1
|
Ullah R, Ali G, Baseer A, Irum Khan S, Akram M, Khan S, Ahmad N, Farooq U, Kanwal Nawaz N, Shaheen S, Kumari G, Ullah I. Tannic acid inhibits lipopolysaccharide-induced cognitive impairment in adult mice by targeting multiple pathological features. Int Immunopharmacol 2022; 110:108970. [DOI: 10.1016/j.intimp.2022.108970] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/05/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022]
|
2
|
Li X, Xiu W, Xiao H, Li Y, Yang K, Yuwen L, Yang D, Weng L, Wang L. Fluorescence and ratiometric photoacoustic imaging of endogenous furin activity via peptide functionalized MoS 2 nanosheets. Biomater Sci 2021; 9:8313-8322. [PMID: 34782897 DOI: 10.1039/d1bm01410b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Furin is an important cellular endoprotease, which is expressed at high levels in various cancer cells. Accurate and real-time detection of endogenous furin with high sensitivity and selectivity is significant for the diagnosis of cancer. Herein an activatable nanoprobe (MoS2@PDA-PEG/peptide, MPPF) with dual-mode near-infrared fluorescence (NIRF)/ratiometric photoacoustic (PA) imaging of endogenous furin activity has been developed. The MPPF nanoprobes were constructed by the covalent functionalization of polydopamine (PDA) coated MoS2 nanosheets (NSs) with Cy7-labeled furin substrate peptides. Upon cleavage of the peptides by furin, Cy7 molecules are released from MPPF nanoprobes and recover their fluorescence, realizing furin activity detection with the limit of detection (LOD) down to 3.73 × 10-4 U mL-1. Meanwhile, the ratio of the PA signal at 768 nm to that at 900 nm (PA768/PA900) decreases over time due to the destruction of fluorescence resonance energy transfer effect from Cy7 to MoS2 NSs and the rapid clearance of small Cy7 molecules from tissues. Thus, the simultaneous change in NIRF and ratiometric PA signals enables the imaging of endogenous furin activity in real time, and with high sensitivity, and high selectivity in both tumor cells and tumor-bearing mice.
Collapse
Affiliation(s)
- Xiao Li
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Weijun Xiu
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Hang Xiao
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Yuqing Li
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Kaili Yang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Lihui Yuwen
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| | - Dongliang Yang
- School of Physical and Mathematical Sciences & Institute of Advanced Materials (IAM), Nanjing Tech University, Nanjing 211800, China
| | - Lixing Weng
- School of Geography and Biological Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Lianhui Wang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Centre for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China.
| |
Collapse
|
3
|
Al-Yozbaki M, Acha-Sagredo A, George A, Liloglou T, Wilson CM. Balancing neurotrophin pathway and sortilin function: Its role in human disease. Biochim Biophys Acta Rev Cancer 2020; 1874:188429. [DOI: 10.1016/j.bbcan.2020.188429] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/12/2020] [Accepted: 09/02/2020] [Indexed: 01/03/2023]
|
4
|
Bolus H, Crocker K, Boekhoff-Falk G, Chtarbanova S. Modeling Neurodegenerative Disorders in Drosophila melanogaster. Int J Mol Sci 2020; 21:E3055. [PMID: 32357532 PMCID: PMC7246467 DOI: 10.3390/ijms21093055] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/14/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Drosophila melanogaster provides a powerful genetic model system in which to investigate the molecular mechanisms underlying neurodegenerative diseases. In this review, we discuss recent progress in Drosophila modeling Alzheimer's Disease, Parkinson's Disease, Amyotrophic Lateral Sclerosis (ALS), Huntington's Disease, Ataxia Telangiectasia, and neurodegeneration related to mitochondrial dysfunction or traumatic brain injury. We close by discussing recent progress using Drosophila models of neural regeneration and how these are likely to provide critical insights into future treatments for neurodegenerative disorders.
Collapse
Affiliation(s)
- Harris Bolus
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA;
| | - Kassi Crocker
- Genetics Graduate Training Program, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI 53705, USA;
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Grace Boekhoff-Falk
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI 53705, USA
| | | |
Collapse
|
5
|
Chen W, Li R, Zhu S, Ma J, Pang L, Ma B, Du L, Jin Y. Nasal timosaponin BII dually sensitive in situ hydrogels for the prevention of Alzheimer's disease induced by lipopolysaccharides. Int J Pharm 2020; 578:119115. [PMID: 32045690 DOI: 10.1016/j.ijpharm.2020.119115] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/13/2020] [Accepted: 02/01/2020] [Indexed: 02/01/2023]
Abstract
Alzheimer's disease (AD) is a common and severe brain disease with a high mortality among the elders, but no highly efficient medications are currently available. For example, timosaponin BII, an efficient anti-AD agent, has low oral bioavailability. Here, timosaponin BII was formulated in a temperature/ion-sensitive in situ hydrogel (ISG) that was well transformed into gels in the nasal environment. Timosaponin BII protected the PC12 cells injured by lipopolysaccharides (LPS) by decreasing TNF-α and IL-1β and stabilizing F-actin. Timosaponin BII ISGs were intranasally administered to the mice every day for 38 days. On Day 36, LPS was injected to the mice to establish an AD model. Morris water maze experiments showed that the number of the animals that were able to cross the platform returned to normal and the total distance over which the animals moved in the open field also increased, which demonstrated that the spatial memory and spontaneous behavior were improved after treatment compared to the model. Moreover, an AD improver, inducible nitric oxide synthase (iNOS) in the brain, was reduced after treatment. High brain targeting effect of timosaponin BII ISGs was confirmed by in vivo fluorescence imaging. The nasal timosaponin BII dually sensitive ISGs can serve as a promising medication for local prevention of AD.
Collapse
Affiliation(s)
- Wenyuan Chen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ruiteng Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Siqing Zhu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; Anhui Medical University, Hefei 230032, China
| | - Jinqiu Ma
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lulu Pang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Baiping Ma
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Lina Du
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Anhui Medical University, Hefei 230032, China.
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
6
|
Effects of Single Amino Acid Substitutions on Aggregation and Cytotoxicity Properties of Amyloid β Peptide. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-018-9693-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
7
|
Li X, Cao C, Wei P, Xu M, Liu Z, Liu L, Zhong Y, Li R, Zhou Y, Yi T. Self-Assembly of Amphiphilic Peptides for Recognizing High Furin-Expressing Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:12327-12334. [PMID: 30864434 DOI: 10.1021/acsami.9b01281] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Self-assembled nanostructures of amphiphilic peptides have a wide range of applications in bioimaging and delivery systems. In this study, we design and synthesize a biocompatible amphiphilic peptide (C-3) consisting of an RVRRFFF sequence and a nitrobenzoxadiazole fluorophore that can self-assemble into stable micelles for specifically detecting furin, a kind of proprotein convertase with promoting tumor progression. The self-assembly of C-3 with a β-sheet nanostructure is capable of a rapid and specific response to furin in only 5 min in aqueous solution because of the existence of the RVRR motif in the C-3 molecule. The C-3 nanostructures thus can selectively distinguish high furin-expressing cancer cells, like MDA-MB-231 cells, a kind of human breast cancer cells, from normal cells. Furthermore, the C-3 self-assembly can stay in living cells for a long time and are capable of durable detection of intracellular furin, being good for tracer analysis. To our knowledge, this is the first example of self-assembly of a soluble amphiphilic peptide that can selectively detect furin in high furin-expressing live cells.
Collapse
Affiliation(s)
- Xiang Li
- School of Chemical and Environmental Engineering , Shanghai Institute of Technology , 100 Haiquan Road , Shanghai 201418 , P. R. China
| | - Chunyan Cao
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| | - Peng Wei
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| | - Mengyin Xu
- School of Chemical and Environmental Engineering , Shanghai Institute of Technology , 100 Haiquan Road , Shanghai 201418 , P. R. China
| | - Zhongkuan Liu
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| | - Lingyan Liu
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| | - Yaping Zhong
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| | - Ruohan Li
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| | - Yifeng Zhou
- School of Chemical and Environmental Engineering , Shanghai Institute of Technology , 100 Haiquan Road , Shanghai 201418 , P. R. China
| | - Tao Yi
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| |
Collapse
|
8
|
Yao D, Zhao W, Zhang L, Tian Y. A ratiometric electrochemical strategy for sensitive determination of Furin activity based on dual signal amplification and antifouling nanosurfaces. Analyst 2018; 142:4215-4220. [PMID: 29058010 DOI: 10.1039/c7an01295k] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Developing a sensitive and accurate method for Furin activity is still the bottleneck for understanding the role played by Furin in cell-surface systems and even in Alzheimer's disease. In this work, a ratiometric electrochemical biosensor was developed for sensitive and accurate determination of Furin activity in the cell based on dual signal amplification stemming from a peptide with multiple response sites and the antifouling gold nano-bellflowers (GBFs). A new peptide, HS-CMRVRR↓YKDFDFG (P3), was designed for the first time to be selectively cleaved by Furin at site↓. More importantly, this peptide P3 constitutes three amino acid residues with the -COOH group subsequently used to bind with the response molecule of ferrocene, and can remarkably improve the determination sensitivity by about 2.3 fold. Meanwhile, GBFs stabilized by PEG were taken as a second element to magnify the signal of the ferrocene group via a large ratio surface area and good conductivity, as well as an antibiofouling nanosurface to reduce the biofouling of the electrode surface in cells. This double amplification strategy can greatly enhance the sensitivity of Furin detection by 6.5-fold, which is favorable for detection of low amounts of Furin. In addition, 5'-MB-GGCGCGA(T)13-SH-3' was co-assembled as an inner reference to provide a built-in element to correct the determination error resulting from a complicated analysis environment. Finally, this sensitive and accurate Furin biosensor was successfully applied to detect Furin activity in Furin overexpressed U251 and MDA-MB-468 cells. As far as we know, this is the first report to mention an electrochemical strategy to detect Furin activity in cells.
Collapse
Affiliation(s)
- Dazhi Yao
- Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Dong Chuan Road 500, Shanghai 200241, P.R. China.
| | | | | | | |
Collapse
|
9
|
Plasma β-amyloid 1–42 reference values in cognitively normal subjects. J Neurol Sci 2018; 391:120-126. [DOI: 10.1016/j.jns.2018.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/18/2018] [Accepted: 06/12/2018] [Indexed: 12/17/2022]
|
10
|
Early Contextual Fear Memory Deficits in a Double-Transgenic Amyloid- β Precursor Protein/Presenilin 2 Mouse Model of Alzheimer's Disease. Int J Alzheimers Dis 2017; 2017:8584205. [PMID: 29333315 PMCID: PMC5733185 DOI: 10.1155/2017/8584205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/31/2017] [Accepted: 11/06/2017] [Indexed: 11/17/2022] Open
Abstract
Presenilin 1 and presenilin 2 (PS1 and PS2) play a critical role in γ-secretase-mediated cleavage of amyloid-β precursor protein (APP) and the subsequent generation of β-amyloid peptides. The purpose of the present study was to test whether PS2 mutation accelerates the onset of contextual fear memory deficits in a mouse model of AD that expresses a mutation (K670N/M671L) of the human APP with the Swedish mutation (Tg2576 mice). In the present study, an APP/PS2 double-transgenic mouse model (PS2Tg2576) was generated by crossbreeding transgenic mice carrying the human mutant PS2 (N141I) with Tg2576 mice. Contextual fear conditioning was tested in PS2Tg2576 mice aged 3, 4, 6, and 10-12 months. PS2Tg2576 mice showed a tendency of lower freezing behavior as early as 3 months of age, but significant memory impairment was observed from the age of 4 months. The cognitive impairment was more prominent at ages of 6 and 10-12 months. In contrast, Tg2576 mice aged 3 and 4 months exhibited successful acquisition of contextual fear learning, but Tg2576 mice aged 6 months or older showed significantly impaired fear memory. These results show that PS2 mutation significantly accelerates the onset of fear memory deficits in the APP AD model mice.
Collapse
|
11
|
Walter J, Kemmerling N, Wunderlich P, Glebov K. γ-Secretase in microglia - implications for neurodegeneration and neuroinflammation. J Neurochem 2017; 143:445-454. [PMID: 28940294 DOI: 10.1111/jnc.14224] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/16/2017] [Accepted: 09/18/2017] [Indexed: 12/13/2022]
Abstract
γ-Secretase is an intramembrane cleaving protease involved in the generation of the Alzheimer's disease (AD)-associated amyloid β peptide (Aβ). γ-Secretase is ubiquitously expressed in different organs, and also in different cell types of the human brain. Besides the involvement in the proteolytic generation of Aβ from the amyloid precursor protein, γ-secretase cleaves many additional protein substrates, suggesting pleiotropic functions under physiological and pathophysiological conditions. Microglia exert important functions during brain development and homeostasis in adulthood, and accumulating evidence indicates that microglia and neuroinflammatory processes contribute to the pathogenesis of neurodegenerative diseases. Recent studies demonstrate functional implications of γ-secretase in microglia, suggesting that alterations in γ-secretase activity could contribute to AD pathogenesis by modulation of microglia and related neuroinflammatory processes during neurodegeneration. In this review, we discuss the involvement of γ-secretase in the regulation of microglial functions, and the potential relevance of these processes under physiological and pathophysiological conditions. This article is part of the series "Beyond Amyloid".
Collapse
Affiliation(s)
- Jochen Walter
- Department of Neurology, University of Bonn, Bonn, Germany
| | | | | | | |
Collapse
|
12
|
Panza F, Seripa D, Lozupone M, Solfrizzi V, Imbimbo BP, Barulli MR, Tortelli R, Capozzo R, Bisceglia P, Dimitri A, Stallone R, Dibello V, Quaranta N, Daniele A, Bellomo A, Greco A, Logroscino G. The potential of solanezumab and gantenerumab to prevent Alzheimer’s disease in people with inherited mutations that cause its early onset. Expert Opin Biol Ther 2017; 18:25-35. [DOI: 10.1080/14712598.2018.1389885] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Francesco Panza
- Unit of Neurodegenerative Disease, Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Davide Seripa
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Madia Lozupone
- Unit of Neurodegenerative Disease, Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Vincenzo Solfrizzi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Bruno P. Imbimbo
- Research & Development Department, Chiesi Farmaceutici, Parma, Italy
| | - Maria Rosaria Barulli
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
| | - Rosanna Tortelli
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
| | - Rosa Capozzo
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
| | - Paola Bisceglia
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Andrea Dimitri
- Psychiatric Unit, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Roberta Stallone
- Unit of Neurodegenerative Disease, Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Vittorio Dibello
- Interdisciplinary Department of Medicine (DIM), Section of Dentistry, University of Bari Aldo, Moro, Italy
| | - Nicola Quaranta
- Otolaryngology Unit, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Antonio Greco
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Giancarlo Logroscino
- Unit of Neurodegenerative Disease, Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
| |
Collapse
|
13
|
Pascual AC, Gaveglio VL, Giusto NM, Pasquaré SJ. 2-Arachidonoylglycerol metabolism is differently modulated by oligomeric and fibrillar conformations of amyloid beta in synaptic terminals. Neuroscience 2017; 362:168-180. [PMID: 28844762 DOI: 10.1016/j.neuroscience.2017.08.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 08/19/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent disorder of senile dementia mainly characterized by amyloid-beta peptide (Aβ) deposits in the brain. Cannabinoids are relevant to AD as they exert several beneficial effects in many models of this disease. Still, whether the endocannabinoid system is either up- or down-regulated in AD has not yet been fully elucidated. Thus, the aim of the present paper was to analyze endocannabinoid 2-arachidonoylglycerol (2-AG) metabolism in cerebral cortex synaptosomes incubated with Aβ oligomers or fibrils. These Aβ conformations were obtained by "aging" the 1-40 fragment of the peptide under different agitation and time conditions. A diminished availability of 2-AG resulting from a significant decrease in diacylglycerol lipase (DAGL) activity was observed in the presence of large Aβ1-40 oligomers along with synaptosomal membrane damage, as judged by transmission electron microscopy and LDH release. Conversely, a high availability of 2-AG resulting from an increase in DAGL and lysophosphatidic acid phosphohydrolase activities occurred in the presence of Aβ1-40 fibrils although synaptosomal membrane disruption was also observed. Interestingly, neither synaptosomal mitochondrial viability assayed by MTT reduction nor membrane lipid peroxidation assayed by TBARS formation measurements were altered by Aβ1-40 oligomers or fibrils. These results show a differential effect of Aβ1-40 peptide on 2-AG metabolism depending on its conformation.
Collapse
Affiliation(s)
- Ana C Pascual
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB, UNS-CONICET), Edificio E1, Camino La Carrindanga Km 7, 8000 Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, San Juan 670, 8000 Bahía Blanca, Argentina
| | - Virginia L Gaveglio
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB, UNS-CONICET), Edificio E1, Camino La Carrindanga Km 7, 8000 Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, San Juan 670, 8000 Bahía Blanca, Argentina
| | - Norma M Giusto
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB, UNS-CONICET), Edificio E1, Camino La Carrindanga Km 7, 8000 Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, San Juan 670, 8000 Bahía Blanca, Argentina
| | - Susana J Pasquaré
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB, UNS-CONICET), Edificio E1, Camino La Carrindanga Km 7, 8000 Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, San Juan 670, 8000 Bahía Blanca, Argentina.
| |
Collapse
|
14
|
Dysregulation of intracellular trafficking and endosomal sorting in Alzheimer's disease: controversies and unanswered questions. Biochem J 2017; 473:1977-93. [PMID: 27407168 DOI: 10.1042/bcj20160147] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/18/2016] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of amyloid plaques in the brain consisting of an aggregated form of amyloid β-peptide (Aβ) derived from sequential amyloidogenic processing of the amyloid precursor protein (APP) by membrane-bound proteases β-site APP-cleaving enzyme 1 (BACE1) and γ-secretase. The initial processing of APP by BACE1 is re-gulated by intracellular sorting events of the enzyme, which is a prime target for therapeutic intervention. GWAS (genome-wide sequencing studies) have identified several AD-susceptibility genes that are associated with the regulation of membrane trafficking, and substantial evidence now indicates that AD is likely to arise from defective membrane trafficking in either or both of the secretory and endocytic pathways. Considerable progress has been made in defining the intracellular trafficking pathways of BACE1 and APP and the sorting signals of these membrane proteins that define their itineraries. In this review we highlight recent advances in understanding the regulation of the intracellular sorting of BACE1 and APP, discuss how dysregulation of these trafficking events may lead to enhanced generation of the neurotoxic Aβ products in AD and highlight the unresolved questions in the field.
Collapse
|
15
|
Zakaria R, Wan Yaacob WM, Othman Z, Long I, Ahmad AH, Al-Rahbi B. Lipopolysaccharide-induced memory impairment in rats: a model of Alzheimer's disease. Physiol Res 2017; 66:553-565. [PMID: 28406691 DOI: 10.33549/physiolres.933480] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a primary cause of dementia in the middle-aged and elderly worldwide. Animal models for AD are widely used to study the disease mechanisms as well as to test potential therapeutic agents for disease modification. Among the non-genetically manipulated neuroinflammation models for AD, lipopolysaccharide (LPS)-induced animal model is commonly used. This review paper aims to discuss the possible factors that influence rats' response following LPS injection. Factors such as dose of LPS, route of administration, nature and duration of exposure as well as age and gender of animal used should be taken into account when designing a study using LPS-induced memory impairment as model for AD.
Collapse
Affiliation(s)
- R Zakaria
- Department of Physiology and Department of Psychiatry, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia.
| | | | | | | | | | | |
Collapse
|
16
|
Gu K, Li Q, Lin H, Zhu J, Mo J, He S, Lu X, Jiang X, Sun H. Gamma secretase inhibitors: a patent review (2013 - 2015). Expert Opin Ther Pat 2017; 27:851-866. [PMID: 28350212 DOI: 10.1080/13543776.2017.1313231] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Gamma secretase (GS) is an intricate and multi-subunits complex, and it can cut various transmembrane proteins. Now it is a therapeutic target for a number of diseases. However, due to some side effects, the clinical development of GSI is not successful. Therefore, searching for effective GSIs has become a key point in drug discovery. Areas covered: This review discusses the structure and function of GS and various types of GSIs. And this article seeks to give an overview of the patents or applications published from 2013 to 2015 in which novel chemical classes are claimed to inhibit the GS. Expert opinion: Firstly, further understanding the structure and function of GS to elucidate the disease mechanism and develop AD therapies is urgent. Secondly, if the bioequivalence, pharmacokinetics and selectivity can be improved greatly, some failed clinical inhibitors still can become the promising compounds for clinical trials. Thirdly, some weaknesses are exposed during the development of GSI, especially the insufficient potency, low brain penetration and poor selectivity. Finally, to find potent and selective GSI is the major direction in future. Moreover, to find new indications and dosing regimens in a trial of GSIs also can be seen as new ways.
Collapse
Affiliation(s)
- Kai Gu
- a Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing , China
| | - Qi Li
- a Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing , China
| | - Hongzhi Lin
- a Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing , China
| | - Jie Zhu
- a Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing , China
| | - Jun Mo
- a Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing , China
| | - Siyu He
- a Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing , China
| | - Xin Lu
- a Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing , China
| | - Xueyang Jiang
- b Key Laboratory of Biomedical Functional Materials, School of Science , China Pharmaceutical University , Nanjing , China
| | - Haopeng Sun
- a Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing , China
| |
Collapse
|
17
|
Copenhaver PF, Kögel D. Role of APP Interactions with Heterotrimeric G Proteins: Physiological Functions and Pathological Consequences. Front Mol Neurosci 2017; 10:3. [PMID: 28197070 PMCID: PMC5281615 DOI: 10.3389/fnmol.2017.00003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/05/2017] [Indexed: 12/27/2022] Open
Abstract
Following the discovery that the amyloid precursor protein (APP) is the source of β-amyloid peptides (Aβ) that accumulate in Alzheimer’s disease (AD), structural analyses suggested that the holoprotein resembles a transmembrane receptor. Initial studies using reconstituted membranes demonstrated that APP can directly interact with the heterotrimeric G protein Gαo (but not other G proteins) via an evolutionarily G protein-binding motif in its cytoplasmic domain. Subsequent investigations in cell culture showed that antibodies against the extracellular domain of APP could stimulate Gαo activity, presumably mimicking endogenous APP ligands. In addition, chronically activating wild type APP or overexpressing mutant APP isoforms linked with familial AD could provoke Go-dependent neurotoxic responses, while biochemical assays using human brain samples suggested that the endogenous APP-Go interactions are perturbed in AD patients. More recently, several G protein-dependent pathways have been implicated in the physiological roles of APP, coupled with evidence that APP interacts both physically and functionally with Gαo in a variety of contexts. Work in insect models has demonstrated that the APP ortholog APPL directly interacts with Gαo in motile neurons, whereby APPL-Gαo signaling regulates the response of migratory neurons to ligands encountered in the developing nervous system. Concurrent studies using cultured mammalian neurons and organotypic hippocampal slice preparations have shown that APP signaling transduces the neuroprotective effects of soluble sAPPα fragments via modulation of the PI3K/Akt pathway, providing a mechanism for integrating the stress and survival responses regulated by APP. Notably, this effect was also inhibited by pertussis toxin, indicating an essential role for Gαo/i proteins. Unexpectedly, C-terminal fragments (CTFs) derived from APP have also been found to interact with Gαs, whereby CTF-Gαs signaling can promote neurite outgrowth via adenylyl cyclase/PKA-dependent pathways. These reports offer the intriguing perspective that G protein switching might modulate APP-dependent responses in a context-dependent manner. In this review, we provide an up-to-date perspective on the model that APP plays a variety of roles as an atypical G protein-coupled receptor in both the developing and adult nervous system, and we discuss the hypothesis that disruption of these normal functions might contribute to the progressive neuropathologies that typify AD.
Collapse
Affiliation(s)
- Philip F Copenhaver
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Sciences University, Portland OR, USA
| | - Donat Kögel
- Experimental Neurosurgery, Goethe University Frankfurt Frankfurt am Main, Germany
| |
Collapse
|
18
|
Kim NY, Cho MH, Won SH, Kang HJ, Yoon SY, Kim DH. Sorting nexin-4 regulates β-amyloid production by modulating β-site-activating cleavage enzyme-1. ALZHEIMERS RESEARCH & THERAPY 2017; 9:4. [PMID: 28109317 PMCID: PMC5251330 DOI: 10.1186/s13195-016-0232-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 12/28/2016] [Indexed: 12/30/2022]
Abstract
BACKGROUND Amyloid precursor protein (APP) is cleaved by β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) to produce β-amyloid (Aβ), a critical pathogenic peptide in Alzheimer's disease (AD). Aβ generation can be affected by the intracellular trafficking of APP or its related secretases, which is thus important to understanding its pathological alterations. Although sorting nexin (SNX) family proteins regulate this trafficking, the relevance and role of sorting nexin-4 (SNX4) regarding AD has not been studied yet. METHODS In this study, human brain tissue and APP/PS1 mouse brain tissue were used to check the disease relevance of SNX4. To investigate the role of SNX4 in AD pathogenesis, several experiments were done, such as coimmunoprecipitation, Western blotting, immunohistochemistry, and gradient fractionation. RESULTS We found that SNX4 protein levels changed in the brains of patients with AD and of AD model mice. Overexpression of SNX4 significantly increased the levels of BACE1 and Aβ. Downregulation of SNX4 had the opposite effect. SNX4 interacts with BACE1 and prevents BACE1 trafficking to the lysosomal degradation system, resulting in an increased half-life of BACE1 and increased production of Aβ. CONCLUSIONS We show that SNX4 regulates BACE1 trafficking. Our findings suggest novel therapeutic implications of modulating SNX4 to regulate BACE1-mediated β-processing of APP and subsequent Aβ generation.
Collapse
Affiliation(s)
- Na-Young Kim
- Alzheimer's Disease Experts Lab (ADEL), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Brain Science, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, SongPa-Gu, Seoul, 05505, Korea.,Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Korea.,Cell Dysfunction Research Center (CDRC), University of Ulsan College of Medicine, Seoul, Korea
| | - Mi-Hyang Cho
- Alzheimer's Disease Experts Lab (ADEL), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Brain Science, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, SongPa-Gu, Seoul, 05505, Korea.,Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Korea.,Cell Dysfunction Research Center (CDRC), University of Ulsan College of Medicine, Seoul, Korea
| | - Se-Hoon Won
- Alzheimer's Disease Experts Lab (ADEL), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Brain Science, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, SongPa-Gu, Seoul, 05505, Korea.,Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Korea.,Cell Dysfunction Research Center (CDRC), University of Ulsan College of Medicine, Seoul, Korea
| | - Hoe-Jin Kang
- Alzheimer's Disease Experts Lab (ADEL), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Brain Science, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, SongPa-Gu, Seoul, 05505, Korea.,Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Korea.,Cell Dysfunction Research Center (CDRC), University of Ulsan College of Medicine, Seoul, Korea
| | - Seung-Yong Yoon
- Alzheimer's Disease Experts Lab (ADEL), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea. .,Department of Brain Science, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, SongPa-Gu, Seoul, 05505, Korea. .,Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Korea. .,Cell Dysfunction Research Center (CDRC), University of Ulsan College of Medicine, Seoul, Korea.
| | - Dong-Hou Kim
- Alzheimer's Disease Experts Lab (ADEL), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea. .,Department of Brain Science, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, SongPa-Gu, Seoul, 05505, Korea. .,Bio-Medical Institute of Technology (BMIT), University of Ulsan College of Medicine, Seoul, Korea. .,Cell Dysfunction Research Center (CDRC), University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
19
|
Panza F, Seripa D, Solfrizzi V, Imbimbo BP, Lozupone M, Leo A, Sardone R, Gagliardi G, Lofano L, Creanza BC, Bisceglia P, Daniele A, Bellomo A, Greco A, Logroscino G. Emerging drugs to reduce abnormal β-amyloid protein in Alzheimer’s disease patients. Expert Opin Emerg Drugs 2016; 21:377-391. [DOI: 10.1080/14728214.2016.1241232] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
20
|
Girvan P, Miyake T, Teng X, Branch T, Ying L. Kinetics of the Interactions between Copper and Amyloid-β with FAD Mutations and Phosphorylation at the N terminus. Chembiochem 2016; 17:1732-7. [PMID: 27356100 PMCID: PMC5096041 DOI: 10.1002/cbic.201600255] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Indexed: 12/27/2022]
Abstract
Mutations and post‐translational modifications of amyloid‐β (Aβ) peptide in its N terminus have been shown to increase fibril formation, yet the molecular mechanism is not clear. Here we investigated the kinetics of the interactions of copper with two Aβ peptides containing Familial Alzheimer's disease (FAD) mutations (English (H6R) and Tottori (D7N)), as well as with Aβ peptide phosphorylated at serine 8 (pS8). All three peptides bind to copper with a similar rate as the wild‐type (wt). The dissociation rates follow the order pS8>H6R>wt>D7N; the interconversion between the two coordinating species occurs 50 % faster for H6R and pS8, whereas D7N had only a negligible effect. Interestingly, the rate of ternary complex (copper‐bridged heterodimer) formation for the modified peptides was significantly faster than that for wt, thus leading us to propose that FAD and sporadic AD might share a kinetic origin for the enhanced oligomerisation of Aβ.
Collapse
Affiliation(s)
- Paul Girvan
- Institute of Chemical Biology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK.,Department of Chemistry, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Toru Miyake
- Molecular Medicine, National Heart and Lung Institute, Imperial College London, Exhibition Road, London, SW7 2AZ, UK.,Faculty of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-0034, Japan
| | - Xiangyu Teng
- Institute of Chemical Biology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK.,Department of Chemistry, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Thomas Branch
- Institute of Chemical Biology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK.,Department of Chemistry, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Liming Ying
- Molecular Medicine, National Heart and Lung Institute, Imperial College London, Exhibition Road, London, SW7 2AZ, UK. .,Institute of Chemical Biology, Imperial College London, Exhibition Road, London, SW7 2AZ, UK.
| |
Collapse
|
21
|
Phosphorylation of the amyloid β-peptide at Ser26 stabilizes oligomeric assembly and increases neurotoxicity. Acta Neuropathol 2016; 131:525-37. [PMID: 26898910 PMCID: PMC4789232 DOI: 10.1007/s00401-016-1546-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 02/10/2016] [Accepted: 02/10/2016] [Indexed: 10/25/2022]
Abstract
Aggregation and toxicity of the amyloid β-peptide (Aβ) are considered as critical events in the initiation and progression of Alzheimer's disease (AD). Recent evidence indicated that soluble oligomeric Aβ assemblies exert pronounced toxicity, rather than larger fibrillar aggregates that deposit in the forms of extracellular plaques. While some rare mutations in the Aβ sequence that cause early-onset AD promote the oligomerization, molecular mechanisms that induce the formation or stabilization of oligomers of the wild-type Aβ remain unclear. Here, we identified an Aβ variant phosphorylated at Ser26 residue (pSer26Aβ) in transgenic mouse models of AD and in human brain that shows contrasting spatio-temporal distribution as compared to non-phosphorylated Aβ (npAβ) or other modified Aβ species. pSer26Aβ is particularly abundant in intraneuronal deposits at very early stages of AD, but much less in extracellular plaques. pSer26Aβ assembles into a specific oligomeric form that does not proceed further into larger fibrillar aggregates, and accumulates in characteristic intracellular compartments of granulovacuolar degeneration together with TDP-43 and phosphorylated tau. Importantly, pSer26Aβ oligomers exert increased toxicity in human neurons as compared to other known Aβ species. Thus, pSer26Aβ could represent a critical species in the neurodegeneration during AD pathogenesis.
Collapse
|
22
|
Mandal M, Wu Y, Misiaszek J, Li G, Buevich A, Caldwell JP, Liu X, Mazzola RD, Orth P, Strickland C, Voigt J, Wang H, Zhu Z, Chen X, Grzelak M, Hyde LA, Kuvelkar R, Leach PT, Terracina G, Zhang L, Zhang Q, Michener MS, Smith B, Cox K, Grotz D, Favreau L, Mitra K, Kazakevich I, McKittrick BA, Greenlee W, Kennedy ME, Parker EM, Cumming JN, Stamford AW. Structure-Based Design of an Iminoheterocyclic β-Site Amyloid Precursor Protein Cleaving Enzyme (BACE) Inhibitor that Lowers Central Aβ in Nonhuman Primates. J Med Chem 2016; 59:3231-48. [DOI: 10.1021/acs.jmedchem.5b01995] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Mihirbaran Mandal
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Yusheng Wu
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jeffrey Misiaszek
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Guoqing Li
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Alexei Buevich
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - John P. Caldwell
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaoxiang Liu
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Robert D. Mazzola
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Peter Orth
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Corey Strickland
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Johannes Voigt
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Hongwu Wang
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Zhaoning Zhu
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xia Chen
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Michael Grzelak
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lynn A. Hyde
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Reshma Kuvelkar
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Prescott T. Leach
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Giuseppe Terracina
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lili Zhang
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Qi Zhang
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Maria S. Michener
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Brad Smith
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Kathleen Cox
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Diane Grotz
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Leonard Favreau
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Kaushik Mitra
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Irina Kazakevich
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Brian A. McKittrick
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - William Greenlee
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Matthew E. Kennedy
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Eric M. Parker
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jared N. Cumming
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Andrew W. Stamford
- Department of Global Chemistry, ‡Department of Neuroscience, §Department of Safety Assessment and
Laboratory Animal Research, ∥Department of Discovery Pharmaceutical Sciences, and ⊥Department of
Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
23
|
Tien NT, Karaca I, Tamboli IY, Walter J. Trehalose Alters Subcellular Trafficking and the Metabolism of the Alzheimer-associated Amyloid Precursor Protein. J Biol Chem 2016; 291:10528-40. [PMID: 26957541 DOI: 10.1074/jbc.m116.719286] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Indexed: 01/28/2023] Open
Abstract
The disaccharide trehalose is commonly considered to stimulate autophagy. Cell treatment with trehalose could decrease cytosolic aggregates of potentially pathogenic proteins, including mutant huntingtin, α-synuclein, and phosphorylated tau that are associated with neurodegenerative diseases. Here, we demonstrate that trehalose also alters the metabolism of the Alzheimer disease-related amyloid precursor protein (APP). Cell treatment with trehalose decreased the degradation of full-length APP and its C-terminal fragments. Trehalose also reduced the secretion of the amyloid-β peptide. Biochemical and cell biological experiments revealed that trehalose alters the subcellular distribution and decreases the degradation of APP C-terminal fragments in endolysosomal compartments. Trehalose also led to strong accumulation of the autophagic marker proteins LC3-II and p62, and decreased the proteolytic activation of the lysosomal hydrolase cathepsin D. The combined data indicate that trehalose decreases the lysosomal metabolism of APP by altering its endocytic vesicular transport.
Collapse
Affiliation(s)
- Nguyen T Tien
- From the Department of Neurology, University of Bonn, 53127 Bonn, Germany
| | - Ilker Karaca
- From the Department of Neurology, University of Bonn, 53127 Bonn, Germany
| | - Irfan Y Tamboli
- From the Department of Neurology, University of Bonn, 53127 Bonn, Germany
| | - Jochen Walter
- From the Department of Neurology, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
24
|
Ben Halima S, Mishra S, Raja KMP, Willem M, Baici A, Simons K, Brüstle O, Koch P, Haass C, Caflisch A, Rajendran L. Specific Inhibition of β-Secretase Processing of the Alzheimer Disease Amyloid Precursor Protein. Cell Rep 2016; 14:2127-2141. [PMID: 26923602 DOI: 10.1016/j.celrep.2016.01.076] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 11/09/2015] [Accepted: 01/26/2016] [Indexed: 01/18/2023] Open
Abstract
Development of disease-modifying therapeutics is urgently needed for treating Alzheimer disease (AD). AD is characterized by toxic β-amyloid (Aβ) peptides produced by β- and γ-secretase-mediated cleavage of the amyloid precursor protein (APP). β-secretase inhibitors reduce Aβ levels, but mechanism-based side effects arise because they also inhibit β-cleavage of non-amyloid substrates like Neuregulin. We report that β-secretase has a higher affinity for Neuregulin than it does for APP. Kinetic studies demonstrate that the affinities and catalytic efficiencies of β-secretase are higher toward non-amyloid substrates than toward APP. We show that non-amyloid substrates are processed by β-secretase in an endocytosis-independent manner. Exploiting this compartmentalization of substrates, we specifically target the endosomal β-secretase by an endosomally targeted β-secretase inhibitor, which blocked cleavage of APP but not non-amyloid substrates in many cell systems, including induced pluripotent stem cell (iPSC)-derived neurons. β-secretase inhibitors can be designed to specifically inhibit the Alzheimer process, enhancing their potential as AD therapeutics without undesired side effects.
Collapse
Affiliation(s)
- Saoussen Ben Halima
- Systems and Cell Biology of Neurodegeneration, Institute of Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; Graduate Program in Neuroscience, Neuroscience Center Zurich, 8057 Zurich, Switzerland; Graduate Program of the Zurich Center for Integrative Human Physiology, University of Zurich, 8057 Zurich, Switzerland
| | - Sabyashachi Mishra
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - K Muruga Poopathi Raja
- Department of Physical Chemistry, School of Chemistry, Madurai Kamaraj University, Tamil Nadu 625002, Madurai, India
| | - Michael Willem
- Biomedical Center, Ludwig-Maximilians-University, 81337 Munich, Germany
| | - Antonio Baici
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Kai Simons
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn, 53127 Bonn, Germany; German Center for Neurodegenerative Diseases, 53175 Bonn, Germany; Life & Brain, 53127 Bonn, Germany
| | - Philipp Koch
- Institute of Reconstructive Neurobiology, University of Bonn, 53127 Bonn, Germany
| | - Christian Haass
- Biomedical Center, Ludwig-Maximilians-University, 81337 Munich, Germany; German Center for Neurodegenerative Diseases, 53175 Bonn, Germany; Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Lawrence Rajendran
- Systems and Cell Biology of Neurodegeneration, Institute of Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; Graduate Program in Neuroscience, Neuroscience Center Zurich, 8057 Zurich, Switzerland; Graduate Program of the Zurich Center for Integrative Human Physiology, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
25
|
Lyons B, Friedrich M, Raftery M, Truscott R. Amyloid Plaque in the Human Brain Can Decompose from Aβ(1-40/1-42) by Spontaneous Nonenzymatic Processes. Anal Chem 2016; 88:2675-84. [DOI: 10.1021/acs.analchem.5b03891] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Brian Lyons
- Illawarra
Health and Medical Research Institute, University of Wollongong, Northfields
Avenue, Wollongong, New South
Wales 2522, Australia
- Save
Sight Institute, Sydney Eye Hospital, University of Sydney, 8 Macquarie
Street, Sydney, New South
Wales 2001, Australia
| | - Michael Friedrich
- Illawarra
Health and Medical Research Institute, University of Wollongong, Northfields
Avenue, Wollongong, New South
Wales 2522, Australia
| | - Mark Raftery
- Biological
Mass Spectrometry Facility, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Roger Truscott
- Illawarra
Health and Medical Research Institute, University of Wollongong, Northfields
Avenue, Wollongong, New South
Wales 2522, Australia
| |
Collapse
|
26
|
Vergés M. Retromer in Polarized Protein Transport. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 323:129-79. [PMID: 26944621 DOI: 10.1016/bs.ircmb.2015.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Retromer is an evolutionary conserved protein complex required for endosome-to-Golgi retrieval of receptors for lysosomal hydrolases. It is constituted by a heterotrimer encoded by the vacuolar protein sorting (VPS) gene products Vps26, Vps35, and Vps29, which selects cargo, and a dimer of phosphoinositide-binding sorting nexins, which deforms the membrane. Recent progress in the mechanism of retromer assembly and functioning has strengthened the link between sorting at the endosome and cytoskeleton dynamics. Retromer is implicated in endosomal sorting of many cargos and plays an essential role in plant and animal development. Although it is best known for endosome sorting to the trans-Golgi network, it also intervenes in recycling to the plasma membrane. In polarized cells, such as epithelial cells and neurons, retromer may also be utilized for transcytosis and long-range transport. Considerable evidence implicates retromer in establishment and maintenance of cell polarity. That includes sorting of the apical polarity module Crumbs; regulation of retromer function by the basolateral polarity module Scribble; and retromer-dependent recycling of various cargoes to a certain surface domain, thus controlling polarized location and cell homeostasis. Importantly, altered retromer function has been linked to neurodegeneration, such as in Alzheimer's or Parkinson's disease. This review will underline how alterations in retromer localization and function may affect polarized protein transport and polarity establishment, thereby causing developmental defects and disease.
Collapse
Affiliation(s)
- Marcel Vergés
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI), Girona, Spain; Medical Sciences Department, University of Girona, Girona, Spain.
| |
Collapse
|
27
|
Walter J. The Triggering Receptor Expressed on Myeloid Cells 2: A Molecular Link of Neuroinflammation and Neurodegenerative Diseases. J Biol Chem 2015; 291:4334-41. [PMID: 26694609 DOI: 10.1074/jbc.r115.704981] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The triggering receptor expressed on myeloid cells (TREM) 2 is a member of the immunoglobulin superfamily of receptors and mediates signaling in immune cells via engagement of its co-receptor DNAX-activating protein of 12 kDa (DAP12). Homozygous mutations in TREM2 or DAP12 cause Nasu-Hakola disease, which is characterized by bone abnormalities and dementia. Recently, a variant of TREM2 has also been associated with an increased risk for Alzheimer disease. The selective expression of TREM2 on immune cells and its association with different forms of dementia indicate a contribution of this receptor in common pathways of neurodegeneration.
Collapse
Affiliation(s)
- Jochen Walter
- From the Department of Neurology, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
28
|
Walter J. Twenty Years of Presenilins--Important Proteins in Health and Disease. Mol Med 2015; 21 Suppl 1:S41-8. [PMID: 26605647 DOI: 10.2119/molmed.2015.00163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 07/01/2015] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive decline in cognitive functions associated with depositions of aggregated proteins in the form of extracellular plaques and neurofibrillary tangles in the brain. Extracellular plaques contain characteristic fibrils of amyloid β peptides (Aβ); tangles consist of paired helical filaments of the microtubuli-associated protein tau. Although AD manifests predominantly at ages above 65 years, rare cases show a much earlier onset of disease symptoms with very similar neuropathological characteristics. In 1995, two homologous genes were identified, in which mutations are associated with dominantly inherited familial forms of early onset AD. The genes therefore were dubbed presenilins (PS) and encode polytopic transmembrane proteins. At this time the role of these proteins in the pathogenesis of AD and their biological function in general were completely unknown. However, individuals carrying PS mutations showed alterations in the composition of different length variants of Aβ peptides in blood and cerebrospinal fluid, which indicated the potential involvement of presenilins in the metabolism of Aβ. After 20 years of intense research, the roles of presenilins in Aβ generation as well as important functions in biological processes have been identified. Presenilins represent the catalytic components of protease complexes that directly cleave the amyloid precursor protein (APP) but also many other proteins with important physiological functions. Here, the progress in presenilin research from basic characterization of their cellular functions to the targeting in clinical trials for AD therapy, and potential future directions, will be discussed.
Collapse
Affiliation(s)
- Jochen Walter
- Department of Neurology, University of Bonn, Bonn, Germany
| |
Collapse
|
29
|
Molecular dynamics simulation study reveals potential substrate entry path into γ-secretase/presenilin-1. J Struct Biol 2015; 191:120-9. [DOI: 10.1016/j.jsb.2015.07.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 05/29/2015] [Accepted: 07/01/2015] [Indexed: 11/20/2022]
|
30
|
Barage SH, Sonawane KD. Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer's disease. Neuropeptides 2015; 52:1-18. [PMID: 26149638 DOI: 10.1016/j.npep.2015.06.008] [Citation(s) in RCA: 375] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 06/24/2015] [Accepted: 06/24/2015] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease is an irreversible, progressive neurodegenerative disorder. Various therapeutic approaches are being used to improve the cholinergic neurotransmission, but their role in AD pathogenesis is still unknown. Although, an increase in tau protein concentration in CSF has been described in AD, but several issues remains unclear. Extensive and accurate analysis of CSF could be helpful to define presence of tau proteins in physiological conditions, or released during the progression of neurodegenerative disease. The amyloid cascade hypothesis postulates that the neurodegeneration in AD caused by abnormal accumulation of amyloid beta (Aβ) plaques in various areas of the brain. The amyloid hypothesis has continued to gain support over the last two decades, particularly from genetic studies. Therefore, current research progress in several areas of therapies shall provide an effective treatment to cure this devastating disease. This review critically evaluates general biochemical and physiological functions of Aβ directed therapeutics and their relevance.
Collapse
Affiliation(s)
- Sagar H Barage
- Department of Biotechnology, Shivaji University, Kolhapur 416004, Maharashtra (M.S.), India
| | - Kailas D Sonawane
- Structural Bioinformatics Unit, Department of Biochemistry, Shivaji University, Kolhapur 416004, Maharashtra (M.S.), India; Department of Microbiology, Shivaji University, Kolhapur 416004, Maharashtra (M.S.), India.
| |
Collapse
|
31
|
Oskarsson ME, Paulsson JF, Schultz SW, Ingelsson M, Westermark P, Westermark GT. In vivo seeding and cross-seeding of localized amyloidosis: a molecular link between type 2 diabetes and Alzheimer disease. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:834-46. [PMID: 25700985 DOI: 10.1016/j.ajpath.2014.11.016] [Citation(s) in RCA: 222] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 10/31/2014] [Accepted: 11/06/2014] [Indexed: 01/21/2023]
Abstract
Several proteins have been identified as amyloid forming in humans, and independent of protein origin, the fibrils are morphologically similar. Therefore, there is a potential for structures with amyloid seeding ability to induce both homologous and heterologous fibril growth; thus, molecular interaction can constitute a link between different amyloid forms. Intravenous injection with preformed fibrils from islet amyloid polypeptide (IAPP), proIAPP, or amyloid-beta (Aβ) into human IAPP transgenic mice triggered IAPP amyloid formation in pancreas in 5 of 7 mice in each group, demonstrating that IAPP amyloid could be enhanced through homologous and heterologous seeding with higher efficiency for the former mechanism. Proximity ligation assay was used for colocalization studies of IAPP and Aβ in islet amyloid in type 2 diabetic patients and Aβ deposits in brains of patients with Alzheimer disease. Aβ reactivity was not detected in islet amyloid although islet β cells express AβPP and convertases necessary for Aβ production. By contrast, IAPP and proIAPP were detected in cerebral and vascular Aβ deposits, and presence of proximity ligation signal at both locations showed that the peptides were <40 nm apart. It is not clear whether IAPP present in brain originates from pancreas or is locally produced. Heterologous seeding between IAPP and Aβ shown here may represent a molecular link between type 2 diabetes and Alzheimer disease.
Collapse
Affiliation(s)
- Marie E Oskarsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Johan F Paulsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Martin Ingelsson
- Department of Public Health/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Per Westermark
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
32
|
Panza F, Solfrizzi V, Barulli MR, Bonfiglio C, Guerra V, Osella A, Seripa D, Sabbà C, Pilotto A, Logroscino G. Coffee, tea, and caffeine consumption and prevention of late-life cognitive decline and dementia: a systematic review. J Nutr Health Aging 2015; 19:313-28. [PMID: 25732217 DOI: 10.1007/s12603-014-0563-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A prolonged preclinical phase of more than two decades before the onset of dementia suggested that initial brain changes of Alzheimer's disease (AD) and the symptoms of advanced AD may represent a unique continuum. Given the very limited therapeutic value of drugs currently used in the treatment of AD and dementia, preventing or postponing the onset of AD and delaying or slowing its progression are becoming mandatory. Among possible reversible risk factors of dementia and AD, vascular, metabolic, and lifestyle-related factors were associated with the development of dementia and late-life cognitive disorders, opening new avenues for the prevention of these diseases. Among diet-associated factors, coffee is regularly consumed by millions of people around the world and owing to its caffeine content, it is the best known psychoactive stimulant resulting in heightened alertness and arousal and improvement of cognitive performance. Besides its short-term effect, some case-control and cross-sectional and longitudinal population-based studies evaluated the long-term effects on brain function and provided some evidence that coffee, tea, and caffeine consumption or higher plasma caffeine levels may be protective against cognitive impairment/decline and dementia. In particular, several cross-sectional and longitudinal population-based studies suggested a protective effect of coffee, tea, and caffeine use against late-life cognitive impairment/decline, although the association was not found in all cognitive domains investigated and there was a lack of a distinct dose-response association, with a stronger effect among women than men. The findings on the association of coffee, tea, and caffeine consumption or plasma caffeine levels with incident mild cognitive impairment and its progression to dementia were too limited to draw any conclusion. Furthermore, for dementia and AD prevention, some studies with baseline examination in midlife pointed to a lack of association, although other case-control and longitudinal population-based studies with briefer follow-up periods supported favourable effects of coffee, tea, and caffeine consumption against AD. Larger studies with longer follow-up periods should be encouraged, addressing other potential bias and confounding sources, so hopefully opening new ways for diet-related prevention of dementia and AD.
Collapse
Affiliation(s)
- F Panza
- Francesco Panza, MD, PhD, Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy and Department of Clinical Research in Neurology, University of Bari Aldo Moro, "Pia Fondazione Cardinale G. Panico", Tricase, Lecce, Italy,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Bakele M, Lotz-Havla AS, Jakowetz A, Carevic M, Marcos V, Muntau AC, Gersting SW, Hartl D. An interactive network of elastase, secretases, and PAR-2 protein regulates CXCR1 receptor surface expression on neutrophils. J Biol Chem 2015; 289:20516-25. [PMID: 24914212 DOI: 10.1074/jbc.m114.575803] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
CXCL8 (IL-8) recruits and activates neutrophils through the G protein-coupled chemokine receptor CXCR1. We showed previously that elastase cleaves CXCR1 and thereby impairs antibacterial host defense. However, the molecular intracellular machinery involved in this process remained undefined. Here we demonstrate by using flow cytometry, confocal microscopy, subcellular fractionation, co-immunoprecipitation, and bioluminescence resonance energy transfer that combined α- and γ-secretase activities are functionally involved in elastase-mediated regulation of CXCR1 surface expression on human neutrophils, whereas matrix metalloproteases are dispensable. We further demonstrate that PAR-2 is stored in mobilizable compartments in neutrophils. Bioluminescence resonance energy transfer and co-immunoprecipitation studies showed that secretases, PAR-2, and CXCR1 colocalize and physically interact in a novel protease/secretase-chemokine receptor network. PAR-2 blocking experiments provided evidence that elastase increased intracellular presenilin-1 expression through PAR-2 signaling. When viewed in combination, these studies establish a novel functional network of elastase, secretases, and PAR-2 that regulate CXCR1 expression on neutrophils. Interfering with this network could lead to novel therapeutic approaches in neutrophilic diseases, such as cystic fibrosis or rheumatoid arthritis.
Collapse
|
34
|
Panza F, Solfrizzi V, Imbimbo BP, Giannini M, Santamato A, Seripa D, Logroscino G. Efficacy and safety studies of gantenerumab in patients with Alzheimer's disease. Expert Rev Neurother 2014; 14:973-86. [PMID: 25081412 DOI: 10.1586/14737175.2014.945522] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Among active and passive anti-β-amyloid (Aβ) immunotherapies for Alzheimer's disease (AD), bapineuzumab and solanezumab, two humanized monoclonal antibodies, failed to show significant clinical benefits in mild-to-moderate AD patients in large Phase III clinical trials. Another ongoing Phase III trial of solanezumab aims to confirm positive findings in mild AD patients. Gantenerumab is the first fully human anti-Aβ monoclonal antibody directed to both N-terminal and central regions of Aβ. A 6-month PET study in 16 AD patients showed that gantenerumab treatment dose-dependently reduced brain Aβ deposition, possibly stimulating microglial-mediated phagocytosis. Two ongoing Phase III trials of gantenerumab in patients with prodromal or mild dementia due to AD will determine if any reduction in brain Aβ levels will translate into clinical benefits. An ongoing secondary prevention trial of gantenerumab in presymptomatic subjects with genetic mutations for autosomal-dominant AD will verify the utility of anti-Aβ monoclonal antibodies as prevention therapy.
Collapse
Affiliation(s)
- Francesco Panza
- Department of Basic Medicine, Neuroscience, and Sense Organs, Neurodegenerative Disease Unit, University of Bari Aldo Moro, Bari, Italy
| | | | | | | | | | | | | |
Collapse
|
35
|
Panza F, Solfrizzi V, Imbimbo BP, Logroscino G. Amyloid-directed monoclonal antibodies for the treatment of Alzheimer's disease: the point of no return? Expert Opin Biol Ther 2014; 14:1465-76. [PMID: 24981190 DOI: 10.1517/14712598.2014.935332] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Two humanized monoclonal antibodies, bapineuzumab and solanezumab, directed against the N terminus and mid-region of β-amyloid (Aβ), respectively, were recently tested in large, long-term Phase III trials in patients with mild-to-moderate Alzheimer's disease (AD). AREAS COVERED This review discusses current clinical data on solanezumab, bapineuzumab and their failure in Phase III trials to show significant clinical benefits, as well as other monoclonal antibodies under investigation for AD. EXPERT OPINION Solanezumab showed some beneficial cognitive effects in mildly affected AD patients and this subgroup of AD patients is currently being tested in another Phase III trial to this subgroup of AD patients to confirm previous encouraging observations. Two other monoclonal antibodies, gantenerumab, which preferentially binds to fibrillar Aβ, and crenezumab, which preferentially binds to soluble, oligomeric and fibrillar Aβ deposits, are being tested in secondary prevention trials in presymptomatic subjects with autosomal dominant AD mutations. Solanezumab is also being tested in a prevention study in asymptomatic older subjects, who have positive positron emission tomography scans for brain amyloid deposits. These ongoing secondary prevention trials will tell us if Aβ really plays a crucial role in the pathophysiology of AD.
Collapse
Affiliation(s)
- Francesco Panza
- University of Bari Aldo Moro, Department of Basic Medicine, Neuroscience, and Sense Organs, Neurodegenerative Disease Unit , Bari , Italy
| | | | | | | |
Collapse
|
36
|
Karaca I, Tamboli IY, Glebov K, Richter J, Fell LH, Grimm MO, Haupenthal VJ, Hartmann T, Gräler MH, van Echten-Deckert G, Walter J. Deficiency of sphingosine-1-phosphate lyase impairs lysosomal metabolism of the amyloid precursor protein. J Biol Chem 2014; 289:16761-72. [PMID: 24808180 DOI: 10.1074/jbc.m113.535500] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Progressive accumulation of the amyloid β protein in extracellular plaques is a neuropathological hallmark of Alzheimer disease. Amyloid β is generated during sequential cleavage of the amyloid precursor protein (APP) by β- and γ-secretases. In addition to the proteolytic processing by secretases, APP is also metabolized by lysosomal proteases. Here, we show that accumulation of intracellular sphingosine-1-phosphate (S1P) impairs the metabolism of APP. Cells lacking functional S1P-lyase, which degrades intracellular S1P, strongly accumulate full-length APP and its potentially amyloidogenic C-terminal fragments (CTFs) as compared with cells expressing the functional enzyme. By cell biological and biochemical methods, we demonstrate that intracellular inhibition of S1P-lyase impairs the degradation of APP and CTFs in lysosomal compartments and also decreases the activity of γ-secretase. Interestingly, the strong accumulation of APP and CTFs in S1P-lyase-deficient cells was reversed by selective mobilization of Ca(2+) from the endoplasmic reticulum or lysosomes. Intracellular accumulation of S1P also impairs maturation of cathepsin D and degradation of Lamp-2, indicating a general impairment of lysosomal activity. Together, these data demonstrate that S1P-lyase plays a critical role in the regulation of lysosomal activity and the metabolism of APP.
Collapse
Affiliation(s)
- Ilker Karaca
- From the Department of Neurology, University of Bonn, 53127 Bonn, Germany
| | - Irfan Y Tamboli
- From the Department of Neurology, University of Bonn, 53127 Bonn, Germany
| | - Konstantin Glebov
- From the Department of Neurology, University of Bonn, 53127 Bonn, Germany
| | - Josefine Richter
- German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany
| | - Lisa H Fell
- the Department of Experimental Neurology, University of the Saarland, 66421 Homburg/Saar, Germany
| | - Marcus O Grimm
- the Department of Experimental Neurology, University of the Saarland, 66421 Homburg/Saar, Germany
| | - Viola J Haupenthal
- the Department of Experimental Neurology, University of the Saarland, 66421 Homburg/Saar, Germany
| | - Tobias Hartmann
- the Department of Experimental Neurology, University of the Saarland, 66421 Homburg/Saar, Germany
| | - Markus H Gräler
- the Department of Anaesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care, and Center for Molecular Biomedicine, University Hospital Jena, 07740 Jena, Germany, and
| | - Gerhild van Echten-Deckert
- the Life and Medical Sciences, Membrane Biology and Lipid Biochemistry Unit at the Kekulé-Institute, University of Bonn, 53121 Bonn, Germany
| | - Jochen Walter
- From the Department of Neurology, University of Bonn, 53127 Bonn, Germany,
| |
Collapse
|
37
|
Su Y, Wang Q, Wang C, Chan K, Sun Y, Kuang H. The treatment of Alzheimer's disease using Chinese medicinal plants: from disease models to potential clinical applications. JOURNAL OF ETHNOPHARMACOLOGY 2014; 152:403-423. [PMID: 24412377 DOI: 10.1016/j.jep.2013.12.053] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 11/22/2013] [Accepted: 12/30/2013] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD) is characterized by the sustained higher nervous disorders of the activities and functions of the brain. Due to its heavy burden on society and the patients' families, it is urgent to review the treatments for AD to provide basic data for further research and new drug development. Among these treatments, Chinese Material Medica (CMM) has been traditionally clinical used in China to treat AD for a long time with obvious efficacy. With the further research reports of CMM, new therapeutic materials may be recovered from troves of CMM. However, So far, little or no review work has been reported to conclude anti-AD drugs from CMM in literature. Therefore, a systematic introduction of CMM anti-AD research progress is of great importance and necessity. This paper strives to systematically describe the progress of CMM in the treatment of AD, and lays a basis data for anti-AD drug development from CMM, and provides the essential theoretical support for the further development and utilization of CMM resources through a more comprehensive research of the variety of databases regarding CMM anti-AD effects reports. MATERIAL AND METHODS Literature survey was performed via electronic search (SciFinder®, Pubmed®, Google Scholar and Web of Science) on papers and patents and by systematic research in ethnopharmacological literature at various university libraries. RESULTS This review mainly introduces the current research on the Chinese Material Medica (CMM) theoretical research on Alzheimer's disease (AD), anti-AD active constituent of CMM, anti-AD effects on AD models, anti-AD mechanism of CMM, and anti-AD effect of CMM formula. CONCLUSION Scholars around the world have made studies on the anti-AD molecular mechanism of CMM from different pathways, and have made substantial progress. The progress not only enriched the anti-AD theory of CMM, but also provided clinical practical significance and development prospects in using CMM to treat AD. Western pure drugs cannot replace the advantages of CMM in the anti-AD aspect. Therefore, in the near future, the development of CMM anti-AD drugs with a more clearly role and practical data will be a major trend in the field of AD drug development, and it will promote the use of CMM.
Collapse
Affiliation(s)
- Yang Su
- Key Laboratory of Ministry of Education, Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Qiuhong Wang
- Key Laboratory of Ministry of Education, Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| | - Changfu Wang
- Key Laboratory of Ministry of Education, Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Kelvin Chan
- Centre for Complementary Medicine Research, University of Western Sydney, NSW 2560, Australia; Faculty of Pharmacy, The University of Sydney, NSW 2006, Australia
| | - Yanping Sun
- Key Laboratory of Ministry of Education, Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Haixue Kuang
- Key Laboratory of Ministry of Education, Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW We reviewed clinical trials on active and passive anti-β-amyloid (Aβ) immunotherapy for the treatment of Alzheimer's disease with a particular focus on monoclonal antibodies against Aβ. RECENT FINDINGS Studies on anti-Alzheimer's disease immunotherapy published in the period from January 2012 to October 2013 were reviewed. SUMMARY Both active and passive anti-Aβ immunotherapies were shown to clear brain Aβ deposits. However, an active anti-Aβ vaccine (AN1792) has been discontinued because it caused meningoencephalitis in 6% of Alzheimer's disease patients treated. Among passive immunotherapeutics, two Phase III clinical trials in mild-to-moderate Alzheimer's disease patients with bapineuzumab, a humanized monoclonal antibody directed at the N-terminal sequence of Aβ, were disappointing. Another antibody, solanezumab, directed at the mid-region of Aβ, failed in two Phase III clinical trials in mild-to-moderate Alzheimer's disease patients. A third Phase III study with solanezumab is ongoing in mildly affected Alzheimer's disease patients based on encouraging results in this subgroup of patients. Second-generation active Aβ vaccines (ACC-001, CAD106, and Affitope AD02) and new passive anti-Aβ immunotherapies (gantenerumab and crenezumab) are being tested in prodromal Alzheimer's disease patients, in presymptomatic individuals with Alzheimer's disease-related mutations, or in asymptomatic individuals at risk of developing Alzheimer's disease to definitely test the Aβ cascade hypothesis of Alzheimer's disease.
Collapse
|
39
|
Panza F, Solfrizzi V, Imbimbo BP, Tortelli R, Santamato A, Logroscino G. Amyloid-based immunotherapy for Alzheimer's disease in the time of prevention trials: the way forward. Expert Rev Clin Immunol 2014; 10:405-19. [DOI: 10.1586/1744666x.2014.883921] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
40
|
Rivabene R, Visentin S, Piscopo P, De Nuccio C, Crestini A, Svetoni F, Rosa P, Confaloni A. Thapsigargin affects presenilin-2 but not presenilin-1 regulation in SK-N-BE cells. Exp Biol Med (Maywood) 2013; 239:213-24. [PMID: 24363250 DOI: 10.1177/1535370213514317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Presenilin-1 (PS1) and presenilin-2 (PS2) are transmembrane proteins widely expressed in the central nervous system, which function as the catalytic subunits of γ-secretase, the enzyme that releases amyloid-β protein (Aβ) from ectodomain cleaved amyloid precursor protein (APP) by intramembrane proteolysis. Mutations in PS1, PS2, and Aβ protein precursor are involved in the etiology of familial Alzheimer's disease (FAD), while the cause of the sporadic form of AD (SAD) is still not known. However, since similar neuropathological changes have been observed in both FAD and SAD, a common pathway in the etiology of the disease has been suggested. Given that age-related deranged Ca(2+) regulation has been hypothesized to play a role in SAD pathogenesis via PS gene regulation and γ-secretase activity, we studied the in vitro regulation of PS1 and PS2 in the human neuron-like SK-N-BE cell line treated with the specific endoplasmic reticulum (ER) calcium ATPase inhibitor Thapsigargin (THG), to introduce intracellular Ca(2+) perturbations and mimic the altered Ca(2+) homeostasis observed in AD. Our results showed a consistent and significant down-regulation of PS2, while PS1 appeared to be unmodulated. These events were accompanied by oxidative stress and a number of morphological alterations suggestive of the induction of apoptotic machinery. The administration of the antioxidant N-acetylcysteine (NAC) did not revert the THG-induced effects reported, while treatment with the Ca(2+)-independent ER stressor Brefeldin A did not modulate basal PS1 and PS2 expression. Collectively, these results suggest that Ca(2+) fluctuation rather than ER stress and/or oxidative imbalance seems to play an essential role in PS2 regulation and confirm that, despite their strong homology, PS1 and PS2 could play different roles in AD.
Collapse
Affiliation(s)
- Roberto Rivabene
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena, 299 00161 Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Wunderlich P, Glebov K, Kemmerling N, Tien NT, Neumann H, Walter J. Sequential proteolytic processing of the triggering receptor expressed on myeloid cells-2 (TREM2) protein by ectodomain shedding and γ-secretase-dependent intramembranous cleavage. J Biol Chem 2013; 288:33027-36. [PMID: 24078628 DOI: 10.1074/jbc.m113.517540] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Triggering receptor expressed on myeloid cells-2 (TREM2) and its signaling adaptor protein TYROBP/DAP12 play important roles in signal transduction in dendritic cells, osteoclasts, tissue macrophages, and microglia. Recently, TREM2 variants have been shown to be linked to late onset Alzheimer disease. Here, we demonstrate that TREM2 undergoes sequential proteolytic processing by ectodomain shedding and intramembrane proteolysis. The C-terminal fragment (CTF) of TREM2 generated by ectodomain shedding is cleaved by γ-secretase. Importantly, pharmacologic and genetic γ-secretase inhibition resulted in accumulation of TREM2 CTF at the plasma membrane that also interacts with the signaling adaptor protein DAP12. Thus, the accumulated TREM2 CTF thereby might limit the interaction of DAP12 with the functional full-length receptor, resulting in decreased DAP12 phosphorylation and impaired metabolism of phosphatidylinositol 4,5-bisphosphate. Together, these data demonstrate γ-secretase-mediated intramembranous proteolysis of TREM2 and functionally link two Alzheimer disease-associated proteins in one signaling pathway.
Collapse
Affiliation(s)
- Patrick Wunderlich
- From the Department of Neurology, University of Bonn, 53127 Bonn, Germany and
| | | | | | | | | | | |
Collapse
|
42
|
Guan X, Yang J, Gu H, Zou J, Yao Z. Immunotherapeutic efficiency of a tetravalent Aβ1-15 vaccine in APP/PS1 transgenic mice as mouse model for Alzheimer's disease. Hum Vaccin Immunother 2013; 9:1643-53. [PMID: 23732905 PMCID: PMC3906261 DOI: 10.4161/hv.24830] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 04/16/2013] [Accepted: 04/24/2013] [Indexed: 01/22/2023] Open
Abstract
Immunization with synthetic, preaggregated β-amyloid (Aβ) was the first treatment approach able to dramatically reduce brain Aβ pathology in Alzheimer's disease (AD) animal models. For the development of a safe vaccine, we investigated whether 4Aβ1-15 (four tandem repeats of GPGPG-linked Aβ1-15 sequences) had therapeutic effects in the APP/PS1 transgenic mice model of AD. We described the production of anti-Aβ antibodies in APP/PS1 mice immunized with 4Aβ1-15 mixed with MF59 adjuvant. The anti-Aβ antibody concentrations were increased which bound to AD plaques, markedly reduced Aβ pathology in transgenic AD mice and levels of intracerebral Aβ (soluble and insoluble), whereas increased serum Aβ levels. Immunization via 4Aβ1-15 (mainly of the IgG1 Class) may induce a non-inflammatory Th2 reaction. Immunohistochemistry analysis of MHC Class II and CD45 revealed that microglial cells were in a less activated state. Of note, 4Aβ1-15-immunized mice showed improved acquisition of memory compared with controls in a reference-memory Morris water-maze behavior test. The data identify the novel immunogen 4Aβ1-15 as a promising new tool for AD immunotherapy.
Collapse
Affiliation(s)
- Xiaoying Guan
- Department of Anatomy and Neurobiology; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou, PR China
| | - Junhua Yang
- Department of Anatomy and Neurobiology; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou, PR China
| | - Huaiyu Gu
- Department of Anatomy and Neurobiology; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou, PR China
| | - Juntao Zou
- Department of Anatomy and Neurobiology; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou, PR China
| | - Zhibin Yao
- Department of Anatomy and Neurobiology; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou, PR China
| |
Collapse
|
43
|
Kumar S, Wirths O, Theil S, Gerth J, Bayer TA, Walter J. Early intraneuronal accumulation and increased aggregation of phosphorylated Abeta in a mouse model of Alzheimer's disease. Acta Neuropathol 2013; 125:699-709. [PMID: 23525537 DOI: 10.1007/s00401-013-1107-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 03/06/2013] [Accepted: 03/09/2013] [Indexed: 12/13/2022]
Abstract
The progressive accumulation of extracellular amyloid plaques in the brain is a common hallmark of Alzheimer's disease (AD). We recently identified a novel species of Aβ phosphorylated at serine residue 8 with increased propensity to form toxic aggregates as compared to non-phosphorylated species. The age-dependent analysis of Aβ depositions using novel monoclonal phosphorylation-state specific antibodies revealed that phosphorylated Aβ variants accumulate first inside of neurons in a mouse model of AD already at 2 month of age. At higher ages, phosphorylated Aβ is also abundantly detected in extracellular plaques. Besides a large overlap in the spatiotemporal deposition of phosphorylated and non-phosphorylated Aβ species, fractionized extraction of Aβ from brains revealed an increased accumulation of phosphorylated Aβ in oligomeric assemblies as compared to non-phosphorylated Aβ in vivo. Thus, phosphorylated Aβ could represent an important species in the formation and stabilization of neurotoxic aggregates, and might be targeted for AD therapy and diagnosis.
Collapse
Affiliation(s)
- Sathish Kumar
- Department of Neurology, University of Bonn, 53127, Bonn, Germany
| | | | | | | | | | | |
Collapse
|
44
|
Presenilin 2 mutation accelerates the onset of impairment in trace eyeblink conditioning in a mouse model of Alzheimer's disease overexpressing human mutant amyloid precursor protein. Neurosci Lett 2013; 538:15-9. [PMID: 23370287 DOI: 10.1016/j.neulet.2013.01.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 01/06/2013] [Accepted: 01/16/2013] [Indexed: 01/16/2023]
Abstract
Missense mutations in 2 homologous genes, presenilin 1 (PS1) and presenilin 2 (PS2), cause dementia in a subset of early-onset familial Alzheimer's disease (FAD) pedigrees. The purpose of the present study was to investigate whether PS2 mutation accelerates the onset of trace eyeblink conditioning deficits in an Alzheimer's disease (AD) mouse model overexpressing human amyloid precursor protein (APP) with the Swedish mutation (K670N, M671L) (Tg2576 mice). For this purpose, a double-transgenic mouse (PS2Tg2576 mice) was produced by cross-breeding transgenic mice carrying human mutant PS2 (N141I) with Tg2576 mice. Long-trace interval (trace interval=500ms) eyeblink conditioning was tested in the PS2Tg2576 mice at ages 3, 4, 6, and 12 months. At 3 months, PS2Tg2576 mice exhibited normal acquisition of conditioned responses (CRs) during trace eyeblink conditioning, whereas trace conditioning was significantly impaired in PS2Tg2576 mice at ages 4, 6, and 12 months. In contrast, Tg2576 mice showed intact memory performance during trace conditioning at 4 months. This cross-sectional study clearly indicates that PS2 mutation significantly accelerates the onset of cognitive impairment in associative trace eyeblink memory.
Collapse
|
45
|
Wang XY, Tang SS, Hu M, Long Y, Li YQ, Liao MX, Ji H, Hong H. Leukotriene D4 induces amyloid-β generation via CysLT(1)R-mediated NF-κB pathways in primary neurons. Neurochem Int 2013; 62:340-7. [PMID: 23318673 DOI: 10.1016/j.neuint.2013.01.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/29/2012] [Accepted: 01/04/2013] [Indexed: 12/29/2022]
Abstract
Although the pathogenesis of sporadic Alzheimer's disease (AD) is not clearly understood, neuroinflammation has been known to play a role in the pathogenesis of AD. To investigate a functional link between the neuroinflammation and AD, the effect of leukotriene D4 (LTD4), an inflammatory lipid mediator, was studied on amyloid-β generation in vitro. Application of LTD4 to cell monolayers at concentrations up to 40 nM LTD4 caused increases in the Aβ releases. Concentrations ≥ 40 nM LTD4 decreased neuronal viability. Application of 20 nM LTD4 caused a significant increase in Aβ generation, as assessed by ELISA or Western blotting, without significant cytotoxicity. At this concentration, exposure of neurons to LTD4 for 24h produced maximal effect in the Aβ generation, and significant increases in the expressions of cysteinyl leukotriene 1 receptor (CysLT(1)R) and activity of β- or γ-secretase with complete abrogation by the selective CysLT(1)R antagonist pranlukast. Exposure of neurons to LTD4 for 1h showed activation of NF-κB pathway, by assessing the levels of p65 or phospho-p65 in the nucleus, and either CysLT(1)R antagonist pranlukast or NF-κB inhibitor PDTC prevented the nuclear translocation of p65 and the consequent phosphorylation. PDTC also inhibited LTD4-induced elevations of β- or γ-secretase activity and Aβ generation in vitro. Overall, our data show for the first time that LTD4 causes Aβ production by enhancement of β- or γ-secretase resulting from activation of CysLT(1)R-mediated NF-κB signaling pathway. These findings provide a novel pathologic link between neuroinflammation and AD.
Collapse
Affiliation(s)
- Xiao Yun Wang
- Department of Pharmacology, Laboratory for Diabetes & Brain Diseases, China Pharmaceutical University, Nanjing 210009, China
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Park HJ, Shabashvili D, Nekorchuk MD, Shyqyriu E, Jung JI, Ladd TB, Moore BD, Felsenstein KM, Golde TE, Kim SH. Retention in endoplasmic reticulum 1 (RER1) modulates amyloid-β (Aβ) production by altering trafficking of γ-secretase and amyloid precursor protein (APP). J Biol Chem 2012; 287:40629-40. [PMID: 23043097 PMCID: PMC3504776 DOI: 10.1074/jbc.m112.418442] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 10/05/2012] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Aβ production is influenced by intracellular trafficking of secretases and amyloid precursor protein (APP). RESULTS Retention in endoplasmic reticulum 1 (RER1) regulates the trafficking of γ-secretase and APP, thereby influences Aβ production. CONCLUSION RER1, an ER retention/retrieval factor for γ-secretase and APP, modulates Aβ production. SIGNIFICANCE RER1 and its influence on γ-secretase and APP may be implicated for a safe strategy to target Aβ production. The presence of neuritic plaques containing aggregated amyloid-β (Aβ) peptides in the brain parenchyma is a pathological hallmark of Alzheimer disease (AD). Aβ is generated by sequential cleavage of the amyloid β precursor protein (APP) by β- and γ-secretase, respectively. As APP processing to Aβ requires transport through the secretory pathway, trafficking of the substrate and access to the secretases are key factors that can influence Aβ production (Thinakaran, G., and Koo, E. H. (2008) Amyloid precursor protein trafficking, processing, and function. J. Biol. Chem. 283, 29615-29619). Here, we report that retention in endoplasmic reticulum 1 (RER1) associates with γ-secretase in early secretory compartments and regulates the intracellular trafficking of γ-secretase. RER1 overexpression decreases both γ-secretase localization on the cell surface and Aβ secretion and conversely RER1 knockdown increases the level of cell surface γ-secretase and increases Aβ secretion. Furthermore, we find that increased RER1 levels decrease mature APP and increase immature APP, resulting in less surface accumulation of APP. These data show that RER1 influences the trafficking and localization of both γ-secretase and APP, thereby regulating the production and secretion of Aβ peptides.
Collapse
Affiliation(s)
- Hyo-Jin Park
- From the Department of Pharmacology and Therapeutics, and
- the Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | | | | | - Eva Shyqyriu
- From the Department of Pharmacology and Therapeutics, and
| | - Joo In Jung
- the Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Thomas B. Ladd
- the Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Brenda D. Moore
- the Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Kevin M. Felsenstein
- the Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Todd E. Golde
- the Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Seong-Hun Kim
- From the Department of Pharmacology and Therapeutics, and
| |
Collapse
|
47
|
van Echten-Deckert G, Walter J. Sphingolipids: Critical players in Alzheimer’s disease. Prog Lipid Res 2012; 51:378-93. [DOI: 10.1016/j.plipres.2012.07.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 07/06/2012] [Indexed: 12/20/2022]
|
48
|
Cuartero Y, Mellado M, Capell A, Álvarez-Dolado M, Verges M. Retromer Regulates Postendocytic Sorting of β-Secretase in Polarized Madin-Darby Canine Kidney Cells. Traffic 2012; 13:1393-410. [DOI: 10.1111/j.1600-0854.2012.01392.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 06/28/2012] [Accepted: 07/03/2012] [Indexed: 01/24/2023]
Affiliation(s)
- Yasmina Cuartero
- Laboratory of Epithelial Cell Biology; Centro de Investigación Príncipe Felipe; Valencia; Spain
| | - Maravillas Mellado
- Laboratory of Epithelial Cell Biology; Centro de Investigación Príncipe Felipe; Valencia; Spain
| | - Anja Capell
- German Center for Neurodegenerative Diseases & Adolf Butenandt Institute - Biochemistry; Ludwig Maximilians University; Munich; Germany
| | - Manuel Álvarez-Dolado
- Department of Cell Therapy and Regenerative Medicine; Andalusian Center for Molecular Biology and Regenerative Medicine; Seville; Spain
| | | |
Collapse
|
49
|
Serum fatty-acid composition and the risk of Alzheimer's disease: a longitudinal population-based study. Eur J Clin Nutr 2012; 66:885-90. [PMID: 22713770 DOI: 10.1038/ejcn.2012.63] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND/OBJECTIVES It is unknown if a specific fatty-acid composition influences the development of Alzheimer's disease (AD). Nutrition is a possible target for prevention of dementia and especially omega-3-based fatty acids (n-3 FAs) have previously been suggested to be beneficial for cognition. The objective was to ascertain whether serum FAs predicts the risk of incident AD and dementia in a longitudinal population-based cohort. SUBJECTS/METHODS Uppsala Longitudinal Study of Adult Men started in 1970. The proportions of FAs in serum cholesteryl esters were estimated in men (n=2009) who were 50 years old at baseline. During a 35 year follow-up time, 213 men had developed dementia, out of which 91 AD. The associations were analyzed with Cox proportional hazards and logistic regression; adjusted for age, education and vascular risk factors. RESULTS Subjects with a higher proportion of saturated FAs had a decreased risk of AD in crude and multi-adjusted models (hazard ratio for 1-s.d. increase in palmitic acid 0.72; 95% confidence intervals: 0.59-0.89). These associations persisted even in the group of approximately 85-year-old survivors. n-3 FAs FAs were not associated with decreased risk of AD or dementia. CONCLUSIONS In contrast to experimental studies, saturated FAs were inversely associated with risk of AD. No evidence of a protective effect of n-3 FAs against dementia was found. The results remained essentially unchanged if competing risk from mortality was taken into account.
Collapse
|
50
|
Panza F, Frisardi V, Solfrizzi V, Imbimbo BP, Logroscino G, Santamato A, Greco A, Seripa D, Pilotto A. Immunotherapy for Alzheimer's disease: from anti-β-amyloid to tau-based immunization strategies. Immunotherapy 2012; 4:213-38. [PMID: 22339463 DOI: 10.2217/imt.11.170] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The exact mechanisms leading to Alzheimer's disease (AD) are largely unknown, limiting the identification of effective disease-modifying therapies. The two principal neuropathological hallmarks of AD are extracellular β-amyloid (Aβ), peptide deposition (senile plaques) and intracellular neurofibrillary tangles containing hyperphosphorylated tau protein. During the last decade, most of the efforts of the pharmaceutical industry were directed against the production and accumulation of Aβ. The most innovative of the pharmacological approaches was the stimulation of Aβ clearance from the brain of AD patients via the administration of Aβ antigens (active vaccination) or anti-Aβ antibodies (passive vaccination). Several active and passive anti-Aβ vaccines are under clinical investigation. Unfortunately, the first active vaccine (AN1792, consisting of preaggregate Aβ and an immune adjuvant, QS-21) was abandoned because it caused meningoencephalitis in approximately 6% of treated patients. Anti-Aβ monoclonal antibodies (bapineuzumab and solanezumab) are now being developed. The clinical results of the initial studies with bapineuzumab were equivocal in terms of cognitive benefit. The occurrence of vasogenic edema after bapineuzumab, and more rarely brain microhemorrhages (especially in Apo E ε4 carriers), has raised concerns on the safety of these antibodies directed against the N-terminus of the Aβ peptide. Solanezumab, a humanized anti-Aβ monoclonal antibody directed against the midregion of the Aβ peptide, was shown to neutralize soluble Aβ species. Phase II studies showed a good safety profile of solanezumab, while studies on cerebrospinal and plasma biomarkers documented good signals of pharmacodynamic activity. Although some studies suggested that active immunization may be effective against tau in animal models of AD, very few studies regarding passive immunization against tau protein are currently available. The results of the large, ongoing Phase III trials with bapineuzumab and solanezumab will tell us if monoclonal anti-Aβ antibodies may slow down the rate of deterioration of AD. Based on the new diagnostic criteria of AD and on recent major failures of anti-Aβ drugs in mild-to-moderate AD patients, one could argue that clinical trials on potential disease-modifying drugs, including immunological approaches, should be performed in the early stages of AD.
Collapse
Affiliation(s)
- Francesco Panza
- Geriatric Unit & Gerontology-Geriatric Research Laboratory, IRCCS Casa Sollievo della Sofferenza, Foggia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|