1
|
Lerose V, Ponticelli M, Benedetto N, Carlucci V, Lela L, Tzvetkov NT, Milella L. Withania somnifera (L.) Dunal, a Potential Source of Phytochemicals for Treating Neurodegenerative Diseases: A Systematic Review. PLANTS (BASEL, SWITZERLAND) 2024; 13:771. [PMID: 38592845 PMCID: PMC10976061 DOI: 10.3390/plants13060771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 04/11/2024]
Abstract
Withania somnifera (L.) Dunal is a medicinal plant belonging to the traditional Indian medical system, showing various therapeutic effects such as anti-cancer, anti-inflammatory, anti-microbial, anti-diabetic, and hepatoprotective activity. Of great interest is W. somnifera's potential beneficial effect against neurodegenerative diseases, since the authorized medicinal treatments can only delay disease progression and provide symptomatic relief and are not without side effects. A systematic search of PubMed and Scopus databases was performed to identify preclinical and clinical studies focusing on the applications of W. somnifera in preventing neurodegenerative diseases. Only English articles and those containing the keywords (Withania somnifera AND "neurodegenerative diseases", "neuroprotective effects", "Huntington", "Parkinson", "Alzheimer", "Amyotrophic Lateral Sclerosis", "neurological disorders") in the title or abstract were considered. Reviews, editorials, letters, meta-analyses, conference papers, short surveys, and book chapters were not considered. Selected articles were grouped by pathologies and summarized, considering the mechanism of action. The quality assessment and the risk of bias were performed using the Cochrane Handbook for Systematic Reviews of Interventions checklist. This review uses a systematic approach to summarize the results from 60 investigations to highlight the potential role of W. somnifera and its specialized metabolites in treating or preventing neurodegenerative diseases.
Collapse
Affiliation(s)
- Valentina Lerose
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (V.L.); (N.B.); (V.C.); (L.L.)
| | - Maria Ponticelli
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (V.L.); (N.B.); (V.C.); (L.L.)
| | - Nadia Benedetto
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (V.L.); (N.B.); (V.C.); (L.L.)
| | - Vittorio Carlucci
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (V.L.); (N.B.); (V.C.); (L.L.)
| | - Ludovica Lela
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (V.L.); (N.B.); (V.C.); (L.L.)
| | - Nikolay T. Tzvetkov
- Institute of Molecular Biology “Roumen Tsanev”, Department of Biochemical Pharmacology & Drug Design, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 21, 1113 Sofia, Bulgaria;
| | - Luigi Milella
- Department of Science, University of Basilicata, V.le Ateneo Lucano 10, 85100 Potenza, Italy; (V.L.); (N.B.); (V.C.); (L.L.)
| |
Collapse
|
2
|
Guo Y, Fan Z, Zhao S, Yu W, Hou X, Nie S, Xu S, Zhao C, Han J, Liu X. Brain-targeted lycopene-loaded microemulsion modulates neuroinflammation, oxidative stress, apoptosis and synaptic plasticity in β-amyloid-induced Alzheimer's disease mice. Neurol Res 2023:1-12. [PMID: 37068195 DOI: 10.1080/01616412.2023.2203615] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
OBJECTIVES β-Amyloid protein (Aβ) plays pivotal roles in pathogenesis of Alzheimer's disease (AD) and triggers various pathophysiological events. Lycopene is a promising neuroprotector with multiple bioactivities, while its bioavailability is limited. Lycopene-loaded microemulsion (LME) possessing superior bioavailability and brain-targeting efficiency was developed in our previous study. In this investigation, we aimed to comprehensively evaluate its neuroprotective effects and underlying mechanisms using intracerebroventricular (ICV) Aβ1-42 injection mice. METHODS Mice were assigned to the Sham, Aβ, Aβ + LME and Aβ + lycopene dissolved in olive oil (LOO) groups. ICV Aβ1-42 administration was performed, followed by oral gavage of brain-targeted LME or conventional LOO formulation for 3 weeks. Brain samples were harvested for immunohistochemistry, biochemical assays and western blotting analyses. RESULTS Our findings verified Aβ-induced neurotoxicity on neuroinflammation, oxidative stress, apoptosis, Aβ metabolisms and synaptic plasticity. LME supplementation dramatically attenuated astrocytosis and microgliosis, decreased malondialdehyde production and rescued antioxidant capacities, normalized apoptotic parameters and alleviated neuronal loss, inhibited amyloidogenic processing and activated non-amyloidogenic pathway, together with upregulating synaptic protein expressions and restoring synaptic plasticity. Nevertheless, most of these phenomena were not observed for mice treated with LOO, implying that LME showed significantly higher therapeutic efficacy against Aβ injury. DISCUSSION In summary, brain-targeted LME could exert neuroprotective function via suppressing a series of cascades triggered by Aβ aggregates, thus ameliorating Aβ neurotoxicity and associated abnormalities. Given this, LME may serve as an attractive candidate for AD prevention and treatment, and superiority of brain-targeting delivery is highlighted.
Collapse
Affiliation(s)
- Yunliang Guo
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
- Anti-aging Monitoring Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Zhongyu Fan
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
- Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Shuo Zhao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Wei Yu
- Department of Biochemistry and Molecular Biology, Jining Medical University, Jining, Shandong, PR China
| | - Xunyao Hou
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
- Anti-aging Monitoring Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Shanjing Nie
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
- Anti-aging Monitoring Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Song Xu
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
- Anti-aging Monitoring Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Cheng Zhao
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Junting Han
- Rehabilitation Ward II, Shandong Provincial Third Hospital, Jinan, Shandong, PR China
| | - Xueping Liu
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
- Anti-aging Monitoring Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| |
Collapse
|
3
|
Zhou G, Ye Q, Xu Y, He B, Wu L, Zhu G, Xie J, Yao L, Xiao Z. Mitochondrial calcium uptake 3 mitigates cerebral amyloid angiopathy-related neuronal death and glial inflammation by reducing mitochondrial dysfunction. Int Immunopharmacol 2023; 117:109614. [PMID: 36878048 DOI: 10.1016/j.intimp.2022.109614] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 03/06/2023]
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by the cerebrovascular amyloid-β (Aβ) accumulation, and always accompanied by Alzheimer's disease (AD). Mitochondrial dysfunction-associated cellular events including cell death, inflammation and oxidative stress are implicated in the progression of CAA. Unfortunately, the molecular mechanisms revealing CAA pathogenesis are still obscure, thus requiring further studies. Mitochondrial calcium uptake 3 (MICU3), a regulator of the mitochondrial Ca2+ uniporter (MCU), mediates various biological functions, but its expression and influence on CAA are largely unknown. In the present study, we found that MICU3 expression was gradually declined in cortex and hippocampus of Tg-SwDI transgenic mice. Using stereotaxic operation with AAV9 encoding MICU3, we showed that AAV-MICU3 improved the behavioral performances and cerebral blood flow (CBF) in Tg-SwDI mice, along with markedly reduced Aβ deposition through mediating Aβ metabolism process. Importantly, we found that AAV-MICU3 remarkably improved neuronal death and mitigated glial activation and neuroinflammation in cortex and hippocampus of Tg-SwDI mice. Furthermore, excessive oxidative stress, mitochondrial impairment and dysfunction, decreased ATP and mitochondrial DNA (mtDNA) were detected in Tg-SwDI mice, while being considerably ameliorated upon MICU3 over-expression. More importantly, our in vitro experiments suggested that MICU3-attenuated neuronal death, activation of glial cells and oxidative stress were completely abrogated upon PTEN induced putative kinase 1 (PINK1) knockdown, indicating that PINK1 was required for MICU3 to perform its protective effects against CAA. Mechanistic experiment confirmed an interaction between MICU3 and PINK1. Together, these findings demonstrated that MICU3-PINK1 axis may serve as a key target for CAA treatment mainly through improving mitochondrial dysfunction.
Collapse
Affiliation(s)
- Guijuan Zhou
- Department of Neurology, the First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan, PR China; Department of Rehabilitation Medicine, the First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan, PR China
| | - Qing Ye
- Department of Neurology, the First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan, PR China
| | - Yan Xu
- Department of Neurology, the First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan, PR China
| | - Bing He
- Department of Neurology, the First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan, PR China
| | - Lin Wu
- Department of Neurology, the First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan, PR China
| | - Guanghua Zhu
- Department of Neurology, the First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan, PR China
| | - Juan Xie
- Department of Neurology, the First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan, PR China
| | - Lan Yao
- Department of Neurology, the First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan, PR China
| | - Zijian Xiao
- Department of Neurology, the First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan, PR China.
| |
Collapse
|
4
|
Zhou G, Xiang T, Xu Y, He B, Wu L, Zhu G, Xie J, Yao L, Xiao Z. Fruquintinib/HMPL-013 ameliorates cognitive impairments and pathology in a mouse model of cerebral amyloid angiopathy (CAA). Eur J Pharmacol 2023; 939:175446. [PMID: 36470443 DOI: 10.1016/j.ejphar.2022.175446] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by the cerebrovascular amyloid-β (Aβ) accumulation, and always accompanied by Alzheimer's disease (AD). The mechanisms revealing CAA pathogenesis are still unclear, and it is challenging to develop an efficient therapeutic strategy for its treatment. Vascular endothelial growth factor (VEGF) and its receptors including VEGFR-1,-2,-3 activation are involved in Aβ processing, and modulate numerous cellular events associated with central nervous system (CNS) diseases. In the present study, we attempted to explore the regulatory function of fruquintinib (also named as HMPL-013), a highly selective inhibitor of VEGFR-1,-2,-3 tyrosine kinases, on CAA progression in Tg-SwDI mice. Here, we found that HMPL-013-rich diet consumption for 12 months significantly improved the behavioral performances and cerebral blood flow (CBF) of Tg-SwDI mice compared with the vehicle group. Importantly, HMPL-013 administration considerably reduced Aβ1-40 and Aβ1-42 burden in cortex and hippocampus of Tg-SwDI mice through regulating Aβ metabolism process. Congo red staining confirmed Aβ deposition in vessel walls, reflecting CAA formation, which was, however, strongly ameliorated after HMPL-013 treatment. Neuron death, aberrant glial activation and pro-inflammatory response in brain tissues of Tg-SwDI mice were dramatically alleviated after HMPL-013 consumption. More studies showed that the protective effects of HMPL-013 against CAA might be partially attributed to its regulation on the expression of genes associated with blood vasculature. Intriguingly, VEGF and phosphorylated VEGFR-1,-2 protein expression levels were remarkably decreased by HMPL-013 in cortex and hippocampus of Tg-SwDI mice, which were validated in HMPL-013-treated brain vascular endothelial cells (BVECs) under hypoxia. Finally, we found that VEGF-induced human umbilical vein endothelial cells (HUVEC) proliferation and tube formation were strongly abolished upon HMPL-013 exposure. Collectively, all these findings demonstrated that oral administration of HMPL-013 had therapeutic potential against CAA by reducing Aβ deposition, inflammation and neuron death via suppressing VEGF/VEGFR-1,-2 signaling.
Collapse
Affiliation(s)
- Guijuan Zhou
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China; Department of Rehabilitation Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Tao Xiang
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Yan Xu
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Bing He
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Lin Wu
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Guanghua Zhu
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Juan Xie
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Lan Yao
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China
| | - Zijian Xiao
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421099, Hunan Province, China.
| |
Collapse
|
5
|
Guanidine-based β amyloid precursor protein cleavage enzyme 1 (BACE-1) inhibitors for the Alzheimer's disease (AD): A review. Bioorg Med Chem 2022; 74:117047. [DOI: 10.1016/j.bmc.2022.117047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/16/2022] [Accepted: 10/04/2022] [Indexed: 11/02/2022]
|
6
|
Nopparat C, Boontor A, Panmanee J, Govitrapong P. Melatonin Attenuates Methamphetamine-Induced Alteration of Amyloid β Precursor Protein Cleaving Enzyme Expressions via Melatonin Receptor in Human Neuroblastoma Cells. Neurotox Res 2022; 40:1086-1095. [PMID: 35648367 DOI: 10.1007/s12640-022-00522-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/26/2022] [Accepted: 05/21/2022] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is the most prominent neurodegenerative disease represented by the loss of memory and cognitive impairment symptoms and is one of the major health imperilments among the elderly. Amyloid (Aβ) deposit inside the neuron is one of the characteristic pathological hallmarks of this disease, leading to neuronal cell death. In the amyloidogenic processing, the amyloid precursor protein (APP) is cleaved by beta-secretase and γ-secretase to generate Aβ. Methamphetamine (METH) is a psychostimulant drug that causes neurodegeneration and detrimental cognitive deficits. The analogy between the neurotoxic and neurodegenerative profile of METH and AD pathology necessitates an exploration of the underlying molecular mechanisms. In the present study, we found that METH ineluctably affects APP processing, which might contribute to the marked production of Aβ in human neuroblastoma cells. Melatonin, an indolamine produced and released by the pineal gland as well as other extrapineal, has been protective against METH-induced neurodegenerative processes, thus rescuing neuronal cell death. However, the precise action of melatonin on METH has yet to be determined. We further propose to investigate the protective properties of melatonin on METH-induced APP-cleaving secretases. Pretreatment with melatonin significantly reversed METH-induced APP-cleaving secretases and Aβ production. In addition, pretreatment with luzindole, a melatonin receptor antagonist, significantly prevented the protective effect of melatonin, suggesting that the attenuation of the toxic effect on METH-induced APP processing by melatonin was mediated via melatonin receptor. The present results suggested that melatonin has a beneficial role in preventing Aβ generation in a cellular model of METH-induced AD.
Collapse
Affiliation(s)
- Chutikorn Nopparat
- Innovative Learning Center, Srinakharinwirot University, Sukhumvit 23, Bangkok, 10110, Thailand
| | - Anuttree Boontor
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, 10210, Thailand
| | - Jiraporn Panmanee
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhonpathom, 73170, Thailand
| | - Piyarat Govitrapong
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok, 10210, Thailand.
| |
Collapse
|
7
|
Hsia HE, Tüshaus J, Feng X, Hofmann LI, Wefers B, Marciano DK, Wurst W, Lichtenthaler SF. Endoglycan (PODXL2) is proteolytically processed by ADAM10 (a disintegrin and metalloprotease 10) and controls neurite branching in primary neurons. FASEB J 2021; 35:e21813. [PMID: 34390512 DOI: 10.1096/fj.202100475r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/22/2021] [Accepted: 07/07/2021] [Indexed: 01/24/2023]
Abstract
Cell adhesion is tightly controlled in multicellular organisms, for example, through proteolytic ectodomain shedding of the adhesion-mediating cell surface transmembrane proteins. In the brain, shedding of cell adhesion proteins is required for nervous system development and function, but the shedding of only a few adhesion proteins has been studied in detail in the mammalian brain. One such adhesion protein is the transmembrane protein endoglycan (PODXL2), which belongs to the CD34-family of highly glycosylated sialomucins. Here, we demonstrate that endoglycan is broadly expressed in the developing mouse brains and is proteolytically shed in vitro in mouse neurons and in vivo in mouse brains. Endoglycan shedding in primary neurons was mediated by the transmembrane protease a disintegrin and metalloprotease 10 (ADAM10), but not by its homolog ADAM17. Functionally, endoglycan deficiency reduced the branching of neurites extending from primary neurons in vitro, whereas deletion of ADAM10 had the opposite effect and increased neurite branching. Taken together, our study discovers a function for endoglycan in neurite branching, establishes endoglycan as an ADAM10 substrate and suggests that ADAM10 cleavage of endoglycan may contribute to neurite branching.
Collapse
Affiliation(s)
- Hung-En Hsia
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Johanna Tüshaus
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Xiao Feng
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Laura I Hofmann
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Benedikt Wefers
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Developmental Genetics, Helmholtz Center Munich, Neuherberg/Munich, Germany
| | - Denise K Marciano
- Departments of Cell Biology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Developmental Genetics, Helmholtz Center Munich, Neuherberg/Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Technical University of Munich-Weihenstephan, Neuherberg/Munich, Neuherberg, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
8
|
Taketa TB, Mahl CRA, Calais GB, Beppu MM. Amino acid-functionalized chitosan beads for in vitro copper ions uptake in the presence of histidine. Int J Biol Macromol 2021; 188:421-431. [PMID: 34371051 DOI: 10.1016/j.ijbiomac.2021.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 11/16/2022]
Abstract
One of the hallmarks of Alzheimer's Disease (AD) is the anomalous binding involving amyloid-β (Aβ) peptide and metal ions, such as copper, formed through histidine (His) residues. Herein, adsorption experiments were performed to test the in vitro ability of chitosan to uptake copper ions in the presence of histidine. The characterization of the beads was assessed before and after the adsorption process by scanning electron microscope, X-ray diffraction and Fourier-transform infrared spectroscopy. Amino acid functionalization of chitosan-based beads promoted an increase in the copper ions adsorption capacity (2.47 mmol of Cu(II)/gram of adsorbent). Nevertheless, depending on the order of addition of histidine to the system, different adsorption behaviors were observed. The kinetics showed that, once the Cu(II)-His bond was established, functionalized beads were less efficient to capture Cu(II), which promoted a decrease in the overall adsorption capacity. However, when chitosan and histidine were simultaneously added to the Cu(II) solution, there was no decrease in adsorption capacity. To sum up, chitosan-based materials are an interesting model to provide a better understanding on the biomolecules‑copper interactions that occur in AD, as well as a possible chelating agent that can interfere in the bonds between Aβ residues and copper ions.
Collapse
Affiliation(s)
- Thiago B Taketa
- School of Chemical Engineering, University of Campinas, SP, Brazil
| | - Cynthia R A Mahl
- School of Chemical Engineering, University of Campinas, SP, Brazil
| | | | - Marisa M Beppu
- School of Chemical Engineering, University of Campinas, SP, Brazil.
| |
Collapse
|
9
|
Tobys D, Kowalski LM, Cziudaj E, Müller S, Zentis P, Pach E, Zigrino P, Blaeske T, Höning S. Inhibition of clathrin-mediated endocytosis by knockdown of AP-2 leads to alterations in the plasma membrane proteome. Traffic 2020; 22:6-22. [PMID: 33225555 DOI: 10.1111/tra.12770] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 01/05/2023]
Abstract
In eukaryotic cells, clathrin-mediated endocytosis (CME) is a central pathway for the internalization of proteins from the cell surface, thereby contributing to the maintenance of the plasma membrane protein composition. A key component for the formation of endocytic clathrin-coated vesicles (CCVs) is AP-2, as it sequesters cargo membrane proteins, recruits a multitude of other endocytic factors and initiates clathrin polymerization. Here, we inhibited CME by depletion of AP-2 and explored the consequences for the plasma membrane proteome. Quantitative analysis revealed accumulation of major constituents of the endosomal-lysosomal system reflecting a block in retrieval by compensatory CME. The noticeable enrichment of integrins and blockage of their turnover resulted in severely impaired cell migration. Rare proteins such as the anti-cancer drug target CA9 and tumor markers (CD73, CD164, CD302) were significantly enriched. The AP-2 knockdown attenuated the global endocytic capacity, but clathrin-independent entry pathways were still operating, as indicated by persistent internalization of specific membrane-spanning and GPI-anchored receptors (PVR, IGF1R, CD55, TNAP). We hypothesize that blocking AP-2 function and thus inhibiting CME may be a novel approach to identify new druggable targets, or to increase their residence time at the plasma membrane, thereby increasing the probability for efficient therapeutic intervention.
Collapse
Affiliation(s)
- David Tobys
- Institute for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Lisa Maria Kowalski
- Institute for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Eva Cziudaj
- Institute for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Stefan Müller
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Peter Zentis
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Elke Pach
- Department of Dermatology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Paola Zigrino
- Department of Dermatology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Tobias Blaeske
- Department of Plant Physiology and Biochemistry, University of Constance, Constance, Germany
| | - Stefan Höning
- Institute for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
10
|
Bibi N, Rizvi SMD, Batool A, Kamal MA. Inhibitory Mechanism of An Anticancer Drug, Bexarotene Against Amyloid β Peptide Aggregation: Repurposing Via Neuroinformatics Approach. Curr Pharm Des 2020; 25:2989-2995. [PMID: 31368868 DOI: 10.2174/1381612825666190801123235] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/23/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Aggregation of Amyloid β (Aβ) peptide is a crucial feature of Alzheimer disease (AD) pathogenesis. In fact, Aβ peptides are misfolded and aggregated to frame Amyloid fibrils, which is considered as one of the major contributing events in the onset of AD. All these observations have prompted the researchers to design therapeutic molecules with robust anti-Aβ aggregation potential. Interestingly, in the last few decades, drug repurposing has turned into a fruitful and savvy approach for the treatment of several diseases. Bexarotene is an anticancer drug that has been under consideration for its ability to suppress Aβ-peptide aggregation. However, the exact mechanistic aspect of suppression of Aβ-peptide accumulation has not yet been completely revealed. METHODS In the present study, we have attempted to decipher the mechanistic aspects of the anti-aggregation potential of bexarotene by using the computational biology approach. RESULTS We have observed the effect of 'Aβ-bexarotene' interaction on the aggregation ability of the Aβ-peptide and decoded the involvement of receptor for advanced glycation end products (RAGE) and beta-secretase (BACE-1). A deep structural analysis of Aβ upon binding with bexarotene revealed critical binding sites and structural twists involved in Aβ aggregation. It is evident from the present that bexarotene could significantly restrain the process of primary nucleation of Aβ. In addition, bexarotene showed a strong interaction with RAGE and BACE-1, suggesting them as plausible targets for the neuro-therapeutic action of bexarotene. CONCLUSION Hence, we could safely suggest that bexarotene is a potent drug candidate that could reduce Aβ- peptide aggregation by applying different mechanistic pathways. These results might boost the portfolio of pharmaceutical companies looking for the development of new chemical entities against AD.
Collapse
Affiliation(s)
- Nousheen Bibi
- Shaheed Benazir Bhutto Women University, Peshawar, Pakistan
| | - Syed M D Rizvi
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Abida Batool
- Shaheed Benazir Bhutto Women University, Peshawar, Pakistan
| | - Mohammad A Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia.,Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770, Australia.,Novel Global Community Educational Foundation, NSW, Australia
| |
Collapse
|
11
|
Zhang R, Liu Y, Chen Y, Li Q, Marshall C, Wu T, Hu G, Xiao M. Aquaporin 4 deletion exacerbates brain impairments in a mouse model of chronic sleep disruption. CNS Neurosci Ther 2019; 26:228-239. [PMID: 31364823 PMCID: PMC6978250 DOI: 10.1111/cns.13194] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 06/20/2019] [Accepted: 06/26/2019] [Indexed: 12/24/2022] Open
Abstract
AIMS As a normal physiological process, sleep has recently been shown to facilitate clearance of macromolecular metabolic wastes from the brain via the glymphatic system. The aim of the present study was to investigate pathophysiological roles of astroglial aquaporin 4 (AQP4), a functional regulator of glymphatic clearance, in a mouse model of chronic sleep disruption (SD). METHODS Adult AQP4 null mice and wild-type (WT) mice were given 7 days of SD using the improved rotating rod method, and then received behavioral, neuropathological, and neurochemical analyses. RESULTS Aquaporin 4 deletion resulted in an impairment of glymphatic transport and accumulation of β-amyloid and Tau proteins in the brain following SD. AQP4 null SD mice exhibited severe activation of microglia, neuroinflammation, and synaptic protein loss in the hippocampus, as well as decreased working memory, compared with WT-SD mice. CONCLUSION These results demonstrate that AQP4-mediated glymphatic clearance ameliorates brain impairments caused by abnormal accumulation of metabolic wastes following chronic SD, thus serving as a potential target for sleep-related disorders.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Yun Liu
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Yan Chen
- Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.,Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Li
- Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.,Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Charles Marshall
- Department of Rehabilitation Sciences, University of Kentucky Center of Excellence in Rural Health, Hazard, KY, USA
| | - Ting Wu
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Gang Hu
- Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Ming Xiao
- Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.,Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
Chalatsa I, Arvanitis DA, Koulakiotis NS, Giagini A, Skaltsounis AL, Papadopoulou-Daifoti Z, Tsarbopoulos A, Sanoudou D. The Crocus sativus Compounds trans-Crocin 4 and trans-Crocetin Modulate the Amyloidogenic Pathway and Tau Misprocessing in Alzheimer Disease Neuronal Cell Culture Models. Front Neurosci 2019; 13:249. [PMID: 30971876 PMCID: PMC6443833 DOI: 10.3389/fnins.2019.00249] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 03/04/2019] [Indexed: 11/23/2022] Open
Abstract
Crocus sativus L. natural compounds have been extensively used in traditional medicine for thousands of years. Recent research evidence is now emerging in support of its therapeutic potential for different pathologies including neurodegenerative diseases. Herein, the C. sativus L. natural compounds trans-crocin 4 and trans-crocetin were selected for in depth molecular characterization of their potentially protective effects against Alzheimer’s Disease (AD), utilizing two AD neuronal cell culture models (SH-SY5Y overexpressing APP and PC12 expressing hyperphosphorylated tau). Biologically relevant concentrations, ranging from 0.1 μM to 1 mM, applied for 24 h or 72 h, were well tolerated by differentiated wild type SH-SY5Y and PC12 cells. When tested on neuronally differentiated SH-SY5Y-APP both trans-crocin 4 and trans-crocetin had significant effects against amyloidogenic pathways. Trans-crocin 4 significantly decreased of β-secretase, a key enzyme of the amyloidogenic pathway, and APP-C99, while it decreased γ-secretases that generate toxic beta-amyloid peptides. Similarly, trans-crocetin treatment led to a reduction in β- and γ-secretases, as well as to accumulation of cellular AβPP. When tested on the neuronally differentiated PC12-htau cells, both compounds proved effective in suppressing the active forms of GSK3β and ERK1/2 kinases, as well as significantly reducing total tau and tau phosphorylation. Collectively, our data demonstrate a potent effect of trans-crocin 4 and trans-crocetin in suppressing key molecular pathways of AD pathogenesis, rendering them a promising tool in the prevention and potentially the treatment of AD.
Collapse
Affiliation(s)
- Ioanna Chalatsa
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Demetrios A Arvanitis
- Molecular Biology Division, Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | - Athina Giagini
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexios Leandros Skaltsounis
- Department of Pharmacognosy and Natural Product Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Zeta Papadopoulou-Daifoti
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anthony Tsarbopoulos
- GAIA Research Center, Bioanalytical Department, The Goulandris Natural History Museum, Athens, Greece.,Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Molecular Biology Division, Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece.,Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
13
|
Wang L, Zhang Y, Zhao Y, Marshall C, Wu T, Xiao M. Deep cervical lymph node ligation aggravates AD-like pathology of APP/PS1 mice. Brain Pathol 2018; 29:176-192. [PMID: 30192999 DOI: 10.1111/bpa.12656] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022] Open
Abstract
The imbalance between production and clearance of amyloid-beta (Aβ) is a key step in the onset and development of Alzheimer's disease (AD). Therefore, reducing Aβ accumulation in the brain is a promising therapeutic strategy for AD. The recently discovered glymphatic system and meningeal lymphatic vasculature have been shown to be critical for the elimination of interstitial waste products, especially Aβ, from the brain. In the present study, ligation of deep cervical lymph nodes was performed to block drainage of this system and explore the consequences on Aβ-related pathophysiology. Five-month-old APP/PS1 mice and their wild-type littermates received deep cervical lymphatic node ligation. One month later, behavioral testing and pathological analysis were conducted. Results demonstrated that ligation of dcLNs exacerbated AD-like phenotypes of APP/PS1 mice, showing more severe brain Aβ accumulation, neuroinflammation, synaptic protein loss, impaired polarization of aquaporin-4 and deficits in cognitive and exploratory behaviors. These results suggest that brain lymphatic clearance malfunction is one of the deteriorating factors in the progression of AD, and restoring its function is a potential therapeutic target against AD.
Collapse
Affiliation(s)
- Linmei Wang
- Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanli Zhang
- Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ying Zhao
- Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Charles Marshall
- Department of Rehabilitation Sciences, University of Kentucky Center of Excellence in Rural Health, Hazard, KY
| | - Ting Wu
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Ming Xiao
- Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
14
|
Murru L, Moretto E, Martano G, Passafaro M. Tetraspanins shape the synapse. Mol Cell Neurosci 2018; 91:76-81. [DOI: 10.1016/j.mcn.2018.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/29/2018] [Accepted: 04/01/2018] [Indexed: 01/01/2023] Open
|
15
|
Hesse R, von Einem B, Wagner F, Bott P, Schwanzar D, Jackson RJ, Föhr KJ, Lausser L, Kroker KS, Proepper C, Walther P, Kestler HA, Spires-Jones TL, Boeckers T, Rosenbrock H, von Arnim CAF. sAPPβ and sAPPα increase structural complexity and E/I input ratio in primary hippocampal neurons and alter Ca 2+ homeostasis and CREB1-signaling. Exp Neurol 2018; 304:1-13. [PMID: 29466703 DOI: 10.1016/j.expneurol.2018.02.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/09/2018] [Accepted: 02/14/2018] [Indexed: 12/23/2022]
Abstract
One major pathophysiological hallmark of Alzheimer's disease (AD) is senile plaques composed of amyloid β (Aβ). In the amyloidogenic pathway, cleavage of the amyloid precursor protein (APP) is shifted towards Aβ production and soluble APPβ (sAPPβ) levels. Aβ is known to impair synaptic function; however, much less is known about the physiological functions of sAPPβ. The neurotrophic properties of sAPPα, derived from the non-amyloidogenic pathway of APP cleavage, are well-established, whereas only a few, conflicting studies on sAPPβ exist. The intracellular pathways of sAPPβ are largely unknown. Since sAPPβ is generated alongside Aβ by β-secretase (BACE1) cleavage, we tested the hypothesis that sAPPβ effects differ from sAPPα effects as a neurotrophic factor. We therefore performed a head-to-head comparison of both mammalian recombinant peptides in developing primary hippocampal neurons (PHN). We found that sAPPα significantly increases axon length (p = 0.0002) and that both sAPPα and sAPPβ increase neurite number (p < 0.0001) of PHN at 7 days in culture (DIV7) but not at DIV4. Moreover, both sAPPα- and sAPPβ-treated neurons showed a higher neuritic complexity in Sholl analysis. The number of glutamatergic synapses (p < 0.0001), as well as layer thickness of postsynaptic densities (PSDs), were significantly increased, and GABAergic synapses decreased upon sAPP overexpression in PHN. Furthermore, we showed that sAPPα enhances ERK and CREB1 phosphorylation upon glutamate stimulation at DIV7, but not DIV4 or DIV14. These neurotrophic effects are further associated with increased glutamate sensitivity and CREB1-signaling. Finally, we found that sAPPα levels are significantly reduced in brain homogenates of AD patients compared to control subjects. Taken together, our data indicate critical stage-dependent roles of sAPPs in the developing glutamatergic system in vitro, which might help to understand deleterious consequences of altered APP shedding in AD patients, beyond Aβ pathophysiology.
Collapse
Affiliation(s)
- Raphael Hesse
- Department of Neurology, Ulm University, Ulm, Germany
| | | | | | - Patricia Bott
- Department of Neurology, Ulm University, Ulm, Germany
| | | | - Rosemary J Jackson
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | | | - Ludwig Lausser
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Katja S Kroker
- Boehringer Ingelheim Pharma GmbH & Co KG, Dept. of Drug Discovery Sciences, Biberach, Germany
| | | | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | | | - Tobias Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Holger Rosenbrock
- Boehringer Ingelheim Pharma GmbH & Co KG, Dept. of CNS Diseases Research, Biberach, Germany
| | | |
Collapse
|
16
|
Liu Y, He X, Li Y, Wang T. Cerebrospinal fluid CD4+ T lymphocyte-derived miRNA-let-7b can enhances the diagnostic performance of Alzheimer's disease biomarkers. Biochem Biophys Res Commun 2018; 495:1144-1150. [DOI: 10.1016/j.bbrc.2017.11.122] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 11/19/2017] [Indexed: 01/05/2023]
|
17
|
Klein T, Eckhard U, Dufour A, Solis N, Overall CM. Proteolytic Cleavage-Mechanisms, Function, and "Omic" Approaches for a Near-Ubiquitous Posttranslational Modification. Chem Rev 2017; 118:1137-1168. [PMID: 29265812 DOI: 10.1021/acs.chemrev.7b00120] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Proteases enzymatically hydrolyze peptide bonds in substrate proteins, resulting in a widespread, irreversible posttranslational modification of the protein's structure and biological function. Often regarded as a mere degradative mechanism in destruction of proteins or turnover in maintaining physiological homeostasis, recent research in the field of degradomics has led to the recognition of two main yet unexpected concepts. First, that targeted, limited proteolytic cleavage events by a wide repertoire of proteases are pivotal regulators of most, if not all, physiological and pathological processes. Second, an unexpected in vivo abundance of stable cleaved proteins revealed pervasive, functionally relevant protein processing in normal and diseased tissue-from 40 to 70% of proteins also occur in vivo as distinct stable proteoforms with undocumented N- or C-termini, meaning these proteoforms are stable functional cleavage products, most with unknown functional implications. In this Review, we discuss the structural biology aspects and mechanisms of catalysis by different protease classes. We also provide an overview of biological pathways that utilize specific proteolytic cleavage as a precision control mechanism in protein quality control, stability, localization, and maturation, as well as proteolytic cleavage as a mediator in signaling pathways. Lastly, we provide a comprehensive overview of analytical methods and approaches to study activity and substrates of proteolytic enzymes in relevant biological models, both historical and focusing on state of the art proteomics techniques in the field of degradomics research.
Collapse
Affiliation(s)
- Theo Klein
- Life Sciences Institute, Department of Oral Biological and Medical Sciences, and ‡Department of Biochemistry and Molecular Biology, University of British Columbia , Vancouver, British Columbia V6T 1Z4, Canada
| | - Ulrich Eckhard
- Life Sciences Institute, Department of Oral Biological and Medical Sciences, and ‡Department of Biochemistry and Molecular Biology, University of British Columbia , Vancouver, British Columbia V6T 1Z4, Canada
| | - Antoine Dufour
- Life Sciences Institute, Department of Oral Biological and Medical Sciences, and ‡Department of Biochemistry and Molecular Biology, University of British Columbia , Vancouver, British Columbia V6T 1Z4, Canada
| | - Nestor Solis
- Life Sciences Institute, Department of Oral Biological and Medical Sciences, and ‡Department of Biochemistry and Molecular Biology, University of British Columbia , Vancouver, British Columbia V6T 1Z4, Canada
| | - Christopher M Overall
- Life Sciences Institute, Department of Oral Biological and Medical Sciences, and ‡Department of Biochemistry and Molecular Biology, University of British Columbia , Vancouver, British Columbia V6T 1Z4, Canada
| |
Collapse
|
18
|
Liu CB, Wang R, Yi YF, Gao Z, Chen YZ. Lycopene mitigates β-amyloid induced inflammatory response and inhibits NF-κB signaling at the choroid plexus in early stages of Alzheimer's disease rats. J Nutr Biochem 2017; 53:66-71. [PMID: 29195132 DOI: 10.1016/j.jnutbio.2017.10.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 09/28/2017] [Accepted: 10/24/2017] [Indexed: 11/17/2022]
Abstract
The choroid plexus is able to modulate the cognitive function, through changes in the neuroinflammatory response and in brain immune surveillance. However, whether lycopene is involved in inflammatory responses at the choroid plexus in the early stages of Alzheimer's disease, and its molecular underpinnings are elusive. In this rat study, lycopene was used to investigate its protective effects on inflammation caused by β-amyloid. We characterized the learning and memory abilities, cytokine profiles of circulating TNF-α, IL-1β and IL-6β in the serum and the expressions of Toll like receptor 4 and nuclear factor-κB p65 mRNA and protein at the choroid plexus. The results showed that functional deficits of learning and memory in lycopene treatment groups were significantly improved compared to the control group without lycopene treatment in water maze test. The levels of serum TNF-α, IL-1β and IL-6β were significantly increased, and the expressions of TLR4 and NF-κB p65 mRNA and protein at the choroid plexus were up-regulated, indicating inflammation response was initiated following administration of Aβ1-42. After intragastric pretreatment with lycopene, inflammatory cytokines were significantly reduced and lycopene also reversed the Aβ1-42 induced up-regulation of TLR4 and NF-κB p65 mRNA and protein expressions at the choroid plexus. These results provided a novel evidence that lycopene significantly improved cognitive deficits and were accompanied by the attenuation of inflammatory injury via blocking the activation of NF-κB p65 and TLR4 expressions and production of cytokines, thereby endorsing its usefulness for diminishing β-amyloid deposition in the hippocampus tissues.
Collapse
Affiliation(s)
- Chong-Bin Liu
- Department of Physiology, Basic Medical Science Building, Huzhou University, 313000, Huzhou, People's Republic of China; Department of Physiology, Basic Medical Science College, Wenzhou Medical University, 325035, Wenzhou, People's Republic of China.
| | - Rui Wang
- Department of Physiology, Basic Medical Science Building, Huzhou University, 313000, Huzhou, People's Republic of China
| | - Yan-Feng Yi
- Department of Physiology, Basic Medical Science Building, Huzhou University, 313000, Huzhou, People's Republic of China
| | - Zhen Gao
- Department of Physiology, Basic Medical Science Building, Huzhou University, 313000, Huzhou, People's Republic of China
| | - Yi-Zhu Chen
- Central Blood Station of Huzhou City, 313020, Huzhou, People's Republic of China.
| |
Collapse
|
19
|
Chiarini A, Armato U, Liu D, Dal Prà I. Calcium-Sensing Receptor Antagonist NPS 2143 Restores Amyloid Precursor Protein Physiological Non-Amyloidogenic Processing in Aβ-Exposed Adult Human Astrocytes. Sci Rep 2017; 7:1277. [PMID: 28455519 PMCID: PMC5430644 DOI: 10.1038/s41598-017-01215-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 03/27/2017] [Indexed: 12/11/2022] Open
Abstract
Physiological non-amyloidogenic processing (NAP) of amyloid precursor holoprotein (hAPP) by α-secretases (e.g., ADAM10) extracellularly sheds neurotrophic/neuroprotective soluble (s)APPα and precludes amyloid-β peptides (Aβs) production via β-secretase amyloidogenic processing (AP). Evidence exists that Aβs interact with calcium-sensing receptors (CaSRs) in human astrocytes and neurons, driving the overrelease of toxic Aβ42/Aβ42-os (oligomers), which is completely blocked by CaSR antagonist (calcilytic) NPS 2143. Here, we investigated the mechanisms underlying NPS 2143 beneficial effects in human astrocytes. Moreover, because Alzheimer's disease (AD) involves neuroinflammation, we examined whether NPS 2143 remained beneficial when both fibrillary (f)Aβ25-35 and a microglial cytokine mixture (CMT) were present. Thus, hAPP NAP prevailed over AP in untreated astrocytes, which extracellularly shed all synthesized sAPPα while secreting basal Aβ40/42 amounts. Conversely, fAβ25-35 alone dramatically reduced sAPPα extracellular shedding while driving Aβ42/Aβ42-os oversecretion that CMT accelerated but not increased, despite a concurring hAPP overexpression. NPS 2143 promoted hAPP and ADAM10 translocation to the plasma membrane, thereby restoring sAPPα extracellular shedding and fully suppressing any Aβ42/Aβ42-os oversecretion, but left hAPP expression unaffected. Therefore, as anti-AD therapeutics calcilytics support neuronal viability by safeguarding astrocytes neurotrophic/neuroprotective sAPPα shedding, suppressing neurons and astrocytes Aβ42/Aβ42-os build-up/secretion, and remaining effective even under AD-typical neuroinflammatory conditions.
Collapse
Affiliation(s)
- Anna Chiarini
- Human Histology & Embryology Unit, Medical School, University of Verona, Verona, Venetia, Italy.
| | - Ubaldo Armato
- Human Histology & Embryology Unit, Medical School, University of Verona, Verona, Venetia, Italy
| | - Daisong Liu
- The Third Xiangya Hospital of Central South University, Department of Plastic Surgery, Changsha, Hunan, China
| | - Ilaria Dal Prà
- Human Histology & Embryology Unit, Medical School, University of Verona, Verona, Venetia, Italy.
| |
Collapse
|
20
|
Overexpression of MTERF4 promotes the amyloidogenic processing of APP by inhibiting ADAM10. Biochem Biophys Res Commun 2017; 482:928-934. [DOI: 10.1016/j.bbrc.2016.11.135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 11/24/2016] [Indexed: 12/21/2022]
|
21
|
Schröder B, Saftig P. Intramembrane proteolysis within lysosomes. Ageing Res Rev 2016; 32:51-64. [PMID: 27143694 DOI: 10.1016/j.arr.2016.04.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 04/01/2016] [Accepted: 04/26/2016] [Indexed: 11/26/2022]
Abstract
Regulated intramembrane proteolysis is of pivotal importance in a diverse set of developmental and physiological processes. Altered intramembrane substrate turnover may be associated with neurodegeneration, cancer and impaired immune function. In this review we will focus on the intramembrane proteases which have been localized in the lysosomal membrane. Members of the γ-secretase complex and γ-secretase activity are found in the lysosomal membrane and are discussed to contribute to intracellular amyloid β production. Mutant or deficient γ-secretase may cause disturbed lysosomal function. The signal peptide peptidase-like (SPPL) protease 2a is a lysosomal membrane component and cleaves CD74, the invariant chain of the MHC II complex, as well as FasL, TNF, ITM2B and TMEM106, type II transmembrane proteins involved in the regulation of immunity and neurodegeneration. Therefore, it can be concluded, that not only proteolysis within the lysosomal lumen but also within lysosomal membranes regulates important cellular functions and contributes essentially to proteostasis of membrane proteins what may become increasingly compromised in the aged individual.
Collapse
|
22
|
Alves S, Fol R, Cartier N. Gene Therapy Strategies for Alzheimer's Disease: An Overview. Hum Gene Ther 2016; 27:100-7. [PMID: 26838997 DOI: 10.1089/hum.2016.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Key neuropathological hallmarks of Alzheimer's disease (AD) are extracellular amyloid plaques and intracellular accumulation of hyperphosphorylated Tau protein. The mechanisms underlying these neuropathological changes remain unclear. So far, research on AD therapy has had limited success in terms of symptomatic treatments although it has also had several failures for disease-modifying drugs. Gene transfer strategies to the brain have contributed to evaluate in animal models many interesting tracks, some of which should deserve clinical applications in AD patients in the future.
Collapse
Affiliation(s)
- Sandro Alves
- INSERM U1169/MIRCen CEA 92265 Fontenay aux Roses and Université Paris-Sud, Université Paris-Saclay , Orsay, France
| | - Romain Fol
- INSERM U1169/MIRCen CEA 92265 Fontenay aux Roses and Université Paris-Sud, Université Paris-Saclay , Orsay, France
| | - Nathalie Cartier
- INSERM U1169/MIRCen CEA 92265 Fontenay aux Roses and Université Paris-Sud, Université Paris-Saclay , Orsay, France
| |
Collapse
|
23
|
Plummer S, Van den Heuvel C, Thornton E, Corrigan F, Cappai R. The Neuroprotective Properties of the Amyloid Precursor Protein Following Traumatic Brain Injury. Aging Dis 2016; 7:163-79. [PMID: 27114849 PMCID: PMC4809608 DOI: 10.14336/ad.2015.0907] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 09/07/2015] [Indexed: 01/16/2023] Open
Abstract
Despite the significant health and economic burden that traumatic brain injury (TBI) places on society, the development of successful therapeutic agents have to date not translated into efficacious therapies in human clinical trials. Injury to the brain is ongoing after TBI, through a complex cascade of primary and secondary injury events, providing a valuable window of opportunity to help limit and prevent some of the severe consequences with a timely treatment. Of note, it has been suggested that novel treatments for TBI should be multifactorial in nature, mimicking the body's own endogenous repair response. Whilst research has historically focused on the role of the amyloid precursor protein (APP) in the pathogenesis of Alzheimer's disease, recent advances in trauma research have demonstrated that APP offers considerable neuroprotective properties following TBI, suggesting that APP is an ideal therapeutic candidate. Its acute upregulation following TBI has been shown to serve a beneficial role following trauma and has lead to significant advances in understanding the neuroprotective and neurotrophic functions of APP and its metabolites. Research has focused predominantly on the APP derivative sAPPα, which has consistently demonstrated neuroprotective and neurotrophic functions both in vitro and in vivo following various traumatic insults. Its neuroprotective activity has been narrowed down to a 15 amino acid sequence, and this region is linked to both heparan binding and growth-factor-like properties. It has been proposed that APP binds to heparan sulfate proteoglycans to exert its neuroprotective action. APP presents us with a novel therapeutic compound that could overcome many of the challenges that have stalled development of efficacious TBI treatments previously.
Collapse
Affiliation(s)
- Stephanie Plummer
- Adelaide Centre for Neuroscience Research, the University of Adelaide, South Australia, Australia
| | - Corinna Van den Heuvel
- Adelaide Centre for Neuroscience Research, the University of Adelaide, South Australia, Australia
| | - Emma Thornton
- Adelaide Centre for Neuroscience Research, the University of Adelaide, South Australia, Australia
| | - Frances Corrigan
- Adelaide Centre for Neuroscience Research, the University of Adelaide, South Australia, Australia
| | - Roberto Cappai
- Department of Pathology, the University of Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Fol R, Braudeau J, Ludewig S, Abel T, Weyer SW, Roederer JP, Brod F, Audrain M, Bemelmans AP, Buchholz CJ, Korte M, Cartier N, Müller UC. Viral gene transfer of APPsα rescues synaptic failure in an Alzheimer's disease mouse model. Acta Neuropathol 2016; 131:247-266. [PMID: 26538149 DOI: 10.1007/s00401-015-1498-9] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/07/2015] [Accepted: 10/15/2015] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is characterized by synaptic failure, dendritic and axonal atrophy, neuronal death and progressive loss of cognitive functions. It is commonly assumed that these deficits arise due to β-amyloid accumulation and plaque deposition. However, increasing evidence indicates that loss of physiological APP functions mediated predominantly by neurotrophic APPsα produced in the non-amyloidogenic α-secretase pathway may contribute to AD pathogenesis. Upregulation of APPsα production via induction of α-secretase might, however, be problematic as this may also affect substrates implicated in tumorigenesis. Here, we used a gene therapy approach to directly overexpress APPsα in the brain using AAV-mediated gene transfer and explored its potential to rescue structural, electrophysiological and behavioral deficits in APP/PS1∆E9 AD model mice. Sustained APPsα overexpression in aged mice with already preexisting pathology and amyloidosis restored synaptic plasticity and partially rescued spine density deficits. Importantly, AAV-APPsα treatment also resulted in a functional rescue of spatial reference memory in the Morris water maze. Moreover, we demonstrate a significant reduction of soluble Aβ species and plaque load. In addition, APPsα induced the recruitment of microglia with a ramified morphology into the vicinity of plaques and upregulated IDE and TREM2 expression suggesting enhanced plaque clearance. Collectively, these data indicate that APPsα can mitigate synaptic and cognitive deficits, despite established pathology. Increasing APPsα may therefore be of therapeutic relevance for AD.
Collapse
Affiliation(s)
- Romain Fol
- INSERM U1169/MIRCen CEA, 92265, Fontenay aux Roses, France
- University Paris Sud, University Paris-Saclay, 91400, Orsay, France
- Université Paris Descartes, 75006, Paris, France
| | - Jerome Braudeau
- INSERM U1169/MIRCen CEA, 92265, Fontenay aux Roses, France
- University Paris Sud, University Paris-Saclay, 91400, Orsay, France
| | - Susann Ludewig
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Brunswick, Germany
| | - Tobias Abel
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | - Sascha W Weyer
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120, Heidelberg, Germany
| | - Jan-Peter Roederer
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120, Heidelberg, Germany
| | - Florian Brod
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | - Mickael Audrain
- INSERM U1169/MIRCen CEA, 92265, Fontenay aux Roses, France
- University Paris Sud, University Paris-Saclay, 91400, Orsay, France
- Université Paris Descartes, 75006, Paris, France
| | - Alexis-Pierre Bemelmans
- University Paris Sud, University Paris-Saclay, 91400, Orsay, France
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Département des Sciences du Vivant (DSV), Institut d'Imagerie Biomédicale (I2BM), Molecular Imaging Research Center (MIRCen), 92260, Fontenay aux Roses, France
- Centre National de la Recherche Scientifique (CNRS), UMR 9199, Neurodegenerative Diseases Laboratory, 92260, Fontenay aux Roses, France
| | - Christian J Buchholz
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225, Langen, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Brunswick, Germany
- Helmholtz Centre for Infection Research, AG NIND, Inhoffenstr. 7, 38124, Brunswick, Germany
| | - Nathalie Cartier
- INSERM U1169/MIRCen CEA, 92265, Fontenay aux Roses, France.
- University Paris Sud, University Paris-Saclay, 91400, Orsay, France.
| | - Ulrike C Müller
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120, Heidelberg, Germany.
| |
Collapse
|
25
|
Kumalo HM, Soliman ME. A comparative molecular dynamics study on BACE1 and BACE2 flap flexibility. J Recept Signal Transduct Res 2016; 36:505-14. [PMID: 26804314 DOI: 10.3109/10799893.2015.1130058] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Beta-amyloid precursor protein cleavage enzyme1 (BACE1) and beta-amyloid precursor protein cleavage enzyme2 (BACE2), members of aspartyl protease family, are close homologs and have high similarity in their protein crystal structures. However, their enzymatic properties are different, which leads to different clinical outcomes. In this study, we performed sequence analysis and all-atom molecular dynamic (MD) simulations for both enzymes in their ligand-free states in order to compare their dynamical flap behaviors. This is to enhance our understanding of the relationship between sequence, structure and the dynamics of this protein family. Sequence analysis shows that in BACE1 and BACE2, most of the ligand-binding sites are conserved, indicative of their enzymatic property as aspartyl protease members. The other conserved residues are more or less unsystematically localized throughout the structure. Herein, we proposed and applied different combined parameters to define the asymmetric flap motion; the distance, d1, between the flap tip and the flexible region; the dihedral angle, φ, to account for the twisting motion and the TriCα angle, θ2 and θ1. All four combined parameters were found to appropriately define the observed "twisting" motion during the flaps different conformational states. Additional analysis of the parameters indicated that the flaps can exist in an ensemble of conformations, i.e. closed, semi-open and open conformations for both systems. However, the behavior of the flap tips during simulations is different between BACE1 and BACE2. The BACE1 active site cavity is more spacious as compared to that of BACE2. The analysis of 10S loop and 113S loop showed a similar trend to that of flaps, with the BACE1 loops being more flexible and less stable than those of BACE2. We believe that the results, methods and perspectives highlighted in this report would assist researchers in the discovery of BACE inhibitors as potential Alzheimer's disease therapies.
Collapse
Affiliation(s)
- H M Kumalo
- a Molecular Modelling & Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal , Westville , Durban , South Africa
| | - Mahmoud E Soliman
- a Molecular Modelling & Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal , Westville , Durban , South Africa
| |
Collapse
|
26
|
Puzzo D, Gulisano W, Arancio O, Palmeri A. The keystone of Alzheimer pathogenesis might be sought in Aβ physiology. Neuroscience 2015; 307:26-36. [PMID: 26314631 PMCID: PMC4591241 DOI: 10.1016/j.neuroscience.2015.08.039] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/15/2015] [Accepted: 08/18/2015] [Indexed: 01/17/2023]
Abstract
For several years Amyloid-beta peptide (Aβ) has been considered the main pathogenetic factor of Alzheimer's disease (AD). According to the so called Amyloid Cascade Hypothesis the increase of Aβ triggers a series of events leading to synaptic dysfunction and memory loss as well as to the structural brain damage in the later stage of the disease. However, several evidences suggest that this hypothesis is not sufficient to explain AD pathogenesis, especially considering that most of the clinical trials aimed to decrease Aβ levels have been unsuccessful. Moreover, Aβ is physiologically produced in the healthy brain during neuronal activity and it is needed for synaptic plasticity and memory. Here we propose a model interpreting AD pathogenesis as an alteration of the negative feedback loop between Aβ and its physiological receptors, focusing on alpha7 nicotinic acetylcholine receptors (α7-nAchRs). According to this vision, when Aβ cannot exert its physiological function a negative feedback mechanism would induce a compensatory increase of its production leading to an abnormal accumulation that reduces α7-nAchR function, leading to synaptic dysfunction and memory loss. In this perspective, the indiscriminate Aβ removal might worsen neuronal homeostasis, causing a further impoverishment of learning and memory. Even if further studies are needed to better understand and validate these mechanisms, we believe that to deepen the role of Aβ in physiological conditions might represent the keystone to elucidate important aspects of AD pathogenesis.
Collapse
Affiliation(s)
- D Puzzo
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, Viale A. Doria 6 (ed. 2), University of Catania, Catania 95125, Italy.
| | - W Gulisano
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, Viale A. Doria 6 (ed. 2), University of Catania, Catania 95125, Italy
| | - O Arancio
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, 630 West 168th Street, Columbia University, New York, NY 10032, USA
| | - A Palmeri
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, Viale A. Doria 6 (ed. 2), University of Catania, Catania 95125, Italy
| |
Collapse
|
27
|
Conant K, Allen M, Lim ST. Activity dependent CAM cleavage and neurotransmission. Front Cell Neurosci 2015; 9:305. [PMID: 26321910 PMCID: PMC4531370 DOI: 10.3389/fncel.2015.00305] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/27/2015] [Indexed: 12/13/2022] Open
Abstract
Spatially localized proteolysis represents an elegant means by which neuronal activity dependent changes in synaptic structure, and thus experience dependent learning and memory, can be achieved. In vitro and in vivo studies suggest that matrix metalloproteinase and adamalysin activity is concentrated at the cell surface, and emerging evidence suggests that increased peri-synaptic expression, release and/or activation of these proteinases occurs with enhanced excitatory neurotransmission. Synaptically expressed cell adhesion molecules (CAMs) could therefore represent important targets for neuronal activity-dependent proteolysis. Several CAM subtypes are expressed at the synapse, and their cleavage can influence the efficacy of synaptic transmission through a variety of non-mutually exclusive mechanisms. In the following review, we discuss mechanisms that regulate neuronal activity-dependent synaptic CAM shedding, including those that may be calcium dependent. We also highlight CAM targets of activity-dependent proteolysis including neuroligin and intercellular adhesion molecule-5 (ICAM-5). We include discussion focused on potential consequences of synaptic CAM shedding, with an emphasis on interactions between soluble CAM cleavage products and specific pre- and post-synaptic receptors.
Collapse
Affiliation(s)
- Katherine Conant
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| | - Megan Allen
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| | - Seung T Lim
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| |
Collapse
|
28
|
Abstract
Proteases regulate a myriad of cell functions, both in normal and disease states. In addition to protein turnover, they regulate a range of signaling processes, including those mediated by Eph receptors and their ephrin ligands. A variety of proteases is reported to directly cleave Ephs and/or ephrins under different conditions, to promote receptor and/or ligand shedding, and regulate receptor/ligand internalisation and signaling. They also cleave other adhesion proteins in response to Eph-ephrin interactions, to indirectly facilitate Eph-mediated functions. Proteases thus contribute to Eph/ephrin mediated changes in cell-cell and cell-matrix interactions, in cell morphology and in cell migration and invasion, in a manner which appears to be tightly regulated by, and co-ordinated with, Eph signaling. This review summarizes the current literature describing the function and regulation of protease activities during Eph/ephrin-mediated cell signaling.
Collapse
Affiliation(s)
- Lakmali Atapattu
- a Department of Biochemistry and Molecular Biology ; Monash University , Victoria ; Australia
| | | | | |
Collapse
|
29
|
Tetrahydroxystilbene glucoside extends mouse life span via upregulating neural klotho and downregulating neural insulin or insulin-like growth factor 1. Neurobiol Aging 2015; 36:1462-70. [DOI: 10.1016/j.neurobiolaging.2014.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 10/26/2014] [Accepted: 11/04/2014] [Indexed: 02/08/2023]
|
30
|
Hick M, Herrmann U, Weyer SW, Mallm JP, Tschäpe JA, Borgers M, Mercken M, Roth FC, Draguhn A, Slomianka L, Wolfer DP, Korte M, Müller UC. Acute function of secreted amyloid precursor protein fragment APPsα in synaptic plasticity. Acta Neuropathol 2015; 129:21-37. [PMID: 25432317 DOI: 10.1007/s00401-014-1368-x] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 11/11/2014] [Accepted: 11/12/2014] [Indexed: 11/24/2022]
Abstract
The key role of APP in the pathogenesis of Alzheimer disease is well established. However, postnatal lethality of double knockout mice has so far precluded the analysis of the physiological functions of APP and the APLPs in the brain. Previously, APP family proteins have been implicated in synaptic adhesion, and analysis of the neuromuscular junction of constitutive APP/APLP2 mutant mice showed deficits in synaptic morphology and neuromuscular transmission. Here, we generated animals with a conditional APP/APLP2 double knockout (cDKO) in excitatory forebrain neurons using NexCre mice. Electrophysiological recordings of adult NexCre cDKOs indicated a strong synaptic phenotype with pronounced deficits in the induction and maintenance of hippocampal LTP and impairments in paired pulse facilitation, indicating a possible presynaptic deficit. These deficits were also reflected in impairments in nesting behavior and hippocampus-dependent learning and memory tasks, including deficits in Morris water maze and radial maze performance. Moreover, while no gross alterations of brain morphology were detectable in NexCre cDKO mice, quantitative analysis of adult hippocampal CA1 neurons revealed prominent reductions in total neurite length, dendritic branching, reduced spine density and reduced spine head volume. Strikingly, the impairment of LTP could be selectively rescued by acute application of exogenous recombinant APPsα, but not APPsβ, indicating a crucial role for APPsα to support synaptic plasticity of mature hippocampal synapses on a rapid time scale. Collectively, our analysis reveals an essential role of APP family proteins in excitatory principal neurons for mediating normal dendritic architecture, spine density and morphology, synaptic plasticity and cognition.
Collapse
Affiliation(s)
- Meike Hick
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kovacs GG, Adle-Biassette H, Milenkovic I, Cipriani S, van Scheppingen J, Aronica E. Linking pathways in the developing and aging brain with neurodegeneration. Neuroscience 2014; 269:152-72. [PMID: 24699227 DOI: 10.1016/j.neuroscience.2014.03.045] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 03/21/2014] [Accepted: 03/21/2014] [Indexed: 12/12/2022]
Abstract
The molecular and cellular mechanisms, which coordinate the critical stages of brain development to reach a normal structural organization with appropriate networks, are progressively being elucidated. Experimental and clinical studies provide evidence of the occurrence of developmental alterations induced by genetic or environmental factors leading to the formation of aberrant networks associated with learning disabilities. Moreover, evidence is accumulating that suggests that also late-onset neurological disorders, even Alzheimer's disease, might be considered disorders of aberrant neural development with pathological changes that are set up at early stages of development before the appearance of the symptoms. Thus, evaluating proteins and pathways that are important in age-related neurodegeneration in the developing brain together with the characterization of mechanisms important during brain development with relevance to brain aging are of crucial importance. In the present review we focus on (1) aspects of neurogenesis with relevance to aging; (2) neurodegenerative disease (NDD)-associated proteins/pathways in the developing brain; and (3) further pathways of the developing or neurodegenerating brains that show commonalities. Elucidation of complex pathogenetic routes characterizing the earliest stage of the detrimental processes that result in pathological aging represents an essential first step toward a therapeutic intervention which is able to reverse these pathological processes and prevent the onset of the disease. Based on the shared features between pathways, we conclude that prevention of NDDs of the elderly might begin during the fetal and childhood life by providing the mothers and their children a healthy environment for the fetal and childhood development.
Collapse
Affiliation(s)
- G G Kovacs
- Institute of Neurology, Medical University of Vienna, Austria.
| | - H Adle-Biassette
- Inserm U1141, F-75019 Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMRS 676, F-75019 Paris, France; Department of Pathology, Lariboisière Hospital, APHP, Paris, France
| | - I Milenkovic
- Institute of Neurology, Medical University of Vienna, Austria
| | | | - J van Scheppingen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, The Netherlands
| | - E Aronica
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, The Netherlands; SEIN - Stichting Epilepsie Instellingen Nederland, Heemstede, The Netherlands; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, The Netherlands
| |
Collapse
|
32
|
Weyer SW, Zagrebelsky M, Herrmann U, Hick M, Ganss L, Gobbert J, Gruber M, Altmann C, Korte M, Deller T, Müller UC. Comparative analysis of single and combined APP/APLP knockouts reveals reduced spine density in APP-KO mice that is prevented by APPsα expression. Acta Neuropathol Commun 2014; 2:36. [PMID: 24684730 PMCID: PMC4023627 DOI: 10.1186/2051-5960-2-36] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 03/07/2014] [Indexed: 11/21/2022] Open
Abstract
Synaptic dysfunction and synapse loss are key features of Alzheimer's pathogenesis. Previously, we showed an essential function of APP and APLP2 for synaptic plasticity, learning and memory. Here, we used organotypic hippocampal cultures to investigate the specific role(s) of APP family members and their fragments for dendritic complexity and spine formation of principal neurons within the hippocampus. Whereas CA1 neurons from APLP1-KO or APLP2-KO mice showed normal neuronal morphology and spine density, APP-KO mice revealed a highly reduced dendritic complexity in mid-apical dendrites. Despite unaltered morphology of APLP2-KO neurons, combined APP/APLP2-DKO mutants showed an additional branching defect in proximal apical dendrites, indicating redundancy and a combined function of APP and APLP2 for dendritic architecture. Remarkably, APP-KO neurons showed a pronounced decrease in spine density and reductions in the number of mushroom spines. No further decrease in spine density, however, was detectable in APP/APLP2-DKO mice. Mechanistically, using APPsα-KI mice lacking transmembrane APP and expressing solely the secreted APPsα fragment we demonstrate that APPsα expression alone is sufficient to prevent the defects in spine density observed in APP-KO mice. Collectively, these studies reveal a combined role of APP and APLP2 for dendritic architecture and a unique function of secreted APPs for spine density.
Collapse
Affiliation(s)
- Sascha W Weyer
- Department of Bioinformatics and Functional Genomics, Ruprecht-Karls University Heidelberg, Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, Heidelberg D-69120, Germany
| | - Marta Zagrebelsky
- TU Braunschweig, Zoological Institute, Cellular Neurobiology, Spielmannstr. 7, Braunschweig D-38106, Germany
| | - Ulrike Herrmann
- TU Braunschweig, Zoological Institute, Cellular Neurobiology, Spielmannstr. 7, Braunschweig D-38106, Germany
| | - Meike Hick
- Department of Bioinformatics and Functional Genomics, Ruprecht-Karls University Heidelberg, Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, Heidelberg D-69120, Germany
| | - Lennard Ganss
- Department of Bioinformatics and Functional Genomics, Ruprecht-Karls University Heidelberg, Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, Heidelberg D-69120, Germany
- Present address: Department of Applied Tumor Biology, Ruprecht-Karls University Heidelberg, Institute of Pathology, University of Heidelberg, Heidelberg D-69120, Germany
| | - Julia Gobbert
- Department of Bioinformatics and Functional Genomics, Ruprecht-Karls University Heidelberg, Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, Heidelberg D-69120, Germany
| | - Morna Gruber
- Goethe University Frankfurt, Institute of Clinical Neuroanatomy, Neuroscience Center, Theodor-Stern-Kai 7, Frankfurt am Main D-60596, Germany
| | - Christine Altmann
- Goethe University Frankfurt, Institute of Clinical Neuroanatomy, Neuroscience Center, Theodor-Stern-Kai 7, Frankfurt am Main D-60596, Germany
| | - Martin Korte
- TU Braunschweig, Zoological Institute, Cellular Neurobiology, Spielmannstr. 7, Braunschweig D-38106, Germany
| | - Thomas Deller
- Goethe University Frankfurt, Institute of Clinical Neuroanatomy, Neuroscience Center, Theodor-Stern-Kai 7, Frankfurt am Main D-60596, Germany
| | - Ulrike C Müller
- Department of Bioinformatics and Functional Genomics, Ruprecht-Karls University Heidelberg, Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, Heidelberg D-69120, Germany
| |
Collapse
|
33
|
Becker-Pauly C, Broder C, Prox J, Koudelka T, Tholey A. Mapping orphan proteases by proteomics: Meprin metalloproteases deciphered as potential therapeutic targets. Proteomics Clin Appl 2014; 8:382-8. [DOI: 10.1002/prca.201300079] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 10/14/2013] [Accepted: 11/18/2013] [Indexed: 01/02/2023]
Affiliation(s)
- Christoph Becker-Pauly
- Institute of Biochemistry; Unit for Degradomics of the Protease Web; University of Kiel; Kiel Germany
| | - Claudia Broder
- Institute of Biochemistry; Unit for Degradomics of the Protease Web; University of Kiel; Kiel Germany
| | - Johannes Prox
- Institute of Biochemistry; Unit for Degradomics of the Protease Web; University of Kiel; Kiel Germany
| | - Tomas Koudelka
- AG Systematic Proteome Research & Bioanalytics - Institute for Experimental Medicine; Christian-Albrechts-Universität; Kiel Germany
| | - Andreas Tholey
- AG Systematic Proteome Research & Bioanalytics - Institute for Experimental Medicine; Christian-Albrechts-Universität; Kiel Germany
| |
Collapse
|
34
|
Altmeppen HC, Prox J, Puig B, Dohler F, Falker C, Krasemann S, Glatzel M. Roles of endoproteolytic α-cleavage and shedding of the prion protein in neurodegeneration. FEBS J 2013; 280:4338-47. [DOI: 10.1111/febs.12196] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/25/2013] [Accepted: 02/14/2013] [Indexed: 12/11/2022]
Affiliation(s)
- Hermann C. Altmeppen
- Institute of Neuropathology; University Medical Center HH-Eppendorf; Hamburg Germany
| | - Johannes Prox
- Institute of Biochemistry; Christian Albrechts University; Kiel Germany
| | - Berta Puig
- Institute of Neuropathology; University Medical Center HH-Eppendorf; Hamburg Germany
| | - Frank Dohler
- Institute of Neuropathology; University Medical Center HH-Eppendorf; Hamburg Germany
| | - Clemens Falker
- Institute of Neuropathology; University Medical Center HH-Eppendorf; Hamburg Germany
| | - Susanne Krasemann
- Institute of Neuropathology; University Medical Center HH-Eppendorf; Hamburg Germany
| | - Markus Glatzel
- Institute of Neuropathology; University Medical Center HH-Eppendorf; Hamburg Germany
| |
Collapse
|
35
|
Watt B, van Niel G, Raposo G, Marks MS. PMEL: a pigment cell-specific model for functional amyloid formation. Pigment Cell Melanoma Res 2013; 26:300-15. [PMID: 23350640 DOI: 10.1111/pcmr.12067] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 01/15/2013] [Indexed: 12/15/2022]
Abstract
PMEL is a pigment cell-specific protein responsible for the formation of fibrillar sheets within the pigment organelle, the melanosome. The fibrillar sheets serve as a template upon which melanins polymerize as they are synthesized. The PMEL fibrils are required for optimal pigment cell function, as animals that either lack PMEL expression or express mutant PMEL variants show varying degrees of hypopigmentation and pigment cell inviability. The PMEL fibrils have biophysical properties of amyloid, a protein fold that is frequently associated with neurodegenerative and other diseases. However, PMEL is one of a growing number of non-pathogenic amyloid proteins that contribute to the function of the cell and/or organism that produces them. Understanding how PMEL generates amyloid in a non-pathogenic manner might provide insights into how to avoid toxicity due to pathological amyloid formation. In this review, we summarize and reconcile data concerning the fate of PMEL from its site of synthesis in the endoplasmic reticulum to newly formed melanosomes and the role of distinct PMEL subdomains in trafficking and amyloid fibril formation. We then discuss how its progression through the secretory pathway into the endosomal system might allow for the regulated and non-toxic conversion of PMEL into an ordered amyloid polymer.
Collapse
Affiliation(s)
- Brenda Watt
- Department of Pathology and Laboratory Medicine, Department of Physiology, and Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
36
|
Anukulthanakorn K, Malaivijitnond S, Kitahashi T, Jaroenporn S, Parhar I. Molecular events during the induction of neurodegeneration and memory loss in estrogen-deficient rats. Gen Comp Endocrinol 2013; 181:316-23. [PMID: 23036734 DOI: 10.1016/j.ygcen.2012.07.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 07/25/2012] [Indexed: 12/23/2022]
Abstract
This study aims to delineate the relationship among estrogen deficiency, neurodegeneration, and cognitive impairment of ovariectomized rats. Female Sprague-Dawley rats were ovariectomized and euthanized after 1-4 month periods (M(0)-M(4) groups). Blood samples were collected for the determination of serum levels of 17β-estradiol (E(2)), luteinizing hormone (LH), and follicle stimulating hormone (FSH). Five consecutive days before the euthanization, cognitive performance of the rats was examined by Morris water maze test. After euthanization, the hippocampus was collected, and expression of the genes associated with amyloid plaques (App, Adam10 and Bace1) and neurofibrillary tangles (Tau4 and Tau3) were examined by real-time PCR. Serum E(2) levels were declined following 2 weeks of ovariectomy. Conversely, serum FSH and LH levels were profoundly increased by 2 weeks of ovariectomy for approximately 4 and 22 times, respectively. Cognitive impairments, indicated by the longer latency and distance, were observed only in the M(3) and M(4) groups. The Tau4 mRNA levels were significantly increased as early as 1 month after ovariectomy (in the M(1) group; P<0.05), and tended to be increased further with the advancing time. Similarly, the Tau3 mRNA levels were increased by ovariectomy, but with the highest level in the M(1) group, and decreased thereafter. The mRNA levels of App, Adam10 and Bace1 were increased by ovariectomy, but significant differences were observed only in the M(4) group. These results indicate that estrogen deficiency can induce a sequence of events that results in the production of neurofibrillary tangles, amyloid deposition, and spatial memory deficit in rats.
Collapse
Affiliation(s)
- Kanya Anukulthanakorn
- Biological Science Program, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | | | | | | | | |
Collapse
|
37
|
Ortega F, Stott J, Visser SAG, Bendtsen C. Interplay between α-, β-, and γ-secretases determines biphasic amyloid-β protein level in the presence of a γ-secretase inhibitor. J Biol Chem 2012; 288:785-92. [PMID: 23152503 PMCID: PMC3543028 DOI: 10.1074/jbc.m112.419135] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Amyloid-β (Aβ) is produced by the consecutive cleavage of amyloid precursor protein (APP) first by β-secretase, generating C99, and then by γ-secretase. APP is also cleaved by α-secretase. It is hypothesized that reducing the production of Aβ in the brain may slow the progression of Alzheimer disease. Therefore, different γ-secretase inhibitors have been developed to reduce Aβ production. Paradoxically, it has been shown that low to moderate inhibitor concentrations cause a rise in Aβ production in different cell lines, in different animal models, and also in humans. A mechanistic understanding of the Aβ rise remains elusive. Here, a minimal mathematical model has been developed that quantitatively describes the Aβ dynamics in cell lines that exhibit the rise as well as in cell lines that do not. The model includes steps of APP processing through both the so-called amyloidogenic pathway and the so-called non-amyloidogenic pathway. It is shown that the cross-talk between these two pathways accounts for the increase in Aβ production in response to inhibitor, i.e. an increase in C99 will inhibit the non-amyloidogenic pathway, redirecting APP to be cleaved by β-secretase, leading to an additional increase in C99 that overcomes the loss in γ-secretase activity. With a minor extension, the model also describes plasma Aβ profiles observed in humans upon dosing with a γ-secretase inhibitor. In conclusion, this mechanistic model rationalizes a series of experimental results that spans from in vitro to in vivo and to humans. This has important implications for the development of drugs targeting Aβ production in Alzheimer disease.
Collapse
Affiliation(s)
- Fernando Ortega
- Computational Biology, Discovery Sciences, AstraZeneca, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | | | | | | |
Collapse
|
38
|
Abstract
Clathrin is considered the prototype vesicle coat protein whose self-assembly mediates sorting of membrane cargo and recruitment of lipid modifiers. Detailed knowledge of clathrin biochemistry, structure, and interacting proteins has accumulated since the first observation, almost 50 years ago, of its role in receptor-mediated endocytosis of yolk protein. This review summarizes that knowledge, and focuses on properties of the clathrin heavy and light chain subunits and interaction of the latter with Hip proteins, to address the diversity of clathrin function beyond conventional receptor-mediated endocytosis. The distinct functions of the two human clathrin isoforms (CHC17 and CHC22) are discussed, highlighting CHC22's specialized involvement in traffic of the GLUT4 glucose transporter and consequent role in human glucose metabolism. Analysis of clathrin light chain function and interaction with the actin-binding Hip proteins during bacterial infection defines a novel actin-organizing function for CHC17 clathrin. By considering these diverse clathrin functions, along with intracellular sorting roles and influences on mitosis, further relevance of clathrin function to human health and disease is established.
Collapse
Affiliation(s)
- Frances M Brodsky
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94143-0552, USA.
| |
Collapse
|
39
|
Mittendorf KF, Deatherage CL, Ohi MD, Sanders CR. Tailoring of membrane proteins by alternative splicing of pre-mRNA. Biochemistry 2012; 51:5541-56. [PMID: 22708632 DOI: 10.1021/bi3007065] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Alternative splicing (AS) of RNA is a key mechanism for diversification of the eukaryotic proteome. In this process, different mRNA transcripts can be produced through altered excision and/or inclusion of exons during processing of the pre-mRNA molecule. Since its discovery, AS has been shown to play roles in protein structure, function, and localization. Dysregulation of this process can result in disease phenotypes. Moreover, AS pathways are promising therapeutic targets for a number of diseases. Integral membrane proteins (MPs) represent a class of proteins that may be particularly amenable to regulation by alternative splicing because of the distinctive topological restraints associated with their folding, structure, trafficking, and function. Here, we review the impact of AS on MP form and function and the roles of AS in MP-related disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Kathleen F Mittendorf
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | |
Collapse
|
40
|
Schreiber A, Fischer S, Lang T. The amyloid precursor protein forms plasmalemmal clusters via its pathogenic amyloid-β domain. Biophys J 2012; 102:1411-7. [PMID: 22455924 DOI: 10.1016/j.bpj.2012.02.031] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 02/17/2012] [Accepted: 02/21/2012] [Indexed: 11/26/2022] Open
Abstract
The amyloid precursor protein (APP) is a large, ubiquitous integral membrane protein with a small amyloid-β (Aβ) domain. In the human brain, endosomal processing of APP produces neurotoxic Aβ-peptides, which are involved in Alzheimer's disease. Here, we show that the Aβ sequence exerts a physiological function when still present in the unprocessed APP molecule. From the extracellular site, Aβ concentrates APP molecules into plasmalemmal membrane protein clusters. Moreover, Aβ stabilization of clusters is a prerequisite for their targeting to endocytic clathrin structures. Therefore, we conclude that the Aβ domain directly mediates a central step in APP trafficking, driving its own conversion into neurotoxic peptides.
Collapse
Affiliation(s)
- Arne Schreiber
- Department of Membrane Biochemistry, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | | | | |
Collapse
|