1
|
Benitez BA, Wallace CE, Patel M, Nykanen NP, Yuede CM, Eaton SL, Pottier C, Cetin A, Johnson M, Bevan MT, Gardiner WD, Edwards HM, Doherty BM, Harrigan RT, Kurian D, Wishart TM, Smith C, Cirrito JR, Sands MS. Haploinsufficiency of lysosomal enzyme genes in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.16.623962. [PMID: 39605615 PMCID: PMC11601326 DOI: 10.1101/2024.11.16.623962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
There is growing evidence suggesting that the lysosome or lysosome dysfunction is associated with Alzheimer's disease (AD). Pathway analysis of post mortem brain-derived proteomic data from AD patients shows that the lysosomal system is perturbed relative to similarly aged unaffected controls. However, it is unclear if these changes contributed to the pathogenesis or are a response to the disease. Consistent with the hypothesis that lysosome dysfunction contributes to AD pathogenesis, whole genome sequencing data indicate that heterozygous pathogenic mutations and predicted protein-damaging variants in multiple lysosomal enzyme genes are enriched in AD patients compared to matched controls. Heterozygous loss-of-function mutations in the palmitoyl protein thioesterase-1 (PPT1), α-L-iduronidase (IDUA), β-glucuronidase (GUSB), N-acetylglucosaminidase (NAGLU), and galactocerebrosidase (GALC) genes have a gene-dosage effect on Aβ40 levels in brain interstitial fluid in C57BL/6 mice and significantly increase Aβ plaque formation in the 5xFAD mouse model of AD, thus providing in vivo validation of the human genetic data. A more detailed analysis of PPT1 heterozygosity in 18-month-old mice revealed changes in α-, β-, and γ-secretases that favor an amyloidogenic pathway. Proteomic changes in brain tissue from aged PPT1 heterozygous sheep are consistent with both the mouse data and the potential activation of AD pathways. Finally, CNS-directed, AAV-mediated gene therapy significantly decreased Aβ plaques, increased life span, and improved behavioral performance in 5xFAD/PPT1+/- mice. Collectively, these data strongly suggest that heterozygosity of multiple lysosomal enzyme genes represent risk factors for AD and may identify precise therapeutic targets for a subset of genetically-defined AD patients.
Collapse
Affiliation(s)
- Bruno A Benitez
- Department of Medicine, Washington University, St. Louis, MO 63110
- Department of Psychiatry, Washington University, St. Louis, MO 63110
- Current address: Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Clare E Wallace
- Department of Neurology, Washington University, St. Louis, MO 63110
| | - Maulikkumar Patel
- Department of Psychiatry, Washington University, St. Louis, MO 63110
| | | | - Carla M Yuede
- Department of Psychiatry, Washington University, St. Louis, MO 63110
| | | | - Cyril Pottier
- Department of Psychiatry, Washington University, St. Louis, MO 63110
| | - Arda Cetin
- Department of Psychiatry, Washington University, St. Louis, MO 63110
| | - Matthew Johnson
- Department of Psychiatry, Washington University, St. Louis, MO 63110
| | - Mia T Bevan
- Department of Neurology, Washington University, St. Louis, MO 63110
| | | | - Hannah M Edwards
- Department of Neurology, Washington University, St. Louis, MO 63110
| | | | - Ryan T Harrigan
- Department of Neurology, Washington University, St. Louis, MO 63110
| | - Dominic Kurian
- Roslin Institute, University of Edinburgh, Edinburgh, EH25 9RG
| | - Thomas M Wishart
- Roslin Institute, University of Edinburgh, Edinburgh, EH25 9RG
- Current primary address: Centre for Systems Health and Integrated Metabolic Research, Department of Biosciences, School of Science and Technology, Nottingham Trent University, NHB 084, Clifton Campus, NG11 8NS
| | - Colin Smith
- Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - John R Cirrito
- Department of Neurology, Washington University, St. Louis, MO 63110
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110
| | - Mark S Sands
- Department of Medicine, Washington University, St. Louis, MO 63110
- Department of Genetics, Washington University, St. Louis, MO 63110
- Hope Center for Neurologic Disease, Washington University, St. Louis, MO 63110
| |
Collapse
|
2
|
Chiu J, Krupa JM, Seah C, Pasternak SH. Small GTPases control macropinocytosis of amyloid precursor protein and cleavage to amyloid-β. Heliyon 2024; 10:e31077. [PMID: 38799759 PMCID: PMC11126852 DOI: 10.1016/j.heliyon.2024.e31077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
The overproduction of the toxic peptide amyloid-beta (Aβ) generated from the cleavage of amyloid precursor protein (APP) is proposed to be a critical event in the development of Alzheimer's disease. Evidence suggests that the cleavage of APP occurs after its internalization from the cell surface. Previously, we identified a novel pathway for APP internalization, which trafficks cell surface APP directly to lysosomes by macropinocytosis, leading to its processing into Aβ. We also demonstrated that ADP-ribosylation factor 6 (Arf6) is required for the macropinocytosis of APP. Here, we characterized the roles of Arf6's downstream effectors Rac1, Cdc42 and RhoA. Both pharmacological inhibition and siRNA knockdown of these proteins reduced the amount of APP colocalized with LAMP1-labeled lysosomes without affecting APP transport to early endosomes. Decreases in the production of both Aβ40 and Aβ42 were also observed by ELISA in response to inhibitor treatment. These findings together demonstrate that Rac1, Cdc42 and RhoA are components of the mechanism regulating the macropinocytosis of APP and targeting these components can reduce the production of Aβ.
Collapse
Affiliation(s)
- Justin Chiu
- Department of Physiology and Pharmacology, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jordan M. Krupa
- Neuroscience Program, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Claudia Seah
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Stephen H. Pasternak
- Department of Physiology and Pharmacology, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Neuroscience Program, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Clinical Neurological Sciences, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
3
|
Sola M, Rendon-Angel A, Rojo Martinez V, Sgrignani J, Magrin C, Piovesana E, Cavalli A, Paganetti P, Papin S. Tau protein binds to the P53 E3 ubiquitin ligase MDM2. Sci Rep 2023; 13:10208. [PMID: 37353565 PMCID: PMC10290082 DOI: 10.1038/s41598-023-37046-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023] Open
Abstract
Tau gene mutations cause a progressive dementia and neurotoxic Tau forms deposited in neurofibrillary tangles are hallmarks of neurodegenerative tauopathies. Loss of non-canonical Tau functions may contribute to disease. In fact, Tau depletion affects the cellular response to DNA damage and tauopathies exhibit the accumulation of DNA lesions. Moreover, Tau modulates P53 activity and cell fate. Considering that MDM2 is the main antagonist of P53, we investigated, using orthogonal assays, if Tau interacts with MDM2. We report the existence in cells and brain of a Tau-MDM2 complex that, in vitro, exhibits reduced P53 ubiquitination activity in a manner sensitive to a Tau mutation. The Tau-MDM2 interaction involves the microtubule-binding domain of Tau and the acidic domain of MDM2, reminiscent of the binding of Tau to negatively charged microtubules. Notably, MDM2 accumulates aberrantly in neurofibrillary tangles. Aging-associated insults may expose a novel loss-of-function of Tau in neurodegeneration and cancer.
Collapse
Affiliation(s)
- Martina Sola
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Room 102a, Via Chiesa 5, 6500, Bellinzona, Switzerland
- PhD Program in Neurosciences, Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Azucena Rendon-Angel
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Room 102a, Via Chiesa 5, 6500, Bellinzona, Switzerland
| | - Viviana Rojo Martinez
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Room 102a, Via Chiesa 5, 6500, Bellinzona, Switzerland
| | - Jacopo Sgrignani
- Computational Structural Biology, Institute for Research in Biomedicine, Università Della Svizzera Italiana, Bellinzona, Switzerland
| | - Claudia Magrin
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Room 102a, Via Chiesa 5, 6500, Bellinzona, Switzerland
- PhD Program in Neurosciences, Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Ester Piovesana
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Room 102a, Via Chiesa 5, 6500, Bellinzona, Switzerland
- PhD Program in Neurosciences, Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Andrea Cavalli
- Computational Structural Biology, Institute for Research in Biomedicine, Università Della Svizzera Italiana, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Paolo Paganetti
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Room 102a, Via Chiesa 5, 6500, Bellinzona, Switzerland.
- PhD Program in Neurosciences, Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland.
- Neurocentro Della Svizzera Italiana, Ente Ospedaliero Cantonale, Lugano, Switzerland.
| | - Stéphanie Papin
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Room 102a, Via Chiesa 5, 6500, Bellinzona, Switzerland
| |
Collapse
|
4
|
Tautou M, Descamps F, Larchanché PE, Buée L, El Bakali J, Melnyk P, Sergeant N. A Polyaminobiaryl-Based β-secretase Modulator Alleviates Cognitive Impairments, Amyloid Load, Astrogliosis, and Neuroinflammation in APPSwe/PSEN1ΔE9 Mice Model of Amyloid Pathology. Int J Mol Sci 2023; 24:ijms24065285. [PMID: 36982363 PMCID: PMC10048993 DOI: 10.3390/ijms24065285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/03/2023] [Accepted: 02/15/2023] [Indexed: 03/12/2023] Open
Abstract
The progress in Alzheimer’s disease (AD) treatment suggests a combined therapeutic approach targeting the two lesional processes of AD, which include amyloid plaques made of toxic Aβ species and neurofibrillary tangles formed of aggregates of abnormally modified Tau proteins. A pharmacophoric design, novel drug synthesis, and structure-activity relationship enabled the selection of a polyamino biaryl PEL24-199 compound. The pharmacologic activity consists of a non-competitive β-secretase (BACE1) modulatory activity in cells. Curative treatment of the Thy-Tau22 model of Tau pathology restores short-term spatial memory, decreases neurofibrillary degeneration, and alleviates astrogliosis and neuroinflammatory reactions. Modulatory effects of PEL24-199 towards APP catalytic byproducts are described in vitro, but whether PEL24-199 can alleviate the Aβ plaque load and associated inflammatory counterparts in vivo remains to be elucidated. We investigated short- and long-term spatial memory, Aβ plaque load, and inflammatory processes in APPSwe/PSEN1ΔE9 PEL24-199 treated transgenic model of amyloid pathology to achieve this objective. PEL24-199 curative treatment induced the recovery of spatial memory and decreased the amyloid plaque load in association with decreased astrogliosis and neuroinflammation. The present results underline the synthesis and selection of a promising polyaminobiaryl-based drug that modulates both Tau and, in this case, APP pathology in vivo via a neuroinflammatory-dependent process.
Collapse
Affiliation(s)
- Marie Tautou
- Univ. Lille, Inserm, CHU Lille, UMRS1172—LilNCog—Lille Neuroscience & Cognition, 59000 Lille, France
| | - Florian Descamps
- Univ. Lille, Inserm, CHU Lille, UMRS1172—LilNCog—Lille Neuroscience & Cognition, 59000 Lille, France
| | - Paul-Emmanuel Larchanché
- Univ. Lille, Inserm, CHU Lille, UMRS1172—LilNCog—Lille Neuroscience & Cognition, 59000 Lille, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, UMRS1172—LilNCog—Lille Neuroscience & Cognition, 59000 Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, 59045 Lille, France
| | - Jamal El Bakali
- Univ. Lille, Inserm, CHU Lille, UMRS1172—LilNCog—Lille Neuroscience & Cognition, 59000 Lille, France
| | - Patricia Melnyk
- Univ. Lille, Inserm, CHU Lille, UMRS1172—LilNCog—Lille Neuroscience & Cognition, 59000 Lille, France
- Correspondence: (P.M.); (N.S.); Tel.: +33-663101728 (N.S.)
| | - Nicolas Sergeant
- Univ. Lille, Inserm, CHU Lille, UMRS1172—LilNCog—Lille Neuroscience & Cognition, 59000 Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, 59045 Lille, France
- Correspondence: (P.M.); (N.S.); Tel.: +33-663101728 (N.S.)
| |
Collapse
|
5
|
Limone A, Veneruso I, D'Argenio V, Sarnataro D. Endosomal trafficking and related genetic underpinnings as a hub in Alzheimer's disease. J Cell Physiol 2022; 237:3803-3815. [PMID: 35994714 DOI: 10.1002/jcp.30864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/13/2022] [Accepted: 08/08/2022] [Indexed: 01/07/2023]
Abstract
Genetic studies support the amyloid cascade as the leading hypothesis for the pathogenesis of Alzheimer's disease (AD). Although significant efforts have been made in untangling the amyloid and other pathological events in AD, ongoing interventions for AD have not been revealed efficacious for slowing down disease progression. Recent advances in the field of genetics have shed light on the etiology of AD, identifying numerous risk genes associated with late-onset AD, including genes related to intracellular endosomal trafficking. Some of the bases for the development of AD may be explained by the recently emerging AD genetic "hubs," which include the processing pathway of amyloid precursor protein and the endocytic pathway. The endosomal genetic hub may represent a common pathway through which many pathological effects can be mediated and novel, alternative biological targets could be identified for therapeutic interventions. The aim of this review is to focus on the genetic and biological aspects of the endosomal compartments related to AD progression. We report recent studies which describe how changes in endosomal genetics impact on functional events, such as the amyloidogenic and non-amyloidogenic processing, degradative pathways, and the importance of receptors related to endocytic trafficking, including the 37/67 kDa laminin-1 receptor ribosomal protein SA, and their implications for neurodegenerative diseases.
Collapse
Affiliation(s)
- Adriana Limone
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Napoli, Italy
| | - Iolanda Veneruso
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Napoli, Italy.,CEINGE-Biotecnologie Avanzate, Napoli, Italy
| | - Valeria D'Argenio
- CEINGE-Biotecnologie Avanzate, Napoli, Italy.,Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Roma, Italy
| | - Daniela Sarnataro
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Napoli, Italy
| |
Collapse
|
6
|
Ren W, Lou H, Ren X, Wen G, Wu X, Xia X, Wang S, Yu X, Yan L, Zhang G, Yao J, Lu Y, Wu X. Ketamine promotes the amyloidogenic pathway by regulating endosomal pH. Toxicology 2022; 471:153163. [PMID: 35378374 DOI: 10.1016/j.tox.2022.153163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
Abstract
Ketamine is an anesthetic and addictive drug that can cause cognitive dysfunction and neuroinflammation. Studies have shown that carboxy-terminal fragment derived from β-secretase (CTF-β) and amyloid beta (Aβ), the amyloidogenic products of amyloid precursor protein (APP), can also induce neuroinflammation and impair cognitive function. However, it remains unclear whether ketamine regulates the amyloidogenic pathway. In the endosome, APP is cleaved by beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), whose activity is influenced by pH. Endosomal acidification is mainly regulated by sodium hydrogen exchanger 6 (NHE6), which leaks protons out of endosomes, and vacuolar proton translocating ATPases (V-ATPase), which pump protons into endosomes. Therefore, we hypothesized that ketamine lowers the endosomal pH by reducing the endosomal NHE6 protein level, and this hyperacidification promotes the amyloidogenic pathway. We set up C57BL/6 J mouse models using 10, 20, 40, 80, and 100 mg/kg ketamine administration and SH-SY5Y cell models using 1, 10, 100, and 1000 μM ketamine administration to investigate its effects on the amyloidogenic pathway at different doses. Western blotting results showed that 100 mg/kg ketamine treatment in vivo and 1000 μM ketamine treatment in vitro increased endosomal BACE1 and CTF-β protein levels and reduced endosomal NHE6 and APP protein levels. The endosomal accumulation of BACE1 caused by ketamine administration was also observed using confocal imaging. Moreover, flow cytometry indicated that ketamine treatment lowered the endosomal pH value of SH-SY5Y cells. Later, cells were pretreated with monensin to restore the endosomal pH. Monensin did not affect amyloidogenic-related proteins or NHE6 directly; therefore, ketamine-promoted endosomal amyloidogenic processing and BACE1 accumulation were depleted by restoring endosomal acidity through monensin pretreatment. Finally, knockdown of NHE6 promoted the amyloidogenic pathway similarly and prevented further enhancement by ketamine. These results indicated that the effects of ketamine on the amyloidogenic pathway were dependent on the reduction of NHE6 and endosomal pH.
Collapse
Affiliation(s)
- Weishu Ren
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Haoyang Lou
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xinghua Ren
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Gehua Wen
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xue Wu
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xi Xia
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Shuying Wang
- Department of Anesthesiology, the First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaojin Yu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lei Yan
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Guohua Zhang
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Jun Yao
- School of Forensic Medicine, China Medical University, Shenyang, China.
| | - Yan Lu
- Key Laboratory of Health Ministry in Congenital Malformation, Affiliated Shengjing Hospital of China Medical University, Shenyang, China.
| | - Xu Wu
- School of Forensic Medicine, China Medical University, Shenyang, China.
| |
Collapse
|
7
|
Armbrust F, Bickenbach K, Marengo L, Pietrzik C, Becker-Pauly C. The Swedish dilemma - the almost exclusive use of APPswe-based mouse models impedes adequate evaluation of alternative β-secretases. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119164. [PMID: 34699873 DOI: 10.1016/j.bbamcr.2021.119164] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/30/2021] [Accepted: 10/11/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, however incurable so far. It is widely accepted that aggregated amyloid β (Aβ) peptides play a crucial role for the pathogenesis of AD, as they cause neurotoxicity and deposit as so-called Aβ plaques in AD patient brains. Aβ peptides derive from the amyloid precursor protein (APP) upon consecutive cleavage at the β- and γ-secretase site. Hence, mutations in the APP gene are often associated with autosomal dominant inherited AD. Almost thirty years ago, two mutations at the β-secretase site were observed in two Swedish families (termed Swedish APP (APPswe) mutations), which led to early-onset AD. Consequently, APPswe was established in almost every common AD mouse model, as it contributes to early Aβ plaque formation and cognitive impairments. Analyzing these APPswe-based mouse models, the aspartyl protease BACE1 has been evolving as the prominent β-secretase responsible for Aβ release in AD and as the most important therapeutic target for AD treatment. However, with respect to β-secretase processing, the very rare occurring APPswe variant substantially differs from wild-type APP. BACE1 dominates APPswe processing resulting in the release of Aβ1-x, whereas N-terminally truncated Aβ forms are scarcely generated. However, these N-terminally truncated Aβ species such as Aβ2-x, Aβ3-x and Aβ4-x are elevated in AD patient brains and exhibit an increased potential to aggregate compared to Aβ1-x peptides. Proteases such as meprin β, cathepsin B and ADAMTS4 were identified as alternative β-secretases being capable of generating these N-terminally truncated Aβ species from wild-type APP. However, neither meprin β nor cathepsin B are capable of generating N-terminally truncated Aβ peptides from APPswe. Hence, the role of BACE1 for the Aβ formation during AD might be overrepresented through the excessive use of APPswe mouse models. In this review we critically discuss the consideration of BACE1 as the most promising therapeutic target. Shifting the focus of AD research towards alternative β secretases might unveil promising alternatives to BACE1 inhibitors constantly failing in clinical trials due to ineffectiveness and harmful side effects.
Collapse
Affiliation(s)
- Fred Armbrust
- Biochemical Institute, Unit for Degradomics of the Protease Web, University of Kiel, Kiel, Germany
| | - Kira Bickenbach
- Biochemical Institute, Unit for Degradomics of the Protease Web, University of Kiel, Kiel, Germany
| | - Liana Marengo
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Claus Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | - Christoph Becker-Pauly
- Biochemical Institute, Unit for Degradomics of the Protease Web, University of Kiel, Kiel, Germany.
| |
Collapse
|
8
|
Tautou M, Eddarkaoui S, Descamps F, Larchanché PE, El Bakali J, Goveas LM, Dumoulin M, Lamarre C, Blum D, Buée L, Melnyk P, Sergeant N. A ß-Secretase Modulator Decreases Tau Pathology and Preserves Short-Term Memory in a Mouse Model of Neurofibrillary Degeneration. Front Pharmacol 2021; 12:679335. [PMID: 34267657 PMCID: PMC8276176 DOI: 10.3389/fphar.2021.679335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/18/2021] [Indexed: 01/24/2023] Open
Abstract
Identifying which among several in cellulo pharmacological activities is necessary for the proper in vivo activity is essential for further drug development against Alzheimer’s disease pathophysiological processes. An in-depth structure–activity relationship–based study has been carried out, and two molecules, named MAGS02-14 and PEL24-199, that share a ß-secretase modulatory effect associated or not to a lysosomotropic activity in cellulo have been identified. In terms of chemical formulas, MAGS02-14 and PEL24-199 only differ from each other by a single nitrogen atom. The study aimed to elucidate the in vivo pharmacological effects of lysosomotropic and/or the ß-secretase modulatory activity in a tau pathology mouse model. To address this question, the THY-Tau22 transgenic model of tauopathy was treated with both compounds for 6 weeks in a curative paradigm. Short-term memory, tau burden, and inflammatory processes were analyzed using orthogonal methods, and PEL24-199, but not MAGS02-14, was shown to restore the short-term memory and reduce the neurofibrillary degenerating process. These effects were associated with a reduced phosphorylation of tau, an increased phosphatase expression, and decreased astrogliosis. Our results, therefore, suggest that the lysosomotropic activity may be nonessential for the effect on tau pathology.
Collapse
Affiliation(s)
- Marie Tautou
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Sabiha Eddarkaoui
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Florian Descamps
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Paul-Emmanuel Larchanché
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Jamal El Bakali
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Liesel Mary Goveas
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Mélanie Dumoulin
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Chloé Lamarre
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - David Blum
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Luc Buée
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Patricia Melnyk
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Nicolas Sergeant
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| |
Collapse
|
9
|
Prasad H, Rao R. Endosomal Acid-Base Homeostasis in Neurodegenerative Diseases. Rev Physiol Biochem Pharmacol 2020; 185:195-231. [PMID: 32737755 PMCID: PMC7614123 DOI: 10.1007/112_2020_25] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Neurodegenerative disorders are debilitating and largely untreatable conditions that pose a significant burden to affected individuals and caregivers. Overwhelming evidence supports a crucial preclinical role for endosomal dysfunction as an upstream pathogenic hub and driver in Alzheimer's disease (AD) and related neurodegenerative disorders. We present recent advances on the role of endosomal acid-base homeostasis in neurodegeneration and discuss evidence for converging mechanisms. The strongest genetic risk factor in sporadic AD is the ε4 allele of Apolipoprotein E (ApoE4), which potentiates pre-symptomatic endosomal dysfunction and prominent amyloid beta (Aβ) pathology, although how these pathways are linked mechanistically has remained unclear. There is emerging evidence that the Christianson syndrome protein NHE6 is a prominent ApoE4 effector linking endosomal function to Aβ pathologies. By functioning as a dominant leak pathway for protons, the Na+/H+ exchanger activity of NHE6 limits endosomal acidification and regulates β-secretase (BACE)-mediated Aβ production and LRP1 receptor-mediated Aβ clearance. Pathological endosomal acidification may impact both Aβ generation and clearance mechanisms and emerges as a promising therapeutic target in AD. We also offer our perspective on the complex role of endosomal acid-base homeostasis in the pathogenesis of neurodegeneration and its therapeutic implications for neuronal rescue and repair strategies.
Collapse
Affiliation(s)
- Hari Prasad
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India, Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rajini Rao
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
10
|
Bhattacharya A, Limone A, Napolitano F, Cerchia C, Parisi S, Minopoli G, Montuori N, Lavecchia A, Sarnataro D. APP Maturation and Intracellular Localization Are Controlled by a Specific Inhibitor of 37/67 kDa Laminin-1 Receptor in Neuronal Cells. Int J Mol Sci 2020; 21:ijms21051738. [PMID: 32143270 PMCID: PMC7084285 DOI: 10.3390/ijms21051738] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/28/2020] [Accepted: 02/29/2020] [Indexed: 12/11/2022] Open
Abstract
Amyloid precursor protein (APP) is processed along both the nonamyloidogenic pathway preventing amyloid beta peptide (Aβ) production and the amyloidogenic pathway, generating Aβ, whose accumulation characterizes Alzheimer’s disease. Items of evidence report that the intracellular trafficking plays a key role in the generation of Aβ and that the 37/67 kDa LR (laminin receptor), acting as a receptor for Aβ, may mediate Aβ-pathogenicity. Moreover, findings indicating interaction between the receptor and the key enzymes involved in the amyloidogenic pathway suggest a strong link between 37/67 kDa LR and APP processing. We show herein that the specific 37/67 kDa LR inhibitor, NSC48478, is able to reversibly affect the maturation of APP in a pH-dependent manner, resulting in the partial accumulation of the immature APP isoforms (unglycosylated/acetylated forms) in the endoplasmic reticulum (ER) and in transferrin-positive recycling endosomes, indicating alteration of the APP intracellular trafficking. These effects reveal NSC48478 inhibitor as a novel small molecule to be tested in disease conditions, mediated by the 37/67 kDa LR and accompanied by inactivation of ERK1/2 (extracellular signal-regulated kinases) signalling and activation of Akt (serine/threonine protein kinase) with consequent inhibition of GSK3β.
Collapse
Affiliation(s)
- Antaripa Bhattacharya
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy; (A.B.); (A.L.); (S.P.); (G.M.)
| | - Adriana Limone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy; (A.B.); (A.L.); (S.P.); (G.M.)
| | - Filomena Napolitano
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy; (F.N.); (N.M.)
| | - Carmen Cerchia
- Department of Pharmacy, “Drug Discovery Lab”, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (C.C.); (A.L.)
| | - Silvia Parisi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy; (A.B.); (A.L.); (S.P.); (G.M.)
| | - Giuseppina Minopoli
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy; (A.B.); (A.L.); (S.P.); (G.M.)
| | - Nunzia Montuori
- Department of Translational Medical Sciences, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy; (F.N.); (N.M.)
| | - Antonio Lavecchia
- Department of Pharmacy, “Drug Discovery Lab”, University of Naples “Federico II”, Via D. Montesano 49, 80131 Naples, Italy; (C.C.); (A.L.)
| | - Daniela Sarnataro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy; (A.B.); (A.L.); (S.P.); (G.M.)
- Correspondence:
| |
Collapse
|
11
|
Hui L, Soliman ML, Geiger NH, Miller NM, Afghah Z, Lakpa KL, Chen X, Geiger JD. Acidifying Endolysosomes Prevented Low-Density Lipoprotein-Induced Amyloidogenesis. J Alzheimers Dis 2020; 67:393-410. [PMID: 30594929 DOI: 10.3233/jad-180941] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cholesterol dyshomeostasis has been linked to the pathogenesis of sporadic Alzheimer's disease (AD). In furthering the understanding of mechanisms by which increased levels of circulating cholesterol augments the risk of developing sporadic AD, others and we have reported that low-density lipoprotein (LDL) enters brain parenchyma by disrupting the blood-brain barrier and that endolysosome de-acidification plays a role in LDL-induced amyloidogenesis in neurons. Here, we tested the hypothesis that endolysosome de-acidification was central to amyloid-β (Aβ) generation and that acidifying endolysosomes protects against LDL-induced increases in Aβ levels in neurons. We demonstrated that LDL, but not HDL, de-acidified endolysosomes and increased intraneuronal and secreted levels of Aβ. ML-SA1, an agonist of endolysosome-resident TRPML1 channels, acidified endolysosomes, and TRPML1 knockdown attenuated ML-SA1-induced endolysosome acidification. ML-SA1 blocked LDL-induced increases in intraneuronal and secreted levels of Aβ as well as Aβ accumulation in endolysosomes, prevented BACE1 accumulation in endolysosomes, and decreased BACE1 activity levels. LDL downregulated TRPML1 protein levels, and TRPML1 knockdown worsens LDL-induced increases in Aβ. Our findings suggest that endolysosome acidification by activating TRPML1 may represent a protective strategy against sporadic AD.
Collapse
Affiliation(s)
- Liang Hui
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Mahmoud L Soliman
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nicholas H Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nicole M Miller
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Zahra Afghah
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Koffi L Lakpa
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
12
|
Scholz D, Chernyshova Y, Ückert AK, Leist M. Reduced Aβ secretion by human neurons under conditions of strongly increased BACE activity. J Neurochem 2018; 147:256-274. [PMID: 29804308 DOI: 10.1111/jnc.14467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/06/2018] [Accepted: 05/23/2018] [Indexed: 12/20/2022]
Abstract
The initial step in the amyloidogenic cascade of amyloid precursor protein (APP) processing is catalyzed by beta-site APP-cleaving enzyme (BACE), and this protease has increased activities in affected areas of Alzheimer's disease brains. We hypothesized that altered APP processing, because of augmented BACE activity, would affect the actions of direct and indirect BACE inhibitors. We therefore compared post-mitotic human neurons (LUHMES) with their BACE-overexpressing counterparts (BLUHMES). Although β-cleavage of APP was strongly increased in BLUHMES, they produced less full-length and truncated amyloid beta (Aβ) than LUHMES. Moreover, low concentrations of BACE inhibitors decreased cellular BACE activity as expected, but increased Aβ1-40 levels. Several other approaches to modulate BACE activity led to a similar, apparently paradoxical, behavior. For instance, reduction in intracellular acidification by bepridil increased Aβ production in parallel with decreased BACE activity. In contrast to BLUHMES, the respective control cells (LUHMES or BLUHMES with catalytically inactive BACE) showed conventional pharmacological responses. Other non-canonical neurochemical responses (so-called 'rebound effects') are well-documented for the Aβ pathway, especially for γ-secretase: a partial block of its activity leads to an increased Aβ secretion by some cell types. We therefore compared LUHMES and BLUHMES regarding rebound effects of γ-secretase inhibitors and found an Aβ rise in LUHMES but not in BLUHMES. Thus, different cellular factors are responsible for the γ-secretase- versus BACE-related Aβ rebound. We conclude that increased BACE activity, possibly accompanied by an altered cellular localization pattern, can dramatically influence Aβ generation in human neurons and affect pharmacological responses to secretase inhibitors. OPEN PRACTICES: Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Diana Scholz
- Chair for in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Yana Chernyshova
- Chair for in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Anna-Katharina Ückert
- Chair for in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - Marcel Leist
- Chair for in vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| |
Collapse
|
13
|
Gay M, Carato P, Coevoet M, Renault N, Larchanché PE, Barczyk A, Yous S, Buée L, Sergeant N, Melnyk P. New phenylaniline derivatives as modulators of amyloid protein precursor metabolism. Bioorg Med Chem 2018; 26:2151-2164. [PMID: 29559198 DOI: 10.1016/j.bmc.2018.03.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/08/2018] [Accepted: 03/10/2018] [Indexed: 11/25/2022]
Abstract
The chloroquinoline scaffold is characteristic of anti-malarial drugs such as chloroquine (CQ) or amodiaquine (AQ). These drugs are also described for their potential effectiveness against prion disease, HCV, EBV, Ebola virus, cancer, Parkinson or Alzheimer diseases. Amyloid precursor protein (APP) metabolism is deregulated in Alzheimer's disease. Indeed, CQ modifies amyloid precursor protein (APP) metabolism by precluding the release of amyloid-beta peptides (Aβ), which accumulate in the brain of Alzheimer patients to form the so-called amyloid plaques. We showed that AQ and analogs have similar effects although having a higher cytotoxicity. Herein, two new series of compounds were synthesized by replacing 7-chloroquinolin-4-amine moiety of AQ by 2-aminomethylaniline and 2-aminomethylphenyle moieties. Their structure activity relationship was based on their ability to modulate APP metabolism, Aβ release, and their cytotoxicity similarly to CQ. Two compounds 15a, 16a showed interesting and potent effect on the redirection of APP metabolism toward a decrease of Aβ peptide release (in the same range compared to AQ), and a 3-10-fold increased stability of APP carboxy terminal fragments (CTFα and AICD) without obvious cellular toxicity at 100 µM.
Collapse
Affiliation(s)
- Marion Gay
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000 Lille, France
| | - Pascal Carato
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000 Lille, France
| | - Mathilde Coevoet
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000 Lille, France
| | - Nicolas Renault
- Univ. Lille, Inserm, CHU Lille, U995 - LIRIC - Lille Inflammation Research International Center, F-59000 Lille, France
| | - Paul-Emmanuel Larchanché
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000 Lille, France
| | - Amélie Barczyk
- Univ. Lille, Inserm, CHU Lille, U995 - LIRIC - Lille Inflammation Research International Center, F-59000 Lille, France
| | - Saïd Yous
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000 Lille, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000 Lille, France
| | - Nicolas Sergeant
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000 Lille, France
| | - Patricia Melnyk
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000 Lille, France.
| |
Collapse
|
14
|
Wu Z, Ni J, Liu Y, Teeling JL, Takayama F, Collcutt A, Ibbett P, Nakanishi H. Cathepsin B plays a critical role in inducing Alzheimer's disease-like phenotypes following chronic systemic exposure to lipopolysaccharide from Porphyromonas gingivalis in mice. Brain Behav Immun 2017; 65:350-361. [PMID: 28610747 DOI: 10.1016/j.bbi.2017.06.002] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 12/14/2022] Open
Abstract
A number of clinical and experimental studies have revealed a strong association between periodontitis and accelerated cognitive decline in Alzheimer's disease (AD); however, the mechanism of the association is unknown. In the present study, we tested the hypothesis that cathepsin (Cat) B plays a critical role in the initiation of neuroinflammation and neural dysfunction following chronic systemic exposure to lipopolysaccharide from Porphyromonas gingivalis (PgLPS) in mice (1mg/kg, daily, intraperitoneally). Young (2months old) and middle-aged (12months old) wild-type (WT; C57BL/6N) or CatB-deficient (CatB-/-) mice were exposed to PgLPS daily for 5 consecutive weeks. The learning and memory function were assessed using the passive avoidance test, and the expression of amyloid precursor protein (APP), CatB, TLR2 and IL-1β was analyzed in brain tissues by immunohistochemistry and Western blotting. We found that chronic systemic exposure to PgLPS for five consecutive weeks induced learning and memory deficits with the intracellular accumulation of Aβ in neurons in the middle-aged WT mice, but not in young WT or middle-aged CatB-/- mice. PgLPS significantly increased the expression of CatB in both microglia and neurons in middle-aged WT mice, while increased expression of mature IL-1β and TLR2 was restricted to microglia in the hippocampus of middle-aged WT mice, but not in that of the middle-aged CatB-/- ones. In in vitro studies, PgLPS (1µg/ml) stimulation upregulated the mean mRNA expression of IL-1β, TLR2 and downregulated the protein levels of IκBα in the cultured MG6 microglia as well as in the primary microglia from WT mice, which were significantly inhibited by the CatB-specific inhibitor CA-074Me as well as by the primary microglia from CatB-/- mice. Furthermore, the mean mRNA expression of APP and CatB were significantly increased in the primary cultured hippocampal neurons after treatment with conditioned medium from PgLPS-treated WT primary microglia, but not after treatment with conditioned medium neutralized with anti-IL-1beta, and not after treatment with conditioned medium from PgLPS-treated CatB-/- primary microglia or with PgLPS directly. Taken together, these findings indicate that chronic systemic exposure to PgLPS induces AD-like phenotypes, including microglia-mediated neuroinflammation, intracellular Aβ accumulation in neurons and impairment of the learning and memory functions in the middle-aged mice in a CatB-dependent manner. We propose that CatB may be a therapeutic target for preventing periodontitis-associated cognitive decline in AD.
Collapse
Affiliation(s)
- Zhou Wu
- Department of Aging Science and Pharmacology, Kyushu University, Japan; OBT Research Center, Faculty of Dental Science, Kyushu University, Japan.
| | - Junjun Ni
- Department of Aging Science and Pharmacology, Kyushu University, Japan
| | - Yicong Liu
- Department of Aging Science and Pharmacology, Kyushu University, Japan
| | - Jessica L Teeling
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, United Kingdom
| | - Fumiko Takayama
- Department of Aging Science and Pharmacology, Kyushu University, Japan
| | - Alex Collcutt
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, United Kingdom
| | - Paul Ibbett
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, United Kingdom
| | - Hiroshi Nakanishi
- Department of Aging Science and Pharmacology, Kyushu University, Japan
| |
Collapse
|
15
|
Gleevec shifts APP processing from a β-cleavage to a nonamyloidogenic cleavage. Proc Natl Acad Sci U S A 2017; 114:1389-1394. [PMID: 28115709 DOI: 10.1073/pnas.1620963114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Neurotoxic amyloid-β peptides (Aβ) are major drivers of Alzheimer's disease (AD) and are formed by sequential cleavage of the amyloid precursor protein (APP) by β-secretase (BACE) and γ-secretase. Our previous study showed that the anticancer drug Gleevec lowers Aβ levels through indirect inhibition of γ-secretase activity. Here we report that Gleevec also achieves its Aβ-lowering effects through an additional cellular mechanism. It renders APP less susceptible to proteolysis by BACE without inhibiting BACE enzymatic activity or the processing of other BACE substrates. This effect closely mimics the phenotype of APP A673T, a recently discovered mutation that protects carriers against AD and age-related cognitive decline. In addition, Gleevec induces formation of a specific set of APP C-terminal fragments, also observed in cells expressing the APP protective mutation and in cells exposed to a conventional BACE inhibitor. These Gleevec phenotypes require an intracellular acidic pH and are independent of tyrosine kinase inhibition, given that a related compound lacking tyrosine kinase inhibitory activity, DV2-103, exerts similar effects on APP metabolism. In addition, DV2-103 accumulates at high concentrations in the rodent brain, where it rapidly lowers Aβ levels. This study suggests that long-term treatment with drugs that indirectly modulate BACE processing of APP but spare other BACE substrates and achieve therapeutic concentrations in the brain might be effective in preventing or delaying the onset of AD and could be safer than nonselective BACE inhibitor drugs.
Collapse
|
16
|
Becker-Pauly C, Pietrzik CU. The Metalloprotease Meprin β Is an Alternative β-Secretase of APP. Front Mol Neurosci 2017; 9:159. [PMID: 28105004 PMCID: PMC5215381 DOI: 10.3389/fnmol.2016.00159] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/09/2016] [Indexed: 01/08/2023] Open
Abstract
The membrane bound metalloprotease meprin β is important for collagen fibril assembly in connective tissue formation and for the detachment of the intestinal mucus layer for proper barrier function. Recent proteomic studies revealed dozens of putative new substrates of meprin β, including the amyloid precursor protein (APP). It was shown that APP is cleaved by meprin β in distinct ways, either at the β-secretase site resulting in increased levels of Aβ peptides, or at the N-terminus releasing 11 kDa, and 20 kDa peptide fragments. The latter event was discussed to be rather neuroprotective, whereas the ectodomain shedding of APP by meprin β reminiscent to BACE-1 is in line with the amyloid hypothesis of Alzheimer's disease, promoting neurodegeneration. The N-terminal 11 kDa and 20 kDa peptide fragments represent physiological cleavage products, since they are found in human brains under different diseased or non-diseased states, whereas these fragments are completely missing in brains of meprin β knock-out animals. Meprin β is not only a sheddase of adhesion molecules, such as APP, but was additionally demonstrated to cleave within the prodomain of ADAM10. Activated ADAM10, the α-secretase of APP, is then able to shed meprin β from the cell surface thereby abolishing the β-secretase activity. All together meprin β seems to be a novel player in APP processing events, even influencing other enzymes involved in APP cleavage.
Collapse
Affiliation(s)
- Christoph Becker-Pauly
- Unit for Degradomics of the Protease Web, Institute of Biochemistry, University of Kiel Kiel, Germany
| | - Claus U Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz Mainz, Germany
| |
Collapse
|
17
|
Schröder B, Saftig P. Intramembrane proteolysis within lysosomes. Ageing Res Rev 2016; 32:51-64. [PMID: 27143694 DOI: 10.1016/j.arr.2016.04.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/01/2016] [Accepted: 04/26/2016] [Indexed: 11/26/2022]
Abstract
Regulated intramembrane proteolysis is of pivotal importance in a diverse set of developmental and physiological processes. Altered intramembrane substrate turnover may be associated with neurodegeneration, cancer and impaired immune function. In this review we will focus on the intramembrane proteases which have been localized in the lysosomal membrane. Members of the γ-secretase complex and γ-secretase activity are found in the lysosomal membrane and are discussed to contribute to intracellular amyloid β production. Mutant or deficient γ-secretase may cause disturbed lysosomal function. The signal peptide peptidase-like (SPPL) protease 2a is a lysosomal membrane component and cleaves CD74, the invariant chain of the MHC II complex, as well as FasL, TNF, ITM2B and TMEM106, type II transmembrane proteins involved in the regulation of immunity and neurodegeneration. Therefore, it can be concluded, that not only proteolysis within the lysosomal lumen but also within lysosomal membranes regulates important cellular functions and contributes essentially to proteostasis of membrane proteins what may become increasingly compromised in the aged individual.
Collapse
|
18
|
Tam JHK, Cobb MR, Seah C, Pasternak SH. Tyrosine Binding Protein Sites Regulate the Intracellular Trafficking and Processing of Amyloid Precursor Protein through a Novel Lysosome-Directed Pathway. PLoS One 2016; 11:e0161445. [PMID: 27776132 PMCID: PMC5077117 DOI: 10.1371/journal.pone.0161445] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 08/07/2016] [Indexed: 01/18/2023] Open
Abstract
The amyloid hypothesis posits that the production of β-amyloid (Aβ) aggregates leads to neurodegeneration and cognitive decline associated with AD. Aβ is produced by sequential cleavage of the amyloid precursor protein (APP) by β- and γ-secretase. While nascent APP is well known to transit to the endosomal/ lysosomal system via the cell surface, we have recently shown that APP can also traffic to lysosomes intracellularly via its interaction with AP-3. Because AP-3 interacts with cargo protein via interaction with tyrosine motifs, we mutated the three tyrosines motif in the cytoplasmic tail of APP. Here, we show that the YTSI motif interacts with AP-3, and phosphorylation of the serine in this motif disrupts the interaction and decreases APP trafficking to lysosomes. Furthermore, we show that phosphorylation at this motif can decrease the production of neurotoxic Aβ 42. This demonstrates that reducing APP trafficking to lysosomes may be a strategy to reduce Aβ 42 in Alzheimer’s disease.
Collapse
Affiliation(s)
- Joshua H. K. Tam
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, Western University, London Ontario, Canada, N6A 5B7
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada, N6A 5B7
| | - M. Rebecca Cobb
- Program in Neuroscience, Western University, London, Ontario, Canada, N6A 5B7
| | - Claudia Seah
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, Western University, London Ontario, Canada, N6A 5B7
| | - Stephen H. Pasternak
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, Western University, London Ontario, Canada, N6A 5B7
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada, N6A 5B7
- Program in Neuroscience, Western University, London, Ontario, Canada, N6A 5B7
- Department of Clinical Neurological Sciences, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada, N6A 5B7
- * E-mail:
| |
Collapse
|
19
|
Decreased amyloid-β and increased neuronal hyperactivity by immunotherapy in Alzheimer's models. Nat Neurosci 2015; 18:1725-7. [PMID: 26551546 DOI: 10.1038/nn.4163] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/14/2015] [Indexed: 12/14/2022]
Abstract
Among the most promising approaches for treating Alzheimer's disease is immunotherapy with amyloid-β (Aβ)-targeting antibodies. Using in vivo two-photon imaging in mouse models, we found that two different antibodies to Aβ used for treatment were ineffective at repairing neuronal dysfunction and caused an increase in cortical hyperactivity. This unexpected finding provides a possible cellular explanation for the lack of cognitive improvement by immunotherapy in human studies.
Collapse
|
20
|
Chapman G, Major JA, Iyer K, James AC, Pursglove SE, Moreau JLM, Dunwoodie SL. Notch1 endocytosis is induced by ligand and is required for signal transduction. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:166-77. [PMID: 26522918 DOI: 10.1016/j.bbamcr.2015.10.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/27/2015] [Accepted: 10/28/2015] [Indexed: 01/20/2023]
Abstract
The Notch signalling pathway is widely utilised during embryogenesis in situations where cell-cell interactions are important for cell fate specification and differentiation. DSL ligand endocytosis into the ligand-expressing cell is an important aspect of Notch signalling because it is thought to supply the force needed to separate the Notch heterodimer to initiate signal transduction. A functional role for receptor endocytosis during Notch signal transduction is more controversial. Here we have used live-cell imaging to examine trafficking of the Notch1 receptor in response to ligand binding. Contact with cells expressing ligands induced internalisation and intracellular trafficking of Notch1. Notch1 endocytosis was accompanied by transendocytosis of ligand into the Notch1-expressing signal-receiving cell. Ligand caused Notch1 endocytosis into SARA-positive endosomes in a manner dependent on clathrin and dynamin function. Moreover, inhibition of endocytosis in the receptor-expressing cell impaired ligand-induced Notch1 signalling. Our findings resolve conflicting observations from mammalian and Drosophila studies by demonstrating that ligand-dependent activation of Notch1 signalling requires receptor endocytosis. Endocytosis of Notch1 may provide a force on the ligand:receptor complex that is important for potent signal transduction.
Collapse
Affiliation(s)
- G Chapman
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, Australia.
| | - J A Major
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - K Iyer
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - A C James
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - S E Pursglove
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - J L M Moreau
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - S L Dunwoodie
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, Australia; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
21
|
Tam JHK, Pasternak SH. Imaging the Intracellular Trafficking of APP with Photoactivatable GFP. J Vis Exp 2015:e53153. [PMID: 26555118 DOI: 10.3791/53153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Beta-amyloid (Aβ) is the major constituent of senile plaques found in the brains of Alzheimer's disease patients. Aβ is derived from the sequential cleavage of Amyloid Precursor Protein (APP) by β and γ-secretases. Despite the importance of Aβ to AD pathology, the subcellular localization of these cleavages is not well established. Work in our laboratory and others implicate the endosomal/lysosomal system in APP processing after internalization from the cell surface. However, the intracellular trafficking of APP is relatively understudied. While cell-surface proteins are amendable to many labeling techniques, there are no simple methods for following the trafficking of membrane proteins from the Golgi. To this end, we created APP constructs that were tagged with photo-activatable GFP (paGFP) at the C-terminus. After synthesis, paGFP has low basal fluorescence, but it can be stimulated with 413 nm light to produce a strong, stable green fluorescence. By using the Golgi marker Galactosyl transferase coupled to Cyan Fluorescent Protein (GalT-CFP) as a target, we are able to accurately photoactivate APP in the trans-Golgi network. Photo-activated APP-paGFP can then be followed as it traffics to downstream compartments identified with fluorescently tagged compartment marker proteins for the early endosome (Rab5), the late endosome (Rab9) and the lysosome (LAMP1). Furthermore, using inhibitors to APP processing including chloroquine or the γ-secretase inhibitor L685, 458, we are able to perform pulse-chase experiments to examine the processing of APP in single cells. We find that a large fraction of APP moves rapidly to the lysosome without appearing at the cell surface, and is then cleared from the lysosome by secretase-like cleavages. This technique demonstrates the utility of paGFP for following the trafficking and processing of intracellular proteins from the Golgi to downstream compartments.
Collapse
Affiliation(s)
- Joshua H K Tam
- Department of Physiology and Pharmacology, Robarts Research Institute, Western University
| | - Stephen H Pasternak
- Department of Physiology and Pharmacology, Robarts Research Institute, Western University; Department of Clinical Neurological Sciences, Western University;
| |
Collapse
|
22
|
Tang W, Tam JHK, Seah C, Chiu J, Tyrer A, Cregan SP, Meakin SO, Pasternak SH. Arf6 controls beta-amyloid production by regulating macropinocytosis of the Amyloid Precursor Protein to lysosomes. Mol Brain 2015; 8:41. [PMID: 26170135 PMCID: PMC4501290 DOI: 10.1186/s13041-015-0129-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 06/11/2015] [Indexed: 11/15/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by the deposition of Beta-Amyloid (Aβ) peptides in the brain. Aβ peptides are generated by cleavage of the Amyloid Precursor Protein (APP) by the β − and γ − secretase enzymes. Although this process is tightly linked to the internalization of cell surface APP, the compartments responsible are not well defined. We have found that APP can be rapidly internalized from the cell surface to lysosomes, bypassing early and late endosomes. Here we show by confocal microscopy and electron microscopy that this pathway is mediated by macropinocytosis. APP internalization is enhanced by antibody binding/crosslinking of APP suggesting that APP may function as a receptor. Furthermore, a dominant negative mutant of Arf6 blocks direct transport of APP to lysosomes, but does not affect classical endocytosis to endosomes. Arf6 expression increases through the hippocampus with the development of Alzheimer’s disease, being expressed mostly in the CA1 and CA2 regions in normal individuals but spreading through the CA3 and CA4 regions in individuals with pathologically diagnosed AD. Disruption of lysosomal transport of APP reduces both Aβ40 and Aβ42 production by more than 30 %. Our findings suggest that the lysosome is an important site for Aβ production and that altering APP trafficking represents a viable strategy to reduce Aβ production.
Collapse
Affiliation(s)
- Weihao Tang
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| | - Joshua H K Tam
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| | - Claudia Seah
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada.
| | - Justin Chiu
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| | - Andrea Tyrer
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| | - Sean P Cregan
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| | - Susan O Meakin
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Biochemistry, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| | - Stephen H Pasternak
- J. Allyn Taylor Centre for Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, 1151 Richmond St, London, ON, N6A 5B8, Canada. .,Department of Clinical Neurological Sciences, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada. .,Department of Physiology and Pharmacology, Schulich School of Medicine, the University of Western Ontario, London, ON, N6A 5B7, Canada.
| |
Collapse
|
23
|
Prasad H, Rao R. The Na+/H+ exchanger NHE6 modulates endosomal pH to control processing of amyloid precursor protein in a cell culture model of Alzheimer disease. J Biol Chem 2015; 290:5311-27. [PMID: 25561733 DOI: 10.1074/jbc.m114.602219] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Early intervention may be key to safe and effective therapies in patients with Alzheimer disease. Endosomal dysfunction is an early step in neurodegeneration. Endosomes are a major site of production of Aβ peptide from the processing of amyloid precursor protein (APP) by clipping enzymes (β- and γ-secretases). The β-secretase enzyme BACE1 requires acidic lumen pH for optimum function, and acid pH promotes Aβ aggregation. The Na(+)/H(+) exchanger NHE6 provides a leak pathway for protons, limiting luminal acidification by proton pumps. Like APP, NHE6 expression was induced upon differentiation of SH-SY5Y neuroblastoma cells and localized to an endosomal compartment. Therefore, we investigated whether NHE6 expression altered APP localization and processing in a stably transfected cell culture model of human APP expression. We show that co-expression with NHE6 or treatment with the Na(+)/H(+) ionophore monensin shifted APP away from the trans-Golgi network into early and recycling endosomes in HEK293 cells. NHE6 alkalinized the endosomal lumen, similar to monensin, and significantly attenuated APP processing and Aβ secretion. In contrast, Aβ production was elevated upon NHE6 knockdown. We show that NHE6 transcript and protein levels are lowered in Alzheimer brains relative to control. These findings, taken together with emerging genetic evidence linking endosomal Na(+)/H(+) exchangers with Alzheimer disease, suggest that proton leak pathways may regulate Aβ generation and contribute to disease etiology.
Collapse
Affiliation(s)
- Hari Prasad
- From the Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Rajini Rao
- From the Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
24
|
Tam JHK, Seah C, Pasternak SH. The Amyloid Precursor Protein is rapidly transported from the Golgi apparatus to the lysosome and where it is processed into beta-amyloid. Mol Brain 2014; 7:54. [PMID: 25085554 PMCID: PMC4237969 DOI: 10.1186/s13041-014-0054-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/23/2014] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by cerebral deposition of β-amyloid peptide (Aβ). Aβ is produced by sequential cleavage of the Amyloid Precursor Protein (APP) by β- and γ-secretases. Many studies have demonstrated that the internalization of APP from the cell surface can regulate Aβ production, although the exact organelle in which Aβ is produced remains contentious. A number of recent studies suggest that intracellular trafficking also plays a role in regulating Aβ production, but these pathways are relatively under-studied. The goal of this study was to elucidate the intracellular trafficking of APP, and to examine the site of intracellular APP processing. RESULTS We have tagged APP on its C-terminal cytoplasmic tail with photoactivatable Green Fluorescent Protein (paGFP). By photoactivating APP-paGFP in the Golgi, using the Golgi marker Galactosyltranferase fused to Cyan Fluorescent Protein (GalT-CFP) as a target, we are able to follow a population of nascent APP molecules from the Golgi to downstream compartments identified with compartment markers tagged with red fluorescent protein (mRFP or mCherry); including rab5 (early endosomes) rab9 (late endosomes) and LAMP1 (lysosomes). Because γ-cleavage of APP releases the cytoplasmic tail of APP including the photoactivated GFP, resulting in loss of fluorescence, we are able to visualize the cleavage of APP in these compartments. Using APP-paGFP, we show that APP is rapidly trafficked from the Golgi apparatus to the lysosome; where it is rapidly cleared. Chloroquine and the highly selective γ-secretase inhibitor, L685, 458, cause the accumulation of APP in lysosomes implying that APP is being cleaved by secretases in the lysosome. The Swedish mutation dramatically increases the rate of lysosomal APP processing, which is also inhibited by chloroquine and L685, 458. By knocking down adaptor protein 3 (AP-3; a heterotetrameric protein complex required for trafficking many proteins to the lysosome) using siRNA, we are able to reduce this lysosomal transport. Blocking lysosomal transport of APP reduces Aβ production by more than a third. CONCLUSION These data suggests that AP-3 mediates rapid delivery of APP to lysosomes, and that the lysosome is a likely site of Aβ production.
Collapse
Affiliation(s)
- Joshua HK Tam
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, 100 Perth Drive, London N6A 5K8, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London N6A 5K8, Ontario, Canada
| | - Claudia Seah
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, 100 Perth Drive, London N6A 5K8, Ontario, Canada
| | - Stephen H Pasternak
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, 100 Perth Drive, London N6A 5K8, Ontario, Canada
- Department of Clinical Neurological Sciences, London N6A 5K8, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London N6A 5K8, Ontario, Canada
| |
Collapse
|
25
|
Abstract
Dementias are a varied group of disorders typically associated with memory loss, impaired judgment and/or language and by symptoms affecting other cognitive and social abilities to a degree that interferes with daily functioning. Alzheimer's disease (AD) is the most common cause of a progressive dementia, followed by dementia with Lewy bodies (DLB), frontotemporal dementia (FTD), (VaD) and HIV-associated neurocognitive disorders (HAND). The pathogenesis of this group of disorders has been linked to the abnormal accumulation of proteins in the brains of affected individuals, which in turn has been related to deficits in protein clearance. Autophagy is a key cellular protein clearance pathway with proteolytic cleavage and degradation via the ubiquitin-proteasome pathway representing another important clearance mechanism. Alterations in the levels of autophagy and the proteins associated with the autophagocytic pathway have been reported in various types of dementias. This review will examine recent literature across these disorders and highlight a common theme of altered autophagy across the spectrum of the dementias.
Collapse
|
26
|
Dopamine induces apoptosis in APPswe-expressing Neuro2A cells following Pepstatin-sensitive proteolysis of APP in acid compartments. Brain Res 2012; 1471:102-17. [DOI: 10.1016/j.brainres.2012.06.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 06/14/2012] [Accepted: 06/21/2012] [Indexed: 11/23/2022]
|
27
|
Nuclear signalling by membrane protein intracellular domains: the AICD enigma. Cell Signal 2011; 24:402-409. [PMID: 22024280 DOI: 10.1016/j.cellsig.2011.10.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 10/10/2011] [Indexed: 01/13/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative illness and the leading cause of dementia in the elderly. The accumulation of amyloid-β peptide (Aβ) is a well-known pathological hallmark associated with the disease. However, Aβ is only one of several metabolites produced by β- and γ-secretase actions on the transmembrane protein, the amyloid precursor protein (APP). A proteolytic fragment termed the APP intracellular domain (AICD) is also produced. By analogy with the Notch signalling pathway, AICD has been proposed as a transcriptional regulator although its mechanism of action and the complement of genes regulated remain controversial. This review will focus on the contributions that studies of APP processing have brought to the understanding of a novel nuclear signalling pathway that may contribute to the pathology of AD and may provide new therapeutic opportunities.
Collapse
|
28
|
The second-generation active Aβ immunotherapy CAD106 reduces amyloid accumulation in APP transgenic mice while minimizing potential side effects. J Neurosci 2011; 31:9323-31. [PMID: 21697382 DOI: 10.1523/jneurosci.0293-11.2011] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Immunization against amyloid-β (Aβ) can reduce amyloid accumulation in vivo and is considered a potential therapeutic approach for Alzheimer's disease. However, it has been associated with meningoencephalitis thought to be mediated by inflammatory T-cells. With the aim of producing an immunogenic vaccine without this side effect, we designed CAD106 comprising Aβ1-6 coupled to the virus-like particle Qβ. Immunization with this vaccine did not activate Aβ-specific T-cells. In APP transgenic mice, CAD106 induced efficacious Aβ antibody titers of different IgG subclasses mainly recognizing the Aβ3-6 epitope. CAD106 reduced brain amyloid accumulation in two APP transgenic mouse lines. Plaque number was a more sensitive readout than plaque area, followed by Aβ42 and Aβ40 levels. Studies with very strong overall amyloid reduction showed an increase in vascular Aβ, which atypically was nonfibrillar. The efficacy of Aβ immunotherapy depended on the Aβ levels and thus differed between animal models, brain regions, and stage of amyloid deposition. Therefore, animal studies may not quantitatively predict the effect in human Alzheimer's disease. Our studies provided no evidence for increased microhemorrhages or inflammatory reactions in amyloid-containing brain. In rhesus monkeys, CAD106 induced a similar antibody response as in mice. The antibodies stained amyloid deposits on tissue sections of mouse and human brain but did not label cellular structures containing APP. They reacted with Aβ monomers and oligomers and blocked Aβ toxicity in cell culture. We conclude that CAD106 immunization is suited to interfere with Aβ aggregation and its downstream detrimental effects.
Collapse
|
29
|
Rabe S, Reichwald J, Ammaturo D, de Strooper B, Saftig P, Neumann U, Staufenbiel M. The Swedish APP mutation alters the effect of genetically reduced BACE1 expression on the APP processing. J Neurochem 2011; 119:231-9. [DOI: 10.1111/j.1471-4159.2011.07412.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
30
|
Critical role of astroglial apolipoprotein E and liver X receptor-α expression for microglial Aβ phagocytosis. J Neurosci 2011; 31:7049-59. [PMID: 21562267 DOI: 10.1523/jneurosci.6546-10.2011] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Liver X receptors (LXRs) regulate immune cell function and cholesterol metabolism, both factors that are critically involved in Alzheimer's disease (AD). To investigate the therapeutic potential of long-term LXR activation in amyloid-β (Aβ) peptide deposition in an AD model, 13-month-old, amyloid plaque-bearing APP23 mice were treated with the LXR agonist TO901317. Postmortem analysis demonstrated that TO901317 efficiently crossed the blood-brain barrier. Insoluble and soluble Aβ levels in the treated APP23 mice were reduced by 80% and 40%, respectively, compared with untreated animals. Amyloid precursor protein (APP) processing, however, was hardly changed by the compound, suggesting that the observed effects were instead mediated by Aβ disposal. Despite the profound effect on Aβ levels, spatial learning in the Morris water maze was only slightly improved by the treatment. ABCA1 (ATP-binding cassette transporter 1) and apolipoprotein E (ApoE) protein levels were increased and found to be primarily localized in astrocytes. Experiments using primary microglia demonstrated that medium derived from primary astrocytes exposed to TO901317 stimulated phagocytosis of fibrillar Aβ. Conditioned medium from TO901317-treated ApoE(-/-) or LXRα(-/-) astrocytes did not increase phagocytosis of Aβ. In APP23 mice, long-term treatment with TO901317 strongly increased the association of microglia and Aβ plaques. Short-term treatment of APP/PS1 mice with TO901317 also increased this association, which was dependent on the presence of LXRα and was accompanied by increased ApoE lipidation. Together, these data suggest that astrocytic LXRα activation and subsequent release of ApoE by astrocytes is critical for the ability of microglia to remove fibrillar Aβ in response to treatment with TO901317.
Collapse
|
31
|
Kipfer-Kauer A, McKinnon SJ, Frueh BE, Goldblum D. Distribution of amyloid precursor protein and amyloid-beta in ocular hypertensive C57BL/6 mouse eyes. Curr Eye Res 2010; 35:828-34. [PMID: 20795865 DOI: 10.3109/02713683.2010.494240] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE Amyloid precursor protein (APP) and amyloid-beta (Abeta) appear to participate in the pathophysiology of retinal ganglion cell (RGC) death in glaucoma. We, therefore, determined the distribution of APP and Abeta in the retinas of C57BL/6 mice after induction of chronic ocular hypertension. METHODS Ocular hypertension was induced in one eye of three-month-old C57BL/6 mice by injection of hypertonic saline into episcleral veins. After 6 weeks of documented elevated intraocular pressure (IOP), retinas were fixed with 4% paraformaldehyde and processed for immunohistochemistry with antibodies including a polyclonal antibody to the C-terminus of Abeta 40 (Novartis 17-40/23) and a polyclonal antibody to the APP ectodomain (Novartis 474). Distribution and semiquantitative expression of APP and Abeta immunolabeling in ocular hypertensive and control retinas were graded in a masked fashion and compared. RESULTS APP and Abeta immunoreactivity was found in the pia/dura, optic nerve (ON), and RGC layer of ocular hypertensive retinas, whereas APP and Abeta immunoreactivity in the contralateral control eyes was detected only in the pia/dura. Comparison of ocular hypertensive and control eyes for Abeta immunolabeling was significant in the ON and RGC layer (p < 0.05) whereas no significant difference was found when compared for APP staining. CONCLUSIONS High Abeta and APP levels were seen in ocular hypertensive retinas, probably due to abnormal APP-splicing in the presence of elevated IOP.
Collapse
|
32
|
Winkler DT, Abramowski D, Danner S, Zurini M, Paganetti P, Tolnay M, Staufenbiel M. Rapid cerebral amyloid binding by Aβ antibodies infused into β-amyloid precursor protein transgenic mice. Biol Psychiatry 2010; 68:971-4. [PMID: 20359696 DOI: 10.1016/j.biopsych.2010.01.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Revised: 01/29/2010] [Accepted: 01/30/2010] [Indexed: 11/19/2022]
Abstract
BACKGROUND Passive immunization for the treatment of Alzheimer's disease (AD) was rapidly translated into clinical trials. However, basic mechanisms of AD immunotherapy remain only partially understood. METHODS We analyzed the dynamic changes of amyloid-β (Aβ) levels in plasma, brain, and cerebrospinal fluid (CSF) as well as cerebral amyloid binding by Aβ antibody after a single β1-antibody infusion into APP(Swedish) and APP(wildtype) transgenic mice at preplaque and plaque-bearing age. RESULTS Following intravenous Aβ antibody treatment, plasma Aβ increased rapidly, reaching significantly higher levels in preplaque compared with plaque-bearing mice, whereas cerebral and CSF Aβ remained unchanged. Strikingly, Aβ antibodies exhibited strong cerebral amyloid plaque binding rapidly after intravenous administration in a subset of animals with more severe vascular amyloid. CONCLUSIONS Rapid plasma Aβ increase after Aβ antibody infusion results primarily from stabilization of Aβ. Nevertheless, the smaller plasma Aβ increase in plaque-bearing mice might be of diagnostic use. Importantly, intravenously administered antibodies can rapidly bind to cerebral plaques, potentially facilitated by vascular-amyloid-mediated damage of the blood-brain barrier.
Collapse
Affiliation(s)
- David T Winkler
- Department of Neurology, University Hospital Basel, Basel, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
33
|
Bepridil and amiodarone simultaneously target the Alzheimer's disease beta- and gamma-secretase via distinct mechanisms. J Neurosci 2010; 30:8974-83. [PMID: 20592218 DOI: 10.1523/jneurosci.1199-10.2010] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The two proteases beta-secretase and gamma-secretase generate the amyloid beta peptide and are drug targets for Alzheimer's disease. Here we tested the possibility of targeting the cellular environment of beta-secretase cleavage instead of the beta-secretase enzyme itself. beta-Secretase has an acidic pH optimum and cleaves the amyloid precursor protein in the acidic endosomes. We identified two drugs, bepridil and amiodarone, that are weak bases and are in clinical use as calcium antagonists. Independently of their calcium-blocking activity, both compounds mildly raised the membrane-proximal, endosomal pH and inhibited beta-secretase cleavage at therapeutically achievable concentrations in cultured cells, in primary neurons, and in vivo in guinea pigs. This shows that an alkalinization of the cellular environment could be a novel therapeutic strategy to inhibit beta-secretase. Surprisingly, bepridil and amiodarone also modulated gamma-secretase cleavage independently of endosomal alkalinization. Thus, both compounds act as dual modulators that simultaneously target beta- and gamma-secretase through distinct molecular mechanisms. In addition to Alzheimer's disease, compounds with dual properties may also be useful for drug development targeting other membrane proteins.
Collapse
|
34
|
Lorenzen A, Samosh J, Vandewark K, Anborgh PH, Seah C, Magalhaes AC, Cregan SP, Ferguson SSG, Pasternak SH. Rapid and direct transport of cell surface APP to the lysosome defines a novel selective pathway. Mol Brain 2010; 3:11. [PMID: 20409323 PMCID: PMC2868040 DOI: 10.1186/1756-6606-3-11] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Accepted: 04/21/2010] [Indexed: 12/03/2022] Open
Abstract
Background A central feature of Alzheimer's disease is the cleavage of the amyloid precursor protein (APP) to form beta-amyloid peptide (Aβ) by the β-secretase and γ-secretase enzymes. Although this has been shown to occur after endocytosis of APP from the cell surface, the exact compartments of APP processing are not well defined. We have previously demonstrated that APP and γ-secretase proteins and activity are highly enriched in purified rat liver lysosomes. In order to examine the lysosomal distribution and trafficking of APP in cultured cells, we generated constructs containing APP fused to a C-terminal fluorescent protein tag and N-terminal HA-epitope tag. These were co-transfected with a panel of fluorescent-protein tagged compartment markers. Results Here we demonstrate using laser-scanning confocal microscopy that although APP is present throughout the endosomal/lysosomal system in transfected Cos7 and neuronal SN56 cell lines as well as in immunostained cultured mouse neurons, it is enriched in the lysosome. We also show that the Swedish and London mutations reduce the amount of APP in the lysosome. Surprisingly, in addition to its expected trafficking from the cell surface to the early and then late endosomes, we find that cell-surface labelled APP is transported rapidly and directly from the cell surface to lysosomes in both Cos7 and SN56 cells. This rapid transit to the lysosome is blocked by the presence of either the London or Swedish mutations. Conclusions These results demonstrate the presence of a novel, rapid and specific transport pathway from the cell surface to the lysosomes. This suggests that regulation of lysosomal traffic could regulate APP processing and that the lysosome could play a central role in the pathophysiology of Alzheimer's disease.
Collapse
Affiliation(s)
- Angela Lorenzen
- J, Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, Schulich School of Medicine, the University of Western Ontario, London, Ontario, N6A 5K8, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yan Y, Denef N, Schüpbach T. The vacuolar proton pump, V-ATPase, is required for notch signaling and endosomal trafficking in Drosophila. Dev Cell 2009; 17:387-402. [PMID: 19758563 DOI: 10.1016/j.devcel.2009.07.001] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 05/31/2009] [Accepted: 07/01/2009] [Indexed: 11/17/2022]
Abstract
We have identified Rabconnectin-3alpha and beta (Rbcn-3A and B) as two regulators of Notch signaling in Drosophila. We found that, in addition to disrupting Notch signaling, mutations in Rbcn-3A and B cause defects in endocytic trafficking, where Notch and other membrane proteins accumulate in late endosomal compartments. We show that Notch is transported to the surface of mutant cells and that signaling is disrupted after the S2 cleavage. Interestingly, the yeast homolog of Rbcn-3A, Rav1, regulates the V-ATPase proton pump responsible for acidifying intracellular organelles. We found that, similarly, Rbcn-3A and B appear to regulate V-ATPase function. Moreover, we identified mutants in VhaAC39, a V-ATPase subunit, and showed that they phenocopy Rbcn-3A and Rbcn-3B mutants. Our results demonstrate that Rbcn-3 affects Notch signaling and trafficking through regulating V-ATPase function, which implies that the acidification of an intracellular compartment in the receiving cells is crucial for signaling.
Collapse
Affiliation(s)
- Yan Yan
- Howard Hughes Medical Institute, Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | | | | |
Collapse
|
36
|
Increased exploratory activity of APP23 mice in a novel environment is reversed by siRNA. Brain Res 2008; 1243:124-33. [DOI: 10.1016/j.brainres.2008.09.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Revised: 09/06/2008] [Accepted: 09/11/2008] [Indexed: 12/11/2022]
|
37
|
Abramowski D, Wiederhold KH, Furrer U, Jaton AL, Neuenschwander A, Runser MJ, Danner S, Reichwald J, Ammaturo D, Staab D, Stoeckli M, Rueeger H, Neumann U, Staufenbiel M. Dynamics of Abeta turnover and deposition in different beta-amyloid precursor protein transgenic mouse models following gamma-secretase inhibition. J Pharmacol Exp Ther 2008; 327:411-24. [PMID: 18687920 DOI: 10.1124/jpet.108.140327] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Human beta-amyloid precursor protein (APP) transgenic mice are commonly used to test potential therapeutics for Alzheimer's disease. We have characterized the dynamics of beta-amyloid (Abeta) generation and deposition following gamma-secretase inhibition with compound LY-411575 [N(2)-[(2S)-2-(3,5-difluorophenyl)-2-hydroxyethanoyl]-N(1)-[(7S)-5-methyl-6-oxo-6,7-dihydro-5H-dibenzo[b,d]azepin-7-yl]-L-alaninamide]. Kinetic studies in preplaque mice distinguished a detergent-soluble Abeta pool in brain with rapid turnover (half-lives for Abeta40 and Abeta42 were 0.7 and 1.7 h) and a much more stable, less soluble pool. Abeta in cerebrospinal fluid (CSF) reflected the changes in the soluble brain Abeta pool, whereas plasma Abeta turned over more rapidly. In brain, APP C-terminal fragments (CTF) accumulated differentially. The half-lives for gamma-secretase degradation were estimated as 0.4 and 0.1 h for C99 and C83, respectively. Three different APP transgenic lines responded very similarly to gamma-secretase inhibition regardless of the familial Alzheimer's disease mutations in APP. Amyloid deposition started with Abeta42, whereas Abeta38 and Abeta40 continued to turn over. Chronic gamma-secretase inhibition lowered amyloid plaque formation to a different degree in different brain regions of the same mice. The extent was inversely related to the initial amyloid load in the region analyzed. No evidence for plaque removal below baseline was obtained. gamma-Secretase inhibition led to a redistribution of intracellular Abeta and an elevation of CTFs in neuronal fibers. In CSF, Abeta showed a similar turnover as in preplaque animals demonstrating its suitability as marker of newly generated, soluble Abeta in plaque-bearing brain. This study supports the use of APP transgenic mice as translational models to characterize Abeta-lowering therapeutics.
Collapse
|
38
|
Vingtdeux V, Hamdane M, Loyens A, Gelé P, Drobeck H, Bégard S, Galas MC, Delacourte A, Beauvillain JC, Buée L, Sergeant N. Alkalizing drugs induce accumulation of amyloid precursor protein by-products in luminal vesicles of multivesicular bodies. J Biol Chem 2007; 282:18197-18205. [PMID: 17468104 DOI: 10.1074/jbc.m609475200] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Amyloid precursor protein (APP) metabolism is central to the pathogenesis of Alzheimer disease. We showed recently that the amyloid intracellular domain (AICD), which is released by gamma-secretase cleavage of APP C-terminal fragments (CTFs), is strongly increased in cells treated with alkalizing drugs (Vingtdeux, V., Hamdane, M., Bégard, S., Loyens, A., Delacourte, A., Beauvillain, J.-C., Buée, L., Marambaud, P., and Sergeant, N. (2007) Neurobiol. Dis. 25, 686-696). Herein, we aimed to determine the cell compartment in which AICD accumulates. We show that APP-CTFs and AICD are present in multivesicular structures. Multivesicular bodies contain intraluminal vesicles (known as exosomes) when released in the extracellular space. We demonstrate that APP, APP-CTFs, and AICD are integrated and secreted within exosomes in differentiated neuroblastoma and primary neuronal culture cells. Together with recent data showing that amyloid-beta is also found in exosomes, our data show that multivesicular bodies are essential organelles for APP metabolism and that all APP metabolites can be secreted in the extracellular space.
Collapse
Affiliation(s)
- Valérie Vingtdeux
- INSERM, U837, Neurodegenerative Disorders and Neuronal Death, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France; Facultéde Médecine, Institut de Médecine Prédictive et de Recherche Thérapeutique, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France
| | - Malika Hamdane
- INSERM, U837, Neurodegenerative Disorders and Neuronal Death, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France; Facultéde Médecine, Institut de Médecine Prédictive et de Recherche Thérapeutique, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France
| | - Anne Loyens
- INSERM, U837, Neurodegenerative Disorders and Neuronal Death, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France; Facultéde Médecine, Institut de Médecine Prédictive et de Recherche Thérapeutique, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France
| | - Patrick Gelé
- INSERM, U837, Neurodegenerative Disorders and Neuronal Death, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France; Facultéde Médecine, Institut de Médecine Prédictive et de Recherche Thérapeutique, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France
| | - Hervé Drobeck
- CNRS, UMR 8161, "Lille Institute of Biology," University of Lille 1, Pasteur Institute of Lille, 1, rue du Professeur Calmette, F-59021 Lille Cedex, France
| | - Séverine Bégard
- INSERM, U837, Neurodegenerative Disorders and Neuronal Death, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France; Facultéde Médecine, Institut de Médecine Prédictive et de Recherche Thérapeutique, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France
| | - Marie-Christine Galas
- INSERM, U837, Neurodegenerative Disorders and Neuronal Death, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France; Facultéde Médecine, Institut de Médecine Prédictive et de Recherche Thérapeutique, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France
| | - André Delacourte
- INSERM, U837, Neurodegenerative Disorders and Neuronal Death, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France; Facultéde Médecine, Institut de Médecine Prédictive et de Recherche Thérapeutique, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France
| | - Jean-Claude Beauvillain
- INSERM, U837, Neurodegenerative Disorders and Neuronal Death, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France; Facultéde Médecine, Institut de Médecine Prédictive et de Recherche Thérapeutique, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France
| | - Luc Buée
- INSERM, U837, Neurodegenerative Disorders and Neuronal Death, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France; Facultéde Médecine, Institut de Médecine Prédictive et de Recherche Thérapeutique, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France
| | - Nicolas Sergeant
- INSERM, U837, Neurodegenerative Disorders and Neuronal Death, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France; Facultéde Médecine, Institut de Médecine Prédictive et de Recherche Thérapeutique, Centre de Recherches Jean-Pierre Aubert, Université Lille 2, Place de Verdun, F-59045 Lille, France.
| |
Collapse
|
39
|
Vingtdeux V, Hamdane M, Bégard S, Loyens A, Delacourte A, Beauvillain JC, Buée L, Marambaud P, Sergeant N. Intracellular pH regulates amyloid precursor protein intracellular domain accumulation. Neurobiol Dis 2007; 25:686-96. [PMID: 17207630 DOI: 10.1016/j.nbd.2006.09.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2006] [Revised: 07/26/2006] [Accepted: 09/19/2006] [Indexed: 01/01/2023] Open
Abstract
The amyloid precursor protein (APP) metabolism is central to pathogenesis of Alzheimer's disease (AD). Parenchymal amyloid deposits, a neuropathological hallmark of AD, are composed of amyloid-beta peptides (Abeta). Abeta derives from the amyloid precursor protein (APP) by sequential cleavages by beta- and gamma-secretases. Gamma-secretase cleavage releases the APP intracellular domain (AICD), suggested to mediate a nuclear signaling. Physiologically, AICD is seldom detected and thus supposed to be rapidly degraded. The mechanisms responsible of its degradation remain unknown. We used a pharmacological approach and showed that several alkalizing drugs induce the accumulation of AICD in neuroblastoma SY5Y cell lines stably expressing APP constructs. Moreover, alkalizing drugs induce AICD accumulation in naive SY5Y, HEK and COS cells. This accumulation is not mediated by the proteasome or metallopeptidases and is not the result of an increased gamma-secretase activity since the gamma-secretase cleavage of Notch1 and N-Cadherin is not affected by alkalizing drug treatments. Altogether, our data demonstrate for the first time that alkalizing drugs induce the accumulation of AICD, a mechanism likely mediated by the endosome/lysosome pathway.
Collapse
Affiliation(s)
- Valérie Vingtdeux
- INSERM U815, Centre Jean-Pierre Aubert, Neurodegenerative Disorders and Neuronal Death, 1, Place de Verdun, F-59045 Lille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Dev KK, Chatterjee S, Osinde M, Stauffer D, Morgan H, Kobialko M, Dengler U, Rueeger H, Martoglio B, Rovelli G. Signal peptide peptidase dependent cleavage of type II transmembrane substrates releases intracellular and extracellular signals. Eur J Pharmacol 2006; 540:10-7. [PMID: 16697367 DOI: 10.1016/j.ejphar.2006.04.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Revised: 03/27/2006] [Accepted: 04/03/2006] [Indexed: 10/24/2022]
Abstract
The intramembrane-cleaving proteases (I-CLiPs) presenilin-1 and -2 (PS1 and PS2), signal peptide peptidase (SPP) and the Site-2 protease (S2P) catalyze critical steps in cell signaling and are implicated in diseases such as Alzheimer's disease, hepatitis C virus (HCV) infection and cholesterol homeostasis. Here we describe the development of a cellular assay based on cleavage of the transmembrane sequence of the HCV core protein precursor, releasing intra- and extra-cellular signals that represent sequential signal peptidase and SPP cleavage, respectively. We find that the SPP inhibitor (Z-LL)2-ketone (IC50 = 1.33 microM) and the gamma-secretase potent inhibitors NVP-AHW700-NX (IC50 = 51 nM) and LY411575 (IC50 = 61 nM) but not DAPT dose dependently inhibited SPP but not signal peptidase cleavage. Our data confirm that type II orientated substrates, like the HCV transmembrane sequence, are sequentially cleaved by signal peptidase then SPP. This dual assay provides a powerful tool to pharmacologically analyze sequential cleavage events of signal peptidase and SPP and their regulation.
Collapse
Affiliation(s)
- Kumlesh K Dev
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, CH-4002 Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ji ZS, Müllendorff K, Cheng IH, Miranda RD, Huang Y, Mahley RW. Reactivity of apolipoprotein E4 and amyloid beta peptide: lysosomal stability and neurodegeneration. J Biol Chem 2005; 281:2683-92. [PMID: 16298992 DOI: 10.1074/jbc.m506646200] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We previously demonstrated that apolipoprotein E4 (apoE4) potentiates lysosomal leakage and apoptosis induced by amyloid beta (Abeta) peptide in cultured Neuro-2a cells and hypothesized that the low pH of lysosomes accentuates the conversion of apoE4 to a molten globule, inducing reactive intermediates capable of destabilizing cellular membranes. Here we report that neutralizing lysosomal pH with bafilomycin or NH4Cl abolished the apoE4 potentiation of Abeta-induced lysosomal leakage and apoptosis in Neuro-2a cells. Consistent with these results, apoE4 at acidic pH bound more avidly to phospholipid vesicles and disrupted them to a greater extent than at pH 7.4. Comparison of "Arctic" mutant Abeta, which forms multimers, and GM6 mutant Abeta, which remains primarily monomeric, showed that aggregation is essential for apoE4 to potentiate Abeta-induced lysosomal leakage and apoptosis. Both apoE4 and Abeta1-42 had to be internalized to exert these effects. Blocking the low density lipoprotein receptor-related protein with small interfering RNA abolished the enhanced effects of apoE4 and Abeta on lysosomes and apoptosis. In cultured Neuro-2a cells, Abeta1-42 increased lysosome formation to a greater extent in apoE3- or apoE4-transfected cells than in Neo-transfected cells, as shown by immunostaining for lysosome-associated membrane protein 1. Similarly, in transgenic mice expressing apoE and amyloid precursor protein, hippocampal neurons displayed increased numbers of lysosomes. Thus, apoE4 and Abeta1-42 may work in concert in neurons to increase lysosome formation while increasing the susceptibility of lysosomal membranes to disruption, release of lysosomal enzymes into the cytosol, and neuronal degeneration.
Collapse
Affiliation(s)
- Zhong-Sheng Ji
- Gladstone Institute of Neurological Disease, Gladstone Institute of Cardiovascular Disease, University of California, San Francisco, California 94158,USA
| | | | | | | | | | | |
Collapse
|
42
|
Scragg JL, Fearon IM, Boyle JP, Ball SG, Varadi G, Peers C. Alzheimer's amyloid peptides mediate hypoxic up-regulation of L-type Ca2+ channels. FASEB J 2004; 19:150-2. [PMID: 15494446 DOI: 10.1096/fj.04-2659fje] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We examined the effects of chronic hypoxia on recombinant human L-type Ca2+ channel alpha1C subunits stably expressed in HEK 293 cells, using whole-cell patch-clamp recordings. Current density was dramatically increased following 24 h exposure to chronic hypoxia (CH), and membrane channel protein levels were enhanced. CH also increased the levels of Alzheimer's amyloid beta peptides (AbetaPs), determined immunocytochemically. Pharmacological prevention of AbetaP production (via exposure to inhibitors of secretase enzymes that are required to cleave AbetaP from its precursor protein) prevented hypoxic augmentation of currents, as did inhibition of vesicular trafficking with bafilomycin A1. The enhancing effect of AbetaPs or CH were abolished following incubation with the monoclonal 3D6 antibody, raised against the extracellular N' terminus of AbetaP. Immunolocalization and immunoprecipitation studies provided compelling evidence that AbetaPs physically associated with the alpha1C subunit, and this association was promoted by hypoxia. These data suggest an important role for AbetaPs in mediating the increase in Ca2+ channel activity following CH and show that AbetaPs act post-transcriptionally to promote alpha1C subunit insertion into (and/or retention within) the plasma membrane. Such an action will likely contribute to the Ca2+ dyshomeostasis of Alzheimer's disease and may contribute to the mechanisms underlying the known increased incidence of this neurodegenerative disease following hypoxic episodes.
Collapse
Affiliation(s)
- Jason L Scragg
- Institute for Cardiovascular Research, The University of Leeds, Leeds, UK
| | | | | | | | | | | |
Collapse
|
43
|
Froestl B, Steiner B, Müller WE. Enhancement of proteolytic processing of the β-amyloid precursor protein by hyperforin. Biochem Pharmacol 2003; 66:2177-84. [PMID: 14609742 DOI: 10.1016/j.bcp.2003.08.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We studied the effect of hyperforin, a component of St. John's wort (Hypericum perforatum) extracts, on the processing of the amyloid precursor protein (APP) in rat pheochromocytoma PC12 cells, stably transfected with human wildtype APP. We observed transiently increased release of secretory APP fragments upon hyperforin treatment. Unique features, like a strong reduction of intracellular APP and the time course of soluble APP release, distinguished the effects of hyperforin from those of alkalizing agents and phorbol esters, well known activators of secretory processing of APP. Carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP), a protonophore, induced an almost identical decrease in intracellular pH in PC12 cells as does hyperforin. Despite this, FCCP induced a less pronounced release of soluble APP fragments and only slightly reduced intracellular APP levels. These results suggest that hyperforin is an activator of secretory processing of APP with a novel mechanism of action not solely dependent on its effects on intracellular pH.
Collapse
Affiliation(s)
- Bettina Froestl
- Department of Pharmacology, Biocenter, University of Frankfurt, Marie-Curie-Str. 9, D-60439 Frankfurt, Germany
| | | | | |
Collapse
|
44
|
Khan MT, Joseph SK. Proteolysis of type I inositol 1,4,5-trisphosphate receptor in WB rat liver cells. Biochem J 2003; 375:603-11. [PMID: 12927021 PMCID: PMC1223733 DOI: 10.1042/bj20030828] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2003] [Revised: 08/12/2003] [Accepted: 08/20/2003] [Indexed: 11/17/2022]
Abstract
A comparison of the basal degradation of type I Ins P3Rs [L- myo -inositol 1,4,5-trisphosphate receptor], measured by pulse-chase analysis or by analysis of immunoreactive Ins P3Rs after cycloheximide addition, indicated that the small pool of newly synthesized radioactive Ins P3Rs degraded relatively rapidly compared with the large pool of mature Ins P3Rs. An antibody (Ab) against a peptide sequence within the IL-3 (third intraluminal loop) of the receptor (IL-3 Ab) was used to identify protected proteolytic fragments that may accumulate in cells. The IL-3 Ab recognized a 56 kDa fragment in both WB rat liver cells and A7R5 smooth-muscle cells. Gel filtration experiments indicated that the 56 kDa fragment was monomeric and, based on reactivity to other Abs, was missing the cytosol-exposed N- and C-terminal segments of the receptor. The addition of the lysosomal protease inhibitor chloroquine resulted in the rapid disappearance of the 56 kDa band. This effect was mimicked by the cysteine protease inhibitors leupeptin, N -acetyl-L-leucyl-L-leucyl-L-methioninal and N -acetyl-leucyl-leucyl-norleucinal. Lactacystin and NH4Cl were less effective. A second fragment of 16 kDa containing the C-terminus accumulated only when the cells were treated with NH4Cl, and not with any of the other inhibitors tested. No N-terminal-reactive fragments were observed. We propose that mature Ins P3R tetramers dissociate into monomers and that the 56 kDa fragment is a cleavage intermediate of the monomer representing the six transmembrane domains. Angiotensin-II-stimulated down-regulation of Ins P3Rs in WB cells has been shown to involve the ubiquitin/proteasome pathway. Angiotensin-II treatment of WB cells neither resulted in the accumulation of any new fragments nor increased the levels of the 56 or 16 kDa fragments. We conclude that basal and agonist-stimulated degradations of Ins P3Rs occur by different pathways. The agonist-mediated pathway involves the concerted removal and proteolysis of the entire receptor molecule from the endoplasmic reticulum membrane without the appearance of intermediate intraluminal fragments.
Collapse
Affiliation(s)
- M Tariq Khan
- Department of Pathology & Cell Biology, Thomas Jefferson University School of Medicine, Philadelphia, PA 19107, USA
| | | |
Collapse
|
45
|
Pasternak SH, Bagshaw RD, Guiral M, Zhang S, Ackerley CA, Pak BJ, Callahan JW, Mahuran DJ. Presenilin-1, nicastrin, amyloid precursor protein, and gamma-secretase activity are co-localized in the lysosomal membrane. J Biol Chem 2003; 278:26687-94. [PMID: 12736250 DOI: 10.1074/jbc.m304009200] [Citation(s) in RCA: 222] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Alzheimer's disease (AD) is caused by the cerebral deposition of beta-amyloid (Abeta), a 38-43-amino acid peptide derived by proteolytic cleavage of the amyloid precursor protein (APP). Initial studies indicated that final cleavage of APP by the gamma-secretase (a complex containing presenilin and nicastrin) to produce Abeta occurred in the endosomal/lysosomal system. However, other studies showing a predominant endoplasmic reticulum localization of the gamma-secretase proteins and a neutral pH optimum of in vitro gamma-secretase assays have challenged this conclusion. We have recently identified nicastrin as a major lysosomal membrane protein. In the present work, we use Western blotting and immunogold electron microscopy to demonstrate that significant amounts of mature nicastrin, presenilin-1, and APP are co-localized with lysosomal associated membrane protein-1 (cAMP-1) in the outer membranes of lysosomes. Furthermore, we demonstrate that these membranes contain an acidic gamma-secretase activity, which is immunoprecipitable with an antibody to nicastrin. These experiments establish APP, nicastrin, and presenilin-1 as resident lysosomal membrane proteins and indicate that gamma-secretase is a lysosomal protease. These data reassert the importance of the lysosomal/endosomal system in the generation of Abeta and suggest a role for lysosomes in the pathophysiology of AD.
Collapse
Affiliation(s)
- Stephen H Pasternak
- Research Institute, The Hospital for Sick Children, University of Toronto, Toronto M5G 1X8, Canada
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Weihofen A, Lemberg MK, Friedmann E, Rueeger H, Schmitz A, Paganetti P, Rovelli G, Martoglio B. Targeting presenilin-type aspartic protease signal peptide peptidase with gamma-secretase inhibitors. J Biol Chem 2003; 278:16528-33. [PMID: 12621027 DOI: 10.1074/jbc.m301372200] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Presenilin is implicated in the pathogenesis of Alzheimer's disease. It is thought to constitute the catalytic subunit of the gamma-secretase complex that catalyzes intramembrane cleavage of beta-amyloid precursor protein, the last step in the generation of amyloidogenic Abeta peptides. The latter are major constituents of amyloid plaques in the brain of Alzheimer's disease patients. Inhibitors of gamma-secretase are considered potential therapeutics for the treatment of this disease because they prevent production of Abeta peptides. Recently, we discovered a family of presenilin-type aspartic proteases. The founding member, signal peptide peptidase, catalyzes intramembrane cleavage of distinct signal peptides in the endoplasmic reticulum membrane of animals. In humans, the protease plays a crucial role in the immune system. Moreover, it is exploited by the hepatitis C virus for the processing of the structural components of the virion and hence is an attractive target for anti-infective intervention. Signal peptide peptidase and presenilin share identical active site motifs and both catalyze intramembrane proteolysis. These common features let us speculate that gamma-secretase inhibitors directed against presenilin may also inhibit signal peptide peptidase. Here we demonstrate that some of the most potent known gamma-secretase inhibitors efficiently inhibit signal peptide peptidase. However, we found compounds that showed higher specificity for one or the other protease. Our findings highlight the possibility of developing selective inhibitors aimed at reducing Abeta generation without affecting other intramembrane-cleaving aspartic proteases.
Collapse
Affiliation(s)
- Andreas Weihofen
- Institute of Biochemistry, Swiss Federal Institute of Technology (ETH), ETH-Hoenggerberg, 8093 Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Gordon DJ, Tappe R, Meredith SC. Design and characterization of a membrane permeable N-methyl amino acid-containing peptide that inhibits Abeta1-40 fibrillogenesis. THE JOURNAL OF PEPTIDE RESEARCH : OFFICIAL JOURNAL OF THE AMERICAN PEPTIDE SOCIETY 2002; 60:37-55. [PMID: 12081625 DOI: 10.1034/j.1399-3011.2002.11002.x] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Alzheimer's disease, Huntington's disease and prion diseases are part of a growing list of diseases associated with formation of beta-sheet containing fibrils. In a previous publication, we demonstrated that the self-association of the Alzheimer's beta-amyloid (Abeta) peptide is inhibited by peptides homologous to the central core domain of Abeta, but containing N-methyl amino acids at alternate positions. When these inhibitor peptides are arrayed in an extended, beta-strand conformation, the alternating position of N-methyl amino acids gives the peptide two distinct faces, one exhibiting a normal pattern of peptide backbone hydrogen bonds, but the other face having limited hydrogen-bonding capabilities due to the replacement of the amide protons by N-methyl groups. Here, we demonstrate, through two-dimensional NMR and circular dichroic spectroscopy, that a pentapeptide with two N-methyl amino acids, Abeta16-20m or Ac-K(Me)LV(Me)FF-NH2, does indeed have the intended structure of an extended beta-strand. This structure is remarkably stable to changes in solvent conditions and resists denaturation by heating, changes in pH (from 2.5 to 10.5), and addition of denaturants such as urea and guanindine-HCl. We also show that this peptide, despite its hydrophobic composition, is highly water soluble, to concentrations > 30 mm, in contrast to the nonmethylated congener, Abeta16-20 (Ac-KLVFF-NH2). The striking water solubility, in combination with the hydrophobic composition of the peptide, suggested that the peptide might be able to pass spontaneously through cell membranes and model phospholipid bilayers such as unilamellar vesicles. Thus, we also demonstrate that this peptide is indeed able to pass spontaneously through both synthetic phospholipid bilayer vesicles and cell membranes. Characterization of the biophysical properties of the Abeta16-20m peptide may facilitate the application of this strategy to other systems as diverse as the HIV protease and chemokines, in which there is dimerization through beta-strand domains.
Collapse
Affiliation(s)
- D J Gordon
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|
48
|
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder that is characterized by extracellular deposits of amyloid-beta peptide (Abeta) and a severe depletion of the cholinergic system, although the relationship between these two events is poorly understood. In the neocortex, there is a loss of cholinergic fibers and receptors and a decrease of both choline acetyltransferase (ChAT) and acetylcholinesterase enzyme activities. The nucleus basalis of Meynert (NBM), which provides the major cholinergic input to the neocortex, undergoes profound neuron loss in AD. In the present study, we have examined the cholinergic alterations in amyloid precursor protein transgenic mice (APP23), a mouse model of cerebral beta-amyloidosis. In aged APP23 mice, our results reveal modest decreases in cortical cholinergic enzyme activity compared with age-matched wild-type mice. Total cholinergic fiber length was more severely affected, with 29 and 35% decreases in the neocortex of aged APP23 mice compared with age-matched wild-type mice and young transgenic mice, respectively. However, there was no loss of cholinergic basal forebrain neurons in these aged APP23 mice, suggesting that the cortical cholinergic deficit in APP23 mice is locally induced by the deposition of amyloid and is not caused by a loss of cholinergic basal forebrain neurons. To study the impact of cholinergic basal forebrain degeneration on cortical amyloid deposition, we performed unilateral NBM lesions in adult APP23 mice. Three to 8 months after lesioning, a 38% reduction in ChAT activity and significant cholinergic fiber loss were observed in the ipsilateral frontal cortex. There was a 19% decrease in Abeta levels of the ipsilateral compared with contralateral frontal cortex with no change in the ratio of Abeta40 to Abeta42. We conclude that the severe cholinergic deficit in AD is caused by both the loss of cholinergic basal forebrain neurons and locally by cerebral amyloidosis in the neocortex. Moreover, our results suggest that disruption of the basal cholinergic forebrain system does not promote cerebral amyloidosis in APP23 transgenic mice.
Collapse
|
49
|
Bodendorf U, Fischer F, Bodian D, Multhaup G, Paganetti P. A splice variant of beta-secretase deficient in the amyloidogenic processing of the amyloid precursor protein. J Biol Chem 2001; 276:12019-23. [PMID: 11152683 DOI: 10.1074/jbc.m008861200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
beta-Secretase (BACE) initiates the amyloidogenic processing of the amyloid precursor protein leading to the generation of the beta-amyloid, the main component of Alzheimer's disease senile plaques. BACE is a type I transmembrane aspartyl protease of 501 amino acids. Here we describe a novel BACE mRNA lacking 132 base pairs that is expressed in the pancreas but not in the brain. Sequence alignment indicates that the deleted fragment matches the terminal two-thirds of exon 3. The new BACE variant is short of a 44-amino acid region located between the two catalytic aspartyl residues. Accordingly, a 50-kDa form of BACE (BACE457) is detected in the human pancreas. When expressed in cells, BACE457 colocalizes with the marker for the endoplasmic reticulum BiP. Moreover, BACE457 remains in a proenzymatic and endoglycosidase H-sensitive state, suggesting that its transport along the secretory pathway is blocked at the level of the endoplasmic reticulum. Notably, this novel form of BACE does not contribute to the processing of the amyloid precursor protein. Our findings suggest that tissue-specific splicing of the BACE mRNA may explain the observation that in the human pancreas robust transcription of the BACE gene does not translate into recovered enzymatic activity.
Collapse
Affiliation(s)
- U Bodendorf
- Nervous System, Novartis Pharma AG, CH-4002 Basel, Switzerland
| | | | | | | | | |
Collapse
|
50
|
Cataldo AM, Peterhoff CM, Troncoso JC, Gomez-Isla T, Hyman BT, Nixon RA. Endocytic pathway abnormalities precede amyloid beta deposition in sporadic Alzheimer's disease and Down syndrome: differential effects of APOE genotype and presenilin mutations. THE AMERICAN JOURNAL OF PATHOLOGY 2000; 157:277-86. [PMID: 10880397 PMCID: PMC1850219 DOI: 10.1016/s0002-9440(10)64538-5] [Citation(s) in RCA: 619] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Endocytosis is critical to the function and fate of molecules important to Alzheimer's disease (AD) etiology, including the beta protein precursor (betaPP), amyloid beta (Abeta) peptide, and apolipoprotein E (ApoE). Early endosomes, a major site of Abeta peptide generation, are markedly enlarged within neurons in the Alzheimer brain, suggesting altered endocytic pathway (EP) activity. Here, we show that neuronal EP activation is a specific and very early response in AD. To evaluate endocytic activation, we used markers of internalization (rab5, rabaptin 5) and recycling (rab4), and found that enlargement of rab5-positive early endosomes in the AD brain was associated with elevated levels of rab4 immunoreactive protein and translocation of rabaptin 5 to endosomes, implying that both endocytic uptake and recycling are activated. These abnormalities were evident in pyramidal neurons of the neocortex at preclinical stages of disease when Alzheimer-like neuropathology, such as Abeta deposition, was restricted to the entorhinal region. In Down syndrome, early endosomes were significantly enlarged in some pyramidal neurons as early as 28 weeks of gestation, decades before classical AD neuropathology develops. Markers of EP activity were only minimally influenced by normal aging and other neurodegenerative diseases studied. Inheritance of the epsilon4 allele of APOE, however, accentuated early endosome enlargement at preclinical stages of AD. By contrast, endosomes were normal in size at advanced stages of familial AD caused by mutations of presenilin 1 or 2, indicating that altered endocytosis is not a consequence of Abeta deposition. These results identify EP activation as the earliest known intraneuronal change to occur in sporadic AD, the most common form of AD. Given the important role of the EP in Abeta peptide generation and ApoE function, early endosomal abnormalities provide a mechanistic link between EP alterations, genetic susceptibility factors, and Abeta generation and suggest differences that may be involved in Abeta generation and beta amyloidogenesis in subtypes of AD.
Collapse
Affiliation(s)
- A M Cataldo
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA.
| | | | | | | | | | | |
Collapse
|