Protein Secretases/biosynthesis , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/physiology , Animals , Endopeptidases/biosynthesis , Endopeptidases/physiology , Lipoylation/genetics , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/physiology , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Peptide Fragments/metabolism , Peptide Fragments/physiology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
11.
J Biol Chem ; 285(49): 38042-52, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-20921220

ABSTRACT

The γ-secretase protein complex executes the intramembrane proteolysis of amyloid precursor protein (APP), which releases Alzheimer disease ß-amyloid peptide. In addition to APP, γ-secretase also cleaves several other type I membrane protein substrates including Notch1 and N-cadherin. γ-Secretase is made of four integral transmembrane protein subunits: presenilin (PS), nicastrin, APH1, and PEN2. Multiple lines of evidence indicate that a heteromer of PS-derived N- and C-terminal fragments functions as the catalytic subunit of γ-secretase. Only limited information is available on the domains within each subunit involved in the recognition and recruitment of diverse substrates and the transfer of substrates to the catalytic site. Here, we performed mutagenesis of two domains of PS1, namely the first luminal loop domain (LL1) and the second transmembrane domain (TM2), and analyzed PS1 endoproteolysis as well as the catalytic activities of PS1 toward APP, Notch, and N-cadherin. Our results show that distinct residues within LL1 and TM2 domains as well as the length of the LL1 domain are critical for PS1 endoproteolysis, but not for PS1 complex formation with nicastrin, APH1, and PEN2. Furthermore, our experimental PS1 mutants formed γ-secretase complexes with distinct catalytic properties toward the three substrates examined in this study; however, the mutations did not affect PS1 interaction with the substrates. We conclude that the N-terminal LL1 and TM2 domains are critical for PS1 endoproteolysis and the coordination between the putative substrate-docking site and the catalytic core of the γ-secretase.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Presenilin-1/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor , Animals , Cadherins/genetics , Cadherins/metabolism , Catalytic Domain/physiology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Mutagenesis , Mutation , Presenilin-1/genetics , Protein Structure, Tertiary , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Substrate Specificity/physiology
12.
Int J Alzheimers Dis ; 20102010 Jul 20.
Article in English | MEDLINE | ID: mdl-20798900

ABSTRACT

Mutations in PSEN genes, which encode presenilin proteins, cause familial early-onset Alzheimer's disease (AD). Transgenic mouse models based on coexpression of familial AD-associated presenilin and amyloid precursor protein variants successfully mimic characteristic pathological features of AD, including plaque formation, synaptic dysfunction, and loss of memory. Presenilins function as the catalytic subunit of gamma-secretase, the enzyme that catalyzes intramembraneous proteolysis of amyloid precursor protein to release beta-amyloid peptides. Familial AD-associated mutations in presenilins alter the site of gamma-secretase cleavage in a manner that increases the generation of longer and highly fibrillogenic beta-amyloid peptides. In addition to amyloid precursor protein, gamma-secretase catalyzes intramembrane proteolysis of many other substrates known to be important for synaptic function. This paper focuses on how various animal models have enabled us to elucidate the physiological importance of diverse gamma-secretase substrates, including amyloid precursor protein and discusses their roles in the context of cellular signaling and synaptic function.

13.
Neurobiol Dis ; 32(1): 37-49, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18652896

ABSTRACT

Sequential processing of amyloid precursor protein by beta- and gamma-secretases generates Alzheimer's disease (AD)-associated beta-amyloid peptides. Recently it was reported that the transmembrane protein p23/TMP21 associates with gamma-secretase, and negatively regulates beta-amyloid production. Despite the link between p23 function and AD pathogenesis, the expression of p23 has not been examined in the brain. Here, we describe the detailed immunohistochemical characterization of p23 expression in rodent and human brain. We report that p23 is co-expressed with gamma-secretase subunits in select neuronal cell populations in rodent brain. Interestingly, the steady-state level of p23 in the brain is high during embryonic development and then declines after birth. Furthermore, the steady-state p23 levels are reduced in the brains of individuals with AD. We conclude that p23 is expressed in neurons throughout the brain and the decline in p23 expression during postnatal development may significantly contribute to enhanced beta-amyloid production in the adult brain.


Subject(s)
Membrane Proteins/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Animals, Newborn , Brain Chemistry/genetics , Cell Line, Tumor , Cells, Cultured , Cerebellum/chemistry , Cerebellum/metabolism , Frontal Lobe/chemistry , Frontal Lobe/metabolism , HeLa Cells , Hippocampus/chemistry , Hippocampus/metabolism , Humans , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Middle Aged , Nucleocytoplasmic Transport Proteins , Rats , Rats, Sprague-Dawley
14.
J Biol Chem ; 283(28): 19489-98, 2008 Jul 11.
Article in English | MEDLINE | ID: mdl-18456655

ABSTRACT

Cerebral deposition of beta-amyloid (Abeta) peptides is a pathological hallmark of Alzheimer disease. Intramembranous proteolysis of amyloid precursor protein by a multiprotein gamma-secretase complex generates Abeta. Previously, it was reported that CD147, a glycoprotein that stimulates production of matrix metalloproteinases (MMPs), is a subunit of gamma-secretase and that the levels of secreted Abeta inversely correlate with CD147 expression. Here, we show that the levels and localization of CD147 in fibroblasts, as well as postnatal expression and distribution in brain, are distinct from those of integral gamma-secretase subunits. Notably, we show that although depletion of CD147 increased extracellular Abeta levels in intact cells, membranes isolated from CD147-depleted cells failed to elevate Abeta production in an in vitro gamma-secretase assay. Consistent with an extracellular source that modulates Abeta metabolism, synthetic Abeta was degraded more rapidly in the conditioned medium of cells overexpressing CD147. Moreover, modulation of CD147 expression had no effect on epsilon-site cleavage of amyloid precursor protein and Notch1 receptor. Collectively, our results demonstrate that CD147 modulates Abeta levels not by regulating gamma-secretase activity, but by stimulating extracellular degradation of Abeta. In view of the known function of CD147 in MMP production, we postulate that CD147 expression influences Abeta levels by an indirect mechanism involving MMPs that can degrade extracellular Abeta.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Basigin/metabolism , Matrix Metalloproteinases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Animals , Basigin/genetics , Cell Line , Cerebellum/metabolism , Cerebellum/pathology , Gene Expression Regulation/genetics , Humans , Mice , Receptor, Notch1/genetics , Receptor, Notch1/metabolism
15.
J Neurochem ; 104(6): 1637-48, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17996025

ABSTRACT

Mutations in the genes encoding presenilins (PS1 and PS2) account for the majority of cases of early-onset Alzheimer's disease. PS1 and PS2 form the catalytic center of gamma-secretase, an enzyme responsible for intramembraneous proteolysis of several type I transmembrane proteins. Many gamma-secretase substrates are coupled to intracellular signaling events such as cAMP-response element binding protein and Rac1/p21-activated kinase pathways, which are associated with synaptic function. Here, we have examined the activation of these pathways in neurons lacking PS1 expression or gamma-secretase activity. We found evidence for heightened steady-state activation of cAMP-response element binding protein, Rac1, and p21-activated kinase signaling in PS-deficient neurons. Our study highlights the importance of PS-dependent proteolytic cleavage of gamma-secretase substrates in regulating neuronal signal transduction.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Presenilin-1/genetics , p21-Activated Kinases/metabolism , rac GTP-Binding Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Presenilin-1/metabolism , Signal Transduction/physiology , rac1 GTP-Binding Protein
16.
Nat Clin Pract Neurol ; 3(7): 374-82, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17611486

ABSTRACT

Alzheimer's disease (AD) is the most common cause of age-related dementia. Pathologically, AD is characterized by the deposition in the brain of amyloid-beta peptides derived from proteolysis of amyloid precursor protein (APP) by beta-site APP cleaving enzyme 1 (BACE1) and gamma-secretase. A growing body of evidence implicates cholesterol and cholesterol-rich membrane microdomains in amyloidogenic processing of APP. Here, we review recent findings regarding the association of BACE1, gamma-secretase and APP in lipid rafts, and discuss potential therapeutic strategies for AD that are based on knowledge gleaned from the membrane environment that fosters APP processing.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Protein Precursor/metabolism , Membrane Microdomains/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Humans , Protein Transport/physiology
17.
J Biol Chem ; 280(27): 25892-900, 2005 Jul 08.
Article in English | MEDLINE | ID: mdl-15886206

ABSTRACT

Gamma-secretase facilitates the regulated intramembrane proteolysis of select type I membrane proteins that play diverse physiological roles in multiple cell types and tissue. In this study, we used biochemical approaches to examine the distribution of amyloid precursor protein (APP) and several additional gamma-secretase substrates in membrane microdomains. We report that APP C-terminal fragments (CTFs) and gamma-secretase reside in Lubrol WX detergent-insoluble membranes (DIM) of cultured cells and adult mouse brain. APP CTFs that accumulate in cells lacking gamma-secretase activity preferentially associate with DIM. Cholesterol depletion and magnetic immunoisolation studies indicate recruitment of APP CTFs into cholesterol- and sphingolipid-rich lipid rafts, and co-residence of APP CTFs, PS1, and syntaxin 6 in DIM patches derived from the trans-Golgi network. Photoaffinity cross-linking studies provided evidence for the preponderance of active gamma-secretase in lipid rafts of cultured cells and adult brain. Remarkably, unlike the case of APP, CTFs derived from Notch1, Jagged2, deleted in colorectal cancer (DCC), and N-cadherin remain largely detergent-soluble, indicative of their spatial segregation in non-raft domains. In embryonic brain, the majority of PS1 and nicastrin is present in Lubrol WX-soluble membranes, wherein the CTFs derived from APP, Notch1, DCC, and N-cadherin also reside. We suggest that gamma-secretase residence in non-raft membranes facilitates proteolysis of diverse substrates during embryonic development but that the translocation of gamma-secretase to lipid rafts in adults ensures processing of certain substrates, including APP CTFs, while limiting processing of other potential substrates.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Cell Membrane/enzymology , Membrane Lipids/metabolism , Age Factors , Amyloid Precursor Protein Secretases , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/embryology , Brain/enzymology , Cell Line, Tumor , Cholesterol/metabolism , Endopeptidases , Gene Deletion , Jagged-2 Protein , Membrane Microdomains/enzymology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neuroblastoma , Polyethylene Glycols , Presenilin-1 , Presenilin-2 , Qa-SNARE Proteins , Receptor, Notch1 , Receptors, Cell Surface/metabolism , Sphingolipids/metabolism , Substrate Specificity , Transcription Factors/metabolism
18.
J Neurosci ; 25(6): 1540-9, 2005 Feb 09.
Article in English | MEDLINE | ID: mdl-15703408

ABSTRACT

Presenilin (PS) plays an essential role in intramembranous gamma-secretase processing of amyloid precursor protein (APP) and several membrane-bound proteins. Here we report that selective accumulation of a membrane-tethered deleted in colorectal cancer (DCC) derivative (DCC-alpha) correlates with extensive neurite outgrowth in transfected neuroblastoma cells and axodendritic connectivity associated with increased spine density in cortical neurons derived from PS1(-/-) embryos, as well as wild-type neurons treated with gamma-secretase inhibitors. cAMP-dependent signaling was also increased in both the neuroblastoma and cortical neuron systems. As a physiological consequence of increases in axodendritic connectivity and in the magnitude of cAMP-dependent signaling, short- and long-term glutamatergic synaptic transmission was enhanced in PS-deficient neurons. Together, these results demonstrate for the first time that PS-mediated gamma-secretase activity attenuates receptor-mediated intracellular signaling pathways that are critical in regulating glutamatergic synaptic transmission and memory processes.


Subject(s)
Membrane Proteins/metabolism , Memory/physiology , Neurons/metabolism , Second Messenger Systems/physiology , Synaptic Transmission/physiology , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Carbamates/pharmacology , Carbazoles/pharmacology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Line, Tumor/ultrastructure , Cell Membrane/metabolism , Colforsin/pharmacology , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , DCC Receptor , Dipeptides/pharmacology , Endopeptidases , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Genes, DCC , Glutamic Acid/physiology , Indoles/pharmacology , Lidocaine/analogs & derivatives , Lidocaine/pharmacology , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/physiology , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/metabolism , Neurites/ultrastructure , Neuroblastoma/pathology , Neurons/drug effects , Piperazines/pharmacology , Presenilin-1 , Protein Processing, Post-Translational , Pyrroles/pharmacology , Quinoxalines/pharmacology , Rats , Receptors, Cell Surface , Recombinant Fusion Proteins/metabolism , Second Messenger Systems/drug effects , Synaptic Transmission/drug effects , Transfection , Tumor Suppressor Proteins/metabolism
19.
Pharmacol Res ; 50(4): 411-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15304238

ABSTRACT

Mutations in the genes encoding presenilin 1 (PS1) and presenilin 2 (PS2) account for the majority of the cases of familial early-onset Alzheimer's disease (FAD). Presenilins (PSs) facilitate the intramembraneous cleavage of amyloid precursor protein (APP), coined gamma-secretase cleavage, which generates beta-amyloid peptides (A beta). Considerable evidence suggests that FAD-linked PS variants exert their pathogenic influence by selectively elevating the levels of highly fibrillogenic A beta 42 peptides. In addition, numerous other functions have been ascribed to PSs based on subcellular localization, protein interactions, loss of function studies, and intramembraneous gamma-secretase cleavage of growing number of substrates. This review summarizes the diverse physiological functions that are regulated by PSs beyond APP metabolism.


Subject(s)
Alzheimer Disease/etiology , Membrane Proteins/metabolism , Membrane Proteins/physiology , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases , Endopeptidases/metabolism , Humans , Membrane Proteins/genetics , Mutation , Peptide Fragments/metabolism , Presenilin-1
SELECTION OF CITATIONS
SEARCH DETAIL
...