1
|
Haut F, Argyrousi EK, Arancio O. Re-Arranging the Puzzle between the Amyloid-Beta and Tau Pathology: An APP-Centric Approach. Int J Mol Sci 2023; 25:259. [PMID: 38203429 PMCID: PMC10779219 DOI: 10.3390/ijms25010259] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/04/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
After several years of research in the field of Alzheimer's disease (AD), it is still unclear how amyloid-beta (Aβ) and Tau, two key hallmarks of the disease, mediate the neuropathogenic events that lead to AD. Current data challenge the "Amyloid Cascade Hypothesis" that has prevailed in the field of AD, stating that Aβ precedes and triggers Tau pathology that will eventually become the toxic entity in the progression of the disease. This perspective also led the field of therapeutic approaches towards the development of strategies that target Aβ or Tau. In the present review, we discuss recent literature regarding the neurotoxic role of both Aβ and Tau in AD, as well as their physiological function in the healthy brain. Consequently, we present studies suggesting that Aβ and Tau act independently of each other in mediating neurotoxicity in AD, thereafter, re-evaluating the "Amyloid Cascade Hypothesis" that places Tau pathology downstream of Aβ. More recent studies have confirmed that both Aβ and Tau could propagate the disease and induce synaptic and memory impairments via the amyloid precursor protein (APP). This finding is not only interesting from a mechanistic point of view since it provides better insights into the AD pathogenesis but also from a therapeutic point of view since it renders APP a common downstream effector for both Aβ and Tau. Subsequently, therapeutic strategies that act on APP might provide a more viable and physiologically relevant approach for targeting AD.
Collapse
Affiliation(s)
- Florence Haut
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S, New York, NY 10032, USA; (F.H.); (E.K.A.)
| | - Elentina K. Argyrousi
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S, New York, NY 10032, USA; (F.H.); (E.K.A.)
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S, New York, NY 10032, USA; (F.H.); (E.K.A.)
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
2
|
Antonino M, Marmo P, Freites CL, Quassollo GE, Sánchez MF, Lorenzo A, Bignante EA. Aβ Assemblies Promote Amyloidogenic Processing of APP and Intracellular Accumulation of Aβ42 Through Go/Gβγ Signaling. Front Cell Dev Biol 2022; 10:852738. [PMID: 35445022 PMCID: PMC9013780 DOI: 10.3389/fcell.2022.852738] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/07/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by the deposition of aggregated species of amyloid beta (Aβ) in the brain, which leads to progressive cognitive deficits and dementia. Aβ is generated by the successive cleavage of the amyloid precursor protein (APP), first by β-site APP cleaving enzyme 1 (BACE1) and subsequently by the γ-secretase complex. Those conditions which enhace or reduce its clearance predispose to Aβ aggregation and the development of AD. In vitro studies have demonstrated that Aβ assemblies spark a feed-forward loop heightening Aβ production. However, the underlying mechanism remains unknown. Here, we show that oligomers and fibrils of Aβ enhance colocalization and physical interaction of APP and BACE1 in recycling endosomes of human neurons derived from induced pluripotent stem cells and other cell types, which leads to exacerbated amyloidogenic processing of APP and intracellular accumulation of Aβ42. In cells that are overexpressing the mutant forms of APP which are unable to bind Aβ or to activate Go protein, we have found that treatment with aggregated Aβ fails to increase colocalization of APP with BACE1 indicating that Aβ-APP/Go signaling is involved in this process. Moreover, inhibition of Gβγ subunit signaling with βARKct or gallein prevents Aβ-dependent interaction of APP and BACE1 in endosomes, β-processing of APP, and intracellular accumulation of Aβ42. Collectively, our findings uncover a signaling mechanism leading to a feed-forward loop of amyloidogenesis that might contribute to Aβ pathology in the early stages of AD and suggest that gallein could have therapeutic potential.
Collapse
Affiliation(s)
- Magdalena Antonino
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Paula Marmo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Carlos Leandro Freites
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | | | - Alfredo Lorenzo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
- *Correspondence: Elena Anahi Bignante, ; Alfredo Lorenzo,
| | - Elena Anahi Bignante
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Córdoba, Argentina
- *Correspondence: Elena Anahi Bignante, ; Alfredo Lorenzo,
| |
Collapse
|
3
|
Genetic deletion of α7 nicotinic acetylcholine receptors induces an age-dependent Alzheimer's disease-like pathology. Prog Neurobiol 2021; 206:102154. [PMID: 34453977 DOI: 10.1016/j.pneurobio.2021.102154] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/29/2021] [Accepted: 08/18/2021] [Indexed: 11/22/2022]
Abstract
The accumulation of amyloid-beta peptide (Aβ) and the failure of cholinergic transmission are key players in Alzheimer's disease (AD). However, in the healthy brain, Aβ contributes to synaptic plasticity and memory acting through α7 subtype nicotinic acetylcholine receptors (α7nAChRs). Here, we hypothesized that the α7nAChR deletion blocks Aβ physiological function and promotes a compensatory increase in Aβ levels that, in turn, triggers an AD-like pathology. To validate this hypothesis, we studied the age-dependent phenotype of α7 knock out mice. We found that α7nAChR deletion caused an impairment of hippocampal synaptic plasticity and memory at 12 months of age, paralleled by an increase of Amyloid Precursor Protein expression and Aβ levels. This was accompanied by other classical AD features such as a hyperphosphorylation of tau at residues Ser 199, Ser 396, Thr 205, a decrease of GSK-3β at Ser 9, the presence of paired helical filaments and neurofibrillary tangles, neuronal loss and an increase of GFAP-positive astrocytes. Our findings suggest that α7nAChR malfunction might precede Aβ and tau pathology, offering a different perspective to interpret the failure of anti-Aβ therapies against AD and to find novel therapeutical approaches aimed at restoring α7nAChRs-mediated Aβ function at the synapse.
Collapse
|
4
|
Yamamoto K, Yamamoto R, Kato N. Amyloid β and Amyloid Precursor Protein Synergistically Suppress Large-Conductance Calcium-Activated Potassium Channel in Cortical Neurons. Front Aging Neurosci 2021; 13:660319. [PMID: 34149396 PMCID: PMC8211014 DOI: 10.3389/fnagi.2021.660319] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/20/2021] [Indexed: 12/03/2022] Open
Abstract
Intracellular amyloid β (Aβ) injection suppresses the large-conductance calcium-dependent potassium (BK) channel in cortical pyramidal cells from wild-type (WT) mice. In 3xTg Alzheimer’s disease (AD) model mice, intraneuronal Aβ is genetically programed to accumulate, which suppresses the BK channel. However, the mode of BK channel suppression remained unclarified. The present report revealed that only one (11A1) of the three anti-Aβ-oligomer antibodies that we examined, but not anti-monomer-Aβ-antibodies, was effective in recovering BK channel activity in 3xTg neurons. Antibodies against amyloid precursor protein (APP) were also found to be effective, suggesting that APP plays an essential part in this Aβ-oligomer-induced BK channel suppression in 3xTg neurons. In WT neurons, by contrast, APP suppressed BK channels by itself, which suggests that either APP or Aβ is sufficient to block BK channels, thus pointing to a different co-operativity of Aβ and APP in WT and 3xTg neurons. To clarify this difference, we relied on our previous finding that the scaffold protein Homer1a reverses the BK channel blockade in both WT and 3xTg neurons. In cortical neurons from 3xTg mice that bear Homer1a knockout (4xTg mice), neither anti-APP antibodies nor 11A1, but only the 6E10 antibody that binds both APP and Aβ, rescued the BK channel suppression. Given that Homer1a expression is activity dependent and 3xTg neurons are hyperexcitable, Homer1a is likely to be expressed sufficiently in 3xTg neurons, thereby alleviating the suppressive influence of APP and Aβ on BK channel. A unique way that APP modifies Aβ toxicity is thus proposed.
Collapse
Affiliation(s)
- Kenji Yamamoto
- Department of Physiology, Kanazawa Medical University, Ishikawa, Japan.,Department of Neurology and Clinical Research Center, National Hospital Organization Utano National Hospital, Kyoto, Japan
| | - Ryo Yamamoto
- Department of Physiology, Kanazawa Medical University, Ishikawa, Japan
| | - Nobuo Kato
- Department of Physiology, Kanazawa Medical University, Ishikawa, Japan
| |
Collapse
|
5
|
Puzzo D, Argyrousi EK, Staniszewski A, Zhang H, Calcagno E, Zuccarello E, Acquarone E, Fa' M, Li Puma DD, Grassi C, D'Adamio L, Kanaan NM, Fraser PE, Arancio O. Tau is not necessary for amyloid-β-induced synaptic and memory impairments. J Clin Invest 2021; 130:4831-4844. [PMID: 32544084 DOI: 10.1172/jci137040] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 06/10/2020] [Indexed: 12/20/2022] Open
Abstract
The amyloid hypothesis posits that the amyloid-beta (Aβ) protein precedes and requires microtubule-associated protein tau in a sort of trigger-bullet mechanism leading to Alzheimer's disease (AD) pathology. This sequence of events has become dogmatic in the AD field and is used to explain clinical trial failures due to a late start of the intervention when Aβ already activated tau. Here, using a multidisciplinary approach combining molecular biological, biochemical, histopathological, electrophysiological, and behavioral methods, we demonstrated that tau suppression did not protect against Aβ-induced damage of long-term synaptic plasticity and memory, or from amyloid deposition. Tau suppression could even unravel a defect in basal synaptic transmission in a mouse model of amyloid deposition. Similarly, tau suppression did not protect against exogenous oligomeric tau-induced impairment of long-term synaptic plasticity and memory. The protective effect of tau suppression was, in turn, confined to short-term plasticity and memory. Taken together, our data suggest that therapies downstream of Aβ and tau together are more suitable to combat AD than therapies against one or the other alone.
Collapse
Affiliation(s)
- Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Oasi Research Institute-IRCCS, Troina, Italy
| | - Elentina K Argyrousi
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and.,Department of Medicine, Columbia University, New York, New York, USA
| | - Agnieszka Staniszewski
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and.,Department of Medicine, Columbia University, New York, New York, USA
| | - Hong Zhang
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and.,Department of Medicine, Columbia University, New York, New York, USA
| | - Elisa Calcagno
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and.,Department of Medicine, Columbia University, New York, New York, USA
| | - Elisa Zuccarello
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and.,Department of Medicine, Columbia University, New York, New York, USA
| | - Erica Acquarone
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and.,Department of Medicine, Columbia University, New York, New York, USA
| | - Mauro Fa'
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and.,Department of Medicine, Columbia University, New York, New York, USA
| | - Domenica D Li Puma
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico A. Gemelli-IRCCS, Rome, Italy
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico A. Gemelli-IRCCS, Rome, Italy
| | - Luciano D'Adamio
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University, Newark, New Jersey, USA
| | - Nicholas M Kanaan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, and.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, and.,Department of Medicine, Columbia University, New York, New York, USA
| |
Collapse
|
6
|
Spitzer P, Walter M, Göth C, Oberstein TJ, Linning P, Knölker HJ, Kornhuber J, Maler JM. Pharmacological Inhibition of Amyloidogenic APP Processing and Knock-Down of APP in Primary Human Macrophages Impairs the Secretion of Cytokines. Front Immunol 2020; 11:1967. [PMID: 33013850 PMCID: PMC7494750 DOI: 10.3389/fimmu.2020.01967] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 07/21/2020] [Indexed: 12/18/2022] Open
Abstract
It has been previously shown that the amyloid precursor protein (APP) support the innate immune defense as an immune receptor. Amyloid β (Aβ) peptides seem to have properties of an antimicrobial peptide and can act as opsonines. In APP-deficient mouse models, a reduced secretion of cytokines has been observed. Still, it is unclear whether this can be attributed to the lack of APP or to the missing secretion of Aβ peptides. We inhibited the secretion of Aβ peptides in primary human monocyte derived macrophages with the γ-secretase inhibitor N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine-t-butyl-ester (DAPT) or the β-secretase inhibitor GL-189. Alternatively, we knocked down APP by transfection with siRNA. We measured tumor necrosis factor α (TNFα), interleukin 6 (IL-6) and interleukin (IL-10) by enzyme linked immunosorbent assay (ELISA) and evaluated the phagocytotic activity by flow cytometry. We observed reduced concentrations of TNFα and IL-6 in the media of APPk/d macrophages and after inhibition of the β-, or γ-secretase, especially after additional immunological activation with lipopolysaccharide (LPS). Secretion of IL-10 was increased after pharmacological inhibition of APP processing when the macrophages were not immunologically activated but was decreased during LPS-induced inflammation in APPk/d macrophages. No changes of the phagocytotic activity were observed. We conclude that macrophage APP and Aβ peptides support the initiation of an immune response and are involved in the regulation of TNFα, IL-6, and IL-10 secretion by human monocyte-derived macrophages.
Collapse
Affiliation(s)
- Philipp Spitzer
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Erlangen, Germany
| | - Matthias Walter
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Erlangen, Germany
| | - Caroline Göth
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Erlangen, Germany
| | - Timo Jan Oberstein
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Erlangen, Germany
| | - Philipp Linning
- Faculty of Chemistry, Technische Universität Dresden, Dresden, Germany
| | | | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Erlangen, Germany
| | - Juan Manuel Maler
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
7
|
Effect of Aβ Oligomers on Neuronal APP Triggers a Vicious Cycle Leading to the Propagation of Synaptic Plasticity Alterations to Healthy Neurons. J Neurosci 2020; 40:5161-5176. [PMID: 32444385 DOI: 10.1523/jneurosci.2501-19.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/04/2020] [Accepted: 04/03/2020] [Indexed: 01/04/2023] Open
Abstract
Alterations of excitatory synaptic function are the strongest correlate to the pathologic disturbance of cognitive ability observed in the early stages of Alzheimer's disease (AD). This pathologic feature is driven by amyloid-β oligomers (Aβos) and propagates from neuron to neuron. Here, we investigated the mechanism by which Aβos affect the function of synapses and how these alterations propagate to surrounding healthy neurons. We used complementary techniques ranging from electrophysiological recordings and molecular biology to confocal microscopy in primary cortical cultures, and from acute hippocampal and cortical slices from male wild-type and amyloid precursor protein (APP) knock-out (KO) mice to assess the effects of Aβos on glutamatergic transmission, synaptic plasticity, and dendritic spine structure. We showed that extracellular application of Aβos reduced glutamatergic synaptic transmission and long-term potentiation. These alterations were not observed in APP KO neurons, suggesting that APP expression is required. We demonstrated that Aβos/APP interaction increases the amyloidogenic processing of APP leading to intracellular accumulation of newly produced Aβos. Intracellular Aβos participate in synaptic dysfunctions as shown by pharmacological inhibition of APP processing or by intraneuronal infusion of an antibody raised against Aβos. Furthermore, we provide evidence that following APP processing, extracellular release of Aβos mediates the propagation of the synaptic pathology characterized by a decreased spine density of neighboring healthy neurons in an APP-dependent manner. Together, our data unveil a complementary role for Aβos in AD, while intracellular Aβos alter synaptic function, extracellular Aβos promote a vicious cycle that propagates synaptic pathology from diseased to healthy neurons.SIGNIFICANCE STATEMENT Here we provide the proof that a vicious cycle between extracellular and intracellular pools of Aβ oligomers (Aβos) is required for the spreading of Alzheimer's disease (AD) pathology. We showed that extracellular Aβos propagate excitatory synaptic alterations by promoting amyloid precursor protein (APP) processing. Our results also suggest that subsequent to APP cleavage two pools of Aβos are produced. One pool accumulates inside the cytosol, inducing the loss of synaptic plasticity potential. The other pool is released into the extracellular space and contributes to the propagation of the pathology from diseased to healthy neurons. Pharmacological strategies targeting the proteolytic cleavage of APP disrupt the relationship between extracellular and intracellular Aβ, providing a therapeutic approach for the disease.
Collapse
|
8
|
Acquarone E, Argyrousi EK, van den Berg M, Gulisano W, Fà M, Staniszewski A, Calcagno E, Zuccarello E, D’Adamio L, Deng SX, Puzzo D, Arancio O, Fiorito J. Synaptic and memory dysfunction induced by tau oligomers is rescued by up-regulation of the nitric oxide cascade. Mol Neurodegener 2019; 14:26. [PMID: 31248451 PMCID: PMC6598340 DOI: 10.1186/s13024-019-0326-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/05/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Soluble aggregates of oligomeric forms of tau protein (oTau) have been associated with impairment of synaptic plasticity and memory in Alzheimer's disease. However, the molecular mechanisms underlying the synaptic and memory dysfunction induced by elevation of oTau are still unknown. METHODS This work used a combination of biochemical, electrophysiological and behavioral techniques. Biochemical methods included analysis of phosphorylation of the cAMP-responsive element binding (CREB) protein, a transcriptional factor involved in memory, histone acetylation, and expression immediate early genes c-Fos and Arc. Electrophysiological methods included assessment of long-term potentiation (LTP), a type of synaptic plasticity thought to underlie memory formation. Behavioral studies investigated both short-term spatial memory and associative memory. These phenomena were examined following oTau elevation. RESULTS Levels of phospho-CREB, histone 3 acetylation at lysine 27, and immediate early genes c-Fos and Arc, were found to be reduced after oTau elevation during memory formation. These findings led us to explore whether up-regulation of various components of the nitric oxide (NO) signaling pathway impinging onto CREB is capable of rescuing oTau-induced impairment of plasticity, memory, and CREB phosphorylation. The increase of NO levels protected against oTau-induced impairment of LTP through activation of soluble guanylyl cyclase. Similarly, the elevation of cGMP levels and stimulation of the cGMP-dependent protein kinases (PKG) re-established normal LTP after exposure to oTau. Pharmacological inhibition of cGMP degradation through inhibition of phosphodiesterase 5 (PDE5), rescued oTau-induced LTP reduction. These findings could be extrapolated to memory because PKG activation and PDE5 inhibition rescued oTau-induced memory impairment. Finally, PDE5 inhibition re-established normal elevation of CREB phosphorylation and cGMP levels after memory induction in the presence of oTau. CONCLUSIONS Up-regulation of CREB activation through agents acting on the NO cascade might be beneficial against tau-induced synaptic and memory dysfunctions.
Collapse
Affiliation(s)
- Erica Acquarone
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
- DiMi Department of Internal Medicine and Medical Specialties, University of Genoa, 16132 Genoa, Italy
| | - Elentina K. Argyrousi
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
- Faculty of Psychology and Neuroscience, Maastricht University, 6229 Maastricht, Netherlands
| | - Manon van den Berg
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
- Faculty of Psychology and Neuroscience, Maastricht University, 6229 Maastricht, Netherlands
| | - Walter Gulisano
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95125 Catania, Italy
| | - Mauro Fà
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
| | - Agnieszka Staniszewski
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
| | - Elisa Calcagno
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
- Department of Experimental Medicine, Section of General Pathology, School of Medical and Pharmaceutical Sciences, University of Genoa, 16132 Genoa, Italy
| | - Elisa Zuccarello
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
| | - Luciano D’Adamio
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University, Newark, NJ USA
| | - Shi-Xian Deng
- Department of Medicine, Columbia University, New York, NY 10032 USA
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95125 Catania, Italy
- Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
- Department of Medicine, Columbia University, New York, NY 10032 USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032 USA
| | - Jole Fiorito
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
- Department of Life Sciences, New York Institute of Technology, Northern Boulevard P.O. Box 8000, Theobald Science Center, room 425, Old Westbury, NY 11568 USA
| |
Collapse
|
9
|
Visconte C, Canino J, Guidetti GF, Zarà M, Seppi C, Abubaker AA, Pula G, Torti M, Canobbio I. Amyloid precursor protein is required for in vitro platelet adhesion to amyloid peptides and potentiation of thrombus formation. Cell Signal 2018; 52:95-102. [PMID: 30172024 DOI: 10.1016/j.cellsig.2018.08.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/31/2018] [Accepted: 08/28/2018] [Indexed: 01/08/2023]
Abstract
Amyloid precursor protein (APP) is the precursor of amyloid β (Aβ) peptides, whose accumulation in the brain is associated with Alzheimer's disease. APP is also expressed on the platelet surface and Aβ peptides are platelet agonists. The physiological role of APP is largely unknown. In neurons, APP acts as an adhesive receptor, facilitating integrin-mediated cell adhesion, while in platelets it regulates coagulation and venous thrombosis. In this work, we analyzed platelets from APP KO mice to investigate whether membrane APP supports platelet adhesion to physiological and pathological substrates. We found that APP-null platelets adhered and spread normally on collagen, von Willebrand Factor or fibrinogen. However, adhesion on immobilized Aβ peptides Aβ1-40, Aβ1-42 and Aβ25-35 was completely abolished in platelets lacking APP. By contrast, platelet activation and aggregation induced by Aβ peptides occurred normally in the absence of APP. Adhesion of APP-transfected HEK293 to Aβ peptides was significantly higher than that of control cells expressing low levels of APP. Co-coating of Aβ1-42 and Aβ25-35 with collagen strongly potentiated platelet adhesion when whole blood from wild type mice was perfused at arterial shear rate, but had no effects with blood from APP KO mice. These results demonstrate that APP selectively mediates platelet adhesion to Aβ under static condition but not platelet aggregation, and is responsible for Aβ-promoted potentiation of thrombus formation under flow. Therefore, APP may facilitate an early step in thrombus formation when Aβ peptides accumulate in cerebral vessel walls or atherosclerotic plaques.
Collapse
Affiliation(s)
- Caterina Visconte
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Jessica Canino
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy; Scuola Universitaria Superiore, IUSS, Pavia, Italy
| | | | - Marta Zarà
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Claudio Seppi
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | | | - Giordano Pula
- Institute of Biomedical & Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Mauro Torti
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Ilaria Canobbio
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy.
| |
Collapse
|
10
|
Bignante EA, Ponce NE, Heredia F, Musso J, Krawczyk MC, Millán J, Pigino GF, Inestrosa NC, Boccia MM, Lorenzo A. APP/Go protein Gβγ-complex signaling mediates Aβ degeneration and cognitive impairment in Alzheimer's disease models. Neurobiol Aging 2017; 64:44-57. [PMID: 29331876 DOI: 10.1016/j.neurobiolaging.2017.12.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/05/2017] [Accepted: 12/10/2017] [Indexed: 11/15/2022]
Abstract
Deposition of amyloid-β (Aβ), the proteolytic product of the amyloid precursor protein (APP), might cause neurodegeneration and cognitive decline in Alzheimer's disease (AD). However, the direct involvement of APP in the mechanism of Aβ-induced degeneration in AD remains on debate. Here, we analyzed the interaction of APP with heterotrimeric Go protein in primary hippocampal cultures and found that Aβ deposition dramatically enhanced APP-Go protein interaction in dystrophic neurites. APP overexpression rendered neurons vulnerable to Aβ toxicity by a mechanism that required Go-Gβγ complex signaling and p38-mitogen-activated protein kinase activation. Gallein, a selective pharmacological inhibitor of Gβγ complex, inhibited Aβ-induced dendritic and axonal dystrophy, abnormal tau phosphorylation, synaptic loss, and neuronal cell death in hippocampal neurons expressing endogenous protein levels. In the 3xTg-AD mice, intrahippocampal application of gallein reversed memory impairment associated with early Aβ pathology. Our data provide further evidence for the involvement of APP/Go protein in Aβ-induced degeneration and reveal that Gβγ complex is a signaling target potentially relevant for developing therapies for halting Aβ degeneration in AD.
Collapse
Affiliation(s)
- Elena Anahi Bignante
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina; Instituto Universitario de Ciencias Biomédicas de Córdoda (IUCBC), Argentina
| | - Nicolás Eric Ponce
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Florencia Heredia
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Juliana Musso
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María C Krawczyk
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Julieta Millán
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gustavo F Pigino
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Punta Arenas, Chile
| | - Mariano M Boccia
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alfredo Lorenzo
- Instituto de Investigación Médica "Mercedes y Martín Ferreyra", INIMEC-CONICET- Universidad Nacional de Córdoba, Córdoba, Argentina; Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina.
| |
Collapse
|
11
|
Human Brain-Derived Aβ Oligomers Bind to Synapses and Disrupt Synaptic Activity in a Manner That Requires APP. J Neurosci 2017; 37:11947-11966. [PMID: 29101243 DOI: 10.1523/jneurosci.2009-17.2017] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/19/2017] [Accepted: 09/29/2017] [Indexed: 12/20/2022] Open
Abstract
Compelling genetic evidence links the amyloid precursor protein (APP) to Alzheimer's disease (AD) and several theories have been advanced to explain the relationship. A leading hypothesis proposes that a small amphipathic fragment of APP, the amyloid β-protein (Aβ), self-associates to form soluble aggregates that impair synaptic and network activity. Here, we used the most disease-relevant form of Aβ, protein isolated from AD brain. Using this material, we show that the synaptotoxic effects of Aβ depend on expression of APP and that the Aβ-mediated impairment of synaptic plasticity is accompanied by presynaptic effects that disrupt the excitatory/inhibitory (E/I) balance. The net increase in the E/I ratio and inhibition of plasticity are associated with Aβ localizing to synapses and binding of soluble Aβ aggregates to synapses requires the expression of APP. Our findings indicate a role for APP in AD pathogenesis beyond the generation of Aβ and suggest modulation of APP expression as a therapy for AD.SIGNIFICANCE STATEMENT Here, we report on the plasticity-disrupting effects of amyloid β-protein (Aβ) isolated from Alzheimer's disease (AD) brain and the requirement of amyloid precursor protein (APP) for these effects. We show that Aβ-containing AD brain extracts block hippocampal LTP, augment glutamate release probability, and disrupt the excitatory/inhibitory balance. These effects are associated with Aβ localizing to synapses and genetic ablation of APP prevents both Aβ binding and Aβ-mediated synaptic dysfunctions. Our results emphasize the importance of APP in AD and should stimulate new studies to elucidate APP-related targets suitable for pharmacological manipulation.
Collapse
|
12
|
Sosa LJ, Cáceres A, Dupraz S, Oksdath M, Quiroga S, Lorenzo A. The physiological role of the amyloid precursor protein as an adhesion molecule in the developing nervous system. J Neurochem 2017; 143:11-29. [PMID: 28677143 DOI: 10.1111/jnc.14122] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 06/28/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
Abstract
The amyloid precursor protein (APP) is a type I transmembrane glycoprotein better known for its participation in the physiopathology of Alzheimer disease as the source of the beta amyloid fragment. However, the physiological functions of the full length protein and its proteolytic fragments have remained elusive. APP was first described as a cell-surface receptor; nevertheless, increasing evidence highlighted APP as a cell adhesion molecule. In this review, we will focus on the current knowledge of the physiological role of APP as a cell adhesion molecule and its involvement in key events of neuronal development, such as migration, neurite outgrowth, growth cone pathfinding, and synaptogenesis. Finally, since APP is over-expressed in Down syndrome individuals because of the extra copy of chromosome 21, in the last section of the review, we discuss the potential contribution of APP to the neuronal and synaptic defects described in this genetic condition. Read the Editorial Highlight for this article on page 9. Cover Image for this issue: doi. 10.1111/jnc.13817.
Collapse
Affiliation(s)
- Lucas J Sosa
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Alfredo Cáceres
- Laboratorio Neurobiología, Instituto Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina.,Instituto Universitario Ciencias Biomédicas Córdoba, Córdoba, Argentina
| | - Sebastián Dupraz
- Axonal Growth and Regeneration, German Center for Neurodegenarative Diseases, Bonn, Germany
| | - Mariana Oksdath
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Santiago Quiroga
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Alfredo Lorenzo
- Laboratorio de Neuropatología Experimental, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
13
|
Puzzo D, Piacentini R, Fá M, Gulisano W, Li Puma DD, Staniszewski A, Zhang H, Tropea MR, Cocco S, Palmeri A, Fraser P, D'Adamio L, Grassi C, Arancio O. LTP and memory impairment caused by extracellular Aβ and Tau oligomers is APP-dependent. eLife 2017; 6. [PMID: 28696204 PMCID: PMC5529106 DOI: 10.7554/elife.26991] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 07/10/2017] [Indexed: 12/21/2022] Open
Abstract
The concurrent application of subtoxic doses of soluble oligomeric forms of human amyloid-beta (oAβ) and Tau (oTau) proteins impairs memory and its electrophysiological surrogate long-term potentiation (LTP), effects that may be mediated by intra-neuronal oligomers uptake. Intrigued by these findings, we investigated whether oAβ and oTau share a common mechanism when they impair memory and LTP in mice. We found that as already shown for oAβ, also oTau can bind to amyloid precursor protein (APP). Moreover, efficient intra-neuronal uptake of oAβ and oTau requires expression of APP. Finally, the toxic effect of both extracellular oAβ and oTau on memory and LTP is dependent upon APP since APP-KO mice were resistant to oAβ- and oTau-induced defects in spatial/associative memory and LTP. Thus, APP might serve as a common therapeutic target against Alzheimer's Disease (AD) and a host of other neurodegenerative diseases characterized by abnormal levels of Aβ and/or Tau. DOI:http://dx.doi.org/10.7554/eLife.26991.001
Collapse
Affiliation(s)
- Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Roberto Piacentini
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mauro Fá
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, United States
| | - Walter Gulisano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Domenica D Li Puma
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Agnes Staniszewski
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, United States
| | - Hong Zhang
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, United States
| | - Maria Rosaria Tropea
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sara Cocco
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Agostino Palmeri
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Paul Fraser
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Luciano D'Adamio
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, United States
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ottavio Arancio
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New york, United States
| |
Collapse
|
14
|
Xu F, Davis J, Hoos M, Van Nostrand WE. Mutation of the Kunitz-type proteinase inhibitor domain in the amyloid β-protein precursor abolishes its anti-thrombotic properties in vivo. Thromb Res 2017; 155:58-64. [PMID: 28499154 DOI: 10.1016/j.thromres.2017.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/14/2017] [Accepted: 05/02/2017] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Kunitz proteinase inhibitor (KPI) domain-containing forms of the amyloid β-protein precursor (AβPP) inhibit cerebral thrombosis. KPI domain-lacking forms of AβPP are abundant in brain. Regions of AβPP other than the KPI domain may also be involved with regulating cerebral thrombosis. To determine the contribution of the KPI domain to the overall function of AβPP in regulating cerebral thrombosis we generated a reactive center mutant that was devoid of anti-thrombotic activity and studied its anti-thrombotic function in vitro and in vivo. METHODS To determine the extent of KPI function of AβPP in regulating cerebral thrombosis we generated a recombinant reactive center KPIR13I mutant devoid of anti-thrombotic activity. The anti-proteolytic and anti-coagulant properties of wild-type and R13I mutant KPI were investigated in vitro. Cerebral thrombosis of wild-type, AβPP knock out and AβPP/KPIR13I mutant mice was evaluated in experimental models of carotid artery thrombosis and intracerebral hemorrhage. RESULTS Recombinant mutant KPIR13I domain was ineffective in the inhibition of pro-thrombotic proteinases and did not inhibit the clotting of plasma in vitro. AβPP/KPIR13I mutant mice were similarly deficient as AβPP knock out mice in regulating cerebral thrombosis in experimental models of carotid artery thrombosis and intracerebral hemorrhage. CONCLUSIONS We demonstrate that the anti-thrombotic function of AβPP primarily resides in the KPI activity of the protein.
Collapse
Affiliation(s)
- Feng Xu
- Department of Neurosurgery, Stony Brook University, Stony Brook, NY 11794-8122, United States; Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8122, United States
| | - Judianne Davis
- Department of Neurosurgery, Stony Brook University, Stony Brook, NY 11794-8122, United States; Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8122, United States
| | - Michael Hoos
- Department of Neurosurgery, Stony Brook University, Stony Brook, NY 11794-8122, United States; Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8122, United States
| | - William E Van Nostrand
- Department of Neurosurgery, Stony Brook University, Stony Brook, NY 11794-8122, United States; Department of Medicine, Stony Brook University, Stony Brook, NY 11794-8122, United States.
| |
Collapse
|
15
|
Copenhaver PF, Kögel D. Role of APP Interactions with Heterotrimeric G Proteins: Physiological Functions and Pathological Consequences. Front Mol Neurosci 2017; 10:3. [PMID: 28197070 PMCID: PMC5281615 DOI: 10.3389/fnmol.2017.00003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/05/2017] [Indexed: 12/27/2022] Open
Abstract
Following the discovery that the amyloid precursor protein (APP) is the source of β-amyloid peptides (Aβ) that accumulate in Alzheimer’s disease (AD), structural analyses suggested that the holoprotein resembles a transmembrane receptor. Initial studies using reconstituted membranes demonstrated that APP can directly interact with the heterotrimeric G protein Gαo (but not other G proteins) via an evolutionarily G protein-binding motif in its cytoplasmic domain. Subsequent investigations in cell culture showed that antibodies against the extracellular domain of APP could stimulate Gαo activity, presumably mimicking endogenous APP ligands. In addition, chronically activating wild type APP or overexpressing mutant APP isoforms linked with familial AD could provoke Go-dependent neurotoxic responses, while biochemical assays using human brain samples suggested that the endogenous APP-Go interactions are perturbed in AD patients. More recently, several G protein-dependent pathways have been implicated in the physiological roles of APP, coupled with evidence that APP interacts both physically and functionally with Gαo in a variety of contexts. Work in insect models has demonstrated that the APP ortholog APPL directly interacts with Gαo in motile neurons, whereby APPL-Gαo signaling regulates the response of migratory neurons to ligands encountered in the developing nervous system. Concurrent studies using cultured mammalian neurons and organotypic hippocampal slice preparations have shown that APP signaling transduces the neuroprotective effects of soluble sAPPα fragments via modulation of the PI3K/Akt pathway, providing a mechanism for integrating the stress and survival responses regulated by APP. Notably, this effect was also inhibited by pertussis toxin, indicating an essential role for Gαo/i proteins. Unexpectedly, C-terminal fragments (CTFs) derived from APP have also been found to interact with Gαs, whereby CTF-Gαs signaling can promote neurite outgrowth via adenylyl cyclase/PKA-dependent pathways. These reports offer the intriguing perspective that G protein switching might modulate APP-dependent responses in a context-dependent manner. In this review, we provide an up-to-date perspective on the model that APP plays a variety of roles as an atypical G protein-coupled receptor in both the developing and adult nervous system, and we discuss the hypothesis that disruption of these normal functions might contribute to the progressive neuropathologies that typify AD.
Collapse
Affiliation(s)
- Philip F Copenhaver
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Sciences University, Portland OR, USA
| | - Donat Kögel
- Experimental Neurosurgery, Goethe University Frankfurt Frankfurt am Main, Germany
| |
Collapse
|
16
|
Preat T, Goguel V. Role of Drosophila Amyloid Precursor Protein in Memory Formation. Front Mol Neurosci 2016; 9:142. [PMID: 28008309 PMCID: PMC5143682 DOI: 10.3389/fnmol.2016.00142] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 11/28/2016] [Indexed: 12/22/2022] Open
Abstract
The amyloid precursor protein (APP) is a membrane protein engaged in complex proteolytic pathways. APP and its derivatives have been shown to play a central role in Alzheimer’s disease (AD), a progressive neurodegenerative disease characterized by memory decline. Despite a huge effort from the research community, the primary cause of AD remains unclear, making it crucial to better understand the physiological role of the APP pathway in brain plasticity and memory. Drosophila melanogaster is a model system well-suited to address this issue. Although relatively simple, the fly brain is highly organized, sustains several forms of learning and memory, and drives numerous complex behaviors. Importantly, molecules and mechanisms underlying memory processes are conserved from flies to mammals. The fly encodes a single non-essential APP homolog named APP-Like (APPL). Using in vivo inducible RNA interference strategies, it was shown that APPL knockdown in the mushroom bodies (MB)—the central integrative brain structure for olfactory memory—results in loss of memory. Several APPL derivatives, such as secreted and full-length membrane APPL, may play different roles in distinct types of memory phases. Furthermore, overexpression of Drosophila amyloid peptide exacerbates the memory deficit caused by APPL knockdown, thus potentiating memory decline. Data obtained in the fly support the hypothesis that APP acts as a transmembrane receptor, and that disruption of its normal function may contribute to cognitive impairment during early AD.
Collapse
Affiliation(s)
- Thomas Preat
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, Centre National de la Recherche Scientifique (CNRS), ESPCI Paris, PSL Research University Paris, France
| | - Valérie Goguel
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, Centre National de la Recherche Scientifique (CNRS), ESPCI Paris, PSL Research University Paris, France
| |
Collapse
|
17
|
Manocha GD, Floden AM, Rausch K, Kulas JA, McGregor BA, Rojanathammanee L, Puig KR, Puig KL, Karki S, Nichols MR, Darland DC, Porter JE, Combs CK. APP Regulates Microglial Phenotype in a Mouse Model of Alzheimer's Disease. J Neurosci 2016; 36:8471-86. [PMID: 27511018 PMCID: PMC4978805 DOI: 10.1523/jneurosci.4654-15.2016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 05/20/2016] [Accepted: 06/24/2016] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED Prior work suggests that amyloid precursor protein (APP) can function as a proinflammatory receptor on immune cells, such as monocytes and microglia. Therefore, we hypothesized that APP serves this function in microglia during Alzheimer's disease. Although fibrillar amyloid β (Aβ)-stimulated cytokine secretion from both wild-type and APP knock-out (mAPP(-/-)) microglial cultures, oligomeric Aβ was unable to stimulate increased secretion from mAPP(-/-) cells. This was consistent with an ability of oligomeric Aβ to bind APP. Similarly, intracerebroventricular infusions of oligomeric Aβ produced less microgliosis in mAPP(-/-) mice compared with wild-type mice. The mAPP(-/-) mice crossed to an APP/PS1 transgenic mouse line demonstrated reduced microgliosis and cytokine levels and improved memory compared with wild-type mice despite robust fibrillar Aβ plaque deposition. These data define a novel function for microglial APP in regulating their ability to acquire a proinflammatory phenotype during disease. SIGNIFICANCE STATEMENT A hallmark of Alzheimer's disease (AD) brains is the accumulation of amyloid β (Aβ) peptide within plaques robustly invested with reactive microglia. This supports the notion that Aβ stimulation of microglial activation is one source of brain inflammatory changes during disease. Aβ is a cleavage product of the ubiquitously expressed amyloid precursor protein (APP) and is able to self-associate into a wide variety of differently sized and structurally distinct multimers. In this study, we demonstrate both in vitro and in vivo that nonfibrillar, oligomeric forms of Aβ are able to interact with the parent APP protein to stimulate microglial activation. This provides a mechanism by which metabolism of APP results in possible autocrine or paracrine Aβ production to drive the microgliosis associated with AD brains.
Collapse
Affiliation(s)
- Gunjan D Manocha
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Angela M Floden
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Keiko Rausch
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Joshua A Kulas
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Brett A McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Lalida Rojanathammanee
- Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, 30000 Thailand
| | - Kelley R Puig
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Kendra L Puig
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Sanjib Karki
- Department of Chemistry and Biochemistry, University of Missouri-St. Louis, St. Louis, Missouri 63121-4400, and
| | - Michael R Nichols
- Department of Chemistry and Biochemistry, University of Missouri-St. Louis, St. Louis, Missouri 63121-4400, and
| | - Diane C Darland
- Department of Biology, University of North Dakota, Grand Forks, North Dakota 58202
| | - James E Porter
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203
| | - Colin K Combs
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203,
| |
Collapse
|
18
|
APP Receptor? To Be or Not To Be. Trends Pharmacol Sci 2016; 37:390-411. [PMID: 26837733 DOI: 10.1016/j.tips.2016.01.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 11/22/2022]
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.
Collapse
|
19
|
Van Nostrand WE. The influence of the amyloid ß-protein and its precursor in modulating cerebral hemostasis. Biochim Biophys Acta Mol Basis Dis 2015; 1862:1018-26. [PMID: 26519139 DOI: 10.1016/j.bbadis.2015.10.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/20/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023]
Abstract
Ischemic and hemorrhagic strokes are a significant cause of brain injury leading to vascular cognitive impairment and dementia (VCID). These deleterious events largely result from disruption of cerebral hemostasis, a well-controlled and delicate balance between thrombotic and fibrinolytic pathways in cerebral blood vessels and surrounding brain tissue. Ischemia and hemorrhage are both commonly associated with cerebrovascular deposition of amyloid ß-protein (Aß). In this regard, Aß directly and indirectly modulates cerebral thrombosis and fibrinolysis. Further, major isoforms of the Aß precursor protein (AßPP) function as a potent inhibitor of pro-thrombotic proteinases. The purpose of this review article is to summarize recent research on how cerebral vascular Aß and AßPP influence cerebral hemostasis. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia, edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- William E Van Nostrand
- Department of Neurosurgery, HSC-T12/086, Stony Brook University, Stony Brook, NY 11794-8122, USA; Department of Medicine, HSC-T12/086, Stony Brook University, Stony Brook, NY 11794-8122, USA.
| |
Collapse
|
20
|
Bourdet I, Lampin-Saint-Amaux A, Preat T, Goguel V. Amyloid-β Peptide Exacerbates the Memory Deficit Caused by Amyloid Precursor Protein Loss-of-Function in Drosophila. PLoS One 2015; 10:e0135741. [PMID: 26274614 PMCID: PMC4537105 DOI: 10.1371/journal.pone.0135741] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 07/26/2015] [Indexed: 11/18/2022] Open
Abstract
The amyloid precursor protein (APP) plays a central role in Alzheimer's disease (AD). APP can undergo two exclusive proteolytic pathways: cleavage by the α-secretase initiates the non-amyloidogenic pathway while cleavage by the β-secretase initiates the amyloidogenic pathway that leads, after a second cleavage by the γ-secretase, to amyloid-β (Aβ) peptides that can form toxic extracellular deposits, a hallmark of AD. The initial events leading to AD are still unknown. Importantly, aside from Aβ toxicity whose molecular mechanisms remain elusive, several studies have shown that APP plays a positive role in memory, raising the possibility that APP loss-of-function may participate to AD. We previously showed that APPL, the Drosophila APP ortholog, is required for associative memory in young flies. In the present report, we provide the first analysis of the amyloidogenic pathway's influence on memory in the adult. We show that transient overexpression of the β-secretase in the mushroom bodies, the center for olfactory memory, did not alter memory. In sharp contrast, β-secretase overexpression affected memory when associated with APPL partial loss-of-function. Interestingly, similar results were observed with Drosophila Aβ peptide. Because Aβ overexpression impaired memory only when combined to APPL partial loss-of-function, the data suggest that Aβ affects memory through the APPL pathway. Thus, memory is altered by two connected mechanisms-APPL loss-of-function and amyloid peptide toxicity-revealing in Drosophila a functional interaction between APPL and amyloid peptide.
Collapse
Affiliation(s)
- Isabelle Bourdet
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, CNRS, ESPCI-ParisTech, PSL Research University, 10 rue Vauquelin, 75005 Paris, France
| | - Aurélie Lampin-Saint-Amaux
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, CNRS, ESPCI-ParisTech, PSL Research University, 10 rue Vauquelin, 75005 Paris, France
| | - Thomas Preat
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, CNRS, ESPCI-ParisTech, PSL Research University, 10 rue Vauquelin, 75005 Paris, France
- * E-mail: (VG); (TP)
| | - Valérie Goguel
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, CNRS, ESPCI-ParisTech, PSL Research University, 10 rue Vauquelin, 75005 Paris, France
- * E-mail: (VG); (TP)
| |
Collapse
|
21
|
Canobbio I, Abubaker AA, Visconte C, Torti M, Pula G. Role of amyloid peptides in vascular dysfunction and platelet dysregulation in Alzheimer's disease. Front Cell Neurosci 2015; 9:65. [PMID: 25784858 PMCID: PMC4347625 DOI: 10.3389/fncel.2015.00065] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/11/2015] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative cause of dementia in the elderly. AD is accompanied by the accumulation of amyloid peptides in the brain parenchyma and in the cerebral vessels. The sporadic form of AD accounts for about 95% of all cases. It is characterized by a late onset, typically after the age of 65, with a complex and still poorly understood aetiology. Several observations point towards a central role of cerebrovascular dysfunction in the onset of sporadic AD (SAD). According to the "vascular hypothesis", AD may be initiated by vascular dysfunctions that precede and promote the neurodegenerative process. In accordance to this, AD patients show increased hemorrhagic or ischemic stroke risks. It is now clear that multiple bidirectional connections exist between AD and cerebrovascular disease, and in this new scenario, the effect of amyloid peptides on vascular cells and blood platelets appear to be central to AD. In this review, we analyze the effect of amyloid peptides on vascular function and platelet activation and its contribution to the cerebrovascular pathology associated with AD and the progression of this disease.
Collapse
Affiliation(s)
- Ilaria Canobbio
- Department of Biology and Biotechnology, Unit of Biochemistry, University of Pavia Pavia, Italy
| | - Aisha Alsheikh Abubaker
- Department of Biology and Biotechnology, Unit of Biochemistry, University of Pavia Pavia, Italy
| | - Caterina Visconte
- Department of Biology and Biotechnology, Unit of Biochemistry, University of Pavia Pavia, Italy
| | - Mauro Torti
- Department of Biology and Biotechnology, Unit of Biochemistry, University of Pavia Pavia, Italy
| | - Giordano Pula
- Department of Biology and Biotechnology, Unit of Biochemistry, University of Pavia Pavia, Italy
| |
Collapse
|
22
|
Fogel H, Frere S, Segev O, Bharill S, Shapira I, Gazit N, O'Malley T, Slomowitz E, Berdichevsky Y, Walsh DM, Isacoff EY, Hirsch JA, Slutsky I. APP homodimers transduce an amyloid-β-mediated increase in release probability at excitatory synapses. Cell Rep 2014; 7:1560-1576. [PMID: 24835997 DOI: 10.1016/j.celrep.2014.04.024] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 03/12/2014] [Accepted: 04/12/2014] [Indexed: 10/25/2022] Open
Abstract
Accumulation of amyloid-β peptides (Aβ), the proteolytic products of the amyloid precursor protein (APP), induces a variety of synaptic dysfunctions ranging from hyperactivity to depression that are thought to cause cognitive decline in Alzheimer's disease. While depression of synaptic transmission has been extensively studied, the mechanisms underlying synaptic hyperactivity remain unknown. Here, we show that Aβ40 monomers and dimers augment release probability through local fine-tuning of APP-APP interactions at excitatory hippocampal boutons. Aβ40 binds to the APP, increases the APP homodimer fraction at the plasma membrane, and promotes APP-APP interactions. The APP activation induces structural rearrangements in the APP/Gi/o-protein complex, boosting presynaptic calcium flux and vesicle release. The APP growth-factor-like domain (GFLD) mediates APP-APP conformational changes and presynaptic enhancement. Thus, the APP homodimer constitutes a presynaptic receptor that transduces signal from Aβ40 to glutamate release. Excessive APP activation may initiate a positive feedback loop, contributing to hippocampal hyperactivity in Alzheimer's disease.
Collapse
Affiliation(s)
- Hilla Fogel
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Samuel Frere
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Oshik Segev
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Shashank Bharill
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Ilana Shapira
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Neta Gazit
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Tiernan O'Malley
- Laboratory for Neurodegenerative Research, School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin 4, Republic of Ireland; Laboratory for Neurodegenerative Research, Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Institutes of Medicine, Boston, MA 02115, USA
| | - Edden Slomowitz
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Yevgeny Berdichevsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Dominic M Walsh
- Laboratory for Neurodegenerative Research, Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Institutes of Medicine, Boston, MA 02115, USA
| | - Ehud Y Isacoff
- Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Joel A Hirsch
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
23
|
Immobilized amyloid Aβ peptides support platelet adhesion and activation. FEBS Lett 2013; 587:2606-11. [PMID: 23831058 DOI: 10.1016/j.febslet.2013.06.041] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/10/2013] [Accepted: 06/24/2013] [Indexed: 01/26/2023]
Abstract
Accumulation of amyloidogenic Aβ peptides in the brain contributes to the onset of Alzheimer disease. Aβ peptide deposits are also present in blood vessel walls, mainly deriving from circulating platelets. However, their effect on platelet function is unclear. We demonstrate that immobilized Aβ peptides induce platelet adhesion and spreading through metalloproteinase-sensitive surface receptors. Aβ peptides also fasten platelet spreading on collagen, and support the time- and ADP-dependent activation of adherent platelets, leading to stimulation of several signalling proteins. Our results indicate a potential role for peripheral Aβ peptides in promoting platelet adhesion and activation in the initiation of thrombus formation.
Collapse
|
24
|
Amyloid β precursor protein as a molecular target for amyloid β--induced neuronal degeneration in Alzheimer's disease. Neurobiol Aging 2013; 34:2525-37. [PMID: 23714735 DOI: 10.1016/j.neurobiolaging.2013.04.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/17/2013] [Accepted: 04/20/2013] [Indexed: 11/23/2022]
Abstract
A role of amyloid β (Aβ) peptide aggregation and deposition in Alzheimer's disease (AD) pathogenesis is widely accepted. Significantly, abnormalities induced by aggregated Aβ have been linked to synaptic and neuritic degeneration, consistent with the "dying-back" pattern of degeneration that characterizes neurons affected in AD. However, molecular mechanisms underlying the toxic effect of aggregated Aβ remain elusive. In the last 2 decades, a variety of aggregated Aβ species have been identified and their toxic properties demonstrated in diverse experimental systems. Concurrently, specific Aβ assemblies have been shown to interact and misregulate a growing number of molecular effectors with diverse physiological functions. Such pleiotropic effects of aggregated Aβ posit a mayor challenge for the identification of the most cardinal Aβ effectors relevant to AD pathology. In this review, we discuss recent experimental evidence implicating amyloid β precursor protein (APP) as a molecular target for toxic Aβ assemblies. Based on a significant body of pathologic observations and experimental evidence, we propose a novel pathologic feed-forward mechanism linking Aβ aggregation to abnormalities in APP processing and function, which in turn would trigger the progressive loss of neuronal connectivity observed early in AD.
Collapse
|
25
|
Brinkmalm G, Portelius E, Öhrfelt A, Mattsson N, Persson R, Gustavsson MK, Vite CH, Gobom J, Månsson JE, Nilsson J, Halim A, Larson G, Rüetschi U, Zetterberg H, Blennow K, Brinkmalm A. An online nano-LC-ESI-FTICR-MS method for comprehensive characterization of endogenous fragments from amyloid β and amyloid precursor protein in human and cat cerebrospinal fluid. JOURNAL OF MASS SPECTROMETRY : JMS 2012; 47:591-603. [PMID: 22576872 DOI: 10.1002/jms.2987] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Amyloid precursor protein (APP) is the precursor protein to amyloid β (Aβ), the main constituent of senile plaques in Alzheimer's disease (AD). Endogenous Aβ peptides reflect the APP processing, and greater knowledge of different APP degradation pathways is important to understand the mechanism underlying AD pathology. When one analyzes longer Aβ peptides by low-energy collision-induced dissociation tandem mass spectrometry (MS/MS), mainly long b-fragments are observed, limiting the possibility to determine variations such as amino acid variants or post-translational modifications (PTMs) within the N-terminal half of the peptide. However, by using electron capture dissociation (ECD), we obtained a more comprehensive sequence coverage for several APP/Aβ peptide species, thus enabling a deeper characterization of possible variants and PTMs. Abnormal APP/Aβ processing has also been described in the lysosomal storage disease Niemann-Pick type C and the major large animal used for studying this disease is cat. By ECD MS/MS, a substitution of Asp7 → Glu in cat Aβ was identified. Further, sialylated core 1 like O-glycans at Tyr10, recently discovered in human Aβ (a previously unknown glycosylation type), were identified also in cat cerebrospinal fluid (CSF). It is therefore likely that this unusual type of glycosylation is common for (at least) species belonging to the magnorder Boreoeutheria. We here describe a detailed characterization of endogenous APP/Aβ peptide species in CSF by using an online top-down MS-based method.
Collapse
Affiliation(s)
- Gunnar Brinkmalm
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Libeu CP, Poksay KS, John V, Bredesen DE. Structural and functional alterations in amyloid-β precursor protein induced by amyloid-β peptides. J Alzheimers Dis 2011; 25:547-66. [PMID: 21471643 DOI: 10.3233/jad-2011-101938] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease-associated amyloid-β (Aβ) peptide is neurotoxic as an oligomer, but not as a monomer, by an unknown mechanism. We showed previously that Aβ interacts with the amyloid-β precursor protein (AβPP), leading to caspase cleavage and cell death induction. To characterize this structure and interaction further, we purified the extracellular domain of AβPP695 (eAβPP) and its complex with Aβ oligomers (AβOs) of varying sizes, and then performed small angle X-ray scattering (SAXS). In the absence of any Aβ, eAβPP was a compact homodimer with a tight association between the E1 and E2 domains. Dimeric Aβ oligomers induced monomerization of eAβPP while larger oligomers also bound eAβPP but preserved the homodimer. Efficient binding of the larger oligomers correlated with the presence of prefibrillar oligomers, suggesting that the eAβPP binding is limited to a conformational subset of Aβ oligomers. Both forms of Aβ bound to eAβPP at the Aβ-cognate region and induced dissociation of the E1 and E2 domains. Our data provide the first structural evidence for Aβ-AβPP binding and suggest a mechanism for differential modulation of AβPP processing and cell death signaling by Aβ dimers versus conformationally-specific larger oligomers.
Collapse
|
27
|
Abstract
Alzheimer's disease (AD) is an age-associated disease characterized by increased accumulation of extracellular amyloid-β (Aβ) plaques within the brain. Histological examination has also revealed profound microglial activation in diseased brains often in association with these fibrillar peptide aggregates. The paradoxical presence of increased, reactive microglia yet accumulating extracellular debris suggests that these cells may be phagocytically compromised during disease. Prior work has demonstrated that primary microglia from adult mice are unable to phagocytose fibrillar Aβ1-42 in vitro when compared to microglia cultured from early postnatal animals. These data suggest that microglia undergo an age-associated decrease in microglial ability to interact with Aβ fibrils. In order to better define a temporal profile of microglia-Aβ interaction, acutely isolated, rather than cultured, microglia from 2 month, 6 month, and postnatal day 0 C57BL/6 mice were compared. Postnatal day 0 microglia demonstrated a CD47 dependent ability to phagocytose Aβ fibrils that was lost by 6 months. This corresponded with the ability of postnatal day 0 but not adult microglia to decrease Aβ immunoreactive plaque load from AD sections in vitro. In spite of limited Aβ uptake ability, adult microglia had functional phagocytic uptake of bacterial bioparticles and demonstrated the ability to adhere to both Aβ plaques and in vitro fibrillized Aβ. These data demonstrate a temporal profile of specifically Aβ-microglia interaction with a critical developmental period at 6 months in which cells remain able to interact with Aβ fibrils but lose their ability to phagocytose it.
Collapse
Affiliation(s)
- Angela Marie Floden
- Dept. of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, 504 Hamline Street, Neuroscience Building, Grand Forks, ND 58203-9037, , Phone: 701-777-2873, Fax: 701-777-4490
| | - Colin Kelly Combs
- Dept. of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, 504 Hamline Street, Neuroscience Building, Grand Forks, ND 58203-9037, , Phone: 701-777-4025, Fax: 701-777-4490
| |
Collapse
|
28
|
Kedikian G, Heredia F, Salvador VR, Raimunda D, Isoardi N, Heredia L, Lorenzo A. Secreted amyloid precursor protein and holo-APP bind amyloid beta through distinct domains eliciting different toxic responses on hippocampal neurons. J Neurosci Res 2010; 88:1795-803. [PMID: 20155808 DOI: 10.1002/jnr.22347] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Amyloid beta (Abeta) is a metabolic product of Abeta precursor protein (APP). Deposition of Abeta in the brain and neuronal degeneration are characteristic hallmarks of Alzheimer's disease (AD). Abeta induces neuronal degeneration, but the mechanism of neurotoxicity remains elusive. Increasing evidence implicates APP as a receptor-like protein for Abeta fibrils (fAbeta). In this study, we present further experimental support for the direct interaction of APP with fAbeta and for its involvement in Abeta neurotoxicity. Using recombinant purified holo-APP (h-APP), we have shown that it directly binds fAbeta. Employing deletion mutant forms of APP, we show that two different sequences are involved in the binding of APP to fAbeta. One sequence in the n-terminus of APP is required for binding of fAbeta to secreted APP (s-APP) but not to h-APP. In addition, the extracellular juxtamembrane Abeta-sequence mediates binding of fAbeta to h-APP but not to s-APP. Deletion of the extracellular juxtamembrane Abeta sequence abolishes abnormal h-APP accumulation and toxicity induced by fAbeta deposition, whereas deletions in the n-terminus of APP do not affect Abeta toxicity. These experiments show that interaction of toxic Abeta species with its membrane-anchored parental protein promotes toxicity in hippocampal neurons, adding further support to an Abeta-receptor-like function of APP directly implicated in neuronal degeneration in AD.
Collapse
Affiliation(s)
- Gabriela Kedikian
- Laboratory of Experimental Neuropathology, Instituto de Investigación Médica "Mercedes y Martín Ferreyra," INIMEC-CONICET, Córdoba, Argentina
| | | | | | | | | | | | | |
Collapse
|
29
|
Identification of novel N-terminal fragments of amyloid precursor protein in cerebrospinal fluid. Exp Neurol 2010; 223:351-8. [DOI: 10.1016/j.expneurol.2009.06.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 06/11/2009] [Accepted: 06/17/2009] [Indexed: 11/19/2022]
|
30
|
Amyloid precursor protein mediates a tyrosine kinase-dependent activation response in endothelial cells. J Neurosci 2009; 29:14451-62. [PMID: 19923279 DOI: 10.1523/jneurosci.3107-09.2009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Amyloid precursor protein (APP) is a ubiquitously expressed type 1 integral membrane protein. It has the ability to bind numerous extracellular matrix components and propagate signaling responses via its cytoplasmic phospho-tyrosine, (682)YENPTY(687), binding motif. We recently demonstrated increased protein levels of APP, phosphorylated APP (Tyr682), and beta-amyloid (Abeta) in brain vasculature of atherosclerotic and Alzheimer's disease (AD) tissue colocalizing primarily within the endothelial layer. This study demonstrates similar APP changes in peripheral vasculature from human and mouse apoE(-/-) aorta, suggesting that APP-related changes are not restricted to brain vasculature. Therefore, primary mouse aortic endothelial cells and human umbilical vein endothelial cells were used as a model system to examine the function of APP in endothelial cells. APP multimerization with an anti-N-terminal APP antibody, 22C11, to simulate ligand binding stimulated an Src kinase family-dependent increase in protein phospho-tyrosine levels, APP phosphorylation, and Abeta secretion. Furthermore, APP multimerization stimulated increased protein levels of the proinflammatory proteins, cyclooxygenase-2 and vascular cell adhesion molecule-1 also in an Src kinase family-dependent manner. Endothelial APP was also involved in mediating monocytic cell adhesion. Collectively, these data demonstrate that endothelial APP regulates immune cell adhesion and stimulates a tyrosine kinase-dependent response driving acquisition of a reactive endothelial phenotype. These APP-mediated events may serve as therapeutic targets for intervention in progressive vascular changes common to cerebrovascular disease and AD.
Collapse
|
31
|
Xu F, Previti ML, Nieman MT, Davis J, Schmaier AH, Van Nostrand WE. AbetaPP/APLP2 family of Kunitz serine proteinase inhibitors regulate cerebral thrombosis. J Neurosci 2009; 29:5666-70. [PMID: 19403832 PMCID: PMC2719965 DOI: 10.1523/jneurosci.0095-09.2009] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Revised: 03/31/2009] [Accepted: 04/03/2009] [Indexed: 11/21/2022] Open
Abstract
The amyloid beta-protein precursor (AbetaPP) is best recognized as the precursor to the Abeta peptide that accumulates in the brains of patients with Alzheimer's disease, but less is known about its physiological functions. Isoforms of AbetaPP that contain a Kunitz-type serine proteinase inhibitor (KPI) domain are expressed in brain and, outside the CNS, in circulating blood platelets. Recently, we showed that KPI-containing forms of AbetaPP regulates cerebral thrombosis in vivo (Xu et al., 2005, 2007). Amyloid precursor like protein-2 (APLP2), a closely related homolog to AbetaPP, also possesses a highly conserved KPI domain. Virtually nothing is known of its function. Here, we show that APLP2 also regulates cerebral thrombosis risk. Recombinant purified KPI domains of AbetaPP and APLP2 both inhibit the plasma clotting in vitro. In a carotid artery thrombosis model, both AbetaPP(-/-) and APLP2(-/-) mice exhibit similar significantly shorter times to vessel occlusion compared with wild-type mice indicating a prothrombotic phenotype. Similarly, in an experimental model of intracerebral hemorrhage, both AbetaPP(-/-) and APLP2(-/-) mice produce significantly smaller hematomas with reduced brain hemoglobin content compared with wild-type mice. Together, these results indicate that AbetaPP and APLP2 share overlapping anticoagulant functions with regard to regulating thrombosis after cerebral vascular injury.
Collapse
Affiliation(s)
- Feng Xu
- Department of Medicine, Stony Brook University, Stony Brook, New York 11794-8153, and
| | - Mary Lou Previti
- Department of Medicine, Stony Brook University, Stony Brook, New York 11794-8153, and
| | - Marvin T. Nieman
- Department of Medicine, Division of Hematology/Oncology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Judianne Davis
- Department of Medicine, Stony Brook University, Stony Brook, New York 11794-8153, and
| | - Alvin H. Schmaier
- Department of Medicine, Division of Hematology/Oncology, Case Western Reserve University, Cleveland, Ohio 44106
| | | |
Collapse
|
32
|
Austin SA, Combs CK. Amyloid precursor protein mediates monocyte adhesion in AD tissue and apoE(-)/(-) mice. Neurobiol Aging 2008; 31:1854-66. [PMID: 19058878 DOI: 10.1016/j.neurobiolaging.2008.10.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 10/07/2008] [Accepted: 10/17/2008] [Indexed: 10/21/2022]
Abstract
Amyloid precursor protein (APP) is a type 1 integral membrane protein, which is highly conserved and ubiquitously expressed. Numerous data suggest it functions in cellular adhesion. For example, APP binds components of the extracellular matrix to propagate intracellular signaling responses. In order to investigate adhesion-related changes in inflamed vasculature, brains from apolipoprotein E(-/-) (apoE(-/-)) mice were examined for changes related to APP then compared to human Alzheimer's disease (AD) brains. Cerebrovasculature from mouse apoE(-)/(-) and human AD brains revealed strong immunoreactivity for APP, APP phosphorylated at tyrosine residue 682 (pAPP) and Aβ. Further, Western blot analyses from mouse apoE(-/-) and AD brains showed statistically higher protein levels of APP, pAPP and increased APP association with the tyrosine kinase, Src. Lastly, utilizing a modified Stamper-Woodruff adhesion assay, we demonstrated that adhesion of monocytic cells to apoE(-/-) and AD brain endothelium is partially APP dependent. These data suggest that endothelial APP function coupled with increased Aβ production are involved in the vascular dysfunction associated with atherosclerosis and AD.
Collapse
Affiliation(s)
- Susan A Austin
- Department of Pharmacology, Physiology & Therapeutics, University of North Dakota, School of Medicine and Health Sciences, 504 Hamline St., Room 116, Grand Forks, ND 58203, United States
| | | |
Collapse
|
33
|
Fawzi NL, Phillips AH, Ruscio JZ, Doucleff M, Wemmer DE, Head-Gordon T. Structure and dynamics of the Abeta(21-30) peptide from the interplay of NMR experiments and molecular simulations. J Am Chem Soc 2008; 130:6145-58. [PMID: 18412346 PMCID: PMC3474854 DOI: 10.1021/ja710366c] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We combine molecular dynamics simulations and new high-field NMR experiments to describe the solution structure of the Abeta(21-30) peptide fragment that may be relevant for understanding structural mechanisms related to Alzheimer's disease. By using two different empirical force-field combinations, we provide predictions of the three-bond scalar coupling constants ((3)J(H(N)H(alpha))), chemical-shift values, (13)C relaxation parameters, and rotating-frame nuclear Overhauser effect spectroscopy (ROESY) crosspeaks that can then be compared directly to the same observables measured in the corresponding NMR experiment of Abeta(21-30). We find robust prediction of the (13)C relaxation parameters and medium-range ROESY crosspeaks by using new generation TIP4P-Ew water and Amber ff99SB protein force fields, in which the NMR validates that the simulation yields both a structurally and dynamically correct ensemble over the entire Abeta(21-30) peptide. Analysis of the simulated ensemble shows that all medium-range ROE restraints are not satisfied simultaneously and demonstrates the structural diversity of the Abeta(21-30) conformations more completely than when determined from the experimental medium-range ROE restraints alone. We find that the structural ensemble of the Abeta(21-30) peptide involves a majority population (approximately 60%) of unstructured conformers, lacking any secondary structure or persistent hydrogen-bonding networks. However, the remaining minority population contains a substantial percentage of conformers with a beta-turn centered at Val24 and Gly25, as well as evidence of the Asp23 to Lys28 salt bridge important to the fibril structure. This study sets the stage for robust theoretical work on Abeta(1-40) and Abeta(1-42), for which collection of detailed NMR data on the monomer will be more challenging because of aggregation and fibril formation on experimental timescales at physiological conditions. In addition, we believe that the interplay of modern molecular simulation and high-quality NMR experiments has reached a fruitful stage for characterizing structural ensembles of disordered peptides and proteins in general.
Collapse
Affiliation(s)
- Nicolas L. Fawzi
- UCSF/UC Berkeley Joint Graduate Group in Bioengineering, Berkeley, California 94720
| | - Aaron H. Phillips
- Department of Chemistry, University of California, Berkeley, California 94720
| | - Jory Z. Ruscio
- Department of Bioengineering, University of California, Berkeley, California 94720
| | - Michaeleen Doucleff
- Department of Chemistry, University of California, Berkeley, California 94720
| | - David E. Wemmer
- Department of Chemistry, University of California, Berkeley, California 94720
- Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | - Teresa Head-Gordon
- UCSF/UC Berkeley Joint Graduate Group in Bioengineering, Berkeley, California 94720
- Department of Bioengineering, University of California, Berkeley, California 94720
- Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| |
Collapse
|
34
|
Sola Vigo F, Kedikian G, Heredia L, Heredia F, Añel AD, Rosa AL, Lorenzo A. Amyloid-beta precursor protein mediates neuronal toxicity of amyloid beta through Go protein activation. Neurobiol Aging 2008; 30:1379-92. [PMID: 18187234 DOI: 10.1016/j.neurobiolaging.2007.11.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Revised: 10/25/2007] [Accepted: 11/16/2007] [Indexed: 11/19/2022]
Abstract
Amyloid beta (Abeta) is a metabolic product of amyloid-beta precursor protein (APP). Deposition of Abeta in the brain and neuronal degeneration are characteristic hallmarks of Alzheimer's disease (AD). Abeta induces neuronal degeneration, but the mechanism of neurotoxicity remains elusive. Here we show that overexpression of APP renders hippocampal neurons vulnerable to Abeta toxicity. Deletion of the extracellular Abeta sequence of APP prevents binding of APP to Abeta, and abolishes toxicity. Abeta toxicity is also abrogated by deletion of the cytoplasmic domain of APP, or by deletions comprising the Go protein-binding sequence of APP. Treatment with Pertussis toxin (PTX) abrogates APP-dependent toxicity of Abeta. Overexpression of PTX-insensitive Galpha-o subunit, but not Galpha-i subunit, of G protein restores Abeta toxicity in the presence of PTX, and this requires the integrity of APP-binding site for Go protein. Altogether, these experiments indicate that interaction of APP with toxic Abeta-species promotes toxicity in hippocampal neurons by a mechanism that involves APP-mediated Go protein activation, revealing an Abeta-receptor-like function of APP directly implicated in neuronal degeneration in AD.
Collapse
|
35
|
Gralle M, Ferreira ST. Structure and functions of the human amyloid precursor protein: the whole is more than the sum of its parts. Prog Neurobiol 2007; 82:11-32. [PMID: 17428603 DOI: 10.1016/j.pneurobio.2007.02.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 10/26/2006] [Accepted: 02/01/2007] [Indexed: 12/30/2022]
Abstract
The amyloid precursor protein (APP) is a transmembrane protein that plays major roles in the regulation of several important cellular functions, especially in the nervous system, where it is involved in synaptogenesis and synaptic plasticity. The secreted extracellular domain of APP, sAPPalpha, acts as a growth factor for many types of cells and promotes neuritogenesis in post-mitotic neurons. Alternative proteolytic processing of APP releases potentially neurotoxic species, including the amyloid-beta (Abeta) peptide that is centrally implicated in the pathogenesis of Alzheimer's disease (AD). Reinforcing this biochemical link to neuronal dysfunction and neurodegeneration, APP is also genetically linked to AD. In this review, we discuss the biological functions of APP in the context of tissue morphogenesis and restructuring, where APP appears to play significant roles both as a contact receptor and as a diffusible factor. Structural investigation of APP, which is necessary for a deeper understanding of its roles at a molecular level, has also been advancing rapidly. We summarize recent progress in the determination of the structure of isolated APP fragments and of the conformations of full-length sAPPalpha, in both monomeric and dimeric states. The potential role of APP dimerization for the regulation of its biological functions is also discussed.
Collapse
Affiliation(s)
- Matthias Gralle
- Instituto de Bioquímica Médica, Programa de Bioquímica e Biofísica Celular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.
| | | |
Collapse
|
36
|
Heredia L, Helguera P, de Olmos S, Kedikian G, Solá Vigo F, LaFerla F, Staufenbiel M, de Olmos J, Busciglio J, Cáceres A, Lorenzo A. Phosphorylation of actin-depolymerizing factor/cofilin by LIM-kinase mediates amyloid beta-induced degeneration: a potential mechanism of neuronal dystrophy in Alzheimer's disease. J Neurosci 2006; 26:6533-42. [PMID: 16775141 PMCID: PMC6674046 DOI: 10.1523/jneurosci.5567-05.2006] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Deposition of fibrillar amyloid beta (fAbeta) plays a critical role in Alzheimer's disease (AD). We have shown recently that fAbeta-induced dystrophy requires the activation of focal adhesion proteins and the formation of aberrant focal adhesion structures, suggesting the activation of a mechanism of maladaptative plasticity in AD. Focal adhesions are actin-based structures that provide a structural link between the extracellular matrix and the cytoskeleton. To gain additional insight in the molecular mechanism of neuronal degeneration in AD, here we explored the involvement of LIM kinase 1 (LIMK1), actin-depolymerizing factor (ADF), and cofilin in Abeta-induced dystrophy. ADF/cofilin are actin-binding proteins that play a central role in actin filament dynamics, and LIMK1 is the kinase that phosphorylates and thereby inhibits ADF/cofilin. Our data indicate that treatment of hippocampal neurons with fAbeta increases the level of Ser3-phosphorylated ADF/cofilin and Thr508-phosphorylated LIMK1 (P-LIMK1), accompanied by a dramatic remodeling of actin filaments, neuritic dystrophy, and neuronal cell death. A synthetic peptide, S3 peptide, which acts as a specific competitor for ADF/cofilin phosphorylation by LIMK1, inhibited fAbeta-induced ADF/cofilin phosphorylation, preventing actin filament remodeling and neuronal degeneration, indicating the involvement of LIMK1 in Abeta-induced neuronal degeneration in vitro. Immunofluorescence analysis of AD brain showed a significant increase in the number of P-LIMK1-positive neurons in areas affected with AD pathology. P-LIMK1-positive neurons also showed early signs of AD pathology, such as intracellular Abeta and pretangle phosphorylated tau. Thus, LIMK1 activation may play a key role in AD pathology.
Collapse
|
37
|
Shaked GM, Kummer MP, Lu DC, Galvan V, Bredesen DE, Koo EH. Abeta induces cell death by direct interaction with its cognate extracellular domain on APP (APP 597-624). FASEB J 2006; 20:1254-6. [PMID: 16636103 PMCID: PMC1847355 DOI: 10.1096/fj.05-5032fje] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Amyloid beta-peptide (Abeta) is postulated to play a central role in the pathogenesis of Alzheimer's disease. We recently proposed a pathway of Abeta-induced toxicity that is APP dependent and involves the facilitation of APP complex formation by Abeta. The APP-dependent component requires cleavage of APP at position 664 in the cytoplasmic domain, presumably by caspases or caspase-like proteases, with release of a potentially cytotoxic C31 peptide. In this study we show that Abeta interacted directly and specifically with membrane-bound APP to facilitate APP homo-oligomerization. Using chimeric APP molecules, this interaction was shown to take place between Abeta and its homologous sequence on APP. Consistent with this finding, we demonstrated that Abeta also facilitated the oligomerization of beta-secretase cleaved APP C-terminal fragment (C99). We found that the YENPTY domain in the APP cytoplasmic tail and contained within C31 is critical for this cell death pathway. Deletion or alanine- scanning mutagenesis through this domain significantly attenuated cell death apparently without affecting either APP dimerization or cleavage at position 664. This indicated that sequences within C31 are required after its release from APP. As the YENPTY domain has been shown to interact with a number of cytosolic adaptor molecules, it is possible that the interaction of APP, especially dimeric forms of APP, with these molecules contribute to cell death.
Collapse
Affiliation(s)
- G. M. Shaked
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - M. P. Kummer
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - D. C. Lu
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - V. Galvan
- Buck Institute for Age Research, Novato, California, USA
| | - D. E. Bredesen
- Buck Institute for Age Research, Novato, California, USA
| | - E. H. Koo
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
- Correspondence: Department of Neurosciences, University of California, San Diego, 9500 Gilman Dr., Mail Code 0691, La Jolla, CA 92093-0691, USA. E-mail:
| |
Collapse
|
38
|
Sondag CM, Combs CK. Amyloid precursor protein cross-linking stimulates beta amyloid production and pro-inflammatory cytokine release in monocytic lineage cells. J Neurochem 2006; 97:449-61. [PMID: 16539666 DOI: 10.1111/j.1471-4159.2006.03759.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Beta amyloid peptide-containing neuritic plaques are a defining feature of Alzheimer's disease pathology. Beta amyloid are 38-43 residue peptides derived by proteolytic cleavage of amyloid precursor protein. Although much attention has focused on the proteolytic events leading to beta amyloid generation, the function of amyloid precursor protein remains poorly described. Previously, we reported that amyloid precursor protein functions as a pro-inflammatory receptor on monocytic lineage cells and defined a role for amyloid precursor protein in adhesion by demonstrating that beta(1) integrin-mediated pro-inflammatory activation of monocytes is amyloid precursor protein dependent. We demonstrated that antibody-induced cross-linking of amyloid precursor protein in human THP-1 monocytes and primary mouse microglia stimulates a tyrosine kinase-based pro-inflammatory signaling response leading to acquisition of a reactive phenotype. Here, we have identified pro-inflammatory mediators released upon amyloid precursor protein-dependent activation of monocytes and microglia. We show that amyloid precursor protein cross-linking stimulated tyrosine kinase-dependent increases in pro-inflammatory cytokine release and a tyrosine kinase-independent increase in beta amyloid 1-42 generation. These data provide much needed insight into the function of amyloid precursor protein and provide potential therapeutic targets to limit inflammatory changes associated with the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- C M Sondag
- Department Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, 58202, USA
| | | |
Collapse
|
39
|
Heredia L, Lin R, Vigo FS, Kedikian G, Busciglio J, Lorenzo A. Deposition of amyloid fibrils promotes cell-surface accumulation of amyloid β precursor protein. Neurobiol Dis 2004; 16:617-29. [PMID: 15262274 DOI: 10.1016/j.nbd.2004.04.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2003] [Revised: 04/09/2004] [Accepted: 04/19/2004] [Indexed: 12/21/2022] Open
Abstract
Amyloid beta protein (Abeta) deposition and neuronal degeneration are characteristic pathological features of Alzheimer's disease (AD). In vitro, Abeta fibrils (fAbeta) induce neuronal degeneration reminiscent to AD, but the mechanism of neurotoxicity is unknown. Here we show that amyloid fibrils increase the level of cell-surface full-length amyloid beta precursor protein (h-AbetaPP) and secreted AbetaPP (s-AbetaPP). Pulse-chase analysis indicated that fAbeta selectively inhibited the turnover of cell-surface AbetaPP, without altering its intracellular levels. FAbeta-induced AbetaPP accumulation was not abrogated by cycloheximide, suggesting that increased protein synthesis is not critically required. Abeta fibrils sequester s-AbetaPP from the culture medium and promote its accumulation at the cell surface, indicating that binding of Abeta fibrils mediates AbetaPP accumulation. A time course analysis of Abeta treatment showed that AbetaPP level is elevated before significant cell death can be detected, while other toxic insults do not augment AbetaPP level, suggesting that AbetaPP may be specifically involved in early stages of Abeta-induced neurodegeneration. Finally, Abeta fibrils promote clustering of h-AbetaPP in abnormal focal adhesion-like (FA-like) structures that mediate neuronal dystrophy, increasing its association with the cytoskeleton. These results indicate that the interaction of Abeta fibrils with AbetaPP is an early event in the mechanism of Abeta-induced neurodegeneration that may play a significant role in AD pathogenesis.
Collapse
Affiliation(s)
- Lorena Heredia
- Laboratory of Experimental Neuropathology, Instituto de Investigación Médica Mercedes y Martín Ferreyra-INIMEC/CONICET, Córdoba, 5000 Argentina
| | | | | | | | | | | |
Collapse
|
40
|
McCarron MO, Nicoll JAR. Cerebral amyloid angiopathy and thrombolysis-related intracerebral haemorrhage. Lancet Neurol 2004; 3:484-92. [PMID: 15261609 DOI: 10.1016/s1474-4422(04)00825-7] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Intracerebral haemorrhage is a complication of thrombolytic therapy for acute myocardial infarction, pulmonary embolism, and ischaemic stroke. There is increasing evidence that cerebral amyloid angiopathy (CAA), which itself can cause haemorrhage (CAAH), may be a risk factor for thrombolysis-related intracerebral haemorrhage. CAAH and thrombolysis-related intracerebral haemorrhage share some clinical features, such as predisposition to lobar or superficial regions of the brain, multiple haemorrhages, increasing frequency with age, and an association with dementia. In vitro work showed that accumulation of amyloid-beta peptide causes degeneration of cells in the walls of blood vessels, affects vasoactivity, and improves proteolytic mechanisms, such as fibrinolysis, anticoagulation, and degradation of the extracellular matrix. In a mouse model of CAA there is a low haemorrhagic threshold after thrombolytic therapy compared with that in wild-type mice. To date only a small number of anecdotal clinicopathological relations have been reported; neuroimaging advances and further study of the frequency and role of CAA in patients with thrombolysis-related intracerebral haemorrhage are required.
Collapse
Affiliation(s)
- Mark O McCarron
- Department of Neurology, Altnagelvin Hospital, Londonderry, BT47 6SB, UK.
| | | |
Collapse
|
41
|
Schmechel A, Zentgraf H, Scheuermann S, Fritz G, Pipkorn R, Reed J, Beyreuther K, Bayer TA, Multhaup G. Alzheimer beta-amyloid homodimers facilitate A beta fibrillization and the generation of conformational antibodies. J Biol Chem 2003; 278:35317-24. [PMID: 12840025 DOI: 10.1074/jbc.m303547200] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We reported previously that stabilized beta-amyloid peptide dimers were derived from mutant amyloid precursor protein with a single cysteine in the ectodomain juxtamembrane position. In vivo studies revealed that two forms of SDS-stable A beta homodimers exist, species ending at A beta 40 and A beta 42. The phenomenon of the transformation of the initially deposited 42-residue beta-amyloid peptide into the amyloid fibrils of Alzheimer's disease plaques remains to be explained in physical terms, i.e. energetically and structurally. We therefore performed spectroscopic analyses revealing that engineered dimeric peptides ending at residue 42 displayed a much more pronounced beta-structural transition than corresponding monomers. Specifically, the single chemically induced dimerization of A beta peptides significantly increased the beta-sheet content by a factor of 2. The C-terminal residues Ile-41 and Ala-42 of dimeric forms further increased the beta-sheet content by roughly one-third. In contrast to A beta 42, the beta-sheet content of the alpha- and gamma-secretase-generated p3 fragments did not necessarily correlate with the tendency to form fibrils, although p3/17-42 had a pronounced thread forming character with fibril lengths of up to 2.5 microM. Electron microscopic images show that forms of p3/17-42 generated smaller granular particles than forms ending at residue 40. We discuss these findings in terms of A beta 1-42 dimers representing paranuclei, which self-aggregate into ribbon-like ordered fibrils by elongation. Based on A beta 42 dimer-specific titers of a polyclonal antiserum we propose that the A beta homodimer represents a nidus for plaque formation and a well defined novel therapeutic target.
Collapse
Affiliation(s)
- Ariane Schmechel
- Freie Universitaet Berlin, Institut fuer Chemie/Biochemie, Thielallee 63, Berlin D-14195, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
White AR, Maher F, Brazier MW, Jobling MF, Thyer J, Stewart LR, Thompson A, Gibson R, Masters CL, Multhaup G, Beyreuther K, Barrow CJ, Collins SJ, Cappai R. Diverse fibrillar peptides directly bind the Alzheimer's amyloid precursor protein and amyloid precursor-like protein 2 resulting in cellular accumulation. Brain Res 2003; 966:231-44. [PMID: 12618346 DOI: 10.1016/s0006-8993(02)04173-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The Alzheimer's disease Abeta peptide can increase the levels of cell-associated amyloid precursor protein (APP) in vitro. To determine the specificity of this response for Abeta and whether it is related to cytotoxicity, we tested a diverse range of fibrillar peptides including amyloid-beta (Abeta), the fibrillar prion peptides PrP106-126 and PrP178-193 and human islet-cell amylin. All these peptides increased the levels of APP and amyloid precursor-like protein 2 (APLP2) in primary cultures of astrocytes and neurons. Specificity was shown by a lack of change to amyloid precursor-like protein 1, tau-1 and cellular prion protein (PrP(c)) levels. APP and APLP2 levels were elevated only in cultures exposed to fibrillar peptides as assessed by electron microscopy and not in cultures treated with non-fibrillogenic peptide variants or aggregated lipoprotein. We found that PrP106-126 and the non-toxic but fibril-forming PrP178-193 increased APP levels in cultures derived from both wild-type and PrP(c)-deficient mice indicating that fibrillar peptides up-regulate APP through a non-cytotoxic mechanism and irrespective of parental protein expression. Fibrillar PrP106-126 and Abeta peptides bound recombinant APP and APLP2 suggesting the accumulation of these proteins was mediated by direct binding to the fibrillated peptide. This was supported by decreased APP accumulation following extensive washing of the cultures to remove fibrillar aggregates. Pre-incubation of fibrillar peptide with recombinant APP18-146, the putative fibril binding site, also abrogated the accumulation of APP. These findings show that diverse fibrillogenic peptides can induce accumulation of APP and APLP2 and this mechanism could contribute to pathogenesis in neurodegenerative disorders.
Collapse
Affiliation(s)
- Anthony R White
- Department of Pathology, The University of Melbourne, 3010, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|