1
|
Liu S, Cho MY, Huang YN, Park T, Chaudhuri S, Rosewood TJ, Bice PJ, Chung D, Bennett DA, Ertekin-Taner N, Saykin AJ, Nho K. Multi-Omics Analysis for Identifying Cell-Type-Specific Druggable Targets in Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.08.25320199. [PMID: 39830273 PMCID: PMC11741481 DOI: 10.1101/2025.01.08.25320199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Background Analyzing disease-linked genetic variants via expression quantitative trait loci (eQTLs) is important for identifying potential disease-causing genes. Previous research prioritized genes by integrating Genome-Wide Association Study (GWAS) results with tissue-level eQTLs. Recent studies have explored brain cell type-specific eQTLs, but they lack a systematic analysis across various Alzheimer's disease (AD) GWAS datasets, nor did they compare effects between tissue and cell type levels or across different cell type-specific eQTL datasets. In this study, we integrated brain cell type-specific eQTL datasets with AD GWAS datasets to identify potential causal genes at the cell type level. Methods To prioritize disease-causing genes, we used Summary Data-Based Mendelian Randomization (SMR) and Bayesian Colocalization (COLOC) to integrate AD GWAS summary statistics with cell-type-specific eQTLs. Combining data from five AD GWAS, three single-cell eQTL datasets, and one bulk tissue eQTL meta-analysis, we identified and confirmed both novel and known candidate causal genes. We investigated gene regulation through enhancer activity using H3K27ac and ATAC-seq data, performed protein-protein interaction and pathway enrichment analyses, and conducted a drug/compound enrichment analysis with the Drug Signatures Database (DSigDB) to support drug repurposing for AD. Results We identified 27 candidate causal genes for AD using cell type-specific eQTL datasets, with the highest numbers in microglia, followed by excitatory neurons, astrocytes, inhibitory neurons, oligodendrocytes, and oligodendrocyte precursor cells (OPCs). PABPC1 emerged as a novel astrocyte-specific gene. Our analysis revealed protein-protein interaction (PPI) networks for these causal genes in microglia and astrocytes. We found the "regulation of aspartic-type peptidase activity" pathway being the most enriched among all the causal genes. AD-risk variants associated with candidate causal gene PABPC1 is located near or within enhancers only active in astrocytes. We classified the genes into three drug tiers and identified druggable interactions, with imatinib mesylate emerging as a key candidate. A drug-target gene network was created to explore potential drug targets for AD. Conclusions We systematically prioritized AD candidate causal genes based on cell type-specific molecular evidence. The integrative approach enhances our understanding of molecular mechanisms of AD-related genetic variants and facilitates the interpretation of AD GWAS results.
Collapse
Affiliation(s)
- Shiwei Liu
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Min Young Cho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
- Sungkyunkwan University, Seoul, Republic of Korea
| | - Yen-Ning Huang
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Tamina Park
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Soumilee Chaudhuri
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Thea Jacobson Rosewood
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Paula J Bice
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Dongjun Chung
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, OH, 43210, USA
| | - David A. Bennett
- Department of Neurological Science, Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
| | - Kwangsik Nho
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 550 N. University Blvd. Indianapolis, IN, 46202, USA
- Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, 355 W. 16th Street, Goodman Hall, Suite 4100, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, 340 West 10th Street, Fairbanks Hall, Suite 6200 Indianapolis, Indiana, 46202, USA
| |
Collapse
|
2
|
Attri M, Raghav A, Sinha J. Revolutionising Neurological Therapeutics: Investigating Drug Repurposing Strategies. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:115-131. [PMID: 39323347 DOI: 10.2174/0118715273329531240911075309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 09/27/2024]
Abstract
Repurposing drugs (DR) has become a viable approach to hasten the search for cures for neurodegenerative diseases (NDs). This review examines different off-target and on-target drug discovery techniques and how they might be used to find possible treatments for non-diagnostic depressions. Off-target strategies look at the known or unknown side effects of currently approved drugs for repositioning, whereas on-target strategies connect disease pathways to targets that can be treated with drugs. The review highlights the potential of experimental and computational methodologies, such as machine learning, proteomic techniques, network and genomics-based approaches, and in silico screening, in uncovering new drug-disease correlations. It also looks at difficulties and failed attempts at drug repurposing for NDs, highlighting the necessity of exact and standardised procedures to increase success rates. This review's objectives are to address the purpose of drug repurposing in human disorders, particularly neurological diseases, and to provide an overview of repurposing candidates that are presently undergoing clinical trials for neurological conditions, along with any possible causes and early findings. We then include a list of drug repurposing strategies, restrictions, and difficulties for upcoming research.
Collapse
Affiliation(s)
- Meenakshi Attri
- School of Medical & Allied Sciences, K.R. Mangalam University, Gurugram, Haryana 122103, India
| | - Asha Raghav
- Department of Pharmaceutics, School of Health Sciences, Sushant University, Gurugram, Haryana 122003, India
| | - Jyoti Sinha
- Department of Pharmaceutics, School of Health Sciences, Sushant University, Gurugram, Haryana 122003, India
| |
Collapse
|
3
|
Netzer WJ, Sinha A, Ghias M, Chang E, Gindinova K, Mui E, Seo JS, Sinha SC. Stretching the structural envelope of imatinib to reduce β-amyloid production by modulating both β- and γ-secretase cleavages of APP. Front Chem 2024; 12:1381205. [PMID: 39439934 PMCID: PMC11493595 DOI: 10.3389/fchem.2024.1381205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024] Open
Abstract
We previously showed that the anticancer drug imatinib mesylate (IMT, trade name: Gleevec) and a chemically distinct compound, DV2-103 (a kinase-inactive derivative of the potent Abl and Src kinase inhibitor, PD173955) lower Aβ levels at low micromolar concentrations primarily through a lysosome-dependent mechanism that renders APP less susceptible to proteolysis by BACE1 without directly inhibiting BACE1 enzymatic activity, or broadly inhibiting the processing of other BACE1 substrates. Additionally, IMT indirectly inhibits γ-secretase and stimulates autophagy, and thus may decrease Aβ levels through multiple pathways. In two recent studies we demonstrated similar effects on APP metabolism caused by derivatives of IMT and DV2-103. In the present study, we synthesized and tested radically altered IMT isomers (IMTi's) that possess medium structural similarity to IMT. Independent of structural similarity, these isomers manifest widely differing potencies in altering APP metabolism. These will enable us to choose the most potent isomers for further derivatization.
Collapse
Affiliation(s)
- William J. Netzer
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Anjana Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Mondana Ghias
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Emily Chang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Katherina Gindinova
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Emily Mui
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Ji-Seon Seo
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Subhash C. Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
- Appel Alzheimer’s Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
4
|
Das V, Miller JH, Alladi CG, Annadurai N, De Sanctis JB, Hrubá L, Hajdúch M. Antineoplastics for treating Alzheimer's disease and dementia: Evidence from preclinical and observational studies. Med Res Rev 2024; 44:2078-2111. [PMID: 38530106 DOI: 10.1002/med.22033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024]
Abstract
As the world population ages, there will be an increasing need for effective therapies for aging-associated neurodegenerative disorders, which remain untreatable. Dementia due to Alzheimer's disease (AD) is one of the leading neurological diseases in the aging population. Current therapeutic approaches to treat this disorder are solely symptomatic, making the need for new molecular entities acting on the causes of the disease extremely urgent. One of the potential solutions is to use compounds that are already in the market. The structures have known pharmacokinetics, pharmacodynamics, toxicity profiles, and patient data available in several countries. Several drugs have been used successfully to treat diseases different from their original purposes, such as autoimmunity and peripheral inflammation. Herein, we divulge the repurposing of drugs in the area of neurodegenerative diseases, focusing on the therapeutic potential of antineoplastics to treat dementia due to AD and dementia. We briefly touch upon the shared pathological mechanism between AD and cancer and drug repurposing strategies, with a focus on artificial intelligence. Next, we bring out the current status of research on the development of drugs, provide supporting evidence from retrospective, clinical, and preclinical studies on antineoplastic use, and bring in new areas, such as repurposing drugs for the prion-like spreading of pathologies in treating AD.
Collapse
Affiliation(s)
- Viswanath Das
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - John H Miller
- School of Biological Sciences and Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Charanraj Goud Alladi
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Narendran Annadurai
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - Lenka Hrubá
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| |
Collapse
|
5
|
Bretou M, Sannerud R, Escamilla-Ayala A, Leroy T, Vrancx C, Van Acker ZP, Perdok A, Vermeire W, Vorsters I, Van Keymolen S, Maxson M, Pavie B, Wierda K, Eskelinen EL, Annaert W. Accumulation of APP C-terminal fragments causes endolysosomal dysfunction through the dysregulation of late endosome to lysosome-ER contact sites. Dev Cell 2024; 59:1571-1592.e9. [PMID: 38626765 DOI: 10.1016/j.devcel.2024.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/02/2023] [Accepted: 03/20/2024] [Indexed: 04/18/2024]
Abstract
Neuronal endosomal and lysosomal abnormalities are among the early changes observed in Alzheimer's disease (AD) before plaques appear. However, it is unclear whether distinct endolysosomal defects are temporally organized and how altered γ-secretase function or amyloid precursor protein (APP) metabolism contribute to these changes. Inhibiting γ-secretase chronically, in mouse embryonic fibroblast and hippocampal neurons, led to a gradual endolysosomal collapse initiated by decreased lysosomal calcium and increased cholesterol, causing downstream defects in endosomal recycling and maturation. This endolysosomal demise is γ-secretase dependent, requires membrane-tethered APP cytoplasmic domains, and is rescued by APP depletion. APP C-terminal fragments (CTFs) localized to late endosome/lysosome-endoplasmic reticulum contacts; an excess of APP-CTFs herein reduced lysosomal Ca2+ refilling from the endoplasmic reticulum, promoting cholesterol accretion. Tonic regulation by APP-CTFs provides a mechanistic explanation for their cellular toxicity: failure to timely degrade APP-CTFs sustains downstream signaling, instigating lysosomal dyshomeostasis, as observed in prodromal AD. This is the opposite of substrates such as Notch, which require intramembrane proteolysis to initiate signaling.
Collapse
Affiliation(s)
- Marine Bretou
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Ragna Sannerud
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | | | - Tom Leroy
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Céline Vrancx
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Zoë P Van Acker
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Anika Perdok
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Wendy Vermeire
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Inge Vorsters
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Sophie Van Keymolen
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Michelle Maxson
- Cell Biology Program, The Hospital for Sick Children, Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Benjamin Pavie
- VIB-BioImaging Core, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | - Keimpe Wierda
- Electrophysiology Expertise Unit, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | | | - Wim Annaert
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
6
|
Obaha A, Novinec M. Regulation of Peptidase Activity beyond the Active Site in Human Health and Disease. Int J Mol Sci 2023; 24:17120. [PMID: 38069440 PMCID: PMC10707025 DOI: 10.3390/ijms242317120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
This comprehensive review addresses the intricate and multifaceted regulation of peptidase activity in human health and disease, providing a comprehensive investigation that extends well beyond the boundaries of the active site. Our review focuses on multiple mechanisms and highlights the important role of exosites, allosteric sites, and processes involved in zymogen activation. These mechanisms play a central role in shaping the complex world of peptidase function and are promising potential targets for the development of innovative drugs and therapeutic interventions. The review also briefly discusses the influence of glycosaminoglycans and non-inhibitory binding proteins on enzyme activities. Understanding their role may be a crucial factor in the development of therapeutic strategies. By elucidating the intricate web of regulatory mechanisms that control peptidase activity, this review deepens our understanding in this field and provides a roadmap for various strategies to influence and modulate peptidase activity.
Collapse
Affiliation(s)
| | - Marko Novinec
- Faculty of Chemistry and Chemical Technology, Department of Chemistry and Biochemistry, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia;
| |
Collapse
|
7
|
Majeed J, Sabbagh MN, Kang MH, Lawrence JJ, Pruitt K, Bacus S, Reyna E, Brown M, Decourt B. Cancer drugs with high repositioning potential for Alzheimer's disease. Expert Opin Emerg Drugs 2023; 28:311-332. [PMID: 38100555 PMCID: PMC10877737 DOI: 10.1080/14728214.2023.2296079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/13/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION Despite the recent full FDA approval of lecanemab, there is currently no disease modifying therapy (DMT) that can efficiently slow down the progression of Alzheimer's disease (AD) in the general population. This statement emphasizes the need to identify novel DMTs in the shortest time possible to prevent a global epidemic of AD cases as the world population experiences an increase in lifespan. AREAS COVERED Here, we review several classes of anti-cancer drugs that have been or are being investigated in Phase II/III clinical trials for AD, including immunomodulatory drugs, RXR agonists, sex hormone therapies, tyrosine kinase inhibitors, and monoclonal antibodies. EXPERT OPINION Given the overall course of brain pathologies during the progression of AD, we express a great enthusiasm for the repositioning of anti-cancer drugs as possible AD DMTs. We anticipate an increasing number of combinatorial therapy strategies to tackle AD symptoms and their underlying pathologies. However, we strongly encourage improvements in clinical trial study designs to better assess target engagement and possible efficacy over sufficient periods of drug exposure.
Collapse
Affiliation(s)
- Jad Majeed
- University of Arizona Honors College, Tucson, Arizona, USA
| | - Marwan N. Sabbagh
- Alzheimer’s and Memory Disorders Division, Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Min H. Kang
- Department of Pediatrics, Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - J. Josh Lawrence
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Kevin Pruitt
- Department of Pharmacology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Ellie Reyna
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Maddy Brown
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Boris Decourt
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
- Roseman University of Health Sciences, Las Vegas, Nevada, USA
| |
Collapse
|
8
|
FDA-Approved Kinase Inhibitors in Preclinical and Clinical Trials for Neurological Disorders. Pharmaceuticals (Basel) 2022; 15:ph15121546. [PMID: 36558997 PMCID: PMC9784968 DOI: 10.3390/ph15121546] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/09/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Cancers and neurological disorders are two major types of diseases. We previously developed a new concept termed "Aberrant Cell Cycle Diseases" (ACCD), revealing that these two diseases share a common mechanism of aberrant cell cycle re-entry. The aberrant cell cycle re-entry is manifested as kinase/oncogene activation and tumor suppressor inactivation, which are hallmarks of both tumor growth in cancers and neuronal death in neurological disorders. Therefore, some cancer therapies (e.g., kinase inhibition, tumor suppressor elevation) can be leveraged for neurological treatments. The United States Food and Drug Administration (US FDA) has so far approved 74 kinase inhibitors, with numerous other kinase inhibitors in clinical trials, mostly for the treatment of cancers. In contrast, there are dire unmet needs of FDA-approved drugs for neurological treatments, such as Alzheimer's disease (AD), intracerebral hemorrhage (ICH), ischemic stroke (IS), traumatic brain injury (TBI), and others. In this review, we list these 74 FDA-approved kinase-targeted drugs and identify those that have been reported in preclinical and/or clinical trials for neurological disorders, with a purpose of discussing the feasibility and applicability of leveraging these cancer drugs (FDA-approved kinase inhibitors) for neurological treatments.
Collapse
|
9
|
Hunt NJ, Wahl D, Westwood LJ, Lockwood GP, Le Couteur DG, Cogger VC. Targeting the liver in dementia and cognitive impairment: Dietary macronutrients and diabetic therapeutics. Adv Drug Deliv Rev 2022; 190:114537. [PMID: 36115494 PMCID: PMC10125004 DOI: 10.1016/j.addr.2022.114537] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 01/24/2023]
Abstract
Many people living with dementia and cognitive impairment have dysfunctional mitochondrial and insulin-glucose metabolism resembling type 2 diabetes mellitus and old age. Evidence from human trials shows that nutritional interventions and anti-diabetic medicines that target nutrient-sensing pathways overcome these deficits in glucose and energy metabolism and can improve cognition and/or reduce symptoms of dementia. The liver is the main organ that mediates the systemic effects of diets and many diabetic medicines; therefore, it is an intermediate target for such dementia interventions. A challenge is the efficacy of these treatments in older age. Solutions include the targeted hepatic delivery of diabetic medicines using nanotechnologies and titration of macronutrients to optimize hepatic energy metabolism.
Collapse
Affiliation(s)
- Nicholas J Hunt
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2008, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2008, Australia; Sydney Nano Institute, The University of Sydney, Sydney, NSW 2008, Australia; ANZAC Research Institute & Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW 2139, Australia
| | - Devin Wahl
- Department of Health and Exercise Science & Centre for Healthy Aging, Colorado State University, CO 80523, United States
| | - Lara J Westwood
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2008, Australia; ANZAC Research Institute & Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW 2139, Australia
| | - Glen P Lockwood
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2008, Australia; ANZAC Research Institute & Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW 2139, Australia
| | - David G Le Couteur
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2008, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2008, Australia; ANZAC Research Institute & Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW 2139, Australia
| | - Victoria C Cogger
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2008, Australia; ANZAC Research Institute & Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW 2139, Australia.
| |
Collapse
|
10
|
Hur JY. γ-Secretase in Alzheimer's disease. Exp Mol Med 2022; 54:433-446. [PMID: 35396575 PMCID: PMC9076685 DOI: 10.1038/s12276-022-00754-8] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/05/2022] [Accepted: 01/20/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is caused by synaptic and neuronal loss in the brain. One of the characteristic hallmarks of AD is senile plaques containing amyloid β-peptide (Aβ). Aβ is produced from amyloid precursor protein (APP) by sequential proteolytic cleavages by β-secretase and γ-secretase, and the polymerization of Aβ into amyloid plaques is thought to be a key pathogenic event in AD. Since γ-secretase mediates the final cleavage that liberates Aβ, γ-secretase has been widely studied as a potential drug target for the treatment of AD. γ-Secretase is a transmembrane protein complex containing presenilin, nicastrin, Aph-1, and Pen-2, which are sufficient for γ-secretase activity. γ-Secretase cleaves >140 substrates, including APP and Notch. Previously, γ-secretase inhibitors (GSIs) were shown to cause side effects in clinical trials due to the inhibition of Notch signaling. Therefore, more specific regulation or modulation of γ-secretase is needed. In recent years, γ-secretase modulators (GSMs) have been developed. To modulate γ-secretase and to understand its complex biology, finding the binding sites of GSIs and GSMs on γ-secretase as well as identifying transiently binding γ-secretase modulatory proteins have been of great interest. In this review, decades of findings on γ-secretase in AD are discussed.
Collapse
Affiliation(s)
- Ji-Yeun Hur
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
11
|
Bhargavan B, Woollard SM, McMillan JE, Kanmogne GD. CCR5 antagonist reduces HIV-induced amyloidogenesis, tau pathology, neurodegeneration, and blood-brain barrier alterations in HIV-infected hu-PBL-NSG mice. Mol Neurodegener 2021; 16:78. [PMID: 34809709 PMCID: PMC8607567 DOI: 10.1186/s13024-021-00500-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 11/03/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neurocognitive impairment is present in 50% of HIV-infected individuals and is often associated with Alzheimer's Disease (AD)-like brain pathologies, including increased amyloid-beta (Aβ) and Tau hyperphosphorylation. Here, we aimed to determine whether HIV-1 infection causes AD-like pathologies in an HIV/AIDS humanized mouse model, and whether the CCR5 antagonist maraviroc alters HIV-induced pathologies. METHODS NOD/scid-IL-2Rγcnull mice engrafted with human blood leukocytes were infected with HIV-1, left untreated or treated with maraviroc (120 mg/kg twice/day). Human cells in animal's blood were quantified weekly by flow cytometry. Animals were sacrificed at week-3 post-infection; blood and tissues viral loads were quantified using p24 antigen ELISA, RNAscope, and qPCR. Human (HLA-DR+) cells, Aβ-42, phospho-Tau, neuronal markers (MAP 2, NeuN, neurofilament-L), gamma-secretase activating protein (GSAP), and blood-brain barrier (BBB) tight junction (TJ) proteins expression and transcription were quantified in brain tissues by immunohistochemistry, immunofluorescence, immunoblotting, and qPCR. Plasma Aβ-42, Aβ-42 cellular uptake, release and transendothelial transport were quantified by ELISA. RESULTS HIV-1 significantly decreased human (h)CD4+ T-cells and hCD4/hCD8 ratios; decreased the expression of BBB TJ proteins claudin-5, ZO-1, ZO-2; and increased HLA-DR+ cells in brain tissues. Significantly, HIV-infected animals showed increased plasma and brain Aβ-42 and phospho-Tau (threonine181, threonine231, serine396, serine199), associated with transcriptional upregulation of GSAP, an enzyme that catalyzes Aβ formation, and loss of MAP 2, NeuN, and neurofilament-L. Maraviroc treatment significantly reduced blood and brain viral loads, prevented HIV-induced loss of neuronal markers and TJ proteins; decreased HLA-DR+ cells infiltration in brain tissues, significantly reduced HIV-induced increase in Aβ-42, GSAP, and phospho-Tau. Maraviroc also reduced Aβ retention and increased Aβ release in human macrophages; decreased the receptor for advanced glycation end products (RAGE) and increased low-density lipoprotein receptor-related protein-1 (LRP1) expression in human brain endothelial cells. Maraviroc induced Aβ transendothelial transport, which was blocked by LRP1 antagonist but not RAGE antagonist. CONCLUSIONS Maraviroc significantly reduced HIV-induced amyloidogenesis, GSAP, phospho-Tau, neurodegeneration, BBB alterations, and leukocytes infiltration into the CNS. Maraviroc increased cellular Aβ efflux and transendothelial Aβ transport via LRP1 pathways. Thus, therapeutically targeting CCR5 could reduce viremia, preserve the BBB and neurons, increased brain Aβ efflux, and reduce AD-like neuropathologies.
Collapse
Affiliation(s)
- Biju Bhargavan
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, 985800 Nebraska Medical Center, Omaha, NE 68198-5800 USA
| | - Shawna M. Woollard
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, 985800 Nebraska Medical Center, Omaha, NE 68198-5800 USA
- Huvepharma, 421 W Industrial Lake Drive, Lincoln, NE 68528 USA
| | - Jo Ellyn McMillan
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, 985800 Nebraska Medical Center, Omaha, NE 68198-5800 USA
| | - Georgette D. Kanmogne
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, 985800 Nebraska Medical Center, Omaha, NE 68198-5800 USA
| |
Collapse
|
12
|
Saka R, Chella N, Khan W. Development of Imatinib Mesylate-Loaded Liposomes for Nose to Brain Delivery: In Vitro and In Vivo Evaluation. AAPS PharmSciTech 2021; 22:192. [PMID: 34184160 DOI: 10.1208/s12249-021-02072-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022] Open
Abstract
Neurodegenerative diseases like Alzheimer's disease require treatment where it is essential for drug to reach brain. Nose to brain delivery of drugs enables direct transport to brain bypassing blood brain barrier. Imatinib mesylate, an anti-cancer agent, was found to have potential anti-Alzheimer's activity and thus repurposed for the same. However, the drug has severe side effects, poor brain bioavailability which may hinder effective treatment of Alzheimer's disease. In the current work, imatinib mesylate-loaded liposomes were prepared with particle size below 150 nm with sustained drug release up to 96 h. The liposomal drug formulation was compared with plain drug solution for cytotoxicity on N2a cells and did not show any kind of toxicity at concentrations up to 25 μg/mL. The nanocarrier formulation was then evaluated for brain deposition by nose to brain administration in comparison with drug solution in rats. The liposomes effectively improved the brain deposition of drug in brain from formulation compared to pure drug solution as indicated by AUC from in vivo experiments. These results indicate that the nose to brain delivery of liposomal imatinib mesylate improved the drug deposition and residence time in brain compared to drug solution administered through oral and intranasal routes.
Collapse
|
13
|
Wolfe MS. Probing Mechanisms and Therapeutic Potential of γ-Secretase in Alzheimer's Disease. MOLECULES (BASEL, SWITZERLAND) 2021; 26:molecules26020388. [PMID: 33450968 PMCID: PMC7828430 DOI: 10.3390/molecules26020388] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/02/2021] [Accepted: 01/10/2021] [Indexed: 12/14/2022]
Abstract
The membrane-embedded γ-secretase complex carries out hydrolysis within the lipid bilayer in proteolyzing nearly 150 different membrane protein substrates. Among these substrates, the amyloid precursor protein (APP) has been the most studied, as generation of aggregation-prone amyloid β-protein (Aβ) is a defining feature of Alzheimer's disease (AD). Mutations in APP and in presenilin, the catalytic component of γ-secretase, cause familial AD, strong evidence for a pathogenic role of Aβ. Substrate-based chemical probes-synthetic peptides and peptidomimetics-have been critical to unraveling the complexity of γ-secretase, and small drug-like inhibitors and modulators of γ-secretase activity have been essential for exploring the potential of the protease as a therapeutic target for Alzheimer's disease. Such chemical probes and therapeutic prototypes will be reviewed here, with concluding commentary on the future directions in the study of this biologically important protease complex and the translation of basic findings into therapeutics.
Collapse
Affiliation(s)
- Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, 1567 Irving Hill Road, GLH-2115, Lawrence, KS 66045, USA
| |
Collapse
|
14
|
Wong E, Frost GR, Li YM. γ-Secretase Modulatory Proteins: The Guiding Hand Behind the Running Scissors. Front Aging Neurosci 2020; 12:614690. [PMID: 33343338 PMCID: PMC7738330 DOI: 10.3389/fnagi.2020.614690] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/10/2020] [Indexed: 12/16/2022] Open
Abstract
Described as the "proteasome of the membrane" or the "scissors in the membrane," γ-secretase has notoriously complicated biology, and even after decades of research, the full extent of its regulatory mechanism remains unclear. γ-Secretase is an intramembrane aspartyl protease complex composed of four obligatory subunits: Nicastrin (NCT), Presenilin (PS), Presenilin Enhancer-2 (Pen-2), and Anterior pharynx-defective-1 (Aph-1). γ-Secretase cleaves numerous type 1 transmembrane substrates, with no apparent homology, and plays major roles in broad biological pathways such as development, neurogenesis, and cancer. Notch and the amyloid precursor protein (APP) and are undoubtedly the best-studied γ-secretase substrates because of their role in cancer and Alzheimer's disease (AD) and therefore became the focus of increasing studies as an attractive therapeutic target. The regulation of γ-secretase is intricate and involves the function of multiple cellular entities. Recently, γ-secretase modulatory proteins (GSMPs), which are non-essential subunits and yet modulate γ-secretase activity and specificity, have emerged as an important component in guiding γ-secretase. GSMPs are responsive to cellular and environmental changes and therefore, provide another layer of regulation of γ-secretase. This type of enzymatic regulation allows for a rapid and fine-tuning of γ-secretase activity when appropriate signals appear enabling a temporal level of regulation. In this review article, we discuss the latest developments on GSMPs and implications on the development of effective therapeutics for γ-secretase-associated diseases such as AD and cancer.
Collapse
Affiliation(s)
- Eitan Wong
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
15
|
Sinha A, Chang JC, Xu P, Gindinova K, Cho Y, Sun W, Wu X, Li YM, Greengard P, Kelly JW, Sinha SC. Brain Permeable Tafamidis Amide Analogs for Stabilizing TTR and Reducing APP Cleavage. ACS Med Chem Lett 2020; 11:1973-1979. [PMID: 33062181 PMCID: PMC7549266 DOI: 10.1021/acsmedchemlett.9b00688] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/02/2020] [Indexed: 02/08/2023] Open
Abstract
Tafamidis, 1, a potent transthyretin kinetic stabilizer, weakly inhibits the γ-secretase enzyme in vitro. We have synthesized four amide derivatives of 1. These compounds reduce production of the Aβ peptide in N2a695 cells but do not inhibit the γ-secretase enzyme in cell-free assays. By performing fluorescence correlation spectroscopy, we have shown that TTR inhibits Aβ oligomerization and that addition of tafamidis or its amide derivative does not affect TTR's ability to inhibit Aβ oligomerization. The piperazine amide derivative of tafamidis (1a) efficiently penetrates and accumulates in mouse brain and undergoes proteolysis under physiological conditions in mice to produce tafamidis.
Collapse
Affiliation(s)
- Anjana Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Jerry C Chang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Peng Xu
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Katherina Gindinova
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Younhee Cho
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Weilin Sun
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Xianzhong Wu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Yue Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Subhash C Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| |
Collapse
|
16
|
Luo W, Ip FCF, Fu G, Cheung K, Tian Y, Hu Y, Sinha A, Cheng EYL, Wu X, Bustos V, Greengard P, Li YM, Sinha SC, Ip NY. A Pentacyclic Triterpene from Ligustrum lucidum Targets γ-Secretase. ACS Chem Neurosci 2020; 11:2827-2835. [PMID: 32786303 PMCID: PMC8325170 DOI: 10.1021/acschemneuro.0c00389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyloid-beta peptides generated by β-secretase- and γ-secretase-mediated successive cleavage of amyloid precursor protein are believed to play a causative role in Alzheimer's disease. Thus, reducing amyloid-beta generation by modulating γ-secretase remains a promising approach for Alzheimer's disease therapeutic development. Here, we screened fruit extracts of Ligustrum lucidum Ait. (Oleaceae) and identified active fractions that increase the C-terminal fragment of amyloid precursor protein and reduce amyloid-beta production in a neuronal cell line. These fractions contain a mixture of two isomeric pentacyclic triterpene natural products, 3-O-cis- or 3-O-trans-p-coumaroyl maslinic acid (OCMA), in different ratios. We further demonstrated that trans-OCMA specifically inhibits γ-secretase and decreases amyloid-beta levels without influencing cleavage of Notch. By using photoactivatable probes targeting the subsites residing in the γ-secretase active site, we demonstrated that trans-OCMA selectively affects the S1 subsite of the active site in this protease. Treatment of Alzheimer's disease transgenic model mice with trans-OCMA or an analogous carbamate derivative of a related pentacyclic triterpene natural product, oleanolic acid, rescued the impairment of synaptic plasticity. This work indicates that the naturally occurring compound trans-OCMA and its analogues could become a promising class of small molecules for Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Wenjie Luo
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Fanny C F Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay Road, Kowloon, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, Shenzhen-Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China 518057
| | - Guangmiao Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay Road, Kowloon, Hong Kong, China
| | - Kit Cheung
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay Road, Kowloon, Hong Kong, China
| | - Yuan Tian
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Yueqing Hu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay Road, Kowloon, Hong Kong, China
| | - Anjana Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Elaine Y L Cheng
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay Road, Kowloon, Hong Kong, China
| | - Xianzhong Wu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Victor Bustos
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Subhash C Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Nancy Y Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, and Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay Road, Kowloon, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, Shenzhen-Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China 518057
| |
Collapse
|
17
|
Martins M, Silva R, M. M. Pinto M, Sousa E. Marine Natural Products, Multitarget Therapy and Repurposed Agents in Alzheimer's Disease. Pharmaceuticals (Basel) 2020; 13:E242. [PMID: 32933034 PMCID: PMC7558913 DOI: 10.3390/ph13090242] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial disease characterized by the presence of amyloid plaques, neurofibrillary tangles, and nerve cell death that affects, mainly, older people. After decades of investigation, the search for an efficacious treatment for AD remains and several strategies can be and are being employed in this journey. In this review, four of the most promising strategies, alongside with its most promising agents under investigation or development are highlighted. Marine natural products (MNP) are a source of unique chemical structures with useful biological activities for AD treatment. One of the most promising compounds, a marine-derived acidic oligosaccharide (GV-971) just passed phase III clinical trials with a unique mechanism of action. Combination therapy and multitargeted-directed ligand therapy (MTDL) are also two important strategies, with several examples in clinical trials, based on the belief that the best approach for AD is a therapy capable of modulating multiple target pathways. Drug repurposing, a strategy that requires a smaller investment and is less time consuming, is emerging as a strong contender with a variety of pharmacological agents resurfacing in an attempt to identify a therapeutic candidate capable of modifying the course of this disease.
Collapse
Affiliation(s)
- Márcia Martins
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.M.); (M.M.M.P.)
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Renata Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
| | - Madalena M. M. Pinto
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.M.); (M.M.M.P.)
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Emília Sousa
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.M.); (M.M.M.P.)
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
18
|
Zhang S, Cai F, Wu Y, Bozorgmehr T, Wang Z, Zhang S, Huang D, Guo J, Shen L, Rankin C, Tang B, Song W. A presenilin-1 mutation causes Alzheimer disease without affecting Notch signaling. Mol Psychiatry 2020; 25:603-613. [PMID: 29915376 DOI: 10.1038/s41380-018-0101-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/19/2018] [Accepted: 05/09/2018] [Indexed: 02/01/2023]
Abstract
Presenilin-1 (PSEN1) is the catalytic subunit of the γ-secretase complex, and pathogenic mutations in the PSEN1 gene account for the majority cases of familial AD (FAD). FAD-associated mutant PSEN1 proteins have been shown to affect APP processing and Aβ generation and inhibit Notch1 cleavage and Notch signaling. In this report, we found that a PSEN1 mutation (S169del) altered APP processing and Aβ generation, and promoted neuritic plaque formation as well as learning and memory deficits in AD model mice. However, this mutation did not affect Notch1 cleavage and Notch signaling in vitro and in vivo. Taken together, we demonstrated that PSEN1S169del has distinct effects on APP processing and Notch1 cleavage, suggesting that Notch signaling may not be critical for AD pathogenesis and serine169 could be a critical site as a potential target for the development of novel γ-secretase modulators without affecting Notch1 cleavage to treat AD.
Collapse
Affiliation(s)
- Shuting Zhang
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Fang Cai
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yili Wu
- Department of Psychiatry, Graduate Program in Psychiatry, Jining Medical University, Jining, China
| | - Tahereh Bozorgmehr
- Department of Psychology, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Zhe Wang
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Si Zhang
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Daochao Huang
- Chongqing City Key Lab of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, 400014, Chongqing, China
| | - Jifeng Guo
- Department of Neurology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lu Shen
- Department of Neurology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Catharine Rankin
- Department of Psychology, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Beisha Tang
- Department of Neurology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weihong Song
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
19
|
Bassendine MF, Taylor-Robinson SD, Fertleman M, Khan M, Neely D. Is Alzheimer's Disease a Liver Disease of the Brain? J Alzheimers Dis 2020; 75:1-14. [PMID: 32250293 PMCID: PMC7306895 DOI: 10.3233/jad-190848] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Clinical specialization is not only a force for progress, but it has also led to the fragmentation of medical knowledge. The focus of research in the field of Alzheimer's disease (AD) is neurobiology, while hepatologists focus on liver diseases and lipid specialists on atherosclerosis. This article on AD focuses on the role of the liver and lipid homeostasis in the development of AD. Amyloid-β (Aβ) deposits accumulate as plaques in the brain of an AD patient long before cognitive decline is evident. Aβ generation is a normal physiological process; the steady-state level of Aβ in the brain is determined by balance between Aβ production and its clearance. We present evidence suggesting that the liver is the origin of brain Aβ deposits and that it is involved in peripheral clearance of circulating Aβ in the blood. Hence the liver could be targeted to decrease Aβ production or increase peripheral clearance.
Collapse
Affiliation(s)
- Margaret F. Bassendine
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
- Department of Hepatology & Gastroenterology, Division of Surgery and Cancer, Imperial College London, St Mary’s Campus, UK
| | - Simon D. Taylor-Robinson
- Department of Hepatology & Gastroenterology, Division of Surgery and Cancer, Imperial College London, St Mary’s Campus, UK
| | - Michael Fertleman
- Department of Hepatology & Gastroenterology, Division of Surgery and Cancer, Imperial College London, St Mary’s Campus, UK
- Department of Bioengineering, Imperial College London, UK
| | - Michael Khan
- University of Warwick & University Hospitals of Coventry and Warwickshire NHS Trust, UK
| | - Dermot Neely
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
- Department of Blood Sciences, Newcastle upon Tyne Hospitals NHS Foundation Trust, UK
| |
Collapse
|
20
|
Sinha A, Gindinova K, Mui E, Netzer WJ, Sinha SC. Development of Kinase Inactive PD173955 Analogues for Reducing Production of Aβ Peptides. ACS Med Chem Lett 2019; 10:1430-1435. [PMID: 31620229 DOI: 10.1021/acsmedchemlett.9b00213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 08/29/2019] [Indexed: 01/06/2023] Open
Abstract
Compound 3a, DV2-103, is a kinase inactive analogue of a potent Abl1/Src kinase inhibitor, PD173955, 2. Both compounds, 2 and 3a, are known to reduce production of beta amyloid (Aβ) peptide in cells and animal models. We have now prepared and evaluated a series of PD-173955 analogues, several of which reduced Aβ production potently. This occurs in cells expressing human full-length amyloid precursor protein (APP) and not in cells expressing APP β-C terminal fragment (APP-C99), suggesting that the kinase inactive analogues strongly affect β-secretase (BACE1) cleavage of APP, similarly to Gleevec. A combination of the kinase inactive analogues of PD173955 with a BACE1 inhibitor (BACEi), namely, BACE IV, strongly reduced Aβ levels in cells, as noted previously with Gleevec and analogues. Several potent compounds also penetrated and accumulated in mouse brain in high nanomolar to low micromolar concentration.
Collapse
Affiliation(s)
- Anjana Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Katherina Gindinova
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Emily Mui
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - William J. Netzer
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Subhash C. Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| |
Collapse
|
21
|
Sales TA, Prandi IG, Castro AAD, Leal DHS, Cunha EFFD, Kuca K, Ramalho TC. Recent Developments in Metal-Based Drugs and Chelating Agents for Neurodegenerative Diseases Treatments. Int J Mol Sci 2019; 20:E1829. [PMID: 31013856 PMCID: PMC6514778 DOI: 10.3390/ijms20081829] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/07/2019] [Accepted: 04/09/2019] [Indexed: 02/07/2023] Open
Abstract
The brain has a unique biological complexity and is responsible for important functions in the human body, such as the command of cognitive and motor functions. Disruptive disorders that affect this organ, e.g. neurodegenerative diseases (NDDs), can lead to permanent damage, impairing the patients' quality of life and even causing death. In spite of their clinical diversity, these NDDs share common characteristics, such as the accumulation of specific proteins in the cells, the compromise of the metal ion homeostasis in the brain, among others. Despite considerable advances in understanding the mechanisms of these diseases and advances in the development of treatments, these disorders remain uncured. Considering the diversity of mechanisms that act in NDDs, a wide range of compounds have been developed to act by different means. Thus, promising compounds with contrasting properties, such as chelating agents and metal-based drugs have been proposed to act on different molecular targets as well as to contribute to the same goal, which is the treatment of NDDs. This review seeks to discuss the different roles and recent developments of metal-based drugs, such as metal complexes and metal chelating agents as a proposal for the treatment of NDDs.
Collapse
Affiliation(s)
- Thais A Sales
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras/MG, 37200-000, Brazil.
| | - Ingrid G Prandi
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras/MG, 37200-000, Brazil.
| | - Alexandre A de Castro
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras/MG, 37200-000, Brazil.
| | - Daniel H S Leal
- Department of Health Sciences, Federal University of Espírito Santo, São Mateus/ES, 29932-540, Brazil.
| | - Elaine F F da Cunha
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras/MG, 37200-000, Brazil.
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 500 03, Czech Republic..
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, 500 03 Czech Republic.
| | - Teodorico C Ramalho
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras/MG, 37200-000, Brazil.
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 500 03, Czech Republic..
| |
Collapse
|
22
|
Kumar M, Kulshrestha R, Singh N, Jaggi AS. Expanding spectrum of anticancer drug, imatinib, in the disorders affecting brain and spinal cord. Pharmacol Res 2019; 143:86-96. [PMID: 30902661 DOI: 10.1016/j.phrs.2019.03.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/07/2019] [Accepted: 03/17/2019] [Indexed: 02/07/2023]
Abstract
Imatinib is a tyrosine kinase inhibitor and is used as a first line drug in the treatment of Philadelphia-chromosome-positive chronic myeloid leukaemia and gastrointestinal stromal tumors. Being tyrosine kinase inhibitor, imatinib modulates the activities of Abelson gene (c-Abl), Abelson related gene (ARG), platelet-derived growth factor receptor (PDGFR), FMS-like tyrosine kinase 3 (FLT3), lymphocyte-specific protein (Lck), mitogen activated protein kinase (MAPK), amyloid precursor protein intracellular domain (AICD), α-synuclein and the stem-cell factor receptor (c-kit). Studies have shown the role of imatinib in modulating the pathophysiological state of a number of disorders affecting brain and spinal cord such as Alzheimer's disease, Parkinson's disease, stroke, multiple sclerosis and spinal cord injury. The present review discusses the role of imatinib in the above described disorders and the possible mechanisms involved in these diseases.
Collapse
Affiliation(s)
- Manish Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | | | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India.
| |
Collapse
|
23
|
Sun W, Netzer WJ, Sinha A, Gindinova K, Chang E, Sinha SC. Development of Gleevec Analogues for Reducing Production of β-Amyloid Peptides through Shifting β-Cleavage of Amyloid Precursor Proteins. J Med Chem 2019; 62:3122-3134. [DOI: 10.1021/acs.jmedchem.8b02007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Weilin Sun
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - William J. Netzer
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Anjana Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Katherina Gindinova
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Emily Chang
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| | - Subhash C. Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, New York 10065, United States
| |
Collapse
|
24
|
Gupta MK, Vadde R. In silico identification of natural product inhibitors for γ‐secretase activating protein, a therapeutic target for Alzheimer's disease. J Cell Biochem 2018; 120:10323-10336. [PMID: 30565717 DOI: 10.1002/jcb.28316] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/28/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Manoj Kumar Gupta
- Department of Biotechnology & Bioinformatics Yogi Vemana University, Kadapa Andhra Pradesh India
| | - Ramakrishna Vadde
- Department of Biotechnology & Bioinformatics Yogi Vemana University, Kadapa Andhra Pradesh India
| |
Collapse
|
25
|
Cao J, Hou J, Ping J, Cai D. Advances in developing novel therapeutic strategies for Alzheimer's disease. Mol Neurodegener 2018; 13:64. [PMID: 30541602 PMCID: PMC6291983 DOI: 10.1186/s13024-018-0299-8] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's Disease (AD), the most prevalent neurodegenerative disease of aging, affects one in eight older Americans. Nearly all drug treatments tested for AD today have failed to show any efficacy. There is a great need for therapies to prevent and/or slow the progression of AD. The major challenge in AD drug development is lack of clarity about the mechanisms underlying AD pathogenesis and pathophysiology. Several studies support the notion that AD is a multifactorial disease. While there is abundant evidence that amyloid plays a role in AD pathogenesis, other mechanisms have been implicated in AD such as tangle formation and spread, dysregulated protein degradation pathways, neuroinflammation, and loss of support by neurotrophic factors. Therefore, current paradigms of AD drug design have been shifted from single target approach (primarily amyloid-centric) to developing drugs targeted at multiple disease aspects, and from treating AD at later stages of disease progression to focusing on preventive strategies at early stages of disease development. Here, we summarize current strategies and new trends of AD drug development, including pre-clinical and clinical trials that target different aspects of disease (mechanism-based versus non-mechanism based, e.g. symptomatic treatments, lifestyle modifications and risk factor management).
Collapse
Affiliation(s)
- Jiqing Cao
- James J Peters VA Medical Center, Research & Development, Bronx, NY 10468 USA
- Department of Neurology, Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- The Central Hospital of The Hua Zhong University of Science and Technology, Wuhan, China
| | - Jianwei Hou
- James J Peters VA Medical Center, Research & Development, Bronx, NY 10468 USA
- Department of Neurology, Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Jing Ping
- The Central Hospital of The Hua Zhong University of Science and Technology, Wuhan, China
| | - Dongming Cai
- James J Peters VA Medical Center, Research & Development, Bronx, NY 10468 USA
- Department of Neurology, Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- The Central Hospital of The Hua Zhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Durães F, Pinto M, Sousa E. Old Drugs as New Treatments for Neurodegenerative Diseases. Pharmaceuticals (Basel) 2018; 11:ph11020044. [PMID: 29751602 PMCID: PMC6027455 DOI: 10.3390/ph11020044] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are increasing in number, given that the general global population is becoming older. They manifest themselves through mechanisms that are not fully understood, in many cases, and impair memory, cognition and movement. Currently, no neurodegenerative disease is curable, and the treatments available only manage the symptoms or halt the progression of the disease. Therefore, there is an urgent need for new treatments for this kind of disease, since the World Health Organization has predicted that neurodegenerative diseases affecting motor function will become the second-most prevalent cause of death in the next 20 years. New therapies can come from three main sources: synthesis, natural products, and existing drugs. This last source is known as drug repurposing, which is the most advantageous, since the drug’s pharmacokinetic and pharmacodynamic profiles are already established, and the investment put into this strategy is not as significant as for the classic development of new drugs. There have been several studies on the potential of old drugs for the most relevant neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, Multiple Sclerosis and Amyotrophic Lateral Sclerosis.
Collapse
Affiliation(s)
- Fernando Durães
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- CIIMAR, Interdisciplinary Center of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos P, 4450-208 Matosinhos, Portugal.
| | - Madalena Pinto
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- CIIMAR, Interdisciplinary Center of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos P, 4450-208 Matosinhos, Portugal.
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- CIIMAR, Interdisciplinary Center of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos P, 4450-208 Matosinhos, Portugal.
| |
Collapse
|
27
|
Kumar D, Ganeshpurkar A, Kumar D, Modi G, Gupta SK, Singh SK. Secretase inhibitors for the treatment of Alzheimer's disease: Long road ahead. Eur J Med Chem 2018; 148:436-452. [DOI: 10.1016/j.ejmech.2018.02.035] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/30/2018] [Accepted: 02/10/2018] [Indexed: 10/18/2022]
|
28
|
Monacelli F, Cea M, Borghi R, Odetti P, Nencioni A. Do Cancer Drugs Counteract Neurodegeneration? Repurposing for Alzheimer's Disease. J Alzheimers Dis 2018; 55:1295-1306. [PMID: 27834781 DOI: 10.3233/jad-160840] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In spite of in depth investigations in the field of the amyloid cascade hypothesis, so far, no disease modifying therapy has been developed for Alzheimer's disease (AD). The pathophysiology provides some evidence of the inverse correlation between cancer and AD. Both AD and cancer are characterized by abnormal cellular behaviors; trigger factors along with a meta synchronously action is expected to drive cancer or neurodegeneration, supporting, respectively, progressive neuronal loss or uncontrolled cell proliferation in cancer cells. So far, cancer and AD are seemingly two opposite ends of the same biological spectrum. Basic science increasingly indicates shared molecular mechanisms between cancer and AD and gives weight to key relevant biological theories; according to them, the inverse tuning of clustered gene expression, the sharing of mutual independent pathway or the deregulated unfolded proteins system (UPR) may count for this inverse association. Additionally, the common biological background gave credibility to the recent discovery of a repurposing role for cancer drugs in AD. It refers to the development of new uses for existing pharmaceuticals having the same role as the original mechanism or to the discovery of a new drug action with disease modifying effects. The present review summarizes the most important biological theories that link neurodegeneration and cancer and provides an up-to-date revision of the repurposing cancer agents for AD. The review also addresses the gap of knowledge, since drug cancer repositioning holds an important promise but further investigations are warranted to ascertain the clinical relevance of such attractive clinical candidate compounds for AD.
Collapse
Affiliation(s)
- Fiammetta Monacelli
- Section of Geriatrics, Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| | - Michele Cea
- Section of Haematology, Department of Internal Medicine and Medical Specialties, (DIMI), University of Genoa, Genoa, Italy
| | - Roberta Borghi
- Section of Geriatrics, Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| | - Patrizio Odetti
- Section of Geriatrics, Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| | - Alessio Nencioni
- Section of Geriatrics, Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| |
Collapse
|
29
|
Zhang X, Götte M, Ibig-Rehm Y, Schuffenhauer A, Kamke M, Beisner D, Guerini D, Siebert D, Bonamy GMC, Gabriel D, Bodendorf U. Identification of SPPL2a Inhibitors by Multiparametric Analysis of a High-Content Ultra-High-Throughput Screen. SLAS DISCOVERY 2017; 22:1106-1119. [PMID: 28731783 DOI: 10.1177/2472555217719834] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The intramembrane protease signal peptide peptidase-like 2a (SPPL2a) is a potential drug target for the treatment of autoimmune diseases due to an essential role in B cells and dendritic cells. To screen a library of 1.4 million compounds for inhibitors of SPPL2a, we developed an imaging assay detecting nuclear translocation of the proteolytically released cytosolic substrate fragment. The state-of-the-art hit calling approach based on nuclear translocation resulted in numerous false-positive hits, mainly interrupting intracellular protein trafficking. To filter the false positives, we extracted 340 image-based readouts and developed a novel multiparametric analysis method that successfully triaged the primary hit list. The identified scaffolds were validated by demonstrating activity on endogenous SPPL2a and substrate CD74/p8 in B cells. The multiparametric analysis discovered diverse cellular phenotypes and provided profiles for the whole library. The principle of the presented imaging assay, the screening strategy, and multiparametric analysis are potentially applicable in future screening campaigns.
Collapse
Affiliation(s)
- Xian Zhang
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Marjo Götte
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | | | - Marion Kamke
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Dan Beisner
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, USA.,Vividion Therapeutics, San Diego, CA, USA
| | - Danilo Guerini
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Daniela Siebert
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Daniela Gabriel
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | |
Collapse
|
30
|
Wang DQ, Gao YL, Liu JX, Zheng CH, Kong XZ. Identifying drug-pathway association pairs based on L1L2,1-integrative penalized matrix decomposition. Oncotarget 2017; 8:48075-48085. [PMID: 28624800 PMCID: PMC5564627 DOI: 10.18632/oncotarget.18254] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/01/2017] [Indexed: 01/27/2023] Open
Abstract
The traditional methods of drug discovery follow the "one drug-one target" approach, which ignores the cellular and physiological environment of the action mechanism of drugs. However, pathway-based drug discovery methods can overcome this limitation. This kind of method, such as the Integrative Penalized Matrix Decomposition (iPaD) method, identifies the drug-pathway associations by taking the lasso-type penalty on the regularization term. Moreover, instead of imposing the L1-norm regularization, the L2,1-Integrative Penalized Matrix Decomposition (L2,1-iPaD) method imposes the L2,1-norm penalty on the regularization term. In this paper, based on the iPaD and L2,1-iPaD methods, we propose a novel method named L1L2,1-iPaD (L1L2,1-Integrative Penalized Matrix Decomposition), which takes the sum of the L1-norm and L2,1-norm penalties on the regularization term. Besides, we perform permutation test to assess the significance of the identified drug-pathway association pairs and compute the P-values. Compared with the existing methods, our method can identify more drug-pathway association pairs which have been validated in the CancerResource database. In order to identify drug-pathway associations which are not validated in the CancerResource database, we retrieve published papers to prove these associations. The results on two real datasets prove that our method can achieve better enrichment for identified association pairs than the iPaD and L2,1-iPaD methods.
Collapse
Affiliation(s)
- Dong-Qin Wang
- 1 School of Information Science and Engineering, Qufu Normal University, Rizhao, China
| | - Ying-Lian Gao
- 2 Library of Qufu Normal University, Qufu Normal University, Rizhao, China
| | - Jin-Xing Liu
- 1 School of Information Science and Engineering, Qufu Normal University, Rizhao, China
| | - Chun-Hou Zheng
- 1 School of Information Science and Engineering, Qufu Normal University, Rizhao, China
| | - Xiang-Zhen Kong
- 1 School of Information Science and Engineering, Qufu Normal University, Rizhao, China
| |
Collapse
|
31
|
Hada N, Netzer WJ, Belhassan F, Wennogle LP, Gizurarson S. Nose-to-brain transport of imatinib mesylate: A pharmacokinetic evaluation. Eur J Pharm Sci 2017; 102:46-54. [PMID: 28238945 DOI: 10.1016/j.ejps.2017.02.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 02/01/2017] [Accepted: 02/22/2017] [Indexed: 01/12/2023]
Abstract
The delivery of drugs to the brain is a constant challenge due to limitations imposed by the blood-brain barrier (BBB). Various methods of bypassing the BBB are under investigation. One approach is intranasal administration, where the olfactory region of the nasal cavity extends up to the cranial cavity and provides direct access to the brain. The pharmacokinetics of this transport and factors that determine transport rates and capacity is of vital importance for evaluating the clinical value of this route. Here, the pharmacokinetics of intranasally administered imatinib has been explored. Imatinib is distributed into the brain following intravenous administration, and then rapidly removed. Following intravenous administration, the brain/plasma ratio for imatinib was calculated to be 2% and remained at this ratio for 30min. The brain/plasma ratio following intranasal administration, however, was found to be 5.3% and remained at this ratio for up to 90min. Imatinib was found to be rapidly transported into the brain via the olfactory region, by shutting down the nose-to-blood-to-brain transport with epinephrine. The increased brain concentration of imatinib (0.33μg/g tissue) achieved by intranasal administration, compared with an IV injection, is likely to provide a model for developing a wide range of CNS active molecules that were previously removed from consideration as drug candidates due to their lack of CNS access. Furthermore, brain imatinib levels were increased by co-administration of the p-gp substrates, elacridar and pantoprazole, showing that both compounds were able to inhibit the elimination of imatinib from the brain.
Collapse
Affiliation(s)
- Nobuko Hada
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavík, Iceland
| | - William Joseph Netzer
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Fanny Belhassan
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavík, Iceland
| | | | - Sveinbjörn Gizurarson
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavík, Iceland.
| |
Collapse
|
32
|
Gleevec shifts APP processing from a β-cleavage to a nonamyloidogenic cleavage. Proc Natl Acad Sci U S A 2017; 114:1389-1394. [PMID: 28115709 DOI: 10.1073/pnas.1620963114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Neurotoxic amyloid-β peptides (Aβ) are major drivers of Alzheimer's disease (AD) and are formed by sequential cleavage of the amyloid precursor protein (APP) by β-secretase (BACE) and γ-secretase. Our previous study showed that the anticancer drug Gleevec lowers Aβ levels through indirect inhibition of γ-secretase activity. Here we report that Gleevec also achieves its Aβ-lowering effects through an additional cellular mechanism. It renders APP less susceptible to proteolysis by BACE without inhibiting BACE enzymatic activity or the processing of other BACE substrates. This effect closely mimics the phenotype of APP A673T, a recently discovered mutation that protects carriers against AD and age-related cognitive decline. In addition, Gleevec induces formation of a specific set of APP C-terminal fragments, also observed in cells expressing the APP protective mutation and in cells exposed to a conventional BACE inhibitor. These Gleevec phenotypes require an intracellular acidic pH and are independent of tyrosine kinase inhibition, given that a related compound lacking tyrosine kinase inhibitory activity, DV2-103, exerts similar effects on APP metabolism. In addition, DV2-103 accumulates at high concentrations in the rodent brain, where it rapidly lowers Aβ levels. This study suggests that long-term treatment with drugs that indirectly modulate BACE processing of APP but spare other BACE substrates and achieve therapeutic concentrations in the brain might be effective in preventing or delaying the onset of AD and could be safer than nonselective BACE inhibitor drugs.
Collapse
|
33
|
Yáñez M, Belbin O, Estrada L, Leal N, Contreras P, Lleó A, Burgos P, Zanlungo S, Alvarez A. c-Abl links APP-BACE1 interaction promoting APP amyloidogenic processing in Niemann-Pick type C disease. Biochim Biophys Acta Mol Basis Dis 2016; 1862:2158-2167. [DOI: 10.1016/j.bbadis.2016.08.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 07/31/2016] [Accepted: 08/19/2016] [Indexed: 11/17/2022]
|
34
|
Callahan R, Chestnut BA, Raafat A. Original Research: Featured Article: Imatinib mesylate (Gleevec) inhibits Notch and c-Myc signaling: Five-day treatment permanently rescues mammary development. Exp Biol Med (Maywood) 2016; 242:53-67. [PMID: 27550925 DOI: 10.1177/1535370216665175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/28/2016] [Indexed: 12/31/2022] Open
Abstract
Wap-Int3 transgenic females expressing the Notch4 intracellular domain (designated Int3) from the whey acidic protein promoter exhibit two phenotypes in the mammary gland: blockage of lobuloalveolar development and lactation, and tumor development with 100% penetrance. Previously, we have shown that treatment of Wap-Int3 tumor bearing mice with Imatinib mesylate (Gleevec) is associated with complete regression of the tumor. In the present study, we show that treatment of Wap-Int3 mice during day 1 through day 6 of pregnancy with Gleevec leads to the restoration of their lobuloalveolar development and ability to lactate in subsequent pregnancies in absence of Gleevec treatment. In addition, these mice do not develop mammary tumors. We investigated the mechanism for Gleevec regulation of Notch signaling and found that Gleevec treatment results in a loss of Int3 protein but not of Int3 mRNA in HC11 mouse mammary epithelial cells expressing Int3. The addition of MG-132, a proteasome inhibitor, shows increased ubiquitination of Int3 in the presence of Gleevec. Thus, Gleevec affects the stability of Int3 by promoting the degradation of Int3 via E3 ubiquitin ligases targeting it for the proteasome degradation. Gleevec is a tyrosine kinase inhibitor that acts on c-Kit and PDGFR. Therefore, we investigated the downstream substrate kinase GSK3β to ascertain the possible role that this kinase might play in the stability of Int3. Data show that Gleevec degradation of Int3 is GSK3β dependent. We have expanded our study of the effects Gleevec has on tumorigenesis of other oncogenes. We have found that anchorage-independent growth of HC11-c-Myc cells as well as tumor growth in nude mice is inhibited by Gleevec treatment. As with Int3, Gleevec treatment appears to destabilize the c-Myc protein but not mRNA. These results indicate that Gleevec could be a potential therapeutic drug for patients bearing Notch4 and/or c-Myc positive breast carcinomas.
Collapse
Affiliation(s)
- Robert Callahan
- Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Barry A Chestnut
- Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Ahmed Raafat
- Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| |
Collapse
|
35
|
Harris PWR, Squire C, Young PG, Brimble MA. Chemical synthesis of γ-secretase activating protein using pseudoglutamines as ligation sites. Biopolymers 2016; 104:37-45. [PMID: 25523549 DOI: 10.1002/bip.22600] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/07/2014] [Accepted: 12/08/2014] [Indexed: 01/13/2023]
Abstract
The chemical synthesis of analogue of a novel γ-secretase activating protein, which may play a pivotal role in the formation of amyloid peptides, the precursor to Alzheimer's disease, is described. The linear polypeptide sequence, consisting of 121 amino acids was assembled from four unprotected peptide building blocks using a convergent ligation-based synthesis. A strategic mutation of three glutamine residues to cysteine enabled the ligations, and the cysteines were subsequently converted to pseudoglutamines, to mimic the native glutamine. The full length unfolded protein was obtained in milligram amounts and was demonstrated to be homogeneous by liquid chromatography and mass spectrometry.
Collapse
Affiliation(s)
- Paul W R Harris
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland, 1010, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland, 1010, New Zealand; School of Biological Sciences, The University of Auckland, 3A Symonds Street, Auckland, 1010, New Zealand
| | | | | | | |
Collapse
|
36
|
Part 3: Notch-sparing γ-secretase inhibitors: SAR studies of 2-substituted aminopyridopyrimidinones. Bioorg Med Chem Lett 2016; 26:2138-41. [PMID: 27038496 DOI: 10.1016/j.bmcl.2016.03.077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 03/21/2016] [Accepted: 03/22/2016] [Indexed: 11/23/2022]
Abstract
In search for novel lead compounds as γ-secretase inhibitors, analogs of aminopyrido[2,3-d]pyrimidin-7-ones (I) were synthesized and evaluated for inhibitory effects on amyloid-β-peptide production and cleavage of the Notch1 receptor mediated by γ-secretase. Selected pyridopyrimidines, such as 1, 8, 9, 10, 11 and 16 are γ-secretase inhibitors that did not have an effect on Notch1 receptor processing.
Collapse
|
37
|
Marples B, McGee M, Callan S, Bowen SE, Thibodeau BJ, Michael DB, Wilson GD, Maddens ME, Fontanesi J, Martinez AA. Cranial irradiation significantly reduces beta amyloid plaques in the brain and improves cognition in a murine model of Alzheimer’s Disease (AD). Radiother Oncol 2016; 118:43-51. [DOI: 10.1016/j.radonc.2015.10.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 09/29/2015] [Accepted: 10/19/2015] [Indexed: 10/22/2022]
|
38
|
Barage SH, Sonawane KD. Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer's disease. Neuropeptides 2015; 52:1-18. [PMID: 26149638 DOI: 10.1016/j.npep.2015.06.008] [Citation(s) in RCA: 399] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 06/24/2015] [Accepted: 06/24/2015] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease is an irreversible, progressive neurodegenerative disorder. Various therapeutic approaches are being used to improve the cholinergic neurotransmission, but their role in AD pathogenesis is still unknown. Although, an increase in tau protein concentration in CSF has been described in AD, but several issues remains unclear. Extensive and accurate analysis of CSF could be helpful to define presence of tau proteins in physiological conditions, or released during the progression of neurodegenerative disease. The amyloid cascade hypothesis postulates that the neurodegeneration in AD caused by abnormal accumulation of amyloid beta (Aβ) plaques in various areas of the brain. The amyloid hypothesis has continued to gain support over the last two decades, particularly from genetic studies. Therefore, current research progress in several areas of therapies shall provide an effective treatment to cure this devastating disease. This review critically evaluates general biochemical and physiological functions of Aβ directed therapeutics and their relevance.
Collapse
Affiliation(s)
- Sagar H Barage
- Department of Biotechnology, Shivaji University, Kolhapur 416004, Maharashtra (M.S.), India
| | - Kailas D Sonawane
- Structural Bioinformatics Unit, Department of Biochemistry, Shivaji University, Kolhapur 416004, Maharashtra (M.S.), India; Department of Microbiology, Shivaji University, Kolhapur 416004, Maharashtra (M.S.), India.
| |
Collapse
|
39
|
Inoue M, Hur JY, Kihara T, Teranishi Y, Yamamoto NG, Ishikawa T, Wiehager B, Winblad B, Tjernberg LO, Schedin-Weiss S. Human brain proteins showing neuron-specific interactions with γ-secretase. FEBS J 2015; 282:2587-99. [DOI: 10.1111/febs.13303] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 03/16/2015] [Accepted: 04/17/2015] [Indexed: 11/26/2022]
Affiliation(s)
- Mitsuhiro Inoue
- Karolinska Institutet; Department of Neurobiology; Care Sciences and Society; Center for Alzheimer Research; Division for Neurogeriatrics; Huddinge Sweden
| | - Ji-Yeun Hur
- Karolinska Institutet; Department of Neurobiology; Care Sciences and Society; Center for Alzheimer Research; Division for Neurogeriatrics; Huddinge Sweden
| | - Takahiro Kihara
- Karolinska Institutet; Department of Neurobiology; Care Sciences and Society; Center for Alzheimer Research; Division for Neurogeriatrics; Huddinge Sweden
| | - Yasuhiro Teranishi
- Karolinska Institutet; Department of Neurobiology; Care Sciences and Society; Center for Alzheimer Research; Division for Neurogeriatrics; Huddinge Sweden
| | - Natsuko G. Yamamoto
- Karolinska Institutet; Department of Neurobiology; Care Sciences and Society; Center for Alzheimer Research; Division for Neurogeriatrics; Huddinge Sweden
| | - Taizo Ishikawa
- Karolinska Institutet; Department of Neurobiology; Care Sciences and Society; Center for Alzheimer Research; Division for Neurogeriatrics; Huddinge Sweden
| | - Birgitta Wiehager
- Karolinska Institutet; Department of Neurobiology; Care Sciences and Society; Center for Alzheimer Research; Division for Neurogeriatrics; Huddinge Sweden
| | - Bengt Winblad
- Karolinska Institutet; Department of Neurobiology; Care Sciences and Society; Center for Alzheimer Research; Division for Neurogeriatrics; Huddinge Sweden
| | - Lars O. Tjernberg
- Karolinska Institutet; Department of Neurobiology; Care Sciences and Society; Center for Alzheimer Research; Division for Neurogeriatrics; Huddinge Sweden
| | - Sophia Schedin-Weiss
- Karolinska Institutet; Department of Neurobiology; Care Sciences and Society; Center for Alzheimer Research; Division for Neurogeriatrics; Huddinge Sweden
| |
Collapse
|
40
|
Chu J, Li JG, Hoffman NE, Stough AM, Madesh M, Praticò D. Regulation of gamma-secretase activating protein by the 5Lipoxygenase: in vitro and in vivo evidence. Sci Rep 2015; 5:11086. [PMID: 26076991 PMCID: PMC4468577 DOI: 10.1038/srep11086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/14/2015] [Indexed: 11/10/2022] Open
Abstract
The formation of Aβ is directly controlled by the γ-secretase complex and its activator, γ-secretase activating protein (GSAP). GSAP derives from a C-terminal fragment of a larger precursor protein via a caspase-3 mediated cleavage. However, the mechanism regulating this process remains unknown. Here we provide in vitro experimental evidence that 5-Lipoxygenase (5LO) is as an endogenous regulator for GSAP formation, but not for other known γ-secretase modulators, by directly and specifically activating caspase-3. These results were confirmed in vivo by using transgenic mouse models of Alzheimer’s disease in which 5LO level and activity were modulated genetically or pharmacologically. Taken together, our findings demonstrate that GSAP cleavage via caspase-3 is regulated and depend upon the availability of 5LO further establishing this protein as an attractive and viable therapeutic target for Alzheimer’s disease.
Collapse
Affiliation(s)
- Jin Chu
- Department of Pharmacology, Center for Translational Medicine Temple University School of Medicine, Philadelphia, PA, USA
| | - Jian-Guo Li
- Department of Pharmacology, Center for Translational Medicine Temple University School of Medicine, Philadelphia, PA, USA
| | - Nicholas E Hoffman
- Department of Biochemistry, Center for Translational Medicine Temple University School of Medicine, Philadelphia, PA, USA
| | - Alexandra M Stough
- Department of Biochemistry, Center for Translational Medicine Temple University School of Medicine, Philadelphia, PA, USA
| | - Muniswamy Madesh
- Department of Biochemistry, Center for Translational Medicine Temple University School of Medicine, Philadelphia, PA, USA
| | - Domenico Praticò
- Department of Pharmacology, Center for Translational Medicine Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
41
|
Kong L, Hu Y, Yao Y, Jiao Y, Li S, Yang J. The Coumarin Derivative Osthole Stimulates Adult Neural Stem Cells, Promotes Neurogenesis in the Hippocampus, and Ameliorates Cognitive Impairment in APP/PS1 Transgenic Mice. Biol Pharm Bull 2015; 38:1290-301. [DOI: 10.1248/bpb.b15-00142] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Yu Hu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Yingjia Yao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Yanan Jiao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Shaoheng Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| | - Jingxian Yang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine
| |
Collapse
|
42
|
Wolfe MS. Unlocking truths of γ-secretase in Alzheimer's disease: what is the translational potential? FUTURE NEUROLOGY 2014; 9:419-429. [PMID: 26146489 DOI: 10.2217/fnl.14.35] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Considerable evidence, particularly from genetics, points to the aggregation-prone amyloid β-peptide as a pathogenic entity in Alzheimer's disease. Hence, the proteases that produce this peptide from its precursor protein have been prime targets for the development of potential therapeutics. One of these proteases, γ-secretase, has been a particular focus. Many inhibitors and modulators of this membrane-embedded protease complex have been identified, with some brought into late-stage clinical trials, where they have spectacularly failed. The reasons for these failures will be discussed, along with recent findings on the mechanism of γ-secretase and of Alzheimer-causing mutations that may suggest new strategies for targeting this enzyme.
Collapse
Affiliation(s)
- Michael S Wolfe
- Center for Neurologic Disease, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Tel.: +1 617 525 5511
| |
Collapse
|
43
|
Tenreiro S, Eckermann K, Outeiro TF. Protein phosphorylation in neurodegeneration: friend or foe? Front Mol Neurosci 2014; 7:42. [PMID: 24860424 PMCID: PMC4026737 DOI: 10.3389/fnmol.2014.00042] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/22/2014] [Indexed: 12/15/2022] Open
Abstract
Protein misfolding and aggregation is a common hallmark in neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and fronto-temporal dementia (FTD). In these disorders, the misfolding and aggregation of specific proteins occurs alongside neuronal degeneration in somewhat specific brain areas, depending on the disorder and the stage of the disease. However, we still do not fully understand the mechanisms governing protein aggregation, and whether this constitutes a protective or detrimental process. In PD, alpha-synuclein (aSyn) forms protein aggregates, known as Lewy bodies, and is phosphorylated at serine 129. Other residues have also been shown to be phosphorylated, but the significance of phosphorylation in the biology and pathophysiology of the protein is still controversial. In AD and in FTD, hyperphosphorylation of tau protein causes its misfolding and aggregation. Again, our understanding of the precise consequences of tau phosphorylation in the biology and pathophysiology of the protein is still limited. Through the use of a variety of model organisms and technical approaches, we are now gaining stronger insight into the effects of phosphorylation in the behavior of these proteins. In this review, we cover recent findings in the field and discuss how targeting phosphorylation events might be used for therapeutic intervention in these devastating diseases of the nervous system.
Collapse
Affiliation(s)
- Sandra Tenreiro
- Cell and Molecular Neuroscience Unit, Instituto de Medicina Molecular Lisboa, Portugal
| | - Katrin Eckermann
- Department of Neurology, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen Göttingen, Germany
| | - Tiago F Outeiro
- Cell and Molecular Neuroscience Unit, Instituto de Medicina Molecular Lisboa, Portugal ; Instituto de Fisiologia, Faculdade de Medicina da Universidade de Lisboa Lisboa, Portugal ; Department of NeuroDegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen Göttingen, Germany
| |
Collapse
|
44
|
Berberine attenuates axonal transport impairment and axonopathy induced by Calyculin A in N2a cells. PLoS One 2014; 9:e93974. [PMID: 24713870 PMCID: PMC3979860 DOI: 10.1371/journal.pone.0093974] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/11/2014] [Indexed: 11/19/2022] Open
Abstract
Berberine is a primary component of the most functional extracts of Coptidis rhizome used in traditional Chinese medicine for centuries. Recent reports indicate that Berberine has the potential to prevent and treat Alzheimer's disease (AD). The previous studies reported that Calyculin A (CA) impaired the axonal transport in neuroblastoma-2a (N2a) cells. Berberine attenuated tau hyperphosphorylation and cytotoxicity induced by CA. Our study aimed at investigating the effects of Berberine on the axonal transport impairment induced by CA in N2a cells. The results showed that Berberine could protect the cell from CA -induced toxicity in metabolism and viability, as well as hyperphosphorylation of tau and neurofilaments (NFs). Furthermore, Berberine could reverse CA-induced axonal transport impairment significantly. Berberine also partially reversed the phosphorylation of the catalytic subunit of PP-2A at Tyrosine 307, a crucial site negatively regulating the activity of PP-2A, and reduced the levels of malondialdehyde and the activity of superoxide dismutase, markers of oxidative stress, induced by CA. The present work for the first time demonstrates that Berberine may play a role in protecting against CA-induced axonal transport impairment by modulating the activity of PP-2A and oxidative stress. Our findings also suggest that Berberine may be a potential therapeutic drug for AD.
Collapse
|
45
|
Kerridge C, Belyaev ND, Nalivaeva NN, Turner AJ. The Aβ-clearance protein transthyretin, like neprilysin, is epigenetically regulated by the amyloid precursor protein intracellular domain. J Neurochem 2014; 130:419-31. [PMID: 24528201 DOI: 10.1111/jnc.12680] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 01/23/2014] [Accepted: 02/03/2014] [Indexed: 12/20/2022]
Abstract
Proteolytic cleavage of the amyloid precursor protein (APP) by the successive actions of β- and γ-secretases generates several biologically active metabolites including the amyloid β-peptide (Aβ) and the APP intracellular domain (AICD). By analogy with the Notch signalling pathway, AICD has been proposed to play a role in transcriptional regulation. Among the cohort of genes regulated by AICD is the Aβ-degrading enzyme neprilysin (NEP). AICD binds to the NEP promoter causing transcriptional activation by competitive replacement with histone deacetylases (HDACs) leading to increased levels of NEP activity and hence increased Aβ clearance. We now show that the Aβ-clearance protein transthyretin (TTR) is also epigenetically up-regulated by AICD. Like NEP regulation, AICD derived specifically from the neuronal APP isoform, APP695 , binds directly to the TTR promoter displacing HDAC1 and HDAC3. Cell treatment with the tyrosine kinase inhibitor Gleevec (imatinib) or with the alkalizing agent NH4 Cl causes an accumulation of 'functional' AICD capable of up-regulating both TTR and NEP, leading to a reduction in total cellular Aβ levels. Pharmacological regulation of both NEP and TTR might represent a viable therapeutic target in Alzheimer's disease.
Collapse
Affiliation(s)
- Caroline Kerridge
- School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | | | | | | |
Collapse
|
46
|
Mahul-Mellier AL, Fauvet B, Gysbers A, Dikiy I, Oueslati A, Georgeon S, Lamontanara AJ, Bisquertt A, Eliezer D, Masliah E, Halliday G, Hantschel O, Lashuel HA. c-Abl phosphorylates α-synuclein and regulates its degradation: implication for α-synuclein clearance and contribution to the pathogenesis of Parkinson's disease. Hum Mol Genet 2014; 23:2858-79. [PMID: 24412932 DOI: 10.1093/hmg/ddt674] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Increasing evidence suggests that the c-Abl protein tyrosine kinase could play a role in the pathogenesis of Parkinson's disease (PD) and other neurodegenerative disorders. c-Abl has been shown to regulate the degradation of two proteins implicated in the pathogenesis of PD, parkin and α-synuclein (α-syn). The inhibition of parkin's neuroprotective functions is regulated by c-Abl-mediated phosphorylation of parkin. However, the molecular mechanisms by which c-Abl activity regulates α-syn toxicity and clearance remain unknown. Herein, using NMR spectroscopy, mass spectrometry, in vitro enzymatic assays and cell-based studies, we established that α-syn is a bona fide substrate for c-Abl. In vitro studies demonstrate that c-Abl directly interacts with α-syn and catalyzes its phosphorylation mainly at tyrosine 39 (pY39) and to a lesser extent at tyrosine 125 (pY125). Analysis of human brain tissues showed that pY39 α-syn is detected in the brains of healthy individuals and those with PD. However, only c-Abl protein levels were found to be upregulated in PD brains. Interestingly, nilotinib, a specific inhibitor of c-Abl kinase activity, induces α-syn protein degradation via the autophagy and proteasome pathways, whereas the overexpression of α-syn in the rat midbrains enhances c-Abl expression. Together, these data suggest that changes in c-Abl expression, activation and/or c-Abl-mediated phosphorylation of Y39 play a role in regulating α-syn clearance and contribute to the pathogenesis of PD.
Collapse
|
47
|
Olsson B, Legros L, Guilhot F, Strömberg K, Smith J, Livesey FJ, Wilson DH, Zetterberg H, Blennow K. Imatinib treatment and Aβ42 in humans. Alzheimers Dement 2013; 10:S374-80. [PMID: 24331439 DOI: 10.1016/j.jalz.2013.08.283] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 08/16/2013] [Accepted: 08/30/2013] [Indexed: 10/25/2022]
Abstract
BACKGROUND The first-line treatment in chronic myeloid leukemia (CML), imatinib, has been shown to decrease the production of amyloid-β (Aβ) in vitro and in animal studies. However, whether imatinib has this effect in humans is not known. METHODS Plasma levels of Aβ42 were analyzed in sequential samples from CML patients treated with imatinib (n=51). The effect of imatinib on Aβ production was also investigated in human embryonic kidney 293 (HEK293) cells overexpressing the amyloid precursor protein (APP) with the Swedish mutation, in mouse primary cortical neurons and in human Down syndrome embryonic stem-cell-derived cortical neurons. RESULTS Twelve months of imatinib treatment did not lower plasma Aβ42 levels in CML patients, and imatinib treatment did not lead to less Aβ42 production in any of the in vitro models whereas β- and γ-secretase inhibitors did. CONCLUSION These data question the previously described role of imatinib in inhibiting amyloidogenic APP processing and as a drug candidate for AD.
Collapse
Affiliation(s)
- Bob Olsson
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.
| | - Laurence Legros
- Service d'Hématologie Clinique, Hôpital de l'Archet, Nice, France
| | | | | | - James Smith
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Frederick J Livesey
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| |
Collapse
|
48
|
Appleby BS, Nacopoulos D, Milano N, Zhong K, Cummings JL. A review: treatment of Alzheimer's disease discovered in repurposed agents. Dement Geriatr Cogn Disord 2013; 35:1-22. [PMID: 23307039 DOI: 10.1159/000345791] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/13/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND/AIMS Many compounds that have already been approved for alternate diagnoses have been studied in relation to Alzheimer's disease (AD). The purpose of this review is to summarize these studies and discuss the rationale and benefits of repurposing drugs for AD treatment. METHODS Studies of drugs related to AD treatment that were relevant to a disease-modifying mechanism of action (MOA) and are already approved by the Food and Drug Administration for non-AD diagnoses were collected from PubMed. RESULTS Many drugs already approved for the treatment of other diseases have been studied in relation to AD treatment. Numerous drugs with known toxicity profiles have the potential to be repurposed as a treatment for AD. CONCLUSION Known MOA, toxicology, and pharmacodynamic profiles would accelerate the process and increase the odds of finding a more timely disease-modifying treatment for AD.
Collapse
Affiliation(s)
- Brian S Appleby
- Cleveland Clinic Lou Ruvo Center for Brain Health, Cleveland, OH, USA
| | | | | | | | | |
Collapse
|
49
|
Gandy S, DeKosky ST. Toward the treatment and prevention of Alzheimer's disease: rational strategies and recent progress. Annu Rev Med 2013; 64:367-83. [PMID: 23327526 DOI: 10.1146/annurev-med-092611-084441] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Alzheimer's disease (AD) is the major cause of late-life brain failure. In the past 25 years, autosomal dominant forms of AD were found to be primariy attributable to mutations in one of two presenilins, polytopic proteins that contain the catalytic site of the γ-secretase protease that releases the amyloid beta (Aβ) peptide. Some familial AD is also due to mutations in the amyloid precursor protein (APP), but recently a mutation in APP was discovered that reduces Aβ generation and is protective against AD, further implicating amyloid metabolism. Prion-like seeding of amyloid fibrils and neurofibrillary tangles has been invoked to explain the stereotypical spread of AD within the brain. Treatment trials with anti-Aβ antibodies have shown target engagement, if not significant treatment effects. Attention is increasingly focused on presymptomatic intervention, because Aβ mismetabolism begins up to 25 years before symptoms begin. AD trials deriving from new biological information involve extraordinary international collaboration and may hold the best hope for success in the fight against AD.
Collapse
Affiliation(s)
- Sam Gandy
- Mount Sinai School of Medicine and James J. Peters Veterans Affairs Medical Center, New York, New York 10029, USA.
| | | |
Collapse
|
50
|
De Strooper B, Iwatsubo T, Wolfe MS. Presenilins and γ-secretase: structure, function, and role in Alzheimer Disease. Cold Spring Harb Perspect Med 2013; 2:a006304. [PMID: 22315713 DOI: 10.1101/cshperspect.a006304] [Citation(s) in RCA: 342] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Presenilins were first discovered as sites of missense mutations responsible for early-onset Alzheimer disease (AD). The encoded multipass membrane proteins were subsequently found to be the catalytic components of γ-secretases, membrane-embedded aspartyl protease complexes responsible for generating the carboxyl terminus of the amyloid β-protein (Aβ) from the amyloid protein precursor (APP). The protease complex also cleaves a variety of other type I integral membrane proteins, most notably the Notch receptor, signaling from which is involved in many cell differentiation events. Although γ-secretase is a top target for developing disease-modifying AD therapeutics, interference with Notch signaling should be avoided. Compounds that alter Aβ production by γ-secretase without affecting Notch proteolysis and signaling have been identified and are currently at various stages in the drug development pipeline.
Collapse
Affiliation(s)
- Bart De Strooper
- Center for Human Genetics, Leuven Institute for Neurodegenerative Diseases, KULeuven, 3000 Leuven, Belgium; Department of Molecular and Developmental Genetics, VIB, 3000, Leuven, Belgium
| | | | | |
Collapse
|