1
|
Yao D, Li T, Yu L, Hu M, He Y, Zhang R, Wu J, Li S, Kuang W, Yang X, Liu G, Xie Y. Selective degradation of hyperphosphorylated tau by proteolysis-targeting chimeras ameliorates cognitive function in Alzheimer's disease model mice. Front Pharmacol 2024; 15:1351792. [PMID: 38919259 PMCID: PMC11196765 DOI: 10.3389/fphar.2024.1351792] [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: 12/07/2023] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most common chronic neurodegenerative diseases. Hyperphosphorylated tau plays an indispensable role in neuronal dysfunction and synaptic damage in AD. Proteolysis-targeting chimeras (PROTACs) are a novel type of chimeric molecule that can degrade target proteins by inducing their polyubiquitination. This approach has shown promise for reducing tau protein levels, which is a potential therapeutic target for AD. Compared with traditional drug therapies, the use of PROTACs to reduce tau levels may offer a more specific and efficient strategy for treating AD, with fewer side effects. In the present study, we designed and synthesized a series of small-molecule PROTACs to knock down tau protein. Of these, compound C8 was able to lower both total and phosphorylated tau levels in HEK293 cells with stable expression of wild-type full-length human tau (termed HEK293-htau) and htau-overexpressed mice. Western blot findings indicated that C8 degraded tau protein through the ubiquitin-proteasome system in a time-dependent manner. In htau-overexpressed mice, the results of both the novel object recognition and Morris water maze tests revealed that C8 markedly improved cognitive function. Together, our findings suggest that the use of the small-molecule PROTAC C8 to degrade phosphorylated tau may be a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Dongping Yao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Ting Li
- Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, The Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Mingxing Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Ye He
- Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, The Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruiming Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Junjie Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Shuoyuan Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Weihong Kuang
- Department of Psychiatry and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xifei Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020–2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Gongping Liu
- Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, The Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yongmei Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| |
Collapse
|
2
|
Chu D, Yang X, Wang J, Zhou Y, Gu JH, Miao J, Wu F, Liu F. Tau truncation in the pathogenesis of Alzheimer's disease: a narrative review. Neural Regen Res 2024; 19:1221-1232. [PMID: 37905868 PMCID: PMC11467920 DOI: 10.4103/1673-5374.385853] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/07/2023] [Accepted: 07/25/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Alzheimer's disease is characterized by two major neuropathological hallmarks-the extracellular β-amyloid plaques and intracellular neurofibrillary tangles consisting of aggregated and hyperphosphorylated Tau protein. Recent studies suggest that dysregulation of the microtubule-associated protein Tau, especially specific proteolysis, could be a driving force for Alzheimer's disease neurodegeneration. Tau physiologically promotes the assembly and stabilization of microtubules, whereas specific truncated fragments are sufficient to induce abnormal hyperphosphorylation and aggregate into toxic oligomers, resulting in them gaining prion-like characteristics. In addition, Tau truncations cause extensive impairments to neural and glial cell functions and animal cognition and behavior in a fragment-dependent manner. This review summarizes over 60 proteolytic cleavage sites and their corresponding truncated fragments, investigates the role of specific truncations in physiological and pathological states of Alzheimer's disease, and summarizes the latest applications of strategies targeting Tau fragments in the diagnosis and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xingyue Yang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Jing Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong, Jiangsu Province, China
| | - Jin-Hua Gu
- Department of Clinical Pharmacy, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province, China
| | - Jin Miao
- Laboratory of Animal Center, Nantong University, Nantong, Jiangsu Province, China
| | - Feng Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu Province, China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
3
|
Lin P, Zhang B, Yang H, Yang S, Xue P, Chen Y, Yu S, Zhang J, Zhang Y, Chen L, Fan C, Li F, Ling D. An artificial protein modulator reprogramming neuronal protein functions. Nat Commun 2024; 15:2039. [PMID: 38448420 PMCID: PMC10917760 DOI: 10.1038/s41467-024-46308-6] [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: 08/09/2023] [Accepted: 02/14/2024] [Indexed: 03/08/2024] Open
Abstract
Reversible protein phosphorylation, regulated by protein phosphatases, fine-tunes target protein function and plays a vital role in biological processes. Dysregulation of this process leads to aberrant post-translational modifications (PTMs) and contributes to disease development. Despite the widespread use of artificial catalysts as enzyme mimetics, their direct modulation of proteins remains largely unexplored. To address this gap and enable the reversal of aberrant PTMs for disease therapy, we present the development of artificial protein modulators (APROMs). Through atomic-level engineering of heterogeneous catalysts with asymmetric catalytic centers, these modulators bear structural similarities to protein phosphatases and exhibit remarkable ability to destabilize the bridging μ3-hydroxide. This activation of catalytic centers enables spontaneous hydrolysis of phospho-substrates, providing precise control over PTMs. Notably, APROMs, with protein phosphatase-like characteristics, catalytically reprogram the biological function of α-synuclein by directly hydrolyzing hyperphosphorylated α-synuclein. Consequently, synaptic function is reinforced in Parkinson's disease. Our findings offer a promising avenue for reprogramming protein function through de novo PTMs strategy.
Collapse
Affiliation(s)
- Peihua Lin
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, School of Biomedical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240, China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Zhang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, School of Biomedical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai, 201210, China
| | - Hongli Yang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shengfei Yang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Pengpeng Xue
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ying Chen
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shiyi Yu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jichao Zhang
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, China
| | - Yixiao Zhang
- In-situ Center for Physical Sciences, School of Chemistry and Chemical Engineering, Shanghai Electrochemical Energy Device Research Center (SEED), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Liwei Chen
- In-situ Center for Physical Sciences, School of Chemistry and Chemical Engineering, Shanghai Electrochemical Energy Device Research Center (SEED), Shanghai Jiao Tong University, Shanghai, 200240, China
- Future Battery Research Center, Global Institute of Future Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chunhai Fan
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, School of Biomedical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Fangyuan Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310009, China.
| | - Daishun Ling
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, School of Biomedical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240, China.
- World Laureates Association (WLA) Laboratories, Shanghai, 201210, China.
| |
Collapse
|
4
|
Davidowitz EJ, Lopez P, Jimenez H, Adrien L, Davies P, Moe JG. Small molecule inhibitor of tau self-association in a mouse model of tauopathy: A preventive study in P301L tau JNPL3 mice. PLoS One 2023; 18:e0286523. [PMID: 37556474 PMCID: PMC10411817 DOI: 10.1371/journal.pone.0286523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/17/2023] [Indexed: 08/11/2023] Open
Abstract
Advances in tau biology and the difficulties of amyloid-directed immunotherapeutics have heightened interest in tau as a target for small molecule drug discovery for neurodegenerative diseases. Here, we evaluated OLX-07010, a small molecule inhibitor of tau self-association, for the prevention of tau aggregation. The primary endpoint of the study was statistically significant reduction of insoluble tau aggregates in treated JNPL3 mice compared with Vehicle-control mice. Secondary endpoints were dose-dependent reduction of insoluble tau aggregates, reduction of phosphorylated tau, and reduction of soluble tau. This study was performed in JNPL3 mice, which are representative of inherited forms of 4-repeat tauopathies with the P301L tau mutation (e.g., progressive supranuclear palsy and frontotemporal dementia). The P301L mutation makes tau prone to aggregation; therefore, JNPL3 mice present a more challenging target than mouse models of human tau without mutations. JNPL3 mice were treated from 3 to 7 months of age with Vehicle, 30 mg/kg compound dose, or 40 mg/kg compound dose. Biochemical methods were used to evaluate self-associated tau, insoluble tau aggregates, total tau, and phosphorylated tau in the hindbrain, cortex, and hippocampus. The Vehicle group had higher levels of insoluble tau in the hindbrain than the Baseline group; treatment with 40 mg/kg compound dose prevented this increase. In the cortex, the levels of insoluble tau were similar in the Baseline and Vehicle groups, indicating that the pathological phenotype of these mice was beginning to emerge at the study endpoint and that there was a delay in the development of the phenotype of the model as originally characterized. No drug-related adverse effects were observed during the 4-month treatment period.
Collapse
Affiliation(s)
- Eliot J. Davidowitz
- Oligomerix, Inc., White Plains, NY, United States of America
- Oligomerix, Inc., Bronx, NY, United States of America
| | | | - Heidy Jimenez
- The Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States of America
| | - Leslie Adrien
- The Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States of America
| | - Peter Davies
- The Litwin-Zucker Research Center for the Study of Alzheimer’s Disease, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States of America
| | - James G. Moe
- Oligomerix, Inc., White Plains, NY, United States of America
- Oligomerix, Inc., Bronx, NY, United States of America
| |
Collapse
|
5
|
Rimal S, Tantray I, Li Y, Pal Khaket T, Li Y, Bhurtel S, Li W, Zeng C, Lu B. Reverse electron transfer is activated during aging and contributes to aging and age-related disease. EMBO Rep 2023; 24:e55548. [PMID: 36794623 PMCID: PMC10074108 DOI: 10.15252/embr.202255548] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 12/18/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Mechanisms underlying the depletion of NAD+ and accumulation of reactive oxygen species (ROS) in aging and age-related disorders remain poorly defined. We show that reverse electron transfer (RET) at mitochondrial complex I, which causes increased ROS production and NAD+ to NADH conversion and thus lowered NAD+ /NADH ratio, is active during aging. Genetic or pharmacological inhibition of RET decreases ROS production and increases NAD+ /NADH ratio, extending the lifespan of normal flies. The lifespan-extending effect of RET inhibition is dependent on NAD+ -dependent Sirtuin, highlighting the importance of NAD+ /NADH rebalance, and on longevity-associated Foxo and autophagy pathways. RET and RET-induced ROS and NAD+ /NADH ratio changes are prominent in human induced pluripotent stem cell (iPSC) model and fly models of Alzheimer's disease (AD). Genetic or pharmacological inhibition of RET prevents the accumulation of faulty translation products resulting from inadequate ribosome-mediated quality control, rescues relevant disease phenotypes, and extends the lifespan of Drosophila and mouse AD models. Deregulated RET is therefore a conserved feature of aging, and inhibition of RET may open new therapeutic opportunities in the context of aging and age-related diseases including AD.
Collapse
Affiliation(s)
- Suman Rimal
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | - Ishaq Tantray
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | - Yu Li
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | | | - Yanping Li
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | - Sunil Bhurtel
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | - Wen Li
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | | | - Bingwei Lu
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| |
Collapse
|
6
|
Li L, Miao J, Jiang Y, Dai CL, Iqbal K, Liu F, Chu D. Passive immunization inhibits tau phosphorylation and improves recognition learning and memory in 3xTg-AD mice. Exp Neurol 2023; 362:114337. [PMID: 36717015 DOI: 10.1016/j.expneurol.2023.114337] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/05/2023] [Accepted: 01/24/2023] [Indexed: 01/30/2023]
Abstract
Tau pathology is essential in the pathogenesis of Alzheimer's disease (AD) and related tauopathies. Tau immunotherapy aimed at reducing the progression of tau pathology provides a potential therapeutic strategy for treating these diseases. By screening monoclonal antibodies 43D, 63B, 39E10, and 77G7 that recognize epitopes ranging from tau's N-terminus to C-terminus, we found the 77G7, which targets the microtubule-binding domain promoted tau clearance in a dose-dependent manner by entering neuronal cells in vitro. Intra-cerebroventricular injection of 77G7 antibody reduced tau levels in the wild-type FVB mouse brain. Without influencing the levels of detergent-insoluble and aggregated tau, intravenous injection of 77G7 reduced tau hyperphosphorylation in the brain and improved novel object recognition but not spatial learning and memory in 15-18-month-old 3xTg-AD mice. These studies suggest that epitopes recognized by tau antibodies are crucial for the efficacy of immunotherapy. Immunization with antibody 77G7 provides a novel potential opportunity for tau-directed immunotherapy of AD and related tauopathies.
Collapse
Affiliation(s)
- Longfei Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Jin Miao
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA; Laboratory Animal Center, Nantong University, Nantong 226001, China
| | - Yanli Jiang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Chun-Ling Dai
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA.
| | - Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA.
| |
Collapse
|
7
|
Mota IFL, de Lima LS, Santana BDM, Gobbo GDAM, Bicca JVML, Azevedo JRM, Veras LG, Taveira RDAA, Pinheiro GB, Mortari MR. Alzheimer's Disease: Innovative Therapeutic Approaches Based on Peptides and Nanoparticles. Neuroscientist 2023; 29:78-96. [PMID: 34018874 DOI: 10.1177/10738584211016409] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Alzheimer's disease (AD) is the main cause of dementia in the world and its etiology is not yet fully understood. The pathology of AD is primarily characterized by intracellular neurofibrillary tangles and extracellular amyloid-β plaques. Unfortunately, few treatment options are available, and most treat symptoms, as is the case of acetylcholinesterase inhibitors (IAChE) and N-methyl-d-aspartate receptor antagonists. For more than 20 years pharmaceutical research has targeted the "amyloid cascade hypothesis," but this has not produced meaningful results, leading researchers to focus now on other characteristics of the disease and on multitarget approaches. This review aims to evaluate some new treatments that are being developed and studied. Among these are new treatments based on peptides, which have high selectivity and low toxicity; however, these compounds have a short half-life and encounter challenges when crossing the blood-brain barrier. The present review discusses up-and-coming peptides tested as treatments and explores some nanotechnological strategies to overcome the downsides. These compounds are promising, as they not only act on the symptoms but also aim to prevent progressive neuronal loss.
Collapse
Affiliation(s)
- Isabela F L Mota
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Larissa S de Lima
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Bruna de M Santana
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Giovanna de A M Gobbo
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - João V M L Bicca
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Juliana R M Azevedo
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Letícia G Veras
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Rodrigo de A A Taveira
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Gabriela B Pinheiro
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Márcia R Mortari
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| |
Collapse
|
8
|
Hajjo R, Sabbah DA, Abusara OH, Al Bawab AQ. A Review of the Recent Advances in Alzheimer's Disease Research and the Utilization of Network Biology Approaches for Prioritizing Diagnostics and Therapeutics. Diagnostics (Basel) 2022; 12:diagnostics12122975. [PMID: 36552984 PMCID: PMC9777434 DOI: 10.3390/diagnostics12122975] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is a polygenic multifactorial neurodegenerative disease that, after decades of research and development, is still without a cure. There are some symptomatic treatments to manage the psychological symptoms but none of these drugs can halt disease progression. Additionally, over the last few years, many anti-AD drugs failed in late stages of clinical trials and many hypotheses surfaced to explain these failures, including the lack of clear understanding of disease pathways and processes. Recently, different epigenetic factors have been implicated in AD pathogenesis; thus, they could serve as promising AD diagnostic biomarkers. Additionally, network biology approaches have been suggested as effective tools to study AD on the systems level and discover multi-target-directed ligands as novel treatments for AD. Herein, we provide a comprehensive review on Alzheimer's disease pathophysiology to provide a better understanding of disease pathogenesis hypotheses and decipher the role of genetic and epigenetic factors in disease development and progression. We also provide an overview of disease biomarkers and drug targets and suggest network biology approaches as new tools for identifying novel biomarkers and drugs. We also posit that the application of machine learning and artificial intelligence to mining Alzheimer's disease multi-omics data will facilitate drug and biomarker discovery efforts and lead to effective individualized anti-Alzheimer treatments.
Collapse
Affiliation(s)
- Rima Hajjo
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carlina at Chapel Hill, Chapel Hill, NC 27599, USA
- National Center for Epidemics and Communicable Disease Control, Amman 11118, Jordan
- Correspondence:
| | - Dima A. Sabbah
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan
| | - Osama H. Abusara
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan
| | - Abdel Qader Al Bawab
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan
| |
Collapse
|
9
|
Selvarasu K, Singh AK, Iyaswamy A, Gopalkrishnashetty Sreenivasmurthy S, Krishnamoorthi S, Bera AK, Huang JD, Durairajan SSK. Reduction of kinesin I heavy chain decreases tau hyperphosphorylation, aggregation, and memory impairment in Alzheimer's disease and tauopathy models. Front Mol Biosci 2022; 9:1050768. [PMID: 36387285 PMCID: PMC9641281 DOI: 10.3389/fmolb.2022.1050768] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/10/2022] [Indexed: 08/29/2023] Open
Abstract
Many neurodegenerative diseases, such as Alzheimer's disease (AD) and frontotemporal dementia with Parkinsonism linked to chromosome 17, are characterized by tau pathology. Numerous motor proteins, many of which are involved in synaptic transmission, mediate transport in neurons. Dysfunction in motor protein-mediated neuronal transport mechanisms occurs in several neurodegenerative disorders but remains understudied in AD. Kinesins are the most important molecular motor proteins required for microtubule-dependent transport in neurons, and kinesin-1 is crucial for neuronal transport among all kinesins. Although kinesin-1 is required for normal neuronal functions, the dysfunction of these motor domains leading to neurodegenerative diseases is not fully understood. Here, we reported that the kinesin-I heavy chain (KIF5B), a key molecular motor protein, is involved in tau homeostasis in AD cells and animal models. We found that the levels of KIF5B in P301S tau mice are high. We also found that the knockdown and knockout (KO) of KIFf5B significantly decreased the tau stability, and overexpression of KIF5B in KIF5B-KO cells significantly increased the expression of phosphorylated and total tau levels. This suggested that KIF5B might prevent tau accumulation. By conducting experiments on P301S tau mice, we showed that partially reducing KIF5B levels can reduce hyperphosphorylation of the human tau protein, formation of insoluble aggregates, and memory impairment. Collectively, our results suggested that decreasing KIF5B levels is sufficient to prevent and/or slow down abnormal tau behavior of AD and other tauopathies.
Collapse
Affiliation(s)
- Karthikeyan Selvarasu
- Molecular Mycology and Neurodegenerative Disease Research Laboratory, Department of Microbiology, Central University of Tamil Nadu, Thiruvarur, India
| | - Abhay Kumar Singh
- Molecular Mycology and Neurodegenerative Disease Research Laboratory, Department of Microbiology, Central University of Tamil Nadu, Thiruvarur, India
| | - Ashok Iyaswamy
- Mr. and Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | | | - Senthilkumar Krishnamoorthi
- Centre for Trans-Disciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Chennai, India
| | - Amal Kanti Bera
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Jian-Dong Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Siva Sundara Kumar Durairajan
- Molecular Mycology and Neurodegenerative Disease Research Laboratory, Department of Microbiology, Central University of Tamil Nadu, Thiruvarur, India
| |
Collapse
|
10
|
Durairajan SSK, Selvarasu K, Bera MR, Rajaram K, Iyaswamy A, Li M. Alzheimer's Disease and other Tauopathies: Exploring Efficacy of Medicinal Plant-derived Compounds in Alleviating Tau-mediated Neurodegeneration. Curr Mol Pharmacol 2022; 15:361-379. [PMID: 34488602 DOI: 10.2174/1874467214666210906125318] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/12/2020] [Accepted: 01/27/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD), a major form of dementia, has been reported to affect more than 50 million people worldwide. It is characterized by the presence of amyloid-β (Aβ) plaques and hyperphosphorylated Tau-associated neurofibrillary tangles in the brain. Apart from AD, microtubule (MT)-associated protein Tau is also involved in other neurodegenerative diseases called tauopathies, including Pick's disease, frontotemporal lobar degeneration, progressive supranuclear palsy, and corticobasal degeneration. The recent unsuccessful phase III clinical trials related to Aβ- targeted therapeutic drugs have indicated that alternative targets, such as Tau, should be studied to discover more effective and safer drugs. Recent drug discovery approaches to reduce AD-related Tau pathologies are primarily based on blocking Tau aggregation, inhibiting Tau phosphorylation, compensating impaired Tau function with MT-stabilizing agents, and targeting the degradation pathways in neuronal cells to degrade Tau protein aggregates. Owing to several limitations of the currently available Tau-directed drugs, further studies are required to generate further effective and safer Tau-based disease-modifying drugs. Here, we review the studies focused on medicinal plant- derived compounds capable of modulating the Tau protein, which is significantly elevated and hyperphosphorylated in AD and other tauopathies. We have mainly considered the studies focused on Tau protein as a therapeutic target. We have reviewed several pertinent papers retrieved from PubMed and ScienceDirect using relevant keywords, with a primary focus on the Tau-targeting compounds from medicinal plants. These compounds include indolines, phenolics, flavonoids, coumarins, alkaloids, and iridoids, which have been scientifically proven to be Tau-targeting candidates for the treatment of AD.
Collapse
Affiliation(s)
- Siva Sundara Kumar Durairajan
- Mycobiology and Neurodegenerative Disease Research Lab, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, India
| | - Karthikeyan Selvarasu
- Mycobiology and Neurodegenerative Disease Research Lab, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, India
| | - Minu Rani Bera
- Mycobiology and Neurodegenerative Disease Research Lab, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, India
| | - Kaushik Rajaram
- Mycobiology and Neurodegenerative Disease Research Lab, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, India
| | - Ashok Iyaswamy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Min Li
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| |
Collapse
|
11
|
Li Y, Peng X. Comparison of the force fields on monomeric and fibrillar PHF6 of tau protein. Biophys Chem 2021; 277:106631. [PMID: 34116358 DOI: 10.1016/j.bpc.2021.106631] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/18/2021] [Accepted: 06/01/2021] [Indexed: 11/19/2022]
Abstract
The hexapeptide 306VQIVYK311 (PHF6) plays an important role in the aggregation of Tau protein, which is a hallmark of the Alzheimer's disease (AD). In this article, we systematically compare the effects of eight popular all-atom force fields on the monomeric and fibrillar PHF6 in the molecular dynamics (MD) simulations, which could be helpful in the computer-aided drug design against PHF6. We show that the fibrillar PHF6 prefers β-strand-like structures in all the force fields while the monomer has different structural preferences depending on the force fields. The interactions for stabilizing the fibril are further investigated. In the end, according to the interactions revealed by NMR and the stability of the fibril in the literature, we benchmark the force fields.
Collapse
Affiliation(s)
- Yanchun Li
- Center for Quantum Technology Research, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurements (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China
| | - Xubiao Peng
- Center for Quantum Technology Research, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurements (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China; Beijing Academy of Quantum Information Sciences, Beijing 100193, China.
| |
Collapse
|
12
|
Mahdiabadi S, Momtazmanesh S, Perry G, Rezaei N. Immune modulations and immunotherapies for Alzheimer's disease: a comprehensive review. Rev Neurosci 2021; 33:365-381. [PMID: 34506700 DOI: 10.1515/revneuro-2021-0092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD), the most common cause of dementia, is characterized by progressive cognitive and memory impairment ensued from neuronal dysfunction and eventual death. Intraneuronal deposition of tau proteins and extracellular senile amyloid-β plaques have ruled as the supreme postulations of AD for a relatively long time, and accordingly, a wide range of therapeutics, especially immunotherapies have been implemented. However, none of them resulted in significant positive cognitive outcomes. Especially, the repetitive failure of anti-amyloid therapies proves the inefficiency of the amyloid cascade hypothesis, suggesting that it is time to reconsider this hypothesis. Thus, for the time being, the focus is being shifted to neuroinflammation as a third core pathology in AD. Neuroinflammation was previously considered a result of the two aforementioned phenomena, but new studies suggest that it might play a causal role in the pathogenesis of AD. Neuroinflammation can act as a double-edged sword in the pathogenesis of AD, and the activation of glial cells is indispensable for mediating such attenuating or detrimental effects. The association of immune-related genes polymorphisms with the clinical phenotype of AD as well as the protective effect of anti-inflammatory drugs like nonsteroidal anti-inflammatory drugs supports the possible causal role of neuroinflammation in AD. Here, we comprehensively review immune-based therapeutic approaches toward AD, including monoclonal antibodies and vaccines. We also discuss their efficacy and underlying reasons for shortcomings. Lastly, we highlight the capacity of modulating the neuroimmune interactions and targeting neuroinflammation as a promising opportunity for finding optimal treatments for AD.
Collapse
Affiliation(s)
- Sara Mahdiabadi
- School of Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center, Tehran 1419733151, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran 14194, Iran
| | - Sara Momtazmanesh
- School of Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center, Tehran 1419733151, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran 14194, Iran
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio (UTSA), San Antonio, TX 78249, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center, Tehran 1419733151, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran 14194, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| |
Collapse
|
13
|
Shinohara M, Hirokawa J, Shimodaira A, Tashiro Y, Suzuki K, Gheni G, Fukumori A, Matsubara T, Morishima M, Saito Y, Murayama S, Sato N. ELISA Evaluation of Tau Accumulation in the Brains of Patients with Alzheimer Disease. J Neuropathol Exp Neurol 2021; 80:652-662. [PMID: 34283221 DOI: 10.1093/jnen/nlab047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Despite the routine use of sandwich enzyme-linked immunosorbent assays (ELISAs) for quantifying tau levels in CSF and plasma, tau accumulations in the brains of patients with Alzheimer disease (AD) have rarely been evaluated by this method. Thus, by introducing several tau ELISAs that target different epitopes, we evaluated accumulated tau levels in postmortem brains depending on disease stage, brain areas, and other AD-related changes. Notably, tau levels in insoluble fraction determined by each ELISAs differ depending on the epitopes of antibodies: non-AD control samples yield relatively high signals when an antibody against the N-terminal region of tau is used. On the other hand, ELISAs combining antibodies against the later-middle to C-terminal regions of tau produced substantially increased signals from AD samples, compared to those from non-AD controls. Such ELISAs better distinguish AD and non-AD controls, and the results are more closely associated with Braak neurofibrillary tangles stage, Aβ accumulation, and glial markers. Moreover, these ELISAs can reflect the pattern of tau spread across brain regions. In conclusion, Tau ELISAs that combine antibodies against the later-middle to C-terminal regions of tau can better reflect neuropathological tau accumulation, which would enable to evaluate tau accumulation in the brain at a biochemical level.
Collapse
Affiliation(s)
- Mitsuru Shinohara
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Junko Hirokawa
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Akemi Shimodaira
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Yoshitaka Tashiro
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Kaoru Suzuki
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Ghupurjan Gheni
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Akio Fukumori
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Tomoyasu Matsubara
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Maho Morishima
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Yuko Saito
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Shigeo Murayama
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| | - Naoyuki Sato
- From the Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan (MS, JH, AS, YT, KS, GG, AF, NS).,Department of Aging Neurobiology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan (MS, AF, NS).,Department of Pharmacotherapeutics II, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan (AF).,Department of Neuropathology (The Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Itabashi, Tokyo, Japan (TM, MM, YS, SM).,Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan (SM)
| |
Collapse
|
14
|
Ma D, Huang R, Guo K, Zhao Z, Wei W, Gu L, Li L, Zhang L. Cornel Iridoid Glycoside Protects Against STAT1-Dependent Synapse and Memory Deficits by Increasing N-Methyl-D-aspartate Receptor Expression in a Tau Transgenic Mice. Front Aging Neurosci 2021; 13:671206. [PMID: 34113246 PMCID: PMC8185567 DOI: 10.3389/fnagi.2021.671206] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
P301S transgenic mice are an animal model of tauopathy and Alzheimer’s disease (AD), exhibiting tau pathology and synaptic dysfunction. Cornel iridoid glycoside (CIG) is an active ingredient extracted from Cornus officinalis, a traditional Chinese herb. In the present study, the purpose was to investigate the effects and mechanisms of CIG on tau pathology and synaptic dysfunction using P301S transgenic mice. The results showed that intragastric administration of CIG for 3.5 months improved cognitive impairments and the survival rate of P301S mice. Electrophysiological recordings and transmission electron microscopy study showed that CIG improved synaptic plasticity and increased the ultrastructure and number of synapse. Moreover, CIG increased the expression levels of N-methyl-D-aspartate receptors (NMDAR) subunits GluN1, GluN2A, and GluN2B, and α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) subunit GluA1. We inferred that the major mechanism of CIG involving in the regulation of synaptic dysfunctions was inhibiting the activation of Janus kinase-2 (JAK2)/signal transducer and activator of transcription 1 (STAT1) signaling pathway and alleviating STAT1-induced suppression of NMDAR expressions. Based on our findings, we thought CIG might be a promising candidate for the therapy of tauopathy such as AD.
Collapse
Affiliation(s)
- Denglei Ma
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Department of Pharmacy, Beijing Institute for Brain Disorders, Beijing Engineering Research Center for Nerve System Drugs, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Rui Huang
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Department of Pharmacy, Beijing Institute for Brain Disorders, Beijing Engineering Research Center for Nerve System Drugs, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Kaiwen Guo
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Department of Pharmacy, Beijing Institute for Brain Disorders, Beijing Engineering Research Center for Nerve System Drugs, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zirun Zhao
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, United States
| | - Weipeng Wei
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Department of Pharmacy, Beijing Institute for Brain Disorders, Beijing Engineering Research Center for Nerve System Drugs, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Lihong Gu
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Department of Pharmacy, Beijing Institute for Brain Disorders, Beijing Engineering Research Center for Nerve System Drugs, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Lin Li
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Department of Pharmacy, Beijing Institute for Brain Disorders, Beijing Engineering Research Center for Nerve System Drugs, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Lan Zhang
- Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Department of Pharmacy, Beijing Institute for Brain Disorders, Beijing Engineering Research Center for Nerve System Drugs, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Wu R, Li L, Shi R, Zhou Y, Jin N, Gu J, Tung YC, Liu F, Chu D. Dephosphorylation Passivates the Seeding Activity of Oligomeric Tau Derived From Alzheimer's Brain. Front Mol Neurosci 2021; 14:631833. [PMID: 34054426 PMCID: PMC8155256 DOI: 10.3389/fnmol.2021.631833] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 03/26/2021] [Indexed: 11/13/2022] Open
Abstract
Accumulation of intracellular neurofibrillary tangles (NFTs), which are constituted of abnormally phosphorylated tau, is one of the neuropathological hallmarks of Alzheimer’s disease (AD). The oligomeric aggregates of tau in AD brain (AD O-tau) are believed to trigger NFT spreading by seeding normal tau aggregation as toxic seeds, in a prion-like fashion. Here, we revealed the features of AD O-tau by Western blots using antibodies against various epitopes and determined the effect of dephosphorylation on the seeding activity of AD O-tau by capture and seeded aggregation assays. We found that N-terminal truncated and C-terminalhyperphosphorylated tau species were enriched in AD O-tau. Dephosphorylation of AD O-tau by alkaline phosphatasediminished its activity in capturing tau in vitro and ininducing insoluble aggregates in cultured cells. Our resultssuggested that dephosphorylation passivated the seeding activity ofAD O-tau. Inhibition of phosphorylation may be a potentstrategy to prevent the spreading of tau patho3logy.
Collapse
Affiliation(s)
- Ruozhen Wu
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Longfei Li
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Ruirui Shi
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Yan Zhou
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Nana Jin
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jianlan Gu
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Yunn Chyn Tung
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, United States
| | - Dandan Chu
- NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
16
|
Oberrauch S, Metha JA, Brian ML, Barnes SA, Featherby TJ, Lawrence AJ, Hoyer D, Murawski C, Jacobson LH. Reward motivation and cognitive flexibility in tau null-mutation mice. Neurobiol Aging 2021; 100:106-117. [PMID: 33524848 DOI: 10.1016/j.neurobiolaging.2020.12.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/05/2020] [Accepted: 12/15/2020] [Indexed: 01/21/2023]
Abstract
The reduction of tau or hyperphosphorylated tau (p-tau) has been proposed as a therapeutic strategy for Alzheimer's disease (AD) and frontotemporal dementia (FTD). Cognitive decline and sleep-wake dysregulation seen in AD and FTD patients are mimicked in transgenic and null-mutation mouse models of tauopathy. Alterations in the reward system are additional symptoms of AD and FTD. However, the role of tau in reward processes is not well understood. The present study aimed to examine reward and reward-motivated cognitive processes in male and female tau knockout (tau-/-) and wild-type mice using progressive ratio and reversal learning tasks. Tau-/- mice were heavier, ate more in the home cage, and reached criterion in operant lever training faster than wild-type mice. Tau-/- mice had a higher breakpoint in progressive ratio but were unimpaired in reversal learning or reward sensitivity. These data indicate that tau loss of function alters reward processing. This may help to explain aberrant reward-related behaviors in tauopathy patients and highlights a potentially important area for consideration in the development of anti-tau therapies.
Collapse
Affiliation(s)
- Sara Oberrauch
- The Florey Institute of Neuroscience and Mental Health, Parkville Campus, University of Melbourne, Parkville, Australia; Department of Pharmacology & Therapeutics, School of Medicine, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Jeremy A Metha
- The Florey Institute of Neuroscience and Mental Health, Parkville Campus, University of Melbourne, Parkville, Australia; Department of Pharmacology & Therapeutics, School of Medicine, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia; Department of Finance, Brain, Mind & Markets Laboratory, The University of Melbourne, Melbourne, Victoria, Australia
| | - Maddison L Brian
- The Florey Institute of Neuroscience and Mental Health, Parkville Campus, University of Melbourne, Parkville, Australia; Department of Pharmacology & Therapeutics, School of Medicine, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Samuel A Barnes
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Travis J Featherby
- The Florey Institute of Neuroscience and Mental Health, Parkville Campus, University of Melbourne, Parkville, Australia
| | - Andrew J Lawrence
- The Florey Institute of Neuroscience and Mental Health, Parkville Campus, University of Melbourne, Parkville, Australia
| | - Daniel Hoyer
- The Florey Institute of Neuroscience and Mental Health, Parkville Campus, University of Melbourne, Parkville, Australia; Department of Pharmacology & Therapeutics, School of Medicine, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Carsten Murawski
- Department of Finance, Brain, Mind & Markets Laboratory, The University of Melbourne, Melbourne, Victoria, Australia
| | - Laura H Jacobson
- The Florey Institute of Neuroscience and Mental Health, Parkville Campus, University of Melbourne, Parkville, Australia; Department of Pharmacology & Therapeutics, School of Medicine, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia; Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health and The University of Melbourne, Victoria, Australia.
| |
Collapse
|
17
|
Gu JL, Liu F. Tau in Alzheimer's Disease: Pathological Alterations and an Attractive Therapeutic Target. Curr Med Sci 2021; 40:1009-1021. [PMID: 33428128 DOI: 10.1007/s11596-020-2282-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/03/2020] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease with two major hallmarks: extracellular amyloid plaques made of amyloid-β (Aβ) and intracellular neurofibrillary tangles (NFTs) of abnormally hyperphosphorylated tau. The number of NFTs correlates positively with the severity of dementia in AD patients. However, there is still no efficient therapy available for AD treatment and prevention so far. A deeper understanding of AD pathogenesis has identified novel strategies for the generation of specific therapies over the past few decades. Several studies have suggested that the prion-like seeding and spreading of tau pathology in the brain may be a key driver of AD. Tau protein is considered as a promising candidate target for the development of therapeutic interventions due to its considerable pathological role in a variety of neurodegenerative disorders. Abnormal tau hyperphosphorylation plays a detrimental pathological role, eventually leading to neurodegeneration. In the present review, we describe the recent research progresses in the pathological mechanisms of tau protein in AD and briefly discuss tau-based therapeutic strategies.
Collapse
Affiliation(s)
- Jian-Lan Gu
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong, 226001, China. .,Co-innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration and Ministry of Education of Jiangsu, Nantong, 226001, China.
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| |
Collapse
|
18
|
Duquette A, Pernègre C, Veilleux Carpentier A, Leclerc N. Similarities and Differences in the Pattern of Tau Hyperphosphorylation in Physiological and Pathological Conditions: Impacts on the Elaboration of Therapies to Prevent Tau Pathology. Front Neurol 2021; 11:607680. [PMID: 33488502 PMCID: PMC7817657 DOI: 10.3389/fneur.2020.607680] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022] Open
Abstract
Tau protein, a neuronal microtubule-associated protein, becomes hyperphosphorylated in several neurodegenerative diseases called tauopathies. Hyperphosphorylation of tau is correlated to its redistribution from the axon to the somato-dendritic compartment at early stages of tauopathies. Interestingly, tau hyperphosphorylation begins in different regions of the brain in each tauopathy. In some regions, both neurons and glial cells develop tau hyperphosphorylation. Tau hyperphosphorylation is also observed in physiological conditions such as hibernation and brain development. In the first section of present article, we will review the spatiotemporal and cellular distribution of hyperphosphorylated tau in the most frequent tauopathies. In the second section, we will compare the pattern of tau hyperphosphorylation in physiological and pathological conditions and discuss the sites that could play a pivotal role in the conversion of non-toxic to toxic forms of hyperphosphorylated tau. Furthermore, we will discuss the role of hyperphosphorylated tau in physiological and pathological conditions and the fact that tau hyperphosphorylation is reversible in physiological conditions but not in a pathological ones. In the third section, we will speculate how the differences and similarities between hyperphosphorylated tau in physiological and pathological conditions could impact the elaboration of therapies to prevent tau pathology. In the fourth section, the different therapeutic approaches using tau as a direct or indirect therapeutic target will be presented.
Collapse
Affiliation(s)
- Antoine Duquette
- Research Center of the University of Montreal Hospital (CRCHUM), Montréal, QC, Canada.,Département de Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Camille Pernègre
- Research Center of the University of Montreal Hospital (CRCHUM), Montréal, QC, Canada.,Département de Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Ariane Veilleux Carpentier
- Research Center of the University of Montreal Hospital (CRCHUM), Montréal, QC, Canada.,Département de Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Nicole Leclerc
- Research Center of the University of Montreal Hospital (CRCHUM), Montréal, QC, Canada.,Département de Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
19
|
Colovati MES, Novais IP, Zampol M, Mendes GD, Cernach MCS, Zanesco A. Interaction between physical exercise and APOE gene polymorphism on cognitive function in older people. ACTA ACUST UNITED AC 2020; 54:e10098. [PMID: 33331535 PMCID: PMC7727114 DOI: 10.1590/1414-431x202010098] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 09/18/2020] [Indexed: 12/27/2022]
Abstract
We aimed to present an overview of the literature regarding the interaction between physical exercise and APOE gene polymorphism on cognitive function, particularly in patients with Alzheimer's disease (AD). Firstly, this review focused on the effect of the physical exercise on cognitive function, regardless of APOE gene polymorphism. Some studies have shown that a high level of cardiorespiratory fitness is associated with less neuronal damage with an improvement in memory score tests whereas other studies failed to detect any association between physical exercise and cognitive improvement either in healthy individuals or patients with AD. Taken together, standardized protocols and more longitudinal studies are required to provide a better insight into the effects of physical exercise on cognitive function. Although there is no agreement in the literature regarding the effects of physical exercise on cognitive function, it is well established that it improves social interaction and the feeling of well-being, thereby positively contributing to the quality of life of the elderly. Regarding the influence of physical exercise on cognitive function in APOE ε4 allele carriers, the data trend shows that the carriers of allele ε4 for APOE gene were more responsive to the beneficial effects of physical exercise on cognitive function compared with non-carriers. Nevertheless, studies with larger sample sizes will provide more accuracy about this relationship.
Collapse
Affiliation(s)
- M E S Colovati
- Laboratório de Fisiopatologia do Envelhecimento, Programa de Pós-Graduação em Saúde e Meio Ambiente, Universidade Metropolitana de Santos, Santos, SP, Brasil
| | - I P Novais
- Departamento de Saúde I, Programa de Pós-Graduação em Educação Física UESB/UESC, Universidade Estadual do Sudoeste da Bahia, Jequié, BA, Brasil
| | - M Zampol
- Laboratório de Fisiopatologia do Envelhecimento, Programa de Pós-Graduação em Saúde e Meio Ambiente, Universidade Metropolitana de Santos, Santos, SP, Brasil
| | - G D Mendes
- Laboratório de Fisiopatologia do Envelhecimento, Programa de Pós-Graduação em Saúde e Meio Ambiente, Universidade Metropolitana de Santos, Santos, SP, Brasil
| | - M C S Cernach
- Laboratório de Fisiopatologia do Envelhecimento, Programa de Pós-Graduação em Saúde e Meio Ambiente, Universidade Metropolitana de Santos, Santos, SP, Brasil
| | - A Zanesco
- Laboratório de Fisiopatologia do Envelhecimento, Programa de Pós-Graduação em Saúde e Meio Ambiente, Universidade Metropolitana de Santos, Santos, SP, Brasil
| |
Collapse
|
20
|
Ma D, Luo Y, Huang R, Zhao Z, Wang Q, Li L, Zhang L. Cornel Iridoid Glycoside Suppresses Tau Hyperphosphorylation and Aggregation in a Mouse Model of Tauopathy through Increasing Activity of PP2A. Curr Alzheimer Res 2020; 16:1316-1331. [PMID: 31902362 DOI: 10.2174/1567205017666200103113158] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/12/2019] [Accepted: 12/30/2019] [Indexed: 01/27/2023]
Abstract
BACKGROUND rTg4510 mice are transgenic mice expressing P301L mutant tau and have been developed as an animal model of tauopathy including Alzheimer's Disease (AD). Cornel Iridoid Glycoside (CIG) is an active ingredient extracted from Cornus officinalis, a traditional Chinese herb. The purpose of the present study was to investigate the effects of CIG on tau pathology and underlying mechanisms using rTg4510 mice. METHODS The cognitive functions were detected by Morris water maze and objective recognition tests. Western blotting and immunofluorescence were conducted to measure the levels of phosphorylated tau and related proteins. Serine/threonine phosphatase assay was applied to detect the activity of protein phosphatase 2A (PP2A). RESULTS Intragastric administration of CIG for 3 months improved learning and memory abilities, prevented neuronal and synapse loss, halted brain atrophy, elevated levels of synaptic proteins, protected cytoskeleton, reduced tau hyperphosphorylation and aggregation in the brain of rTg4510 mice. In the mechanism studies, CIG increased the activity of PP2A, elevated the methylation of PP2A catalytic C (PP2Ac) at leucine 309, decreased the phosphorylation of PP2Ac at tyrosine 307, and increased protein expression of leucine carboxyl methyltransferase 1 (LCMT-1), protein tyrosine phosphatase 1B (PTP1B), and protein phosphatase 2A phosphatase activator (PTPA) in the brain of rTg4510 mice. CONCLUSION CIG might have the potential to treat tauopathy such as AD via activating PP2A.
Collapse
Affiliation(s)
- Denglei Ma
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Yi Luo
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Rui Huang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Zirun Zhao
- Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, United States
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Lin Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China
| |
Collapse
|
21
|
Plotkin SS, Cashman NR. Passive immunotherapies targeting Aβ and tau in Alzheimer's disease. Neurobiol Dis 2020; 144:105010. [PMID: 32682954 PMCID: PMC7365083 DOI: 10.1016/j.nbd.2020.105010] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 07/01/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022] Open
Abstract
Amyloid-β (Aβ) and tau proteins currently represent the two most promising targets to treat Alzheimer's disease. The most extensively developed method to treat the pathologic forms of these proteins is through the administration of exogenous antibodies, or passive immunotherapy. In this review, we discuss the molecular-level strategies that researchers are using to design an effective therapeutic antibody, given the challenges in treating this disease. These challenges include selectively targeting a protein that has misfolded or is pathological rather than the more abundant, healthy protein, designing strategic constructs for immunizing an animal to raise an antibody that has the appropriate conformational selectivity to achieve this end, and clearing the pathological protein species before prion-like cell-to-cell spread of misfolded protein has irreparably damaged neurons, without invoking damaging inflammatory responses in the brain that naturally arise when the innate immune system is clearing foreign agents. The various solutions to these problems in current clinical trials will be discussed.
Collapse
Affiliation(s)
- Steven S Plotkin
- University of British Columbia, Department of Physics and Astronomy and Genome Sciences and Technology Program, Vancouver, BC V6T 1Z1, Canada.
| | - Neil R Cashman
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 2B5, Canada.
| |
Collapse
|
22
|
Muralidar S, Ambi SV, Sekaran S, Thirumalai D, Palaniappan B. Role of tau protein in Alzheimer's disease: The prime pathological player. Int J Biol Macromol 2020; 163:1599-1617. [PMID: 32784025 DOI: 10.1016/j.ijbiomac.2020.07.327] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/07/2020] [Accepted: 07/31/2020] [Indexed: 01/11/2023]
Abstract
Alzheimer's disease (AD) is a prevalently found tauopathy characterized by memory loss and cognitive insufficiency. AD is an age-related neurodegenerative disease with two major hallmarks which includes extracellular amyloid plaques made of amyloid-β (Aβ) and intracellular neurofibrillary tangles of hyperphosphorylated tau. With population aging worldwide, there is an indispensable need for treatment strategies that can potentially manage this developing dementia. Despite broad researches on targeting Aβ in the past two decades, research findings on Aβ targeted therapeutics failed to prove efficacy in the treatment of AD. Tau protein with its extensive pathological role in several neurodegenerative diseases can be considered as a promising target candidate for developing therapeutic interventions. The abnormal hyperphosphorylation of tau plays detrimental pathological functions which ultimately lead to neurodegeneration. This review will divulge the importance of tau in AD pathogenesis, the interplay of Aβ and tau, the pathological functions of tau, and potential therapeutic strategies for an effective management of neuronal disorders.
Collapse
Affiliation(s)
- Shibi Muralidar
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| | - Senthil Visaga Ambi
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India.
| | - Saravanan Sekaran
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India; Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| | - Diraviyam Thirumalai
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| | - Balamurugan Palaniappan
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| |
Collapse
|
23
|
Fyn Tyrosine Kinase as Harmonizing Factor in Neuronal Functions and Dysfunctions. Int J Mol Sci 2020; 21:ijms21124444. [PMID: 32580508 PMCID: PMC7352836 DOI: 10.3390/ijms21124444] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 12/25/2022] Open
Abstract
Fyn is a non-receptor or cytoplasmatic tyrosine kinase (TK) belonging to the Src family kinases (SFKs) involved in multiple transduction pathways in the central nervous system (CNS) including synaptic transmission, myelination, axon guidance, and oligodendrocyte formation. Almost one hundred years after the original description of Fyn, this protein continues to attract extreme interest because of its multiplicity of actions in the molecular signaling pathways underlying neurodevelopmental as well as neuropathologic events. This review highlights and summarizes the most relevant recent findings pertinent to the role that Fyn exerts in the brain, emphasizing aspects related to neurodevelopment and synaptic plasticity. Fyn is a common factor in healthy and diseased brains that targets different proteins and shapes different transduction signals according to the neurological conditions. We will primarily focus on Fyn-mediated signaling pathways involved in neuronal differentiation and plasticity that have been subjected to considerable attention lately, opening the fascinating scenario to target Fyn TK for the development of potential therapeutic interventions for the treatment of CNS injuries and certain neurodegenerative disorders like Alzheimer’s disease.
Collapse
|
24
|
Monteiro KL, Alcântara MGDS, de Aquino TM, da Silva-Júnior EF. Tau Protein Aggregation in Alzheimer's Disease: Recent Advances in the Development of Novel Therapeutic Agents. Curr Pharm Des 2020; 26:1682-1692. [DOI: 10.2174/1381612826666200414164038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/26/2020] [Indexed: 12/18/2022]
Abstract
:
Major research in Alzheimer’s disease (AD) related to disease-modifying agents is concentrated on
pharmacological approaches related to diagnostic markers, neurofibrillary tangles and amyloid plaques. Although
most studies focus on anti-amyloid strategies, investigations on tau protein have produced significant advances in
the modulation of the pathophysiology of several neurodegenerative diseases. Since the discovery of phenothiazines
as tau protein aggregation inhibitors (TAGIs), many additional small molecule inhibitors have been discovered
and characterized in biological model systems, which exert their interaction effects by covalent and noncovalent
means. In this paper, we summarize the latest advances in the discovery and development of tau aggregation
inhibitors using a specialized approach in their chemical classes. The design of new TAGIs and their encouraging
use in in vivo and clinical trials support their potential therapeutic use in AD.
Collapse
Affiliation(s)
- Kadja L.C. Monteiro
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Maceió, Brazil
| | - Marcone G. dos S. Alcântara
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Maceió, Brazil
| | - Thiago M. de Aquino
- Chemistry and Biotechnology Institute, Federal University of Alagoas, Maceió, Brazil
| | | |
Collapse
|
25
|
Husna Ibrahim N, Yahaya MF, Mohamed W, Teoh SL, Hui CK, Kumar J. Pharmacotherapy of Alzheimer's Disease: Seeking Clarity in a Time of Uncertainty. Front Pharmacol 2020; 11:261. [PMID: 32265696 PMCID: PMC7105678 DOI: 10.3389/fphar.2020.00261] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is recognized as a major health hazard that mostly affects people older than 60 years. AD is one of the biggest medical, economic, and social concerns to patients and their caregivers. AD was ranked as the 5th leading cause of global deaths in 2016 by the World Health Organization (WHO). Many drugs targeting the production, aggregation, and clearance of Aβ plaques failed to give any conclusive clinical outcomes. This mainly stems from the fact that AD is not a disease attributed to a single-gene mutation. Two hallmarks of AD, Aβ plaques and neurofibrillary tangles (NFTs), can simultaneously induce other AD etiologies where every pathway is a loop of consequential events. Therefore, the focus of recent AD research has shifted to exploring other etiologies, such as neuroinflammation and central hyperexcitability. Neuroinflammation results from the hyperactivation of microglia and astrocytes that release pro-inflammatory cytokines due to the neurological insults caused by Aβ plaques and NFTs, eventually leading to synaptic dysfunction and neuronal death. This review will report the failures and side effects of many anti-Aβ drugs. In addition, emerging treatments targeting neuroinflammation in AD, such as nonsteroidal anti-inflammatory drugs (NSAIDs) and receptor-interacting serine/threonine protein kinase 1 (RIPK1), that restore calcium dyshomeostasis and microglia physiological function in clearing Aβ plaques, respectively, will be deliberately discussed. Other novel pharmacotherapy strategies in treating AD, including disease-modifying agents (DMTs), repurposing of medications used to treat non-AD illnesses, and multi target-directed ligands (MTDLs) are also reviewed. These approaches open new doors to the development of AD therapy, especially combination therapy that can cater for several targets simultaneously, hence effectively slowing or stopping AD.
Collapse
Affiliation(s)
- Nurul Husna Ibrahim
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Wael Mohamed
- Basic Medical Science Department, Kulliyyah of Medicine, International Islamic University Malaysia, Kuantan, Malaysia
- Faculty of Medicine, Department of Clinical Pharmacology, Menoufia University, Shebin El-Kom, Egypt
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Chua Kien Hui
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
- Glycofood Sdn Bhd, Selangor, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
26
|
Wattmo C, Blennow K, Hansson O. Cerebro-spinal fluid biomarker levels: phosphorylated tau (T) and total tau (N) as markers for rate of progression in Alzheimer's disease. BMC Neurol 2020; 20:10. [PMID: 31918679 PMCID: PMC6951013 DOI: 10.1186/s12883-019-1591-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/29/2019] [Indexed: 01/08/2023] Open
Abstract
Background We investigated the potential associations between cerebro-spinal fluid (CSF) levels of phosphorylated tau (P-tau) and total tau (T-tau) with short-term response to cholinesterase inhibitor (ChEI) treatment, longitudinal outcome and progression rates in Alzheimer’s disease (AD). Methods This prospective, observational study included 129 participants clinically diagnosed with mild-to-moderate AD, who underwent a lumbar puncture. The CSF biomarkers amyloid-β1–42 (Aβ42), P-tau and T-tau were analysed with xMAP technology. Cognitive, global, instrumental and basic activities of daily living (ADL) capacities at the start of ChEI therapy and semi-annually over 3 years were evaluated. Results All patients had abnormal Aβ42 (A+). Fifty-eight individuals (45%) exhibited normal P-tau and T-tau (A+ T– (N)–), 12 (9%) abnormal P-tau/normal T-tau (A+ T+ (N)–), 17 (13%) normal P-tau/abnormal T-tau (A+ T– (N)+) and 42 (33%) abnormal P-tau and T-tau (A+ T+ (N)+). The participants with A+ T+ (N)+ were younger than A+ T– (N)+ at the estimated onset of AD and the initiation of ChEIs. The proportion of 6-month responders to ChEI and deterioration/year after start of treatment did not differ between the AT(N) profiles in any scales. A higher percentage of globally improved/unchanged patients was exhibited in the A+ T– (N)– group after 12, 30 and 36 months of ChEI therapy but not at other assessments. In apolipoprotein E (APOE) ε4-carriers, linear relationships were found between greater cognitive decline/year and higher tau; Mini-Mental State Examination score – T-tau (rs = − 0.257, p = 0.014) and Alzheimer’s Disease Assessment Scale–cognitive subscale – P-tau (rs = − 0.242, p = 0.022). A correlation between faster progression in instrumental ADL (IADL) and higher T-tau was also detected (rs = − 0.232, p = 0.028). These associations were not demonstrated in non-ε4-carriers. Conclusions Younger age and faster global deterioration were observed in AD patients with pathologic tau and neurodegeneration, whereas more rapid cognitive and IADL decline were related to higher P-tau or T-tau in APOE ε4-carriers only. The results might indicate an association between more pronounced tau pathology/neuronal injury and the APOE ε4-allele leading to a worse prognosis. Our findings showed that the AT(N) biomarker profiles have limited utility to predict AD progression rates and, thus, measure change and interpreting outcomes from clinical trials of future therapies.
Collapse
Affiliation(s)
- Carina Wattmo
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, SE-205 02, Malmö, Sweden. .,Memory Clinic, Skåne University Hospital, SE-205 02, Malmö, Sweden.
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy, University of Gothenburg, SE-431 80, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, SE-205 02, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, SE-205 02, Malmö, Sweden
| |
Collapse
|
27
|
Shukla R, Munjal NS, Singh TR. Identification of novel small molecules against GSK3β for Alzheimer's disease using chemoinformatics approach. J Mol Graph Model 2019; 91:91-104. [PMID: 31202091 DOI: 10.1016/j.jmgm.2019.06.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 06/06/2019] [Accepted: 06/06/2019] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease is a rapidly increasing neurodegenerative disease. It is a multifactorial disease and also a global threat. Several enzymes are implicated in the disease in which Glycogen Synthase Kinase 3 beta is a key enzyme to increase the disease progression by the hyperphosphorylation of the tau protein. We have used an integrative chemoinformatics and pharmacokinetics approach for the identification of novel small molecules. We have retrieved a subset from the ZINC database (n = 5,36,709) and screened against GSK3β in four steps. From here top 298 potent compounds were selected and employed for their pharmacokinetics analysis. We had seen that 29 compounds showed the key characteristics to be a novel drug candidate therefore, all these compounds were employed for redocking studies using Autodock Vina and Autodock. This analysis revealed that four compounds were showing good binding affinity. All these four compounds were employed for MDS analysis of 100 ns From here using a bunch of MD analyses we have found that out of four compounds GSK3β-ZINC21011059 and GSK3β-ZINC21011066 act as a stable protein-ligand complex. Therefore we proposed ZINC21011059 and ZINC21011066 can serve as a novel compounds against GSK3β and predicted scaffold can be used for further optimization towards the improvement of isoform selectivity, and warranting further investigations towards their in vitro and in vivo validation of the bioactivity.
Collapse
Affiliation(s)
- Rohit Shukla
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Solan, H.P, 173234, India
| | - Nupur S Munjal
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Solan, H.P, 173234, India
| | - Tiratha Raj Singh
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Solan, H.P, 173234, India.
| |
Collapse
|
28
|
González JF, Alcántara AR, Doadrio AL, Sánchez-Montero JM. Developments with multi-target drugs for Alzheimer's disease: an overview of the current discovery approaches. Expert Opin Drug Discov 2019; 14:879-891. [PMID: 31165654 DOI: 10.1080/17460441.2019.1623201] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: Alzheimer's disease (AD), the most common type of dementia among older adults, is a chronic neurodegenerative pathology that causes a progressive loss of cognitive functioning with a decline of rational skills. It is well known that AD is multifactorial, so there are many different pharmacological targets that can be pursued. Areas covered: The authors highlight the strategic value of privileged scaffolds in a multi-target lead compound generation against AD, exploring the concept of multi-target design, with a special emphasis on hybrid compounds. Hence, the most promising building blocks for designing and synthesizing hybrid anti-AD drugs are shown, while also presenting the more advanced hybrid compounds. Expert opinion: The available therapeutic arsenal for AD, designed under the traditional paradigm of 'one-drug/one target/one-disease', is based on the inhibition of brain acetylcholinesterase (AChE) to increase acetylcholine (ACh) levels. However, this classical approach has not been sufficiently effective when used to treat any multifactor-depending pathology (cancer, diabetes or AD). The multi-target drug concept has been quickly adopted by medicinal chemists. The basic research developments reported in recent years are a solid foundation that will pave the way for the construction of future AD therapeutics.
Collapse
Affiliation(s)
- Juan F González
- a Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid , Madrid , Spain
| | - Andrés R Alcántara
- a Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid , Madrid , Spain
| | - Antonio L Doadrio
- a Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid , Madrid , Spain
| | - Jose María Sánchez-Montero
- a Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid , Madrid , Spain
| |
Collapse
|
29
|
Jana S, Singh SK. Identification of human tau-tubulin kinase 1 inhibitors: an integrated e-pharmacophore-based virtual screening and molecular dynamics simulation. J Biomol Struct Dyn 2019; 38:886-900. [DOI: 10.1080/07391102.2019.1590242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Srabanti Jana
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Sushil Kumar Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| |
Collapse
|
30
|
Shukla R, Singh TR. Virtual screening, pharmacokinetics, molecular dynamics and binding free energy analysis for small natural molecules against cyclin-dependent kinase 5 for Alzheimer's disease. J Biomol Struct Dyn 2019; 38:248-262. [PMID: 30688165 DOI: 10.1080/07391102.2019.1571947] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and characterized by brain cell death, memory loss and is the most common form of dementia. Although AD has devastating effects, however, drugs which can treat the AD remain limited. The cyclin-dependent kinase 5 (CDK5) has been recognized as being involved in the pathological hyperphosphorylation of tau protein, which leads to the formation of neurofibrillary tangles (NFTs). We utilized the structure-based virtual screening (SBVS) approach to find the potential inhibitors against HsCDK5. The natural compound subset from the ZINC database (n = 167,741) was retrieved and screened by using SBVS method. From here, we have predicted 297 potent inhibitors. These 297 compounds were evaluated through their pharmacokinetic properties by ADMET (absorption, distribution, metabolism, elimination/excretion and toxicity) descriptors. Finally, 17 compounds were selected and used for re-docking. After the refinement by molecular docking and by using drug-likeness analysis, we have identified four potential inhibitors (ZINC85877721, ZINC96114862, ZINC96115616 and ZINC96116231). All these four ligands were employed for 100 ns MDS study. From the root mean square deviation (RMSD), root mean square fluctuation (RMSF), Rg, number of hydrogen bonds, solvent accessible surface area (SASA), principal component analysis (PCA) and binding free energy analysis we have found that out of four inhibitors ZINC85877721 and ZINC96116231 showed good binding free energy of -198.84 and -159.32 kJ.mol-1, respectively, and also good in other structural analyses. Both compounds displayed excellent pharmacological and structural properties to be the drug candidates. Collectively, these findings recommend that two compounds have great potential to be a promising agent against AD to reduce the CDK5 induced hyperphosphorylation and could be considered as therapeutic agents for the AD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rohit Shukla
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, India
| | - Tiratha Raj Singh
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, India
| |
Collapse
|
31
|
Abstract
Alzheimer's disease (AD), the most common form of dementia, is characterized by extracellular β-amyloid plaques and intracellular neurofibrillary tangles (NFTs), which are considered as major targets for AD therapies. However, no effective therapy is available to cure or prevent the progression of AD up until now. Accumulation of NFTs, which consist of abnormally hyperphosphorylated tau, is directly correlated with the degree of dementia in AD patients. Emerging evidence indicates that the prion-like seeding and spreading of tau pathology may be the key driver of AD. In the past decades, greater understanding of tau pathway reveals new targets for the development of specific therapies. Here, we review the recent research progress in the mechanism underlying tau pathology in AD and briefly introduce tau-based therapeutics.
Collapse
Affiliation(s)
- Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, PR China
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York 10314, United States
| |
Collapse
|
32
|
Alonso AD, Cohen LS, Corbo C, Morozova V, ElIdrissi A, Phillips G, Kleiman FE. Hyperphosphorylation of Tau Associates With Changes in Its Function Beyond Microtubule Stability. Front Cell Neurosci 2018; 12:338. [PMID: 30356756 PMCID: PMC6189415 DOI: 10.3389/fncel.2018.00338] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 09/13/2018] [Indexed: 01/02/2023] Open
Abstract
Tau is a neuronal microtubule associated protein whose main biological functions are to promote microtubule self-assembly by tubulin and to stabilize those already formed. Tau also plays an important role as an axonal microtubule protein. Tau is an amazing protein that plays a key role in cognitive processes, however, deposits of abnormal forms of tau are associated with several neurodegenerative diseases, including Alzheimer disease (AD), the most prevalent, and Chronic Traumatic Encephalopathy (CTE) and Traumatic Brain Injury (TBI), the most recently associated to abnormal tau. Tau post-translational modifications (PTMs) are responsible for its gain of toxic function. Alonso et al. (1996) were the first to show that the pathological tau isolated from AD brains has prion-like properties and can transfer its toxic function to the normal molecule. Furthermore, we reported that the pathological changes are associated with tau phosphorylation at Ser199 and 262 and Thr212 and 231. This pathological version of tau induces subcellular mislocalization in cultured cells and neurons, and translocates into the nucleus or accumulated in the perinuclear region of cells. We have generated a transgenic mouse model that expresses pathological human tau (PH-Tau) in neurons at two different concentrations (4% and 14% of the total endogenous tau). In this model, PH-Tau causes cognitive decline by at least two different mechanisms: one that involves the cytoskeleton with axonal disruption (at high concentration), and another in which the apparent neuronal morphology is not grossly affected, but the synaptic terminals are altered (at lower concentration). We will discuss the putative involvement of tau in proteostasis under these conditions. Understanding tau’s biological activity on and off the microtubules will help shed light to the mechanism of neurodegeneration and of normal neuronal function.
Collapse
Affiliation(s)
- Alejandra D Alonso
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, The City University of New York, Staten Island, NY, United States.,Biology Program, The Graduate Center, The City University of New York, New York, NY, United States.,Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Leah S Cohen
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, The City University of New York, Staten Island, NY, United States
| | - Christopher Corbo
- Department of Biology, Wagner College, Staten Island, NY, United States
| | - Viktoriya Morozova
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, The City University of New York, Staten Island, NY, United States.,Biology Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Abdeslem ElIdrissi
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, The City University of New York, Staten Island, NY, United States.,Biology Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Greg Phillips
- Department of Biology and Center for Developmental Neuroscience, College of Staten Island, The City University of New York, Staten Island, NY, United States.,Biology Program, The Graduate Center, The City University of New York, New York, NY, United States
| | - Frida E Kleiman
- Biochemistry Program, The Graduate Center, The City University of New York, New York, NY, United States.,Department of Chemistry, Hunter College, The City University of New York, New York, NY, United States
| |
Collapse
|