1
|
König S, Schmidt N, Bechberger K, Morris S, Priego M, Zaky H, Song Y, Pielage J, Brunholz S, Brady ST, Kins S, Morfini G. Axon-Autonomous Effects of the Amyloid Precursor Protein Intracellular Domain (AICD) on Kinase Signaling and Fast Axonal Transport. Cells 2023; 12:2403. [PMID: 37830617 PMCID: PMC10572015 DOI: 10.3390/cells12192403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
The amyloid precursor protein (APP) is a key molecular component of Alzheimer's disease (AD) pathogenesis. Proteolytic APP processing generates various cleavage products, including extracellular amyloid beta (Aβ) and the cytoplasmic APP intracellular domain (AICD). Although the role of AICD in the activation of kinase signaling pathways is well established in the context of full-length APP, little is known about intracellular effects of the AICD fragment, particularly within discrete neuronal compartments. Deficits in fast axonal transport (FAT) and axonopathy documented in AD-affected neurons prompted us to evaluate potential axon-autonomous effects of the AICD fragment for the first time. Vesicle motility assays using the isolated squid axoplasm preparation revealed inhibition of FAT by AICD. Biochemical experiments linked this effect to aberrant activation of selected axonal kinases and heightened phosphorylation of the anterograde motor protein conventional kinesin, consistent with precedents showing phosphorylation-dependent regulation of motors proteins powering FAT. Pharmacological inhibitors of these kinases alleviated the AICD inhibitory effect on FAT. Deletion experiments indicated this effect requires a sequence encompassing the NPTY motif in AICD and interacting axonal proteins containing a phosphotyrosine-binding domain. Collectively, these results provide a proof of principle for axon-specific effects of AICD, further suggesting a potential mechanistic framework linking alterations in APP processing, FAT deficits, and axonal pathology in AD.
Collapse
Affiliation(s)
- Svenja König
- Department for Human Biology and Human Genetics, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany (K.B.); (S.K.)
| | - Nadine Schmidt
- Department for Human Biology and Human Genetics, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany (K.B.); (S.K.)
| | - Karin Bechberger
- Department for Human Biology and Human Genetics, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany (K.B.); (S.K.)
| | - Sarah Morris
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA (S.T.B.)
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Mercedes Priego
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA (S.T.B.)
| | - Hannah Zaky
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA (S.T.B.)
| | - Yuyu Song
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
- Department of Neurology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02129, USA
| | - Jan Pielage
- Department of Zoology, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany;
| | - Silke Brunholz
- Department for Human Biology and Human Genetics, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany (K.B.); (S.K.)
| | - Scott T. Brady
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA (S.T.B.)
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Stefan Kins
- Department for Human Biology and Human Genetics, University of Kaiserslautern-Landau, 67663 Kaiserslautern, Germany (K.B.); (S.K.)
| | - Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA (S.T.B.)
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| |
Collapse
|
2
|
Banerjee S, Manisha C, Bharathi J J, Kumar AP, Justin A, Ramanathan M. Structural dynamics and catalytic modulations of Aβ regulating enzymes as future outlook for Alzheimer's. Biochem Biophys Res Commun 2022; 631:1-8. [PMID: 36162324 DOI: 10.1016/j.bbrc.2022.09.068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/15/2022] [Indexed: 11/28/2022]
Abstract
Aβ cascade hypothesis being considered most evident event in AD pathology and even today it holds good. Dysregulation of catalytic events of Aβ regulating enzymes can possibly cause faulty Aβ trafficking; inequity of Aβ formation and clearance resulting in misfolded protein accumulation, neurodegeneration and cognitive impairment. Many novel approaches have been made on this pathway to discover new molecules, unfortunately couldn't reach the terminal phases of clinical trials. Over decades, studies have been more focused on enzyme chemistry and explored the relationship between structural features and catalytic function of Aβ regulating enzymes. However, the modulations of catalytic mechanisms of those enzymes have not been imposed so far to reduce the Aβ load. Hence, in this review, we have critically detailed the knowledge of basic structural dynamics and possible catalytic modulations of enzymes responsible for Aβ formation and clearance that will impart new perspectives in drug discovery process.
Collapse
Affiliation(s)
- Sayani Banerjee
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, 643 001, India
| | - Chennu Manisha
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, 643 001, India
| | - Jeyaram Bharathi J
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, 643 001, India
| | - Ashwini Prem Kumar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, 643 001, India
| | - Antony Justin
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, 643 001, India.
| | - Muthiah Ramanathan
- Department of Pharmacology, PSG College of Pharmacy, Coimbatore, Tamil Nadu, 641 004, India
| |
Collapse
|
3
|
Xu H, Van Remmen H. The SarcoEndoplasmic Reticulum Calcium ATPase (SERCA) pump: a potential target for intervention in aging and skeletal muscle pathologies. Skelet Muscle 2021; 11:25. [PMID: 34772465 PMCID: PMC8588740 DOI: 10.1186/s13395-021-00280-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/26/2021] [Indexed: 01/13/2023] Open
Abstract
As a key regulator of cellular calcium homeostasis, the Sarcoendoplasmic Reticulum Calcium ATPase (SERCA) pump acts to transport calcium ions from the cytosol back to the sarcoplasmic reticulum (SR) following muscle contraction. SERCA function is closely associated with muscle health and function, and SERCA activity is susceptible to muscle pathogenesis. For example, it has been well reported that pathological conditions associated with aging, neurodegeneration, and muscular dystrophy (MD) significantly depress SERCA function with the potential to impair intracellular calcium homeostasis and further contribute to muscle atrophy and weakness. As a result, targeting SERCA activity has attracted attention as a therapeutical method for the treatment of muscle pathologies. The interventions include activation of SERCA activity and genetic overexpression of SERCA. This review will focus on SERCA function and regulation mechanisms and describe how those mechanisms are affected under muscle pathological conditions including elevated oxidative stress induced by aging, muscle disease, or neuromuscular disorders. We also discuss the current progress and therapeutic approaches to targeting SERCA in vivo.
Collapse
Affiliation(s)
- Hongyang Xu
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Holly Van Remmen
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA. .,Oklahoma City VA Medical Center, Oklahoma City, OK, USA. .,Department of Physiology, OUHSC, Oklahoma City, OK, USA.
| |
Collapse
|
4
|
Reveglia P, Nasso R, Angiolillo A, Lecce L, Paolillo C, De Tullio S, Gelzo M, Di Costanzo A, Matrone C, Corso G. Tandem Mass Spectrometry as Strategy for the Selective Identification and Quantification of the Amyloid Precursor Protein Tyr682 Residue Phosphorylation Status in Human Blood Mononuclear Cells. Biomolecules 2021; 11:biom11091297. [PMID: 34572510 PMCID: PMC8471626 DOI: 10.3390/biom11091297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 11/28/2022] Open
Abstract
Background: Alzheimer’s disease (AD) is a devastating neurodegenerative disease without guidelines for early diagnosis or personalized treatment. Previous studies have highlighted a crucial role of increasing phosphorylation levels of the amyloid precursor protein (APP) Tyr682 residue in predicting neuronal deficits in AD patients. However, the lack of a method for the identification and quantification of Tyr682 phosphorylation levels prevents its potential clinical applications. Methods: Here we report a method to identify and quantify APP Tyr682 phosphorylation levels in blood mononuclear cells of AD patients by tandem mass spectrometry (tMS). Results: This method showed excellent sensitivity with detection and quantification limits set respectively at 0.035 and 0.082 ng injected for the phosphorylated peptide and at 0.02 and 0.215 ng injected for the non-phosphorylated peptide. The average levels of both peptides were quantified in transfected HELA cells (2.48 and 3.53 ng/μg of protein, respectively). Preliminary data on 3 AD patients showed quantifiable levels of phosphorylated peptide (0.10–0.15 ng/μg of protein) and below the LOQ level of non-phosphorylated peptide (0.13 ng/μg of protein). Conclusion: This method could allow the identification of patients with increased APP Tyr682 phosphorylation and allow early characterization of molecular changes prior to the appearance of clinical signs.
Collapse
Affiliation(s)
- Pierluigi Reveglia
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (P.R.); (L.L.); (C.P.); (S.D.T.)
| | - Rosarita Nasso
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (R.N.); (C.M.)
| | - Antonella Angiolillo
- Centre for Research and Training in Medicine for Aging, Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, 86100 Campobasso, Italy; (A.A.); (A.D.C.)
| | - Lucia Lecce
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (P.R.); (L.L.); (C.P.); (S.D.T.)
| | - Carmela Paolillo
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (P.R.); (L.L.); (C.P.); (S.D.T.)
| | - Samantha De Tullio
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (P.R.); (L.L.); (C.P.); (S.D.T.)
| | - Monica Gelzo
- CEINGE-Biotecnologie Avanzate, scarl, 80145 Naples, Italy;
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples Federico II, 80131 Naples, Italy
| | - Alfonso Di Costanzo
- Centre for Research and Training in Medicine for Aging, Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, 86100 Campobasso, Italy; (A.A.); (A.D.C.)
| | - Carmela Matrone
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (R.N.); (C.M.)
| | - Gaetano Corso
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (P.R.); (L.L.); (C.P.); (S.D.T.)
- Correspondence:
| |
Collapse
|
5
|
Iannuzzi F, Sirabella R, Canu N, Maier TJ, Annunziato L, Matrone C. Fyn Tyrosine Kinase Elicits Amyloid Precursor Protein Tyr682 Phosphorylation in Neurons from Alzheimer's Disease Patients. Cells 2020; 9:E1807. [PMID: 32751526 PMCID: PMC7463977 DOI: 10.3390/cells9081807] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disorder with a few early detection strategies. We previously proposed the amyloid precursor protein (APP) tyrosine 682 (Tyr682) residue as a valuable target for the development of new innovative pharmacologic or diagnostic interventions in AD. Indeed, when APP is phosphorylated at Tyr682, it is forced into acidic neuronal compartments where it is processed to generate neurotoxic amyloid β peptides. Of interest, Fyn tyrosine kinase (TK) interaction with APP Tyr682 residue increases in AD neurons. Here we proved that when Fyn TK was overexpressed it elicited APP Tyr682 phosphorylation in neurons from healthy donors and promoted the amyloidogenic APP processing with Aβ peptides accumulation and neuronal death. Phosphorylation of APP at Tyr (pAPP-Tyr) increased in neurons of AD patients and AD neurons that exhibited high pAPP-Tyr also had higher Fyn TK activity. Fyn TK inhibition abolished the pAPP-Tyr and reduced Aβ42 secretion in AD neurons. In addition, the multidomain adaptor protein Fe65 controlled the Fyn-mediated pAPP-Tyr, warranting the possibility of targeting the Fe65-APP-Fyn pathway to develop innovative strategies in AD. Altogether, these results strongly emphasize the relevance of focusing on pAPP Tyr682 either for diagnostic purposes, as an early biomarker of the disease, or for pharmacological targeting, using Fyn TKI.
Collapse
Affiliation(s)
- Filomena Iannuzzi
- Department of Biomedicine, Aarhus University, Aarhus C, 8000 Aarhus, Denmark;
| | - Rossana Sirabella
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
| | - Nadia Canu
- Department of System Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
- Institute of Biochemistry and Cell Biology, CNR, 00015 Monterotondo, Rome, Italy
| | - Thorsten J. Maier
- Paul-Ehrlich-Institut, (Federal Institute for Vaccines and Biomedicines), 63225 Langen, Germany;
| | - Lucio Annunziato
- SDN Research Institute Diagnostics and Nuclear (IRCCS SDN), Gianturco, 80131 Naples, Italy
| | - Carmela Matrone
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
| |
Collapse
|
6
|
Rahate K, Bhatt LK, Prabhavalkar KS. SERCA stimulation: A potential approach in therapeutics. Chem Biol Drug Des 2019; 95:5-15. [DOI: 10.1111/cbdd.13620] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/19/2019] [Accepted: 08/26/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Kiran Rahate
- Department of Pharmacology SVKM’s Dr. Bhanuben Nanavati College of Pharmacy Mumbai India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology SVKM’s Dr. Bhanuben Nanavati College of Pharmacy Mumbai India
| | - Kedar S. Prabhavalkar
- Department of Pharmacology SVKM’s Dr. Bhanuben Nanavati College of Pharmacy Mumbai India
| |
Collapse
|
7
|
The role of PTB domain containing adaptor proteins on PICALM-mediated APP endocytosis and localization. Biochem J 2019; 476:2093-2109. [DOI: 10.1042/bcj20180840] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 07/01/2019] [Accepted: 07/12/2019] [Indexed: 02/06/2023]
Abstract
AbstractOne hallmark of Alzheimer's disease (AD) is the presence of amyloid plaques, which mainly consist of the amyloid precursor protein (APP) cleavage product amyloid β (Aβ). For cleavage to occur, the APP must be endocytosed from the cell surface. The phosphatidylinositol binding clathrin assembly protein (PICALM) is involved in clathrin-mediated endocytosis and polymorphisms in and near the gene locus were identified as genetic risk factors for AD. PICALM overexpression enhances APP internalization and Aβ production. Furthermore, PICALM shuttles into the nucleus, but its function within the nucleus is still unknown. Using co-immunoprecipitation, we demonstrated an interaction between PICALM and APP, which is abrogated by mutation of the APP NPXY-motif. Since the NPXY-motif is an internalization signal that binds to phosphotryrosine-binding domain-containing adaptor proteins (PTB-APs), we hypothesized that PTB-APs can modulate the APP-PICALM interaction. We found that interaction between PICALM and the PTB-APs (Numb, JIP1b and GULP1) enhances the APP-PICALM interaction. Fluorescence activated cell sorting analysis and internalization assays revealed differentially altered APP cell surface levels and endocytosis rates that depended upon the presence of PICALM and co-expression of distinct PTB-APs. Additionally, we were able to show an impact of PICALM nuclear shuttling upon co-expression of PTB-APs and PICALM, with the magnitude of the effect depending on which PTB-AP was co-expressed. Taken together, our results indicate a modulating effect of PTB-APs on PICALM-mediated APP endocytosis and localization.
Collapse
|
8
|
Matrone C, Iannuzzi F, Annunziato L. The Y 682ENPTY 687 motif of APP: Progress and insights toward a targeted therapy for Alzheimer's disease patients. Ageing Res Rev 2019; 52:120-128. [PMID: 31039414 DOI: 10.1016/j.arr.2019.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/04/2019] [Accepted: 04/10/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder for which no curative treatments, disease modifying strategies or effective symptomatic therapies exist. Current pharmacologic treatments for AD can only decelerate the progression of the disease for a short time, often at the cost of severe side effects. Therefore, there is an urgent need for biomarkers able to diagnose AD at its earliest stages, to conclusively track disease progression, and to accelerate the clinical development of innovative therapies. Scientific research and economic efforts for the development of pharmacotherapies have recently homed in on the hypothesis that neurotoxic β-amyloid (Aβ) peptides in their oligomeric or fibrillary forms are primarily responsible for the cognitive impairment and neuronal death seen in AD. As such, modern pharmacologic approaches are largely based on reducing production by inhibiting β and γ secretase cleavage of the amyloid precursor protein (APP) or on dissolving existing cerebral Aβ plaques or to favor Aβ clearance from the brain. The following short review aims to persuade the reader of the idea that APP plays a much larger role in AD pathogenesis. APP plays a greater role in AD pathogenesis than its role as the precursor for Aβ peptides: both the abnormal cleavage of APP leading to Aβ peptide accumulation and the disruption of APP physiological functions contribute to AD pathogenesis. We summarize our recent results on the role played by the C-terminal APP motif -the Y682ENPTY68 motif- in APP function and dysfunction, and we provide insights into targeting the Tyr682 residue of APP as putative novel strategy in AD.
Collapse
|
9
|
Furusawa K, Takasugi T, Chiu YW, Hori Y, Tomita T, Fukuda M, Hisanaga SI. CD2-associated protein (CD2AP) overexpression accelerates amyloid precursor protein (APP) transfer from early endosomes to the lysosomal degradation pathway. J Biol Chem 2019; 294:10886-10899. [PMID: 31138646 DOI: 10.1074/jbc.ra118.005385] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 05/16/2019] [Indexed: 12/23/2022] Open
Abstract
A hallmark of Alzheimer's disease (AD) pathology is the appearance of senile plaques, which are composed of β-amyloid (Aβ) peptides. Aβ is produced by sequential cleavages of amyloid precursor protein (APP) by β- and γ-secretases. These cleavages take place in endosomes during intracellular trafficking of APP through the endocytic and recycling pathways. Genome-wide association studies have identified several risk factors for late-onset AD, one of which is CD2-associated protein (CD2AP), an adaptor molecule that regulates membrane trafficking. Although CD2AP's involvement in APP trafficking has recently been reported, how APP trafficking is regulated remains unclear. We sought to address this question by investigating the effect of CD2AP overexpression or knockdown on the intracellular APP distribution and degradation of APP in cultured COS-7 and HEK293 cells. We found that overexpression of CD2AP increases the localization of APP to Rab7-positive late endosomes, and decreases its localization to Rab5-positive early endosomes. CD2AP overexpression accelerated the onset of APP degradation without affecting its degradation rate. Furthermore, nutrient starvation increased the localization of APP to Rab7-positive late endosomes, and CD2AP overexpression stimulated starvation-induced lysosomal APP degradation. Moreover, the effect of CD2AP on the degradation of APP was confirmed by CD2AP overexpression and knockdown in primary cortical neurons from mice. We conclude that CD2AP accelerates the transfer of APP from early to late endosomes. This transfer in localization stimulates APP degradation by reducing the amount of time before degradation initiation. Taken together, these results may explain why impaired CD2AP function is a risk factor for AD.
Collapse
Affiliation(s)
- Kotaro Furusawa
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Minami-Osawa, Hachioji, Tokyo 192-0397
| | - Toshiyuki Takasugi
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Minami-Osawa, Hachioji, Tokyo 192-0397
| | - Yung-Wen Chiu
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033 and
| | - Yukiko Hori
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033 and
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033 and
| | - Mitsunori Fukuda
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Shin-Ichi Hisanaga
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Minami-Osawa, Hachioji, Tokyo 192-0397,.
| |
Collapse
|
10
|
Profiling of Alzheimer’s disease related genes in mild to moderate vitamin D hypovitaminosis. J Nutr Biochem 2019; 67:123-137. [DOI: 10.1016/j.jnutbio.2019.01.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/13/2018] [Accepted: 01/29/2019] [Indexed: 02/01/2023]
|
11
|
Wang N, Cai XY, Gao W, Shen Y. Numb-p72, but not Numb-p65, contributes to the trafficking of group I metabotropic glutamate receptors. Neuroreport 2018; 29:902-906. [PMID: 29762249 DOI: 10.1097/wnr.0000000000001051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The protein Numb localizes to clathrin-coated vesicles and participates in the trafficking of transmembrane receptors. We previous reported that Numb promotes the presence of metabotropic glutamate receptor 1 (mGlu1) on neuronal membrane, and Numb deficiency impairs synaptic expression of mGlu1. However, the actions of different Numb isoforms on mGlu1 trafficking are unknown. Here, we found that Numb-p72 and Numb-p65 are distinctly expressed in HEK293T cells. Interestingly, Numb-p72, but not Numb-p65, binds to mGlu1α and promotes the membrane expression of mGlu1α by antagonizing its internalization. We hypothesize that the incomplete structure of Numb-p65 does not act in the same way as Numb-p72 on mGlu1 trafficking.
Collapse
Affiliation(s)
- Na Wang
- School of Medicine, Zhejiang University City College
| | - Xin-Yu Cai
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Wei Gao
- School of Medicine, Zhejiang University City College
| | - Ying Shen
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
12
|
Trafficking in Alzheimer's Disease: Modulation of APP Transport and Processing by the Transmembrane Proteins LRP1, SorLA, SorCS1c, Sortilin, and Calsyntenin. Mol Neurobiol 2017; 55:5809-5829. [PMID: 29079999 DOI: 10.1007/s12035-017-0806-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/17/2017] [Indexed: 12/11/2022]
Abstract
The amyloid precursor protein (APP), one key player in Alzheimer's disease (AD), is extensively processed by different proteases. This leads to the generation of diverging fragments including the amyloid β (Aβ) peptide, which accumulates in brains of AD patients. Subcellular trafficking of APP is an important aspect for its proteolytic conversion, since the various secretases which cleave APP are located in different cellular compartments. As a consequence, altered subcellular targeting of APP is thought to directly affect the degree to which Aβ is generated. The mechanisms underlying intracellular APP transport are critical to understand AD pathogenesis and can serve as a target for future pharmacological interventions. In the recent years, a number of APP interacting proteins were identified which are implicated in sorting of APP, thereby influencing APP processing at different angles of the secretory or endocytic pathway. This review provides an update on the proteolytic processing of APP and the interplay of the transmembrane proteins low-density lipoprotein receptor-related protein 1, sortilin-receptor with A-type repeats, SorCS1c, sortilin, and calsyntenin. We discuss the specific interactions with APP, the capacity to modulate the intracellular itinerary and the proteolytic conversion of APP, a possible involvement in the clearance of Aβ, and the implications of these transmembrane proteins in AD and other neurodegenerative diseases.
Collapse
|
13
|
Small things matter: Implications of APP intracellular domain AICD nuclear signaling in the progression and pathogenesis of Alzheimer’s disease. Prog Neurobiol 2017; 156:189-213. [DOI: 10.1016/j.pneurobio.2017.05.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/25/2017] [Accepted: 05/30/2017] [Indexed: 01/08/2023]
|
14
|
Poulsen ET, Iannuzzi F, Rasmussen HF, Maier TJ, Enghild JJ, Jørgensen AL, Matrone C. An Aberrant Phosphorylation of Amyloid Precursor Protein Tyrosine Regulates Its Trafficking and the Binding to the Clathrin Endocytic Complex in Neural Stem Cells of Alzheimer's Disease Patients. Front Mol Neurosci 2017; 10:59. [PMID: 28360834 PMCID: PMC5350151 DOI: 10.3389/fnmol.2017.00059] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 02/21/2017] [Indexed: 11/15/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and is likely caused by defective amyloid precursor protein (APP) trafficking and processing in neurons leading to amyloid plaques containing the amyloid-β (Aβ) APP peptide byproducts. Understanding how APP is targeted to selected destinations inside neurons and identifying the mechanisms responsible for the generation of Aβ are thus the keys for the advancement of new therapies. We previously developed a mouse model with a mutation at tyrosine (Tyr) 682 in the C-terminus of APP. This residue is needed for APP to bind to the coating protein Clathrin and to the Clathrin adaptor protein AP2 as well as for the correct APP trafficking and sorting in neurons. By extending these findings to humans, we found that APP binding to Clathrin is decreased in neural stem cells from AD sufferers. Increased APP Tyr phosphorylation alters APP trafficking in AD neurons and it is associated to Fyn Tyr kinase activation. We show that compounds affecting Tyr kinase activity and counteracting defects in AD neurons can control APP location and compartmentalization. APP Tyr phosphorylation is thus a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Ebbe T. Poulsen
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhus, Denmark
| | | | | | - Thorsten J. Maier
- Institute of Biomedicine, Aarhus UniversityAarhus, Denmark
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Goethe UniversityFrankfurt, Germany
| | - Jan J. Enghild
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhus, Denmark
| | | | | |
Collapse
|
15
|
Talenti A, Bertolini F, Pagnacco G, Pilla F, Ajmone-Marsan P, Rothschild MF, Crepaldi P. The Valdostana goat: a genome-wide investigation of the distinctiveness of its selective sweep regions. Mamm Genome 2017; 28:114-128. [PMID: 28255622 DOI: 10.1007/s00335-017-9678-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 01/26/2017] [Indexed: 01/10/2023]
Abstract
The Valdostana goat is an alpine breed, raised only in the northern Italian region of the Aosta Valley. This breed's main purpose is to produce milk and meat, but is peculiar for its involvement in the "Batailles de Chèvres," a recent tradition of non-cruel fight tournaments. At both the genetic and genomic levels, only a very limited number of studies have been performed with this breed and there are no studies about the genomic signatures left by selection. In this work, 24 unrelated Valdostana animals were screened for runs of homozygosity to identify highly homozygous regions. Then, six different approaches (ROH comparison, Fst single SNPs and windows based, Bayesian, Rsb, and XP-EHH) were applied comparing the Valdostana dataset with 14 other Italian goat breeds to confirm regions that were different among the comparisons. A total of three regions of selection that were also unique among the Valdostana were identified and located on chromosomes 1, 7, and 12 and contained 144 genes. Enrichment analyses detected genes such as cytokines and lymphocyte/leukocyte proliferation genes involved in the regulation of the immune system. A genetic link between an aggressive challenge, cytokines, and immunity has been hypothesized in many studies both in humans and in other species. Possible hypotheses associated with the signals of selection detected could be therefore related to immune-related factors as well as with the peculiar battle competition, or other breed-specific traits, and provided insights for further investigation of these unique regions, for the understanding and safeguard of the Valdostana breed.
Collapse
Affiliation(s)
- Andrea Talenti
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Milan, Italy
| | | | - Giulio Pagnacco
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Milan, Italy
| | - Fabio Pilla
- Dipartimento Agricoltura, Ambiente e Alimenti, Università degli Studi del Molise, via Francesco De Sanctis s.n.c., 86100, Campobasso, Italy
| | - Paolo Ajmone-Marsan
- Istituto di Zootecnica, Università Cattolica del Sacro Cuore, via Emilia Parmense, 84, 29122, Piacenza, Italy
| | - Max F Rothschild
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | - Paola Crepaldi
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
16
|
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
|
17
|
Alzheimer disease: modeling an Aβ-centered biological network. Mol Psychiatry 2016; 21:861-71. [PMID: 27021818 DOI: 10.1038/mp.2016.38] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/16/2016] [Accepted: 02/18/2016] [Indexed: 01/15/2023]
Abstract
In genetically complex diseases, the search for missing heritability is focusing on rare variants with large effect. Thanks to next generation sequencing technologies, genome-wide characterization of these variants is now feasible in every individual. However, a lesson from current studies is that collapsing rare variants at the gene level is often insufficient to obtain a statistically significant signal in case-control studies, and that network-based analyses are an attractive complement to classical approaches. In Alzheimer disease (AD), according to the prevalent amyloid cascade hypothesis, the pathology is driven by the amyloid beta (Aβ) peptide. In past years, based on experimental studies, several hundreds of proteins have been shown to interfere with Aβ production, clearance, aggregation or toxicity. Thanks to a manual curation of the literature, we identified 335 genes/proteins involved in this biological network and classified them according to their cellular function. The complete list of genes, or its subcomponents, will be of interest in ongoing AD genetic studies.
Collapse
|
18
|
Dunning CJR, Black HL, Andrews KL, Davenport EC, Conboy M, Chawla S, Dowle AA, Ashford D, Thomas JR, Evans GJO. Multisite tyrosine phosphorylation of the N-terminus of Mint1/X11α by Src kinase regulates the trafficking of amyloid precursor protein. J Neurochem 2016; 137:518-27. [PMID: 26865271 PMCID: PMC4982022 DOI: 10.1111/jnc.13571] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 01/21/2016] [Accepted: 02/03/2016] [Indexed: 12/18/2022]
Abstract
Mint/X11 is one of the four neuronal trafficking adaptors that interact with amyloid precursor protein (APP) and are linked with its cleavage to generate β‐amyloid peptide, a key player in the pathology of Alzheimer's disease. How APP switches between adaptors at different stages of the secretory pathway is poorly understood. Here, we show that tyrosine phosphorylation of Mint1 regulates the destination of APP. A canonical SH2‐binding motif (202YEEI) was identified in the N‐terminus of Mint1 that is phosphorylated on tyrosine by C‐Src and recruits the active kinase for sequential phosphorylation of further tyrosines (Y191 and Y187). A single Y202F mutation in the Mint1 N‐terminus inhibits C‐Src binding and tyrosine phosphorylation. Previous studies observed that co‐expression of wild‐type Mint1 and APP causes accumulation of APP in the trans‐Golgi. Unphosphorylatable Mint1 (Y202F) or pharmacological inhibition of Src reduced the accumulation of APP in the trans‐Golgi of heterologous cells. A similar result was observed in cultured rat hippocampal neurons where Mint1(Y202F) permitted the trafficking of APP to more distal neurites than the wild‐type protein. These data underline the importance of the tyrosine phosphorylation of Mint1 as a critical switch for determining the destination of APP.
The regulation of amyloid precursor protein (APP) trafficking is poorly understood. We have discovered that the APP adapter, Mint1, is phosphorylated by C‐Src kinase. Mint1 causes APP accumulation in the trans‐Golgi network, whereas inhibition of Src or mutation of Mint1‐Y202 permits APP recycling. The phosphorylation status of Mint1 could impact on the pathological trafficking of APP in Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Adam A Dowle
- Department of Biology, University of York, York, UK
| | | | | | | |
Collapse
|
19
|
Moodley S, Hui Bai X, Kapus A, Yang B, Liu M. XB130/Tks5 scaffold protein interaction regulates Src-mediated cell proliferation and survival. Mol Biol Cell 2015; 26:4492-502. [PMID: 26446840 PMCID: PMC4666142 DOI: 10.1091/mbc.e15-07-0483] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/29/2015] [Indexed: 12/20/2022] Open
Abstract
XB130 and Tks5 interact endogenously and form a complex with Src tyrosine kinase. Tks5, like XB130, plays a role in cell proliferation and cell survival, and the interaction between XB130 and Tks5 is critical for regulation of Src-mediated cell proliferation and survival. The scaffold protein XB130 regulates cell growth, survival, and migration. Yeast two-hybrid screening suggests that XB130 interacts with another scaffold protein, Tks5. We hypothesized that XB130 and Tks5 form a macromolecular complex to mediate signal transduction cascades for the regulation of cell growth and survival. Coimmunoprecipitation demonstrated that XB130 and Tks5 interact endogenously and form a complex with Src tyrosine kinase. Structure–function studies showed that the fifth SH3 domain of Tks5 binds to the N-terminus of XB130, which contains polyproline-rich motifs. Cell growth and survival studies revealed that down-regulation of XB130 and/or Tks5 reduced cell proliferation, resulting in cell cycle inhibition at the G1 phase and increased caspase 3 activity and apoptosis. Moreover, cell proliferation and survival were increased by overexpression of XB130 or Tks5 but decreased when XB130/Tks5 binding was disrupted by overexpression of XB130 N-terminal deleted mutant and/or Tks5 fifth SH3 domain W1108A mutant. Furthermore, down-regulation of XB130 and/or Tks5 inhibited serum- and growth factor–induced Src activation and downstream phosphorylation of PI3K and Akt. Our results suggest that Tks5, similar to XB130, plays a role in cell proliferation and cell survival and that the interaction between XB130 and Tks5 appears to be critical for regulation of Src-mediated cellular homeostasis.
Collapse
Affiliation(s)
- Serisha Moodley
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Xiao Hui Bai
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Andras Kapus
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada Department of Surgery, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Burton Yang
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mingyao Liu
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada Department of Surgery, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
20
|
Lee HJ, Yoon JH, Ahn JS, Jo EH, Kim MY, Lee YC, Kim JW, Ann EJ, Park HS. Fe65 negatively regulates Jagged1 signaling by decreasing Jagged1 protein stability through the E3 ligase Neuralized-like 1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2918-28. [PMID: 26276215 DOI: 10.1016/j.bbamcr.2015.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 08/06/2015] [Accepted: 08/09/2015] [Indexed: 11/30/2022]
Abstract
Fe65 is a highly conserved adaptor protein that interacts with several binding partners. Fe65 binds proteins to mediate various cellular processes. But the interacting partner and the regulatory mechanisms controlled by Fe65 are largely unknown. In this study, we found that Fe65 interacts with the C-terminus of Jagged1. Furthermore, Fe65 negatively regulates AP1-mediated Jagged1 intercellular domain transactivation in a Tip60-independent manner. We found that Fe65 triggers the degradation of Jagged1, but not the Jagged1 intracellular domain (JICD), through both proteasome and lysosome pathways. We also showed that Fe65 promotes recruitment of the E3 ligase Neuralized-like 1 (Neurl1) to membrane-tethered Jagged1 and monoubiquitination of Jagged1. These three proteins form a stable trimeric complex, thereby decreasing Jagged1 targeting by ubiquitin-mediated degradation. Consequently, Jagged1 is a novel binding partner of Fe65, and Fe65 may act as a novel effector of Jagged1 signaling.
Collapse
Affiliation(s)
- Hye-Jin Lee
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Ji-Hye Yoon
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Ji-Seon Ahn
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Eun-Hye Jo
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Mi-Yeon Kim
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Young Chul Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jin Woo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Eun-Jung Ann
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea.
| | - Hee-Sae Park
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
21
|
|
22
|
Jiang S, Li Y, Zhang X, Bu G, Xu H, Zhang YW. Trafficking regulation of proteins in Alzheimer's disease. Mol Neurodegener 2014; 9:6. [PMID: 24410826 PMCID: PMC3891995 DOI: 10.1186/1750-1326-9-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 12/15/2013] [Indexed: 12/12/2022] Open
Abstract
The β-amyloid (Aβ) peptide has been postulated to be a key determinant in the pathogenesis of Alzheimer’s disease (AD). Aβ is produced through sequential cleavage of the β-amyloid precursor protein (APP) by β- and γ-secretases. APP and relevant secretases are transmembrane proteins and traffic through the secretory pathway in a highly regulated fashion. Perturbation of their intracellular trafficking may affect dynamic interactions among these proteins, thus altering Aβ generation and accelerating disease pathogenesis. Herein, we review recent progress elucidating the regulation of intracellular trafficking of these essential protein components in AD.
Collapse
Affiliation(s)
| | | | | | | | | | - Yun-wu Zhang
- Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
23
|
Cheung HNM, Dunbar C, Mórotz GM, Cheng WH, Chan HYE, Miller CCJ, Lau KF. FE65 interacts with ADP-ribosylation factor 6 to promote neurite outgrowth. FASEB J 2013; 28:337-49. [PMID: 24056087 DOI: 10.1096/fj.13-232694] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
FE65 is an adaptor protein that binds to the amyloid precursor protein (APP). As such, FE65 has been implicated in the pathogenesis of Alzheimer's disease. In addition, evidence suggests that FE65 is involved in brain development. It is generally believed that FE65 participates in these processes by recruiting various interacting partners to form functional complexes. Here, we show that via its first phosphotyrosine binding (PTB) domain, FE65 binds to the small GTPase ADP-ribosylation factor 6 (ARF6). FE65 preferentially binds to ARF6-GDP, and they colocalize in neuronal growth cones. Interestingly, FE65 stimulates the activation of both ARF6 and its downstream GTPase Rac1, a regulator of actin dynamics, and functions in growth cones to stimulate neurite outgrowth. We show that transfection of FE65 and/or ARF6 promotes whereas small interfering RNA knockdown of FE65 or ARF6 inhibits neurite outgrowth in cultured neurons as compared to the mock-transfected control cells. Moreover, knockdown of ARF6 attenuates FE65 stimulation of neurite outgrowth and defective neurite outgrowth seen in FE65-deficient neurons is partially corrected by ARF6 overexpression. Notably, the stimulatory effect of FE65 and ARF6 on neurite outgrowth is abrogated either by dominant-negative Rac1 or knockdown of Rac1. Thus, we identify FE65 as a novel regulator of neurite outgrowth via controlling ARF6-Rac1 signaling.
Collapse
Affiliation(s)
- Hei Nga Maggie Cheung
- 1School of Life Sciences, Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China.
| | | | | | | | | | | | | |
Collapse
|
24
|
Yang M, Virassamy B, Vijayaraj SL, Lim Y, Saadipour K, Wang YJ, Han YC, Zhong JH, Morales CR, Zhou XF. The intracellular domain of sortilin interacts with amyloid precursor protein and regulates its lysosomal and lipid raft trafficking. PLoS One 2013; 8:e63049. [PMID: 23704887 PMCID: PMC3660575 DOI: 10.1371/journal.pone.0063049] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 03/27/2013] [Indexed: 12/31/2022] Open
Abstract
The processing of Amyloid precursor protein (APP) is multifaceted, comprising of protein transport, internalization and sequential proteolysis. However, the exact mechanism of APP intracellular trafficking and distribution remains unclear. To determine the interaction between sortilin and APP and the effect of sortilin on APP trafficking and processing, we studied the binding site and its function by mapping experiments, colocalization, coimmunoprecipitation and sucrose gradient fractionation. We identified for the first time that sortilin interacts with APP at both N- and C-terminal regions. The sortilin-FLVHRY (residues 787–792) and APP-NPTYKFFE (residues 759–766) motifs are crucial for the C-terminal interaction. We also found that lack of the FLVHRY motif reduces APP lysosomal targeting and increases APP distribution in lipid rafts in co-transfected HEK293 cells. These results are consistent with our in vivo data where sortilin knockout mice showed a decrease of APP lysosomal distribution and an increase of APP in lipid rafts. We further confirmed that overexpression of sortilin-FLVHRY mutants failed to rescue the lysosomal degradation of APP. Thus, our data suggests that sortilin is implicated in APP lysosomal and lipid raft targeting via its carboxyl-terminal F/YXXXXF/Y motif. Our study provides new molecular insights into APP trafficking and processing.
Collapse
Affiliation(s)
- Miao Yang
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Evin G, Li QX. Platelets and Alzheimer’s disease: Potential of APP as a biomarker. World J Psychiatry 2012; 2:102-13. [PMID: 24175176 PMCID: PMC3782192 DOI: 10.5498/wjp.v2.i6.102] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2011] [Revised: 07/13/2012] [Accepted: 07/23/2012] [Indexed: 02/05/2023] Open
Abstract
Platelets are the first peripheral source of amyloid precursor protein (APP). They possess the proteolytic machinery to produce Aβ and fragments similar to those produced in neurons, and thus offer an ex-vivo model to study APP processing and changes associated with Alzheimer’s disease (AD). Platelet process APP mostly through the α-secretase pathway to release soluble APP (sAPP). They produce small amounts of Aβ, predominantly Aβ40 over Aβ42. sAPP and Aβ are stored in α-granules and are released upon platelet activation by thrombin and collagen, and agents inducing platelet degranulation. A small proportion of full-length APP is present at the platelet surface and this increases by 3-fold upon platelet activation. Immunoblotting of platelet lysates detects APP as isoforms of 130 kDa and 106-110 kDa. The ratio of these of APP isoforms is significantly lower in patients with AD and mild cognitive impairment (MCI) than in healthy controls. This ratio follows a decrease that parallels cognitive decline and can predict conversion from MCI to AD. Alterations in the levels of α-secretase ADAM10 and in the enzymatic activities of α- and β-secretase observed in platelets of patients with AD are consistent with increased processing through the amyloidogenic pathway. β-APP cleaving enzyme activity is increased by 24% in platelet membranes of patients with MCI and by 17% in those with AD. Reports of changes in platelet APP expression with MCI and AD have been promising so far and merit further investigation as the search for blood biomarkers in AD, in particular at the prodromal stage, remains a priority and a challenge.
Collapse
Affiliation(s)
- Geneviève Evin
- Geneviève Evin, Qiao-Xin Li, Department of Pathology and Mental Health Research Institute, The University of Melbourne, Parkville 3010, Australia
| | | |
Collapse
|
26
|
Park HJ, Shabashvili D, Nekorchuk MD, Shyqyriu E, Jung JI, Ladd TB, Moore BD, Felsenstein KM, Golde TE, Kim SH. Retention in endoplasmic reticulum 1 (RER1) modulates amyloid-β (Aβ) production by altering trafficking of γ-secretase and amyloid precursor protein (APP). J Biol Chem 2012; 287:40629-40. [PMID: 23043097 PMCID: PMC3504776 DOI: 10.1074/jbc.m112.418442] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 10/05/2012] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Aβ production is influenced by intracellular trafficking of secretases and amyloid precursor protein (APP). RESULTS Retention in endoplasmic reticulum 1 (RER1) regulates the trafficking of γ-secretase and APP, thereby influences Aβ production. CONCLUSION RER1, an ER retention/retrieval factor for γ-secretase and APP, modulates Aβ production. SIGNIFICANCE RER1 and its influence on γ-secretase and APP may be implicated for a safe strategy to target Aβ production. The presence of neuritic plaques containing aggregated amyloid-β (Aβ) peptides in the brain parenchyma is a pathological hallmark of Alzheimer disease (AD). Aβ is generated by sequential cleavage of the amyloid β precursor protein (APP) by β- and γ-secretase, respectively. As APP processing to Aβ requires transport through the secretory pathway, trafficking of the substrate and access to the secretases are key factors that can influence Aβ production (Thinakaran, G., and Koo, E. H. (2008) Amyloid precursor protein trafficking, processing, and function. J. Biol. Chem. 283, 29615-29619). Here, we report that retention in endoplasmic reticulum 1 (RER1) associates with γ-secretase in early secretory compartments and regulates the intracellular trafficking of γ-secretase. RER1 overexpression decreases both γ-secretase localization on the cell surface and Aβ secretion and conversely RER1 knockdown increases the level of cell surface γ-secretase and increases Aβ secretion. Furthermore, we find that increased RER1 levels decrease mature APP and increase immature APP, resulting in less surface accumulation of APP. These data show that RER1 influences the trafficking and localization of both γ-secretase and APP, thereby regulating the production and secretion of Aβ peptides.
Collapse
Affiliation(s)
- Hyo-Jin Park
- From the Department of Pharmacology and Therapeutics, and
- the Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | | | | | - Eva Shyqyriu
- From the Department of Pharmacology and Therapeutics, and
| | - Joo In Jung
- the Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Thomas B. Ladd
- the Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Brenda D. Moore
- the Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Kevin M. Felsenstein
- the Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Todd E. Golde
- the Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Seong-Hun Kim
- From the Department of Pharmacology and Therapeutics, and
| |
Collapse
|
27
|
Abstract
Ubiquitin-dependent proteolysis is a major mechanism that downregulates misfolded proteins or those that have finished a programmed task. In the last two decades, neddylation has emerged as a major regulatory pathway for ubiquitination. Central to the neddylation pathway is the amyloid precursor protein (APP)-binding protein APP-BP1, which together with Uba3, plays an analogous role to the ubiquitin-activating enzyme E1 in nedd8 activation. Activated nedd8 covalently modifies and activates a major class of ubiquitin ligases called Cullin-RING ligases (CRLs). New evidence suggests that neddylation also modifies Type-1 transmembrane receptors such as APP. Here we review the functions of neddylation and summarize evidence suggesting that dysfunction of neddylation is involved in Alzheimer's disease.
Collapse
Affiliation(s)
- Yuzhi Chen
- Department of Geriatrics and Department of Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | | | | |
Collapse
|
28
|
Ntelios D, Berninger B, Tzimagiorgis G. Numb and Alzheimer's disease: the current picture. Front Neurosci 2012; 6:145. [PMID: 23060745 PMCID: PMC3463830 DOI: 10.3389/fnins.2012.00145] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 09/14/2012] [Indexed: 11/23/2022] Open
Abstract
Twenty-three years ago, numb was identified as a critical regulator in Drosophila sensory organ precursor cell asymmetric divisions. Beyond the recently recognized role in carcinogenesis, Numb seems to be important in Alzheimer’s disease. This assertion comes from the involvement in various processes such as synapse morphogenesis, amyloid precursor protein trafficking, notch signaling, and neurogenesis. The purpose of the present mini-review is to provide the current picture of Numb’s participation in mechanisms underlying Alzheimer’s disease pathogenesis and emphasize potential aspects for future research.
Collapse
Affiliation(s)
- Dimitrios Ntelios
- Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki Thessaloniki, Greece
| | | | | |
Collapse
|
29
|
Song P, Pimplikar SW. Knockdown of amyloid precursor protein in zebrafish causes defects in motor axon outgrowth. PLoS One 2012; 7:e34209. [PMID: 22545081 PMCID: PMC3335837 DOI: 10.1371/journal.pone.0034209] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 02/28/2012] [Indexed: 12/18/2022] Open
Abstract
Amyloid precursor protein (APP) plays a pivotal role in Alzheimer's disease (AD) pathogenesis, but its normal physiological functions are less clear. Combined deletion of the APP and APP-like protein 2 (APLP2) genes in mice results in post-natal lethality, suggesting that APP performs an essential, if redundant, function during embryogenesis. We previously showed that injection of antisense morpholino to reduce APP levels in zebrafish embryos caused convergent-extension defects. Here we report that a reduction in APP levels causes defective axonal outgrowth of facial branchiomotor and spinal motor neurons, which involves disorganized axonal cytoskeletal elements. The defective outgrowth is caused in a cell-autonomous manner and both extracellular and intracellular domains of human APP are required to rescue the defective phenotype. Interestingly, wild-type human APP rescues the defective phenotype but APPswe mutation, which causes familial AD, does not. Our results show that the zebrafish model provides a powerful system to delineate APP functions in vivo and to study the biological effects of APP mutations.
Collapse
Affiliation(s)
- Ping Song
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Sanjay W. Pimplikar
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
30
|
Beyer AS, von Einem B, Schwanzar D, Keller IE, Hellrung A, Thal DR, Ingelsson M, Makarova A, Deng M, Chhabra ES, Pröpper C, Böckers TM, Hyman BT, von Arnim CA. Engulfment adapter PTB domain containing 1 interacts with and affects processing of the amyloid-β precursor protein. Neurobiol Aging 2012; 33:732-43. [DOI: 10.1016/j.neurobiolaging.2010.06.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 06/08/2010] [Accepted: 06/09/2010] [Indexed: 01/22/2023]
|
31
|
Xie Z, Dong Y, Maeda U, Xia W, Tanzi RE. RNAi-mediated knock-down of Dab and Numb attenuate Aβ levels via γ-secretase mediated APP processing. Transl Neurodegener 2012; 1:8. [PMID: 23211096 PMCID: PMC3514095 DOI: 10.1186/2047-9158-1-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2011] [Accepted: 03/22/2012] [Indexed: 11/24/2022] Open
Abstract
Amyloid-β-protein (Aβ), the key component of senile plaques in Alzheimer's disease (AD) brain, is produced from amyloid precursor protein (APP) by cleavage of β-secretase and then γ-secretase. APP adaptor proteins with phosphotyrosine-binding (PTB) domains, including Dab (gene: DAB) and Numb (gene: NUMB), can bind to and interact with the conserved YENPTY-motif in the APP C-terminus. Here we describe, for the first time, the effects of RNAi knock-down of Dab and Numb expression on APP processing and Aβ production. RNAi knock-down of Dab and Numb in H4 human neuroglioma cells stably transfected to express either FL-APP (H4-FL-APP cells) or APP-C99 (H4-APP-C99 cells) increased levels of APP-C-terminal fragments (APP-CTFs) and lowered Aβ levels in both cell lines by inhibiting γ-secretase cleavage of APP. Finally, RNAi knock-down of APP also reduced levels of Numb in H4-APP cells. These findings suggest that pharmacologically blocking interaction of APP with Dab and Numb may provide novel therapeutic strategies of AD. The notion of attenuating γ-secretase cleavage of APP via the APP adaptor proteins, Dab and Numb, is particularly attractive with regard to therapeutic potential, given that side effects of γ-secretase inhibition owing to impaired proteolysis of other γ-secretase substrates, e.g. Notch, might be avoided.
Collapse
Affiliation(s)
- Zhongcong Xie
- Department of Neurology, Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129-2060, USA.
| | | | | | | | | |
Collapse
|
32
|
Yang Z, Fan Y, Deng Z, Wu B, Zheng Q. Amyloid precursor protein as a potential marker of malignancy and prognosis in papillary thyroid carcinoma. Oncol Lett 2012; 3:1227-1230. [PMID: 22783423 DOI: 10.3892/ol.2012.639] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 02/22/2012] [Indexed: 11/06/2022] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common carcinoma of the thyroid gland and has a relatively favorable prognosis. However, it is important to identify PTC characteristics that indicate a high risk for recurrence and metastasis. Recent data indicate that the amyloid precursor protein (APP) is involved in cell adhesion, motility and proliferation. At present, the expression levels of APP and their prognostic significance in PTC have not been studied. In the present study, the APP gene expression in 10 PTCs and their adjacent thyroid tissue samples was analyzed using quantitative real time-PCR. Tissue array blocks were obtained from 90 PTC cases. Tumor regions and their adjacent non-tumor regions were analyzed in tissue array blocks and immunohistochemistry was conducted using sectioned slides. Semi-quantitative scores were correlated with the clinicopathological factors of 90 PTC patients. APP-specific mRNA was increased 50-fold in PTC samples compared with their adjacent thyroid tissue. The immunohistochemistry results showed APP expression levels to be significantly increased in PTC samples compared with their adjacent non-tumor thyroid tissue (p<0.001). High APP scores were significantly correlated with large tumor size, extracapsular invasion and lymph node metastasis. The amyloid precursor protein was overexpressed in PTC and a high APP expression was associated with high malignant potential. Therefore, APP may serve as a prognostic marker and potential novel therapeutic target in PTC.
Collapse
Affiliation(s)
- Zhili Yang
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | | | | | | | | |
Collapse
|
33
|
Selective neutralization of APP-C99 with monoclonal antibodies reduces the production of Alzheimer's Aβ peptides. Neurobiol Aging 2012; 33:2704-14. [PMID: 22317957 DOI: 10.1016/j.neurobiolaging.2011.12.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 12/19/2011] [Accepted: 12/28/2011] [Indexed: 12/30/2022]
Abstract
The toxic amyloid-β (Aβ) peptides involved in Alzheimer's disease (AD) are produced after processing of the amyloid precursor protein-C-terminal fragment APP-C99 by γ-secretase. Thus, major therapeutic efforts have been focused on inhibiting the activity of this enzyme. However, preclinical and clinical trials testing γ-secretase inhibitors revealed adverse side effects most likely attributed to impaired processing of the Notch-1 receptor, a γ-secretase substrate critically involved in cell fate decisions. Here we report an innovative approach to selectively target the γ-secretase-mediated processing of APP-C99 with monoclonal antibodies neutralizing this substrate. Generated by immunizing mice with natively folded APP-C99, these antibodies bound N- or C-terminal accessible epitopes of this substrate, and decorated extracellular amyloid deposits in AD brain tissues. In cell-based assays, the same antibodies impaired APP-C99 processing by γ-secretase, and reduced Aβ production. Furthermore, they significantly decreased brain Aβ levels in the APPPS1 mouse model of AD after intracerebroventricular injection. Together, our findings support APP-C99 substrate-targeting antibodies as new immunotherapeutic and Notch-sparing agents to lower the levels of Aβ peptides implicated in AD.
Collapse
|
34
|
Positive evolutionary selection of an HD motif on Alzheimer precursor protein orthologues suggests a functional role. PLoS Comput Biol 2012; 8:e1002356. [PMID: 22319430 PMCID: PMC3271017 DOI: 10.1371/journal.pcbi.1002356] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 12/07/2011] [Indexed: 12/31/2022] Open
Abstract
HD amino acid duplex has been found in the active center of many different enzymes. The dyad plays remarkably different roles in their catalytic processes that usually involve metal coordination. An HD motif is positioned directly on the amyloid beta fragment (Aβ) and on the carboxy-terminal region of the extracellular domain (CAED) of the human amyloid precursor protein (APP) and a taxonomically well defined group of APP orthologues (APPOs). In human Aβ HD is part of a presumed, RGD-like integrin-binding motif RHD; however, neither RHD nor RXD demonstrates reasonable conservation in APPOs. The sequences of CAEDs and the position of the HD are not particularly conserved either, yet we show with a novel statistical method using evolutionary modeling that the presence of HD on CAEDs cannot be the result of neutral evolutionary forces (p<0.0001). The motif is positively selected along the evolutionary process in the majority of APPOs, despite the fact that HD motif is underrepresented in the proteomes of all species of the animal kingdom. Position migration can be explained by high probability occurrence of multiple copies of HD on intermediate sequences, from which only one is kept by selective evolutionary forces, in a similar way as in the case of the “transcription binding site turnover.” CAED of all APP orthologues and homologues are predicted to bind metal ions including Amyloid-like protein 1 (APLP1) and Amyloid-like protein 2 (APLP2). Our results suggest that HDs on the CAEDs are most probably key components of metal-binding domains, which facilitate and/or regulate inter- or intra-molecular interactions in a metal ion-dependent or metal ion concentration-dependent manner. The involvement of naturally occurring mutations of HD (Tottori (D7N) and English (H6R) mutations) in early onset Alzheimer's disease gives additional support to our finding that HD has an evolutionary preserved function on APPOs. HD amino acid duplex can be found in the active center of different metallo-enzymes. An HD motif is positioned directly on the amyloid beta (Aβ) fragment and on the carboxy-terminal region of the extracellular domain of the human amyloid precursor protein (APP) and a taxonomically well defined group of APP orthologues (APPOs). The conservation of the HD dyad is not position specific and it cannot be seen in a multiple alignment. Yet we show with a novel statistical method using evolutionary modeling that HD motif is positively selected by evolution on APPOs, despite the fact that HD dyad is underrepresented in the proteomes of all species of the animal kingdom. CAED of all APP orthologues and homologues are predicted to bind metal ions including Amyloid-like protein 1 (APLP1) and Amyloid-like protein 2 (APLP2). Our results suggest that HDs on the APPOs are most probably key components of metal-binding domains, which facilitate and/or regulate inter- or intra-molecular interactions in a metal ion-dependent or metal ion concentration-dependent manner. The involvement of naturally occurring mutations of HD (Tottori (D7N) and English (H6R)) in early onset Alzheimer's disease gives additional support to our finding that HD has an evolutionary preserved function on APPOs.
Collapse
|
35
|
Jeon YH, Lee JY, Kim S. Chemical modulators working at pharmacological interface of target proteins. Bioorg Med Chem 2011; 20:1893-901. [PMID: 22227462 DOI: 10.1016/j.bmc.2011.12.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 11/30/2011] [Accepted: 12/08/2011] [Indexed: 01/23/2023]
Abstract
For last few decades, the active site cleft and substrate-binding site of enzymes as well as ligand-binding site of the receptors have served as the main pharmacological space for drug discovery. However, rapid accumulation of proteome and protein network analysis data has opened a new therapeutic space that is the interface between the interacting proteins. Due to the complexity of the interaction modes and the numbers of the participating components, it is still challenging to identify the chemicals that can accurately control the protein-protein interactions at desire. Nonetheless, the number of chemical drugs and candidates working at the interface of the interacting proteins are rapidly increasing. This review addresses the current case studies and state-of-the-arts in the development of small chemical modulators controlling the interactions of the proteins that have pathological implications in various human diseases such as cancer, immune disorders, neurodegenerative and infectious diseases.
Collapse
Affiliation(s)
- Young Ho Jeon
- Korea University College of Pharmacy Sejong-ro, Jochiwon, Yeonggi-gun, Chungnam 339-700, Republic of Korea
| | | | | |
Collapse
|
36
|
Ha S, Furukawa R, Fechheimer M. Association of AICD and Fe65 with Hirano bodies reduces transcriptional activation and initiation of apoptosis. Neurobiol Aging 2011; 32:2287-98. [PMID: 20133016 PMCID: PMC2894277 DOI: 10.1016/j.neurobiolaging.2010.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Revised: 12/22/2009] [Accepted: 01/07/2010] [Indexed: 02/03/2023]
Abstract
Hirano bodies are cytoplasmic inclusions predominantly found in the central nervous system associated with various conditions including aging and Alzheimer's disease (AD). Since most studies of Hirano bodies have been performed in post-mortem samples, the physiological roles of Hirano bodies have not been investigated. Astrocytoma H4 cells were employed to test the hypothesis that Hirano bodies interact with and modulate signaling by the C-terminal fragment of amyloid-β precursor protein (AICD). We demonstrated by immunofluorescence and immunoprecipitation that model Hirano bodies accumulate AICD. Since stimulation of transcription by AICD is dependent on its interaction with the nuclear adaptor protein Fe65, we examined localization of Fe65, and employed a dual luciferase reporter assay to test the effects of Hirano bodies on AICD- and Fe65-dependent modulation of gene expression. We find that both AICD and Fe65 are co-localized in model Hirano bodies. Model Hirano bodies also down-regulate both AICD-dependent apoptosis and AICD- and Fe65-dependent transcriptional activity. Thus, association of AICD and Fe65 with Hirano bodies impedes their function in promoting apoptosis and modulating transcription.
Collapse
Affiliation(s)
- Sangdeuk Ha
- Department of Cellular Biology, University of Georgia, Athens, GA 30602 USA
| | - Ruth Furukawa
- Department of Cellular Biology, University of Georgia, Athens, GA 30602 USA
| | - Marcus Fechheimer
- Department of Cellular Biology, University of Georgia, Athens, GA 30602 USA
| |
Collapse
|
37
|
Abstract
The pathogenic model of Alzheimer's disease (AD) posits that aggregates of amyloid β, a product of amyloid precursor protein (APP) processing, cause dementia. However, alterations of normal APP functions could contribute to AD pathogenesis, and it is therefore important to understand the role of APP. APP is a member of a gene family that shows functional redundancy as documented by the evidence that single knock-out mice are viable, whereas mice with combined deletions of APP family genes die shortly after birth. A residue in the APP intracellular region, Y(682), is indispensable for these essential functions of APP. It is therefore important to identify pathways that regulate phosphorylation of Y(682) as well as the role of Y(682) in vivo. TrkA is associated with both phosphorylation of APP-Y(682) and alteration of APP processing, suggesting that tyrosine phosphorylation of APP links APP processing and neurotrophic signaling to intracellular pathways associated with cellular differentiation and survival. Here we have tested whether the NGF/TrkA signaling pathway is a physiological regulator of APP phosphorylation. We find that NGF induces tyrosine phosphorylation of APP, and that APP interacts with TrkA and this interaction requires Y(682). Unpredictably, we also uncover that APP, and specifically Y(682), regulates activation of the NGF/TrkA signaling pathway in vivo, the subcellular distribution of TrkA and the sensitivity of neurons to the trophic action of NGF. This evidence suggests that these two membrane protein's functions are strictly interconnected and that the NGF/TrkA signaling pathway is involved in AD pathogenesis and can be used as a therapeutic target.
Collapse
|
38
|
Suh J, Lyckman A, Wang L, Eckman EA, Guénette SY. FE65 proteins regulate NMDA receptor activation-induced amyloid precursor protein processing. J Neurochem 2011; 119:377-88. [PMID: 21824144 PMCID: PMC3188680 DOI: 10.1111/j.1471-4159.2011.07419.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Amyloid precursor protein (APP) family members and their proteolytic products are implicated in normal nervous system function and Alzheimer's disease pathogenesis. APP processing and Aβ secretion are regulated by neuronal activity. Various data suggest that NMDA receptor (NMDAR) activity plays a role in both non-amyloidogenic and amyloidogenic APP processing depending on whether synaptic or extrasynaptic NMDARs are activated, respectively. The APP-interacting FE65 proteins modulate APP trafficking and processing in cell lines, but little is known about their contribution to APP trafficking and processing in neurons, either in vivo or in vitro. In this study, we examined the contribution of the FE65 protein family to APP trafficking and processing in WT and FE65/FE65L1 double knockout neurons under basal conditions and following NMDAR activation. We report that FE65 proteins facilitate neuronal Aβ secretion without affecting APP fast axonal transport to pre-synaptic terminals. In addition, FE65 proteins facilitate an NMDAR-dependent non-amyloidogenic APP processing pathway. Generation of high-molecular weight (HMW) species bearing an APP C-terminal epitope was also observed following NMDAR activation. These HMW species require proteasomal and calpain activities for their accumulation. Recovery of APP polypeptide fragments from electroeluted HMW species having molecular weights consistent with calpain I cleavage of APP suggests that HMW species are complexes formed from APP metabolic products. Our results indicate that the FE65 proteins contribute to physiological APP processing and accumulation of APP metabolic products resulting from NMDAR activation.
Collapse
Affiliation(s)
- Jaehong Suh
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | | | | | | | | |
Collapse
|
39
|
Abstract
The amyloid precursor protein (APP) has been under intensive study in recent years, mainly due to its critical role in the pathogenesis of Alzheimer's disease (AD). β-Amyloid (Aβ) peptides generated from APP proteolytic cleavage can aggregate, leading to plaque formation in human AD brains. Point mutations of APP affecting Aβ production are found to be causal for hereditary early onset familial AD. It is very likely that elucidating the physiological properties of APP will greatly facilitate the understanding of its role in AD pathogenesis. A number of APP loss- and gain-of-function models have been established in model organisms including Caenorhabditis elegans, Drosophila, zebrafish and mouse. These in vivo models provide us valuable insights into APP physiological functions. In addition, several knock-in mouse models expressing mutant APP at a physiological level are available to allow us to study AD pathogenesis without APP overexpression. This article will review the current physiological and pathophysiological animal models of APP.
Collapse
|
40
|
Minami SS, Clifford TG, Hoe HS, Matsuoka Y, Rebeck GW. Fyn knock-down increases Aβ, decreases phospho-tau, and worsens spatial learning in 3×Tg-AD mice. Neurobiol Aging 2011; 33:825.e15-24. [PMID: 21741124 DOI: 10.1016/j.neurobiolaging.2011.05.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 04/13/2011] [Accepted: 05/25/2011] [Indexed: 10/18/2022]
Abstract
Fyn kinase phosphorylates tau and exacerbates amyloid beta (Aβ)-mediated synaptic dysfunction. However, Fyn also increases the nonpathological cleavage of amyloid precursor protein (APP), suggesting opposing roles for Fyn in the pathogenesis of Alzheimer's disease (AD). To determine the effect of Fyn on both Aβ and tau pathologies, we crossed homozygous Alzheimer's disease triple transgenic (3×Tg) mice harboring mutations in amyloid precursor protein, presenilin-1, and tau with wild-type or Fyn knockout mice to generate Fyn(+/+)3×Tg(+/-) or Fyn(+/-)3×Tg(+/-) mice. We found that Fyn(+/-)3×Tg(+/-) mice had increased soluble and intracellular Aβ, and these changes were accompanied by impaired performance on the Morris water maze at 18 months. Fyn(+/-)3×Tg(+/-) mice had decreased phosphorylated tau at 15-18 months (as did Fyn knockout mice), but Fyn(+/-)3×Tg(+/-) mice had increased phosphorylated tau by 24 months. In addition, we observed that Fyn(+/-)3×Tg(+/-) males were delayed in developing Aβ pathology compared with females, and displayed better spatial learning performance at 18 months. Overall, these findings suggest that loss of Fyn at early stages of disease increases soluble Aβ accumulation and worsens spatial learning in the absence of changes in tau phosphorylation.
Collapse
Affiliation(s)
- S Sakura Minami
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057-1464, USA
| | | | | | | | | |
Collapse
|
41
|
Zheng H, Koo EH. Biology and pathophysiology of the amyloid precursor protein. Mol Neurodegener 2011; 6:27. [PMID: 21527012 PMCID: PMC3098799 DOI: 10.1186/1750-1326-6-27] [Citation(s) in RCA: 236] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Accepted: 04/28/2011] [Indexed: 01/22/2023] Open
Abstract
The amyloid precursor protein (APP) plays a central role in the pathophysiology of Alzheimer's disease in large part due to the sequential proteolytic cleavages that result in the generation of β-amyloid peptides (Aβ). Not surprisingly, the biological properties of APP have also been the subject of great interest and intense investigations. Since our 2006 review, the body of literature on APP continues to expand, thereby offering further insights into the biochemical, cellular and functional properties of this interesting molecule. Sophisticated mouse models have been created to allow in vivo examination of cell type-specific functions of APP together with the many functional domains. This review provides an overview and update on our current understanding of the pathobiology of APP.
Collapse
Affiliation(s)
- Hui Zheng
- Huffington Center on Aging and Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| | | |
Collapse
|
42
|
The intracellular threonine of amyloid precursor protein that is essential for docking of Pin1 is dispensable for developmental function. PLoS One 2011; 6:e18006. [PMID: 21445342 PMCID: PMC3062548 DOI: 10.1371/journal.pone.0018006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 02/17/2011] [Indexed: 12/11/2022] Open
Abstract
Background Processing of Aβ-precursor protein (APP) plays an important role in Alzheimer's Disease (AD) pathogenesis. Thr residue at amino acid 668 of the APP intracellular domain (AID) is highly conserved. When phosphorylated, this residue generates a binding site for Pin1. The interaction of APP with Pin1 has been involved in AD pathogenesis. Methodology/Principal Findings To dissect the functions of this sequence in vivo, we created an APP knock-in allele, in which Thr668 is replaced by an Ala (T668A). Doubly deficient APP/APP-like protein 2 (APLP2) mice present postnatal lethality and neuromuscular synapse defects. Previous work has shown that the APP intracellular domain is necessary for preventing early lethality and neuromuscular junctions (NMJ) defects. Crossing the T668A allele into the APLP2 knockout background showed that mutation of Thr668 does not cause a defective phenotype. Notably, the T668A mutant APP is able to bind Mint1. Conclusions/Significance Our results argue against an important role of the Thr668 residue in the essential function of APP in developmental regulation. Furthermore, they indicate that phosphorylation at this residue is not functionally involved in those APP-mediated functions that prevent (NMJ) defects and early lethality in APLP2 null mice.
Collapse
|
43
|
Chapuis J, Vingtdeux V, Campagne F, Davies P, Marambaud P. Growth arrest-specific 1 binds to and controls the maturation and processing of the amyloid-beta precursor protein. Hum Mol Genet 2011; 20:2026-36. [PMID: 21357679 DOI: 10.1093/hmg/ddr085] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD), the most common neurodegenerative disorder, is characterized by cerebral deposition of amyloid-β (Aβ), a series of peptides derived from the processing of the amyloid-β precursor protein (APP). To identify new candidate genes for AD, we recently performed a transcriptome analysis to screen for genes preferentially expressed in the hippocampus and located in AD linkage regions. This strategy identified CALHM1 (calcium homeostasis modulator 1), a gene modulating AD age at onset and Aβ metabolism. Here, we focused our attention on another candidate identified using this screen, growth arrest-specific 1 (Gas1), a gene involved in the central nervous system development. We found that Gas1 formed a complex with APP and controlled APP maturation and processing. Gas1 expression inhibited APP full glycosylation and routing to the cell surface by leading to a trafficking blockade of APP between the endoplasmic reticulum and the Golgi. Gas1 expression also resulted in a robust inhibition of APP transport into multivesicular bodies, further demonstrating that Gas1 negatively regulated APP intracellular trafficking. Consequently, Gas1 overexpression led to a reduction in Aβ production, and conversely, Gas1 silencing in cells expressing endogenously Gas1 increased Aβ levels. These results suggest that Gas1 is a novel APP-interacting protein involved in the control of APP maturation and processing.
Collapse
Affiliation(s)
- Julien Chapuis
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, North Shore-LIJ, Manhasset, NY 11030, USA
| | | | | | | | | |
Collapse
|
44
|
Abstract
Gonadotropin-releasing hormone-1 (GnRH-1) neurons migrate from the nasal placode to the forebrain where they control gonadal function via the hypothalamic-pituitary-gonadal axis. The birth of GnRH-1-expressing neurons is one of the first neurogenic events in the developing nasal placode. By gene expression screening on single GnRH-1 neurons, amyloid precursor binding protein-1 (FE65) was identified in migratory GnRH-1 neurons. FE65 has been shown to modulate β1-integrin dynamics, actin cytoskeleton, cell motility, and FE65/amyloid precursor protein signaling has been described in neuro/glial cell fate determination as well as in modulating neurogenesis. Analysis of two mouse lines, one deficient for the 97 kDa FE65 isoform and a second deficient for the 97 and 60 kDa forms of FE65, showed overlapping phenotypes. In both lines, no migratory defects of the GnRH-1 neurons were observed, but a 25% increase in GnRH-1 neuronal number during embryonic development was found. Bromodeoxyuridine birth tracing and spatiotemporal tracking of GnRH-1 cell precursors demonstrated that the lack of the N-terminal portion of FE65, which includes part of the functional nuclear translocation/gene transcription domain of FE65 (WW domain), extends the timing of GnRH-1 neurogenesis in the developing nasal placode without affecting proliferation of GnRH-1 neuronal progenitors or cell death. The observed changes in the dynamics of GnRH-1 neurogenesis highlight a unique role for the 97 kDa isoform of FE65 and suggest that GnRH-1 cells, which have a short neurogenic window, originate from multipotent progenitors able to generate distinct cell types as GnRH-1 neurogenesis declines in response to environmental changes.
Collapse
|
45
|
Barbagallo APM, Wang Z, Zheng H, D'Adamio L. A single tyrosine residue in the amyloid precursor protein intracellular domain is essential for developmental function. J Biol Chem 2011; 286:8717-21. [PMID: 21266574 DOI: 10.1074/jbc.c111.219873] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The Aβ-precursor protein (APP) intracellular domain is highly conserved and contains many potentially important residues, in particular the (682)YENPTY(687) motif. To dissect the functions of this sequence in vivo, we created an APP knock-in allele mutating Tyr(682) to Gly (Y682G). Crossing this allele to APP-like protein 2 (APLP2) knock-out background showed that mutation of Tyr(682) results in postnatal lethality and neuromuscular synapse defects similar to doubly deficient APP/APLP2 mice. Our results demonstrate that a single residue in the APP intracellular region, Tyr(682), is indispensable for the essential function of APP in developmental regulation.
Collapse
Affiliation(s)
- Alessia P M Barbagallo
- Department of Microbiology and Immunology, Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | |
Collapse
|
46
|
Zhang YW, Thompson R, Zhang H, Xu H. APP processing in Alzheimer's disease. Mol Brain 2011; 4:3. [PMID: 21214928 PMCID: PMC3022812 DOI: 10.1186/1756-6606-4-3] [Citation(s) in RCA: 617] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 01/07/2011] [Indexed: 12/12/2022] Open
Abstract
An important pathological feature of Alzheimer's disease (AD) is the presence of extracellular senile plaques in the brain. Senile plaques are composed of aggregations of small peptides called β-amyloid (Aβ). Multiple lines of evidence demonstrate that overproduction/aggregation of Aβ in the brain is a primary cause of AD and inhibition of Aβ generation has become a hot topic in AD research. Aβ is generated from β-amyloid precursor protein (APP) through sequential cleavages first by β-secretase and then by γ-secretase complex. Alternatively, APP can be cleaved by α-secretase within the Aβ domain to release soluble APPα and preclude Aβ generation. Cleavage of APP by caspases may also contribute to AD pathologies. Therefore, understanding the metabolism/processing of APP is crucial for AD therapeutics. Here we review current knowledge of APP processing regulation as well as the patho/physiological functions of APP and its metabolites.
Collapse
Affiliation(s)
- Yun-wu Zhang
- Institute for Biomedical Research, Xiamen University, 422 SiMingNanLu, Xiamen 361005, Fujian, PR China
| | | | | | | |
Collapse
|
47
|
Identification of NEEP21 as a ß-amyloid precursor protein-interacting protein in vivo that modulates amyloidogenic processing in vitro. J Neurosci 2010; 30:15677-85. [PMID: 21084623 DOI: 10.1523/jneurosci.4464-10.2010] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease and the most common form of dementia. AD is pathologically characterized by the deposition of pathogenic Aβ peptides that are derived from larger integral membrane proteins, termed β-amyloid precursor proteins (APPs). In an attempt to understand the function of APP, in vitro studies have focused on the identification of interacting proteins. To investigate the APP in vivo interactome in an unbiased manner, we generated mice that harbor a mouse prion protein promoter-driven cDNA encoding human APP-695 fused to a C-terminal affinity tag. Using this tag, we prepared mild detergent lysates from transgenic mouse brain cortical membrane preparations and isolated a number of previously identified APP-interacting proteins. In addition to these factors, mass spectrometric analysis revealed the presence of NEEP21 as a novel interacting protein. We now report that NEEP21 profoundly affects the processing of APP and Aβ production. Thus, this study demonstrates that using proteomic methods on our transgenic model can uncover important in vivo APP-interacting proteins that will provide insights into the biology of APP.
Collapse
|
48
|
Barbagallo APM, Weldon R, Tamayev R, Zhou D, Giliberto L, Foreman O, D'Adamio L. Tyr(682) in the intracellular domain of APP regulates amyloidogenic APP processing in vivo. PLoS One 2010; 5:e15503. [PMID: 21103325 PMCID: PMC2982846 DOI: 10.1371/journal.pone.0015503] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 10/08/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The pathogenesis of Alzheimer's disease is attributed to misfolding of Amyloid-β (Aβ) peptides. Aβ is generated during amyloidogenic processing of Aβ-precursor protein (APP). Another characteristic of the AD brain is increased phosphorylation of APP amino acid Tyr(682). Tyr(682) is part of the Y(682)ENPTY(687) motif, a docking site for interaction with cytosolic proteins that regulate APP metabolism and signaling. For example, normal Aβ generation and secretion are dependent upon Tyr(682) in vitro. However, physiological functions of Tyr(682) are unknown. METHODOLOGY/PRINCIPAL FINDINGS To this end, we have generated an APP Y682G knock-in (KI) mouse to help dissect the role of APP Tyr(682) in vivo. We have analyzed proteolytic products from both the amyloidogenic and non-amyloidogenic processing of APP and measure a profound shift towards non-amyloidogenic processing in APP KI mice. In addition, we demonstrate the essential nature of amino acid Tyr(682) for the APP/Fe65 interaction in vivo. CONCLUSIONS/SIGNIFICANCE Together, these observations point to an essential role of APP intracellular domain for normal APP processing and function in vivo, and provide rationale for further studies into physiological functions associated with this important phosphorylation site.
Collapse
Affiliation(s)
- Alessia P. M. Barbagallo
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Richard Weldon
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Robert Tamayev
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Dawang Zhou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Luca Giliberto
- The Litwin-Zucker Research Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, North Shore – LIJ, Manhasset, New York, United States of America
| | - Oded Foreman
- Department of Laboratory Animal Health, The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Luciano D'Adamio
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
49
|
Wiese M, Antebi A, Zheng H. Intracellular trafficking and synaptic function of APL-1 in Caenorhabditis elegans. PLoS One 2010; 5. [PMID: 20862215 PMCID: PMC2942829 DOI: 10.1371/journal.pone.0012790] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 08/21/2010] [Indexed: 11/24/2022] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disorder primarily characterized by the deposition of β-amyloid plaques in the brain. Plaques are composed of the amyloid-β peptide derived from cleavage of the amyloid precursor protein (APP). Mutations in APP lead to the development of Familial Alzheimer's Disease (FAD), however, the normal function of this protein has proven elusive. The organism Caenorhabditis elegans is an attractive model as the amyloid precursor-like protein (APL-1) is the single ortholog of APP, and loss of apl-1 leads to a severe molting defect and early larval lethality. Methodology/Principal Findings We report here that lethality and molting can be rescued by full length APL-1, C-terminal mutations as well as a C-terminal truncation, suggesting that the extracellular region of the protein is essential for viability. RNAi knock-down of apl-1 followed by drug testing on the acetylcholinesterase inhibitor aldicarb showed that loss of apl-1 leads to aldicarb hypersensitivity, indicating a defect in synaptic function. The aldicarb hypersensitivity can be rescued by full length APL-1 in a dose dependent fashion. At the cellular level, kinesins UNC-104/KIF-1A and UNC-116/kinesin-1 are positive regulators of APL-1 expression in the neurons. Knock-down of the small GTPase rab-5 also leads to a dramatic decrease in the amount of apl-1 expression in neurons, suggesting that trafficking from the plasma membrane to the early endosome is important for apl-1 function. Loss of function of a different small GTPase, UNC-108, on the contrary, leads to the retention of APL-1 in the cell body. Conclusions/Significance Our results reveal novel insights into the intracellular trafficking of APL-1 and we report a functional role for APL-1 in synaptic transmission.
Collapse
Affiliation(s)
- Mary Wiese
- Huffington Center on Aging and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Adam Antebi
- Huffington Center on Aging and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Max Planck Institute for Biology of Aging, Köln, Germany
- * E-mail: (AA); (HZ)
| | - Hui Zheng
- Huffington Center on Aging and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (AA); (HZ)
| |
Collapse
|
50
|
Soluble amyloid precursor protein (APP) regulates transthyretin and Klotho gene expression without rescuing the essential function of APP. Proc Natl Acad Sci U S A 2010; 107:17362-7. [PMID: 20855613 DOI: 10.1073/pnas.1012568107] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Amyloidogenic processing of the amyloid precursor protein (APP) generates a large secreted ectodomain fragment (APPsβ), β-amyloid (Aβ) peptides, and an APP intracellular domain (AICD). Whereas Aβ is viewed as critical for Alzheimer's disease pathogenesis, the role of other APP processing products remains enigmatic. Of interest, the AICD has been implicated in transcriptional regulation, and N-terminal cleavage of APPsβ has been suggested to produce an active fragment that may mediate axonal pruning and neuronal cell death. We previously reported that mice deficient in APP and APP-like protein 2 (APLP2) exhibit early postnatal lethality and neuromuscular synapse defects, whereas mice with neuronal conditional deletion of APP and APLP2 are viable. Using transcriptional profiling, we now identify transthyretin (TTR) and Klotho as APP/APLP2-dependent genes whose expression is decreased in loss-of-function states but increased in gain-of-function states. Significantly, by creating an APP knockin allele that expresses only APPsβ protein, we demonstrate that APPsβ is not normally cleaved in vivo and is fully capable of mediating the APP-dependent regulation of TTR and Klotho gene expression. Despite being an active regulator of gene expression, APPsβ did not rescue the lethality and neuromuscular synapse defects of APP and APLP2 double-KO animals. Our studies identify TTR and Klotho as physiological targets of APP that are regulated by soluble APPsβ independent of developmental APP functions. This unexpected APP-mediated signaling pathway may play an important role in maintaining TTR and Klotho levels and their respective functions in Aβ sequestration and aging.
Collapse
|