51
|
Edmison D, Wang L, Gowrishankar S. Lysosome Function and Dysfunction in Hereditary Spastic Paraplegias. Brain Sci 2021; 11:152. [PMID: 33498913 PMCID: PMC7911997 DOI: 10.3390/brainsci11020152] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/15/2021] [Accepted: 01/21/2021] [Indexed: 12/13/2022] Open
Abstract
Hereditary Spastic Paraplegias (HSPs) are a genetically diverse group of inherited neurological diseases with over 80 associated gene loci. Over the last decade, research into mechanisms underlying HSPs has led to an emerging interest in lysosome dysfunction. In this review, we highlight the different classes of HSPs that have been linked to lysosome defects: (1) a subset of complex HSPs where mutations in lysosomal genes are causally linked to the diseases, (2) other complex HSPs where mutation in genes encoding membrane trafficking adaptors lead to lysosomal defects, and (3) a subset of HSPs where mutations affect genes encoding proteins whose function is primarily linked to a different cellular component or organelle such as microtubule severing and Endoplasmic Reticulum-shaping, while also altering to lysosomes. Interestingly, aberrant axonal lysosomes, associated with the latter two subsets of HSPs, are a key feature observed in other neurodegenerative diseases such as Alzheimer's disease. We discuss how altered lysosome function and trafficking may be a critical contributor to HSP pathology and highlight the need for examining these features in the cortico-spinal motor neurons of HSP mutant models.
Collapse
Affiliation(s)
| | | | - Swetha Gowrishankar
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (D.E.); (L.W.)
| |
Collapse
|
52
|
Syeda T, Cannon JR. Environmental exposures and the etiopathogenesis of Alzheimer's disease: The potential role of BACE1 as a critical neurotoxic target. J Biochem Mol Toxicol 2021; 35:e22694. [PMID: 33393683 DOI: 10.1002/jbt.22694] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a major public health crisis due to devastating cognitive symptoms, a lack of curative treatments, and increasing prevalence. Most cases are sporadic (>95% of cases) after the age of 65 years, implicating an important role of environmental factors in disease pathogenesis. Environmental neurotoxicants have been implicated in neurodegenerative disorders including Parkinson's Disease and AD. Animal models of AD and in vitro studies have shed light on potential neuropathological mechanisms, yet the biochemical and molecular underpinnings of AD-relevant environmental neurotoxicity remain poorly understood. Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) is a potentially critical pathogenic target of environmentally induced neurotoxicity. BACE1 clearly has a critical role in AD pathophysiology: It is required for amyloid beta production and expression and activity of BACE1 are increased in the AD brain. Though the literature on BACE1 in response to environmental insults is limited, current studies, along with extensive AD neurobiology literature suggest that BACE1 deserves attention as an important neurotoxic target. Here, we critically review research on environmental neurotoxicants such as metals, pesticides, herbicides, fungicides, polyfluoroalkyl substances, heterocyclic aromatic amines, advanced glycation end products, and acrolein that modulate BACE1 and potential mechanisms of action. Though more research is needed to clearly understand whether BACE1 is a critical mediator of AD-relevant neurotoxicity, available reports provide convincing evidence that BACE1 is altered by environmental risk factors associated with AD pathology, implying that BACE1 inhibition and its use as a biomarker should be considered in AD management and research.
Collapse
Affiliation(s)
- Tauqeerunnisa Syeda
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA.,Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, Indiana, USA
| | - Jason R Cannon
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA.,Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
53
|
Subramanian J, Savage JC, Tremblay MÈ. Synaptic Loss in Alzheimer's Disease: Mechanistic Insights Provided by Two-Photon in vivo Imaging of Transgenic Mouse Models. Front Cell Neurosci 2020; 14:592607. [PMID: 33408613 PMCID: PMC7780885 DOI: 10.3389/fncel.2020.592607] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/25/2020] [Indexed: 01/05/2023] Open
Abstract
Synapse loss is the strongest correlate for cognitive decline in Alzheimer's disease. The mechanisms underlying synapse loss have been extensively investigated using mouse models expressing genes with human familial Alzheimer's disease mutations. In this review, we summarize how multiphoton in vivo imaging has improved our understanding of synapse loss mechanisms associated with excessive amyloid in the living animal brain. We also discuss evidence obtained from these imaging studies for the role of cell-intrinsic calcium dyshomeostasis and cell-extrinsic activities of microglia, which are the immune cells of the brain, in mediating synapse loss.
Collapse
Affiliation(s)
- Jaichandar Subramanian
- Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS, United States
| | - Julie C Savage
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada.,Department of Molecular Medicine, Université Laval, Québec City, QC, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.,Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
54
|
Chronic BACE-1 Inhibitor Administration in TASTPM Mice (APP KM670/671NL and PSEN1 M146V Mutation): An EEG Study. Int J Mol Sci 2020; 21:ijms21239072. [PMID: 33260655 PMCID: PMC7730584 DOI: 10.3390/ijms21239072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/16/2020] [Accepted: 11/24/2020] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE In this exploratory study, we tested whether electroencephalographic (EEG) rhythms may reflect the effects of a chronic administration (4 weeks) of an anti-amyloid β-site amyloid precursor protein (APP) cleaving enzyme 1 inhibitor (BACE-1; ER-901356; Eisai Co., Ltd., Tokyo, Japan) in TASTPM (double mutation in APP KM670/671NL and PSEN1 M146V) producing Alzheimer's disease (AD) amyloid neuropathology as compared to wild type (WT) mice. METHODS Ongoing EEG rhythms were recorded from a bipolar frontoparietal and two monopolar frontomedial (prelimbic) and hippocampal channels in 11 WT Vehicle, 10 WT BACE-1, 10 TASTPM Vehicle, and 11 TASTPM BACE-1 mice (males; aged 8/9 months old at the beginning of treatment). Normalized EEG power (density) was compared between the first day (Day 0) and after 4 weeks (Week 4) of the BACE-1 inhibitor (10 mg/Kg) or vehicle administration in the 4 mouse groups. Frequency and magnitude of individual EEG delta and theta frequency peaks (IDF and ITF) were considered during animal conditions of behaviorally passive and active wakefulness. Cognitive status was not tested. RESULTS Compared with the WT group, the TASTPM group generally showed a significantly lower reactivity in frontoparietal ITF power during the active over the passive condition (p < 0.05). Notably, there was no other statistically significant effect (e.g., additional electrodes, recording time, and BACE-1 inhibitor). CONCLUSIONS The above EEG biomarkers reflected differences between the WT and TASTPM groups, but no BACE-1 inhibitor effect. The results suggest an enhanced experimental design with the use of younger mice, longer drug administrations, an effective control drug, and neuropathological amyloid markers.
Collapse
|
55
|
Sun BL, Chen Y, Fan DY, Zhu C, Zeng F, Wang YJ. Critical thinking on amyloid-beta-targeted therapy: challenges and perspectives. SCIENCE CHINA-LIFE SCIENCES 2020; 64:926-937. [DOI: 10.1007/s11427-020-1810-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/28/2020] [Indexed: 01/02/2023]
|
56
|
Hampel H, Lista S, Vanmechelen E, Zetterberg H, Giorgi FS, Galgani A, Blennow K, Caraci F, Das B, Yan R, Vergallo A. β-Secretase1 biological markers for Alzheimer's disease: state-of-art of validation and qualification. Alzheimers Res Ther 2020; 12:130. [PMID: 33066807 PMCID: PMC7566058 DOI: 10.1186/s13195-020-00686-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/15/2020] [Indexed: 01/09/2023]
Abstract
β-Secretase1 (BACE1) protein concentrations and rates of enzyme activity, analyzed in human bodily fluids, are promising candidate biological markers for guidance in clinical trials investigating BACE1 inhibitors to halt or delay the dysregulation of the amyloid-β pathway in Alzheimer's disease (AD). A robust body of evidence demonstrates an association between cerebrospinal fluid/blood BACE1 biomarkers and core pathophysiological mechanisms of AD, such as brain protein misfolding and aggregration, neurodegeneration, and synaptic dysfunction.In pharmacological trials, BACE1 candidate biomarkers may be applied to a wide set of contexts of use (CoU), including proof of mechanism, dose-finding, response and toxicity dose estimation. For clinical CoU, BACE1 biomarkers show good performance for prognosis and disease prediction.The roadmap toward validation and qualification of BACE1 biomarkers requires standardized pre-analytical and analytical protocols to reduce inter-site variance that may have contributed to inconsistent results.BACE1 biomarker-drug co-development programs, including biomarker-guided outcomes and endpoints, may support the identification of sub-populations with a higher probability to benefit from BACE1 inhibitors with a reduced risk of adverse effects, in line with the evolving precision medicine paradigm.
Collapse
Affiliation(s)
- Harald Hampel
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Simone Lista
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France
- Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, F-75013, Paris, France
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Filippo Sean Giorgi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessandro Galgani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Filippo Caraci
- Department of Drug Sciences, University of Catania, Catania, Italy
- Oasi Research Institute-IRCCS, Troina, Italy
| | - Brati Das
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Andrea Vergallo
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France.
| |
Collapse
|
57
|
Yao Y, Nzou G, Alle T, Tsering W, Maimaiti S, Trojanowski JQ, Lee VMY, Ballatore C, Brunden KR. Correction of microtubule defects within Aβ plaque-associated dystrophic axons results in lowered Aβ release and plaque deposition. Alzheimers Dement 2020; 16:1345-1357. [PMID: 32918367 DOI: 10.1002/alz.12144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 06/16/2020] [Indexed: 11/09/2022]
Abstract
The hallmark pathologies of the Alzheimer's disease (AD) brain are amyloid beta (Aβ)-containing senile plaques and neurofibrillary tangles formed from the microtubule (MT)-binding tau protein. Tau becomes hyperphosphorylated and disengages from MTs in AD, with evidence of resulting MT structure/function defects. Brain-penetrant MT-stabilizing compounds can normalize MTs and axonal transport in mouse models with tau pathology, thereby reducing neuron loss and decreasing tau pathology. MT dysfunction is also observed in dystrophic axons adjacent to Aβ plaques, resulting in accumulation of amyloid precursor protein (APP) and BACE1 with the potential for enhanced localized Aβ generation. We have examined whether the brain-penetrant MT-stabilizing compound CNDR-51657 might decrease plaque-associated axonal dystrophy and Aβ release in 5XFAD mice that develop an abundance of Aβ plaques. Administration of CNDR-51657 to 1.5-month-old male and female 5XFAD mice for 4 or 7 weeks led to decreased soluble brain Aβ that coincided with reduced APP and BACE1 levels, resulting in decreased formation of insoluble Aβ deposits. These data suggest a vicious cycle whereby initial Aβ plaque formation causes MT disruption in nearby axons, resulting in the local accumulation of APP and BACE1 that facilitates additional Aβ generation and plaque deposition. The ability of a MT-stabilizing compound to attenuate this cycle, and also reduce deficits resulting from reduced tau binding to MTs, suggests that molecules of this type hold promise as potential AD therapeutics.
Collapse
Affiliation(s)
- Yuemang Yao
- Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Goodwell Nzou
- Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Thibault Alle
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
| | - Wangchen Tsering
- Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shaniya Maimaiti
- Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Virginia M-Y Lee
- Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Carlo Ballatore
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California, USA
| | - Kurt R Brunden
- Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
58
|
Pinheiro L, Faustino C. Therapeutic Strategies Targeting Amyloid-β in Alzheimer's Disease. Curr Alzheimer Res 2020; 16:418-452. [PMID: 30907320 DOI: 10.2174/1567205016666190321163438] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/16/2019] [Accepted: 03/17/2019] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder linked to protein misfolding and aggregation. AD is pathologically characterized by senile plaques formed by extracellular Amyloid-β (Aβ) peptide and Intracellular Neurofibrillary Tangles (NFT) formed by hyperphosphorylated tau protein. Extensive synaptic loss and neuronal degeneration are responsible for memory impairment, cognitive decline and behavioral dysfunctions typical of AD. Amyloidosis has been implicated in the depression of acetylcholine synthesis and release, overactivation of N-methyl-D-aspartate (NMDA) receptors and increased intracellular calcium levels that result in excitotoxic neuronal degeneration. Current drugs used in AD treatment are either cholinesterase inhibitors or NMDA receptor antagonists; however, they provide only symptomatic relief and do not alter the progression of the disease. Aβ is the product of Amyloid Precursor Protein (APP) processing after successive cleavage by β- and γ-secretases while APP proteolysis by α-secretase results in non-amyloidogenic products. According to the amyloid cascade hypothesis, Aβ dyshomeostasis results in the accumulation and aggregation of Aβ into soluble oligomers and insoluble fibrils. The former are synaptotoxic and can induce tau hyperphosphorylation while the latter deposit in senile plaques and elicit proinflammatory responses, contributing to oxidative stress, neuronal degeneration and neuroinflammation. Aβ-protein-targeted therapeutic strategies are thus a promising disease-modifying approach for the treatment and prevention of AD. This review summarizes recent findings on Aβ-protein targeted AD drugs, including β-secretase inhibitors, γ-secretase inhibitors and modulators, α-secretase activators, direct inhibitors of Aβ aggregation and immunotherapy targeting Aβ, focusing mainly on those currently under clinical trials.
Collapse
Affiliation(s)
- Lídia Pinheiro
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto 1649-003 Lisboa, Portugal
| | - Célia Faustino
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto 1649-003 Lisboa, Portugal
| |
Collapse
|
59
|
Lozupone M, Solfrizzi V, D'Urso F, Di Gioia I, Sardone R, Dibello V, Stallone R, Liguori A, Ciritella C, Daniele A, Bellomo A, Seripa D, Panza F. Anti-amyloid-β protein agents for the treatment of Alzheimer's disease: an update on emerging drugs. Expert Opin Emerg Drugs 2020; 25:319-335. [PMID: 32772738 DOI: 10.1080/14728214.2020.1808621] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Currently available Alzheimer's disease (AD) therapeutics are only symptomatic, targeting cholinergic and glutamatergic neurotransmissions. Several putative disease-modifying drugs in late-stage clinical development target amyloid-β (Aβ) peptide and tau protein, the principal neurophatological hallmarks of the disease. AREAS COVERED Phase III randomized clinical trials of anti-Aβ drugs for AD treatment were searched in US and EU clinical trial registries and principal biomedical databases until May 2020. EXPERT OPINION At present, compounds in Phase III clinical development for AD include four anti-Ab monoclonal antibodies (solanezumab, gantenerumab, aducanumab, BAN2401), the combination of cromolyn sodium and ibuprofen (ALZT-OP1), and two small molecules (levetiracetam, GV-971). These drugs are mainly being tested in subjects during early AD phases or at preclinical stage of familial AD or even in asymptomatic subjects at high risk of developing AD. The actual results support the hypothesis that elevated Aβ represents an early stage in the AD continuum and demonstrate the feasibility of enrolling these high-risk participants in secondary prevention trials to slow cognitive decline during the AD preclinical stages. However, a series of clinical failures may question further development of Aβ-targeting drugs and the findings from current ongoing Phase III trials will hopefully give light to this critical issue.
Collapse
Affiliation(s)
- Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro , Bari, Italy
| | - Vincenzo Solfrizzi
- "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro" , Bari, Italy
| | - Francesca D'Urso
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia , Foggia, Italy
| | - Ilaria Di Gioia
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia , Foggia, Italy
| | - Rodolfo Sardone
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| | - Vittorio Dibello
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy.,Department of Orofacial Pain and Dysfunction, Academic Centre of Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , The Netherlands
| | - Roberta Stallone
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| | - Angelo Liguori
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| | - Chiara Ciritella
- Physical and Rehabilitation Medicine Department, University of Foggia , Foggia, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart , Rome, Italy.,Institute of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia , Foggia, Italy
| | - Davide Seripa
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo Della Sofferenza , Foggia, Italy.,Hematology and Stem Cell Transplant Unit, Vito Fazzi Hospital, ASL Lecce , Lecce, Italy
| | - Francesco Panza
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| |
Collapse
|
60
|
Ovsepian SV, Horacek J, O'Leary VB, Hoschl C. The Ups and Downs of BACE1: Walking a Fine Line between Neurocognitive and Other Psychiatric Symptoms of Alzheimer's Disease. Neuroscientist 2020; 27:222-234. [PMID: 32713260 DOI: 10.1177/1073858420940943] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although neurocognitive deficit is the best-recognized indicator of Alzheimer's disease (AD), psychotic and other noncognitive symptoms are the prime cause of institutionalization. BACE1 is the rate-limiting enzyme in the production of Aβ of AD, and one of the promising therapeutic targets in countering cognitive decline and amyloid pathology. Changes in BACE1 activity have also emerged to cause significant noncognitive neuropsychiatric symptoms and impairments of circadian rhythms, as evident from clinical trials and reports in transgenic models. In this study, we consider key characteristics of BACE1 with its contribution to neurocognitive deficit and other psychiatric symptoms of AD. We argue that a growing list of noncognitive mental impairments related to pharmacological modulation of BACE1 might present a major obstacle in clinical translation of emerging therapeutic leads targeting this protease. The adverse effects of BACE1 inhibition on mental health call for a revision of treatment strategies that assume indiscriminate inhibition of this key protease, and stress the need for further mechanistic and translational studies.
Collapse
Affiliation(s)
- Saak V Ovsepian
- National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.,International Centre for Neurotherapeutics, Dublin City University, Dublin, Ireland
| | - Jiri Horacek
- National Institute of Mental Health, Klecany, Czech Republic
| | - Valerie B O'Leary
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Cyril Hoschl
- National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
61
|
Lv L, Yang F, Li H, Yuan J. Brain‐targeted co‐delivery of β‐amyloid converting enzyme 1
shRNA
and epigallocatechin‐3‐gallate by multifunctional nanocarriers for Alzheimer's disease treatment. IUBMB Life 2020; 72:1819-1829. [PMID: 32668504 DOI: 10.1002/iub.2330] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/24/2020] [Accepted: 05/24/2020] [Indexed: 12/22/2022]
Affiliation(s)
- Lijie Lv
- Department of Medical and NursingThe First Hospital of Jilin University Changchun China
| | - Fan Yang
- Department of Pediatric SurgeryThe First Hospital of Jilin University Changchun China
| | - He Li
- Department of Pain MedicineThe First Hospital of Jilin University Changchun China
| | - Jiuli Yuan
- Department of Medical and NursingThe First Hospital of Jilin University Changchun China
| |
Collapse
|
62
|
Khalifeh M, Read MI, Barreto GE, Sahebkar A. Trehalose against Alzheimer's Disease: Insights into a Potential Therapy. Bioessays 2020; 42:e1900195. [PMID: 32519387 DOI: 10.1002/bies.201900195] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 04/13/2020] [Indexed: 12/21/2022]
Abstract
Trehalose is a natural disaccharide with a remarkable ability to stabilize biomolecules. In recent years, trehalose has received growing attention as a neuroprotective molecule and has been tested in experimental models for different neurodegenerative diseases. Although the underlying neuroprotective mechanism of trehalose's action is unclear, one of the most important hypotheses is autophagy induction. The chaperone-like activity of trehalose and the ability to modulate inflammatory responses has also been reported. There is compelling evidence that the dysfunction of autophagy and aggregation of misfolded proteins contribute to the pathogenesis of Alzheimer's disease (AD) and other neurodegenerative disorders. Therefore, given the linking between trehalose and autophagy induction, it appears to be a promising therapy for AD. Herein, the published studies concerning the use of trehalose as a potential therapy for AD are summarized, providing a rationale for applying trehalose to reduce Alzheimer's pathology.
Collapse
Affiliation(s)
- Masoomeh Khalifeh
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Morgayn I Read
- Department of Pharmacology, School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
63
|
Barthet G, Mulle C. Presynaptic failure in Alzheimer's disease. Prog Neurobiol 2020; 194:101801. [PMID: 32428558 DOI: 10.1016/j.pneurobio.2020.101801] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/24/2020] [Accepted: 04/03/2020] [Indexed: 12/14/2022]
Abstract
Synaptic loss is the best correlate of cognitive deficits in Alzheimer's disease (AD). Extensive experimental evidence also indicates alterations of synaptic properties at the early stages of disease progression, before synapse loss and neuronal degeneration. A majority of studies in mouse models of AD have focused on post-synaptic mechanisms, including impairment of long-term plasticity, spine structure and glutamate receptor-mediated transmission. Here we review the literature indicating that the synaptic pathology in AD includes a strong presynaptic component. We describe the evidence indicating presynaptic physiological functions of the major molecular players in AD. These include the amyloid precursor protein (APP) and the two presenilin (PS) paralogs PS1 or PS2, genetically linked to the early-onset form of AD, in addition to tau which accumulates in a pathological form in the AD brain. Three main mechanisms participating in presynaptic functions are highlighted. APP fragments bind to presynaptic receptors (e.g. nAChRs and GABAB receptors), presenilins control Ca2+ homeostasis and Ca2+-sensors, and tau regulates the localization of presynaptic molecules and synaptic vesicles. We then discuss how impairment of these presynaptic physiological functions can explain or forecast the hallmarks of synaptic impairment and associated dysfunction of neuronal circuits in AD. Beyond the physiological roles of the AD-related proteins, studies in AD brains also support preferential presynaptic alteration. This review features presynaptic failure as a strong component of pathological mechanisms in AD.
Collapse
Affiliation(s)
- Gael Barthet
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, University of Bordeaux, France
| | - Christophe Mulle
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, University of Bordeaux, France.
| |
Collapse
|
64
|
Miyamoto M, Kuzuya A, Noda Y, Ueda S, Asada-Utsugi M, Ito S, Fukusumi Y, Kawachi H, Takahashi R, Kinoshita A. Synaptic Vesicle Protein 2B Negatively Regulates the Amyloidogenic Processing of AβPP as a Novel Interaction Partner of BACE1. J Alzheimers Dis 2020; 75:173-185. [DOI: 10.3233/jad-200071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Masakazu Miyamoto
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Kuzuya
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuha Noda
- Department of Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sakiho Ueda
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Megumi Asada-Utsugi
- Department of Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinji Ito
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshiyasu Fukusumi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hiroshi Kawachi
- Department of Cell Biology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ayae Kinoshita
- Department of Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
65
|
Taliyan R, Chandran SK, Kakoty V. Therapeutic Approaches to Alzheimer's Type of Dementia: A Focus on FGF21 Mediated Neuroprotection. Curr Pharm Des 2020; 25:2555-2568. [PMID: 31333086 DOI: 10.2174/1381612825666190716101411] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 07/08/2019] [Indexed: 12/31/2022]
Abstract
Neurodegenerative disorders are the most devastating disorder of the nervous system. The pathological basis of neurodegeneration is linked with dysfunctional protein trafficking, mitochondrial stress, environmental factors and aging. With the identification of insulin and insulin receptors in some parts of the brain, it has become evident that certain metabolic conditions associated with insulin dysfunction like Type 2 diabetes mellitus (T2DM), dyslipidemia, obesity etc., are also known to contribute to neurodegeneration mainly Alzheimer's Disease (AD). Recently, a member of the fibroblast growth factor (FGF) superfamily, FGF21 has proved tremendous efficacy in diseases like diabetes mellitus, obesity and insulin resistance (IR). Increased levels of FGF21 have been reported to exert multiple beneficial effects in metabolic syndrome. FGF21 receptors are present in certain areas of the brain involved in learning and memory. However, despite extensive research, its function as a neuroprotectant in AD remains elusive. FGF21 is a circulating endocrine hormone which is mainly secreted by the liver primarily in fasting conditions. FGF21 exerts its effects after binding to FGFR1 and co-receptor, β-klotho (KLB). It is involved in regulating energy via glucose and lipid metabolism. It is believed that aberrant FGF21 signalling might account for various anomalies like neurodegeneration, cancer, metabolic dysfunction etc. Hence, this review will majorly focus on FGF21 role as a neuroprotectant and potential metabolic regulator. Moreover, we will also review its potential as an emerging candidate for combating metabolic stress induced neurodegenerative abnormalities.
Collapse
Affiliation(s)
- Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani-333031, Rajasthan, India
| | - Sarathlal K Chandran
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani-333031, Rajasthan, India
| | - Violina Kakoty
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani-333031, Rajasthan, India
| |
Collapse
|
66
|
Collins JM, Woodhouse A, Bye N, Vickers JC, King AE, Ziebell JM. Pathological Links between Traumatic Brain Injury and Dementia: Australian Pre-Clinical Research. J Neurotrauma 2020; 37:782-791. [PMID: 32046575 DOI: 10.1089/neu.2019.6906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) can cause persistent cognitive changes and ongoing neurodegeneration in the brain. Accumulating epidemiological and pathological evidence implicates TBI in the development of Alzheimer's disease, the most common cause of dementia. Further, the TBI-induced form of dementia, called chronic traumatic encephalopathy, shares many pathological hallmarks present in multiple different diseases which cause dementia. The inflammatory and neuritic responses to TBI and dementia overlap, indicating that they may share common pathological mechanisms and that TBI may ultimately cause a pathological cascade culminating in the development of dementia. This review explores Australian pre-clinical research investigating the pathological links between TBI and dementia.
Collapse
Affiliation(s)
- Jessica M Collins
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Adele Woodhouse
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Nicole Bye
- School of Pharmacy, and College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia.,School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Jenna M Ziebell
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
67
|
Multiple inflammatory profiles of microglia and altered neuroimages in APP/PS1 transgenic AD mice. Brain Res Bull 2020; 156:86-104. [DOI: 10.1016/j.brainresbull.2020.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/14/2019] [Accepted: 01/03/2020] [Indexed: 12/11/2022]
|
68
|
Lei Mon SS, Yoshioka K, Jia C, Kunieda T, Asami Y, Yoshida-Tanaka K, Piao W, Kuwahara H, Nishina K, Nagata T, Yokota T. Highly efficient gene silencing in mouse brain by overhanging-duplex oligonucleotides via intraventricular route. FEBS Lett 2020; 594:1413-1423. [PMID: 31990989 DOI: 10.1002/1873-3468.13742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/10/2020] [Accepted: 01/17/2020] [Indexed: 11/05/2022]
Abstract
Gapmer-type antisense oligonucleotides have not yet been approved for the treatment of central nervous system diseases, whereas steric-blocking-type antisense oligonucleotides have been well-developed for clinical use. We here characterize a new type of double-stranded oligonucleotides, overhanging-duplex oligonucleotides, which are composed of the parent gapmer and its extended complementary RNA. By intracerebroventricular injection, overhanging oligonucleotides show greater silencing potency with more efficient delivery into mouse brains than the parent single-stranded gapmer. Structure-activity relationship analyses reveal that the potency enhancement requires 13-mer or more overhanging oligonucleotides with a phosphorothioate backbone. Overhanging oligonucleotides provide a new platform of therapeutic oligonucleotides for gene modulation in the central nervous system.
Collapse
Affiliation(s)
- Su Su Lei Mon
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences and Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), Japan
| | - Kotaro Yoshioka
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences and Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), Japan
| | - Chunyan Jia
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences and Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), Japan
| | - Taiki Kunieda
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences and Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), Japan
| | - Yutaro Asami
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences and Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), Japan
| | - Kie Yoshida-Tanaka
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences and Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), Japan
| | - Wenying Piao
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences and Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), Japan
| | - Hiroya Kuwahara
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences and Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), Japan
| | - Kazutaka Nishina
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences and Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), Japan
| | - Tetsuya Nagata
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences and Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences and Center for Brain Integration Research, Tokyo Medical and Dental University (TMDU), Japan
| |
Collapse
|
69
|
Proximity ligation assay reveals both pre- and postsynaptic localization of the APP-processing enzymes ADAM10 and BACE1 in rat and human adult brain. BMC Neurosci 2020; 21:6. [PMID: 32019490 PMCID: PMC7001251 DOI: 10.1186/s12868-020-0554-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/27/2020] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Synaptic degeneration and accumulation of amyloid β-peptides (Aβ) are hallmarks of the Alzheimer diseased brain. Aβ is synaptotoxic and produced by sequential cleavage of the amyloid precursor protein (APP) by the β-secretase BACE1 and by γ-secretase. If APP is instead cleaved by the α-secretase ADAM10, Aβ will not be generated. Although BACE1 is considered to be a presynaptic protein and ADAM10 has been reported to mainly localize to the postsynaptic density, we have previously shown that both ADAM10 and BACE1 are highly enriched in synaptic vesicles of rat brain and mouse primary hippocampal neurons. RESULTS Here, using brightfield proximity ligation assay, we expanded our previous result in primary neurons and investigated the in situ synaptic localization of ADAM10 and BACE1 in rat and human adult brain using both pre- and postsynaptic markers. We found that ADAM10 and BACE1 were in close proximity with both the presynaptic marker synaptophysin and the postsynaptic marker PSD-95. The substrate APP was also detected both pre- and postsynaptically. Subcellular fractionation confirmed that ADAM10 and BACE1 are enriched to a similar degree in synaptic vesicles and as well as in the postsynaptic density. CONCLUSIONS We show that the α-secretase ADAM10 and the β-secretase BACE1 are located in both the pre- and postsynaptic compartments in intact brain sections. These findings increase our understanding of the regulation of APP processing, thereby facilitating development of more specific treatment strategies.
Collapse
|
70
|
Bergkvist L, Du Z, Elovsson G, Appelqvist H, Itzhaki LS, Kumita JR, Kågedal K, Brorsson AC. Mapping pathogenic processes contributing to neurodegeneration in Drosophila models of Alzheimer's disease. FEBS Open Bio 2020; 10:338-350. [PMID: 31823504 PMCID: PMC7050262 DOI: 10.1002/2211-5463.12773] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/21/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, affecting millions of people and currently lacking available disease‐modifying treatments. Appropriate disease models are necessary to investigate disease mechanisms and potential treatments. Drosophila melanogaster models of AD include the Aβ fly model and the AβPP‐BACE1 fly model. In the Aβ fly model, the Aβ peptide is fused to a secretion sequence and directly overexpressed. In the AβPP‐BACE1 model, human AβPP and human BACE1 are expressed in the fly, resulting in in vivo production of Aβ peptides and other AβPP cleavage products. Although these two models have been used for almost two decades, the underlying mechanisms resulting in neurodegeneration are not yet clearly understood. In this study, we have characterized toxic mechanisms in these two AD fly models. We detected neuronal cell death and increased protein carbonylation (indicative of oxidative stress) in both AD fly models. In the Aβ fly model, this correlates with high Aβ1–42 levels and down‐regulation of the levels of mRNA encoding lysosomal‐associated membrane protein 1, lamp1 (a lysosomal marker), while in the AβPP‐BACE1 fly model, neuronal cell death correlates with low Aβ1–42 levels, up‐regulation of lamp1 mRNA levels and increased levels of C‐terminal fragments. In addition, a significant amount of AβPP/Aβ antibody (4G8)‐positive species, located close to the endosomal marker rab5, was detected in the AβPP‐BACE1 model. Taken together, this study highlights the similarities and differences in the toxic mechanisms which result in neuronal death in two different AD fly models. Such information is important to consider when utilizing these models to study AD pathogenesis or screening for potential treatments.
Collapse
Affiliation(s)
- Liza Bergkvist
- Division of Molecular Biotechnology, Department of Physics, Chemistry and Biology, Linköping University, Sweden
| | - Zhen Du
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, UK.,Department of Pharmacology, University of Cambridge, UK
| | - Greta Elovsson
- Division of Molecular Biotechnology, Department of Physics, Chemistry and Biology, Linköping University, Sweden
| | - Hanna Appelqvist
- Division of Molecular Biotechnology, Department of Physics, Chemistry and Biology, Linköping University, Sweden.,Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Sweden
| | | | - Janet R Kumita
- Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, UK
| | - Katarina Kågedal
- Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Sweden
| | - Ann-Christin Brorsson
- Division of Molecular Biotechnology, Department of Physics, Chemistry and Biology, Linköping University, Sweden
| |
Collapse
|
71
|
Mouchlis VD, Melagraki G, Zacharia LC, Afantitis A. Computer-Aided Drug Design of β-Secretase, γ-Secretase and Anti-Tau Inhibitors for the Discovery of Novel Alzheimer's Therapeutics. Int J Mol Sci 2020; 21:E703. [PMID: 31973122 PMCID: PMC7038192 DOI: 10.3390/ijms21030703] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 12/14/2022] Open
Abstract
Aging-associated neurodegenerative diseases, which are characterized by progressive neuronal death and synapses loss in human brain, are rapidly growing affecting millions of people globally. Alzheimer's is the most common neurodegenerative disease and it can be caused by genetic and environmental risk factors. This review describes the amyloid-β and Tau hypotheses leading to amyloid plaques and neurofibrillary tangles, respectively which are the predominant pathways for the development of anti-Alzheimer's small molecule inhibitors. The function and structure of the druggable targets of these two pathways including β-secretase, γ-secretase, and Tau are discussed in this review article. Computer-Aided Drug Design including computational structure-based design and ligand-based design have been employed successfully to develop inhibitors for biomolecular targets involved in Alzheimer's. The application of computational molecular modeling for the discovery of small molecule inhibitors and modulators for β-secretase and γ-secretase is summarized. Examples of computational approaches employed for the development of anti-amyloid aggregation and anti-Tau phosphorylation, proteolysis and aggregation inhibitors are also reported.
Collapse
Affiliation(s)
| | - Georgia Melagraki
- Division of Physical Sciences & Applications, Hellenic Military Academy, Vari 16672, Greece;
| | - Lefteris C. Zacharia
- Department of Life and Health Sciences, University of Nicosia, Nicosia 1700, Cyprus;
| | - Antreas Afantitis
- Department of ChemoInformatics, NovaMechanics Ltd., Nicosia 1046, Cyprus
| |
Collapse
|
72
|
Lombardo S, Chiacchiaretta M, Tarr A, Kim W, Cao T, Sigal G, Rosahl TW, Xia W, Haydon PG, Kennedy ME, Tesco G. BACE1 partial deletion induces synaptic plasticity deficit in adult mice. Sci Rep 2019; 9:19877. [PMID: 31882662 PMCID: PMC6934620 DOI: 10.1038/s41598-019-56329-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022] Open
Abstract
BACE1 is the first enzyme involved in APP processing, thus it is a strong therapeutic target candidate for Alzheimer's disease. The observation of deleterious phenotypes in BACE1 Knock-out (KO) mouse models (germline and conditional) raised some concerns on the safety and tolerability of BACE1 inhibition. Here, we have employed a tamoxifen inducible BACE1 conditional Knock-out (cKO) mouse model to achieve a controlled partial depletion of BACE1 in adult mice. Biochemical and behavioural characterization was performed at two time points: 4-5 months (young mice) and 12-13 months (aged mice). A ~50% to ~70% BACE1 protein reduction in hippocampus and cortex, respectively, induced a significant reduction of BACE1 substrates processing and decrease of Aβx-40 levels at both ages. Hippocampal axonal guidance and peripheral nerve myelination were not affected. Aged mice displayed a CA1 long-term potentiation (LTP) deficit that was not associated with memory impairment. Our findings indicate that numerous phenotypes observed in germline BACE1 KO reflect a fundamental role of BACE1 during development while other phenotypes, observed in adult cKO, may be absent when partially rather than completely deleting BACE1. However, we demonstrated that partial depletion of BACE1 still induces CA1 LTP impairment, supporting a role of BACE1 in synaptic plasticity in adulthood.
Collapse
Affiliation(s)
- Sylvia Lombardo
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | - Martina Chiacchiaretta
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | - Andrew Tarr
- Circuits and Behaviour Core, Center for Neuroscience Research, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | - WonHee Kim
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | - Tingyi Cao
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | - Griffin Sigal
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | - Thomas W Rosahl
- External In Vivo Pharmacology, Merck & Co. Inc., Kenilworth, NJ, 07033, USA
| | - Weiming Xia
- Geriatric Research, Education and Clinic Center, Bedford Veterans Affairs Medical Center, Bedford, MA, 01730, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Philip G Haydon
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | | | - Giuseppina Tesco
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA.
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA.
| |
Collapse
|
73
|
Maesako M, Zoltowska KM, Berezovska O. Synapsin 1 promotes Aβ generation via BACE1 modulation. PLoS One 2019; 14:e0226368. [PMID: 31830091 PMCID: PMC6907790 DOI: 10.1371/journal.pone.0226368] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/25/2019] [Indexed: 12/26/2022] Open
Abstract
It has been revealed that β-amyloid (Aβ) is generated and released from the presynaptic terminals in activity-dependent manner. However, molecules modulating the presynaptic Aβ generation remain elusive. Here we test the hypothesis that Synapsin 1 (Syn1) may acts as a modulator of the Aβ production. Using biochemical and Förster resonance energy transfer (FRET)-based imaging approaches we have found that Syn1 knock down decreases, whereas (over)expression of Syn1 in cells increases the Aβ levels. Mechanistically, Syn1 does not seem to affect the activity of Presenilin 1 (PS1)/γ-secretase, PS1 conformation, or the proximity between PS1 and amyloid precursor protein (APP). However, we found that Syn1 is involved in up-regulation of the β-site APP cleaving enzyme 1 (BACE1)/β-secretase activity and increases the APP/BACE1 interaction. Therefore, we conclude that Syn1 may promote Aβ production via the modulation of BACE1.
Collapse
Affiliation(s)
- Masato Maesako
- MassGeneral Institute for Neurodegenerative Disease, Alzheimer’s Disease Research Unit, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States of America
| | - Katarzyna M. Zoltowska
- MassGeneral Institute for Neurodegenerative Disease, Alzheimer’s Disease Research Unit, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States of America
| | - Oksana Berezovska
- MassGeneral Institute for Neurodegenerative Disease, Alzheimer’s Disease Research Unit, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States of America
- * E-mail:
| |
Collapse
|
74
|
Peters F, Salihoglu H, Pratsch K, Herzog E, Pigoni M, Sgobio C, Lichtenthaler SF, Neumann U, Herms J. Tau deletion reduces plaque-associated BACE1 accumulation and decelerates plaque formation in a mouse model of Alzheimer's disease. EMBO J 2019; 38:e102345. [PMID: 31701556 PMCID: PMC6885735 DOI: 10.15252/embj.2019102345] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/29/2019] [Accepted: 10/08/2019] [Indexed: 01/24/2023] Open
Abstract
In Alzheimer's disease, BACE1 protease initiates the amyloidogenic processing of amyloid precursor protein (APP) that eventually results in synthesis of β-amyloid (Aβ) peptide. Aβ deposition in turn causes accumulation of BACE1 in plaque-associated dystrophic neurites, thereby potentiating progressive Aβ deposition once initiated. Since systemic pharmacological BACE inhibition causes adverse effects in humans, it is important to identify strategies that specifically normalize overt BACE1 activity around plaques. The microtubule-associated protein tau regulates axonal transport of proteins, and tau deletion rescues Aβ-induced transport deficits in vitro. In the current study, long-term in vivo two-photon microscopy and immunohistochemistry were performed in tau-deficient APPPS1 mice. Tau deletion reduced plaque-associated axonal pathology and BACE1 accumulation without affecting physiological BACE1 expression distant from plaques. Thereby, tau deletion effectively decelerated formation of new plaques and reduced plaque compactness. The data revealed that tau reinforces Aβ deposition, presumably by contributing to accumulation of BACE1 in plaque-associated dystrophies. Targeting tau-dependent mechanisms could become a suitable strategy to specifically reduce overt BACE1 activity around plaques, thereby avoiding adverse effects of systemic BACE inhibition.
Collapse
Affiliation(s)
- Finn Peters
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
| | - Hazal Salihoglu
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
| | - Katrin Pratsch
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
| | - Etienne Herzog
- IINS, UMR 5297Université BordeauxBordeauxFrance
- CNRS, IINS, UMR 5297BordeauxFrance
| | - Martina Pigoni
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
| | - Carmelo Sgobio
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
- NeuroproteomicsSchool of MedicineKlinikum rechts der Isar and Institute for Advanced StudyTechnical University of MunichMunichGermany
| | - Ulf Neumann
- NeuroscienceNovartis Institutes for BioMedical Research (NIBR)BaselSwitzerland
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
- Center for Neuropathology and Prion ResearchLudwig‐Maximilians UniversityMunichGermany
| |
Collapse
|
75
|
Meilandt WJ, Maloney JA, Imperio J, Lalehzadeh G, Earr T, Crowell S, Bainbridge TW, Lu Y, Ernst JA, Fuji RN, Atwal JK. Characterization of the selective in vitro and in vivo binding properties of crenezumab to oligomeric Aβ. ALZHEIMERS RESEARCH & THERAPY 2019; 11:97. [PMID: 31787113 PMCID: PMC6886224 DOI: 10.1186/s13195-019-0553-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/06/2019] [Indexed: 02/01/2023]
Abstract
Background Accumulation of amyloid β (Aβ) in the brain is proposed as a cause of Alzheimer’s disease (AD), with Aβ oligomers hypothesized to be the primary mediators of neurotoxicity. Crenezumab is a humanized immunoglobulin G4 monoclonal antibody that has been shown to bind to synthetic monomeric and aggregated Aβ in vitro; however, less is known about the binding characteristic in vivo. In this study, we evaluated the binding patterns of crenezumab to synthetic and native forms of Aβ both in vitro and in vivo. Methods Crenezumab was used to immunoprecipitate Aβ from synthetic Aβ preparations or brain homogenates from a PS2APP mouse model of AD to determine the forms of Aβ that crenezumab interacts with. Following systemic dosing in PS2APP or nontransgenic control mice, immunohistochemistry was used to localize crenezumab and assess its relative distribution in the brain, compared with amyloid plaques and markers of neuritic dystrophies (BACE1; LAMP1). Pharmacodynamic correlations were performed to investigate the relationship between peripheral and central target engagement. Results In vitro, crenezumab immunoprecipitated Aβ oligomers from both synthetic Aβ preparations and endogenous brain homogenates from PS2APP mice. In vivo studies in the PS2APP mouse showed that crenezumab localizes to regions surrounding the periphery of amyloid plaques in addition to the hippocampal mossy fibers. These regions around the plaques are reported to be enriched in oligomeric Aβ, actively incorporate soluble Aβ, and contribute to Aβ-induced neurotoxicity and axonal dystrophy. In addition, crenezumab did not appear to bind to the dense core region of plaques or vascular amyloid. Conclusions Crenezumab binds to multiple forms of amyloid β (Aβ), particularly oligomeric forms, and localizes to brain areas rich in Aβ oligomers, including the halo around plaques and hippocampal mossy fibers, but not to vascular Aβ. These insights highlight a unique mechanism of action for crenezumab of engaging Aβ oligomers.
Collapse
Affiliation(s)
- William J Meilandt
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Janice A Maloney
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Jose Imperio
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Guita Lalehzadeh
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Tim Earr
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Susan Crowell
- Department of Preclinical and Translational Pharmacokinetics/Pharmacodynamics, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Travis W Bainbridge
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Yanmei Lu
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - James A Ernst
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Reina N Fuji
- Department of Safety Assessment Pathology, Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Jasvinder K Atwal
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
| |
Collapse
|
76
|
Imbimbo BP, Watling M. Investigational BACE inhibitors for the treatment of Alzheimer's disease. Expert Opin Investig Drugs 2019; 28:967-975. [PMID: 31661331 DOI: 10.1080/13543784.2019.1683160] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: The amyloid hypothesis of Alzheimer's disease (AD) states that brain accumulation of amyloid-β (Aβ) oligomers and soluble aggregates represents the major causal event of the disease. Several small organic molecules have been synthesized and developed to inhibit the enzyme (β-site amyloid precursor protein cleaving enzyme-1 or BACE1) whose action represents the rate-limiting step in Aβ production.Areas covered: We reviewed the pharmacology and clinical trials of major BACE1 inhibitors.Expert opinion: In transgenic mouse models of AD, BACE1 inhibitors dose-dependently lower Aβ levels in brain and cerebrospinal fluid (CSF) but the evidence for attenuation or reversal cognitive or behavioral deficits is very scanty. In AD patients, BACE1 inhibitors robustly lower plasma and CSF Aβ levels and reduce brain plaques but without cognitive, clinical, or functional benefit. To date, seventeen BACE1 inhibitors have failed in double-blind, placebo-controlled clinical trials in patients with mild-to-moderate or prodromal AD, or in cognitively normal subjects at risk of developing AD. Several of these studies were prematurely interrupted due to toxicity or cognitive and behavioral worsening compared to placebo-treated patients. Elenbecestat, the last BACE1 inhibitor remaining in late clinical testing for AD, was recently discontinued due to safety concerns.
Collapse
Affiliation(s)
| | - Mark Watling
- CNS & Pain Department, TranScrip Partners, Reading, UK
| |
Collapse
|
77
|
Xie Y, Ba L, Wang M, Deng SY, Chen SM, Huang LF, Zhang M, Wang W, Ding FF. Chronic sleep fragmentation shares similar pathogenesis with neurodegenerative diseases: Endosome-autophagosome-lysosome pathway dysfunction and microglia-mediated neuroinflammation. CNS Neurosci Ther 2019; 26:215-227. [PMID: 31549780 PMCID: PMC6978272 DOI: 10.1111/cns.13218] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/22/2019] [Accepted: 08/24/2019] [Indexed: 12/15/2022] Open
Abstract
Aims Insufficient sleep has been found to result in varying degrees of cognitive impairment and emotional changes. Sleep was reported probably responsible for cleaning metabolic wastes in brain by increasing extracellular bulk flow. Herein, we propose that chronic sleep insufficiency in young adult wild‐type mice is also linked with dysfunction of intracellular protein degradation pathways and microglia‐mediated neuroinflammation, which are potentially important mechanisms in the initiation of neurodegeneration. Methods We applied the chronic sleep fragmentation (CSF) model to induce chronic sleep insufficiency in wild‐type mice. After 2 months of CSF, cognitive function, amyloid‐β accumulation, dysfunction of endosome‐autophagosome‐lysosome pathway, and microglia activation were evaluated. Results Following CSF, impairment of spatial learning and memory, and aggravated anxiety‐like behavior in mice were identified by behavioral experiments. Increased intracellular amyloid‐β accumulation was observed in cortex and hippocampus. Mechanistically, CSF could significantly enhance the expression of Rab5 (early endosome marker), Rab7 (late endosome marker), as well as LC3B (autophagosome marker), and autophagy‐positive regulatory factors in brain detected by immunofluorescent staining and Western blot. In addition, activation of microglia was evident by enhanced CD68, CD16/32, and CD206 levels after CSF treatment. Conclusions Chronic sleep fragmentation could initiate pathogenetic processes similar to the early stage of neurodegeneration, including dysfunction of endosome‐autophagosome‐lysosome pathway and microglia‐mediated neuroinflammation. Our findings further strengthen the link between chronic sleep insufficiency and the initiation of neurodegeneration even if lack of genetic predisposition.
Collapse
Affiliation(s)
- Yi Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Ba
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sai-Yue Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si-Miao Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Fang Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Neurological Diseases of Chinese Ministry of Education, The School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng-Fei Ding
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
78
|
Liang ZM, Peng YH, Chen Y, Long LL, Luo HJ, Chen YJ, Liang YL, Tian YH, Li SJ, Shi YS, Zhang XM. The BACE1-Specific DNA Aptamer A1 Rescues Amyloid-β Pathology and Behavioral Deficits in a Mouse Model of Alzheimer's Disease. Nucleic Acid Ther 2019; 29:359-366. [PMID: 31513457 DOI: 10.1089/nat.2019.0812] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Amyloid-β (Aβ) plaque deposits in the brain are considered to be one of the main pathological markers of Alzheimer's disease (AD). The sequential proteolytic cleavage of amyloid precursor protein (APP) by the aspartyl proteases β-site APP-cleaving enzyme 1 (BACE1) and γ-secretase produces Aβ. Therefore, BACE1 inhibition is a very attractive target for the treatment of AD. Our previous work identified a DNA aptamer named A1 that can bind to BACE1 with high affinity and specificity and exhibits a distinct inhibitory effect on BACE1 activity in an AD cell model. The purpose of this research was to test the effect of aptamer A1 in Tg6799 mice. Four-month-old Tg6799 mice were randomly divided into two groups and treated with aptamer A1 and ineffective aptamer A1scr, respectively, by intracerebroventricular injection. Subsequent behavioral experiments showed that treatment with the aptamer A1 improved the cognitive abilities of the AD mice. Western blot indicated that BACE1 and soluble amyloid precursor protein β (sAPPβ) expression significantly decreased in the A1-treated mice. Moreover, aptamer A1 reduced the content of Aβ42 and the number and density of senile plaques in AD mice. Therefore, our results indicate that aptamer A1 is a novel specific and potent BACE1 inhibitor and is a promising potential target for the treatment of AD.
Collapse
Affiliation(s)
- Zhi-Man Liang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Pathology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Yong-Hua Peng
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yue Chen
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Li-Li Long
- The First Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Hong-Jie Luo
- The First Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Ya-Jun Chen
- The First Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Yan-Ling Liang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ying-Hong Tian
- Experiment Teaching & Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shu-Ji Li
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yu-Sheng Shi
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xing-Mei Zhang
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
79
|
Saadipour K, Tiberi A, Lombardo S, Grajales E, Montroull L, Mañucat-Tan NB, LaFrancois J, Cammer M, Mathews PM, Scharfman HE, Liao FF, Friedman WJ, Zhou XF, Tesco G, Chao MV. Regulation of BACE1 expression after injury is linked to the p75 neurotrophin receptor. Mol Cell Neurosci 2019; 99:103395. [PMID: 31422108 DOI: 10.1016/j.mcn.2019.103395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/24/2019] [Accepted: 08/08/2019] [Indexed: 12/25/2022] Open
Abstract
BACE1 is a transmembrane aspartic protease that cleaves various substrates and it is required for normal brain function. BACE1 expression is high during early development, but it is reduced in adulthood. Under conditions of stress and injury, BACE1 levels are increased; however, the underlying mechanisms that drive BACE1 elevation are not well understood. One mechanism associated with brain injury is the activation of injurious p75 neurotrophin receptor (p75), which can trigger pathological signals. Here we report that within 72 h after controlled cortical impact (CCI) or laser injury, BACE1 and p75 are increased and tightly co-expressed in cortical neurons of mouse brain. Additionally, BACE1 is not up-regulated in p75 null mice in response to focal cortical injury, while p75 over-expression results in BACE1 augmentation in HEK-293 and SY5Y cell lines. A luciferase assay conducted in SY5Y cell line revealed that BACE1 expression is regulated at the transcriptional level in response to p75 transfection. Interestingly, this effect does not appear to be dependent upon p75 ligands including mature and pro-neurotrophins. In addition, BACE1 activity on amyloid precursor protein (APP) is enhanced in SY5Y-APP cells transfected with a p75 construct. Lastly, we found that the activation of c-jun n-terminal kinase (JNK) by p75 contributes to BACE1 up-regulation. This study explores how two injury-induced molecules are intimately connected and suggests a potential link between p75 signaling and the expression of BACE1 after brain injury.
Collapse
Affiliation(s)
- Khalil Saadipour
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA.
| | - Alexia Tiberi
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA; Bio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7, Pisa, 56126, Italy
| | - Sylvia Lombardo
- Alzheimer's Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Elena Grajales
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA
| | - Laura Montroull
- Department of Biological Sciences, Rutgers Life Sciences Center, Rutgers University, Newark, NJ 07102, USA
| | - Noralyn B Mañucat-Tan
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, South Australia, 5000, Australia
| | - John LaFrancois
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, Orangeburg, NY 10962, USA
| | - Michael Cammer
- DART Microscopy Laboratory, NYU Langone Medical Center, New York, NY 10016, USA
| | - Paul M Mathews
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, Orangeburg, NY 10962, USA
| | - Helen E Scharfman
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, Orangeburg, NY 10962, USA
| | - Francesca-Fang Liao
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Wilma J Friedman
- Department of Biological Sciences, Rutgers Life Sciences Center, Rutgers University, Newark, NJ 07102, USA
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, South Australia, 5000, Australia
| | - Giueseppina Tesco
- Alzheimer's Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Moses V Chao
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA.
| |
Collapse
|
80
|
Gaudreault R, Mousseau N. Mitigating Alzheimer’s Disease with Natural Polyphenols: A Review. Curr Alzheimer Res 2019; 16:529-543. [DOI: 10.2174/1567205016666190315093520] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/14/2019] [Accepted: 03/13/2019] [Indexed: 11/22/2022]
Abstract
:According to Alzheimer’s Disease International (ADI), nearly 50 million people worldwide were living with dementia in 2017, and this number is expected to triple by 2050. Despite years of research in this field, the root cause and mechanisms responsible for Alzheimer’s disease (AD) have not been fully elucidated yet. Moreover, promising preclinical results have repeatedly failed to translate into patient treatments. Until now, none of the molecules targeting AD has successfully passed the Phase III trial. Although natural molecules have been extensively studied, they normally require high concentrations to be effective; alternately, they are too large to cross the blood-brain barrier (BBB).:In this review, we report AD treatment strategies, with a virtually exclusive focus on green chemistry (natural phenolic molecules). These include therapeutic strategies for decreasing amyloid-β (Aβ) production, preventing and/or altering Aβ aggregation, and reducing oligomers cytotoxicity such as curcumin, (-)-epigallocatechin-3-gallate (EGCG), morin, resveratrol, tannic acid, and other natural green molecules. We also examine whether consideration should be given to potential candidates used outside of medicine and nutrition, through a discussion of two intermediate-sized green molecules, with very similar molecular structures and key properties, which exhibit potential in mitigating Alzheimer’s disease.
Collapse
Affiliation(s)
- Roger Gaudreault
- Department of Physics, Universit�© de Montr�©al, Case Postale 6128, Succursale Centre-ville, Montreal (QC), Canada
| | - Normand Mousseau
- Department of Physics, Universit�© de Montr�©al, Case Postale 6128, Succursale Centre-ville, Montreal (QC), Canada
| |
Collapse
|
81
|
Li P, Marshall L, Oh G, Jakubowski JL, Groot D, He Y, Wang T, Petronis A, Labrie V. Epigenetic dysregulation of enhancers in neurons is associated with Alzheimer's disease pathology and cognitive symptoms. Nat Commun 2019; 10:2246. [PMID: 31113950 PMCID: PMC6529540 DOI: 10.1038/s41467-019-10101-7] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 04/18/2019] [Indexed: 12/23/2022] Open
Abstract
Epigenetic control of enhancers alters neuronal functions and may be involved in Alzheimer’s disease (AD). Here, we identify enhancers in neurons contributing to AD by comprehensive fine-mapping of DNA methylation at enhancers, genome-wide. We examine 1.2 million CpG and CpH sites in enhancers in prefrontal cortex neurons of individuals with no/mild, moderate, and severe AD pathology (n = 101). We identify 1224 differentially methylated enhancer regions; most of which are hypomethylated at CpH sites in AD neurons. CpH methylation losses occur in normal aging neurons, but are accelerated in AD. Integration of epigenetic and transcriptomic data demonstrates a pro-apoptotic reactivation of the cell cycle in post-mitotic AD neurons. Furthermore, AD neurons have a large cluster of significantly hypomethylated enhancers in the DSCAML1 gene that targets BACE1. Hypomethylation of these enhancers in AD is associated with an upregulation of BACE1 transcripts and an increase in amyloid plaques, neurofibrillary tangles, and cognitive decline. Epigenetic control of enhancers may contribute to neurological disease. Here the authors carry out genome-wide analysis of DNA methylation in neurons isolated postmortem from patients with Alzheimer’s disease.
Collapse
Affiliation(s)
- Peipei Li
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Lee Marshall
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Gabriel Oh
- Centre for Addiction and Mental Health, Toronto, M5T 1R8, ON, Canada
| | - Jennifer L Jakubowski
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Daniel Groot
- Centre for Addiction and Mental Health, Toronto, M5T 1R8, ON, Canada
| | - Yu He
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Ting Wang
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Arturas Petronis
- Centre for Addiction and Mental Health, Toronto, M5T 1R8, ON, Canada.,Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257, Vilnius, Lithuania
| | - Viviane Labrie
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, 49503, USA. .,Centre for Addiction and Mental Health, Toronto, M5T 1R8, ON, Canada. .,Division of Psychiatry and Behavioral Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
82
|
Quercetin-modified gold-palladium nanoparticles as a potential autophagy inducer for the treatment of Alzheimer's disease. J Colloid Interface Sci 2019; 552:388-400. [PMID: 31151017 DOI: 10.1016/j.jcis.2019.05.066] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/15/2019] [Accepted: 05/20/2019] [Indexed: 01/14/2023]
Abstract
At present, autophagic dysfunction has been considered to be involved in the pathogenesis of Alzheimer's disease (AD). Thus, the activation of autophagy provides a potential means of eliminating the intracellular amyloid-β (Aβ) and slows down the neurotoxicity induced by Aβ. Here, we synthesize a Quercetin (Qu) modified polysorbate 80 (P-80)-coated AuPd core-shell structure. Our results indicate that Concave cubic Qu@P-80@AuPd can activate autophagy of SH-SY5Y cells, promote the fusion of autophagosomes and lysosomes, accelerate the clearance of Aβ, and protect SH-SY5Y cells from Aβ-induced cytotoxicity damage. Furthermore, Concave cubic Qu@P-80@AuPd also has good biocompatibility and high blood-brain barrier (BBB) permeability. Therefore, we anticipate that Concave cubic Qu@P-80@AuPd will be used as a potential autophagy inducer to treat AD.
Collapse
|
83
|
Liebsch F, Kulic L, Teunissen C, Shobo A, Ulku I, Engelschalt V, Hancock MA, van der Flier WM, Kunach P, Rosa-Neto P, Scheltens P, Poirier J, Saftig P, Bateman RJ, Breitner J, Hock C, Multhaup G. Aβ34 is a BACE1-derived degradation intermediate associated with amyloid clearance and Alzheimer's disease progression. Nat Commun 2019; 10:2240. [PMID: 31110178 PMCID: PMC6527709 DOI: 10.1038/s41467-019-10152-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 04/17/2019] [Indexed: 12/13/2022] Open
Abstract
The beta-site APP cleaving enzyme 1 (BACE1) is known primarily for its initial cleavage of the amyloid precursor protein (APP), which ultimately leads to the generation of Aβ peptides. Here, we provide evidence that altered BACE1 levels and activity impact the degradation of Aβ40 and Aβ42 into a common Aβ34 intermediate. Using human cerebrospinal fluid (CSF) samples from the Amsterdam Dementia Cohort, we show that Aβ34 is elevated in individuals with mild cognitive impairment who later progressed to dementia. Furthermore, Aβ34 levels correlate with the overall Aβ clearance rates in amyloid positive individuals. Using CSF samples from the PREVENT-AD cohort (cognitively normal individuals at risk for Alzheimer’s disease), we further demonstrate that the Aβ34/Aβ42 ratio, representing Aβ degradation and cortical deposition, associates with pre-clinical markers of neurodegeneration. We propose that Aβ34 represents a marker of amyloid clearance and may be helpful for the characterization of Aβ turnover in clinical samples. Aβ34 is generated from degradation of Aβ40 and Aβ42 by β-secretase. Here, the authors show that Aβ34 is a marker for amyloid clearance and is elevated in the CSF of patients that go on to convert from mild cognitive impairment to Alzheimer’s disease, suggesting it may be a useful biomarker.
Collapse
Affiliation(s)
- Filip Liebsch
- Department of Pharmacology and Therapeutics and Integrated Program in Neuroscience, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Luka Kulic
- Institute for Regenerative Medicine, University of Zurich, CH-8952, Schlieren, Switzerland
| | - Charlotte Teunissen
- Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081HZ, Amsterdam, The Netherlands
| | - Adeola Shobo
- Department of Pharmacology and Therapeutics and Integrated Program in Neuroscience, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Irem Ulku
- Department of Pharmacology and Therapeutics and Integrated Program in Neuroscience, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Vivienne Engelschalt
- Institut für Chemie und Biochemie, Freie Universität Berlin, 14195, Berlin, Germany
| | - Mark A Hancock
- SPR-MS Facility, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, 1081HZ, The Netherlands
| | - Peter Kunach
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, McGill University, Montreal, H4H 1R3, QC, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute, McGill University, Montreal, H4H 1R3, QC, Canada
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, 1081HZ, The Netherlands
| | - Judes Poirier
- Department of Psychiatry, McGill University, Montreal, QC, H4H 1R3, Canada
| | - Paul Saftig
- Biochemisches Institut, Christian-Albrechts-Universität-Kiel, 24118, Kiel, Germany
| | - Randall J Bateman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - John Breitner
- Department of Psychiatry, McGill University, Montreal, QC, H4H 1R3, Canada
| | - Christoph Hock
- Institute for Regenerative Medicine, University of Zurich, CH-8952, Schlieren, Switzerland.,Neurimmune, CH-8952, Schlieren, Switzerland
| | - Gerhard Multhaup
- Department of Pharmacology and Therapeutics and Integrated Program in Neuroscience, McGill University, Montreal, QC, H3G 1Y6, Canada.
| |
Collapse
|
84
|
Zhao L, Zhao Y, Tang FL, Xiong L, Su C, Mei L, Zhu XJ, Xiong WC. pHluorin-BACE1-mCherry Acts as a Reporter for the Intracellular Distribution of Active BACE1 In Vitro and In Vivo. Cells 2019; 8:E474. [PMID: 31108937 PMCID: PMC6562731 DOI: 10.3390/cells8050474] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/09/2019] [Accepted: 05/15/2019] [Indexed: 12/23/2022] Open
Abstract
β-site APP-cleaving enzyme 1 (BACE1) initiates amyloid precursor protein (APP) cleavage and β-amyloid (Aβ) production, a critical step in the pathogenesis of Alzheimer's disease (AD). It is thus of considerable interest to investigate how BACE1 activity is regulated. BACE1 has its maximal activity at acidic pH and GFP variant-pHluorin-displays pH dependence. In light of these observations, we generated three tandem fluorescence-tagged BACE1 fusion proteins, named pHluorin-BACE1-mCherry, BACE1-mCherry-pHluorin and BACE1-mCherry-EGFP. Comparing the fluorescence characteristics of these proteins in response to intracellular pH changes induced by chloroquine or bafilomycin A1, we found that pHluorin-BACE1-mCherry is a better pH sensor for BACE1 because its fluorescence intensity responds to pH changes more dramatically and more quickly. Additionally, we found that (pro)renin receptor (PRR), a subunit of the v-ATPase complex, which is critical for maintaining vesicular pH, regulates pHluorin's fluorescence and BACE1 activity in pHluorin-BACE1-mCherry expressing cells. Finally, we found that the expression of Swedish mutant APP (APPswe) suppresses pHluorin fluorescence in pHluorin-BACE1-mCherry expressing cells in culture and in vivo, implicating APPswe not only as a substrate but also as an activator of BACE1. Taken together, these results suggest that the pHluorin-BACE1-mCherry fusion protein may serve as a useful tool for visualizing active/inactive BACE1 in culture and in vivo.
Collapse
Affiliation(s)
- Lu Zhao
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun 130024, China.
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Yang Zhao
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun 130024, China.
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Fu-Lei Tang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Lei Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Ce Su
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun 130024, China.
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Xiao-Juan Zhu
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Cytology and Genetics, Northeast Normal University, Changchun 130024, China.
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
85
|
Guix FX, Sartório CL, Ill-Raga G. BACE1 Translation: At the Crossroads Between Alzheimer's Disease Neurodegeneration and Memory Consolidation. J Alzheimers Dis Rep 2019; 3:113-148. [PMID: 31259308 PMCID: PMC6597968 DOI: 10.3233/adr-180089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human life unfolds not only in time and space, but also in the recollection and interweaving of memories. Therefore, individual human identity depends fully on a proper access to the autobiographical memory. Such access is hindered under pathological conditions such as Alzheimer’s disease, which affects millions of people worldwide. Unfortunately, no effective cure exists to prevent this disorder, the impact of which will rise alarmingly within the next decades. While Alzheimer’s disease is largely considered to be the outcome of amyloid-β (Aβ) peptide accumulation in the brain, conceiving this complex disorder strictly as the result of Aβ-neurotoxicity is perhaps a too straight-line simplification. Instead, complementary to this view, the tableau of molecular disarrangements in the Alzheimer’s disease brain may be reflecting, at least in part, a loss of function phenotype in memory processing. Here we take BACE1 translation and degradation as a gateway to study molecular mechanisms putatively involved in the transition between memory and neurodegeneration. BACE1 participates in the excision of Aβ-peptide from its precursor holoprotein, but plays a role in synaptic plasticity too. Its translation is governed by eIF2α phosphorylation: a hub integrating cellular responses to stress, but also a critical switch in memory consolidation. Paralleling these dualities, the eIF2α-kinase HRI has been shown to be a nitric oxide-dependent physiological activator of hippocampal BACE1 translation. Finally, beholding BACE1 as a representative protease active in the CNS, we venture a new perspective on the cellular basis of memory, which may incorporate neurodegeneration in itself as a drift in memory consolidating systems.
Collapse
Affiliation(s)
- Francesc X Guix
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa-CSIC, Madrid, Spain
| | - Carmem L Sartório
- Division of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Gerard Ill-Raga
- Division of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| |
Collapse
|
86
|
Flores-León M, Pérez-Domínguez M, González-Barrios R, Arias C. Palmitic Acid-Induced NAD + Depletion is Associated with the Reduced Function of SIRT1 and Increased Expression of BACE1 in Hippocampal Neurons. Neurochem Res 2019; 44:1745-1754. [PMID: 31073968 DOI: 10.1007/s11064-019-02810-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/29/2019] [Accepted: 05/02/2019] [Indexed: 12/15/2022]
Abstract
Increased levels of circulating fatty acids, such as palmitic acid (PA), are associated with the development of obesity, insulin resistance, type-2 diabetes and metabolic syndrome. Furthermore, these diseases are linked to an increased risk of cancer, cardiovascular diseases, mild cognitive impairment and even Alzheimer's disease (AD). However, the precise actions of elevated PA levels on neurons and their association with neuronal metabolic disruption that leads to the expression of pathological markers of AD, such as the overproduction and accumulation of the amyloid-β peptide, represent an area of intense investigation. A possible molecular mechanism involved in the effects of PA may be through dysfunction of the NAD+ sensor enzyme, SIRT1. Therefore, the aim of the present study was to analyze the relationship between the effects of PA metabolism on the function of SIRT1 and the upregulation of BACE1 in cultured hippocampal neurons. PA reduced the total amount of NAD+ in neurons that caused an increase in p65 K310 acetylation due to inhibition of SIRT1 activity and low protein content. Furthermore, BACE1 protein and its activity were increased, and BACE1 was relocated in neurites after PA exposure.
Collapse
Affiliation(s)
- Manuel Flores-León
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico
| | - Martha Pérez-Domínguez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico
| | - Rodrigo González-Barrios
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología (INCan)-Instituto de Investigaciones Biomédicas (IIB), Universidad Nacional Autónoma de México (UNAM), 14080, México, DF, Mexico
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico.
| |
Collapse
|
87
|
Smith AJ, Duan T, Verkman AS. Aquaporin-4 reduces neuropathology in a mouse model of Alzheimer's disease by remodeling peri-plaque astrocyte structure. Acta Neuropathol Commun 2019; 7:74. [PMID: 31068220 PMCID: PMC6506955 DOI: 10.1186/s40478-019-0728-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 04/23/2019] [Indexed: 01/01/2023] Open
Abstract
Redistribution of the water channel aquaporin-4 (AQP4) away from astrocyte endfeet and into parenchymal processes is a striking histological feature in mouse models of Alzheimer’s disease (AD) and other neurological conditions with prominent astrogliosis. AQP4 redistribution has been proposed to impair bulk Aβ clearance in AD, resulting in increased amyloid deposition in the brain; however, this finding is controversial. Here, we provide evidence in support of a different and novel role of AQP4 in AD. We found that Aqp4 deletion significantly increased amyloid deposition in cerebral cortex of 5xFAD mice, with an increase in the relative number of fibrillar vs. dense core plaques. AQP4 deficient 5xFAD mice also showed a significant reduction in the density of GFAP labeled peri-plaque astrocyte processes. Microglial plaque coverage was also significantly reduced, suggesting astrocyte involvement in organizing the peri-plaque glial response. The alterations in peri-plaque glial structure were accompanied by increased neuronal uptake of Aβ and an increase in the number of dystrophic neurites surrounding plaques. On the basis of these findings, we propose that redistribution of AQP4 into the parenchymal processes facilitates astrocyte structural plasticity and the formation of a reactive glial net around plaques that protects neurons from the deleterious effects of Aβ aggregates. AQP4 redistribution may thus facilitate plaque containment and reduce neuropathology in AD.
Collapse
|
88
|
MiR-16 attenuates β-amyloid-induced neurotoxicity through targeting β-site amyloid precursor protein-cleaving enzyme 1 in an Alzheimer's disease cell model. Neuroreport 2019; 29:1365-1372. [PMID: 30142113 DOI: 10.1097/wnr.0000000000001118] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The aberrant deposition of β-amyloid (Aβ) is closely linked to the pathogenesis and development of Alzheimer's disease (AD). MiR-16 was abnormally downregulated and may be related to the development of AD. However, the functional role and molecular mechanism of miR-16 in AD pathogenesis are still not well elucidated. The expressions of miR-16 and β-site amyloid precursor protein-cleaving enzyme 1 (BACE1) mRNA and protein levels in AD brain tissues and Aβ-treated PC12 cellular AD model were examined by qRT-PCR and western blot analyses. Luciferase reporter assay was used to verify the potential target of miR-16. The cell viability, apoptosis, and caspase-3 activity in PC12 cells were determined by the MTT assay, flow cytometry analysis, and caspase-3 activity assay, respectively. Downregulation of miR-16 and upregulation of BACE1 existed in AD tissues and the cellular AD model of PC12. In addition, miR-16 directly suppressed BACE1 expression. Moreover, miR-16 overexpression and BACE1 knockdown facilitated Aβ-induced cell toxicity, apoptosis, and caspase-3 activity in N2a cells, which was partially eliminated by overexpression of BACE1. In contrast, BACE1 knockdown reversed the miR-16 inhibition-mediated inhibitory effect on Aβ-induced cell toxicity, apoptosis, and caspase-3 activity in PC12 cells. Collectively, miR-16 attenuated Aβ-induced neurotoxicity through targeting BACE1 in an Aβ insult cellular AD model, providing a potential therapeutic target for AD treatment.
Collapse
|
89
|
Schaeverbeke J, Gille B, Adamczuk K, Vanderstichele H, Chassaing E, Bruffaerts R, Neyens V, Stoops E, Tournoy J, Vandenberghe R, Poesen K. Cerebrospinal fluid levels of synaptic and neuronal integrity correlate with gray matter volume and amyloid load in the precuneus of cognitively intact older adults. J Neurochem 2019; 149:139-157. [PMID: 30720873 DOI: 10.1111/jnc.14680] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 12/10/2018] [Accepted: 02/01/2019] [Indexed: 12/18/2022]
Abstract
The main pathophysiological alterations of Alzheimer's disease (AD) include loss of neuronal and synaptic integrity, amyloidogenic processing, and neuroinflammation. Similar alterations can, however, also be observed in cognitively intact older subjects and may prelude the clinical manifestation of AD. The objectives of this prospective cross-sectional study in a cohort of 38 cognitively intact older adults were twofold: (i) to investigate the latent relationship among cerebrospinal fluid (CSF) biomarkers reflecting the main pathophysiological processes of AD, and (ii) to assess the correlation between these biomarkers and gray matter volume as well as amyloid load. All subjects underwent extensive neuropsychological examinations, CSF sampling, [18 F]-flutemetamol amyloid positron emission tomography, and T1 -weighted magnetic resonance imaging. A factor analysis revealed one factor that explained most of the variance in the CSF biomarker dataset clustering t-tau, α-synuclein, p-tau181 , neurogranin, BACE1, visinin-like protein 1, chitinase-3-like protein 1 (YKL-40), Aβ1-40 and Aβ1-38 . Higher scores on this factor correlated with lower gray matter volume and with higher amyloid load in the precuneus. At the level of individual CSF biomarkers, levels of visinin-like protein 1, neurogranin, BACE1, Aβ1-40 , Aβ1-38, and YKL-40 all correlated inversely with gray matter volume of the precuneus. These findings demonstrate that in cognitively intact older subjects, CSF levels of synaptic and neuronal integrity biomarkers, amyloidogenic processing and measures of innate immunity (YKL-40) display a latent structure of common variance, which is associated with loss of structural integrity of brain regions implicated in the earliest stages of AD. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript, and for *Preregistration* because the study was pre-registered at https://osf.io/7qm9t/. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Jolien Schaeverbeke
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Alzheimer Research Centre KU Leuven, Leuven Institute of Neuroscience and Disease, Leuven, Belgium
| | - Benjamin Gille
- Laboratory for Molecular Neurobiomarker Research, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Department of Chronic disease, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Katarzyna Adamczuk
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Alzheimer Research Centre KU Leuven, Leuven Institute of Neuroscience and Disease, Leuven, Belgium.,Bioclinica LAB, Newark, California, USA
| | | | | | - Rose Bruffaerts
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Veerle Neyens
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Alzheimer Research Centre KU Leuven, Leuven Institute of Neuroscience and Disease, Leuven, Belgium
| | | | - Jos Tournoy
- Alzheimer Research Centre KU Leuven, Leuven Institute of Neuroscience and Disease, Leuven, Belgium.,Department of Chronic disease, Metabolism and Ageing, KU Leuven, Leuven, Belgium.,Department of Geriatric Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Alzheimer Research Centre KU Leuven, Leuven Institute of Neuroscience and Disease, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Koen Poesen
- Laboratory for Molecular Neurobiomarker Research, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Department of Chronic disease, Metabolism and Ageing, KU Leuven, Leuven, Belgium.,Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
90
|
Abstract
Alzheimer's disease (AD), the most common cause of age-dependent dementia, is one of the most significant healthcare problems worldwide. Aggravating this situation, drugs that are currently US Food and Drug Administration (FDA)-approved for AD treatment do not prevent or delay disease progression. Therefore, developing effective therapies for AD patients is of critical urgency. Human genetic and clinical studies over the past three decades have indicated that abnormal generation or accumulation of amyloid-β (Aβ) peptides is a likely culprit in AD pathogenesis. Aβ is generated from amyloid precursor protein (APP) via proteolytic cleavage by β-site APP cleaving enzyme 1 (BACE1) (memapsin 2, β-secretase, Asp 2 protease) and γ-secretase. Mice deficient in BACE1 show abrogated production of Aβ. Therefore, pharmacological inhibition of BACE1 is being intensively pursued as a therapeutic approach to treat AD patients. Recent setbacks in clinical trials with BACE1 inhibitors have highlighted the critical importance of understanding how to properly inhibit BACE1 to treat AD patients. This review summarizes the recent studies on the role of BACE1 in synaptic functions as well as our views on BACE1 inhibition as an effective AD treatment.
Collapse
Affiliation(s)
- Brati Das
- Department of Neuroscience, Room E4032, UConn Health, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA
| | - Riqiang Yan
- Department of Neuroscience, Room E4032, UConn Health, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA.
| |
Collapse
|
91
|
Kidana K, Tatebe T, Ito K, Hara N, Kakita A, Saito T, Takatori S, Ouchi Y, Ikeuchi T, Makino M, Saido TC, Akishita M, Iwatsubo T, Hori Y, Tomita T. Loss of kallikrein-related peptidase 7 exacerbates amyloid pathology in Alzheimer's disease model mice. EMBO Mol Med 2019; 10:emmm.201708184. [PMID: 29311134 PMCID: PMC5840542 DOI: 10.15252/emmm.201708184] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Deposition of amyloid‐β (Aβ) as senile plaques is one of the pathological hallmarks in the brains of Alzheimer's disease (AD) patients. In addition, glial activation has been found in AD brains, although the precise pathological role of astrocytes remains unclear. Here, we identified kallikrein‐related peptidase 7 (KLK7) as an astrocyte‐derived Aβ degrading enzyme. Expression of KLK7 mRNA was significantly decreased in the brains of AD patients. Ablation of Klk7 exacerbated the thioflavin S‐positive Aβ pathology in AD model mice. The expression of Klk7 was upregulated by Aβ treatment in the primary astrocyte, suggesting that Klk7 is homeostatically modulated by Aβ‐induced responses. Finally, we found that the Food and Drug Administration‐approved anti‐dementia drug memantine can increase the expression of Klk7 and Aβ degradation activity specifically in the astrocytes. These data suggest that KLK7 is an important enzyme in the degradation and clearance of deposited Aβ species by astrocytes involved in the pathogenesis of AD.
Collapse
Affiliation(s)
- Kiwami Kidana
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Internal Medicine, Komeikai Hospital, Tokyo, Japan
| | - Takuya Tatebe
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kaori Ito
- Venture Science Laboratories, R&D Division, Daiichi-Sankyo Co. Ltd., Tokyo, Japan
| | - Norikazu Hara
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan
| | - Sho Takatori
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuyoshi Ouchi
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Federation of National Public Service Personnel Mutual Aid Associations, Toranomon Hospital, Tokyo, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Mitsuhiro Makino
- Venture Science Laboratories, R&D Division, Daiichi-Sankyo Co. Ltd., Tokyo, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan
| | - Masahiro Akishita
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukiko Hori
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
92
|
Vnencak M, Schölvinck ML, Schwarzacher SW, Deller T, Willem M, Jedlicka P. Lack of β-amyloid cleaving enzyme-1 (BACE1) impairs long-term synaptic plasticity but enhances granule cell excitability and oscillatory activity in the dentate gyrus in vivo. Brain Struct Funct 2019; 224:1279-1290. [PMID: 30701309 DOI: 10.1007/s00429-019-01836-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 01/16/2019] [Indexed: 12/11/2022]
Abstract
BACE1 is a β-secretase involved in the cleavage of amyloid precursor protein and the pathogenesis of Alzheimer's disease (AD). The entorhinal cortex and the dentate gyrus are important for learning and memory, which are affected in the early stages of AD. Since BACE1 is a potential target for AD therapy, it is crucial to understand its physiological role in these brain regions. Here, we examined the function of BACE1 in the dentate gyrus. We show that loss of BACE1 in the dentate gyrus leads to increased granule cell excitability, indicated by enhanced efficiency of synaptic potentials to generate granule cell spikes. The increase in granule cell excitability was accompanied by prolonged paired-pulse inhibition, altered network gamma oscillations, and impaired synaptic plasticity at entorhinal-dentate synapses of the perforant path. In summary, this is the first detailed electrophysiological study of BACE1 deletion at the network level in vivo. The results suggest that BACE1 is important for normal dentate gyrus network function. This has implications for the use of BACE1 inhibitors as therapeutics for AD therapy, since BACE1 inhibition could similarly disrupt synaptic plasticity and excitability in the entorhinal-dentate circuitry.
Collapse
Affiliation(s)
- Matej Vnencak
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt am Main, Germany. .,Otorhinolaryngology, Head and Neck Surgery, Turku University Hospital, University of Turku, PL 52, 20521, Turku, Finland.
| | - Marieke L Schölvinck
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Frankfurt am Main, Germany
| | - Stephan W Schwarzacher
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt am Main, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt am Main, Germany
| | - Michael Willem
- BioMedical Center, Biochemistry, Ludwig-Maximilians-University, Munich, Germany
| | - Peter Jedlicka
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University, Frankfurt am Main, Germany. .,ICAR3R-Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus-Liebig-University, Rudolf-Buchheim-Str. 6, 35392, Giessen, Germany.
| |
Collapse
|
93
|
Consequences of Pharmacological BACE Inhibition on Synaptic Structure and Function. Biol Psychiatry 2018; 84:478-487. [PMID: 29945719 DOI: 10.1016/j.biopsych.2018.04.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 04/28/2018] [Accepted: 04/28/2018] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease is the most prevalent neurodegenerative disorder among elderly persons. Overt accumulation and aggregation of the amyloid-β peptide (Aβ) is thought to be the initial causative factor for Alzheimer's disease. Aβ is produced by sequential proteolytic cleavage of the amyloid precursor protein. Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) is the initial and rate-limiting protease for the generation of Aβ. Therefore, inhibiting BACE1 is considered one of the most promising therapeutic approaches for potential treatment of Alzheimer's disease. Currently, several drugs blocking this enzyme (BACE inhibitors) are being evaluated in clinical trials. However, high-dosage BACE-inhibitor treatment interferes with structural and functional synaptic plasticity in mice. These adverse side effects may mask the therapeutic benefit of lowering the Aβ concentration. In this review, we focus on the consequences of BACE inhibition-mediated synaptic deficits and the potential clinical implications.
Collapse
|
94
|
Antidiabetic and Neuroprotective Effect of the N-Butanol Extract of Fragaria nilgerrensis Schlecht. in STZ-Induced Diabetic Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:6938370. [PMID: 30254687 PMCID: PMC6142753 DOI: 10.1155/2018/6938370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/28/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022]
Abstract
Diabetes has been associated with neurodegenerative disorders that are accompanied by memory loss and cognitive impairments, but there is no effective treatment for it at present. Fragaria nilgerrensis Schlecht. (FNS), a well-known Chinese materia medica, has been traditionally used for the folkloric treatment of diabetes and other diseases. However, its effects are poorly documented. Here, we investigated the antidiabetic and neuroprotective effect of FNS in diabetic mice. Thin layer chromatography (TLC) and high performance liquid chromatography (HPLC) evaluations of N-butanol extract of Fragaria nilgerrensis Schlecht. (N-FNS) showed the presence of flavonoid and its structure is similar to scutellarin. For the first time, we show the potential neuroprotective and antidiabetic effects of FNS. After 4 weeks of FNS intervention, a significant decrease in blood glucose, increase in body weight, and amelioration in glucose tolerance were observed in FNS treated diabetic mice. In the acute study, FNS enhanced motor activity in the open field task and significantly prevented spatial-learning deficits in Morris water maze tests. Besides, synapse ultrastructure of the hippocampus showed that the mitochondrial morphology was basically restored and all the synaptic structural parameters were gradually normalized after treatment with FNS. Importantly, we found that the activities of SOD and CAT in liver and hippocampus of diabetic mice significantly increased after FNS administration. In vitro, FNS and scutellarin showed high DPPH radical scavenging activity. The study suggests that FNS exerted significant antidiabetic and neuroprotective effects which may be attributed to its antioxidant property.
Collapse
|
95
|
Ou-Yang MH, Kurz JE, Nomura T, Popovic J, Rajapaksha TW, Dong H, Contractor A, Chetkovich DM, Tourtellotte WG, Vassar R. Axonal organization defects in the hippocampus of adult conditional BACE1 knockout mice. Sci Transl Med 2018; 10:eaao5620. [PMID: 30232227 PMCID: PMC11017370 DOI: 10.1126/scitranslmed.aao5620] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 03/09/2018] [Accepted: 05/21/2018] [Indexed: 12/14/2022]
Abstract
β-Site APP (amyloid precursor protein) cleaving enzyme 1 (BACE1) is the β-secretase enzyme that initiates production of the toxic amyloid-β peptide that accumulates in the brains of patients with Alzheimer's disease (AD). Hence, BACE1 is a prime therapeutic target, and several BACE1 inhibitor drugs are currently being tested in clinical trials for AD. However, the safety of BACE1 inhibition is unclear. Germline BACE1 knockout mice have multiple neurological phenotypes, although these could arise from BACE1 deficiency during development. To address this question, we report that tamoxifen-inducible conditional BACE1 knockout mice in which the Bace1 gene was ablated in the adult largely lacked the phenotypes observed in germline BACE1 knockout mice. However, one BACE1-null phenotype was induced after Bace1 gene deletion in the adult mouse brain. This phenotype showed reduced length and disorganization of the hippocampal mossy fiber infrapyramidal bundle, the axonal pathway of dentate gyrus granule cells that is maintained by neurogenesis in the mouse brain. This defect in axonal organization correlated with reduced BACE1-mediated cleavage of the neural cell adhesion protein close homolog of L1 (CHL1), which has previously been associated with axon guidance. Although our results indicate that BACE1 inhibition in the adult mouse brain may avoid phenotypes associated with BACE1 deficiency during embryonic and postnatal development, they also suggest that BACE1 inhibitor drugs developed for treating AD may disrupt the organization of an axonal pathway in the hippocampus, an important structure for learning and memory.
Collapse
Affiliation(s)
- Ming-Hsuan Ou-Yang
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jonathan E Kurz
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Toshihiro Nomura
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Jelena Popovic
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Tharinda W Rajapaksha
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Anis Contractor
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Dane M Chetkovich
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Warren G Tourtellotte
- Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Division of Neuropathology, Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Robert Vassar
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
96
|
Ba L, Chen XH, Chen YL, Nie Q, Li ZJ, Ding FF, Zhang M. Distinct Rab7-related Endosomal-Autophagic-Lysosomal Dysregulation Observed in Cortex and Hippocampus in APPswe/PSEN1dE9 Mouse Model of Alzheimer's Disease. Chin Med J (Engl) 2018; 130:2941-2950. [PMID: 29237927 PMCID: PMC5742922 DOI: 10.4103/0366-6999.220311] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Amyloid-β deposition and accumulation of autophagic vacuoles are pathologic features of Alzheimer's disease (AD). Dysregulation of the endosomal-autophagic-lysosomal (EAL) pathway, which impairs amyloid precursor protein processing, is one of the earliest changes in AD. However, the precise role of EAL pathway in neurodegeneration remains unclear. This study aimed to investigate the role of EAL pathway in AD and further study the mechanism of EAL dysfunction. METHODS We used 3-, 7-, and 12-month-old APPswe/PSEN1dE9 (APP/PS1) mice to model different stages of AD with age- and gender-matched wild-type littermates as controls (4-7 mice per group) and detected the changes of EAL markers, endosomal organizers Rab5 and Rab7, autophagosome marker LC3B, and lysosomal proteins Lamp1/2 in cortex and hippocampus by immunohistochemistry and Western blotting analysis. To further explore the mechanism of EAL dysregulation in AD, components of the class III phosphatidylinositol 3-kinase (PI3KC3) complex, activators of Rab7 (Beclin1 and UVRAG), and the negative regulator of Rab7 (Rubicon) were also measured in this two brain regions. RESULTS In 7-month-old APP/PS1 brain that amyloid beta initiated to accumulate intracellularly, EAL pathway, and related PI3KC3 members, UVRAG and Beclin1 were upregulated both in cortex and hippocampus (all P < 0.05). By the age of 12 months old, when abundant amyloid plaques formed, EAL markers, UVRAG, and Beclin1 were also upregulated in the cortex (all P < 0.05). However, Rab7 was decreased significantly (P = 0.0447), accompanied by a reduction of its activating PI3KC complex component Beclin1 (P = 0.0215) and enhancement of its inhibiting component Rubicon (P = 0.0055) in the hippocampus. CONCLUSIONS Our study implies that EAL pathway, represented as Rab7 and its PI3KC3 regulators' expressions, showed temporal and spatial variation in brains at different stages of AD. It provides new insights into the role of EAL pathway in pathogenesis and indicates potential therapeutic targets in neurodegenerative diseases.
Collapse
Affiliation(s)
- Li Ba
- Department of Neurology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiao-Hua Chen
- Department of Neurology, Huangshi Central Hospital, Huangshi, Hubei 435000, China
| | - Yan-Lin Chen
- Department of Neurology, Wuhan Central Hospital, Wuhan, Hubei 430014, China
| | - Qing Nie
- Department of Neurology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhi-Jun Li
- Department of Neurology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Feng-Fei Ding
- Department of Neurology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Min Zhang
- Department of Neurology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
97
|
Schipke CG, De Vos A, Fuentes M, Jacobs D, Vanmechelen E, Peters O. Neurogranin and BACE1 in CSF as Potential Biomarkers Differentiating Depression with Cognitive Deficits from Early Alzheimer's Disease: A Pilot Study. Dement Geriatr Cogn Dis Extra 2018; 8:277-289. [PMID: 30186306 PMCID: PMC6120408 DOI: 10.1159/000489847] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/04/2018] [Indexed: 12/20/2022] Open
Abstract
Background/Aims Major depressive disorder (MDD) can cooccur with early Alzheimer's disease (AD) or may cause memory problems independently of AD. Previous studies have suggested that the AD-related cerebrospinal fluid (CSF) biomarkers tau and Aβ(1–42) could help discriminate between early AD and depression unrelated to AD. Moreover, the postsynaptic protein neurogranin and presynaptic BACE1 have increasingly gained attention as potential new AD biomarkers, but they have not yet been investigated concerning depression. Methods Using ELISAs, we studied CSF neurogranin and BACE1 levels in patients with mild (n = 21) and moderate (n = 19) AD, as well as in MDD patients with (n = 20) and without (n = 20) cognitive deficits. The clinical examinations included analyses of t-tau, Aβ(1–42), and Aβ(1–40), besides neuropsychological tests and cranial magnetic resonance imaging. Depressive symptom severity was assessed using the Geriatric Depression Scale (GDS). Results Along with classic AD biomarkers, neurogranin and BACE1 CSF levels differed between moderate AD and MDD (p ≤ 0.01). MDD associated with cognitive deficits was distinguished from mild AD through the CSF neurogranin/BACE1 ratio (p < 0.05), which was strongly correlated with GDS scores (ρ = −0.656; p < 0.01). Conclusion The neurogranin/BACE1 ratio in CSF can distinguish between depression and AD among patients with similar cognitive deficits, along with the classic AD biomarkers. Further longitudinal studies are ongoing to identify which biomarkers have prognostic value.
Collapse
Affiliation(s)
- Carola G Schipke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Neuropathology, Berlin, Germany
| | | | - Manuel Fuentes
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH) and German Center for Neurodegenerative Diseases (DZNE), Department of Psychiatry and Psychotherapy, Berlin, Germany
| | - Dirk Jacobs
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Neuropathology, Berlin, Germany
| | - Eugeen Vanmechelen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Neuropathology, Berlin, Germany
| | - Oliver Peters
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH) and German Center for Neurodegenerative Diseases (DZNE), Department of Psychiatry and Psychotherapy, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| |
Collapse
|
98
|
Lichtenthaler SF, Lemberg MK, Fluhrer R. Proteolytic ectodomain shedding of membrane proteins in mammals-hardware, concepts, and recent developments. EMBO J 2018; 37:e99456. [PMID: 29976761 PMCID: PMC6068445 DOI: 10.15252/embj.201899456] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/05/2018] [Accepted: 06/18/2018] [Indexed: 12/14/2022] Open
Abstract
Proteolytic removal of membrane protein ectodomains (ectodomain shedding) is a post-translational modification that controls levels and function of hundreds of membrane proteins. The contributing proteases, referred to as sheddases, act as important molecular switches in processes ranging from signaling to cell adhesion. When deregulated, ectodomain shedding is linked to pathologies such as inflammation and Alzheimer's disease. While proteases of the "a disintegrin and metalloprotease" (ADAM) and "beta-site APP cleaving enzyme" (BACE) families are widely considered as sheddases, in recent years a much broader range of proteases, including intramembrane and soluble proteases, were shown to catalyze similar cleavage reactions. This review demonstrates that shedding is a fundamental process in cell biology and discusses the current understanding of sheddases and their substrates, molecular mechanisms and cellular localizations, as well as physiological functions of protein ectodomain shedding. Moreover, we provide an operational definition of shedding and highlight recent conceptual advances in the field. While new developments in proteomics facilitate substrate discovery, we expect that shedding is not a rare exception, but rather the rule for many membrane proteins, and that many more interesting shedding functions await discovery.
Collapse
Affiliation(s)
- Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, Klinikum rechts der Isar, School of Medicine, and Institute for Advanced Study, Technical University Munich, Munich, Germany
- Munich Center for Systems Neurology (SyNergy), Munich, Germany
| | - Marius K Lemberg
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Regina Fluhrer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Biomedizinisches Centrum (BMC), Ludwig-Maximilians University of Munich, Munich, Germany
| |
Collapse
|
99
|
Ovsepian SV, O'Leary VB, Zaborszky L, Ntziachristos V, Dolly JO. Amyloid Plaques of Alzheimer's Disease as Hotspots of Glutamatergic Activity. Neuroscientist 2018; 25:288-297. [PMID: 30051750 DOI: 10.1177/1073858418791128] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Deposition of amyloid plaques in limbic and associative cortices is amongst the most recognized histopathologic hallmarks of Alzheimer's disease. Despite decades of research, there is a lack of consensus over the impact of plaques on neuronal function, with their role in cognitive decline and memory loss undecided. Evidence has emerged suggesting complex and localized axonal pathology around amyloid plaques, with a significant fraction of swellings and dystrophies becoming enriched with putative synaptic vesicles and presynaptic proteins normally colocalized at hotspots of transmitter release. In the absence of hallmark active zone proteins and postsynaptic receptive elements, the axonal swellings surrounding amyloid plaques have been suggested as sites for ectopic release of glutamate, which under reduced clearance can lead to elevated local excitatory drive. Throughout this review, we consider the emerging data suggestive of amyloid plaques as hotspots of compulsive glutamatergic activity. Evidence for local and long-range effects of nonsynaptic glutamate is discussed in the context of circuit dysfunctions and neurodegenerative changes of Alzheimer's disease.
Collapse
Affiliation(s)
- Saak V Ovsepian
- Institute for Biological and Medical Imaging, Helmholtz Zentrum Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Munich School of Bioengineering, Technical University Munich, Munich, Germany.,International Centre for Neurotherapeutics, Dublin City University, Dublin, Ireland
| | - Valerie B O'Leary
- International Centre for Neurotherapeutics, Dublin City University, Dublin, Ireland
| | - Laszlo Zaborszky
- Center for Molecular and Behavioral Neuroscience, Rutgers, the State University of New Jersey, Newark, NJ, USA
| | - Vasilis Ntziachristos
- Institute for Biological and Medical Imaging, Helmholtz Zentrum Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Munich School of Bioengineering, Technical University Munich, Munich, Germany
| | - J Oliver Dolly
- International Centre for Neurotherapeutics, Dublin City University, Dublin, Ireland
| |
Collapse
|
100
|
Karch S, Broichhagen J, Schneider J, Böning D, Hartmann S, Schmid B, Tripal P, Palmisano R, Alzheimer C, Johnsson K, Huth T. A New Fluorogenic Small-Molecule Labeling Tool for Surface Diffusion Analysis and Advanced Fluorescence Imaging of β-Site Amyloid Precursor Protein-Cleaving Enzyme 1 Based on Silicone Rhodamine: SiR-BACE1. J Med Chem 2018; 61:6121-6139. [PMID: 29939737 DOI: 10.1021/acs.jmedchem.8b00387] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
β-site APP-cleaving enzyme 1 (BACE1) is a major player in the pathogenesis of Alzheimer's disease. Structural and functional fluorescence microscopy offers a powerful approach to learn about the physiology and pathophysiology of this protease. Up to now, however, common labeling techniques require genetic manipulation, use large antibodies, or are not compatible with live cell imaging. Fluorescent small molecules that specifically bind to the protein of interest can overcome these limitations. Herein, we introduce SiR-BACE1, a conjugate of the BACE1 inhibitor S-39 and SiR647, as a novel fluorogenic, tag-free, and antibody-free label for BACE1. We present its chemical development, characterize its photophysical and pharmacologic properties, and evaluate its behavior in solution, in overexpression systems, and in native brain tissue. We demonstrate its applicability in confocal, stimulated emission depletion and dynamic single-molecule microscopy. The first functional studies with SiR-BACE1 on the surface mobility of BACE1 revealed a markedly confined diffusion pattern.
Collapse
Affiliation(s)
- Sandra Karch
- Institute of Physiology and Pathophysiology , Friedrich-Alexander-Universität Erlangen-Nürnberg , Universitaetsstrasse 17 , 91054 Erlangen , Germany
| | - Johannes Broichhagen
- Department of Chemical Biology , Max Planck Institute for Medical Research , Jahnstrasse 29 , 69120 Heidelberg , Germany.,Laboratory of Protein Engineering, Institut des Sciences et Ingénierie Chimiques, Sciences de Base , École Polytechnique Fédérale Lausanne , 1015 Lausanne , Switzerland
| | - Julia Schneider
- Institute of Physiology and Pathophysiology , Friedrich-Alexander-Universität Erlangen-Nürnberg , Universitaetsstrasse 17 , 91054 Erlangen , Germany
| | - Daniel Böning
- Max Planck Institute for the Science of Light , Staudtstrasse 2 , 91058 Erlangen , Germany
| | - Stephanie Hartmann
- Institute of Physiology and Pathophysiology , Friedrich-Alexander-Universität Erlangen-Nürnberg , Universitaetsstrasse 17 , 91054 Erlangen , Germany
| | - Benjamin Schmid
- Optical Imaging Centre , Friedrich-Alexander-Universität Erlangen-Nürnberg , Hartmannstrasse 14 , 91052 Erlangen , Germany
| | - Philipp Tripal
- Optical Imaging Centre , Friedrich-Alexander-Universität Erlangen-Nürnberg , Hartmannstrasse 14 , 91052 Erlangen , Germany
| | - Ralf Palmisano
- Optical Imaging Centre , Friedrich-Alexander-Universität Erlangen-Nürnberg , Hartmannstrasse 14 , 91052 Erlangen , Germany
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology , Friedrich-Alexander-Universität Erlangen-Nürnberg , Universitaetsstrasse 17 , 91054 Erlangen , Germany
| | - Kai Johnsson
- Department of Chemical Biology , Max Planck Institute for Medical Research , Jahnstrasse 29 , 69120 Heidelberg , Germany.,Laboratory of Protein Engineering, Institut des Sciences et Ingénierie Chimiques, Sciences de Base , École Polytechnique Fédérale Lausanne , 1015 Lausanne , Switzerland
| | - Tobias Huth
- Institute of Physiology and Pathophysiology , Friedrich-Alexander-Universität Erlangen-Nürnberg , Universitaetsstrasse 17 , 91054 Erlangen , Germany
| |
Collapse
|