51
|
Johannsen J, Weiss D, Daubmann A, Schmitz L, Denecke J. Evaluation of putative CSF biomarkers in paediatric spinal muscular atrophy (SMA) patients before and during treatment with nusinersen. J Cell Mol Med 2021; 25:8419-8431. [PMID: 34312963 PMCID: PMC8419176 DOI: 10.1111/jcmm.16802] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/31/2021] [Accepted: 07/07/2021] [Indexed: 01/22/2023] Open
Abstract
Spinal muscular atrophy (SMA) is a genetic neurodegenerative disorder leading to immobilization and premature death. Currently, three alternative therapeutic options are available. Therefore, biomarkers that might reflect or predict the clinical course of the individual patient with treatment are of great potential use. Currently, the antisense oligonucleotide nusinersen is the prevalent and longest validated therapy for SMA. We analysed CSF candidate biomarkers for degenerative CNS processes (namely phosphorylated heavy chain (pNf-H), light-chain neurofilaments (NfL), total tau protein (T-Tau), neurogranin, β-secretase BACE-1 and alpha-synuclein) in 193 CSF samples of 44 paediatric SMA types 1, 2 and 3 patients before and under nusinersen treatment and related them to standardized clinical outcome scores in a single-centre pilot study. pNf-H and NfL correlated with disease severity and activity, emphasizing their relevance as marker of neuronal loss and clinical outcome. T-Tau was significantly correlated with motor function scores in SMA type 1 making it an interesting marker for treatment response. Additionally, baseline T-Tau levels were elevated in most SMA patients possibly reflecting the extension of neuronal degeneration in paediatric-onset SMA. Further investigations of these CSF proteins might be beneficial for paediatric SMA subtypes and treatment modalities as an indicator for clinical outcome and should be analysed in larger cohorts.
Collapse
Affiliation(s)
- Jessika Johannsen
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Deike Weiss
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Daubmann
- Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leonie Schmitz
- Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonas Denecke
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
52
|
Vilela AFL, Narciso Dos Reis VE, Cardoso CL. Co-Immobilized Capillary Enzyme Reactor Based on Beta-Secretase1 and Acetylcholinesterase: A Model for Dual-Ligand Screening. Front Chem 2021; 9:708374. [PMID: 34307303 PMCID: PMC8295500 DOI: 10.3389/fchem.2021.708374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/23/2021] [Indexed: 11/13/2022] Open
Abstract
We have developed a dual enzymatic system assay involving liquid chromatography-mass spectrometry (LC–MS) to screen AChE and BACE1 ligands. A fused silica capillary (30 cm × 0.1 mm i.d. × 0.362 mm e.d.) was used as solid support. The co-immobilization procedure encompassed two steps and random immobilization. The resulting huAChE+BACE1-ICER/MS was characterized by using acetylcholine (ACh) and JMV2236 as substrates. The best conditions for the dual enzymatic system assay were evaluated and compared to the conditions of the individual enzymatic system assays. Analysis was performed in series for each enzyme. The kinetic parameters (KMapp) and inhibition assays were evaluated. To validate the system, galantamine and a β-secretase inhibitor were employed as standard inhibitors, which confirmed that the developed screening assay was able to identify reference ligands and to provide quantitative parameters. The combination of these two enzymes in a single on-line system allowed possible multi-target inhibitors to be screened and identified. The innovative huAChE+BACE1-ICER/MS dual enzymatic system reported herein proved to be a reliable tool to identify and to characterize hit ligands for AChE and BACE1 in an enzymatic competitive environment. This innovative system assay involved lower costs; measured the product from enzymatic hydrolysis directly by MS; enabled immediate recovery of the enzymatic activity; showed specificity, selectivity, and sensitivity; and mimicked the cellular process.
Collapse
Affiliation(s)
- Adriana Ferreira Lopes Vilela
- Departamento de Química, Grupo de Cromatografia de Bioafinidade e Produtos Naturais, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Vitor Eduardo Narciso Dos Reis
- Departamento de Química, Grupo de Cromatografia de Bioafinidade e Produtos Naturais, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Carmen Lúcia Cardoso
- Departamento de Química, Grupo de Cromatografia de Bioafinidade e Produtos Naturais, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
53
|
Lomoio S, Willen R, Kim W, Ho KZ, Robinson EK, Prokopenko D, Kennedy ME, Tanzi RE, Tesco G. Gga3 deletion and a GGA3 rare variant associated with late onset Alzheimer's disease trigger BACE1 accumulation in axonal swellings. Sci Transl Med 2021; 12:12/570/eaba1871. [PMID: 33208500 DOI: 10.1126/scitranslmed.aba1871] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 05/18/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022]
Abstract
Axonal dystrophy, indicative of perturbed axonal transport, occurs early during Alzheimer's disease (AD) pathogenesis. Little is known about the mechanisms underlying this initial sign of the pathology. This study proves that Golgi-localized γ-ear-containing ARF binding protein 3 (GGA3) loss of function, due to Gga3 genetic deletion or a GGA3 rare variant that cosegregates with late-onset AD, disrupts the axonal trafficking of the β-site APP-cleaving enzyme 1 (BACE1) resulting in its accumulation in axonal swellings in cultured neurons and in vivo. We show that BACE pharmacological inhibition ameliorates BACE1 axonal trafficking and diminishes axonal dystrophies in Gga3 null neurons in vitro and in vivo. These data indicate that axonal accumulation of BACE1 engendered by GGA3 loss of function results in local toxicity leading to axonopathy. Gga3 deletion exacerbates axonal dystrophies in a mouse model of AD before β-amyloid (Aβ) deposition. Our study strongly supports a role for GGA3 in AD pathogenesis, where GGA3 loss of function triggers BACE1 axonal accumulation independently of extracellular Aβ, and initiates a cascade of events leading to the axonal damage distinctive of the early stage of AD.
Collapse
Affiliation(s)
- Selene Lomoio
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Rachel Willen
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - WonHee Kim
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Kevin Z Ho
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Edward K Robinson
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Dmitry Prokopenko
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | | | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Giuseppina Tesco
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA.
| |
Collapse
|
54
|
Qiao O, Ji H, Zhang Y, Zhang X, Zhang X, Liu N, Huang L, Liu C, Gao W. New insights in drug development for Alzheimer's disease based on microglia function. Biomed Pharmacother 2021; 140:111703. [PMID: 34083109 DOI: 10.1016/j.biopha.2021.111703] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/26/2022] Open
Abstract
One of the biggest challenges in drug development for Alzheimer's disease (AD) is how to effectively remove deposits of amyloid-beta (Aβ). Recently, the relationship between microglia and Aβ has become a research hotspot. Emerging evidence suggests that Aβ-induced microglia-mediated neuroinflammation further aggravates the decline of cognitive function, while microglia are also involved in the process of Aβ clearance. Hence, microglia have become a potential therapeutic target for the treatment or prevention of AD. An in-depth understanding of the role played by microglia in the development of AD will help us to broaden therapeutic strategies for AD. In this review, we provide an overview of the dual roles of microglia in AD progression: the positive effect of phagocytosis of Aβ and its negative effect on neuroinflammation after over-activation. With the advantages of novel structure, high efficiency, and low toxicity, small-molecule compounds as modulators of microglial function have attracted considerable attention in the therapeutic areas of AD. In this review, we also summarize the therapeutic potential of small molecule compounds (SMCs) and their structure-activity relationship for AD treatment through modulating microglial phagocytosis and inhibiting neuroinflammation. For example, the position and number of phenolic hydroxyl groups on the B ring are the key to the activity of flavonoids, and the substitution of hydroxyl groups on the benzene ring enhances the anti-inflammatory activity of phenolic acids. This review is expected to be useful for developing effective modulators of microglial function from SMCs for the amelioration and treatment of AD.
Collapse
Affiliation(s)
- Ou Qiao
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Haixia Ji
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Yi Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Xinyu Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Xueqian Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Na Liu
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Luqi Huang
- Chinese Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Changxiao Liu
- The State Key Laboratories of Pharmacodynamics and Pharmacokinetics, Tianjin 300193, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China.
| |
Collapse
|
55
|
Structural Studies Providing Insights into Production and Conformational Behavior of Amyloid-β Peptide Associated with Alzheimer's Disease Development. MOLECULES (BASEL, SWITZERLAND) 2021; 26:molecules26102897. [PMID: 34068293 PMCID: PMC8153327 DOI: 10.3390/molecules26102897] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease is the most common type of neurodegenerative disease in the world. Genetic evidence strongly suggests that aberrant generation, aggregation, and/or clearance of neurotoxic amyloid-β peptides (Aβ) triggers the disease. Aβ accumulates at the points of contact of neurons in ordered cords and fibrils, forming the so-called senile plaques. Aβ isoforms of different lengths are found in healthy human brains regardless of age and appear to play a role in signaling pathways in the brain and to have neuroprotective properties at low concentrations. In recent years, different substances have been developed targeting Aβ production, aggregation, interaction with other molecules, and clearance, including peptide-based drugs. Aβ is a product of sequential cleavage of the membrane glycoprotein APP (amyloid precursor protein) by β- and γ-secretases. A number of familial mutations causing an early onset of the disease have been identified in the APP, especially in its transmembrane domain. The mutations are reported to influence the production, oligomerization, and conformational behavior of Aβ peptides. This review highlights the results of structural studies of the main proteins involved in Alzheimer's disease pathogenesis and the molecular mechanisms by which perspective therapeutic substances can affect Aβ production and nucleation.
Collapse
|
56
|
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: 411] [Impact Index Per Article: 102.8] [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
|
57
|
Rueeger H, Lueoend R, Machauer R, Veenstra SJ, Holzer P, Hurth K, Voegtle M, Frederiksen M, Rondeau JM, Tintelnot-Blomley M, Jacobson LH, Staufenbiel M, Laue G, Neumann U. Synthesis of the Potent, Selective, and Efficacious β-Secretase (BACE1) Inhibitor NB-360. J Med Chem 2021; 64:4677-4696. [PMID: 33844524 DOI: 10.1021/acs.jmedchem.0c02143] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Starting from lead compound 4, the 1,4-oxazine headgroup was optimized to improve potency and brain penetration. Focusing at the 6-position of the 5-amino-1,4-oxazine, the insertion of a Me and a CF3 group delivered an excellent pharmacological profile with a pKa of 7.1 and a very low P-gp efflux ratio enabling high central nervous system (CNS) penetration and exposure. Various synthetic routes to access BACE1 inhibitors bearing a 5-amino-6-methyl-6-(trifluoromethyl)-1,4-oxazine headgroup were investigated. Subsequent optimization of the P3 fragment provided the highly potent N-(3-((3R,6R)-5-amino-3,6-dimethyl-6-(trifluoromethyl)-3,6-dihydro-2H-1,4-oxazin-3-yl)-4-fluorophenyl)-5-cyano-3-methylpicolinamide 54 (NB-360), able to reduce significantly Aβ levels in mice, rats, and dogs in acute and chronic treatment regimens.
Collapse
|
58
|
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
|
59
|
Tsumagari K, Chang CH, Ishihama Y. Exploring the landscape of ectodomain shedding by quantitative protein terminomics. iScience 2021; 24:102259. [PMID: 33796845 PMCID: PMC7995609 DOI: 10.1016/j.isci.2021.102259] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/18/2021] [Accepted: 02/26/2021] [Indexed: 02/08/2023] Open
Abstract
Ectodomain shedding is a proteolytic process that regulates the levels and functions of membrane proteins. Dysregulated shedding is linked to severe diseases, including cancer and Alzheimer's disease. However, the exact cleavage sites of shedding substrates remain largely unknown. Here, we explore the landscape of ectodomain shedding by generating large-scale, cell-type-specific maps of shedding cleavage sites. By means of N- and C-terminal peptide enrichment and quantitative mass spectrometry, we quantified protein termini in the culture media of 10 human cell lines and identified 489 cleavage sites on 163 membrane proteins whose proteolytic terminal fragments are downregulated in the presence of a broad-spectrum metalloprotease inhibitor. A major fraction of the presented cleavage sites was identified in a cell-type-specific manner and mapped onto receptors, cell adhesion molecules, and protein kinases and phosphatases. We confidently identified 86 cleavage sites as metalloprotease substrates by means of knowledge-based scoring. Secretomes across 10 human cell lines were investigated by protein terminomics Cell-type-specific maps of shedding cleavage sites were generated Most of the cleavage sites were identified in a cell-type-specific manner Knowledge-based scoring enabled prediction of responsible sheddases
Collapse
Affiliation(s)
- Kazuya Tsumagari
- Department of Molecular and Cellular BioAnalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
- Eisai-Keio Innovation Laboratory for Dementia, Center for Integrated Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Chih-Hsiang Chang
- Department of Molecular and Cellular BioAnalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yasushi Ishihama
- Department of Molecular and Cellular BioAnalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
- Department of Proteomics and Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
- Corresponding author
| |
Collapse
|
60
|
Barber J, Sikakana P, Sadler C, Baud D, Valentin JP, Roberts R. A target safety assessment of the potential toxicological risks of targeting plasmepsin IX/X for the treatment of malaria. Toxicol Res (Camb) 2021; 10:203-213. [PMID: 33884171 DOI: 10.1093/toxres/tfaa106] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/28/2022] Open
Abstract
The aspartic proteases plasmepsin IX/X are important antimalarial drug targets due to their specificity to the malaria parasite and their vital role as mediators of disease progression. Focusing on parasite-specific targets where no human homologue exists reduces the possibility of on-target drug toxicity. However, there is a risk of toxicity driven by inadequate selectivity for plasmepsins IX/X in Plasmodium over related mammalian aspartic proteases. Of these, CatD/E may be of most toxicological relevance as CatD is a ubiquitous lysosomal enzyme present in most cell types and CatE is found in the gut and in erythrocytes, the clinically significant site of malarial infection. Based on mammalian aspartic protease physiology and adverse drug reactions (ADRs) to FDA-approved human immunodeficiency virus (HIV) aspartic protease inhibitors, we predicted several potential toxicities including β-cell and congenital abnormalities, hypotension, hypopigmentation, hyperlipidaemia, increased infection risk and respiratory, renal, gastrointestinal, dermatological, and other epithelial tissue toxicities. These ADRs to the HIV treatments are likely to be a result of host aspartic protease inhibition due a lack of specificity for the HIV protease; plasmepsins are much more closely related to human CatD than to HIV proteinase. Plasmepsin IX/X inhibition presents an opportunity to specifically target Plasmodium as an effective antimalarial treatment, providing adequate selectivity can be obtained. Potential plasmepsin IX/X inhibitors should be assayed for inhibitory activity against the main human aspartic proteases and particularly CatD/E. An investigative rodent study conducted early in drug discovery would serve as an initial risk assessment of the potential hazards identified.
Collapse
Affiliation(s)
- Jane Barber
- ApconiX, Alderley Park, Alderley Edge, SK10 4TG, UK
| | | | | | - Delphine Baud
- Medicines for Malaria Venture, 20 Route de Pré-Bois, Geneva 1215, Switzerland
| | - Jean-Pierre Valentin
- UCB Biopharma SRL, Building R9, Chemin du Foriest, 1420 Braine-l'Alleud, Belgium
| | - Ruth Roberts
- ApconiX, Alderley Park, Alderley Edge, SK10 4TG, UK
| |
Collapse
|
61
|
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
|
62
|
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
|
63
|
Wen W, Li P, Liu P, Xu S, Wang F, Huang JH. Post-Translational Modifications of BACE1 in Alzheimer's Disease. Curr Neuropharmacol 2021; 20:211-222. [PMID: 33475074 PMCID: PMC9199555 DOI: 10.2174/1570159x19666210121163224] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/27/2020] [Accepted: 01/21/2021] [Indexed: 11/23/2022] Open
Abstract
Beta-Amyloid Cleaving Enzyme1 (BACE1) is a monospecific enzyme for the key rate-limiting step in the synthesis of beta-amyloid(Aβ) from cleavage of amyloid precursor protein (APP), to form senile plaques and causes cognitive dysfunction in Alzheimer's disease (AD). Post-translation modifications of BACE1, such as acetylation, glycosylation, palmitoylation, phosphorylation, play a crucial role in the trafficking and maturation process of BACE1. The study of BACE1 is of great importance not only for understanding the formation of toxic Aβ but also for the development of an effective therapeutic target for the treatment of AD. This paper review recent advances in the studies about BACE1, with focuses being paid to the relationship of Aβ, BACE1 with post- translational regulation of BACE1. In addition, we specially reviewed studies about the compounds that can be used to affect post-translational regulation of BACE1 or regulate BACE1 in the literature, which can be used for subsequent research on whether BACE1 is a post-translationally modified drug.
Collapse
Affiliation(s)
- Wen Wen
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137. China
| | - Ping Li
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137. China
| | - Panwang Liu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137. China
| | - Shijun Xu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137. China
| | - Fushun Wang
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu, Sichuan 610000. China
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott & White Health Science Center, Temple, TX 79409. United States
| |
Collapse
|
64
|
Syeda T, Cannon JR. Environmental exposures and the etiopathogenesis of Alzheimer's disease: The potential role of BACE1 as a critical neurotoxic target. J Biochem Mol Toxicol 2021; 35:e22694. [PMID: 33393683 DOI: 10.1002/jbt.22694] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a major public health crisis due to devastating cognitive symptoms, a lack of curative treatments, and increasing prevalence. Most cases are sporadic (>95% of cases) after the age of 65 years, implicating an important role of environmental factors in disease pathogenesis. Environmental neurotoxicants have been implicated in neurodegenerative disorders including Parkinson's Disease and AD. Animal models of AD and in vitro studies have shed light on potential neuropathological mechanisms, yet the biochemical and molecular underpinnings of AD-relevant environmental neurotoxicity remain poorly understood. Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) is a potentially critical pathogenic target of environmentally induced neurotoxicity. BACE1 clearly has a critical role in AD pathophysiology: It is required for amyloid beta production and expression and activity of BACE1 are increased in the AD brain. Though the literature on BACE1 in response to environmental insults is limited, current studies, along with extensive AD neurobiology literature suggest that BACE1 deserves attention as an important neurotoxic target. Here, we critically review research on environmental neurotoxicants such as metals, pesticides, herbicides, fungicides, polyfluoroalkyl substances, heterocyclic aromatic amines, advanced glycation end products, and acrolein that modulate BACE1 and potential mechanisms of action. Though more research is needed to clearly understand whether BACE1 is a critical mediator of AD-relevant neurotoxicity, available reports provide convincing evidence that BACE1 is altered by environmental risk factors associated with AD pathology, implying that BACE1 inhibition and its use as a biomarker should be considered in AD management and research.
Collapse
Affiliation(s)
- Tauqeerunnisa Syeda
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA.,Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, Indiana, USA
| | - Jason R Cannon
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA.,Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
65
|
Hassan SA, Kadry MO. Neurodegenerative and Hepatorenal Disorders Induced Via Aluminum Chloride in Murine System: Impact of β-Secretase, MAPK, and KIM. Biol Trace Elem Res 2021; 199:227-236. [PMID: 32279189 DOI: 10.1007/s12011-020-02132-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/23/2020] [Indexed: 12/11/2022]
Abstract
Aluminum chloride (AlCl3) is commonly used in daily life; meanwhile, it is the potential etiology of various neurodegenerative as well as hepatorenal diseases. Therefore, the present study was carried out to investigate the correlation between AlCl3-induced biochemical alterations and the toxicity induced in various organs such as the brain, liver, and kidney. Male mice received AlCl3 in an oral dose of 50 mg kg-1 in addition to (50 mg) in drinking water for 2 weeks. Two weeks post-AlCl3 intoxication, the brain, liver, and kidney biochemical indices were assessed via molecular and western blot analysis. The results are as follows: AlCl3 intoxication induced a significant elevation in serum malondialdehyde in addition to a significant reduction in serum glutathione (GSH) and superoxide dismutase (SOD) levels. Brain β-secretase (tubulin-binding protein) and tau proteins which are responsible for the synthesis of β-amyloid protein that may interfere with neuronal communication in Alzheimer's disease (AD) were also upregulated; regarding hepatic function, AlCl3 elevated serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activities. Moreover, it upregulated hepatic mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) protein expressions as well as renal kidney-inducible molecule-1 (KIM-1) which indicated the deleterious effect of AlCl3 on these organs. These results were confirmed by histopathological investigations. So, we hypothesize that acute AlCl3 administration is responsible for oxidative cell damage that interferes with brain function inducing β-amyloid accumulation, Alzheimer's disease, and neurodegenerative damage as well as hepatorenal injuries.
Collapse
Affiliation(s)
- Sohair Aly Hassan
- Therapeutic Chemistry Department, National Research Center, Al Bouhooth Street, Dokki, Giza, Egypt
| | - Mai O Kadry
- Therapeutic Chemistry Department, National Research Center, Al Bouhooth Street, Dokki, Giza, Egypt.
| |
Collapse
|
66
|
[The future of dementia prevention and treatment strategies]. Nihon Ronen Igakkai Zasshi 2020; 57:374-396. [PMID: 33268621 DOI: 10.3143/geriatrics.57.374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
67
|
Chronic BACE-1 Inhibitor Administration in TASTPM Mice (APP KM670/671NL and PSEN1 M146V Mutation): An EEG Study. Int J Mol Sci 2020; 21:ijms21239072. [PMID: 33260655 PMCID: PMC7730584 DOI: 10.3390/ijms21239072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/16/2020] [Accepted: 11/24/2020] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE In this exploratory study, we tested whether electroencephalographic (EEG) rhythms may reflect the effects of a chronic administration (4 weeks) of an anti-amyloid β-site amyloid precursor protein (APP) cleaving enzyme 1 inhibitor (BACE-1; ER-901356; Eisai Co., Ltd., Tokyo, Japan) in TASTPM (double mutation in APP KM670/671NL and PSEN1 M146V) producing Alzheimer's disease (AD) amyloid neuropathology as compared to wild type (WT) mice. METHODS Ongoing EEG rhythms were recorded from a bipolar frontoparietal and two monopolar frontomedial (prelimbic) and hippocampal channels in 11 WT Vehicle, 10 WT BACE-1, 10 TASTPM Vehicle, and 11 TASTPM BACE-1 mice (males; aged 8/9 months old at the beginning of treatment). Normalized EEG power (density) was compared between the first day (Day 0) and after 4 weeks (Week 4) of the BACE-1 inhibitor (10 mg/Kg) or vehicle administration in the 4 mouse groups. Frequency and magnitude of individual EEG delta and theta frequency peaks (IDF and ITF) were considered during animal conditions of behaviorally passive and active wakefulness. Cognitive status was not tested. RESULTS Compared with the WT group, the TASTPM group generally showed a significantly lower reactivity in frontoparietal ITF power during the active over the passive condition (p < 0.05). Notably, there was no other statistically significant effect (e.g., additional electrodes, recording time, and BACE-1 inhibitor). CONCLUSIONS The above EEG biomarkers reflected differences between the WT and TASTPM groups, but no BACE-1 inhibitor effect. The results suggest an enhanced experimental design with the use of younger mice, longer drug administrations, an effective control drug, and neuropathological amyloid markers.
Collapse
|
68
|
Muppirala AN, Limbach LE, Bradford EF, Petersen SC. Schwann cell development: From neural crest to myelin sheath. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e398. [PMID: 33145925 DOI: 10.1002/wdev.398] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022]
Abstract
Vertebrate nervous system function requires glial cells, including myelinating glia that insulate axons and provide trophic support that allows for efficient signal propagation by neurons. In vertebrate peripheral nervous systems, neural crest-derived glial cells known as Schwann cells (SCs) generate myelin by encompassing and iteratively wrapping membrane around single axon segments. SC gliogenesis and neurogenesis are intimately linked and governed by a complex molecular environment that shapes their developmental trajectory. Changes in this external milieu drive developing SCs through a series of distinct morphological and transcriptional stages from the neural crest to a variety of glial derivatives, including the myelinating sublineage. Cues originate from the extracellular matrix, adjacent axons, and the developing SC basal lamina to trigger intracellular signaling cascades and gene expression changes that specify stages and transitions in SC development. Here, we integrate the findings from in vitro neuron-glia co-culture experiments with in vivo studies investigating SC development, particularly in zebrafish and mouse, to highlight critical factors that specify SC fate. Ultimately, we connect classic biochemical and mutant studies with modern genetic and visualization tools that have elucidated the dynamics of SC development. This article is categorized under: Signaling Pathways > Cell Fate Signaling Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Anoohya N Muppirala
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neuroscience, Kenyon College, Gambier, Ohio, USA
| | | | | | - Sarah C Petersen
- Department of Neuroscience, Kenyon College, Gambier, Ohio, USA.,Department of Biology, Kenyon College, Gambier, Ohio, USA
| |
Collapse
|
69
|
Sun BL, Chen Y, Fan DY, Zhu C, Zeng F, Wang YJ. Critical thinking on amyloid-beta-targeted therapy: challenges and perspectives. SCIENCE CHINA-LIFE SCIENCES 2020; 64:926-937. [DOI: 10.1007/s11427-020-1810-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/28/2020] [Indexed: 01/02/2023]
|
70
|
Bukke VN, Villani R, Archana M, Wawrzyniak A, Balawender K, Orkisz S, Ferraro L, Serviddio G, Cassano T. The Glucose Metabolic Pathway as A Potential Target for Therapeutics: Crucial Role of Glycosylation in Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21207739. [PMID: 33086751 PMCID: PMC7589651 DOI: 10.3390/ijms21207739] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/12/2020] [Accepted: 10/15/2020] [Indexed: 01/17/2023] Open
Abstract
Glucose uptake in the brain decreases because of normal aging but this decline is accelerated in Alzheimer’s disease (AD) patients. In fact, positron emission tomography (PET) studies have shown that metabolic reductions in AD patients occur decades before the onset of symptoms, suggesting that metabolic deficits may be an upstream event in at least some late-onset cases. A decrease in availability of glucose content induces a considerable impairment/downregulation of glycosylation, which is an important post-translational modification. Glycosylation is an important and highly regulated mechanism of secondary protein processing within cells and it plays a crucial role in modulating stability of proteins, as carbohydrates are important in achieving the proper three-dimensional conformation of glycoproteins. Moreover, glycosylation acts as a metabolic sensor that links glucose metabolism to normal neuronal functioning. All the proteins involved in β-amyloid (Aβ) precursor protein metabolism have been identified as candidates of glycosylation highlighting the possibility that Aβ metabolism could be regulated by their glycosylation. Within this framework, the present review aims to summarize the current understanding on the role of glycosylation in the etiopathology of AD, emphasizing the idea that glucose metabolic pathway may represent an alternative therapeutic option for targeting AD. From this perspective, the pharmacological modulation of glycosylation levels may represent a ‘sweet approach’ to treat AD targeting new mechanisms independent of the amyloid cascade and with comparable impacts in familial and sporadic AD.
Collapse
Affiliation(s)
- Vidyasagar Naik Bukke
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Rosanna Villani
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (R.V.); (M.A.); (G.S.)
| | - Moola Archana
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (R.V.); (M.A.); (G.S.)
| | - Agata Wawrzyniak
- Morphological Science Department of Human Anatomy, Medical Faculty University of Rzeszów, 35-036 Rzeszów, Poland; (A.W.); (K.B.); (S.O.)
| | - Krzysztof Balawender
- Morphological Science Department of Human Anatomy, Medical Faculty University of Rzeszów, 35-036 Rzeszów, Poland; (A.W.); (K.B.); (S.O.)
| | - Stanislaw Orkisz
- Morphological Science Department of Human Anatomy, Medical Faculty University of Rzeszów, 35-036 Rzeszów, Poland; (A.W.); (K.B.); (S.O.)
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology, University of Ferrara, 44100 Ferrara, Italy;
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (R.V.); (M.A.); (G.S.)
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
- Correspondence:
| |
Collapse
|
71
|
Fissel JA, Farah MH. Macrophage-specific deletion of BACE1 does not enhance macrophage recruitment to the injured peripheral nerve. J Neuroimmunol 2020; 349:577423. [PMID: 33074142 DOI: 10.1016/j.jneuroim.2020.577423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/02/2020] [Accepted: 10/03/2020] [Indexed: 02/06/2023]
Abstract
Following peripheral nerve injury, macrophages are recruited to the injury site from circulation to clear cellular debris. Injured β-secretase 1 (BACE1) knockout mice have enhanced macrophage recruitment and debris clearance, which may be due to BACE1 activity in macrophages or the hypomyelination observed in BACE1 knockout mice. To assess if BACE1 expression by macrophages mediates enhanced macrophage recruitment we utilized mice with macrophage specific deletion of BACE1 and saw no increase in macrophage recruitment following injury. This study suggests that expression of BACE1 by macrophages may not be essential for increased recruitment observed previously in global BACE1 KO mice.
Collapse
Affiliation(s)
- John A Fissel
- Department of Neurology at Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohamed H Farah
- Department of Neurology at Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
72
|
Kent SA, Spires-Jones TL, Durrant CS. The physiological roles of tau and Aβ: implications for Alzheimer's disease pathology and therapeutics. Acta Neuropathol 2020; 140:417-447. [PMID: 32728795 PMCID: PMC7498448 DOI: 10.1007/s00401-020-02196-w] [Citation(s) in RCA: 265] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 01/18/2023]
Abstract
Tau and amyloid beta (Aβ) are the prime suspects for driving pathology in Alzheimer's disease (AD) and, as such, have become the focus of therapeutic development. Recent research, however, shows that these proteins have been highly conserved throughout evolution and may have crucial, physiological roles. Such functions may be lost during AD progression or be unintentionally disrupted by tau- or Aβ-targeting therapies. Tau has been revealed to be more than a simple stabiliser of microtubules, reported to play a role in a range of biological processes including myelination, glucose metabolism, axonal transport, microtubule dynamics, iron homeostasis, neurogenesis, motor function, learning and memory, neuronal excitability, and DNA protection. Aβ is similarly multifunctional, and is proposed to regulate learning and memory, angiogenesis, neurogenesis, repair leaks in the blood-brain barrier, promote recovery from injury, and act as an antimicrobial peptide and tumour suppressor. This review will discuss potential physiological roles of tau and Aβ, highlighting how changes to these functions may contribute to pathology, as well as the implications for therapeutic development. We propose that a balanced consideration of both the physiological and pathological roles of tau and Aβ will be essential for the design of safe and effective therapeutics.
Collapse
Affiliation(s)
- Sarah A. Kent
- Translational Neuroscience PhD Programme, Centre for Discovery Brain Sciences and the UK Dementia Research Institute, The University of Edinburgh, 1 George Square, Edinburgh, EH8 9JZ Scotland, UK
| | - Tara L. Spires-Jones
- Centre for Discovery Brain Sciences and the UK Dementia Research Institute, The University of Edinburgh, 1 George Square, Edinburgh, EH8 9JZ Scotland, UK
| | - Claire S. Durrant
- Centre for Discovery Brain Sciences and the UK Dementia Research Institute, The University of Edinburgh, 1 George Square, Edinburgh, EH8 9JZ Scotland, UK
| |
Collapse
|
73
|
Nocera G, Jacob C. Mechanisms of Schwann cell plasticity involved in peripheral nerve repair after injury. Cell Mol Life Sci 2020; 77:3977-3989. [PMID: 32277262 PMCID: PMC7532964 DOI: 10.1007/s00018-020-03516-9] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 03/09/2020] [Accepted: 03/30/2020] [Indexed: 01/01/2023]
Abstract
The great plasticity of Schwann cells (SCs), the myelinating glia of the peripheral nervous system (PNS), is a critical feature in the context of peripheral nerve regeneration following traumatic injuries and peripheral neuropathies. After a nerve damage, SCs are rapidly activated by injury-induced signals and respond by entering the repair program. During the repair program, SCs undergo dynamic cell reprogramming and morphogenic changes aimed at promoting nerve regeneration and functional recovery. SCs convert into a repair phenotype, activate negative regulators of myelination and demyelinate the damaged nerve. Moreover, they express many genes typical of their immature state as well as numerous de-novo genes. These genes modulate and drive the regeneration process by promoting neuronal survival, damaged axon disintegration, myelin clearance, axonal regrowth and guidance to their former target, and by finally remyelinating the regenerated axon. Many signaling pathways, transcriptional regulators and epigenetic mechanisms regulate these events. In this review, we discuss the main steps of the repair program with a particular focus on the molecular mechanisms that regulate SC plasticity following peripheral nerve injury.
Collapse
Affiliation(s)
- Gianluigi Nocera
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Mainz, Germany
| | - Claire Jacob
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
74
|
Dcf1 alleviates C99-mediated deficits in drosophila by reducing the cleavage of C99. Biochem Biophys Res Commun 2020; 530:410-417. [DOI: 10.1016/j.bbrc.2020.05.063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/07/2020] [Indexed: 12/23/2022]
|
75
|
Lozupone M, Solfrizzi V, D'Urso F, Di Gioia I, Sardone R, Dibello V, Stallone R, Liguori A, Ciritella C, Daniele A, Bellomo A, Seripa D, Panza F. Anti-amyloid-β protein agents for the treatment of Alzheimer's disease: an update on emerging drugs. Expert Opin Emerg Drugs 2020; 25:319-335. [PMID: 32772738 DOI: 10.1080/14728214.2020.1808621] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Currently available Alzheimer's disease (AD) therapeutics are only symptomatic, targeting cholinergic and glutamatergic neurotransmissions. Several putative disease-modifying drugs in late-stage clinical development target amyloid-β (Aβ) peptide and tau protein, the principal neurophatological hallmarks of the disease. AREAS COVERED Phase III randomized clinical trials of anti-Aβ drugs for AD treatment were searched in US and EU clinical trial registries and principal biomedical databases until May 2020. EXPERT OPINION At present, compounds in Phase III clinical development for AD include four anti-Ab monoclonal antibodies (solanezumab, gantenerumab, aducanumab, BAN2401), the combination of cromolyn sodium and ibuprofen (ALZT-OP1), and two small molecules (levetiracetam, GV-971). These drugs are mainly being tested in subjects during early AD phases or at preclinical stage of familial AD or even in asymptomatic subjects at high risk of developing AD. The actual results support the hypothesis that elevated Aβ represents an early stage in the AD continuum and demonstrate the feasibility of enrolling these high-risk participants in secondary prevention trials to slow cognitive decline during the AD preclinical stages. However, a series of clinical failures may question further development of Aβ-targeting drugs and the findings from current ongoing Phase III trials will hopefully give light to this critical issue.
Collapse
Affiliation(s)
- Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro , Bari, Italy
| | - Vincenzo Solfrizzi
- "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro" , Bari, Italy
| | - Francesca D'Urso
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia , Foggia, Italy
| | - Ilaria Di Gioia
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia , Foggia, Italy
| | - Rodolfo Sardone
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| | - Vittorio Dibello
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy.,Department of Orofacial Pain and Dysfunction, Academic Centre of Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , The Netherlands
| | - Roberta Stallone
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| | - Angelo Liguori
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| | - Chiara Ciritella
- Physical and Rehabilitation Medicine Department, University of Foggia , Foggia, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart , Rome, Italy.,Institute of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia , Foggia, Italy
| | - Davide Seripa
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo Della Sofferenza , Foggia, Italy.,Hematology and Stem Cell Transplant Unit, Vito Fazzi Hospital, ASL Lecce , Lecce, Italy
| | - Francesco Panza
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| |
Collapse
|
76
|
Baloh RH. The Nerve to Give BACE Inhibitors a Second Chance? Neurotherapeutics 2020; 17:966-967. [PMID: 32514890 PMCID: PMC7609629 DOI: 10.1007/s13311-020-00876-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Robert H Baloh
- Department of Neurology, Center for Neural Science and Medicine, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA.
| |
Collapse
|
77
|
Zhang Z, Li Y, He F, Cui Y, Zheng Y, Li R. Sex differences in circulating neuregulin1-β1 and β-secretase 1 expression in childhood-onset schizophrenia. Compr Psychiatry 2020; 100:152176. [PMID: 32430144 DOI: 10.1016/j.comppsych.2020.152176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE Early-onset schizophrenia is a severe and rare form of schizophrenia that is clinically and neurobiologically continuous with the adult form of schizophrenia. Neuregulin1 (NRG1)-mediated signaling is crucial for early neurodevelopment, which exerts its function by limited β-secretase 1 (BACE1) proteolysis processing. However, circulating neuregulin1-β1 (NRG1-β1), an isoform of NRG1, and its cleavage enzyme BACE1 have not been studied in early-onset patients with schizophrenia. METHODS In this study, we collected plasma and clinical information from 71 young patients (7 ≤ age years ≤20) with schizophrenia and 53 age- and sex-matched healthy controls. Immunoassay was used to test levels of circulating NRG1-β1 and BACE1 expression. We further analyzed the relationship of disease-onset age and gender with NRG1-β1 and BACE1 levels. RESULTS We found that circulating plasma levels of NRG1-β1 were significantly decreased in young patients with early-onset schizophrenia. In males with childhood onset schizophrenia (COS), NRG1-β1 was reduced and was inversely correlated with positive symptom of PANSS; moreover, these male patients with higher plasma BACE1 levels showed more severe general symptoms of PANSS and defective social functioning; whereas, no aforementioned results were found in adolescent-onset schizophrenia (AOS). Notably, young female patients with COS and AOS had no significant change in NRG1-β1 and BACE1, which demonstrated a sex-dependent effect in early-onset schizophrenia. CONCLUSION Our results suggest that decreased levels of NRG1-β1 and its cleavage enzyme BACE1 contribute to increased risk of etiology of schizophrenia. Synthetic biomarkers may have clinical applications for the early diagnosis of male COS.
Collapse
Affiliation(s)
- Zhengrong Zhang
- National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Yuhong Li
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Fan He
- National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Yonghua Cui
- National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Yi Zheng
- National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China.
| | - Rena Li
- National Clinical Research Center for Mental Disorders, Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing 100088, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
78
|
Tallon C, Marshall KL, Kennedy ME, Hyde LA, Farah MH. Pharmacological BACE Inhibition Improves Axonal Regeneration in Nerve Injury and Disease Models. Neurotherapeutics 2020; 17:973-988. [PMID: 32236823 PMCID: PMC7609814 DOI: 10.1007/s13311-020-00852-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
While the peripheral nervous system is able to repair itself following injury and disease, recovery is often slow and incomplete, with no available treatments to enhance the effectiveness of regeneration. Using knock-out and transgenic overexpressor mice, we previously reported that BACE1, an aspartyl protease, as reported by Hemming et al. (PLoS One 4:12, 2009), negatively regulates peripheral nerve regeneration. Here, we investigated whether pharmacological inhibition of BACE may enhance peripheral nerve repair following traumatic nerve injury or neurodegenerative disease. BACE inhibitor-treated mice had increased numbers of regenerating axons and enhanced functional recovery after a sciatic nerve crush while inhibition increased axonal sprouting following a partial nerve injury. In the SOD1G93A ALS mouse model, BACE inhibition increased axonal regeneration with improved muscle re-innervation. CHL1, a BACE1 substrate, was elevated in treated mice and may mediate enhanced regeneration. Our data demonstrates that pharmacological BACE inhibition accelerates peripheral axon regeneration after varied nerve injuries and could be used as a potential therapy.
Collapse
Affiliation(s)
- Carolyn Tallon
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, The John G. Rangos Sr. Building, Room 239, 855 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Katherine L Marshall
- Department of Neurology, Neuromuscular Division, 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, Neuromuscular Division, Johns Hopkins University School of Medicine, The John G. Rangos Sr. Building, Room 239, 855 N. Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
79
|
Li C, Zhao B, Fan YN, Jia X, Liu Y. Expression of BACE1 in the Rat Carotid Body. Front Physiol 2020; 11:505. [PMID: 32508682 PMCID: PMC7251150 DOI: 10.3389/fphys.2020.00505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/24/2020] [Indexed: 11/13/2022] Open
Abstract
This study explored the expression of BACE1 (β-amyloid precursor protein cleaving enzyme 1) in the rat carotid body and the effect of CIH (cyclic intermittent hypoxia) on the expression of BACE1. We found that BACE1 was expressed in the rat carotid body and located in the nerve endings and type II cells but not in type I cells. CIH reduced BACE1 level in the carotid body, and reoxygenation or ROS scavenger alleviated this reduction. Furthermore, we found that CIH augmented the mRNA level of PGC-1α but attenuated the mRNA level of BACE1 in the carotid body. Taken together, our results suggest that CIH promotes the production of ROS that upregulates the level of PGC-1α, which may in turn inhibits the transcription of BACE1, and that a reduction in the BACE1 level may be related to CIH-induced reversible and ROS-dependent carotid body plasticity. Our study provides a new candidate molecule for further study of the mechanism of carotid body plasticity.
Collapse
Affiliation(s)
- Chaohong Li
- Henan Key Laboratory of Neural Regeneration and Repairment, Henan Neurology Institute, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Baosheng Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Ya-Nan Fan
- Henan Key Laboratory of Neural Regeneration and Repairment, Henan Neurology Institute, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xianglei Jia
- Henan Key Laboratory of Neural Regeneration and Repairment, Henan Neurology Institute, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yuzhen Liu
- Henan Key Laboratory of Neural Regeneration and Repairment, Henan Neurology Institute, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
80
|
Bera S, Camblor‐Perujo S, Calleja Barca E, Negrete‐Hurtado A, Racho J, De Bruyckere E, Wittich C, Ellrich N, Martins S, Adjaye J, Kononenko NL. AP-2 reduces amyloidogenesis by promoting BACE1 trafficking and degradation in neurons. EMBO Rep 2020; 21:e47954. [PMID: 32323475 PMCID: PMC7271323 DOI: 10.15252/embr.201947954] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/13/2022] Open
Abstract
Cleavage of amyloid precursor protein (APP) by BACE-1 (β-site APP cleaving enzyme 1) is the rate-limiting step in amyloid-β (Aβ) production and a neuropathological hallmark of Alzheimer's disease (AD). Despite decades of research, mechanisms of amyloidogenic APP processing remain highly controversial. Here, we show that in neurons, APP processing and Aβ production are controlled by the protein complex-2 (AP-2), an endocytic adaptor known to be required for APP endocytosis. Now, we find that AP-2 prevents amyloidogenesis by additionally functioning downstream of BACE1 endocytosis, regulating BACE1 endosomal trafficking and its delivery to lysosomes. AP-2 is decreased in iPSC-derived neurons from patients with late-onset AD, while conditional AP-2 knockout (KO) mice exhibit increased Aβ production, resulting from accumulation of BACE1 within late endosomes and autophagosomes. Deletion of BACE1 decreases amyloidogenesis and mitigates synapse loss in neurons lacking AP-2. Taken together, these data suggest a mechanism for BACE1 intracellular trafficking and degradation via an endocytosis-independent function of AP-2 and reveal a novel role for endocytic proteins in AD.
Collapse
Affiliation(s)
- Sujoy Bera
- CECAD Research CenterUniversity of CologneCologneGermany
- Present address:
Centre for Neuroscience and Regenerative MedicineFaculty of ScienceUniversity of Technology SydneySydneyNSWAustralia
| | | | | | | | - Julia Racho
- CECAD Research CenterUniversity of CologneCologneGermany
| | | | | | - Nina Ellrich
- CECAD Research CenterUniversity of CologneCologneGermany
| | - Soraia Martins
- Institute for Stem Cell Research and Regenerative MedicineMedical FacultyHeinrich Heine UniversityDüsseldorfGermany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative MedicineMedical FacultyHeinrich Heine UniversityDüsseldorfGermany
| | | |
Collapse
|
81
|
SPON1 Can Reduce Amyloid Beta and Reverse Cognitive Impairment and Memory Dysfunction in Alzheimer's Disease Mouse Model. Cells 2020; 9:cells9051275. [PMID: 32455709 PMCID: PMC7290723 DOI: 10.3390/cells9051275] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/08/2020] [Accepted: 05/18/2020] [Indexed: 01/07/2023] Open
Abstract
Alzheimer’s disease (AD) is a complex, age-related neurodegenerative disease that is the most common form of dementia. However, the cure for AD has not yet been founded. The accumulation of amyloid beta (Aβ) is considered to be a hallmark of AD. Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), also known as beta secretase is the initiating enzyme in the amyloidogenic pathway. Blocking BACE1 could reduce the amount of Aβ, but this would also prohibit the other functions of BACE1 in brain physiological activity. SPONDIN1 (SPON1) is known to bind to the BACE1 binding site of the amyloid precursor protein (APP) and blocks the initiating amyloidogenesis. Here, we show the effect of SPON1 in Aβ reduction in vitro in neural cells and in an in vivo AD mouse model. We engineered mouse induced neural stem cells (iNSCs) to express Spon1. iNSCs harboring mouse Spon1 secreted SPON1 protein and reduced the quantity of Aβ when co-cultured with Aβ-secreting Neuro 2a cells. The human SPON1 gene itself also reduced Aβ in HEK 293T cells expressing the human APP transgene with AD-linked mutations through lentiviral-mediated delivery. We also demonstrated that injecting SPON1 reduced the amount of Aβ and ameliorated cognitive dysfunction and memory impairment in 5xFAD mice expressing human APP and PSEN1 transgenes with five AD-linked mutations.
Collapse
|
82
|
Scapin C, Ferri C, Pettinato E, Zambroni D, Bianchi F, Del Carro U, Belin S, Caruso D, Mitro N, Pellegatta M, Taveggia C, Schwab MH, Nave KA, Feltri ML, Wrabetz L, D'Antonio M. Enhanced axonal neuregulin-1 type-III signaling ameliorates neurophysiology and hypomyelination in a Charcot-Marie-Tooth type 1B mouse model. Hum Mol Genet 2020; 28:992-1006. [PMID: 30481294 DOI: 10.1093/hmg/ddy411] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/30/2018] [Accepted: 11/22/2018] [Indexed: 12/11/2022] Open
Abstract
Charcot-Marie-Tooth (CMT) neuropathies are a group of genetic disorders that affect the peripheral nervous system with heterogeneous pathogenesis and no available treatment. Axonal neuregulin 1 type III (Nrg1TIII) drives peripheral nerve myelination by activating downstream signaling pathways such as PI3K/Akt and MAPK/Erk that converge on master transcriptional regulators of myelin genes, such as Krox20. We reasoned that modulating Nrg1TIII activity may constitute a general therapeutic strategy to treat CMTs that are characterized by reduced levels of myelination. Here we show that genetic overexpression of Nrg1TIII ameliorates neurophysiological and morphological parameters in a mouse model of demyelinating CMT1B, without exacerbating the toxic gain-of-function that underlies the neuropathy. Intriguingly, the mechanism appears not to be related to Krox20 or myelin gene upregulation, but rather to a beneficial rebalancing in the stoichiometry of myelin lipids and proteins. Finally, we provide proof of principle that stimulating Nrg1TIII signaling, by pharmacological suppression of the Nrg1TIII inhibitor tumor necrosis factor-alpha-converting enzyme (TACE/ADAM17), also ameliorates the neuropathy. Thus, modulation of Nrg1TIII by TACE/ADAM17 inhibition may represent a general treatment for hypomyelinating neuropathies.
Collapse
Affiliation(s)
| | | | | | | | - Francesca Bianchi
- INSPE, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Ubaldo Del Carro
- INSPE, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | | | - Donatella Caruso
- DiSFeB-Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nico Mitro
- DiSFeB-Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Marta Pellegatta
- INSPE, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Carla Taveggia
- INSPE, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Markus H Schwab
- Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany.,Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Klaus-Armin Nave
- Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
| | - M Laura Feltri
- DIBIT, Divisions of Genetics and Cell Biology.,Hunter James Kelly Research Institute.,Department of Neurology.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Lawrence Wrabetz
- DIBIT, Divisions of Genetics and Cell Biology.,Hunter James Kelly Research Institute.,Department of Neurology.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | | |
Collapse
|
83
|
Iraji A, Khoshneviszadeh M, Firuzi O, Khoshneviszadeh M, Edraki N. Novel small molecule therapeutic agents for Alzheimer disease: Focusing on BACE1 and multi-target directed ligands. Bioorg Chem 2020; 97:103649. [PMID: 32101780 DOI: 10.1016/j.bioorg.2020.103649] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/05/2020] [Accepted: 02/03/2020] [Indexed: 12/17/2022]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder that effects 50 million people worldwide. In this review, AD pathology and the development of novel therapeutic agents targeting AD were fully discussed. In particular, common approaches to prevent Aβ production and/or accumulation in the brain including α-secretase activators, specific γ-secretase modulators and small molecules BACE1 inhibitors were reviewed. Additionally, natural-origin bioactive compounds that provide AD therapeutic advances have been introduced. Considering AD is a multifactorial disease, the therapeutic potential of diverse multi target-directed ligands (MTDLs) that combine the efficacy of cholinesterase (ChE) inhibitors, MAO (monoamine oxidase) inhibitors, BACE1 inhibitors, phosphodiesterase 4D (PDE4D) inhibitors, for the treatment of AD are also reviewed. This article also highlights descriptions on the regulator of serotonin receptor (5-HT), metal chelators, anti-aggregants, antioxidants and neuroprotective agents targeting AD. Finally, current computational methods for evaluating the structure-activity relationships (SAR) and virtual screening (VS) of AD drugs are discussed and evaluated.
Collapse
Affiliation(s)
- Aida Iraji
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsima Khoshneviszadeh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Khoshneviszadeh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medicinal Chemistry, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Edraki
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
84
|
Dutta MS, Basu S. Identifying the key residues instrumental in imparting stability to amyloid beta protofibrils - a comparative study using MD simulations of 17-42 residues. J Biomol Struct Dyn 2020; 39:431-456. [PMID: 31900057 DOI: 10.1080/07391102.2019.1711192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Extracellular plaques, the hallmark of Alzheimer's disease brains, consist of insoluble amyloid fibrils that result from the aggregation of amyloid beta peptides. None of the few therapeutic options currently adopted, address the cause of the disease. Instead, they reduce symptom of the disease. Inhibition of aggregation or destabilization of aggregates therefore, emerges as a preferable therapeutic approach. Designing inhibitors or destabilizers demands comprehensive knowledge of the residues of amyloid beta responsible for the phenomenal structural stability of the aggregate. For the purpose, we have compared the effect on structural destabilization of 13 in silico mutations (single and double) with the wild type counterpart of beta-strand-turn-beta-strand motif of the amyloid beta protofibrils by molecular dynamics simulation. Besides the already known salt bridge interaction between K28 and D23, our analyses expose more significant role of K28 as the only positive charge present in the vicinity. Amongst the two consecutive aromatic residues, F19 is involved in stacking interaction; although effect of F20 mutation is more pronounced. Face to face arrangement of A21 and V36 acts as a pillar maintaining the necessary optimum distance between consecutive chains to promote stabilizing interactions. In addition to providing stability to the first beta-strand, large sized negatively charged E22 facilitates salt bridge formation by ensuring fixed relative position of D23 and in turn K28. Likewise, the hydrophobic residues I32 and L34 pack the protofibril core, once again fostering salt bridge interaction. Prospectively, these findings may be compiled for efficient identification or design of scaffolds accountable for protofibril destabilization.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Soumalee Basu
- Department of Microbiology, University of Calcutta, Kolkata, India
| |
Collapse
|
85
|
Abstract
The β-site APP Cleaving enzyme 1 (BACE1) is a membrane-associated aspartyl protease which mediates the production of amyloid-β (Aβ), a major component of amyloid plaques in the Alzheimer’s disease brain. We have synthesised and characterised a series of peptidomimetic analogues of BACE substrates that incorporate two distinct stabilising structures. To demonstrate the potential activity of these compounds, a variety of assaying strategies were used to investigate cleavage susceptibility and inhibition potency under competitive and non-competitive conditions. β-Amino acids and scissile site N-methylation were incorporated into peptide substrate templates as transition state isostere (TSI) substitutes by positional scanning to generate series of non-TSI β-peptidomimetics. The amino acid sequences flanking the β-cleavage site within APP carrying the Swedish double mutation (APPSW), Neuregulin, the synthetic hydroxyethylene-based TSI peptide inhibitor OM99-2, and the high affinity peptide sequence SEISYEVEFR, served as the four substrate templates from which over 60 peptides were designed and synthesised by solid phase peptide synthesis. A quenched fluorescent substrate BACE1 assay in conjunction with liquid chromatography–mass spectrometry (LC-MS) analysis was established to investigate cleavage susceptibility and inhibition potency under competitive and non-competitive conditions. It was determined that β-amino acids substituted at the P1 scissile site position within known peptide substrates were resistant to proteolysis, and particular substitutions induced a concentration-dependent stimulation of BACE1, indicating a possible modulatory role of native BACE1 substrates.
Collapse
|
86
|
Rudan Njavro J, Klotz J, Dislich B, Wanngren J, Shmueli MD, Herber J, Kuhn PH, Kumar R, Koeglsperger T, Conrad M, Wurst W, Feederle R, Vlachos A, Michalakis S, Jedlicka P, Müller SA, Lichtenthaler SF. Mouse brain proteomics establishes MDGA1 and CACHD1 as in vivo substrates of the Alzheimer protease BACE1. FASEB J 2019; 34:2465-2482. [PMID: 31908000 DOI: 10.1096/fj.201902347r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/22/2019] [Accepted: 12/03/2019] [Indexed: 01/18/2023]
Abstract
The protease beta-site APP cleaving enzyme 1 (BACE1) has fundamental functions in the nervous system. Its inhibition is a major therapeutic approach in Alzheimer's disease, because BACE1 cleaves the amyloid precursor protein (APP), thereby catalyzing the first step in the generation of the pathogenic amyloid beta (Aβ) peptide. Yet, BACE1 cleaves numerous additional membrane proteins besides APP. Most of these substrates have been identified in vitro, but only few were further validated or characterized in vivo. To identify BACE1 substrates with in vivo relevance, we used isotope label-based quantitative proteomics of wild type and BACE1-deficient (BACE1 KO) mouse brains. This approach identified known BACE1 substrates, including Close homolog of L1 and contactin-2, which were found to be enriched in the membrane fraction of BACE1 KO brains. VWFA and cache domain-containing protein 1 (CACHD)1 and MAM domain-containing glycosylphosphatidylinositol anchor protein 1 (MDGA1), which have functions in synaptic transmission, were identified and validated as new BACE1 substrates in vivo by immunoblots using primary neurons and mouse brains. Inhibition or deletion of BACE1 from primary neurons resulted in a pronounced inhibition of substrate cleavage and a concomitant increase in full-length protein levels of CACHD1 and MDGA1. The BACE1 cleavage site in both proteins was determined to be located within the juxtamembrane domain. In summary, this study identifies and validates CACHD1 and MDGA1 as novel in vivo substrates for BACE1, suggesting that cleavage of both proteins may contribute to the numerous functions of BACE1 in the nervous system.
Collapse
Affiliation(s)
- Jasenka Rudan Njavro
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jakob Klotz
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Bastian Dislich
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Pathology, University of Bern, Switzerland
| | - Johanna Wanngren
- Division of Neurogeriatrics, Department of NVS, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Merav D Shmueli
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Julia Herber
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Peer-Hendrik Kuhn
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Rohit Kumar
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Neurology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Thomas Koeglsperger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Neurology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Genome Engineering, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Developmental Genetics, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Regina Feederle
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,German Research Center for Environmental Health, Institute for Diabetes and Obesity, Monoclonal Antibody Core Facility, Helmholtz Zentrum München, Neuherberg, Germany.,Core Facility Monoclonal Antibodies, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Germany.,Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Germany
| | - Stylianos Michalakis
- Department of Ophthalmology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Peter Jedlicka
- Faculty of Medicine, ICAR3R - Interdisciplinary Centre for 3Rs in Animal Research, Justus-Liebig-University, Giessen, Germany.,Neuroscience Center, Institute of Clinical Neuroanatomy, Goethe University, Frankfurt am Main, Germany.,Frankfurt Institute for Advanced Studies, Frankfurt am Main, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
87
|
Lombardo S, Chiacchiaretta M, Tarr A, Kim W, Cao T, Sigal G, Rosahl TW, Xia W, Haydon PG, Kennedy ME, Tesco G. BACE1 partial deletion induces synaptic plasticity deficit in adult mice. Sci Rep 2019; 9:19877. [PMID: 31882662 PMCID: PMC6934620 DOI: 10.1038/s41598-019-56329-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022] Open
Abstract
BACE1 is the first enzyme involved in APP processing, thus it is a strong therapeutic target candidate for Alzheimer's disease. The observation of deleterious phenotypes in BACE1 Knock-out (KO) mouse models (germline and conditional) raised some concerns on the safety and tolerability of BACE1 inhibition. Here, we have employed a tamoxifen inducible BACE1 conditional Knock-out (cKO) mouse model to achieve a controlled partial depletion of BACE1 in adult mice. Biochemical and behavioural characterization was performed at two time points: 4-5 months (young mice) and 12-13 months (aged mice). A ~50% to ~70% BACE1 protein reduction in hippocampus and cortex, respectively, induced a significant reduction of BACE1 substrates processing and decrease of Aβx-40 levels at both ages. Hippocampal axonal guidance and peripheral nerve myelination were not affected. Aged mice displayed a CA1 long-term potentiation (LTP) deficit that was not associated with memory impairment. Our findings indicate that numerous phenotypes observed in germline BACE1 KO reflect a fundamental role of BACE1 during development while other phenotypes, observed in adult cKO, may be absent when partially rather than completely deleting BACE1. However, we demonstrated that partial depletion of BACE1 still induces CA1 LTP impairment, supporting a role of BACE1 in synaptic plasticity in adulthood.
Collapse
Affiliation(s)
- Sylvia Lombardo
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | - Martina Chiacchiaretta
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | - Andrew Tarr
- Circuits and Behaviour Core, Center for Neuroscience Research, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | - WonHee Kim
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | - Tingyi Cao
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | - Griffin Sigal
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | - Thomas W Rosahl
- External In Vivo Pharmacology, Merck & Co. Inc., Kenilworth, NJ, 07033, USA
| | - Weiming Xia
- Geriatric Research, Education and Clinic Center, Bedford Veterans Affairs Medical Center, Bedford, MA, 01730, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Philip G Haydon
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA
| | | | - Giuseppina Tesco
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA.
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111, USA.
| |
Collapse
|
88
|
Fledrich R, Kungl T, Nave KA, Stassart RM. Axo-glial interdependence in peripheral nerve development. Development 2019; 146:146/21/dev151704. [PMID: 31719044 DOI: 10.1242/dev.151704] [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] [Indexed: 01/01/2023]
Abstract
During the development of the peripheral nervous system, axons and myelinating Schwann cells form a unique symbiotic unit, which is realized by a finely tuned network of molecular signals and reciprocal interactions. The importance of this complex interplay becomes evident after injury or in diseases in which aspects of axo-glial interaction are perturbed. This Review focuses on the specific interdependence of axons and Schwann cells in peripheral nerve development that enables axonal outgrowth, Schwann cell lineage progression, radial sorting and, finally, formation and maintenance of the myelin sheath.
Collapse
Affiliation(s)
- Robert Fledrich
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany .,Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Theresa Kungl
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany.,Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Ruth M Stassart
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany .,Department of Neuropathology, University Clinic Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
89
|
Performance evaluation of molecular docking and free energy calculations protocols using the D3R Grand Challenge 4 dataset. J Comput Aided Mol Des 2019; 33:1031-1043. [PMID: 31677003 DOI: 10.1007/s10822-019-00232-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 10/09/2019] [Indexed: 12/11/2022]
Abstract
Using the D3R Grand Challenge 4 dataset containing Beta-secretase 1 (BACE) and Cathepsin S (CatS) inhibitors, we have evaluated the performance of our in-house docking workflow that involves in the first step the selection of the most suitable docking software for the system of interest based on structural and functional information available in public databases, followed by the docking of the dataset to predict the binding modes and ranking of ligands. The macrocyclic nature of the BACE ligands brought additional challenges, which were dealt with by a careful preparation of the three-dimensional input structures for ligands. This provided top-performing predictions for BACE, in contrast with CatS, where the predictions in the absence of guiding constraints provided poor results. These results highlight the importance of previous structural knowledge that is needed for correct predictions on some challenging targets. After the end of the challenge, we also carried out free energy calculations (i.e. in a non-blinded manner) for CatS using the pmx software and several force fields (AMBER, Charmm). Using knowledge-based starting pose construction allowed reaching remarkable accuracy for the CatS free energy estimates. Interestingly, we show that the use of a consensus result, by averaging the results from different force fields, increases the prediction accuracy.
Collapse
|
90
|
Long JM, Holtzman DM. Alzheimer Disease: An Update on Pathobiology and Treatment Strategies. Cell 2019; 179:312-339. [PMID: 31564456 PMCID: PMC6778042 DOI: 10.1016/j.cell.2019.09.001] [Citation(s) in RCA: 1886] [Impact Index Per Article: 314.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022]
Abstract
Alzheimer disease (AD) is a heterogeneous disease with a complex pathobiology. The presence of extracellular β-amyloid deposition as neuritic plaques and intracellular accumulation of hyperphosphorylated tau as neurofibrillary tangles remains the primary neuropathologic criteria for AD diagnosis. However, a number of recent fundamental discoveries highlight important pathological roles for other critical cellular and molecular processes. Despite this, no disease-modifying treatment currently exists, and numerous phase 3 clinical trials have failed to demonstrate benefits. Here, we review recent advances in our understanding of AD pathobiology and discuss current treatment strategies, highlighting recent clinical trials and opportunities for developing future disease-modifying therapies.
Collapse
Affiliation(s)
- Justin M Long
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
91
|
Javier-Torrent M, Marco S, Rocandio D, Pons-Vizcarra M, Janes PW, Lackmann M, Egea J, Saura CA. Presenilin/γ-secretase-dependent EphA3 processing mediates axon elongation through non-muscle myosin IIA. eLife 2019; 8:43646. [PMID: 31577226 PMCID: PMC6774734 DOI: 10.7554/elife.43646] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 09/18/2019] [Indexed: 01/08/2023] Open
Abstract
EphA/ephrin signaling regulates axon growth and guidance of neurons, but whether this process occurs also independently of ephrins is unclear. We show that presenilin-1 (PS1)/γ-secretase is required for axon growth in the developing mouse brain. PS1/γ-secretase mediates axon growth by inhibiting RhoA signaling and cleaving EphA3 independently of ligand to generate an intracellular domain (ICD) fragment that reverses axon defects in PS1/γ-secretase- and EphA3-deficient hippocampal neurons. Proteomic analysis revealed that EphA3 ICD binds to non-muscle myosin IIA (NMIIA) and increases its phosphorylation (Ser1943), which promotes NMIIA filament disassembly and cytoskeleton rearrangement. PS1/γ-secretase-deficient neurons show decreased phosphorylated NMIIA and NMIIA/actin colocalization. Moreover, pharmacological NMII inhibition reverses axon retraction in PS-deficient neurons suggesting that NMIIA mediates PS/EphA3-dependent axon elongation. In conclusion, PS/γ-secretase-dependent EphA3 cleavage mediates axon growth by regulating filament assembly through RhoA signaling and NMIIA, suggesting opposite roles of EphA3 on inhibiting (ligand-dependent) and promoting (receptor processing) axon growth in developing neurons.
Collapse
Affiliation(s)
- Míriam Javier-Torrent
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sergi Marco
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Daniel Rocandio
- Institut de Recerca Biomédica de Lleida, Universitat de Lleida, Lleida, Spain
| | - Maria Pons-Vizcarra
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Peter W Janes
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Martin Lackmann
- Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Joaquim Egea
- Institut de Recerca Biomédica de Lleida, Universitat de Lleida, Lleida, Spain
| | - Carlos A Saura
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Facultat de Medicina, Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
92
|
β-Secretase BACE1 Is Required for Normal Cochlear Function. J Neurosci 2019; 39:9013-9027. [PMID: 31527119 DOI: 10.1523/jneurosci.0028-19.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 08/30/2019] [Accepted: 09/04/2019] [Indexed: 12/20/2022] Open
Abstract
Cleavage of amyloid precursor protein (APP) by β-secretase BACE1 initiates the production and accumulation of neurotoxic amyloid-β peptides, which is widely considered an essential pathogenic mechanism in Alzheimer's disease (AD). Here, we report that BACE1 is essential for normal auditory function. Compared with wild-type littermates, BACE1-/- mice of either sex exhibit significant hearing deficits, as indicated by increased thresholds and reduced amplitudes in auditory brainstem responses (ABRs) and decreased distortion product otoacoustic emissions (DPOAEs). Immunohistochemistry revealed aberrant synaptic organization in the cochlea and hypomyelination of auditory nerve fibers as predominant neuropathological substrates of hearing loss in BACE1-/- mice. In particular, we found that fibers of spiral ganglion neurons (SGN) close to the organ of Corti are disorganized and abnormally swollen. BACE1 deficiency also engenders organization defects in the postsynaptic compartment of SGN fibers with ectopic overexpression of PSD95 far outside the synaptic region. During postnatal development, auditory fiber myelination in BACE1-/- mice lags behind dramatically and remains incomplete into adulthood. We relate the marked hypomyelination to the impaired processing of Neuregulin-1 when BACE1 is absent. To determine whether the cochlea of adult wild-type mice is susceptible to AD treatment-like suppression of BACE1, we administered the established BACE1 inhibitor NB-360 for 6 weeks. The drug suppressed BACE1 activity in the brain, but did not impair hearing performance and, upon neuropathological examination, did not produce the characteristic cochlear abnormalities of BACE1-/- mice. Together, these data strongly suggest that the hearing loss of BACE1 knock-out mice represents a developmental phenotype.SIGNIFICANCE STATEMENT Given its crucial role in the pathogenesis of Alzheimer's disease (AD), BACE1 is a prime pharmacological target for AD prevention and therapy. However, the safe and long-term administration of BACE1-inhibitors as envisioned in AD requires a comprehensive understanding of the various physiological functions of BACE1. Here, we report that BACE1 is essential for the processing of auditory signals in the inner ear, as BACE1-deficient mice exhibit significant hearing loss. We relate this deficit to impaired myelination and aberrant synapse formation in the cochlea, which manifest during postnatal development. By contrast, prolonged pharmacological suppression of BACE1 activity in adult wild-type mice did not reproduce the hearing deficit or the cochlear abnormalities of BACE1 null mice.
Collapse
|
93
|
Early Intervention in Alzheimer's Disease: How Early is Early Enough? Neurosci Bull 2019; 36:195-197. [PMID: 31494835 DOI: 10.1007/s12264-019-00429-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022] Open
|
94
|
Saadipour K, Tiberi A, Lombardo S, Grajales E, Montroull L, Mañucat-Tan NB, LaFrancois J, Cammer M, Mathews PM, Scharfman HE, Liao FF, Friedman WJ, Zhou XF, Tesco G, Chao MV. Regulation of BACE1 expression after injury is linked to the p75 neurotrophin receptor. Mol Cell Neurosci 2019; 99:103395. [PMID: 31422108 DOI: 10.1016/j.mcn.2019.103395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/24/2019] [Accepted: 08/08/2019] [Indexed: 12/25/2022] Open
Abstract
BACE1 is a transmembrane aspartic protease that cleaves various substrates and it is required for normal brain function. BACE1 expression is high during early development, but it is reduced in adulthood. Under conditions of stress and injury, BACE1 levels are increased; however, the underlying mechanisms that drive BACE1 elevation are not well understood. One mechanism associated with brain injury is the activation of injurious p75 neurotrophin receptor (p75), which can trigger pathological signals. Here we report that within 72 h after controlled cortical impact (CCI) or laser injury, BACE1 and p75 are increased and tightly co-expressed in cortical neurons of mouse brain. Additionally, BACE1 is not up-regulated in p75 null mice in response to focal cortical injury, while p75 over-expression results in BACE1 augmentation in HEK-293 and SY5Y cell lines. A luciferase assay conducted in SY5Y cell line revealed that BACE1 expression is regulated at the transcriptional level in response to p75 transfection. Interestingly, this effect does not appear to be dependent upon p75 ligands including mature and pro-neurotrophins. In addition, BACE1 activity on amyloid precursor protein (APP) is enhanced in SY5Y-APP cells transfected with a p75 construct. Lastly, we found that the activation of c-jun n-terminal kinase (JNK) by p75 contributes to BACE1 up-regulation. This study explores how two injury-induced molecules are intimately connected and suggests a potential link between p75 signaling and the expression of BACE1 after brain injury.
Collapse
Affiliation(s)
- Khalil Saadipour
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA.
| | - Alexia Tiberi
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA; Bio@SNS Laboratory, Scuola Normale Superiore, Piazza dei Cavalieri 7, Pisa, 56126, Italy
| | - Sylvia Lombardo
- Alzheimer's Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Elena Grajales
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA
| | - Laura Montroull
- Department of Biological Sciences, Rutgers Life Sciences Center, Rutgers University, Newark, NJ 07102, USA
| | - Noralyn B Mañucat-Tan
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, South Australia, 5000, Australia
| | - John LaFrancois
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, Orangeburg, NY 10962, USA
| | - Michael Cammer
- DART Microscopy Laboratory, NYU Langone Medical Center, New York, NY 10016, USA
| | - Paul M Mathews
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, Orangeburg, NY 10962, USA
| | - Helen E Scharfman
- The Nathan Kline Institute of Psychiatric Research, Center for Dementia Research, Orangeburg, NY 10962, USA
| | - Francesca-Fang Liao
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Wilma J Friedman
- Department of Biological Sciences, Rutgers Life Sciences Center, Rutgers University, Newark, NJ 07102, USA
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Adelaide, South Australia, 5000, Australia
| | - Giueseppina Tesco
- Alzheimer's Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Moses V Chao
- Departments of Cell Biology, Physiology & Neuroscience, and Psychiatry, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, New York 10016, USA.
| |
Collapse
|
95
|
Przybyłowska M, Kowalski S, Dzierzbicka K, Inkielewicz-Stepniak I. Therapeutic Potential of Multifunctional Tacrine Analogues. Curr Neuropharmacol 2019; 17:472-490. [PMID: 29651948 PMCID: PMC6520589 DOI: 10.2174/1570159x16666180412091908] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/25/2018] [Accepted: 04/04/2018] [Indexed: 12/12/2022] Open
Abstract
Abstract: Tacrine is a potent inhibitor of cholinesterases (acetylcholinesterase and butyrylcholinesterase) that shows limiting clinical application by liver toxicity. In spite of this, analogues of tacrine are considered as a model inhibitor of cholinesterases in the therapy of Alzheimer’s disease. The interest in these compounds is mainly related to a high variety of their structure and biological properties. In the present review, we have described the role of cholinergic transmission and treatment strategies in Alzheimer’s disease as well as the synthesis and biological activity of several recently developed classes of multifunctional tacrine analogues and hybrids, which consist of a new paradigm to treat Alzheimer’s disease. We have also reported potential of these analogues in the treatment of Alzheimer’s diseases in various experimental systems.
Collapse
Affiliation(s)
- Maja Przybyłowska
- Department of Organic Chemistry, Gdansk University of Technology, 11/12 G. Narutowicza Street, 80-233, Gdansk, Poland
| | - Szymon Kowalski
- Department of Medical Chemistry, Medical University of Gdansk, Debinki 1 Street, 80-211 Gdansk, Poland
| | - Krystyna Dzierzbicka
- Department of Organic Chemistry, Gdansk University of Technology, 11/12 G. Narutowicza Street, 80-233, Gdansk, Poland
| | | |
Collapse
|
96
|
Vullhorst D, Buonanno A. NMDA Receptors Regulate Neuregulin 2 Binding to ER-PM Junctions and Ectodomain Release by ADAM10 [corrected]. Mol Neurobiol 2019; 56:8345-8363. [PMID: 31240601 DOI: 10.1007/s12035-019-01659-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/20/2019] [Indexed: 12/13/2022]
Abstract
Unprocessed pro-neuregulin 2 (pro-NRG2) accumulates on neuronal cell bodies at junctions between the endoplasmic reticulum and plasma membrane (ER-PM junctions). NMDA receptors (NMDARs) trigger NRG2 ectodomain shedding from these sites followed by activation of ErbB4 receptor tyrosine kinases, and ErbB4 signaling cell-autonomously downregulates intrinsic excitability of GABAergic interneurons by reducing voltage-gated sodium channel currents. NMDARs also promote dispersal of Kv2.1 clusters from ER-PM junctions and cause a hyperpolarizing shift in its voltage-dependent channel activation, suggesting that NRG2/ErbB4 and Kv2.1 work together to regulate intrinsic interneuron excitability in an activity-dependent manner. Here we explored the cellular processes underlying NMDAR-dependent NRG2 shedding in cultured rat hippocampal neurons. We report that NMDARs control shedding by two separate but converging mechanisms. First, NMDA treatment disrupts binding of pro-NRG2 to ER-PM junctions by post-translationally modifying conserved Ser/Thr residues in its intracellular domain. Second, using a mutant NRG2 protein that cannot be modified at these residues and that fails to accumulate at ER-PM junctions, we demonstrate that NMDARs also directly promote NRG2 shedding by ADAM-type metalloproteinases. Using pharmacological and shRNA-mediated knockdown, and metalloproteinase overexpression, we unexpectedly find that ADAM10, but not ADAM17/TACE, is the major NRG2 sheddase acting downstream of NMDAR activation. Together, these findings reveal how NMDARs exert tight control over the NRG2/ErbB4 signaling pathway, and suggest that NRG2 and Kv2.1 are co-regulated components of a shared pathway that responds to elevated extracellular glutamate levels.
Collapse
Affiliation(s)
- Detlef Vullhorst
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 35 Lincoln Drive, Room 2C-1000, Bethesda, MD, 20892, USA
| | - Andres Buonanno
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 35 Lincoln Drive, Room 2C-1000, Bethesda, MD, 20892, USA.
| |
Collapse
|
97
|
Kataria H, Alizadeh A, Karimi-Abdolrezaee S. Neuregulin-1/ErbB network: An emerging modulator of nervous system injury and repair. Prog Neurobiol 2019; 180:101643. [PMID: 31229498 DOI: 10.1016/j.pneurobio.2019.101643] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 12/20/2022]
Abstract
Neuregulin-1 (Nrg-1) is a member of the Neuregulin family of growth factors with essential roles in the developing and adult nervous system. Six different types of Nrg-1 (Nrg-1 type I-VI) and over 30 isoforms have been discovered; however, their specific roles are not fully determined. Nrg-1 signals through a complex network of protein-tyrosine kinase receptors, ErbB2, ErbB3, ErbB4 and multiple intracellular pathways. Genetic and pharmacological studies of Nrg-1 and ErbB receptors have identified a critical role for Nrg-1/ErbB network in neurodevelopment including neuronal migration, neural differentiation, myelination as well as formation of synapses and neuromuscular junctions. Nrg-1 signaling is best known for its characterized role in development and repair of the peripheral nervous system (PNS) due to its essential role in Schwann cell development, survival and myelination. However, our knowledge of the impact of Nrg-1/ErbB on the central nervous system (CNS) has emerged in recent years. Ongoing efforts have uncovered a multi-faceted role for Nrg-1 in regulating CNS injury and repair processes. In this review, we provide a timely overview of the most recent updates on Nrg-1 signaling and its role in nervous system injury and diseases. We will specifically highlight the emerging role of Nrg-1 in modulating the glial and immune responses and its capacity to foster neuroprotection and remyelination in CNS injury. Nrg-1/ErbB network is a key regulatory pathway in the developing nervous system; therefore, unraveling its role in neuropathology and repair can aid in development of new therapeutic approaches for nervous system injuries and associated disorders.
Collapse
Affiliation(s)
- Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Arsalan Alizadeh
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
98
|
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: 15] [Impact Index Per Article: 2.5] [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
|
99
|
Neumann U, Ufer M, Jacobson LH, Rouzade-Dominguez ML, Huledal G, Kolly C, Lüönd RM, Machauer R, Veenstra SJ, Hurth K, Rueeger H, Tintelnot-Blomley M, Staufenbiel M, Shimshek DR, Perrot L, Frieauff W, Dubost V, Schiller H, Vogg B, Beltz K, Avrameas A, Kretz S, Pezous N, Rondeau JM, Beckmann N, Hartmann A, Vormfelde S, David OJ, Galli B, Ramos R, Graf A, Lopez Lopez C. The BACE-1 inhibitor CNP520 for prevention trials in Alzheimer's disease. EMBO Mol Med 2019; 10:emmm.201809316. [PMID: 30224383 PMCID: PMC6220303 DOI: 10.15252/emmm.201809316] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The beta‐site amyloid precursor protein cleaving enzyme‐1 (BACE‐1) initiates the generation of amyloid‐β (Aβ), and the amyloid cascade leading to amyloid plaque deposition, neurodegeneration, and dementia in Alzheimer's disease (AD). Clinical failures of anti‐Aβ therapies in dementia stages suggest that treatment has to start in the early, asymptomatic disease states. The BACE‐1 inhibitor CNP520 has a selectivity, pharmacodynamics, and distribution profile suitable for AD prevention studies. CNP520 reduced brain and cerebrospinal fluid (CSF) Aβ in rats and dogs, and Aβ plaque deposition in APP‐transgenic mice. Animal toxicology studies of CNP520 demonstrated sufficient safety margins, with no signs of hair depigmentation, retina degeneration, liver toxicity, or cardiovascular effects. In healthy adults ≥ 60 years old, treatment with CNP520 was safe and well tolerated and resulted in robust and dose‐dependent Aβ reduction in the cerebrospinal fluid. Thus, long‐term, pivotal studies with CNP520 have been initiated in the Generation Program.
Collapse
Affiliation(s)
- Ulf Neumann
- Neuroscience, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Mike Ufer
- Translational Medicine, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Laura H Jacobson
- Neuroscience, Novartis Institute for BioMedical Research, Basel, Switzerland
| | | | - Gunilla Huledal
- PK Sciences, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Carine Kolly
- Preclinical Safety, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Rainer M Lüönd
- Global Discovery Chemistry, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Rainer Machauer
- Global Discovery Chemistry, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Siem J Veenstra
- Global Discovery Chemistry, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Konstanze Hurth
- Global Discovery Chemistry, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Heinrich Rueeger
- Global Discovery Chemistry, Novartis Institute for BioMedical Research, Basel, Switzerland
| | | | | | - Derya R Shimshek
- Neuroscience, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Ludovic Perrot
- Neuroscience, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Wilfried Frieauff
- Preclinical Safety, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Valerie Dubost
- Preclinical Safety, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Hilmar Schiller
- PK Sciences, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Barbara Vogg
- PK Sciences, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Karen Beltz
- PK Sciences, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Alexandre Avrameas
- Biomarker Discovery, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Sandrine Kretz
- Biomarker Discovery, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Nicole Pezous
- Translational Medicine, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Jean-Michel Rondeau
- Chemical Biology and Therapeutics, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Nicolau Beckmann
- Musculoskeletal Diseases, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Andreas Hartmann
- Preclinical Safety, Novartis Institute for BioMedical Research, Basel, Switzerland
| | - Stefan Vormfelde
- Translational Medicine, Novartis Institute for BioMedical Research, Basel, Switzerland
| | | | - Bruno Galli
- Global Drug Development, Novartis, Basel, Switzerland
| | - Rita Ramos
- Global Drug Development, Novartis, Basel, Switzerland
| | - Ana Graf
- Global Drug Development, Novartis, Basel, Switzerland
| | | |
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
|