51
|
Martinez NW, Gómez FE, Matus S. The Potential Role of Protein Kinase R as a Regulator of Age-Related Neurodegeneration. Front Aging Neurosci 2021; 13:638208. [PMID: 33994991 PMCID: PMC8113420 DOI: 10.3389/fnagi.2021.638208] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 03/10/2021] [Indexed: 01/25/2023] Open
Abstract
There is a growing evidence describing a decline in adaptive homeostasis in aging-related diseases affecting the central nervous system (CNS), many of which are characterized by the appearance of non-native protein aggregates. One signaling pathway that allows cell adaptation is the integrated stress response (ISR), which senses stress stimuli through four kinases. ISR activation promotes translational arrest through the phosphorylation of the eukaryotic translation initiation factor 2 alpha (eIF2α) and the induction of a gene expression program to restore cellular homeostasis. However, depending on the stimulus, ISR can also induce cell death. One of the ISR sensors is the double-stranded RNA-dependent protein kinase [protein kinase R (PKR)], initially described as a viral infection sensor, and now a growing evidence supports a role for PKR on CNS physiology. PKR has been largely involved in the Alzheimer’s disease (AD) pathological process. Here, we reviewed the antecedents supporting the role of PKR on the efficiency of synaptic transmission and cognition. Then, we review PKR’s contribution to AD and discuss the possible participation of PKR as a player in the neurodegenerative process involved in aging-related pathologies affecting the CNS.
Collapse
Affiliation(s)
- Nicolás W Martinez
- Fundación Ciencia & Vida, Santiago, Chile.,Departamento de Ciencias Básicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | | | - Soledad Matus
- Fundación Ciencia & Vida, Santiago, Chile.,Departamento de Ciencias Básicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| |
Collapse
|
52
|
Hampel H, Vassar R, De Strooper B, Hardy J, Willem M, Singh N, Zhou J, Yan R, Vanmechelen E, De Vos A, Nisticò R, Corbo M, Imbimbo BP, Streffer J, Voytyuk I, Timmers M, Tahami Monfared AA, Irizarry M, Albala B, Koyama A, Watanabe N, Kimura T, Yarenis L, Lista S, Kramer L, Vergallo A. The β-Secretase BACE1 in Alzheimer's Disease. Biol Psychiatry 2021; 89:745-756. [PMID: 32223911 PMCID: PMC7533042 DOI: 10.1016/j.biopsych.2020.02.001] [Citation(s) in RCA: 410] [Impact Index Per Article: 102.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/29/2020] [Accepted: 02/05/2020] [Indexed: 01/18/2023]
Abstract
BACE1 (beta-site amyloid precursor protein cleaving enzyme 1) was initially cloned and characterized in 1999. It is required for the generation of all monomeric forms of amyloid-β (Aβ), including Aβ42, which aggregates into bioactive conformational species and likely initiates toxicity in Alzheimer's disease (AD). BACE1 concentrations and rates of activity are increased in AD brains and body fluids, thereby supporting the hypothesis that BACE1 plays a critical role in AD pathophysiology. Therefore, BACE1 is a prime drug target for slowing down Aβ production in early AD. Besides the amyloidogenic pathway, BACE1 has other substrates that may be important for synaptic plasticity and synaptic homeostasis. Indeed, germline and adult conditional BACE1 knockout mice display complex neurological phenotypes. Despite BACE1 inhibitor clinical trials conducted so far being discontinued for futility or safety reasons, BACE1 remains a well-validated therapeutic target for AD. A safe and efficacious compound with high substrate selectivity as well as a more accurate dose regimen, patient population, and disease stage may yet be found. Further research should focus on the role of Aβ and BACE1 in physiological processes and key pathophysiological mechanisms of AD. The functions of BACE1 and the homologue BACE2, as well as the biology of Aβ in neurons and glia, deserve further investigation. Cellular and molecular studies of BACE1 and BACE2 knockout mice coupled with biomarker-based human research will help elucidate the biological functions of these important enzymes and identify their substrates and downstream effects. Such studies will have critical implications for BACE1 inhibition as a therapeutic approach for AD.
Collapse
Affiliation(s)
- Harald Hampel
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey; Sorbonne University, GRC No. 21, Alzheimer Precision Medicine, Pitié-Salpêtrière Hospital, Paris, France.
| | - Robert Vassar
- Department of Neurology, Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Bart De Strooper
- Department of Neurosciences, KU Leuven, Leuven, Belgium; Centre for Brain and Disease Research, VIB (Flanders Institute for Biotechnology), Leuven, Belgium; Dementia Research Institute, University College London, London, United Kingdom
| | - John Hardy
- Department of Molecular Neuroscience and Reta Lilla Weston Laboratories, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Michael Willem
- Chair of Metabolic Biochemistry, Biomedical Center, Faculty of Medicine, Ludwig Maximilians University Munich, Munich, Germany
| | - Neeraj Singh
- Department of Neuroscience, University of Connecticut Health, Farmington, Connecticut
| | - John Zhou
- Department of Neuroscience, University of Connecticut Health, Farmington, Connecticut
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health, Farmington, Connecticut
| | | | | | - Robert Nisticò
- Laboratory of Neuropharmacology, EBRI Rita Levi-Montalcini Foundation, Rome, Italy; School of Pharmacy, Department of Biology, University of Rome "Tor Vergata," Rome, Italy
| | - Massimo Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | | | - Johannes Streffer
- Reference Center for Biological Markers of Dementia, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; UCB Biopharma SPRL, Braine-l'Alleud, Belgium
| | - Iryna Voytyuk
- Department of Neurosciences, KU Leuven, Leuven, Belgium; Centre for Brain and Disease Research, VIB (Flanders Institute for Biotechnology), Leuven, Belgium; ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge, United Kingdom
| | - Maarten Timmers
- Reference Center for Biological Markers of Dementia, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Janssen Research and Development, a division of Janssen Pharmaceutica, Beerse, Belgium
| | - Amir Abbas Tahami Monfared
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Michael Irizarry
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey
| | - Bruce Albala
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey
| | - Akihiko Koyama
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey
| | | | | | - Lisa Yarenis
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey
| | - Simone Lista
- Sorbonne University, GRC No. 21, Alzheimer Precision Medicine, Pitié-Salpêtrière Hospital, Paris, France; Institute of Memory and Alzheimer's Disease, Department of Neurology, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute, INSERM U 1127, CNRS UMR 7225, Paris, France
| | - Lynn Kramer
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey
| | - Andrea Vergallo
- Neurology Business Group, Eisai Inc., Woodcliff Lake, New Jersey; Sorbonne University, GRC No. 21, Alzheimer Precision Medicine, Pitié-Salpêtrière Hospital, Paris, France; Institute of Memory and Alzheimer's Disease, Department of Neurology, Pitié-Salpêtrière Hospital, Paris, France; Brain & Spine Institute, INSERM U 1127, CNRS UMR 7225, Paris, France.
| |
Collapse
|
53
|
Jin X, Yang L, Yan X, Wang Q. Screening Platform Based on Inductively Coupled Plasma Mass Spectrometry for β-Site Amyloid Protein Cleaving Enzyme 1 (BACE1) Inhibitors. ACS Chem Neurosci 2021; 12:1093-1099. [PMID: 33764738 DOI: 10.1021/acschemneuro.0c00816] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
β-Site amyloid protein cleaving enzyme 1 (BACE1) is a promising therapeutic target for developing inhibitors to alleviate Alzheimer's disease (AD). Herein, we established an inductively coupled plasma mass spectrometry (ICPMS)-based inhibitor screening platform. A biotin-labeled lanthanide-coded peptide probe (LCPP; biotin-PEG2-EVNLDAEC-DOTA-Ln) was designed to determine the activity of BACE1 and evaluate the degree of inhibition of inhibitors. The platform was first validated with two commercially available inhibitors (BSI I and BSI IV) in terms of IC50 values and then applied to two newly designed inhibitors (inhibitors II and III) based on the crystal structure of BACE1 interacting with inhibitor I, and each of them contained an acylguanidine core structure. We found that their inhibition effects were improved as evaluated by the sensitive and accurate LCPP-ICPMS platform, demonstrating its ability for new drug screening.
Collapse
Affiliation(s)
- Xin Jin
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Limin Yang
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xiaowen Yan
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Qiuquan Wang
- Department of Chemistry and the MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
54
|
Liu QF, Kanmani S, Lee J, Kim GW, Jeon S, Koo BS. Neoline Improves Memory Impairment and Reduces Amyloid-β Level and Tau Phosphorylation Through AMPK Activation in the Mouse Alzheimer's Disease Model. J Alzheimers Dis 2021; 81:507-516. [PMID: 33814448 DOI: 10.3233/jad-201614] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most general, chronic, and progressive neurodegenerative senile disorder characterized clinically by progressive cognitive deterioration and memory impairment. Neoline is effective against neuropathic pain models, but the effects of neoline against AD-like phenotypes have not been investigated. OBJECTIVE We offer the investigation of the effects of neoline in AD. METHODS In this study, a Tg-APPswe/PS1dE9 AD mouse model was treated orally with neoline at a concentration of 0.5 mg/kg or 0.1 mg/kg starting at 7.5 months and administered for three months, and its anti-AD effects were evaluated. RESULTS Neoline improved memory and cognition impairments and reduced the number of amyloid-beta plaque and the amount of amyloid-β in the brain of AD mice. Furthermore, neoline reduced the anxiety behavior in the AD mouse model. The chronic administration of neoline also induced AMPK phosphorylation and decreased tau, amyloid-β, and BACE1 expression in the hippocampus. These findings indicate that chronic administration of neoline has therapeutic effects via AMPK activation, and BACE1 downregulation resulted in a decrease in the amyloid-β levels in the brain of Tg-APPswe/PS1dE9 AD mice. CONCLUSION Our results suggest that neoline is a therapeutic agent for the cure of neurodegenerative diseases like AD.
Collapse
Affiliation(s)
- Quan Feng Liu
- Department of Oriental Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, Goyang, Republic of Korea
| | - Suganya Kanmani
- Department of Oriental Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, Goyang, Republic of Korea
| | - Jinhyuk Lee
- Woobo Research Institute of Oriental Medicine Co. Ltd, Seoul, Republic of Korea
| | - Geun-Woo Kim
- Department of Neuropsychiatry, Dongguk University Bundang Oriental Hospital, Seongnam, Republic of Korea
| | - Songhee Jeon
- Department of Biomedical Sciences, Center for Glocal Future Biomedical Scientists at Chonnam National University, Gwangju, Republic of Korea
| | - Byung-Soo Koo
- Department of Oriental Neuropsychiatry, Graduate School of Oriental Medicine, Dongguk University, Goyang, Republic of Korea.,Department of Korean Neuropsychiatry, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| |
Collapse
|
55
|
Carter B, Justin HS, Gulick D, Gamsby JJ. The Molecular Clock and Neurodegenerative Disease: A Stressful Time. Front Mol Biosci 2021; 8:644747. [PMID: 33889597 PMCID: PMC8056266 DOI: 10.3389/fmolb.2021.644747] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Circadian rhythm dysfunction occurs in both common and rare neurodegenerative diseases. This dysfunction manifests as sleep cycle mistiming, alterations in body temperature rhythms, and an increase in symptomatology during the early evening hours known as Sundown Syndrome. Disruption of circadian rhythm homeostasis has also been implicated in the etiology of neurodegenerative disease. Indeed, individuals exposed to a shifting schedule of sleep and activity, such as health care workers, are at a higher risk. Thus, a bidirectional relationship exists between the circadian system and neurodegeneration. At the heart of this crosstalk is the molecular circadian clock, which functions to regulate circadian rhythm homeostasis. Over the past decade, this connection has become a focal point of investigation as the molecular clock offers an attractive target to combat both neurodegenerative disease pathogenesis and circadian rhythm dysfunction, and a pivotal role for neuroinflammation and stress has been established. This review summarizes the contributions of molecular clock dysfunction to neurodegenerative disease etiology, as well as the mechanisms by which neurodegenerative diseases affect the molecular clock.
Collapse
Affiliation(s)
- Bethany Carter
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States
| | - Hannah S Justin
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States
| | - Danielle Gulick
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Joshua J Gamsby
- Gamsby Laboratory, USF Health Byrd Alzheimer's Center and Research Institute, University of South Florida Health, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
56
|
Fissel JA, Farah MH. The influence of BACE1 on macrophage recruitment and activity in the injured peripheral nerve. J Neuroinflammation 2021; 18:71. [PMID: 33722254 PMCID: PMC7962400 DOI: 10.1186/s12974-021-02121-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 02/26/2021] [Indexed: 01/13/2023] Open
Abstract
Following peripheral nerve injury, multiple cell types, including axons, Schwann cells, and macrophages, coordinate to promote nerve regeneration. However, this capacity for repair is limited, particularly in older populations, and current treatments are insufficient. A critical component of the regeneration response is the network of cell-to-cell signaling in the injured nerve microenvironment. Sheddases are expressed in the peripheral nerve and play a role in the regulation if this cell-to-cell signaling through cleavage of transmembrane proteins, enabling the regulation of multiple pathways through cis- and trans-cellular regulatory mechanisms. Enhanced axonal regeneration has been observed in mice with deletion of the sheddase beta-secretase (BACE1), a transmembrane aspartyl protease that has been studied in the context of Alzheimer’s disease. BACE1 knockout (KO) mice display enhanced macrophage recruitment and activity following nerve injury, although it is unclear whether this plays a role in driving the enhanced axonal regeneration. Further, it is unknown by what mechanism(s) BACE1 increases macrophage recruitment and activity. BACE1 has many substrates, several of which are known to have immunomodulatory activity. This review will discuss current knowledge of the role of BACE1 and other sheddases in peripheral nerve regeneration and outline known immunomodulatory BACE1 substrates and what potential roles they could play in peripheral nerve regeneration. Currently, the literature suggests that BACE1 and substrates that are expressed by neurons and Schwann cells are likely to be more important for this process than those expressed by macrophages. More broadly, BACE1 may play a role as an effector of immunomodulation beyond the peripheral nerve.
Collapse
Affiliation(s)
- John A Fissel
- Department of Neurology, Johns Hopkins University School of Medicine, The John G. Rangos Sr. Building, Room 239, 855 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Mohamed H Farah
- Department of Neurology, Johns Hopkins University School of Medicine, The John G. Rangos Sr. Building, Room 239, 855 N. Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
57
|
Chen X, Zhang Y, Wang Q, Qin Y, Yang X, Xing Z, Shen Y, Wu H, Qi Y. The function of SUMOylation and its crucial roles in the development of neurological diseases. FASEB J 2021; 35:e21510. [PMID: 33710677 DOI: 10.1096/fj.202002702r] [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: 12/12/2020] [Revised: 02/02/2021] [Accepted: 02/22/2021] [Indexed: 11/11/2022]
Abstract
Neurological diseases are relatively complex diseases of a large system; however, the detailed mechanism of their pathogenesis has not been completely elucidated, and effective treatment methods are still lacking for some of the diseases. The SUMO (small ubiquitin-like modifier) modification is a dynamic and reversible process that is catalyzed by SUMO-specific E1, E2, and E3 ligases and reversed by a family of SENPs (SUMO/Sentrin-specific proteases). SUMOylation covalently conjugates numerous cellular proteins, and affects their cellular localization and biological activity in numerous cellular processes. A wide range of neuronal proteins have been identified as SUMO substrates, and the disruption of SUMOylation results in defects in synaptic plasticity, neuronal excitability, and neuronal stress responses. SUMOylation disorders cause many neurodegenerative diseases, such as Parkinson's disease, Alzheimer's disease, and Huntington's disease. By modulating the ion channel subunit, SUMOylation imbalance is responsible for the development of various channelopathies. The regulation of protein SUMOylation in neurons may provide a new strategy for the development of targeted therapeutic drugs for neurodegenerative diseases and channelopathies.
Collapse
Affiliation(s)
- Xu Chen
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yuhong Zhang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Qiqi Wang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yuanyuan Qin
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xinyi Yang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Zhengcao Xing
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yajie Shen
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Hongmei Wu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yitao Qi
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
58
|
Thapa P, Upadhyay SP, Suo WZ, Singh V, Gurung P, Lee ES, Sharma R, Sharma M. Chalcone and its analogs: Therapeutic and diagnostic applications in Alzheimer's disease. Bioorg Chem 2021; 108:104681. [PMID: 33571811 PMCID: PMC7928223 DOI: 10.1016/j.bioorg.2021.104681] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/15/2020] [Accepted: 01/20/2021] [Indexed: 02/08/2023]
Abstract
Chalcone [(E)-1,3-diphenyl-2-propene-1-one], a small molecule with α, β unsaturated carbonyl group is a precursor or component of many natural flavonoids and isoflavonoids. It is one of the privileged structures in medicinal chemistry. It possesses a wide range of biological activities encouraging many medicinal chemists to study this scaffold for its usefulness to oncology, infectious diseases, virology and neurodegenerative diseases including Alzheimer's disease (AD). Small molecular size, convenient and cost-effective synthesis, and flexibility for modifications to modulate lipophilicity suitable for blood brain barrier (BBB) permeability make chalcones a preferred candidate for their therapeutic and diagnostic potential in AD. This review summarizes and highlights the importance of chalcone and its analogs as single target small therapeutic agents, multi-target directed ligands (MTDLs) as well as molecular imaging agents for AD. The information summarized here will guide many medicinal chemist and researchers involved in drug discovery to consider chalcone as a potential scaffold for the development of anti-AD agents including theranostics.
Collapse
Affiliation(s)
- Pritam Thapa
- Drug Discovery Program, Midwest Veterans' Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO 64128, USA.
| | - Sunil P Upadhyay
- Drug Discovery Program, Midwest Veterans' Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO 64128, USA
| | - William Z Suo
- Laboratory for Alzheimer's Disease & Aging Research, Veterans Affairs Medical Center, Kansas City, MO 64128, USA
| | - Vikas Singh
- Division of Neurology, KCVA Medical Center, Kansas City, MO, USA
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, Iowa City, IA 52242, USA
| | - Eung Seok Lee
- College of Pharmacy, Yeungnam University, Gyeongsan 712-749, Republic of Korea
| | - Ram Sharma
- Drug Discovery Program, Midwest Veterans' Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO 64128, USA
| | - Mukut Sharma
- Drug Discovery Program, Midwest Veterans' Biomedical Research Foundation, KCVA Medical Center, Kansas City, MO 64128, USA
| |
Collapse
|
59
|
Yen YC, Kammeyer AM, Tirlangi J, Ghosh AK, Mesecar AD. A Structure-Based Discovery Platform for BACE2 and the Development of Selective BACE Inhibitors. ACS Chem Neurosci 2021; 12:581-588. [PMID: 33544569 DOI: 10.1021/acschemneuro.0c00629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The ability to perform routine structure-guided drug design for selective BACE inhibitors has been limited because of the lack of robust platform for BACE2 expression, purification, and crystallization. To overcome this limitation, we developed a platform that produces 2-3 mg of pure BACE2 protein per liter of E. coli culture, and we used this protein to design macrocyclic compounds that potently and selectively inhibit BACE1 over BACE2. Compound 2 was found to potently inhibit BACE 1 (Ki = 5 nM) with a selectivity of 214-fold over BACE2. The X-ray crystal structures of unbound BACE2 (2.2 Å) and BACE2 bound to compound 3 (3.0 Å and Ki = 7 nM) were determined and compared to the X-ray structures of BACE1 revealing the S1-S3 subsite as a selectivity determinant. This platform should enable a more rapid development of new and selective BACE inhibitors for the treatment of Alzheimer's disease or type II diabetes.
Collapse
|
60
|
Spatiotemporal processing of neural cell adhesion molecules 1 and 2 by BACE1 in vivo. J Biol Chem 2021; 296:100372. [PMID: 33548223 PMCID: PMC7949136 DOI: 10.1016/j.jbc.2021.100372] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
Neural cell adhesion molecules 1 (NCAM1) and 2 (NCAM2) belong to the cell adhesion molecules of the immunoglobulin superfamily and have been shown to regulate formation, maturation, and maintenance of synapses. NCAM1 and NCAM2 undergo proteolysis, but the identity of all the proteases involved and how proteolysis is used to regulate their functions are not known. We report here that NCAM1 and NCAM2 are BACE1 substrates in vivo. NCAM1 and NCAM2 overexpressed in HEK cells were both cleaved by metalloproteinases or BACE1, and NCAM2 was also processed by γ-secretase. We identified the BACE1 cleavage site of NCAM1 (at Glu 671) and NCAM2 (at Glu 663) using mass spectrometry and site-directed mutagenesis. Next, we assessed BACE1-mediated processing of NCAM1 and NCAM2 in the mouse brain during aging. NCAM1 and NCAM2 were cleaved in the olfactory bulb of BACE1+/+ but not BACE1−/− mice at postnatal day 10 (P10), 4 and 12 months of age. In the hippocampus, a BACE1-specific soluble fragment of NCAM1 (sNCAM1β) was only detected at P10. However, we observed an accumulation of full-length NCAM1 in hippocampal synaptosomes in 4-month-old BACE1−/− mice. We also found that polysialylated NCAM1 (PSA-NCAM1) levels were increased in BACE1−/− mice at P10 and demonstrated that BACE1 cleaves both NCAM1 and PSA-NCAM1 in vitro. In contrast, we did not find evidence for BACE1-dependent NCAM2 processing in the hippocampus at any age analyzed. In summary, our data demonstrate that BACE1 differentially processes NCAM1 and NCAM2 depending on the region of brain, subcellular localization, and age in vivo.
Collapse
|
61
|
Vijayan D, Chandra R. Amyloid Beta Hypothesis in Alzheimer's Disease: Major Culprits and Recent Therapeutic Strategies. Curr Drug Targets 2021; 21:148-166. [PMID: 31385768 DOI: 10.2174/1389450120666190806153206] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 07/13/2019] [Accepted: 07/26/2019] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is one of the most common forms of dementia and has been a global concern for several years. Due to the multi-factorial nature of the disease, AD has become irreversible, fatal and imposes a tremendous socio-economic burden. Even though experimental medicines suggested moderate benefits, AD still lacks an effective treatment strategy for the management of symptoms or cure. Among the various hypotheses that describe development and progression of AD, the amyloid hypothesis has been a long-term adherent to the AD due to the involvement of various forms of Amyloid beta (Aβ) peptides in the impairment of neuronal and cognitive functions. Hence, majority of the drug discovery approaches in the past have focused on the prevention of the accumulation of Aβ peptides. Currently, there are several agents in the phase III clinical trials that target Aβ or the various macromolecules triggering Aβ deposition. In this review, we present the state of the art knowledge on the functional aspects of the key players involved in the amyloid hypothesis. Furthermore, we also discuss anti-amyloid agents present in the Phase III clinical trials.
Collapse
Affiliation(s)
- Dileep Vijayan
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| | - Remya Chandra
- Department of Biotechnology and Microbiology, Thalassery Campus, Kannur University, Kerala Pin 670 661, India
| |
Collapse
|
62
|
Zhu H, Dronamraju V, Xie W, More SS. Sulfur-containing therapeutics in the treatment of Alzheimer's disease. Med Chem Res 2021; 30:305-352. [PMID: 33613018 PMCID: PMC7889054 DOI: 10.1007/s00044-020-02687-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/06/2020] [Indexed: 12/12/2022]
Abstract
Sulfur is widely existent in natural products and synthetic organic compounds as organosulfur, which are often associated with a multitude of biological activities. OBenzothiazole, in which benzene ring is fused to the 4,5-positions of the thiazolerganosulfur compounds continue to garner increasing amounts of attention in the field of medicinal chemistry, especially in the development of therapeutic agents for Alzheimer's disease (AD). AD is a fatal neurodegenerative disease and the primary cause of age-related dementia posing severe societal and economic burdens. Unfortunately, there is no cure for AD. A lot of research has been conducted on sulfur-containing compounds in the context of AD due to their innate antioxidant potential and some are currently being evaluated in clinical trials. In this review, we have described emerging trends in the field, particularly the concept of multi-targeting and formulation of disease-modifying strategies. SAR, pharmacological targets, in vitro/vivo ADMET, efficacy in AD animal models, and applications in clinical trials of such sulfur compounds have also been discussed. This article provides a comprehensive review of organosulfur-based AD therapeutic agents and provides insights into their future development.
Collapse
Affiliation(s)
- Haizhou Zhu
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Venkateshwara Dronamraju
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Wei Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Swati S. More
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
63
|
Latimer CS, Lucot KL, Keene CD, Cholerton B, Montine TJ. Genetic Insights into Alzheimer's Disease. ANNUAL REVIEW OF PATHOLOGY 2021; 16:351-376. [PMID: 33497263 PMCID: PMC8664069 DOI: 10.1146/annurev-pathmechdis-012419-032551] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is a pervasive, relentlessly progressive neurodegenerative disorder that includes both hereditary and sporadic forms linked by common underlying neuropathologic changes and neuropsychological manifestations. While a clinical diagnosis is often made on the basis of initial memory dysfunction that progresses to involve multiple cognitive domains, definitive diagnosis requires autopsy examination of the brain to identify amyloid plaques and neurofibrillary degeneration. Over the past 100 years, there has been remarkable progress in our understanding of the underlying pathophysiologic processes, pathologic changes, and clinical phenotypes of AD, largely because genetic pathways that include but expand beyond amyloid processing have been uncovered. This review discusses the current state of understanding of the genetics of AD with a focus on how these advances are both shaping our understanding of the disease and informing novel avenues and approaches for development of potential therapeutic targets.
Collapse
Affiliation(s)
- Caitlin S Latimer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington 98104, USA
| | - Katherine L Lucot
- Department of Pathology, Stanford University, Stanford, California 94304, USA;
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington 98104, USA
| | - Brenna Cholerton
- Department of Pathology, Stanford University, Stanford, California 94304, USA;
| | - Thomas J Montine
- Department of Pathology, Stanford University, Stanford, California 94304, USA;
| |
Collapse
|
64
|
Tian X, Murfin LC, Wu L, Lewis SE, James TD. Fluorescent small organic probes for biosensing. Chem Sci 2021; 12:3406-3426. [PMID: 34163615 PMCID: PMC8179477 DOI: 10.1039/d0sc06928k] [Citation(s) in RCA: 216] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 01/16/2021] [Indexed: 12/26/2022] Open
Abstract
Small-molecule based fluorescent probes are increasingly important for the detection and imaging of biological signaling molecules due to their simplicity, high selectivity and sensitivity, whilst being non-invasive, and suitable for real-time analysis of living systems. With this perspective we highlight sensing mechanisms including Förster resonance energy transfer (FRET), intramolecular charge transfer (ICT), photoinduced electron transfer (PeT), excited state intramolecular proton transfer (ESIPT), aggregation induced emission (AIE) and multiple modality fluorescence approaches including dual/triple sensing mechanisms (DSM or TSM). Throughout the perspective we highlight the remaining challenges and suggest potential directions for development towards improved small-molecule fluorescent probes suitable for biosensing.
Collapse
Affiliation(s)
- Xue Tian
- Department of Chemistry, University of Bath Bath BA2 7AY UK
| | - Lloyd C Murfin
- Department of Chemistry, University of Bath Bath BA2 7AY UK
| | - Luling Wu
- Department of Chemistry, University of Bath Bath BA2 7AY UK
| | - Simon E Lewis
- Department of Chemistry, University of Bath Bath BA2 7AY UK
| | - Tony D James
- Department of Chemistry, University of Bath Bath BA2 7AY UK
- School of Chemistry and Chemical Engineering, Henan Normal University Xinxiang 453007 P. R. China
| |
Collapse
|
65
|
Panda SS, Jhanji N. Natural Products as Potential Anti-Alzheimer Agents. Curr Med Chem 2021; 27:5887-5917. [PMID: 31215372 DOI: 10.2174/0929867326666190618113613] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/20/2019] [Accepted: 05/28/2019] [Indexed: 01/18/2023]
Abstract
Medicinal plants have curative properties due to the presence of various complex chemical substances of different composition, which are found as secondary metabolites in one or more parts of the plant. The diverse secondary metabolites play an important role in the prevention and cure of various diseases including neurodegenerative diseases like Alzheimer's disease. Naturally occurring compounds such as flavonoids, polyphenols, alkaloids, and glycosides found in various parts of the plant and/or marine sources may potentially protect neurodegeneration as well as improve memory and cognitive function. Many natural compounds show anti-Alzheimer activity through specific pharmacological mechanisms like targeting β-amyloid, Beta-secretase 1 and Acetylcholinesterase. In this review, we have compiled more than 130 natural products with a broad diversity in the class of compounds, which were isolated from different sources showing anti- Alzheimer properties.
Collapse
Affiliation(s)
- Siva S Panda
- Department of Chemistry & Physics, Augusta University, Augusta, Georgia 30912, United States
| | - Nancy Jhanji
- Department of Chemistry & Physics, Augusta University, Augusta, Georgia 30912, United States
| |
Collapse
|
66
|
Pradeepkiran JA, Reddy AP, Yin X, Manczak M, Reddy PH. Protective effects of BACE1 inhibitory ligand molecules against amyloid beta-induced synaptic and mitochondrial toxicities in Alzheimer's disease. Hum Mol Genet 2020; 29:49-69. [PMID: 31595293 DOI: 10.1093/hmg/ddz227] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 08/25/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022] Open
Abstract
Amyloid-β (Aβ) peptides are the major drivers of Alzheimer's disease (AD) pathogenesis, and are formed by successive cleavage of the amyloid precursor protein (APP) by the beta and gamma secretases. Mounting evidence suggests that Aβ and mitochondrial structural and functional abnormalities are critically involved in the loss of synapses and cognitive decline, in patients with AD. In AD brain, state the sequential proteolytic cleavage of APP by beta secretase 1 enzyme (BACE1) and γ-secretase leads to the production and release of Aβ40 and 42. BACE1 expression and activity increased in the brains of AD patients. Structurally, β-secretase has a very large binding site (1000 Å) with fewer hydrophobic domains that makes a challenge to identify the specific targets/binding sites of BACE1. In the present study, we constructed a BACE1 pharmacophore with pepstatin and screened through molecular docking studies. We found one potential candidate (referred as ligand 1) that binds to the key catalytic residues of BACE1 and predicts to inhibit abnormal APP processing and reduce Aβ levels in AD neurons. Using biochemical, molecular, transmission electron microscopy, immunoblotting and immunofluorescence analyses, we studied the protective effects of ligand 1 against Aβ-induced synaptic and mitochondrial toxicities in mouse neuroblastoma (N2a) cells that express mutant APP. We found interaction between ligand 1 and BACE1 and this interaction decreased BACE1 activity, Aβ40 and 42 levels. We also found increased mitochondrial biogenesis, mitochondrial fusion and synaptic activity and reduced mitochondrial fission in ligand 1-treated mutant APP cells. Based on these results, we cautiously conclude that ligand 1 reduces Aβ-induced mitochondrial and synaptic toxicities, and maintains mitochondrial dynamics and neuronal function in AD.
Collapse
Affiliation(s)
- Jangampalli Adi Pradeepkiran
- Internal Medicine Department, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
| | - Arubala P Reddy
- Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
| | - Xiangling Yin
- Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Suite E, Lubbock, TX 79413, USA
| | - Maria Manczak
- Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Suite E, Lubbock, TX 79413, USA
| | - P Hemachandra Reddy
- Internal Medicine Department, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA.,Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA.,Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Suite E, Lubbock, TX 79413, USA.,Cell Biology & Biochemistry Department, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA.,Neurology Department, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA.,Speech, Language and Hearing Sciences Departments, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA.,Public Health Department, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX 79430, USA
| |
Collapse
|
67
|
Lopez-Barbosa N, Garcia JG, Cifuentes J, Castro LM, Vargas F, Ostos C, Cardona-Gomez GP, Hernandez AM, Cruz JC. Multifunctional magnetite nanoparticles to enable delivery of siRNA for the potential treatment of Alzheimer's. Drug Deliv 2020; 27:864-875. [PMID: 32515999 PMCID: PMC8216449 DOI: 10.1080/10717544.2020.1775724] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 01/12/2023] Open
Abstract
Therapeutic drugs for Alzheimer's disease have been extensively studied due to its recurrence and abundance among neurodegenerative diseases. It is thought that the accumulation of amyloid precursor protein (APP) products, a consequence of an up-regulation of the β-site APP-cleaving enzyme 1 (BACE1), is the main triggering mechanism during the early stages of the disease. This study aims to explore the ability of a multifunctional conjugate based on magnetite nanoparticles for the cellular delivery of siRNA against the expression of the BACE1 gene. We immobilized the siRNA strand on PEGylated magnetite nanoparticles and investigated the effects on biocompatibility and efficacy of the conjugation. Similarly, we co-immobilized the translocating protein OmpA on PEGylated nanoparticles to enhance cellular uptake and endosomal escape. BACE1 suppression was statistically significant in HFF-1 cells, without any presence of a cytotoxic effect. The delivery of the nanoconjugate was achieved through endocytosis pathways, where endosome formation was likely escaped due to the proton-sponge effect characteristic of PEGylated nanoparticles or mainly by direct translocation in the case of OmpA/PEGylated nanoparticles.
Collapse
Affiliation(s)
| | - Juan G. Garcia
- Institute of Chemistry, CATALAD Research Group, Universidad de Antioquia UdeA, Medellin, Colombia
| | - Javier Cifuentes
- Department of Biomedical Engineering, Universidad de los Andes, Bogota, Colombia
| | - Lina M. Castro
- Institute of Chemistry, CATALAD Research Group, Universidad de Antioquia UdeA, Medellin, Colombia
| | - Felipe Vargas
- School of Medicine, Universidad de Antioquia UdeA, Medellin, Colombia
| | - Carlos Ostos
- Institute of Chemistry, CATALAD Research Group, Universidad de Antioquia UdeA, Medellin, Colombia
| | | | - Alher Mauricio Hernandez
- Engineering Faculty, Bioinstrumentation and Clinical Engineering Research Group – GIBIC, Bioengineering Department, Universidad de Antioquia UdeA, Medellin, Colombia
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogota, Colombia
- Engineering Faculty, Bioinstrumentation and Clinical Engineering Research Group – GIBIC, Bioengineering Department, Universidad de Antioquia UdeA, Medellin, Colombia
| |
Collapse
|
68
|
Multiple proteases are involved in mesothelin shedding by cancer cells. Commun Biol 2020; 3:728. [PMID: 33262421 PMCID: PMC7708464 DOI: 10.1038/s42003-020-01464-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 10/19/2020] [Indexed: 11/09/2022] Open
Abstract
Mesothelin (MSLN) is a lineage restricted cell surface protein expressed in about 30% of human cancers and high MSLN expression is associated with poor survival in several different cancers. The restricted expression of MSLN in normal tissue and its frequent expression in cancers make MSLN an excellent target for antibody-based therapies. Many clinical trials with agents targeting MSLN have been carried out but to date none of these agents have produced enough responses to obtain FDA approval. MSLN shedding is an important factor that may contribute to the failure of these therapies, because shed MSLN acts as a decoy receptor and allows release of antibodies bound to cell-surface MSLN. We have investigated the mechanism of shedding and show here that members of the ADAM, MMP and BACE families of proteases all participate in shedding, that more than one protease can produce shedding in the same cell, and that inhibition of shedding greatly enhances killing of cells by an immunotoxin targeting MSLN. Our data indicates that controlling MSLN shedding could greatly increase the activity of therapies that target MSLN.
Collapse
|
69
|
Wessels AM, Tariot PN, Zimmer JA, Selzler KJ, Bragg SM, Andersen SW, Landry J, Krull JH, Downing AM, Willis BA, Shcherbinin S, Mullen J, Barker P, Schumi J, Shering C, Matthews BR, Stern RA, Vellas B, Cohen S, MacSweeney E, Boada M, Sims JR. Efficacy and Safety of Lanabecestat for Treatment of Early and Mild Alzheimer Disease: The AMARANTH and DAYBREAK-ALZ Randomized Clinical Trials. JAMA Neurol 2020; 77:199-209. [PMID: 31764959 PMCID: PMC6902191 DOI: 10.1001/jamaneurol.2019.3988] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Importance Alzheimer disease (AD) is a neurodegenerative disorder characterized by cognitive deterioration and impaired activities of daily living. Current treatments provide only minor symptomatic improvements with limited benefit duration. Lanabecestat, a brain-permeable inhibitor of human beta-site amyloid precursor protein-cleaving enzyme 1 (BACE1/β-secretase), was developed to modify the clinical course of AD by slowing disease progression. Objective To assess whether lanabecestat slows the progression of AD compared with placebo in patients with early AD (mild cognitive impairment) and mild AD dementia. Design, Setting, and Participants AMARANTH (first patient visit on September 30, 2014; last patient visit on October 4, 2018) and DAYBREAK-ALZ (first patient visit on July 1, 2016; last patient visit on September 28, 2018) were randomized, placebo-controlled, phase 2/3 and phase 3 clinical trials lasting 104 weeks and 78 weeks, respectively. AMARANTH and DAYBREAK-ALZ were multicenter, global, double-blind studies conducted at 257 and 251 centers, respectively, located in 15 and 18 countries or territories, respectively. A population-based sample of men and women aged 55 to 85 years who met National Institute on Aging-Alzheimer's Association criteria for early AD or mild AD dementia was screened using cognitive assessments, and the presence of amyloid was confirmed. Patients were excluded for unstable medical conditions or medication use, significant cerebrovascular pathologic findings, or a history of vitiligo and/or current evidence of postinflammatory hypopigmentation. AMARANTH screened 6871 patients; 2218 (32.3%) were randomized, and 539 patients completed the study. DAYBREAK-ALZ screened 5706 patients; 1722 (30.2%) were randomized, and 76 patients completed the study. Interventions Patients were randomized (1:1:1) to once-daily oral doses of lanabecestat (20 mg), lanabecestat (50 mg), or placebo. Main Outcomes and Measures The primary outcome measure was change from baseline on the 13-item Alzheimer Disease Assessment Scale-cognitive subscale. Secondary outcomes included Alzheimer's Disease Cooperative Study-Instrumental Activities of Daily Living Inventory, Clinical Dementia Rating, Functional Activities Questionnaire, Mini-Mental State Examination, and Neuropsychiatric Inventory. Efficacy analyses were conducted on the intent-to-treat population. Results Among 2218 AMARANTH patients, the mean (SD) age was 71.3 (7.1) years, and 1177 of 2218 (53.1%) were women. Among 1722 DAYBREAK-ALZ patients, the mean (SD) age was 72.3 (7.0) years, and 1023 of 1722 (59.4%) were women. Both studies were terminated early after futility analysis. There were no consistent, reproducible dose-related findings on primary or secondary efficacy measures. Psychiatric adverse events, weight loss, and hair color changes were reported in a higher percentage of patients receiving lanabecestat than placebo. Conclusions and Relevance Treatment with lanabecestat was well tolerated and did not slow cognitive or functional decline. Trial Registration ClinicalTrials.gov identifiers: NCT02245737 and NCT02783573.
Collapse
Affiliation(s)
| | - Pierre N Tariot
- Banner Alzheimer's Institute, University of Arizona College of Medicine, Phoenix
| | | | | | | | | | - John Landry
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | | | | | - Jamie Mullen
- AstraZeneca, Waltham, Massachusetts, and Gaithersburg, Maryland.,Now retired
| | - Peter Barker
- AstraZeneca, Waltham, Massachusetts, and Gaithersburg, Maryland
| | - Jennifer Schumi
- AstraZeneca, Waltham, Massachusetts, and Gaithersburg, Maryland
| | - Craig Shering
- AstraZeneca, Waltham, Massachusetts, and Gaithersburg, Maryland
| | | | - Robert A Stern
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, Massachusetts
| | - Bruno Vellas
- Gérontopôle, Centre Hospitalier Universitaire de Toulouse, Unités Mixtes de Recherche Institut National de la Santé et de la Recherche Médicale 1027, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Sharon Cohen
- Toronto Memory Program, Toronto, Ontario, Canada
| | | | - Mercè Boada
- Research Center and Memory Clinic, Fundació Alzheimer Centre Educacional, Institut Català de Neurociències Aplicades, Barcelona-Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - John R Sims
- Eli Lilly and Company, Indianapolis, Indiana
| |
Collapse
|
70
|
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: 26] [Impact Index Per Article: 5.2] [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
|
71
|
Dungan CM, Valentino T, Vechetti IJ, Zdunek CJ, Murphy MP, Lin AL, McCarthy JJ, Peterson CA. Exercise-mediated alteration of hippocampal Dicer mRNA and miRNAs is associated with lower BACE1 gene expression and Aβ 1-42 in female 3xTg-AD mice. J Neurophysiol 2020; 124:1571-1577. [PMID: 33052800 DOI: 10.1152/jn.00503.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Changes to cerebral miRNA expression have been implicated in the progression of Alzheimer's disease (AD), as miRNAs that regulate the expression of gene products involved in amyloid beta (Aβ) processing, such as BACE1, are dysregulated in those that suffer from AD. Exercise training improves cognition and reduces BACE1 and Aβ-plaque burden; however, the mechanisms are not fully understood. Using our progressive weighted wheel running (PoWeR) exercise program, we assessed the effect of 20 wk of exercise training on changes in hippocampal miRNA expression in female 3xTg-AD (3xTg) mice. PoWeR was sufficient to promote muscle hypertrophy and increase myonuclear abundance. Furthermore, PoWeR elevated hippocampal Dicer gene expression in 3xTg mice, while altering miRNA expression toward a more wild-type profile. Specifically, miR-29, which is validated to target BACE1, was significantly lower in sedentary 3xTg mice when compared with wild-type but was elevated following PoWeR. Accordingly, BACE1 gene expression, along with detergent-soluble Aβ1-42, was lower in PoWeR-trained 3xTg mice. Our data suggest that PoWeR training upregulates Dicer gene expression to alter cerebral miRNA expression, which may contribute to reduced Aβ accumulation and delay AD progression.NEW & NOTEWORTHY Previous studies have outlined the beneficial effects of exercise on lowering BACE1 expression and reducing Aβ plaques. This study extends upon the work of others by outlining a new potential mechanism by which exercise elicits beneficial effects on Alzheimer's disease pathology, specifically through modulation of Dicer and miRNA expression. This is the first study to examine Dicer and miRNA expression in the hippocampus of the 3xTg model within the context of exercise.
Collapse
Affiliation(s)
- Cory M Dungan
- Department of Physical Therapy, University of Kentucky, Lexington, Kentucky.,The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
| | - Taylor Valentino
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Ivan J Vechetti
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, University of Kentucky, Lexington, Kentucky
| | | | - Michael P Murphy
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky.,Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky
| | - Ai-Ling Lin
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky.,Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - John J McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, University of Kentucky, Lexington, Kentucky
| | - Charlotte A Peterson
- Department of Physical Therapy, University of Kentucky, Lexington, Kentucky.,The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
72
|
Huang YY, Fang N, Luo HR, Gao F, Zou Y, Zhou LL, Zeng QP, Fang SS, Xiao F, Zheng Q. RP1, a RAGE antagonist peptide, can improve memory impairment and reduce Aβ plaque load in the APP/PS1 mouse model of Alzheimer's disease. Neuropharmacology 2020; 180:108304. [PMID: 32931813 DOI: 10.1016/j.neuropharm.2020.108304] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/01/2020] [Accepted: 09/08/2020] [Indexed: 12/25/2022]
Abstract
Amyloid-β (Aβ) accumulation is a pathological hallmark of Alzheimer's disease (AD). The receptor for advanced glycation end products (RAGE) is involved in the production and accumulation of Aβ. RP1, a peptide antagonist of RAGE, was screened by phage display technology in our previous studies, and its neuroprotective effects on an AD cell model have been confirmed. However, its efficacy in vivo remains unclear. Here, the intranasal delivery of RP1 to APPSwe/PS1dE9 (APP/PS1) mice significantly improved memory impairment and relieved the Aβ burden by decreasing the expression of amyloid precursor protein and β-secretase. RNA-sequencing (RNA-seq) was utilized to identify differentially expressed genes (DEGs) in APP/PS1 mice after RP1 administration. Several DEGs in RAGE downstream signalling pathways were downregulated. Some transcription factors (such as Fos) and the pathways enriched in the remarkable modules may also be related to the efficacy of RP1. In conclusion, RP1 significantly improves the AD symptoms of APP/PS1 mice, and the RNA-seq results provide new ideas for elucidating the possible mechanisms of RP1 treatment.
Collapse
Affiliation(s)
- Yi-Yun Huang
- Department of Microbial and Biochemical Pharmacy, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Nian Fang
- Department of Microbial and Biochemical Pharmacy, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Hui-Ru Luo
- Department of Microbial and Biochemical Pharmacy, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Feng Gao
- Department of Microbial and Biochemical Pharmacy, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Yao Zou
- Department of Microbial and Biochemical Pharmacy, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Li-Li Zhou
- Department of Microbial and Biochemical Pharmacy, College of Pharmacy, Jinan University, Guangzhou, China.
| | - Qing-Ping Zeng
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Shi-Song Fang
- Major Infectious Disease Control Key Laboratory, Key Reference Laboratory of Pathogen and Biosafety, Shenzhen Center for Disease Control and Prevention, Shenzhen, China.
| | - Fei Xiao
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China.
| | - Qing Zheng
- Department of Microbial and Biochemical Pharmacy, College of Pharmacy, Jinan University, Guangzhou, China.
| |
Collapse
|
73
|
Lackie RE, Marques-Lopes J, Ostapchenko VG, Good S, Choy WY, van Oosten-Hawle P, Pasternak SH, Prado VF, Prado MAM. Increased levels of Stress-inducible phosphoprotein-1 accelerates amyloid-β deposition in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2020; 8:143. [PMID: 32825842 PMCID: PMC7441634 DOI: 10.1186/s40478-020-01013-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 08/01/2020] [Indexed: 02/08/2023] Open
Abstract
Molecular chaperones and co-chaperones, which are part of the protein quality control machinery, have been shown to regulate distinct aspects of Alzheimer's Disease (AD) pathology in multiple ways. Notably, the co-chaperone STI1, which presents increased levels in AD, can protect mammalian neurons from amyloid-β toxicity in vitro and reduced STI1 levels worsen Aβ toxicity in C. elegans. However, whether increased STI1 levels can protect neurons in vivo remains unknown. We determined that overexpression of STI1 and/or Hsp90 protected C. elegans expressing Aβ(3-42) against Aβ-mediated paralysis. Mammalian neurons were also protected by elevated levels of endogenous STI1 in vitro, and this effect was mainly due to extracellular STI1. Surprisingly, in the 5xFAD mouse model of AD, by overexpressing STI1, we find increased amyloid burden, which amplifies neurotoxicity and worsens spatial memory deficits in these mutants. Increased levels of STI1 disturbed the expression of Aβ-regulating enzymes (BACE1 and MMP-2), suggesting potential mechanisms by which amyloid burden is increased in mice. Notably, we observed that STI1 accumulates in dense-core AD plaques in both 5xFAD mice and human brain tissue. Our findings suggest that elevated levels of STI1 contribute to Aβ accumulation, and that STI1 is deposited in AD plaques in mice and humans. We conclude that despite the protective effects of STI1 in C. elegans and in mammalian cultured neurons, in vivo, the predominant effect of elevated STI1 is deleterious in AD.
Collapse
Affiliation(s)
- Rachel E Lackie
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
- Program in Neuroscience, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada
| | - Jose Marques-Lopes
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
| | - Valeriy G Ostapchenko
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
| | - Sarah Good
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Wing-Yiu Choy
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, Medical Sciences Building, 1151 Richmond St. N, London, N6A 5B7, Canada
| | - Patricija van Oosten-Hawle
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Stephen H Pasternak
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada
- St. Joseph's Health Care London-Parkwood Institute, St. Joseph's Hospital, 268 Grosvenor St Room A1-015, London, N6A 4V2, Canada
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, 1151 Richmond St, London, N6A 3K7, Canada
| | - Vania F Prado
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada.
- Program in Neuroscience, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Anatomy & Cell Biology, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Ontario, Canada.
| | - Marco A M Prado
- Robarts Research Institute, The University of Western Ontario, 1151 Richmond St. N., London, Ontario, N6A 5B7, Canada.
- Program in Neuroscience, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Anatomy & Cell Biology, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Canada.
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond St, London, N6A 3K7, Ontario, Canada.
| |
Collapse
|
74
|
Uddin MS, Kabir MT, Rahman MS, Behl T, Jeandet P, Ashraf GM, Najda A, Bin-Jumah MN, El-Seedi HR, Abdel-Daim MM. Revisiting the Amyloid Cascade Hypothesis: From Anti-Aβ Therapeutics to Auspicious New Ways for Alzheimer's Disease. Int J Mol Sci 2020; 21:5858. [PMID: 32824102 PMCID: PMC7461598 DOI: 10.3390/ijms21165858] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder related to age, characterized by the cerebral deposition of fibrils, which are made from the amyloid-β (Aβ), a peptide of 40-42 amino acids. The conversion of Aβ into neurotoxic oligomeric, fibrillar, and protofibrillar assemblies is supposed to be the main pathological event in AD. After Aβ accumulation, the clinical symptoms fall out predominantly due to the deficient brain clearance of the peptide. For several years, researchers have attempted to decline the Aβ monomer, oligomer, and aggregate levels, as well as plaques, employing agents that facilitate the reduction of Aβ and antagonize Aβ aggregation, or raise Aβ clearance from brain. Unluckily, broad clinical trials with mild to moderate AD participants have shown that these approaches were unsuccessful. Several clinical trials are running involving patients whose disease is at an early stage, but the preliminary outcomes are not clinically impressive. Many studies have been conducted against oligomers of Aβ which are the utmost neurotoxic molecular species. Trials with monoclonal antibodies directed against Aβ oligomers have exhibited exciting findings. Nevertheless, Aβ oligomers maintain equivalent states in both monomeric and aggregation forms; so, previously administered drugs that precisely decrease Aβ monomer or Aβ plaques ought to have displayed valuable clinical benefits. In this article, Aβ-based therapeutic strategies are discussed and several promising new ways to fight against AD are appraised.
Collapse
Affiliation(s)
- Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka 1213, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka 1207, Bangladesh
| | - Md. Tanvir Kabir
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Md. Sohanur Rahman
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi 6205, Bangladesh;
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687 Reims CEDEX 2, France;
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Agnieszka Najda
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland;
| | - May N. Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia;
| | - Hesham R. El-Seedi
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China;
- Pharmacognosy Group, Department of Pharmaceutical Biosciences, Uppsala University, SE-751 23 Uppsala, Sweden
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt
| | - Mohamed M. Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
75
|
Paroni G, Bisceglia P, Seripa D. Understanding the Amyloid Hypothesis in Alzheimer's Disease. J Alzheimers Dis 2020; 68:493-510. [PMID: 30883346 DOI: 10.3233/jad-180802] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The amyloid hypothesis (AH) is still the most accepted model to explain the pathogenesis of inherited Alzheimer's disease (IAD). However, despite the neuropathological overlapping with the non-inherited form (NIAD), AH waver in explaining NIAD. Thus, 30 years after its first statement several questions are still open, mainly regarding the role of amyloid plaques (AP) and apolipoprotein E (APOE). Accordingly, a pathogenetic model including the role of AP and APOE unifying IAD and NIAD pathogenesis is still missing. In the present understanding of the AH, we suggested that amyloid-β (Aβ) peptides production and AP formation is a physiological aging process resulting from a systemic age-related decrease in the efficiency of the proteins catabolism/clearance machinery. In this pathogenetic model Aβ peptides act as neurotoxic molecules, but only above a critical concentration [Aβ]c. A threshold mechanism triggers IAD/NIAD onset only when [Aβ]≥[Aβ]c. In this process, APOE modifies [Aβ]c threshold in an isoform-specific way. Consequently, all factors influencing Aβ anabolism, such as amyloid beta precursor protein (APP), presenilin 1 (PSEN1), and presenilin 2 (PSEN2) gene mutations, and/or Aβ catabolism/clearance could contribute to exceed the threshold [Aβ]c, being characteristic of each individual. In this model, AP formation does not depend on [Aβ]c. The present interpretation of the AH, unifying the pathogenetic theories for IAD and NIAD, will explain why AP and APOE4 may be observed in healthy aging and why they are not the cause of AD. It is clear that further studies are needed to confirm our pathogenetic model. Nevertheless, our suggestion may be useful to better understand the pathogenesis of AD.
Collapse
Affiliation(s)
- Giulia Paroni
- Research Laboratory, Complex Structure of Geriatrics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Paola Bisceglia
- Research Laboratory, Complex Structure of Geriatrics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Davide Seripa
- Research Laboratory, Complex Structure of Geriatrics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| |
Collapse
|
76
|
Suire CN, Abdul-Hay SO, Sahara T, Kang D, Brizuela MK, Saftig P, Dickson DW, Rosenberry TL, Leissring MA. Cathepsin D regulates cerebral Aβ42/40 ratios via differential degradation of Aβ42 and Aβ40. ALZHEIMERS RESEARCH & THERAPY 2020; 12:80. [PMID: 32631408 PMCID: PMC7339583 DOI: 10.1186/s13195-020-00649-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/24/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Cathepsin D (CatD) is a lysosomal protease that degrades both the amyloid β-protein (Aβ) and the microtubule-associated protein, tau, and has been genetically linked to late-onset Alzheimer disease (AD). Here, we sought to examine the consequences of genetic deletion of CatD on Aβ proteostasis in vivo and to more completely characterize the degradation of Aβ42 and Aβ40 by CatD. METHODS We quantified Aβ degradation rates and levels of endogenous Aβ42 and Aβ40 in the brains of CatD-null (CatD-KO), heterozygous null (CatD-HET), and wild-type (WT) control mice. CatD-KO mice die by ~ 4 weeks of age, so tissues from younger mice, as well as embryonic neuronal cultures, were investigated. Enzymological assays and surface plasmon resonance were employed to quantify the kinetic parameters (KM, kcat) of CatD-mediated degradation of monomeric human Aβ42 vs. Aβ40, and the degradation of aggregated Aβ42 species was also characterized. Competitive inhibition assays were used to interrogate the relative inhibition of full-length human and mouse Aβ42 and Aβ40, as well as corresponding p3 fragments. RESULTS Genetic deletion of CatD resulted in 3- to 4-fold increases in insoluble, endogenous cerebral Aβ42 and Aβ40, exceeding the increases produced by deletion of an insulin-degrading enzyme, neprilysin or both, together with readily detectable intralysosomal deposits of endogenous Aβ42-all by 3 weeks of age. Quite significantly, CatD-KO mice exhibited ~ 30% increases in Aβ42/40 ratios, comparable to those induced by presenilin mutations. Mechanistically, the perturbed Aβ42/40 ratios were attributable to pronounced differences in the kinetics of degradation of Aβ42 vis-à-vis Aβ40. Specifically, Aβ42 shows a low-nanomolar affinity for CatD, along with an exceptionally slow turnover rate that, together, renders Aβ42 a highly potent competitive inhibitor of CatD. Notably, the marked differences in the processing of Aβ42 vs. Aβ40 also extend to p3 fragments ending at positions 42 vs. 40. CONCLUSIONS Our findings identify CatD as the principal intracellular Aβ-degrading protease identified to date, one that regulates Aβ42/40 ratios via differential degradation of Aβ42 vs. Aβ40. The finding that Aβ42 is a potent competitive inhibitor of CatD suggests a possible mechanistic link between elevations in Aβ42 and downstream pathological sequelae in AD.
Collapse
Affiliation(s)
- Caitlin N Suire
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA.,Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, 92697, USA
| | - Samer O Abdul-Hay
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Tomoko Sahara
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Dongcheul Kang
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Monica K Brizuela
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098, Kiel, Germany
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | | | - Malcolm A Leissring
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA. .,Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA.
| |
Collapse
|
77
|
Suh J, Romano DM, Nitschke L, Herrick SP, DiMarzio BA, Dzhala V, Bae JS, Oram MK, Zheng Y, Hooli B, Mullin K, Gennarino VA, Wasco W, Schmahmann JD, Albers MW, Zoghbi HY, Tanzi RE. Loss of Ataxin-1 Potentiates Alzheimer's Pathogenesis by Elevating Cerebral BACE1 Transcription. Cell 2020; 178:1159-1175.e17. [PMID: 31442405 DOI: 10.1016/j.cell.2019.07.043] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 12/21/2018] [Accepted: 07/24/2019] [Indexed: 01/28/2023]
Abstract
Expansion of CAG trinucleotide repeats in ATXN1 causes spinocerebellar ataxia type 1 (SCA1), a neurodegenerative disease that impairs coordination and cognition. While ATXN1 is associated with increased Alzheimer's disease (AD) risk, CAG repeat number in AD patients is not changed. Here, we investigated the consequences of ataxin-1 loss of function and discovered that knockout of Atxn1 reduced CIC-ETV4/5-mediated inhibition of Bace1 transcription, leading to increased BACE1 levels and enhanced amyloidogenic cleavage of APP, selectively in AD-vulnerable brain regions. Elevated BACE1 expression exacerbated Aβ deposition and gliosis in AD mouse models and impaired hippocampal neurogenesis and olfactory axonal targeting. In SCA1 mice, polyglutamine-expanded mutant ataxin-1 led to the increase of BACE1 post-transcriptionally, both in cerebrum and cerebellum, and caused axonal-targeting deficit and neurodegeneration in the hippocampal CA2 region. These findings suggest that loss of ataxin-1 elevates BACE1 expression and Aβ pathology, rendering it a potential contributor to AD risk and pathogenesis.
Collapse
Affiliation(s)
- Jaehong Suh
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute of Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA.
| | - Donna M Romano
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute of Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Larissa Nitschke
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Scott P Herrick
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Britt A DiMarzio
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute of Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Volodymyr Dzhala
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Jun-Seok Bae
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute of Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Mary K Oram
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute of Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Yuejiao Zheng
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute of Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Basavaraj Hooli
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute of Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Kristina Mullin
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute of Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Vincenzo A Gennarino
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Wilma Wasco
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute of Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Jeremy D Schmahmann
- Ataxia Unit, Cognitive Behavioral Neurology Unit, Laboratory for Neuroanatomy and Cerebellar Neurobiology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Mark W Albers
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Huda Y Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute of Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA.
| |
Collapse
|
78
|
Li S, Zhao H, Li J, Hao J, Yu H. A series of molecular modeling techniques to reveal selective mechanisms of inhibitors to β-Site amyloid precursor protein cleaving enzyme 1 (BACE1) and β-site amyloid precursor protein cleaving enzyme 2 (BACE2). J Biomol Struct Dyn 2020; 39:2824-2837. [PMID: 32276567 DOI: 10.1080/07391102.2020.1754917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inhibition of β-Site amyloid precursor protein cleaving enzyme 1 (BACE1) has been shown to be an effective treatment for Alzheimer's disease. A wealth of research has focused on finding highly selective small-molecule inhibitors targeting the BACE1 over its close homologue BACE2 to avoid potential side effects. However, given the highly structural similarities of BACE1 and BACE2, designing highly selective BACE1 inhibitors remains a huge challenge. Recently, it has been reported that a potential BACE1 inhibitor named C28 (∼52-fold selectivity) exhibited greater selectivity to BACE1 over BACE2 than the previously reported inhibitors AZD3293 and AZD3839 (∼1.5-fold and 14-fold selectivity). However, few computational studies have been performed to reveal its underlying mechanisms. In this study, a series of molecular modeling techniques were performed to reveal the selective mechanisms. Classical molecular dynamics (cMD) simulations indicated that the major variations appeared to be controlled by overall protein dynamics. Free energy calculations further suggested that the binding affinities of AZD3293 to BACE1 and BACE2 are similar, but the binding affinity of AZD3839 and C28 to BACE1 is much higher than to BACE2, and that the major variations are electrostatic interactions. The protein dynamics and energy differences were further observed in accelerated molecular dynamics (aMD) simulations. In addition, the umbrella sampling simulations revealed the inhibitors' different patterns of dissociation from the binding pockets of BACE1 and BACE2, and that different energy barriers were responsible for the selectivity. The physical principles revealed by this study may facilitate the rational design of more potent BACE1 selective inhibitors. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shipeng Li
- Department of Neurosurgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Hexiang Zhao
- Department of Neurosurgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Jinghui Li
- Department of Neurosurgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Jiajia Hao
- Kunming Medical University Haiyuan College, Kunming, Yunnan Province, China
| | - Hualin Yu
- Department of Neurosurgery, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
79
|
Scearce-Levie K, Sanchez PE, Lewcock JW. Leveraging preclinical models for the development of Alzheimer disease therapeutics. Nat Rev Drug Discov 2020; 19:447-462. [PMID: 32612262 DOI: 10.1038/s41573-020-0065-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2020] [Indexed: 02/06/2023]
Abstract
A large number of mouse models have been engineered, characterized and used to advance biomedical research in Alzheimer disease (AD). Early models simply damaged the rodent brain through toxins or lesions. Later, the spread of genetic engineering technology enabled investigators to develop models of familial AD by overexpressing human genes such as those encoding amyloid precursor protein (APP) or presenilins (PSEN1 or PSEN2) carrying mutations linked to early-onset AD. Recently, more complex models have sought to explore the impact of multiple genetic risk factors in the context of different biological challenges. Although none of these models has proven to be a fully faithful reproduction of the human disease, models remain essential as tools to improve our understanding of AD biology, conduct thorough pharmacokinetic and pharmacodynamic analyses, discover translatable biomarkers and evaluate specific therapeutic approaches. To realize the full potential of animal models as new technologies and knowledge become available, it is critical to define an optimal strategy for their use. Here, we review progress and challenges in the use of AD mouse models, highlight emerging scientific innovations in model development, and introduce a conceptual framework for use of preclinical models for therapeutic development.
Collapse
|
80
|
Galanis C, Vlachos A. Hebbian and Homeostatic Synaptic Plasticity-Do Alterations of One Reflect Enhancement of the Other? Front Cell Neurosci 2020; 14:50. [PMID: 32256317 PMCID: PMC7093376 DOI: 10.3389/fncel.2020.00050] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/21/2020] [Indexed: 01/07/2023] Open
Abstract
During the past 50 years, the cellular and molecular mechanisms of synaptic plasticity have been studied in great detail. A plethora of signaling pathways have been identified that account for synaptic changes based on positive and negative feedback mechanisms. Yet, the biological significance of Hebbian synaptic plasticity (= positive feedback) and homeostatic synaptic plasticity (= negative feedback) remains a matter of debate. Specifically, it is unclear how these opposing forms of plasticity, which share common downstream mechanisms, operate in the same networks, neurons, and synapses. Based on the observation that rapid and input-specific homeostatic mechanisms exist, we here discuss a model that is based on signaling pathways that may adjust a balance between Hebbian and homeostatic synaptic plasticity. Hence, “alterations” in Hebbian plasticity may, in fact, resemble “enhanced” homeostasis, which rapidly returns synaptic strength to baseline. In turn, long-lasting experience-dependent synaptic changes may require attenuation of homeostatic mechanisms or the adjustment of homeostatic setpoints at the single-synapse level. In this context, we propose a role for the proteolytic processing of the amyloid precursor protein (APP) in setting a balance between the ability of neurons to express Hebbian and homeostatic synaptic plasticity.
Collapse
Affiliation(s)
- Christos Galanis
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
81
|
Ge L, Liu Z, Tian Y. A novel two-photon ratiometric fluorescent probe for imaging and sensing of BACE1 in different regions of AD mouse brain. Chem Sci 2020; 11:2215-2224. [PMID: 32180930 PMCID: PMC7058092 DOI: 10.1039/c9sc05256a] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/09/2020] [Indexed: 01/16/2023] Open
Abstract
β-Secretase (BACE1) is the vital enzyme in the pathogenic processes of Alzheimer's disease (AD). However, the development of a powerful tool with high selectivity and sensitivity for BACE1 determination in vivo is a challenge in understanding the pathogenesis of AD. In this work, a novel two-photon ratiometric fluorescent probe (AF633mCyd) was first developed for imaging and sensing of BACE1 in live cells and deep tissues, in which the fluorescence resonance energy transfer (FRET) system was designed and synthesized by a novel two-photon donor, merocyanine derivative (mCyd), connected with an acceptor, Alexa Fluor 633 (AF633), through a peptide substrate (EVNL-DAEFRHDSGYK) with a length of less than 10 nm. The emission spectrum of mCyd possessed sufficient overlap with the absorption spectrum of AF633, resulting in the high sensitivity of the developed AF633mCyd probe. The peptide substrate which can be specifically cleaved by BACE1 was inserted between the donor and acceptor, leading to the high selectivity of the present fluorescent probe. The fluorescence emission peaks of the AF633mCyd probe were observed at 578 nm and 651 nm and the emission ratio demonstrated good linearity with the concentration of BACE1 varying from 0.1 to 40.0 nM with a detection limit down to 65.3 ± 0.1 pM. Considering the advantages of high selectivity and sensitivity, as well as long-term stability and good biocompatibility, the developed probe was successfully applied in imaging and sensing of BACE1 in different regions of AD mouse brain tissue with a depth greater than 300 μm. Using this powerful tool, it was clear that the level of BACE1 was different in various brain regions of AD mouse such as S1BF, CPu, LD, and CA1. The up-regulation of BACE1 was observed especially in the regions S1BF and CA1 in AD mouse brain. Moreover, BACE1 was also found to be closely related to AD pathogenesis caused by oxidative stress.
Collapse
Affiliation(s)
- Lihong Ge
- Department of Chemistry , School of Chemistry and Molecular Engineering , East China Normal University , Dongchuan Road 500 , Shanghai 200241 , China .
| | - Zhichao Liu
- Department of Chemistry , School of Chemistry and Molecular Engineering , East China Normal University , Dongchuan Road 500 , Shanghai 200241 , China .
| | - Yang Tian
- Department of Chemistry , School of Chemistry and Molecular Engineering , East China Normal University , Dongchuan Road 500 , Shanghai 200241 , China .
| |
Collapse
|
82
|
Kosel F, Pelley JMS, Franklin TB. Behavioural and psychological symptoms of dementia in mouse models of Alzheimer's disease-related pathology. Neurosci Biobehav Rev 2020; 112:634-647. [PMID: 32070692 DOI: 10.1016/j.neubiorev.2020.02.012] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 02/08/2020] [Accepted: 02/11/2020] [Indexed: 12/12/2022]
Abstract
Transgenic mouse models have been used extensively to model the cognitive impairments arising from Alzheimer's disease (AD)-related pathology. However, less is known about the relationship between AD-related pathology and the behavioural and psychological symptoms of dementia (BPSD) commonly presented by patients. This review discusses the BPSD-like behaviours recapitulated by several mouse models of AD-related pathology, including the APP/PS1, Tg2576, 3xTg-AD, 5xFAD, and APP23 models. Current evidence suggests that social withdrawal and depressive-like behaviours increase with progressive neuropathology, and increased aggression and sleep-wake disturbances are present even at early stages; however, there is no clear evidence to support increased anxiety-like behaviours, agitation (hyperactivity), or general apathy. Overall, transgenic mouse models of AD-related pathology recapitulate some of the BPSD-like behaviours associated with AD, but these behaviours vary by model. This reflects the patient population, where AD patients typically exhibit one or more BPSD, but rarely all symptoms at once. As a result, we suggest that transgenic mouse models are an important tool to investigate the pathology underlying BPSD in human AD patients.
Collapse
Affiliation(s)
- Filip Kosel
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Jessica M S Pelley
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Tamara B Franklin
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada.
| |
Collapse
|
83
|
Lauretti E, Dincer O, Praticò D. Glycogen synthase kinase-3 signaling in Alzheimer's disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118664. [PMID: 32006534 DOI: 10.1016/j.bbamcr.2020.118664] [Citation(s) in RCA: 331] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/21/2020] [Accepted: 01/24/2020] [Indexed: 01/19/2023]
Abstract
Alzheimer's disease (AD) is the most common form of neurodegenerative disorder with dementia, accounting for approximately 70% of the all cases. Currently, 5.8 million people in the U.S. are living with AD and by 2050 this number is expected to double resulting in a significant socio-economic burden. Despite intensive research, the exact mechanisms that trigger AD are still not known and at the present there is no cure for it. In recent years, many signaling pathways associated with AD neuropathology have been explored as possible candidate targets for the treatment of this condition including glycogen synthase kinase-3β (GSK3-β). GSK3-β is considered a key player in AD pathophysiology since dysregulation of this kinase influences all the major hallmarks of the disease including: tau phosphorylation, amyloid-β production, memory, neurogenesis and synaptic function. The present review summarizes the current understanding of the GSK3-β neurobiology with particular emphasis on its effects on specific signaling pathways associated with AD pathophysiology. Moreover, it discusses the feasibility of targeting GSK3-β for AD treatment and provides a summary of the current research effort to develop GSK3-β inhibitors in preclinical and clinical studies.
Collapse
Affiliation(s)
- Elisabetta Lauretti
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, United States of America
| | - Ozlem Dincer
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, United States of America
| | - Domenico Praticò
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, United States of America.
| |
Collapse
|
84
|
Lafon PA, Wang Y, Arango-Lievano M, Torrent J, Salvador-Prince L, Mansuy M, Mestre-Francès N, Givalois L, Liu J, Mercader JV, Jeanneteau F, Desrumaux C, Perrier V. Fungicide Residues Exposure and β-amyloid Aggregation in a Mouse Model of Alzheimer's Disease. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:17011. [PMID: 31939705 PMCID: PMC7015540 DOI: 10.1289/ehp5550] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Pesticide residues have contaminated our environment and nutrition over the last century. Although these compounds are present at very low concentrations, their long-term effects on human health is of concern. The link between pesticide residues and Alzheimer's disease is not clear and difficult to establish. To date, no in vivo experiments have yet modeled the impact of this chronic contamination on neurodegenerative disorders. OBJECTIVES We investigated the impact of fungicide residues on the pathological markers of Alzheimer's disease in a transgenic mouse model. METHODS Transgenic (J20, hAPP Sw / Ind ) mice were chronically exposed to a cocktail of residues of cyprodinil, mepanipyrim, and pyrimethanil at 0.1 μ g / L in their drinking water for 9 months. We assessed the effects of fungicide residues on the pathological markers of the disease including A β aggregates, neuroinflammation, and neuronal loss. Then, we studied the dynamics of A β aggregation in vivo via a longitudinal study using two-photon microscopy. Finally, we investigated the molecular mechanisms involved in the production and clearance of A β peptides. RESULTS We found that a chronic exposure to three fungicide residues exacerbated aggregation, microgliosis, and neuronal loss. These fungicides also increased vascular amyloid aggregates reminiscent of cerebral amyloid angiopathy between 6 and 9 months of treatment. The mechanism of action revealed that fungicides promoted A β peptide fibril formation in vitro and involved an in vivo overexpression of the levels of the β -secretase -cleaving enzyme (BACE1) combined with impairment of A β clearance through neprylisin (NEP). CONCLUSIONS Chronic exposure of the J20 mouse model of Alzheimer's disease to a cocktail of fungicides, at the regulatory concentration allowed in tap water (0.1 μ g / L ), strengthened the preexisting pathological markers: neuroinflammation, A β aggregation, and APP β -processing . We hypothesize prevention strategies toward pesticide long-term exposure may be an alternative to counterbalance the lack of treatment and to slow down the worldwide Alzheimer's epidemic. https://doi.org/10.1289/EHP5550.
Collapse
Affiliation(s)
| | - Yunyun Wang
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
- Cellular Signaling Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Margarita Arango-Lievano
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Joan Torrent
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
| | | | - Marine Mansuy
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
| | | | - Laurent Givalois
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
| | - Jianfeng Liu
- Cellular Signaling Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Josep Vicent Mercader
- Institute of Agrochemistry and Food Technology, Consejo Superior de Investigaciones Científicas (IATA-CSIC), Paterna, València, Spain
| | - Freddy Jeanneteau
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Catherine Desrumaux
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
- LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France
| | | |
Collapse
|
85
|
Menon PK, Koistinen NA, Iverfeldt K, Ström AL. Phosphorylation of the amyloid precursor protein (APP) at Ser-675 promotes APP processing involving meprin β. J Biol Chem 2019; 294:17768-17776. [PMID: 31604820 PMCID: PMC6879340 DOI: 10.1074/jbc.ra119.008310] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 10/02/2019] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by abnormal deposition of β-amyloid (Aβ) peptides. Aβ is a cleavage product of the amyloid precursor protein (APP), and aberrant posttranslational modifications of APP can alter APP processing and increase Aβ generation. In the AD brain, seven different residues, including Ser-675 (APP695 numbering) in the APP cytoplasmic domain has been found to be phosphorylated. Here, we show that expression of a phosphomimetic variant of Ser-675 in APP (APP-S675E), in human neuroblastoma SK-N-AS cells, reduces secretion of the soluble APP ectodomain (sAPPα), even though the total plasma membrane level of APP was unchanged compared with APP levels in cells expressing APPwt or APP-S675A. Moreover, the level of an alternative larger C-terminal fragment (CTF) increased in the APP-S675E cells, whereas the CTF form that was most abundant in cells expressing APPwt or APP-S675A decreased in the APP-S675E cells. Upon siRNA-mediated knockdown of the astacin metalloprotease meprin β, the levels of the alternative CTF decreased and the CTF ratio was restored back to APPwt levels. Our findings suggest that APP-Ser-675 phosphorylation alters the balance of APP processing, increasing meprin β-mediated and decreasing α-secretase-mediated processing of APP at the plasma membrane. As meprin β cleavage of APP has been shown to result in formation of highly aggregation-prone, truncated Aβ2-40/42 peptides, enhanced APP processing by this enzyme could contribute to AD pathology. We propose that it would be of interest to clarify in future studies how APP-Ser-675 phosphorylation promotes meprin β-mediated APP cleavage.
Collapse
Affiliation(s)
- Preeti Kumaran Menon
- Department of Biochemistry and Biophysics, Stockholm University, 106 91 Stockholm, Sweden
| | - Niina Anneli Koistinen
- Department of Biochemistry and Biophysics, Stockholm University, 106 91 Stockholm, Sweden
| | - Kerstin Iverfeldt
- Department of Biochemistry and Biophysics, Stockholm University, 106 91 Stockholm, Sweden
| | - Anna-Lena Ström
- Department of Biochemistry and Biophysics, Stockholm University, 106 91 Stockholm, Sweden
| |
Collapse
|
86
|
Cervellati C, Trentini A, Rosta V, Passaro A, Bosi C, Sanz JM, Bonazzi S, Pacifico S, Seripa D, Valacchi G, Guerini R, Zuliani G. Serum beta-secretase 1 (BACE1) activity as candidate biomarker for late-onset Alzheimer's disease. GeroScience 2019; 42:159-167. [PMID: 31745860 DOI: 10.1007/s11357-019-00127-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/17/2019] [Indexed: 01/02/2023] Open
Abstract
Beta-secretase (BACE1) is a key enzyme in the formation of amyloid-β; its activity/concentration is increased in brain and cerebrospinal fluid of patients with late-onset Alzheimer's disease (LOAD). Since BACE1 was found also in blood, we evaluated its potential as peripheral biomarker. To this aim, serum BACE1 activity was assessed in 115 subjects with LOAD and 151 controls. We found that BACE1 changed across groups (p < 0.001) with a 25% increase in LOAD versus controls. High levels of BACE1 (IV quartile) were independently associated with the diagnosis of LOAD (OR 2.8; 1.4-5.7). Diagnostic accuracy was 76% for LOAD. Our data suggest that increased BACE1 activity in serum may represent a potential biomarker for LOAD. Additional studies are needed to confirm the usefulness of BACE1, alone or in combination with other markers, in discriminating patients and predicting LOAD onset and progression.
Collapse
Affiliation(s)
- Carlo Cervellati
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy.
| | - Alessandro Trentini
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Valentina Rosta
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Angelina Passaro
- Department of Morphology, Surgery, and Medical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Cristina Bosi
- Department of Morphology, Surgery, and Medical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Juana Maria Sanz
- Department of Morphology, Surgery, and Medical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Stefania Bonazzi
- Department of Morphology, Surgery, and Medical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Salvatore Pacifico
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Davide Seripa
- Research Laboratory, Complex Structure of Geriatrics, Department of Medical Sciences, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini, 1, 71013, San Giovanni Rotondo, Italy
| | - Giuseppe Valacchi
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy.,Plants for Human Health Institute, Animal Science Department, NC State University, 600 Laureate Way, Kannapolis, NC, 28081, USA.,Department of Food and Nutrition, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Remo Guerini
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| | - Giovanni Zuliani
- Department of Morphology, Surgery, and Medical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121, Ferrara, Italy
| |
Collapse
|
87
|
Yen YC, Kammeyer AM, Jensen KC, Tirlangi J, Ghosh AK, Mesecar AD. Development of an Efficient Enzyme Production and Structure-Based Discovery Platform for BACE1 Inhibitors. Biochemistry 2019; 58:4424-4435. [PMID: 31549827 PMCID: PMC7284891 DOI: 10.1021/acs.biochem.9b00714] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACE1 (Beta-site Amyloid Precursor Protein (APP) Cleaving Enzyme 1) is a promising therapeutic target for Alzheimer's Disease (AD). However, efficient expression, purification, and crystallization systems are not well described or detailed in the literature nor are approaches for treatment of enzyme kinetic data for potent inhibitors well described. We therefore developed a platform for expression and purification of BACE1, including protein refolding from E.coli inclusion bodies, in addition to optimizing a reproducible crystallization procedure of BACE1 bound with inhibitors. We also report a detailed approach to the proper analysis of enzyme kinetic data for compounds that exhibit either rapid-equilibrium or tight-binding mechanisms. Our methods allow for the purification of ∼15 mg of BACE1 enzyme from 1 L of culture which is higher than reported yields in the current literature. To evaluate the data analysis approach developed here, a well-known potent inhibitor and two of its derivatives were tested, analyzed, and compared. The inhibitory constants (Ki) obtained from the kinetic studies are in agreement with dissociation constants (Kd) that were also determined using isothermal titration calorimetry (ITC) experiments. The X-ray structures of these three compounds in complex with BACE1 were readily obtained and provide important insight into the structure and thermodynamics of the BACE1-inhibitor interactions.
Collapse
Affiliation(s)
- Yu-Chen Yen
- Department of Biological Sciences, Purdue University, West Lafayette Indiana 47907, United States
| | - Annalissa M. Kammeyer
- Department of Biological Sciences, Purdue University, West Lafayette Indiana 47907, United States
| | - Katherine C. Jensen
- Department of Biological Sciences, Purdue University, West Lafayette Indiana 47907, United States
| | | | - Arun K. Ghosh
- Department of Chemistry, Purdue University, West Lafayette Indiana 47907, United States, Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette Indiana 47907, United States
| | - Andrew D. Mesecar
- Department of Biological Sciences, Purdue University, West Lafayette Indiana 47907, United States, Department of Chemistry, Purdue University, West Lafayette Indiana 47907, United States, Department of Biochemistry, Purdue University, West Lafayette Indiana 47907, United States,Corresponding Author:. Tel.: (765) 494-1924
| |
Collapse
|
88
|
Discovery of a series of selective and cell permeable beta-secretase (BACE1) inhibitors by fragment linking with the assistance of STD-NMR. Bioorg Chem 2019; 92:103253. [DOI: 10.1016/j.bioorg.2019.103253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 06/27/2019] [Accepted: 09/04/2019] [Indexed: 11/23/2022]
|
89
|
Molecular insights into the inhibitory mechanism of bi-functional bis-tryptoline triazole against β-secretase (BACE1) enzyme. Amino Acids 2019; 51:1593-1607. [PMID: 31654211 DOI: 10.1007/s00726-019-02797-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023]
Abstract
The β-site amyloid precursor protein-cleaving enzyme 1 (β-secretase, BACE1) is involved in the formation of amyloid-β (Aβ) peptide that aggregates into soluble oligomers, amyloid fibrils, and plaques responsible for the neurodegeneration in Alzheimer disease (AD). BACE1 is one of the prime therapeutic targets for the design of inhibitors against AD as BACE1 participate in the rate-limiting step in Aβ production. Jiaranaikulwanitch et al. reported bis-tryptoline triazole (BTT) compound as a potent inhibitor against BACE1, Aβ aggregation as well as possessing metal chelation and antioxidant activity. However, the molecular mechanism of BACE1 inhibition by BTT remains unclear. Thus, molecular docking and molecular dynamics (MD) simulations were performed to elucidate the inhibitory mechanism of BTT against BACE1. MD simulations highlight that BTT interact with catalytic aspartic dyad residues (Asp32 and Asp228) and active pocket residues of BACE1. The hydrogen-bond interactions, hydrophobic contacts, and π-π stacking interactions of BTT with flap residues (Val67-Asp77) of BACE1 confine the movement of the flap and help to achieve closed (non-active) conformation. The PCA analysis highlights lower conformational fluctuations for BACE1-BTT complex, which suggests enhanced conformational stability in comparison to apo-BACE1. The results of the present study provide key insights into the underlying inhibitory mechanism of BTT against BACE1 and will be helpful for the rational design of novel inhibitors with enhanced potency against BACE1.
Collapse
|
90
|
Barrett T, Marchalant Y, Park KH. p35 Hemizygous Deletion in 5xFAD Mice Increases Aβ Plaque Load in Males but Not in Females. Neuroscience 2019; 417:45-56. [DOI: 10.1016/j.neuroscience.2019.08.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 07/25/2019] [Accepted: 08/08/2019] [Indexed: 12/29/2022]
|
91
|
Li HM, Yu SP, Fan TY, Zhong Y, Gu T, Wu WY, Zhao C, Chen Z, Chen M, Li NG, Wang XL. Design, synthesis, and biological activity evaluation of BACE1 inhibitors with antioxidant activity. Drug Dev Res 2019; 81:206-214. [PMID: 31397505 DOI: 10.1002/ddr.21585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/06/2019] [Accepted: 07/24/2019] [Indexed: 01/26/2023]
Abstract
The proteolytic enzyme β-secretase (BACE1) plays a central role in the synthesis of the pathogenic β-amyloid peptides (Aβ) in Alzheimer's disease (AD), antioxidants could attenuate the AD syndrome and prevent the disease progression. In this study, BACE1 inhibitors (D1-D18) with free radical-scavenging activities were synthesized by molecular hybridization of 2-aminopyridine with natural antioxidants. The biological activity evaluation showed that D1 had obvious inhibitory activity against BACE1, and strong antioxidant activity in 1,1-diphenyl-2-picrylhydrazyl (DPPH) and 2,2'-azinobis-(3-ethylbenzthiazoline-6-sulphonate) (ABTS+• ) assay, which could be used as a lead compound for further study.
Collapse
Affiliation(s)
- He-Min Li
- Department of Medicinal Chemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shao-Peng Yu
- Department of Medicinal Chemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tian-Yuan Fan
- Department of Medicinal Chemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Zhong
- Department of Medicinal Chemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ting Gu
- Department of Medicinal Chemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wen-Yu Wu
- Department of Medicinal Chemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chao Zhao
- Department of Medicinal Chemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhi Chen
- Department of Medicinal Chemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Chen
- Department of Medicinal Chemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Nian-Guang Li
- Department of Medicinal Chemistry, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiao-Long Wang
- Department of Medicinal Chemistry, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
92
|
Scharfenberg F, Armbrust F, Marengo L, Pietrzik C, Becker-Pauly C. Regulation of the alternative β-secretase meprin β by ADAM-mediated shedding. Cell Mol Life Sci 2019; 76:3193-3206. [PMID: 31201463 PMCID: PMC11105663 DOI: 10.1007/s00018-019-03179-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 05/23/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022]
Abstract
Alzheimer's Disease (AD) is the sixth-leading cause of death in industrialized countries. Neurotoxic amyloid-β (Aβ) plaques are one of the pathological hallmarks in AD patient brains. Aβ accumulates in the brain upon sequential, proteolytic processing of the amyloid precursor protein (APP) by β- and γ-secretases. However, so far disease-modifying drugs targeting β- and γ-secretase pathways seeking a decrease in the production of toxic Aβ peptides have failed in clinics. It has been demonstrated that the metalloproteinase meprin β acts as an alternative β-secretase, capable of generating truncated Aβ2-x peptides that have been described to be increased in AD patients. This indicates an important β-site cleaving enzyme 1 (BACE-1)-independent contribution of the metalloprotease meprin β within the amyloidogenic pathway and may lead to novel drug targeting avenues. However, meprin β itself is embedded in a complex regulatory network. Remarkably, the anti-amyloidogenic α-secretase a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) is a direct competitor for APP at the cell surface, but also a sheddase of inactive pro-meprin β. Overall, we highlight the current cellular, molecular and structural understanding of meprin β as alternative β-secretase within the complex protease web, regulating APP processing in health and disease.
Collapse
Affiliation(s)
- Franka Scharfenberg
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Fred Armbrust
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Liana Marengo
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Claus Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | - Christoph Becker-Pauly
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany.
| |
Collapse
|
93
|
Multi-target design strategies for the improved treatment of Alzheimer's disease. Eur J Med Chem 2019; 176:228-247. [DOI: 10.1016/j.ejmech.2019.05.020] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/06/2019] [Accepted: 05/06/2019] [Indexed: 12/13/2022]
|
94
|
Rodríguez-Soacha DA, Scheiner M, Decker M. Multi-target-directed-ligands acting as enzyme inhibitors and receptor ligands. Eur J Med Chem 2019; 180:690-706. [PMID: 31401465 DOI: 10.1016/j.ejmech.2019.07.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/04/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022]
Abstract
In this review, we present the latest advances in the field of multi-target-directed ligand (MTDL) design for the treatment of various complex pathologies of multifactorial origin. In particular, latest findings in the field of MTDL design targeting both an enzyme and a receptor are presented for different diseases such as Alzheimer's disease (AD), depression, addiction, glaucoma, non-alcoholic steatohepatitis and pain and inflammation. The ethology of the diseases is briefly described, with special emphasis on how the MTDL can evolve into novel therapies that replace the classic pharmacological dogma "one target one disease". Considering the current needs for therapy adherence improvement, it is exposed as from the medicinal chemistry, different molecular scaffolds are studied. With the use of structure activity relationship studies and molecular optimization, new hybrid molecules are generated with improved biological properties acting at two biologically very distinct targets.
Collapse
Affiliation(s)
- Diego Alejandro Rodríguez-Soacha
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Matthias Scheiner
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074, Würzburg, Germany.
| |
Collapse
|
95
|
Arya R, Gupta SP, Paliwal S, Sharma S, Madan K, Chauhan M. Pharmacophore Modeling and Docking Studies to Investigate Potential Leads for the Development of β -Secretase APP Cleavage Enzyme-1 (BACE-1) Inhibitors. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1570180815666181023110736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Alzheimer’s disease is a medical condition with detrimental brain health. It is
majorly diagnosed in aging individuals plaque in β) characterized by accumulated Amyloidal beta (A
1 BACE) 1 secretase APP cleavage enzyme βneurological areas. The ) is the target of choice that can
be exploited to find drugs against Alzheimer’s disease.
Methods:
A series of BACE-1 inhibitors with reported binding constant were considered for the development
of a feature based pharmacophore model.
Results:
The good correlation coefficient (r=0.91) and RMSD of 0.93 was observed with 30 compounds
in training set. The model was validated internally (r2test=0.76) as well as externally by Fischer validation.
The pharmacophore based virtual screening retrieved compounds that were docked and biologically
evaluated.
Conclusion:
The three structurally diverse molecules were tested by in-vitro method. The pyridine
derivative with highest fit value (6.9) exhibited IC50 value of 2.70 µM and thus was found to be the most
promising lead molecule as BACE-1 inhibitor.
Collapse
Affiliation(s)
- Richa Arya
- Banasthali University, 304022, Banasthali, (Raj.), India
| | - Satya Prakash Gupta
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, 250005, Meerut, India
| | | | - Swapnil Sharma
- Banasthali University, 304022, Banasthali, (Raj.), India
| | - Kirtika Madan
- Banasthali University, 304022, Banasthali, (Raj.), India
| | - Monika Chauhan
- Banasthali University, 304022, Banasthali, (Raj.), India
| |
Collapse
|
96
|
Forman M, Palcza J, Tseng J, Stone JA, Walker B, Swearingen D, Troyer MD, Dockendorf MF. Safety, Tolerability, and Pharmacokinetics of the β-Site Amyloid Precursor Protein-Cleaving Enzyme 1 Inhibitor Verubecestat (MK-8931) in Healthy Elderly Male and Female Subjects. Clin Transl Sci 2019; 12:545-555. [PMID: 31215755 PMCID: PMC6742941 DOI: 10.1111/cts.12645] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/10/2019] [Indexed: 12/21/2022] Open
Abstract
β‐site amyloid precursor protein‐cleaving enzyme 1 (BACE1) is required for the production of β‐amyloid peptides, which are implicated in the etiology of Alzheimer's disease. The safety and pharmacokinetics of the BACE1 inhibitor verubecestat have previously been studied in young adults aged 19–45 years. In this randomized, placebo‐controlled, phase I study (protocol MK‐8931‐006), we investigated the safety, tolerability, and pharmacokinetics of a single dose (100 mg) or multiple doses (30, 80, and 120 mg) once daily for 28 days of verubecestat in healthy elderly subjects. Safety end points were assessed at baseline and during the duration of the study period and indicated that verubecestat was generally well tolerated. Verubecestat pharmacokinetics were similar between healthy elderly male and female subjects and similar to those reported in healthy young males in previous studies. These data supported subsequent studies to assess the potential efficacy of verubecestat in subjects with Alzheimer's disease.
Collapse
Affiliation(s)
- Mark Forman
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - John Palcza
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Jack Tseng
- Merck & Co., Inc., Kenilworth, New Jersey, USA.,Purdue Pharma, Stamford, Connecticut, USA
| | | | | | | | - Matthew D Troyer
- Merck & Co., Inc., Kenilworth, New Jersey, USA.,Denali Therapies, South San Francisco, California, USA
| | | |
Collapse
|
97
|
Xiang J, Zhang W, Cai XF, Cai M, Yu ZH, Yang F, Zhu W, Li XT, Wu T, Zhang JS, Cai DF. DNA Aptamers Targeting BACE1 Reduce Amyloid Levels and Rescue Neuronal Deficiency in Cultured Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:302-312. [PMID: 30959405 PMCID: PMC6453838 DOI: 10.1016/j.omtn.2019.02.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 02/03/2019] [Accepted: 02/04/2019] [Indexed: 01/02/2023]
Abstract
β-amyloid (Aβ) plays an essential role in the pathogenesis of Alzheimer's disease (AD). Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) is indispensable for Aβ production, and knockout of BACE1 has no overt phenotypes in mouse. Thus, fine modulation of BACE1 may be a safe and effective treatment for AD patients. However, the large active site of BACE1 makes it challenging to target BACE1 with classical small-molecule inhibitors. DNA aptamer can have high affinity and specificity against diverse targets, and it provides an alternative strategy to target BACE1. In this study, we used a novel cell-systematic evolution of ligands by exponential enrichment (SELEX) strategy to select specific DNA aptamers optimized to target BACE1 under physiological status. After 17 rounds of selection, we identified two DNA aptamers against BACE1: BI1 and BI2. The identified aptamers interacted with BACE1 in pull-down assay, inhibited BACE1 activity in in vitro fluorescence resonance energy transfer (FRET) assay and HEK293-APP stable cell line, reduced Aβ in the culture medium of HEK293-amyloid protein precursor (APP) stable cell line and APP-PS1 primary cultured neurons, and rescued Aβ-induced neuronal deficiency in APP-PS1 primary cultured neurons. In contrast, the identified aptamers had no effect on α- or γ-secretase. In addition, cholesteryl tetraetylene glycol (TEG) modification further improved the potency of the identified aptamers. Our study suggests that it is feasible and effective to target BACE1 with DNA aptamers, and the therapeutic potential of the identified aptamers deserves further investigation.
Collapse
Affiliation(s)
- Jun Xiang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Wen Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Xiao-Fang Cai
- Department of Stomatology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Min Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Zhong-Hai Yu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Feng Yang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Wen Zhu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Xiang-Ting Li
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Ting Wu
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai 200032, China
| | - Jing-Si Zhang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai 200032, China.
| | - Ding-Fang Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Laboratory of Neurology, Institute of Integrative Medicine, Fudan University, Shanghai 200032, China.
| |
Collapse
|
98
|
NRF2/ARE pathway negatively regulates BACE1 expression and ameliorates cognitive deficits in mouse Alzheimer's models. Proc Natl Acad Sci U S A 2019; 116:12516-12523. [PMID: 31164420 DOI: 10.1073/pnas.1819541116] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACE1 is the rate-limiting enzyme for amyloid-β peptides (Aβ) generation, a key event in the pathogenesis of Alzheimer's disease (AD). By an unknown mechanism, levels of BACE1 and a BACE1 mRNA-stabilizing antisense RNA (BACE1-AS) are elevated in the brains of AD patients, implicating that dysregulation of BACE1 expression plays an important role in AD pathogenesis. We found that nuclear factor erythroid-derived 2-related factor 2 (NRF2/NFE2L2) represses the expression of BACE1 and BACE1-AS through binding to antioxidant response elements (AREs) in their promoters of mouse and human. NRF2-mediated inhibition of BACE1 and BACE1-AS expression is independent of redox regulation. NRF2 activation decreases production of BACE1 and BACE1-AS transcripts and Aβ production and ameliorates cognitive deficits in animal models of AD. Depletion of NRF2 increases BACE1 and BACE1-AS expression and Aβ production and worsens cognitive deficits. Our findings suggest that activation of NRF2 can prevent a key early pathogenic process in AD.
Collapse
|
99
|
Yuksel M, Tacal O. Trafficking and proteolytic processing of amyloid precursor protein and secretases in Alzheimer's disease development: An up-to-date review. Eur J Pharmacol 2019; 856:172415. [PMID: 31132354 DOI: 10.1016/j.ejphar.2019.172415] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/26/2019] [Accepted: 05/23/2019] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD), which is predicted to affect 1 in 85 persons worldwide by 2050, results in progressive loss of neuronal functions, leading to impairments in memory and cognitive abilities. As being one of the major neuropathological hallmarks of AD, senile plaques mainly consist of amyloid-β (Aβ) peptides, which are derived from amyloid precursor protein (APP) via the sequential cleavage by β- and γ-secretases. Although the overproduction and accumulation of Aβ peptides are at the center of AD research, the new discoveries point out to the complexity of the disease development. In this respect, it is crucial to understand the processing and the trafficking of APP, the enzymes involved in its processing, the cleavage products and their therapeutic potentials. This review summarizes the salient features of APP processing focusing on APP, the canonical secretases as well as the novel secretases and the cleavage products with an update of the recent developments. We also discussed the intracellular trafficking of APP and secretases in addition to their potential in AD therapy.
Collapse
Affiliation(s)
- Melike Yuksel
- Department of Biochemistry, School of Pharmacy, Hacettepe University, 06100, Ankara, Turkey.
| | - Ozden Tacal
- Department of Biochemistry, School of Pharmacy, Hacettepe University, 06100, Ankara, Turkey.
| |
Collapse
|
100
|
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: 1.2] [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
|