1
|
Xing X, Liu X, Li X, Li M, Wu X, Huang X, Xu A, Liu Y, Zhang J. Insights into spinal muscular atrophy from molecular biomarkers. Neural Regen Res 2025; 20:1849-1863. [PMID: 38934395 DOI: 10.4103/nrr.nrr-d-24-00067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/11/2024] [Indexed: 06/28/2024] Open
Abstract
Spinal muscular atrophy is a devastating motor neuron disease characterized by severe cases of fatal muscle weakness. It is one of the most common genetic causes of mortality among infants aged less than 2 years. Biomarker research is currently receiving more attention, and new candidate biomarkers are constantly being discovered. This review initially discusses the evaluation methods commonly used in clinical practice while briefly outlining their respective pros and cons. We also describe recent advancements in research and the clinical significance of molecular biomarkers for spinal muscular atrophy, which are classified as either specific or non-specific biomarkers. This review provides new insights into the pathogenesis of spinal muscular atrophy, the mechanism of biomarkers in response to drug-modified therapies, the selection of biomarker candidates, and would promote the development of future research. Furthermore, the successful utilization of biomarkers may facilitate the implementation of gene-targeting treatments for patients with spinal muscular atrophy.
Collapse
Affiliation(s)
- Xiaodong Xing
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xinzhu Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiandeng Li
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Mi Li
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xian Wu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Xiaohui Huang
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ajing Xu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Zhang
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
El-Desouky S, Abdel-Halim M, Fathalla RK, Abadi AH, Piazza GA, Salama M, El-Khodery SA, Youssef MA, Elfarrash S. A novel phosphodiesterase 5 inhibitor, RF26, improves memory impairment and ameliorates tau aggregation and neuroinflammation in the P301S tauopathy mouse model of Alzheimer's disease. Exp Neurol 2025; 384:115058. [PMID: 39549949 DOI: 10.1016/j.expneurol.2024.115058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/10/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
Phosphodiesterase-5 (PDE5) inhibitors are primarily used in the treatment of erectile dysfunction and pulmonary hypertension, but have also been reported to have a potential therapeutic effect for the treatment of Alzheimer's disease (AD). This is likely to be through stimulation of nitric oxide (NO)/cyclic guanosine monophosphate (cGMP) signaling by elevating cGMP, a secondary messenger involved in processes of neuroplasticity. In the present study, we evaluated the efficacy of a novel PDE5 inhibitor, RF26, using P301S tauopathy mice model. A body of experimental evidence suggests that the development of tau inclusions leads to the neurodegeneration observed in tauopathies, including AD, Frontotemporal dementia (FTD), Supranuclear palsy and others. RF26 successfully targeted NO/cGMP signaling pathway and showed a significant improvement of spatial memory task performance of P301S mice using Morris Water Maze and T-maze. Furthermore, RF26 -treated mice showed a significant reduction of phosphorylated tau load, gliosis and downregulated pro-inflammatory cytokines. The presented data support the efficacy of RF26 as a potent PDE5 inhibitor and calls for further investigation as a potential therapeutic drug for Alzheimer's and other tauopathy related neurological disorders.
Collapse
Affiliation(s)
- Sara El-Desouky
- Medical experimental research center (MERC), Faculty of Medicine, Mansoura University, 35116 Mansoura, Egypt
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Reem K Fathalla
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Gary A Piazza
- Department of Drug discovery and development, Harrison Collage of Pharmacy, Auburn University, Auburn, AL 36832, USA
| | - Mohamed Salama
- Institute of Global health and Human ecology, American University in Cairo, Egypt; Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, 35116 Mansoura, Egypt
| | - Sabry Ahmed El-Khodery
- Department of internal medicine, Faculty of Veterinary Medicine, Mansoura University, 35116 Mansoura, Egypt
| | - Mohamed A Youssef
- Department of internal medicine, Faculty of Veterinary Medicine, Mansoura University, 35116 Mansoura, Egypt
| | - Sara Elfarrash
- Medical experimental research center (MERC), Faculty of Medicine, Mansoura University, 35116 Mansoura, Egypt; Department of Medical Physiology, Faculty of Medicine, Mansoura University, 35116 Mansoura, Egypt.
| |
Collapse
|
3
|
Ding X, Shang D, Cui Y, Dong X, Chen C, Zhao Y, Li X, Liang X. Tandem HILIC-IMAC strategy for simultaneous N-glycoproteomics and phosphoproteomics in aging mouse brain. J Chromatogr A 2025; 1739:465525. [PMID: 39566287 DOI: 10.1016/j.chroma.2024.465525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/25/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
Abnormal glycosylation and phosphorylation are strongly associated with brain aging. N-glycosylation and phosphorylation are closely involved in pathological processes in a crosstalk dependent manner. However, simultaneous characterization of glycosylation and phosphorylation together in aging brain was uncommon. Herein we developed a novel tandem HILIC-IMAC strategy for simultaneous analysis of N-glycoproteomics and phosphoproteomics. This tandem method showed higher enrichment repeatability, more identifications of glycopeptides and phosphopeptides, and lower overlap. Application of the established method to mouse brain at two ages (8 weeks and 65 weeks) to explore changes in glycosylation and phosphorylation during aging. Up to 10,990 N-glycopeptides and 11,409 phosphopeptides were identified from mouse brain. Among these, differentially expressed phosphoproteins were involved in regulation of microtubule depolymerization, synapse, and transmission of nerve impulse. And glycoproteins differentially expressed with age were mostly related to cell adhesion processes and extracellular matrix. Furthermore, we found the opposite expression trends in glycosylation and phosphorylation during aging on Grin2b. Together, the HILIC-IMAC strategy has potential to discover aging biomarkers and analyze complex biosamples, paving the way for in-depth investigations for the changes of protein glycosylation and phosphorylation in aging brain.
Collapse
Affiliation(s)
- Xinlian Ding
- Pharmacy College, Dalian Medical University, Dalian, 116044, PR China; Key Laboratory of Phytochemistry and Natural Medicines. Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China; Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, PR China
| | - Danyi Shang
- Key Laboratory of Phytochemistry and Natural Medicines. Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China; Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, PR China
| | - Yun Cui
- Key Laboratory of Phytochemistry and Natural Medicines. Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China; Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, PR China
| | - Xuefang Dong
- Key Laboratory of Phytochemistry and Natural Medicines. Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China; Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, PR China
| | - Cheng Chen
- Key Laboratory of Phytochemistry and Natural Medicines. Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China; Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, PR China
| | - Yanyan Zhao
- Pharmacy College, Dalian Medical University, Dalian, 116044, PR China.
| | - Xiuling Li
- Key Laboratory of Phytochemistry and Natural Medicines. Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China; Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, PR China.
| | - Xinmiao Liang
- Key Laboratory of Phytochemistry and Natural Medicines. Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, PR China; Ganjiang Chinese Medicine Innovation Center, Nanchang, 330000, PR China
| |
Collapse
|
4
|
Salari A, Roghani M, Khalili M. HMG-CoA reductase inhibitor simvastatin ameliorates trimethyltin neurotoxicity and cognitive impairment through reversal of Alzheimer's-associated markers. Metab Brain Dis 2024; 40:74. [PMID: 39704877 DOI: 10.1007/s11011-024-01515-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 12/18/2024] [Indexed: 12/21/2024]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder in elderly. The neurotoxicant trimethyltin (TMT) induces neurodegenerative changes, as observed in AD. The 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor simvastatin (SV) has shown protective and promising therapeutic effects in neurological disorders such as AD and Parkinson's disease. The present study aimed to assess neuroprotective effect of simvastatin (SV) against trimethyltin (TMT) memory decline and hippocampal neurodegeneration. For inducing AD-like phenotype, rats were i.p. injected with TMT at 8 mg/kg and were treated with simvastatin daily for 3 weeks at 10 or 30 mg/kg. Our analysis of data indicated that simvastatin-treated TMT group has lower learning and memory deficits in behavioral tasks comprising Barnes maze, Y maze, and novel object discrimination (NOD). In addition, hippocampal inflammatory, oxidative, and apoptotic factors were attenuated besides reduction of acetylcholinesterase (AChE) activity and Alzheimer's pathology factors including presenilin-1 and hyperphorphorylated Tau (p-Tau) upon simvastatin. Moreover, simvastatin treatment of TMT group inverted hippocampal changes of Wnt, β-catenin, ERK, and Akt, ameliorated astrocytic and microglial reactivity, and also prevented injury of CA1 neurons. This study unraveled that simvastatin is capable to prevent TMT-induced Alzheimer's-like phenotype in association with Wnt/β-catenin/ERK/Akt as well as restraining hippocampal neurodegeneration.
Collapse
Affiliation(s)
- Adel Salari
- Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran.
| | - Mohsen Khalili
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| |
Collapse
|
5
|
Meng R, Li Y, Yang X, Cheng Y, Xu M, Zhou L, Wu C, Yu S, Huang W, Wang T, Zhang Q. Polyphenol Mediated Assembly: Tailored Nano-Dredger Unblocks Axonal Autophagosomes Retrograde Transport Traffic Jam for Accelerated Alzheimer's Waste Clearance. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2413614. [PMID: 39686827 DOI: 10.1002/adma.202413614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/01/2024] [Indexed: 12/18/2024]
Abstract
Clear-cut evidence has linked defective autophagy to Alzheimer's disease (AD). Recent studies underscore a unique hurdle in AD neuronal autophagy: impaired retrograde axonal transport of autophagosomes, potent enough to induce autophagic stress and neurodegeneration. Nonetheless, pertinent therapy is unavailable. Here, a novel combinational therapy composed of siROCK2 and lithospermic acid B (LA) is introduced, tailored to dredge blocked axonal autophagy by multi-mitigating microtubule disruption, ATP depletion, oxidative stress, and autophagy initiation impediments in AD. Leveraging the recent discovery of multi-interactions between polyphenol LA and siRNA, ε-Poly-L-lysine, and anionic lipid nanovacuoles, LA and siROCK2 are successfully co-loaded into a fresh nano-drug delivery system, LIP@PL-LA/siRC, via a ratio-flexible and straightforward fabrication process. Further modification with the TPL peptide onto LIP@PL-LA/siRC creates a brain-neuron targeted, biocompatible, and pluripotent nanomedicine, named "Nano-dredger" (T-LIP@PL-LA/siRC). Nano-dredger efficiently accelerates axonal retrograde transport and lysosomal degradation of autophagosomes, thereby facilitating the clearance of neurotoxic proteins, improving neuronal complexity, and alleviating memory defects in 3×Tg-AD transgenic mice. This study provides a fresh and flexible polyphenol/siRNA co-delivery paradigm and furnishes conceptual proof that dredging axonal autophagy represents a promising AD therapeutic avenue.
Collapse
Affiliation(s)
- Ran Meng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Xiyu Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yunlong Cheng
- Shanxi Academy of Traditional Chinese Medicine, Xi'an, 710003, P. R. China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - LingLing Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Chengqin Wu
- Guangzhou CSR Biotech Co. Ltd, Guangzhou, 510700, P. R. China
| | - Shuai Yu
- Guangzhou CSR Biotech Co. Ltd, Guangzhou, 510700, P. R. China
| | - Wenyi Huang
- Guangzhou CSR Biotech Co. Ltd, Guangzhou, 510700, P. R. China
| | - Tianying Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| |
Collapse
|
6
|
Ferretti S, Zanella I. The Underestimated Role of Iron in Frontotemporal Dementia: A Narrative Review. Int J Mol Sci 2024; 25:12987. [PMID: 39684697 DOI: 10.3390/ijms252312987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The term frontotemporal dementia (FTD) comprises a group of neurodegenerative disorders characterized by the progressive degeneration of the frontal and temporal lobes of the brain with language impairment and changes in cognitive, behavioral and executive functions, and in some cases motor manifestations. A high proportion of FTD cases are due to genetic mutations and inherited in an autosomal-dominant manner with variable penetrance depending on the implicated gene. Iron is a crucial microelement that is involved in several cellular essential functions in the whole body and plays additional specialized roles in the central nervous system (CNS) mainly through its redox-cycling properties. Such a feature may be harmful under aerobic conditions, since it may lead to the generation of highly reactive hydroxyl radicals. Dysfunctions of iron homeostasis in the CNS are indeed involved in several neurodegenerative disorders, although it is still challenging to determine whether the dyshomeostasis of this essential but harmful metal is a direct cause of neurodegeneration, a contributor factor or simply a consequence of other neurodegenerative mechanisms. Unlike many other neurodegenerative disorders, evidence of the dysfunction in brain iron homeostasis in FTD is still scarce; nonetheless, the recent literature intriguingly suggests its possible involvement. The present review aims to summarize what is currently known about the contribution of iron dyshomeostasis in FTD based on clinical, imaging, histological, biochemical and molecular studies, further suggesting new perspectives and offering new insights for future investigations on this underexplored field of research.
Collapse
Affiliation(s)
- Sara Ferretti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Isabella Zanella
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Medical Genetics Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| |
Collapse
|
7
|
Fernandez F, Griffiths LR, Sutherland HG, Cole MH, Fitton JH, Winberg P, Schweitzer D, Hopkins LN, Meyer BJ. Sirtuin Proteins and Memory: A Promising Target in Alzheimer's Disease Therapy? Nutrients 2024; 16:4088. [PMID: 39683482 DOI: 10.3390/nu16234088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Sirtuins (SIRTs), nicotine adenine dinucleotide (+)-dependent histone deacetylases, have emerged as critical regulators in many signalling pathways involved in a wide range of biological processes. Currently, seven mammalian SIRTs have been characterized and are found across a number of cellular compartments. There has been considerable interest in the role of SIRTs in the brain due to their role in a plethora of metabolic- and age-related diseases, including their involvement in learning and memory function in physiological and pathophysiological conditions. Although cognitive function declines over the course of healthy ageing, neurological disorders including Alzheimer's disease (AD) can be associated with progressive cognitive impairments. This review aimed to report and integrate recent advances in the understanding of the role of SIRTs in cognitive function and dysfunction in the context of AD. We have also reviewed the use of selective and/or natural SIRT activators as potential therapeutic agents and/or adjuvants for AD.
Collapse
Affiliation(s)
- Francesca Fernandez
- School of Behavioural and Health Sciences, Faculty of Heath Sciences, Australian Catholic University, Banyo, QLD 4014, Australia
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, QLD 4059, Australia
- Healthy Brain and Mind Research Centre, Australian Catholic University, Fitzroy, VIC 3065, Australia
| | - Lyn R Griffiths
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, QLD 4059, Australia
| | - Heidi G Sutherland
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, QLD 4059, Australia
| | - Michael H Cole
- School of Behavioural and Health Sciences, Faculty of Heath Sciences, Australian Catholic University, Banyo, QLD 4014, Australia
- Healthy Brain and Mind Research Centre, Australian Catholic University, Fitzroy, VIC 3065, Australia
| | - J Helen Fitton
- Venus Shell Systems Pty Ltd., Huskisson, NSW 2540, Australia
| | - Pia Winberg
- Venus Shell Systems Pty Ltd., Huskisson, NSW 2540, Australia
- School of Medical, Indigenous and Health Science, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Daniel Schweitzer
- Mater Centre of Neuroscience, 53 Raymond Terrace, South Brisbane, QLD 4066, Australia
- Department of Neurology, Wesley Hospital, 451 Coronation Drive, Auchenflower, QLD 4066, Australia
| | - Lloyd N Hopkins
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, QLD 4059, Australia
| | - Barbara J Meyer
- School of Medical, Indigenous and Health Science, University of Wollongong, Wollongong, NSW 2522, Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
8
|
Alexandre-Silva V, Soares-Silva B, Pereira GC, Custódio-Silva AC, Carvalhinho-Lopes PS, Taliano LO, Lambertucci RH, Cavalcante MD, de Souza Araújo AA, Quintans-Júnior L, Dos Santos JR, Ribeiro AM. Eplingiella fruticosa leaf essential oil complexed with β-cyclodextrin exerts a neuroprotective effect in an Alzheimer's disease animal model induced by Streptozotocin. Metab Brain Dis 2024; 40:40. [PMID: 39579243 DOI: 10.1007/s11011-024-01484-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 11/13/2024] [Indexed: 11/25/2024]
Abstract
Alzheimer's Disease (AD) is physiopathologically marked by an accumulation of beta-amyloid peptide (Aβ), hyperphosphorylation of tau protein, inflammation, and oxidative stress in the brain tissue. While new drugs for AD have been approved, novel treatments are still needed. Eplingiella fruticosa (EF) has demonstrated anti-inflammatory and antioxidant properties, which may be beneficial against AD. This study aimed to evaluate the effects of EF leaf essential oil complexed with β-cyclodextrin in a sporadic AD model induced by streptozotocin (STZ). Male Wistar rats (5-6 months old) received an intracerebroventricular STZ injection (3 mg/kg) or vehicle, and were orally treated with vehicle, EF (5 mg/kg), or donepezil (5 mg/kg) for 14 days. Behavioral tests included olfactory discrimination, open field, novel object recognition, sucrose preference, and spontaneous alternation. Upon completion, rats were euthanatized, and their brains were analyzed for Aβ, tau, and IL-1β via immunohistochemistry, and for oxidative stress markers. STZ-treated rats showed memory deficits and anhedonia, accompanied by increased Aβ, tau, and IL-1β immunoreactivity in the olfactory bulb, cortex, hippocampus, and increased TBARS levels in the hippocampus. On the other hand, EF treatment improved short-term and working memory (p < 0.001), and reduced depressive-like behavior (p = 0.02). Additionally, EF treatment decreased Aβ, tau, and IL-1β immunoreactivity in the olfactory bulb, hippocampus and cortex (p < 0.05), and reduced TBARS levels (p = 0.04) and total oxidant status in the hippocampus (p = 0.03), and increased total antioxidant status in the cortex (p = 0.04). These findings suggest EF has neuroprotective effects against STZ-induced damage, indicating its potential as a novel compound for AD treatment.
Collapse
Affiliation(s)
- Vanessa Alexandre-Silva
- Department of Biosciences, Federal University of São Paulo, Santos, São Paulo, Brazil
- Department of Gerontology, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| | - Beatriz Soares-Silva
- Department of Biosciences, Federal University of São Paulo, Santos, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Merino-Serrais P, Soria JM, Arrabal CA, Ortigado-López A, Esparza MÁG, Muñoz A, Hernández F, Ávila J, DeFelipe J, León-Espinosa G. Protein tau phosphorylation in the proline rich region and its implication in the progression of Alzheimer's disease. Exp Neurol 2024; 383:115049. [PMID: 39522802 DOI: 10.1016/j.expneurol.2024.115049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Tau has a wide variety of essential functions in the brain, but this protein also plays a determining role in the development of Alzheimer's disease (AD) and other neurodegenerative diseases called tauopathies. This is due to its abnormal aggregation and the subsequent formation of neurofibrillary tangles. Tau hyperphosphorylation appears to be a critical step in its transformation into an aggregated protein. However, the exact process, including the cellular events that trigger it, remains unclear. In this study, we employed immunocytochemistry assays on hippocampal sections from AD cases and from tauopathy cases (Braak stage III) with no evidence of cognitive decline, and the P301S mouse model to investigate the colocalization patterns of Tau phosphorylated (p) at specific residues (S202-T205, S214, and T231) within the proline-rich region. Our results show pyramidal neurons in the hippocampus of P301S mice in which Tau is intensely phosphorylated at residues S202 and T205 (recognized by the AT8 antibody), but with no detectable phosphorylation at S214 or T231. These non-colocalizing neurons displayed intensely labeled aggregated pTau deposits distributed through the soma and dendritic processes. However, most of the hippocampal pyramidal neurons are labeled with pTauS214 or pTauT231 antibodies and typically showed a homogeneous and diffuse pTau distribution (not aggregated). This different labeling likely reflects a Tau conformational step, potentially related to the transition from a diffuse tau phosphorylation phenotype (Type 2) into an NFT-like or Type 1 phenotype. We further observed that dendrites of CA3 pyramidal cells are intensely labeled with pTau214 in the stratum lucidum, but not with AT8 or pTauT231. By contrast, analysis of tissue from AD patients or other human tauopathy cases (Braak stage III) with no evidence of cognitive decline revealed extensive colocalization with both antibody combinations in CA1. The complete or mature tangle development may follow a different mechanism in the P301S mouse model or may require more time to achieve the maturity state found in AD cases. Further studies would be necessary to address this question.
Collapse
Affiliation(s)
- Paula Merino-Serrais
- Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain; Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcón, 28223 Madrid, Spain; CIBER de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - José Miguel Soria
- Department of Biomedical Sciences, Cardenal Herrera University-CEU Universities, 46001, Valencia, Spain
| | - Cristina Aguirre Arrabal
- Departamento de Matemática Aplicada y Estadística, Universidad San Pablo-CEU, CEU Universities, Julian Romea 22, 28003 Madrid, Spain
| | - Alfonso Ortigado-López
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain
| | | | - Alberto Muñoz
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcón, 28223 Madrid, Spain; Department of Cell Biology, Universidad Complutense de Madrid, Madrid, Spain
| | - Félix Hernández
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Jesús Ávila
- CIBER de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain; Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Javier DeFelipe
- Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain; Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcón, 28223 Madrid, Spain; CIBER de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Gonzalo León-Espinosa
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcón, 28223 Madrid, Spain; Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain.
| |
Collapse
|
10
|
Illodo S, Al-Soufi W, Novo M. Critical aggregation concentration and reversibility of amyloid-β (1-40) oligomers. Arch Biochem Biophys 2024; 761:110179. [PMID: 39393664 DOI: 10.1016/j.abb.2024.110179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/13/2024] [Accepted: 10/09/2024] [Indexed: 10/13/2024]
Abstract
Amyloid-beta (Aβ) aggregation is a critical factor in the pathogenesis of Alzheimer's disease, with distinct aggregation behaviours observed between its isoforms Amyloid-β 1-40 (Aβ40) and 1-42 (Aβ42). In this study, we investigated the aggregation properties of Aβ40 using fluorescence correlation spectroscopy (FCS) and detailed data analysis. Our results reveal that Aβ40 undergoes a two-step cooperative aggregation process. The first step, characterized by a critical aggregation concentration (cac) of 0.5 ± 0.3 μM, results in the formation of metastable oligomers of 5-25 monomers and stable oligomers of 50-100 monomers, with less than 10 % of the total amyloid aggregated. The second step, with a cac of 19 ± 2 μM, leads to the formation of much larger aggregates, consistent with protofibrils, and approximately 50 % aggregated amyloid. Notably, the cac for Aβ40 is significantly higher, and the fraction of aggregated amyloid is much lower compared to Aβ42, indicating a lower propensity for aggregation. Additionally, our findings suggest that Aβ40 early oligomers are reversible upon dilution, albeit with a kinetic barrier to disaggregation. These insights into the aggregation mechanisms of Aβ40 enhance our understanding of its role in Alzheimer's disease and may inform therapeutic strategies targeting amyloid aggregation.
Collapse
Affiliation(s)
- Sara Illodo
- Facultade de Ciencias, Departamento de Química Física, Campus Terra, Universidade de Santiago de Compostela, 27002, Lugo, Spain; Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS) and Facultade de Química, Departamento de Química Física, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Wajih Al-Soufi
- Facultade de Ciencias, Departamento de Química Física, Campus Terra, Universidade de Santiago de Compostela, 27002, Lugo, Spain
| | - Mercedes Novo
- Facultade de Ciencias, Departamento de Química Física, Campus Terra, Universidade de Santiago de Compostela, 27002, Lugo, Spain.
| |
Collapse
|
11
|
Fang F, Xu T, Chien Hagar HT, Hovde S, Kuo MH, Sun L. Pilot Study for Deciphering Post-Translational Modifications and Proteoforms of Tau Protein by Capillary Electrophoresis-Mass Spectrometry. J Proteome Res 2024; 23:5085-5095. [PMID: 39327902 PMCID: PMC11536466 DOI: 10.1021/acs.jproteome.4c00587] [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: 07/09/2024] [Revised: 09/11/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Abnormal accumulation of tau protein in the brain is one pathological hallmark of Alzheimer's disease (AD). Many tau protein post-translational modifications (PTMs) are associated with the development of AD, such as phosphorylation, acetylation, and methylation. Therefore, a complete picture of the PTM landscape of tau is critical for understanding the molecular mechanisms of AD progression. Here, we offered a pilot study of combining two complementary analytical techniques, capillary zone electrophoresis (CZE)-tandem mass spectrometry (MS/MS) and reversed-phase liquid chromatography (RPLC)-MS/MS, for bottom-up proteomics of recombinant human tau-0N3R. We identified 50 phosphorylation sites of tau-0N3R in total, which is about 25% higher than that from RPLC-MS/MS alone. CZE-MS/MS provided more PTM sites (i.e., phosphorylation) and modified peptides of tau-0N3R than RPLC-MS/MS, and its predicted electrophoretic mobility helped improve the confidence of the identified modified peptides. We developed a highly efficient capillary isoelectric focusing (cIEF)-MS technique to offer a bird's-eye view of tau-0N3R proteoforms, with 11 putative tau-0N3R proteoforms carrying up to nine phosphorylation sites and lower pI values from more phosphorylated proteoforms detected. Interestingly, under native-like cIEF-MS conditions, we observed three putative tau-0N3R dimers carrying phosphate groups. The findings demonstrate that CE-MS is a valuable analytical technique for the characterization of tau PTMs, proteoforms, and even oligomerization.
Collapse
Affiliation(s)
- Fei Fang
- Department
of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
| | - Tian Xu
- Department
of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
| | - Hsiao-Tien Chien Hagar
- Department
of Biochemistry and Molecular Biology, Michigan
State University, 603 Wilson Road, Room 401, East Lansing, Michigan 48824, United States
| | - Stacy Hovde
- Department
of Biochemistry and Molecular Biology, Michigan
State University, 603 Wilson Road, Room 401, East Lansing, Michigan 48824, United States
| | - Min-Hao Kuo
- Department
of Biochemistry and Molecular Biology, Michigan
State University, 603 Wilson Road, Room 401, East Lansing, Michigan 48824, United States
| | - Liangliang Sun
- Department
of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
| |
Collapse
|
12
|
Chen X, Song Y, Hong Y, Zhang X, Li Q, Zhou H. "NO" controversy?: A controversial role in insulin signaling of diabetic encephalopathy. Mol Cell Endocrinol 2024; 593:112346. [PMID: 39151653 DOI: 10.1016/j.mce.2024.112346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/14/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Insulin, a critical hormone in the human body, exerts its effects by binding to insulin receptors and regulating various cellular processes. While nitric oxide (NO) plays an important role in insulin secretion and acts as a mediator in the signal transduction pathway between upstream molecules and downstream effectors, holds a significant position in the downstream signal network of insulin. Researches have shown that the insulin-NO system exhibits a dual regulatory effect within the central nervous system, which is crucial in the regulation of diabetic encephalopathy (DE). Understanding this system holds immense practical importance in comprehending the targets of existing drugs and the development of potential therapeutic interventions. This review extensively examines the characterization of insulin, NO, Nitric oxide synthase (NOS), specific NO pathway, their interconnections, and the mechanisms underlying their regulatory effects in DE, providing a reference for new therapeutic targets of DE.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Ying Song
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China; Hangzhou King's Bio-pharmaceutical Technology Co., Ltd, Hangzhou, Zhejiang, 310007, China.
| | - Ye Hong
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Xiaomin Zhang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qisong Li
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Hongling Zhou
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
13
|
Li L, Xiong Y, Zhang Y, Yan Y, Zhao R, Yang F, Xie M. Biofilm-camouflaged Prussian blue synergistic mitochondrial mass enhancement for Alzheimer's disease based on Cu 2+ chelation and photothermal therapy. J Control Release 2024; 375:269-284. [PMID: 39245418 DOI: 10.1016/j.jconrel.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/19/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases characterized by cognitive and memory impairment. Metal ion imbalance and Mitochondrial dysfunction, leading to abnormal aggregation of β-amyloid protein (Aβ), are key factors in the pathogenesis of AD. Therefore, we designed a composite nanometer system of red blood cell (RBC) membranes-encapsulated Prussian blue nanoparticles (PB/RBC). Prussian blue nanoparticles (PBNPs) can chelate Cu2+ and reduce reactive oxygen species (ROS). The RBC membranes are a kind of natural long-lasting circulating carrier. At the same time, through NIR irradiation, the excellent photothermal ability of PBNPs can also temporarily open the blood-brain barrier (BBB), enhance the transmission efficiency of PB/RBC across the BBB, and depolymerize the formed Aβ deposits, thereby achieving the optimal therapeutic effect. In vitro and in vivo studies demonstrated that PB/RBC could inhibit Cu2+-induced Aβ monomers aggregation, eliminate the deposition of Aβ plaques, improve the quality of mitochondria, restore the phagocytic function of microglia, alleviate neuroinflammation in APP/PS1 mice, and repair memory damage. In conclusion, our biofilm-camouflaged nano-delivery system provides significant neuroprotection by inhibiting Cu2+-induced Aβ monomers aggregation, photothermally depolymerizing Aβ fibrils and reducing the level of ROS, thus effectively ameliorating and treating AD.
Collapse
Affiliation(s)
- Lianxin Li
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Yu Xiong
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Yuewen Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Yujiao Yan
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Ruixin Zhao
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Fengmei Yang
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Meng Xie
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China.
| |
Collapse
|
14
|
Yuan J, Dong X, Zhou S, Nao J. Pharmacological activities and therapeutic potential of Hyperoside in the treatment of Alzheimer's and Parkinson's diseases: A systemic review. Neuroscience 2024; 563:136-147. [PMID: 39489478 DOI: 10.1016/j.neuroscience.2024.10.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are neurodegenerative disorders that significantly impact well-being. Hyperoside (HYP), a flavonoid found in various plant species, particularly within the genus Hypericin, exhibits diverse pharmacological properties. However, the precise mechanisms underlying the anti-AD and anti-PD effects of HYP remain unclear. This systematic review consolidated existing preclinical research on HYP by conducting a comprehensive literature survey and analysis. The objective was to corroborate the therapeutic efficacy of HYP in AD and PD models and to synthesize its potential therapeutic mechanisms. Searches were conducted in the PubMed, CNKI, and Web of Science databases. Reliability assessment of the 17 included studies confirmed the credibility of the mechanisms of action of HYP against AD and PD. We systematically assessed the neuroprotective potential of HYP in in vivo and in vitro models of AD and PD. Our findings indicated that HYP can mitigate, intervene in, and treat AD and PD animal models and associated cells through various mechanisms, including anti-oxidative, anti-inflammatory, anti-apoptotic, anti-Aβ aggregation, and cholinesterase inhibitory activities. Therefore, HYP potentially exerts anti-AD and anti-PD effects through diverse mechanisms, making it a promising candidate for therapeutic intervention in both AD and PD.
Collapse
Affiliation(s)
- Jiayu Yuan
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Siyu Zhou
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
15
|
Aschner M, Skalny AV, Santamaria A, Rocha JBT, Mansouri B, Tizabi Y, Madeddu R, Lu R, Lee E, Tinkov AA. Epigenetic Mechanisms of Aluminum-Induced Neurotoxicity and Alzheimer's Disease: A Focus on Non-Coding RNAs. Neurochem Res 2024; 49:2988-3005. [PMID: 39060769 DOI: 10.1007/s11064-024-04214-9] [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: 04/12/2024] [Revised: 06/16/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
Aluminum (Al) is known to induce neurotoxic effects, potentially contributing to Alzheimer's disease (AD) pathogenesis. Recent studies suggest that epigenetic modification may contribute to Al neurotoxicity, although the mechanisms are still debatable. Therefore, the objective of the present study was to summarize existing data on the involvement of epigenetic mechanisms in Al-induced neurotoxicity, especially AD-type pathology. Existing data demonstrate that Al exposure induces disruption in DNA methylation, histone modifications, and non-coding RNA expression in brains. Alterations in DNA methylation following Al exposure were shown to be mediated by changes in expression and activity of DNA methyltransferases (DNMTs) and ten-eleven translocation proteins (TETs). Al exposure was shown to reduce histone acetylation by up-regulating expression of histone deacetylases (HDACs) and impair histone methylation, ultimately contributing to down-regulation of brain-derived neurotrophic factor (BDNF) expression and activation of nuclear factor κB (NF-κB) signaling. Neurotoxic effects of Al exposure were also associated with aberrant expression of non-coding RNAs, especially microRNAs (miR). Al-induced patterns of miR expression were involved in development of AD-type pathology by increasing amyloid β (Aβ) production through up-regulation of Aβ precursor protein (APP) and β secretase (BACE1) expression (down-regulation of miR-29a/b, miR-101, miR-124, and Let-7c expression), increasing in neuroinflammation through NF-κB signaling (up-regulation of miR-9, miR-125b, miR-128, and 146a), as well as modulating other signaling pathways. Furthermore, reduced global DNA methylation, altered histone modification, and aberrant miRNA expression were associated with cognitive decline in Al-exposed subjects. However, further studies are required to evaluate the contribution of epigenetic mechanisms to Al-induced neurotoxicity and/or AD development.
Collapse
Affiliation(s)
- Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Anatoly V Skalny
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Sovetskaya Str. 14, Yaroslavl, 150000, Russia
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Bolshaya Pirogovskaya St., 2-4, Moscow, 119146, Russia
| | - Abel Santamaria
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, 04960, Mexico
| | - Joao B T Rocha
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Borhan Mansouri
- Substance Abuse Prevention Research Center, Research Institute for Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, 20059, USA
| | - Roberto Madeddu
- Department of Biomedical Sciences-Histology, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Rongzu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, People's Republic of China
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Alexey A Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Sovetskaya Str. 14, Yaroslavl, 150000, Russia.
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Bolshaya Pirogovskaya St., 2-4, Moscow, 119146, Russia.
| |
Collapse
|
16
|
Parra Bravo C, Naguib SA, Gan L. Cellular and pathological functions of tau. Nat Rev Mol Cell Biol 2024; 25:845-864. [PMID: 39014245 DOI: 10.1038/s41580-024-00753-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 07/18/2024]
Abstract
Tau protein is involved in various cellular processes, including having a canonical role in binding and stabilization of microtubules in neurons. Tauopathies are neurodegenerative diseases marked by the abnormal accumulation of tau protein aggregates in neurons, as seen, for example, in conditions such as frontotemporal dementia and Alzheimer disease. Mutations in tau coding regions or that disrupt tau mRNA splicing, tau post-translational modifications and cellular stress factors (such as oxidative stress and inflammation) increase the tendency of tau to aggregate and interfere with its clearance. Pathological tau is strongly implicated in the progression of neurodegenerative diseases, and the propagation of tau aggregates is associated with disease severity. Recent technological advancements, including cryo-electron microscopy and disease models derived from human induced pluripotent stem cells, have increased our understanding of tau-related pathology in neurodegenerative conditions. Substantial progress has been made in deciphering tau aggregate structures and the molecular mechanisms that underlie protein aggregation and toxicity. In this Review, we discuss recent insights into the diverse cellular functions of tau and the pathology of tau inclusions and explore the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Celeste Parra Bravo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Sarah A Naguib
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
17
|
Ren H, Tang L, Yuan Z, Liu Y, Zhou X, Xiao X, Wu X, Chen W, Chen Y, Wang H, Xue Q, Xu X. Combined administration of catalpol, puerarin, gastrodin, and borneol modulates the Tlr4/Myd88/NF-κB signaling pathway and alleviates microglia inflammation in Alzheimer's disease. Front Pharmacol 2024; 15:1492237. [PMID: 39545064 PMCID: PMC11560463 DOI: 10.3389/fphar.2024.1492237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder affecting millions of people worldwide, with no effective treatment currently available. In recent decades, various traditional Chinese medicines (TCMs) and their active ingredients have shown the potential to attenuate the pathogenesis of AD in cellular and animal models. However, the effects of TCM formulas, which are typically administered in practice, have been less studied. This study aims to investigate the therapeutic effects of several formulas consisting of 4 components herbal components: catalpol, puerarin, gastrodin, and borneol, on streptozotocin (STZ)-induced AD models in cells and rats. The new object recognition (NOR), elevated plus maze (EMP), and Morris water maze (MWM) tests were used to evaluate the cognitive functions of rats. Golgi staining, Haematoxylin and Eosin (HE) staining, and Nissl staining analyses were employed assess the physiology of hippocampal tissues. Gene expression profiles were analyzed used transcriptomics and reverse transcription quantitative polymerase chain reaction analysis, while protein expression levels were determined using immunoblotting, immunohistochemical, and immunofluorescence. The production of cytokines was evaluated with enzyme-linked immunosorbent assay. The results demonstrated that the combined administration of these 4 components (CPGB) had superior mitigating effects on AD cell model, as evidenced by the reduced pro-inflammatory cytokine production and decreased deposition of Aβ protein. Further in vivo and in vitro experiments confirmed that varying doses of CPGB formula effectively ameliorated STZ-induced cognitive deficits, as shown by NOR, MWM, and EMP tests, as well as pathological changes in hippocampal tissues and a 3-dimensional brain neurovascular unit (3D-NVU) model, including decreased deposition of Aβ protein and formation of plaques. Transcriptome sequencing and analysis identified 35 genes with significantly altered expression levels due to STZ and CPGB treatment in hippocampal tissues, which were enriched in the Tlr4/Myd88/NF-κB signaling pathway. Interference with this pathway significantly influenced the therapeutic effects of CPGB in the 3D-NVU model. Collectively, these findings suggest that the combined administration of catalpol, puerarin, gastrodin, and borneol offers superior therapeutic effects on AD by modulating the Tlr4/Myd88/NF-κB signaling pathway. This study strengthens the theoretical foundation for using TCMs to treat AD, proving new insights and references for alleviating and treating AD.
Collapse
Affiliation(s)
- Huijing Ren
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
- Tongren Polytechnic College, Tongren, Guizhou, China
| | - Ling Tang
- Shapingba District People’s Hospital of Chongqing, Chongqing, China
| | - Zhiying Yuan
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Yang Liu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xuejiao Zhou
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xiao Xiao
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xingyu Wu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Weihai Chen
- Faculty of Psychology, Southwest University, Chongqing, China
| | - Yi Chen
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Hongjin Wang
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Qiang Xue
- Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Xiaoyu Xu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
- Southwest University Hospital, Chongqing, China
- Chongqing Key Laboratory of New Drug Screening form Traditional Chinese Medicine, Chongqing, China
- Key Disciplines of Traditional Chinese Medicine of Chongqing City, Rehabilitation Medicine of Southwest University, Chongqing, China
| |
Collapse
|
18
|
Isidro F. Brain aging and Alzheimer's disease, a perspective from non-human primates. Aging (Albany NY) 2024; 16:13145-13171. [PMID: 39475348 PMCID: PMC11552644 DOI: 10.18632/aging.206143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 06/03/2024] [Indexed: 11/07/2024]
Abstract
Brain aging is compared between Cercopithecinae (macaques and baboons), non-human Hominidae (chimpanzees, orangutans, and gorillas), and their close relative, humans. β-amyloid deposition in the form of senile plaques (SPs) and cerebral β-amyloid angiopathy (CAA) is a frequent neuropathological change in non-human primate brain aging. SPs are usually diffuse, whereas SPs with dystrophic neurites are rare. Tau pathology, if present, appears later, and it is generally mild or moderate, with rare exceptions in rhesus macaques and chimpanzees. Behavior and cognitive impairment are usually mild or moderate in aged non-human primates. In contrast, human brain aging is characterized by early tau pathology manifested as neurofibrillary tangles (NFTs), composed of paired helical filaments (PHFs), progressing from the entorhinal cortex, hippocampus, temporal cortex, and limbic system to other brain regions. β-amyloid pathology appears decades later, involves the neocortex, and progresses to the paleocortex, diencephalon, brain stem, and cerebellum. SPs with dystrophic neurites containing PHFs and CAA are common. Cognitive impairment and dementia of Alzheimer's type occur in about 1-5% of humans aged 65 and about 25% aged 85. In addition, other proteinopathies, such as limbic-predominant TDP-43 encephalopathy, amygdala-predominant Lewy body disease, and argyrophilic grain disease, primarily affecting the archicortex, paleocortex, and amygdala, are common in aged humans but non-existent in non-human primates. These observations show that human brain aging differs from brain aging in non-human primates, and humans constitute the exception among primates in terms of severity and extent of brain aging damage.
Collapse
Affiliation(s)
- Ferrer Isidro
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Barcelona, Spain
- Reial Acadèmia de Medicina de Catalunya, Barcelona, Spain
| |
Collapse
|
19
|
Kitani A, Matsui Y. Integrative Network Analysis Reveals Novel Moderators of Aβ-Tau Interaction in Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.599092. [PMID: 39554095 PMCID: PMC11565825 DOI: 10.1101/2024.06.14.599092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Background Although interactions between amyloid-beta and tau proteins have been implicated in Alzheimer's disease (AD), the precise mechanisms by which these interactions contribute to disease progression are not yet fully understood. Moreover, despite the growing application of deep learning in various biomedical fields, its application in integrating networks to analyze disease mechanisms in AD research remains limited. In this study, we employed BIONIC, a deep learning-based network integration method, to integrate proteomics and protein-protein interaction data, with an aim to uncover factors that moderate the effects of the Aβ-tau interaction on mild cognitive impairment (MCI) and early-stage AD. Methods Proteomic data from the ROSMAP cohort were integrated with protein-protein interaction (PPI) data using a Deep Learning-based model. Linear regression analysis was applied to histopathological and gene expression data, and mutual information was used to detect moderating factors. Statistical significance was determined using the Benjamini-Hochberg correction (p < 0.05). Results Our results suggested that astrocytes and GPNMB+ microglia moderate the Aβ-tau interaction. Based on linear regression with histopathological and gene expression data, GFAP and IBA1 levels and GPNMB gene expression positively contributed to the interaction of tau with Aβ in non-dementia cases, replicating the results of the network analysis. Conclusions These findings indicate that GPNMB+ microglia moderate the Aβ-tau interaction in early AD and therefore are a novel therapeutic target. To facilitate further research, we have made the integrated network available as a visualization tool for the scientific community (URL: https://igcore.cloud/GerOmics/AlzPPMap).
Collapse
Affiliation(s)
- Akihiro Kitani
- Biomedical and Health Informatics Unit, Department of Integrated Health Science, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yusuke Matsui
- Biomedical and Health Informatics Unit, Department of Integrated Health Science, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, 461-8673 Nagoya, Aichi, Japan
| |
Collapse
|
20
|
Basheer N, Muhammadi MK, Freites CL, Avila M, Momand MUD, Hryntsova N, Smolek T, Katina S, Zilka N. TLR4-mediated chronic neuroinflammation has no effect on tangle pathology in a tauopathy mouse model. Front Aging Neurosci 2024; 16:1468602. [PMID: 39503044 PMCID: PMC11536299 DOI: 10.3389/fnagi.2024.1468602] [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: 07/22/2024] [Accepted: 08/26/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Alzheimer's disease (AD) is marked by the accumulation of fibrillary aggregates composed of pathological tau protein. Although neuroinflammation is frequently observed in conjunction with tau pathology, current preclinical evidence does not sufficiently establish a direct causal role in tau tangle formation. This study aimed to evaluate whether chronic Toll-like receptor 4 (TLR4) stimulation, induced by a high dose of lipopolysaccharide (LPS, 5 mg/kg), exacerbates neurofibrillary tangle (NFT) pathology in a transgenic mouse model of tauopathy that expresses human truncated 151-391/3R tau, an early feature of sporadic AD. Methods We utilized a transgenic mouse model of tauopathy subjected to chronic TLR4 stimulation via weekly intraperitoneal injections of LPS over nine consecutive weeks. Neurofibrillary tangle formation, microglial activation, and tau hyperphosphorylation in the brainstem and hippocampus were assessed through immunohistochemistry, immunofluorescence, and detailed morphometric analysis of microglia. Results Chronic LPS treatment led to a significant increase in the number of Iba-1+ microglia in the LPS-treated group compared to the sham group (p < 0.0001). Notably, there was a 1.5- to 1.7-fold increase in microglia per tangle-bearing neuron in the LPS-treated group. These microglia exhibited a reactive yet exhausted phenotype, characterized by a significant reduction in cell area (p < 0.0001) without significant changes in other morphometric parameters, such as perimeter, circumference, solidity, aspect ratio, or arborization degree. Despite extensive microglial activation, there was no observed reduction in tau hyperphosphorylation or a decrease in tangle formation in the brainstem, where pathology predominantly develops in this model. Discussion These findings suggest that chronic TLR4 stimulation in tau-transgenic mice results in significant microglial activation but does not influence tau tangle formation. This underscores the complexity of the relationship between neuroinflammation and tau pathology, indicating that additional mechanisms may be required for neuroinflammation to directly contribute to tau tangle formation.
Collapse
Affiliation(s)
- Neha Basheer
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | - Carlos Leandro Freites
- Institute of Histology and Embryology of Mendoza (IHEM), National University of Cuyo, National Scientific and Technical Research Council (CONICET), Mendoza, Argentina
| | - Martin Avila
- Institute of Histology and Embryology of Mendoza (IHEM), National University of Cuyo, National Scientific and Technical Research Council (CONICET), Mendoza, Argentina
| | - Miraj Ud Din Momand
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Natalia Hryntsova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomas Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Stanislav Katina
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Mathematics and Statistics, Faculty of Science, Masaryk University, Brno, Czechia
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
21
|
Deng X, Qiu Z, Chen X, Liu J, Wang X, Li J, Zhang J, Cui X, Fu Y, Jiang M. Exploring the potential mechanism of ginsenoside Rg1 to regulate ferroptosis in Alzheimer's disease based on network pharmacology. Eur J Pharmacol 2024; 979:176859. [PMID: 39067563 DOI: 10.1016/j.ejphar.2024.176859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVES To explore the pathogenesis of Alzheimer's disease (AD), the potential targets and signaling pathways of ginsenoside Rg1 against AD were investigated by network pharmacology. METHODS Ginsenoside Rg1 targets were identified through PubChem, PharmMapper, and Uniprot databases, while the GeneCards database was used to examine the respective targets of amyloid precursor protein (APP) and AD. Then, the common targets between ginsenoside Rg1 and APP were explored by the Venny tool, the interaction network diagram between the active components and the targets was built via Cytoscape software, as well as GO enrichment and KEGG pathway annotation analysis were performed. Furthermore, genes associated with ferroptosis were found by the GeneCards and FerrDb databases. Besides, the connection among ginsenoside Rg1, APP, ferroptosis, and AD was predicted and analyzed. Finally, the effects of ginsenosides Rg1 and liproxstain-1 on the proliferation and differentiation of APP/PS1 mice were evaluated by immunohistochemistry. RESULTS Ginsenoside Rg1, APP, ferroptosis, and AD had 12 hub genes. GO enrichment and KEGG pathway annotation analysis showed that EGFR, SRC, protein hydrolysis, protein phosphorylation, the Relaxin pathway, and the FoxO signaling pathway play an important role in the potential mechanism of ginsenoside Rg1's under regulation of ferroptosis anti-AD through the modulation of APP-related signaling pathways. The APP/PS1 mice experiment verified that ginsenosides Rg1 and liproxstain-1 can promote the proliferation and differentiation. CONCLUSION Ginsenoside Rg1, APP and ferroptosis may act on EGFR, SRC, the Relaxin and FoxO signaling pathways to regulate protein metabolism, protein phosphorylation and other pathways to improve AD symptoms.
Collapse
Affiliation(s)
- Xu Deng
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Zixiong Qiu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaoshuai Chen
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiangxiu Liu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaowei Wang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jie Li
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiankai Zhang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaojun Cui
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Yuan Fu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Mei Jiang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
22
|
Lin N, Gao XY, Li X, Chu WM. Involvement of ubiquitination in Alzheimer's disease. Front Neurol 2024; 15:1459678. [PMID: 39301473 PMCID: PMC11412110 DOI: 10.3389/fneur.2024.1459678] [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: 07/04/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024] Open
Abstract
The hallmark pathological features of Alzheimer's disease (AD) consist of senile plaques, which are formed by extracellular β-amyloid (Aβ) deposition, and neurofibrillary tangles, which are formed by the hyperphosphorylation of intra-neuronal tau proteins. With the increase in clinical studies, the in vivo imbalance of iron homeostasis and the dysfunction of synaptic plasticity have been confirmed to be involved in AD pathogenesis. All of these mechanisms are constituted by the abnormal accumulation of misfolded or conformationally altered protein aggregates, which in turn drive AD progression. Proteostatic imbalance has emerged as a key mechanism in the pathogenesis of AD. Ubiquitination modification is a major pathway for maintaining protein homeostasis, and protein degradation is primarily carried out by the ubiquitin-proteasome system (UPS). In this review, we provide an overview of the ubiquitination modification processes and related protein ubiquitination degradation pathways in AD, focusing on the microtubule-associated protein Tau, amyloid precursor protein (APP), divalent metal transporter protein 1 (DMT1), and α-amino-3-hyroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors. We also discuss recent advances in ubiquitination-based targeted therapy for AD, with the aim of contributing new ideas to the development of novel therapeutic interventions for AD.
Collapse
Affiliation(s)
- Nan Lin
- College of Acupuncture and Tuina of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xi-Yan Gao
- The Third Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiao Li
- College of Acupuncture and Tuina of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Wen-Ming Chu
- College of Acupuncture and Tuina of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
23
|
Lan WH, He H, Bayley H, Qing Y. Location of Phosphorylation Sites within Long Polypeptide Chains by Binder-Assisted Nanopore Detection. J Am Chem Soc 2024; 146:24265-24270. [PMID: 38986019 PMCID: PMC11378271 DOI: 10.1021/jacs.4c03912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
The detection and mapping of protein phosphorylation sites are essential for understanding the mechanisms of various cellular processes and for identifying targets for drug development. The study of biopolymers at the single-molecule level has been revolutionized by nanopore technology. In this study, we detect protein phosphorylation within long polypeptides (>700 amino acids), after the attachment of binders that interact with phosphate monoesters; electro-osmosis is used to drive the tagged chains through engineered protein nanopores. By monitoring the ionic current carried by a nanopore, phosphorylation sites are located within individual polypeptide chains, providing a valuable step toward nanopore proteomics.
Collapse
Affiliation(s)
- Wei-Hsuan Lan
- Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K
| | - Hanxiao He
- Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K
| | - Hagan Bayley
- Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K
| | - Yujia Qing
- Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, U.K
| |
Collapse
|
24
|
León-Espinosa G, Murillo AMM, Turegano-Lopez M, DeFelipe J, Holgado M. Phosphorylated Tau at T181 accumulates in the serum of hibernating Syrian hamsters and rapidly disappears after arousal. Sci Rep 2024; 14:20562. [PMID: 39232030 PMCID: PMC11375040 DOI: 10.1038/s41598-024-71481-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024] Open
Abstract
The search for biomarkers for the early diagnosis of neurodegenerative diseases is a growing area. Numerous investigations are exploring minimally invasive and cost-effective biomarkers, with the detection of phosphorylated Tau (pTau) protein emerging as one of the most promising fields. pTau is the main component of the paired helical filaments found in the brains of Alzheimer's disease cases and serves as a precursor in the formation of neurofibrillary tangles (NFTs). Recent research has revealed that analysis of p-Tau181, p-Tau217 and p-Tau231 in blood may be an option for detecting the preclinical stage of Alzheimer's disease. In this study, we have analyzed the values of pTau 181 in the serum of Syrian hamsters during hibernation. Naturally, over the course of hibernation, these animals exhibit a reversible accumulation of pTau in the brain tissue, which rapidly disappears upon awakening. A biosensing system based on the interferometric optical detection method was used to measure the concentration of pTau181 protein in serum samples from Syrian hamsters. This method eliminates the matrix effect and amplifies the signal obtained by using silicon dioxide nanoparticles (SiO2 NPs) biofunctionalized with the αpTau181 antibody. Our results indicate a substantial increase in the serum concentration of pTau in threonine-181 during hibernation, which disappears completely 2-3 h after awakening. Investigating the mechanism by which pTau protein appears in the blood non-pathologically may enhance current diagnostic techniques. Furthermore, since this process is reversible, and no tangles are detected in the brains of hibernating hamsters, additional analysis may contribute to the discovery of improved biomarkers. Additionally, exploring drugs targeting pTau to prevent the formation of tangles or studying the outcomes of any pTau-targeted treatment could be valuable.
Collapse
Affiliation(s)
- G León-Espinosa
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28660, Boadilla del Monte, Madrid, Spain.
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain.
| | - A M M Murillo
- Optics, Photonics and Biophotonics Group, Centre for Biomedical Technology, Campus de Montegancedo Universidad Politécnica de Madrid, 28223, Pozuelo de Alarcón, Madrid, Spain
- Group of Organ and Tissue on-a-chip and In-Vitro Detection, Health Research Institute of the Hospital Clínico San Carlos, IdISSC. C/Profesor Martín Lagos s/n, 4ª Planta Sur, 28040, Madrid, Spain
| | - M Turegano-Lopez
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031, Madrid, Spain
| | - J DeFelipe
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28031, Madrid, Spain
| | - M Holgado
- Optics, Photonics and Biophotonics Group, Centre for Biomedical Technology, Campus de Montegancedo Universidad Politécnica de Madrid, 28223, Pozuelo de Alarcón, Madrid, Spain
- Group of Organ and Tissue on-a-chip and In-Vitro Detection, Health Research Institute of the Hospital Clínico San Carlos, IdISSC. C/Profesor Martín Lagos s/n, 4ª Planta Sur, 28040, Madrid, Spain
- Applied Physics and Materials Engineering Department, Escuela Técnica Superior de Ingenieros Industriales, Universidad Politécnica de Madrid, C/José Gutierrez Abascal, 2, 28006, Madrid, Spain
| |
Collapse
|
25
|
Deng Q, Zhang J, Liu J, Liu Y, Dai Z, Zou X, Li Z. Identifying Protein Phosphorylation Site-Disease Associations Based on Multi-Similarity Fusion and Negative Sample Selection by Convolutional Neural Network. Interdiscip Sci 2024; 16:649-664. [PMID: 38457108 DOI: 10.1007/s12539-024-00615-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 03/09/2024]
Abstract
As one of the most important post-translational modifications (PTMs), protein phosphorylation plays a key role in a variety of biological processes. Many studies have shown that protein phosphorylation is associated with various human diseases. Therefore, identifying protein phosphorylation site-disease associations can help to elucidate the pathogenesis of disease and discover new drug targets. Networks of sequence similarity and Gaussian interaction profile kernel similarity were constructed for phosphorylation sites, as well as networks of disease semantic similarity, disease symptom similarity and Gaussian interaction profile kernel similarity were constructed for diseases. To effectively combine different phosphorylation sites and disease similarity information, random walk with restart algorithm was used to obtain the topology information of the network. Then, the diffusion component analysis method was utilized to obtain the comprehensive phosphorylation site similarity and disease similarity. Meanwhile, the reliable negative samples were screened based on the Euclidean distance method. Finally, a convolutional neural network (CNN) model was constructed to identify potential associations between phosphorylation sites and diseases. Based on tenfold cross-validation, the evaluation indicators were obtained including accuracy of 93.48%, specificity of 96.82%, sensitivity of 90.15%, precision of 96.62%, Matthew's correlation coefficient of 0.8719, area under the receiver operating characteristic curve of 0.9786 and area under the precision-recall curve of 0.9836. Additionally, most of the top 20 predicted disease-related phosphorylation sites (19/20 for Alzheimer's disease; 20/16 for neuroblastoma) were verified by literatures and databases. These results show that the proposed method has an outstanding prediction performance and a high practical value.
Collapse
Affiliation(s)
- Qian Deng
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jing Zhang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jie Liu
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yuqi Liu
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zong Dai
- School of Biomedical Engineering, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Xiaoyong Zou
- School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, China.
| | - Zhanchao Li
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
26
|
Shen Y, Liu F, Zhang M. Therapeutic potential of plant-derived natural compounds in Alzheimer's disease: Targeting microglia-mediated neuroinflammation. Biomed Pharmacother 2024; 178:117235. [PMID: 39094545 DOI: 10.1016/j.biopha.2024.117235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
Microglia are resident immune cells of the central nervous system (CNS) with roles in sensing, housekeeping, and defense. Exploring the role of microglia in the occurrence and development of Alzheimer's disease (AD) and the possible therapeutic mechanism of plant-derived natural compounds (PDNCs) that regulate microglia-associated neuroinflammation may potentially help in elucidating the pathogenesis of AD and provide novel insights for its treatment. This review explores the role of abnormal microglial activation and its dominant neuroinflammatory response, as well as the activation of their target receptors and signaling pathways in AD pathogenesis. Additionally, we report an update on the potential pharmacological mechanisms of multiple PDNCs in modulating microglia-associated neuroinflammation in AD treatment. Dysregulated activation of microglial receptors and their downstream pathways impaired immune homeostasis in animal models of AD. Multiple signaling pathways, such as mitogen-activated protein kinase (MAPK), nuclear factor kappa light chain enhancer of activated B cells (NF-κB), and Toll-like receptors, play important roles in microglial activation and can exacerbate microglia-mediated neuroinflammation. PDNCs, such as magnolol, stigmasterol, matrine, naringenin, naringin, and resveratrol, can delay the progression of AD by inhibiting the proinflammatory receptors of microglia, activating its anti-inflammatory receptors, regulating the receptors related to β-amyloid (Aβ) clearance, reversing immune dysregulation, and maintaining the immune homeostasis of microglial downstream pathways. This review summarizes the mechanisms by which microglia cause chronic inflammation in AD and evaluates the beneficial effects of PDNCs on immune regulation in AD by regulating microglial receptors and their downstream pathways.
Collapse
Affiliation(s)
- Yanyan Shen
- Department of Neurosurgery, Shengjing Hospital of China Medical University, China.
| | - Fang Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, China
| | - Mingjie Zhang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
27
|
Escamilla S, Salas-Lucia F. Thyroid Hormone and Alzheimer Disease: Bridging Epidemiology to Mechanism. Endocrinology 2024; 165:bqae124. [PMID: 39276028 DOI: 10.1210/endocr/bqae124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/12/2024] [Accepted: 09/12/2024] [Indexed: 09/16/2024]
Abstract
The identification of critical factors that can worsen the mechanisms contributing to the pathophysiology of Alzheimer disease is of paramount importance. Thyroid hormones (TH) fit this criterion. Epidemiological studies have identified an association between altered circulating TH levels and Alzheimer disease. The study of human and animal models indicates that TH can affect all the main cellular, molecular, and genetic mechanisms known as hallmarks of Alzheimer disease. This is true not only for the excessive production in the brain of protein aggregates leading to amyloid plaques and neurofibrillary tangles but also for the clearance of these molecules from the brain parenchyma via the blood-brain barrier and for the escalated process of neuroinflammation-and even for the effects of carrying Alzheimer-associated genetic variants. Suboptimal TH levels result in a greater accumulation of protein aggregates in the brain. The direct TH regulation of critical genes involved in amyloid beta production and clearance is remarkable, affecting the expression of multiple genes, including APP (related to amyloid beta production), APOE, LRP1, TREM2, AQP4, and ABCB1 (related to amyloid beta clearance). TH also affects microglia by increasing their migration and function and directly regulating the immunosuppressor gene CD73, impacting the immune response of these cells. Studies aiming to understand the mechanisms that could explain how changes in TH levels can contribute to the brain alterations seen in patients with Alzheimer disease are ongoing. These studies have potential implications for the management of patients with Alzheimer disease and ultimately can contribute to devising new interventions for these conditions.
Collapse
Affiliation(s)
- Sergio Escamilla
- Instituto de Neurociencias, CSIC-Universidad Miguel Hernández, Alicante 03550, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Alicante 03550, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante 03010, Spain
| | | |
Collapse
|
28
|
Fryncel A, Madetko-Alster N, Krępa Z, Kuch M, Alster P. The Possible Associations between Tauopathies and Atherosclerosis, Diabetes Mellitus, Dyslipidemias, Metabolic Syndrome and Niemann-Pick Disease. Diagnostics (Basel) 2024; 14:1831. [PMID: 39202319 PMCID: PMC11354139 DOI: 10.3390/diagnostics14161831] [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: 07/09/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Clinical evaluation and treatment of tauopathic syndromes remain a challenge. There is a growing interest in theories concerning their possible associations with metabolic diseases. The possible connection between those diseases might be linked with cerebrovascular dysfunction. The endothelial cell damage and impairment of the blood-brain barrier observed in atherosclerosis or diabetes may play a role in contributing to tauopathic syndrome development. Additionally, the inflammation evoked by pathological metabolic changes may also be involved in this process. Multiple cases indicate the coexistence of metabolic disorders and tauopathic syndromes. These findings suggest that modifying the evolution of metabolic and cerebrovascular diseases may impact the course of neurodegenerative diseases. Obtained data could indicate the possible benefits of introducing routine carotid artery sonography, revascularization operation or antihypertensive medications among patients at high risk for tauopathies. This review has identified this understudied area, which is currently associated with several diseases for which there is no treatment. Due to the pathomechanisms linking metabolic diseases and tauopathies, further investigation of this area of research, including cohort studies, is recommended and may provide new pharmacological perspectives for treatment.
Collapse
Affiliation(s)
- Aleksandra Fryncel
- Students’ Scientific Circle, Department of Neurology, Mazovian Brodno Hospital, Medical University of Warsaw, Ludwika Kondratowicza 8, 03-242 Warsaw, Poland
| | - Natalia Madetko-Alster
- Department of Neurology, Mazovian Brodno Hospital, Medical University of Warsaw, Ludwika Kondratowicza 8, 03-242 Warsaw, Poland; (N.M.-A.); (P.A.)
| | - Zuzanna Krępa
- Department of Cardiology, Hypertension and Internal Disease, Mazovian Brodno Hospital, Medical University of Warsaw, Ludwika Kondratowicza 8, 03-242 Warsaw, Poland; (Z.K.); (M.K.)
| | - Marek Kuch
- Department of Cardiology, Hypertension and Internal Disease, Mazovian Brodno Hospital, Medical University of Warsaw, Ludwika Kondratowicza 8, 03-242 Warsaw, Poland; (Z.K.); (M.K.)
| | - Piotr Alster
- Department of Neurology, Mazovian Brodno Hospital, Medical University of Warsaw, Ludwika Kondratowicza 8, 03-242 Warsaw, Poland; (N.M.-A.); (P.A.)
| |
Collapse
|
29
|
Cheng F, Fransson LÅ, Mani K. Interplay between glypican-1, amyloid-β and tau phosphorylation in human neural stem cells. Neuroscience 2024; 553:121-127. [PMID: 38992568 DOI: 10.1016/j.neuroscience.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/19/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is characterized by accumulation of amyloid beta (Aβ) and hyperphosphorylated tau (Tau-P) in the brain. Aβ enhances the activity of kinases involved in the formation of Tau-P. Phosphorylation at Thr 181 determines the propagation of multiple tau phosphorylations. Aβ is derived from the amyloid precursor protein (APP). Cleavage of APP by β-secretase also initiates release of heparan sulfate (HS) from the proteoglycan glypican-1 (GPC1). OBJECTIVES In this study, we have explored possible connections between GPC1 expression, HS release, APP processing and Tau-P formation in human neural stem cells. METHODS GPC1 formation was suppressed by using CRISPR/Cas9 and increased by using a vector encoding GPC1. HS release from GPC1 was increased by growing cells in medium containing Arg and ascorbate. Effects were monitored by immunofluorescence microscopy and slot immunoblotting using antibodies/antisera recognizing Aβ, GPC1, HS released from GPC1, total Tau, and Tau phosphorylated at Thr-181, 217 or 231. The latter have been used as blood biomarkers for AD. RESULTS Suppression of GPC1 expression resulted in increased phosphorylation at Thr 181 and Thr 217. When GPC1 was overexpressed, phosphorylation at Thr 217 decreased. Stimulation of HS release from GPC1 diminished tau phosphorylation at all of the three Thr positions, while expression of GPC1 was unaffected. Simultaneous stimulation of HS release and APP processing by the cytokine TNF-α also suppressed tau phosphorylation. CONCLUSION The increased release of GPC1-derived HS may interfere with Aβ formation and/or Aβ interaction with tau.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Experimental Medical Science, Division of Neuroscience, Glycobiology Group, Lund University, Biomedical Center A13, SE-221 84 Lund, Sweden
| | - Lars-Åke Fransson
- Department of Experimental Medical Science, Division of Neuroscience, Glycobiology Group, Lund University, Biomedical Center A13, SE-221 84 Lund, Sweden
| | - Katrin Mani
- Department of Experimental Medical Science, Division of Neuroscience, Glycobiology Group, Lund University, Biomedical Center A13, SE-221 84 Lund, Sweden.
| |
Collapse
|
30
|
Meng L, Wang B, Zhang S, Zhang S, Cai T, Ding CF, Yan Y. One-step fabrication of dipeptide-based bifunctional polymer for individual enrichment of glycopeptides and phosphopeptides from serum. J Chromatogr A 2024; 1730:465173. [PMID: 39025024 DOI: 10.1016/j.chroma.2024.465173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
A dipeptide-based bifunctional material immobilized with Ti4+ (denoted as APE-MBA-VPA-Ti4+) was developed using precipitation polymerization. This polymer combines hydrophilic interaction liquid chromatography (HILIC) and immobilized metal affinity chromatography (IMAC) enrichment strategies, allowing for the individual and simultaneous enrichment of glycopeptides and phosphopeptides. It demonstrated high sensitivity (0.1 fmol μL-1 for glycopeptides, 0.005 fmol μL-1 for phosphopeptides), strong selectivity (molar ratio HRP: BSA = 1:1000, β-casein: BSA = 1:2500), consistent reusability (10 cycles) and satisfactory recovery rate (93.5 ± 1.8 % for glycopeptides, 91.6 ± 0.6 % for phosphopeptides) in the individual enrichment. Utilizing nano LC-MS/MS technology, the serum of liver cancer patients was analyzed after enrichment individually, resulting in the successful capture of 333 glycopeptides covering 262 glycosylation sites, corresponding to 131 glycoproteins, as well as 67 phosphopeptides covering 57 phosphorylation sites, related to 48 phosphoproteins. In comparison, the serum of normal healthy individuals yielded a total of 283 glycopeptides covering 244 glycosylation sites corresponding to 126 glycoproteins, as well as 66 phosphopeptides covering 56 phosphorylation sites related to 37 phosphoproteins. Label-free quantification identified 10 differentially expressed glycoproteins and 8 differentially expressed phosphoproteins in the serum of liver cancer patients. Among them, glycoproteins (HP, BCHE, AGT, C3, and PROC) and phosphoproteins (ZYX, GOLM1, GP1BB, CLU, and TNXB) showed upregulation and displayed potential as biomarkers for liver cancer.
Collapse
Affiliation(s)
- Luyan Meng
- Ningbo No.2 Hospital, Ningbo, Zhejiang, 315099, PR China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang, 315020, PR China
| | - Bing Wang
- School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang, 315211, PR China
| | - Sijia Zhang
- Ningbo No.2 Hospital, Ningbo, Zhejiang, 315099, PR China
| | - Shun Zhang
- Ningbo No.2 Hospital, Ningbo, Zhejiang, 315099, PR China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang, 315020, PR China
| | - Ting Cai
- Ningbo No.2 Hospital, Ningbo, Zhejiang, 315099, PR China; Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang, 315020, PR China.
| | - Chuan-Fan Ding
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang, 315020, PR China; School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang, 315211, PR China.
| | - Yinghua Yan
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang, 315020, PR China; School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang, 315211, PR China.
| |
Collapse
|
31
|
Wang L, Tang Z, Li B, Peng Y, Yang X, Xiao Y, Ni R, Qi XL. Myricetin ameliorates cognitive impairment in 3×Tg Alzheimer's disease mice by regulating oxidative stress and tau hyperphosphorylation. Biomed Pharmacother 2024; 177:116963. [PMID: 38889642 DOI: 10.1016/j.biopha.2024.116963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Alzheimer's disease is characterized by abnormal β-amyloid (Aβ) plaque accumulation, tau hyperphosphorylation, reactive oxidative stress, mitochondrial dysfunction and synaptic loss. Myricetin, a dietary flavonoid, has been shown to exert neuroprotective effects in vitro and in vivo. Here, we aimed to elucidate the mechanism and pathways involved in the protective effect of myricetin. METHODS The effect of myricetin was assessed on Aβ42 oligomer-treated neuronal SH-SY5Y cells and in 3×Tg mice. Behavioral tests were performed to assess the cognitive effects of myricetin (14 days, ip) in 3×Tg mice. The levels of beta-amyloid precursor protein (APP), synaptic and mitochondrial proteins, glycogen synthase kinase3β (GSK3β) and extracellular regulated kinase (ERK) 2 were assessed via Western blotting. Flow cytometry assays, immunofluorescence staining, and transmission electron microscopy were used to assess mitochondrial dysfunction and reactive oxidative stress. RESULTS We found that, compared with control treatment, myricetin treatment improved spatial cognition and learning and memory in 3×Tg mice. Myricetin ameliorated tau phosphorylation and the reduction in pre- and postsynaptic proteins in Aβ42 oligomer-treated neuronal SH-SY5Y cells and in 3×Tg mice. In addition, myricetin reduced reactive oxygen species generation, lipid peroxidation, and DNA oxidation, and rescued mitochondrial dysfunction via the associated GSK3β and ERK 2 signalling pathways. CONCLUSIONS This study provides new insight into the neuroprotective mechanism of myricetin in vitro in cell culture and in vivo in a mouse model of Alzheimer's disease.
Collapse
Affiliation(s)
- Li Wang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Zhi Tang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Bo Li
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Yaqian Peng
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Xi Yang
- Guiyang Healthcare Vocational University, Guizhou ERC for Medical Resources & Healthcare Products (Guizhou Engineering Research Center for Medical Resources and Healthcare Products), Guiyang, Guizhou, China
| | - Yan Xiao
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland; Institute for Biomedical Engineering, ETH Zurich & University of Zurich, Zurich, Switzerland.
| | - Xiao-Lan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Key Laboratory of Molecular Biology of Guizhou Medical University, Guiyang, China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Constructed by the Province and Ministry, Guiyang, China.
| |
Collapse
|
32
|
Ramazi S, Dadzadi M, Darvazi M, Seddigh N, Allahverdi A. Protein modification in neurodegenerative diseases. MedComm (Beijing) 2024; 5:e674. [PMID: 39105197 PMCID: PMC11298556 DOI: 10.1002/mco2.674] [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: 12/05/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Posttranslational modifications play a crucial role in governing cellular functions and protein behavior. Researchers have implicated dysregulated posttranslational modifications in protein misfolding, which results in cytotoxicity, particularly in neurodegenerative diseases such as Alzheimer disease, Parkinson disease, and Huntington disease. These aberrant posttranslational modifications cause proteins to gather in certain parts of the brain that are linked to the development of the diseases. This leads to neuronal dysfunction and the start of neurodegenerative disease symptoms. Cognitive decline and neurological impairments commonly manifest in neurodegenerative disease patients, underscoring the urgency of comprehending the posttranslational modifications' impact on protein function for targeted therapeutic interventions. This review elucidates the critical link between neurodegenerative diseases and specific posttranslational modifications, focusing on Tau, APP, α-synuclein, Huntingtin protein, Parkin, DJ-1, and Drp1. By delineating the prominent aberrant posttranslational modifications within Alzheimer disease, Parkinson disease, and Huntington disease, the review underscores the significance of understanding the interplay among these modifications. Emphasizing 10 key abnormal posttranslational modifications, this study aims to provide a comprehensive framework for investigating neurodegenerative diseases holistically. The insights presented herein shed light on potential therapeutic avenues aimed at modulating posttranslational modifications to mitigate protein aggregation and retard neurodegenerative disease progression.
Collapse
Affiliation(s)
- Shahin Ramazi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Maedeh Dadzadi
- Department of BiotechnologyFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mona Darvazi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Nasrin Seddigh
- Department of BiochemistryFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Abdollah Allahverdi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| |
Collapse
|
33
|
Canet G, Gratuze M, Zussy C, Bouali ML, Diaz SD, Rocaboy E, Laliberté F, El Khoury NB, Tremblay C, Morin F, Calon F, Hébert SS, Julien C, Planel E. Age-dependent impact of streptozotocin on metabolic endpoints and Alzheimer's disease pathologies in 3xTg-AD mice. Neurobiol Dis 2024; 198:106526. [PMID: 38734152 DOI: 10.1016/j.nbd.2024.106526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/13/2024] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease with a complex origin, thought to involve a combination of genetic, biological and environmental factors. Insulin dysfunction has emerged as a potential factor contributing to AD pathogenesis, particularly in individuals with diabetes, and among those with insulin deficiency or undergoing insulin therapy. The intraperitoneal administration of streptozotocin (STZ) is widely used in rodent models to explore the impact of insulin deficiency on AD pathology, although prior research predominantly focused on young animals, with no comparative analysis across different age groups. Our study aimed to fill this gap by analyzing the impact of insulin dysfunction in 7 and 23 months 3xTg-AD mice, that exhibit both amyloid and tau pathologies. Our objective was to elucidate the age-specific consequences of insulin deficiency on AD pathology. STZ administration led to insulin deficiency in the younger mice, resulting in an increase in cortical amyloid-β (Aβ) and tau aggregation, while tau phosphorylation was not significantly affected. Conversely, older mice displayed an unexpected resilience to the peripheral metabolic impact of STZ, while exhibiting an increase in both tau phosphorylation and aggregation without significantly affecting amyloid pathology. These changes were paralleled with alterations in signaling pathways involving tau kinases and phosphatases. Several markers of blood-brain barrier (BBB) integrity declined with age in 3xTg-AD mice, which might have facilitated a direct neurotoxic effect of STZ in older mice. Overall, our research confirms the influence of insulin signaling dysfunction on AD pathology, but also advises careful interpretation of data related to STZ-induced effects in older animals.
Collapse
Affiliation(s)
- Geoffrey Canet
- Laval University, Faculty of Medicine, Neurosciences and Psychiatry department, Québec, QC G1V 0A6, Canada; Neurosciences axis, CHU de Québec Research Center, Québec, QC G1V 4G2, Canada.
| | - Maud Gratuze
- Laval University, Faculty of Medicine, Neurosciences and Psychiatry department, Québec, QC G1V 0A6, Canada; Institute of Neurophysiopathology (INP), University of Aix-Marseille, CNRS UMR 7051, 13385 Marseille, France.
| | - Charleine Zussy
- Laval University, Faculty of Medicine, Neurosciences and Psychiatry department, Québec, QC G1V 0A6, Canada; Neurosciences axis, CHU de Québec Research Center, Québec, QC G1V 4G2, Canada.
| | - Mohamed Lala Bouali
- Laval University, Faculty of Medicine, Neurosciences and Psychiatry department, Québec, QC G1V 0A6, Canada.
| | - Sofia Diego Diaz
- Laval University, Faculty of Medicine, Neurosciences and Psychiatry department, Québec, QC G1V 0A6, Canada.
| | - Emma Rocaboy
- Laval University, Faculty of Medicine, Neurosciences and Psychiatry department, Québec, QC G1V 0A6, Canada.
| | - Francis Laliberté
- Neurosciences axis, CHU de Québec Research Center, Québec, QC G1V 4G2, Canada
| | - Noura B El Khoury
- Laval University, Faculty of Medicine, Neurosciences and Psychiatry department, Québec, QC G1V 0A6, Canada; University of Balamand, Faculty of Arts and Sciences, Departement of Psychology, Tueini Building Kalhat, Al-Kurah, P.O. Box 100, Tripoli, Lebanon.
| | - Cyntia Tremblay
- Neurosciences axis, CHU de Québec Research Center, Québec, QC G1V 4G2, Canada.
| | - Françoise Morin
- Neurosciences axis, CHU de Québec Research Center, Québec, QC G1V 4G2, Canada.
| | - Frédéric Calon
- Neurosciences axis, CHU de Québec Research Center, Québec, QC G1V 4G2, Canada; Laval University, Faculty of Pharmacy, Neurosciences and Psychiatry department, Québec, QC G1V 0A6, Canada.
| | - Sébastien S Hébert
- Laval University, Faculty of Medicine, Neurosciences and Psychiatry department, Québec, QC G1V 0A6, Canada; Neurosciences axis, CHU de Québec Research Center, Québec, QC G1V 4G2, Canada.
| | - Carl Julien
- Research Center in Animal Sciences of Deschambault, Québec, QC G0A 1S0, Canada; Laval University, Faculty of Agricultural and Food Sciences, Québec, QC G1V 0A6, Canada.
| | - Emmanuel Planel
- Laval University, Faculty of Medicine, Neurosciences and Psychiatry department, Québec, QC G1V 0A6, Canada; Neurosciences axis, CHU de Québec Research Center, Québec, QC G1V 4G2, Canada.
| |
Collapse
|
34
|
Tang Z, Peng Y, Jiang Y, Wang L, Guo M, Chen Z, Luo C, Zhang T, Xiao Y, Ni R, Qi X. Gastrodin ameliorates synaptic impairment, mitochondrial dysfunction and oxidative stress in N2a/APP cells. Biochem Biophys Res Commun 2024; 719:150127. [PMID: 38761634 DOI: 10.1016/j.bbrc.2024.150127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
Alzheimer's disease is characterized by abnormal β-amyloid and tau accumulation, mitochondrial dysfunction, oxidative stress, and synaptic dysfunction. Here, we aimed to assess the mechanisms and signalling pathways in the neuroprotective effect of gastrodin, a phenolic glycoside, on murine neuroblastoma N2a cells expressing human Swedish mutant APP (N2a/APP). We found that gastrodin increased the levels of presynaptic-SNAP, synaptophysin, and postsynaptic-PSD95 and reduced phospho-tau Ser396, APP and Aβ1-42 levels in N2a/APP cells. Gastrodin treatment reduced reactive oxygen species generation, lipid peroxidation, mitochondrial fragmentation and DNA oxidation; restored mitochondrial membrane potential and intracellular ATP production. Upregulated phospho-GSK-3β and reduced phospho-ERK and phospho-JNK were involved in the protective effect of gastrodin. In conclusion, we demonstrated the neuroprotective effect of gastrodin in the N2a/APP cell line by ameliorating the impairment on synaptic and mitochondrial function, reducing tau phosphorylation, Aβ1-42 levels as well as reactive oxygen species generation. These results provide new mechanistic insights into the potential effect of gastrodin in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Zhi Tang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China; Basic Medical College, Guizhou Medical University, Guiyang, China
| | - Yaqian Peng
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China; Basic Medical College, Guizhou Medical University, Guiyang, China
| | - Yi Jiang
- Department of Pathology, Affiliated Hospital of Traditional Chinese Medicine of Guangzhou Medical University, Guangzhou, China
| | - Li Wang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China; Basic Medical College, Guizhou Medical University, Guiyang, China
| | - Min Guo
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China; Basic Medical College, Guizhou Medical University, Guiyang, China
| | - Zhuyi Chen
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China; Basic Medical College, Guizhou Medical University, Guiyang, China
| | - Chao Luo
- Basic Medical College, Guizhou Medical University, Guiyang, China
| | - Ting Zhang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China; Basic Medical College, Guizhou Medical University, Guiyang, China
| | - Yan Xiao
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China; Basic Medical College, Guizhou Medical University, Guiyang, China
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland; Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland.
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education and Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, China; Basic Medical College, Guizhou Medical University, Guiyang, China.
| |
Collapse
|
35
|
Yang C, Zhu Z, Wang G, Liu Z, Shu C, Guo J, Wan Q. Fluoxetin suppresses tau phosphorylation and modulates the interaction between tau and tubulin in the hippocampus of CUMS rats. Neurosci Lett 2024; 836:137870. [PMID: 38852764 DOI: 10.1016/j.neulet.2024.137870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/24/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Depression is considered a crucial psychiatric disease correlated with neuronal-dysfunctions induced by stress-stimuli. This study aimed to investigate effect of Fluoxetine (FL) on chronic unpredictable mild stress (CUMS) and explore the associated mechanisms. CUMS rat model was established by treating with lots of stresses. CUMS rats were administered FL, SB216763 (SB), Wortmannin (WT) alone or in combination. CUMS rats were administered 1 % sugar water to conduct sugar water consumption experiment. Acet-Tub, Tyr-Tub, tau46, p-tau-Ser199/202, p-tau-Ser396, p-tau-Ser231, expression was examined using immunohistochemical assay and western blotassay. Interaction between tau and tubulin was evaluated with immunoprecipitation assay. Double immunohistochemical assay was used to identify interaction between Nestin and Tau. The results indicated that FL treatment only increased sugar consumption of CUMS rats (P < 0.05), but also strengthened effects of SB and WT. FL significantly treatment decreased tau phosphorylation (p-tau) in hippocampal tissues of rats compared to those of rats in CUMS group (P < 0.05). FL treatment markedly decreased Acet-Tub and increased Tyr-Tub expression in hippocampal tissues of rats compared to those of rats in CUMS group (P < 0.05). The effects of FL treatment on p-tau down-regulation and tubulin modulation in hippocampal tissues were independent from PI3K and GSK-3 signaling pathways. FL treatment could also enhance proliferation and total tau of newborn neurons of CUMS rats. FL treatment strengthened interaction between tau and botulin in hippocampal tissues of CUMS rats. In conclusion, Fluoxetin suppressed phosphorylation of tau and modulated the interaction between tau and tubulin in hippocampus of adult CUMS rats.
Collapse
Affiliation(s)
- Can Yang
- Department of Psychiatry, Renmin hospital of Wuhan University, Wuhan, China
| | - Zhixian Zhu
- Department of Psychiatry, Renmin hospital of Wuhan University, Wuhan, China.
| | - Gaohua Wang
- Department of Psychiatry, Renmin hospital of Wuhan University, Wuhan, China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin hospital of Wuhan University, Wuhan, China
| | - Chang Shu
- Department of Psychiatry, Renmin hospital of Wuhan University, Wuhan, China
| | - Junhui Guo
- Department of Psychiatry, Renmin hospital of Wuhan University, Wuhan, China
| | - Qirong Wan
- Department of Psychiatry, Renmin hospital of Wuhan University, Wuhan, China
| |
Collapse
|
36
|
Ferrer I. Alzheimer's Disease Neuropathological Change in Aged Non-Primate Mammals. Int J Mol Sci 2024; 25:8118. [PMID: 39125687 PMCID: PMC11311584 DOI: 10.3390/ijms25158118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Human brain aging is characterized by the production and deposition of β-amyloid (Aβ) in the form of senile plaques and cerebral amyloid angiopathy and the intracellular accumulation of hyper-phosphorylated tau (Hp-tau) to form neurofibrillary tangles (NFTs) and dystrophic neurites of senile plaques. The process progresses for years and eventually manifests as cognitive impairment and dementia in a subgroup of aged individuals. Aβ is produced and deposited first in the neocortex in most aged mammals, including humans; it is usually not accompanied by altered behavior and cognitive impairment. Hp-tau is less frequent than Aβ pathology, and NFTs are rare in most mammals. In contrast, NFTs are familiar from middle age onward in humans; NFTs first appear in the paleocortex and selected brain stem nuclei. NFTs precede for decades or years Aβ deposition and correlate with dementia in about 5% of individuals at the age of 65 and 25% at the age of 85. Based on these comparative data, (a) Aβ deposition is the most common Alzheimer's disease neuropathological change (ADNC) in the brain of aged mammals; (b) Hp-tau is less common, and NFTs are rare in most aged mammals; however, NFTs are the principal cytoskeletal pathology in aged humans; (c) NFT in aged humans starts in selected nuclei of the brain stem and paleocortical brain regions progressing to the most parts of the neocortex and other regions of the telencephalon; (d) human brain aging is unique among mammalian species due to the early appearance and dramatic progression of NFTs from middle age onward, matching with cognitive impairment and dementia in advanced cases; (e) neither mammalian nor human brain aging supports the concept of the amyloid cascade hypothesis.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, carrer Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain;
- Reial Acadèmia de Medicina de Catalunya, carrer del Carme 47, 08001 Barcelona, Spain
| |
Collapse
|
37
|
Sun F, Huang X, Wang H, Lin B, Li H, Wang X, Liu Q. Exploring Dimethylsulfoniopropionate as a potential treatment for Alzheimer's disease: A study using the 3 × Tg-AD mouse model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155788. [PMID: 38838634 DOI: 10.1016/j.phymed.2024.155788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/14/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Alzheimer's disease (AD), the most common neurodegenerative disorder, affects a broad spectrum of aging populations. AD is characterized by pathological amyloid-β (Aβ) plaques and neurofibrillary tangles, leading to neural degeneration and cognitive decline. The lack of effective treatments for AD highlights the urgent need for novel therapeutic agents, particularly in the early stages. Dimethylsulfoniopropionate (DMSP) is a natural marine compound with antioxidant and neuroprotective properties. However, studies on the efficacy of DMSP in the treatment of AD and its associated mechanisms are limited. PURPOSE This study aimed to explore the therapeutic effects and mechanisms of action of DMSP as an AD treatment using a preclinical 3 × Tg-AD mouse model. METHODS The research involved administering DMSP (7 μg/mL and 11 μg/mL in drinking water) to four-month-old 3 × Tg-AD mice consecutively for three months. The Y-maze test, novel object recognition test, and Morris water maze test were used to assess memory and learning ability. The relative expression levels and distribution of proteins relevant to Aβ and tau pathology, synapses, and glial cells were analyzed using western blotting and immunofluorescence assays. Additionally, proteomic and bioinformatics approaches were used to explore the potential targets of DMSP treatment. RESULTS DMSP-treated AD mice showed significantly enhanced cognitive function, suggesting that DMSP mitigates memory and learning impairments in AD. Moreover, DMSP diminished the abnormal accumulation of Aβ and phosphorylated tau in both the cortex and hippocampus, which are crucial hallmarks of AD pathology. In addition to its neuroprotective properties, DMSP restored synaptic density and the expression of synaptic and neuronal proteins, which are essential for proper brain function. DMSP displayed anti-inflammatory properties, as evidenced by its ability to suppress inflammatory astrocytes and maintain microglial homeostasis. Notably, DMSP facilitated the maturation of oligodendrocytes (OLs) from oligodendrocyte progenitor cells (OPCs), a critical process in the development of the brain myelination architecture. Proteomic analysis revealed that DMSP positively influenced biological processes crucial for oligodendrocyte development, myelination, and axonal ensheathment, which are often compromised in patients with AD. Protein validation and brain tissue staining supported the role of DMSP in preserving myelin enrichment and sheath integrity. These therapeutic effects were largely attributed to the enhanced expression of myelin-associated glycoprotein (Mag) and tetraspanin Cd9. CONCLUSION Overall, our findings highlight DMSP as a promising novel therapeutic candidate for AD, offering multifaceted benefits in cognitive and memory enhancement, reduction of Aβ and tau pathology, neuronal synapse protection, anti-inflammatory effects, and myelin sheath restoration as an innovative target compared to other studies. In addition to being a potentially effective treatment for AD, DMSP may also have the potential to address other neurodegenerative diseases that are closely associated with myelin impairment.
Collapse
Affiliation(s)
- Fanfan Sun
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences & Oceanography, Shenzhen University, Shenzhen 518055, China; Key Laboratory of Optoelectronic Devices and System of Ministry of Education and Guangdong Province, College Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Xuelian Huang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences & Oceanography, Shenzhen University, Shenzhen 518055, China
| | - Hongshuang Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Baoyi Lin
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences & Oceanography, Shenzhen University, Shenzhen 518055, China
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China; Beijing National Laboratory for Molecular Sciences, Beijing 100190, China.
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences & Oceanography, Shenzhen University, Shenzhen 518055, China; Key Laboratory of Optoelectronic Devices and System of Ministry of Education and Guangdong Province, College Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China; Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518033, China.
| |
Collapse
|
38
|
Ahamad S, Junaid IT, Gupta D. Computational Design of Novel Tau-Tubulin Kinase 1 Inhibitors for Neurodegenerative Diseases. Pharmaceuticals (Basel) 2024; 17:952. [PMID: 39065802 PMCID: PMC11280166 DOI: 10.3390/ph17070952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
The tau-tubulin kinase 1 (TTBK1) protein is a casein kinase 1 superfamily member located at chromosome 6p21.1. It is expressed explicitly in the brain, particularly in the cytoplasm of cortical and hippocampal neurons. TTBK1 has been implicated in the phosphorylation and aggregation of tau in Alzheimer's disease (AD). Considering its significance in AD, TTBK1 has emerged as a promising target for AD treatment. In the present study, we identified novel TTBK1 inhibitors using various computational techniques. We performed a virtual screening-based docking study followed by E-pharmacophore modeling, cavity-based pharmacophore, and ligand design techniques and found ZINC000095101333, LD7, LD55, and LD75 to be potential novel TTBK1 lead inhibitors. The docking results were complemented by Molecular Mechanics/Generalized Born Surface Area (MMGBSA) calculations. The molecular dynamics (MD) simulation studies at a 500 ns scale were carried out to monitor the behavior of the protein toward the identified ligands. Pharmacological and ADME/T studies were carried out to check the drug-likeness of the compounds. In summary, we identified a new series of compounds that could effectively bind the TTBK1 receptor. The newly designed compounds are promising candidates for developing therapeutics targeting TTBK1 for AD.
Collapse
Affiliation(s)
- Shahzaib Ahamad
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Iqbal Taliy Junaid
- Malaria Biology, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India;
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| |
Collapse
|
39
|
Fang F, Xu T, Hagar HTC, Hovde S, Kuo MH, Sun L. A pilot study for deciphering post-translational modifications and proteoforms of tau protein by capillary electrophoresis-mass spectrometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.04.602093. [PMID: 39026802 PMCID: PMC11257423 DOI: 10.1101/2024.07.04.602093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Abnormal accumulation of tau proteins is one pathological hallmark of Alzheimer□s disease (AD). Many tau protein post-translational modifications (PTMs) are associated with the development of AD, such as phosphorylation, acetylation, and methylation. Therefore, a complete picture of PTM landscape of tau is critical for understanding the molecular mechanisms of AD progression. Here, we offered a pilot study of combining two complementary analytical techniques, capillary zone electrophoresis (CZE)-tandem mass spectrometry (MS/MS) and reversed-phase liquid chromatography (RPLC)-MS/MS, for bottom-up proteomics of recombinant human tau-0N3R. We identified 53 phosphorylation sites of tau-0N3R in total, which is about 30% higher than that from RPLC-MS/MS alone. CZE-MS/MS provided more PTM sites (i.e., phosphorylation) and modified peptides of tau-0N3R than RPLC-MS/MS, and its predicted electrophoretic mobility helped improve the confidence of the identified modified peptides. We developed a highly efficient capillary isoelectric focusing (cIEF)-MS technique to offer a bird's-eye view of tau-0N3R proteoforms, with 11 putative tau-0N3R proteoforms carrying up to nine phosphorylation sites and lower pI values from more phosphorylated proteoforms detected. Interestingly, under a native-like cIEF-MS condition, we observed three putative tau-0N3R dimers carrying phosphate groups. The findings demonstrate that CE-MS is a valuable analytical technique for the characterization of tau PTMs, proteoforms, and even oligomerization.
Collapse
|
40
|
Cheng Y, Chen S, Zhang Y, Guo Y, Wu K, Huang Y, Aerqin Q, Kuo K, Li H, Chen S, Liu W, Dong Q, Yu J. Novel diagnostic and prognostic approach for rapidly progressive dementias: Indicators based on amyloid/tau/neurodegeneration (ATN) framework. CNS Neurosci Ther 2024; 30:e14857. [PMID: 39014454 PMCID: PMC11251870 DOI: 10.1111/cns.14857] [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: 02/12/2024] [Revised: 06/17/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
AIMS Apply established cerebrospinal fluid (CSF) and serum biomarkers and novel combined indicators based on the amyloid/tau/neurodegeneration (ATN) framework to improve diagnostic and prognostic power in patients with rapidly progressive dementias (RPDs). METHODS CSF and serum biomarkers of Alzheimer's disease (AD) common neuropathology including Aβ42, Aβ40, p-Tau, and t-Tau were measured in cognitively normal (CN) controls (n = 33) and three RPD groups with rapidly progressive AD (rpAD, n = 23), autoimmune encephalitis (AE, n = 25), and Creutzfeldt-Jakob disease (CJD, n = 28). Logistic regression and multiple linear regression were used for producing combined indicators and prognostic assessment, respectively, including A&T, A&N, T&N, A&T&N, etc. RESULTS: Combined diagnostic indicator with A&T&N had the potential for differentiating AE from other types of RPDs, identifying 62.51% and 75% of AE subjects based on CSF and serum samples, respectively, compared to 39.13% and 37.5% when using autoantibodies. CSF t-Tau was associated with survival in the CJD group (adjusted R-Square = 0.16, p = 0.02), and its prognosis value improved when using combined predictors based on the ATN framework (adjusted R-Square = 0.273, p = 0.014). CONCLUSION Combined indicators based on the ATN framework provide a novel perspective for establishing biomarkers for early recognition of RPDs due to treatment-responsive causes.
Collapse
Affiliation(s)
- Yuan Cheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| | - Shu‐Fen Chen
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| | - Ya‐Ru Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| | - Yu Guo
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| | - Kai‐Min Wu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| | - Yu‐Yuan Huang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| | - Qiaolifan Aerqin
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| | - Kevin Kuo
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| | - Hong‐Qi Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| | - Shi‐Dong Chen
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| | - Wei‐Shi Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| | - Qiang Dong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| | - Jin‐Tai Yu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Center for Neurological DisordersShanghaiChina
| |
Collapse
|
41
|
Ye J, Wan H, Chen S, Liu GP. Targeting tau in Alzheimer's disease: from mechanisms to clinical therapy. Neural Regen Res 2024; 19:1489-1498. [PMID: 38051891 PMCID: PMC10883484 DOI: 10.4103/1673-5374.385847] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/16/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT Alzheimer's disease is the most prevalent neurodegenerative disease affecting older adults. Primary features of Alzheimer's disease include extracellular aggregation of amyloid-β plaques and the accumulation of neurofibrillary tangles, formed by tau protein, in the cells. While there are amyloid-β-targeting therapies for the treatment of Alzheimer's disease, these therapies are costly and exhibit potential negative side effects. Mounting evidence suggests significant involvement of tau protein in Alzheimer's disease-related neurodegeneration. As an important microtubule-associated protein, tau plays an important role in maintaining the stability of neuronal microtubules and promoting axonal growth. In fact, clinical studies have shown that abnormal phosphorylation of tau protein occurs before accumulation of amyloid-β in the brain. Various therapeutic strategies targeting tau protein have begun to emerge, and are considered possible methods to prevent and treat Alzheimer's disease. Specifically, abnormalities in post-translational modifications of the tau protein, including aberrant phosphorylation, ubiquitination, small ubiquitin-like modifier (SUMO)ylation, acetylation, and truncation, contribute to its microtubule dissociation, misfolding, and subcellular missorting. This causes mitochondrial damage, synaptic impairments, gliosis, and neuroinflammation, eventually leading to neurodegeneration and cognitive deficits. This review summarizes the recent findings on the underlying mechanisms of tau protein in the onset and progression of Alzheimer's disease and discusses tau-targeted treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Jinwang Ye
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Huali Wan
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Sihua Chen
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Gong-Ping Liu
- Co-innovation Center of Neurodegeneration, Nantong University, Nantong, Jiangsu Province, China
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
42
|
Zhou Y, Zhou F, Xu S, Shi D, Ding D, Wang S, Poongavanam V, Tang K, Liu X, Zhan P. Hydrophobic tagging of small molecules: an overview of the literature and future outlook. Expert Opin Drug Discov 2024; 19:799-813. [PMID: 38825802 DOI: 10.1080/17460441.2024.2360416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/23/2024] [Indexed: 06/04/2024]
Abstract
INTRODUCTION Hydrophobic tagging (HyT) technology presents a distinct therapeutic strategy diverging from conventional small molecule drugs, providing an innovative approach to drug design. This review aims to provide an overview of the HyT literature and future outlook to offer guidance for drug design. AREAS COVERED In this review, the authors introduce the composition, mechanisms and advantages of HyT technology, as well as summarize the detailed applications of HyT technology in anti-cancer, neurodegenerative diseases (NDs), autoimmune disorders, cardiovascular diseases (CVDs), and other fields. Furthermore, this review discusses key aspects of the future development of HyT molecules. EXPERT OPINION HyT emerges as a highly promising targeted protein degradation (TPD) strategy, following the successful development of proteolysis targeting chimeras (PROTAC) and molecular glue. Based on exploring new avenues, modification of the HyT molecule itself potentially enhances the technology. Improved synthetic pathways and emphasis on pharmacokinetic (PK) properties will facilitate the development of HyT. Furthermore, elucidating the biochemical basis by which the compound's hydrophobic moiety recruits the protein homeostasis network will enable the development of more precise assays that can guide the optimization of the linker and hydrophobic moiety.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Fan Zhou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Shujing Xu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Dazhou Shi
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Dang Ding
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Shuo Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | | | - Kai Tang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
43
|
Mazahir F, Alam MI, Yadav AK. Development of nanomedicines for the treatment of Alzheimer's disease: Raison d'être, strategies, challenges and regulatory aspects. Ageing Res Rev 2024; 98:102318. [PMID: 38705362 DOI: 10.1016/j.arr.2024.102318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/04/2024] [Accepted: 04/27/2024] [Indexed: 05/07/2024]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder characterized by progressive loss of memory. Presently, AD is challenging to treat with current drug therapy as their delivery to the brain is restricted by the presence of the blood-brain barrier. Nanomedicines, due to their size, high surface volume ratio, and ease of tailoring drug release characteristics, showed their potential to treat AD. The nanotechnology-based formulations for brain targeting are expected to enter the market in the near future. So, regulatory frameworks are required to ensure the quality, safety, and effectiveness of the nanomedicines to treat AD. In this review, we discuss different strategies, in-vitro blood-brain permeation models, in-vivo permeation assessment, and regulatory aspects for the development of nanomedicine to treat AD.
Collapse
Affiliation(s)
- Farhan Mazahir
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raebareli, India
| | - Md Imtiyaz Alam
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raebareli, India
| | - Awesh Kumar Yadav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raebareli, India.
| |
Collapse
|
44
|
Zhao J, Wei M, Guo M, Wang M, Niu H, Xu T, Zhou Y. GSK3: A potential target and pending issues for treatment of Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14818. [PMID: 38946682 PMCID: PMC11215492 DOI: 10.1111/cns.14818] [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: 04/22/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 07/02/2024] Open
Abstract
Glycogen synthase kinase-3 (GSK3), consisting of GSK3α and GSK3β subtypes, is a complex protein kinase that regulates numerous substrates. Research has observed increased GSK3 expression in the brains of Alzheimer's disease (AD) patients and models. AD is a neurodegenerative disorder with diverse pathogenesis and notable cognitive impairments, characterized by Aβ aggregation and excessive tau phosphorylation. This article provides an overview of GSK3's structure and regulation, extensively analyzing its relationship with AD factors. GSK3 overactivation disrupts neural growth, development, and function. It directly promotes tau phosphorylation, regulates amyloid precursor protein (APP) cleavage, leading to Aβ formation, and directly or indirectly triggers neuroinflammation and oxidative damage. We also summarize preclinical research highlighting the inhibition of GSK3 activity as a primary therapeutic approach for AD. Finally, pending issues like the lack of highly specific and affinity-driven GSK3 inhibitors, are raised and expected to be addressed in future research. In conclusion, GSK3 represents a target in AD treatment, filled with hope, challenges, opportunities, and obstacles.
Collapse
Affiliation(s)
- Jiahui Zhao
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Mengying Wei
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Future Health Laboratory, Innovation Center of Yangtze River DeltaZhejiang UniversityJiaxingChina
| | - Minsong Guo
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Cangnan County Qiushi Innovation Research Institute of Traditional Chinese MedicineWenzhouChina
| | - Mengyao Wang
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Hongxia Niu
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Laboratory of Blood‐stasis‐toxin Syndrome of Zhejiang ProvinceHangzhouChina
| | - Tengfei Xu
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouChina
- Cangnan County Qiushi Innovation Research Institute of Traditional Chinese MedicineWenzhouChina
| | - Yuan Zhou
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhouChina
- Key Laboratory of Blood‐stasis‐toxin Syndrome of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
45
|
Wang K, Chen X. Protective effect of flavonoids on oxidative stress injury in Alzheimer's disease. Nat Prod Res 2024:1-28. [PMID: 38910339 DOI: 10.1080/14786419.2024.2345760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/14/2024] [Indexed: 06/25/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, which is mainly caused by the damage of the structure and function of the central nervous system. At present, there are many adverse reactions in market-available drugs, which can't significantly inhibit the occurrence of AD. Therefore, the current focus of research is to find safe and effective therapeutic drugs to improve the clinical treatment of AD. Oxidative stress bridges different mechanism hypotheses of AD and plays a key role in AD. Numerous studies have shown that natural flavonoids have good antioxidant effects. They can directly or indirectly resist -oxidative stress, inhibit Aβ aggregation and Tau protein hyperphosphorylation by activating Nrf2 and other oxidation-antioxidation-related signals, regulating synaptic function-related pathways, promoting mitochondrial autophagy, etc., and play a neuroprotective role in AD. In this review, we summarised the mechanism of flavonoids inhibiting oxidative stress injury in AD in recent years. Moreover, because of the shortcomings of poor biofilm permeability and low bioavailability of flavonoids, the advantages and recent research progress of nano-drug delivery systems such as liposomes and solid lipid nanoparticles were highlighted. We hope this review provides a useful way to explore safe and effective AD treatments.
Collapse
Affiliation(s)
- Kaixuan Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinmei Chen
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
46
|
Ricci A, Carradori S, Cataldi A, Zara S. Eg5 and Diseases: From the Well-Known Role in Cancer to the Less-Known Activity in Noncancerous Pathological Conditions. Biochem Res Int 2024; 2024:3649912. [PMID: 38939361 PMCID: PMC11211015 DOI: 10.1155/2024/3649912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/06/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024] Open
Abstract
Eg5 is a protein encoded by KIF11 gene and is primarily involved in correct mitotic cell division. It is also involved in nonmitotic processes such as polypeptide synthesis, protein transport, and angiogenesis. The scientific literature sheds light on the ubiquitous functions of KIF11 and its involvement in the onset and progression of different pathologies. This review focuses attention on two main points: (1) the correlation between Eg5 and cancer and (2) the involvement of Eg5 in noncancerous conditions. Regarding the first point, several tumors revealed an overexpression of this kinesin, thus pushing to look for new Eg5 inhibitors for clinical practice. In addition, the evaluation of Eg5 expression represents a crucial step, as its overexpression could predict a poor prognosis for cancer patients. Referring to the second point, in specific pathological conditions, the reduced activity of Eg5 can be one of the causes of pathological onset. This is the case of Alzheimer's disease (AD), in which Aβ and Tau work as Eg5 inhibitors, or in acquired immune deficiency syndrome (AIDS), in which Tat-mediated Eg5 determines the loss of CD4+ T-lymphocytes. Reduced Eg5 activity, due to mutations of KIF11 gene, is also responsible for pathological conditions such as microcephaly with or without chorioretinopathy, lymphedema, or intellectual disability (MCLRI) and familial exudative vitreous retinopathy (FEVR). In conclusion, this review highlights the double impact that overexpression or loss of function of Eg5 could have in the onset and progression of different pathological situations. This emphasizes, on one hand, a possible role of Eg5 as a potential biomarker and new target in cancer and, on the other hand, the promotion of Eg5 expression/activity as a new therapeutic strategy in different noncancerous diseases.
Collapse
Affiliation(s)
- Alessia Ricci
- Department of Pharmacy, University “G. d'Annunzio” Chieti-Pescara, Chieti, 66100, Italy
| | - Simone Carradori
- Department of Pharmacy, University “G. d'Annunzio” Chieti-Pescara, Chieti, 66100, Italy
| | - Amelia Cataldi
- Department of Pharmacy, University “G. d'Annunzio” Chieti-Pescara, Chieti, 66100, Italy
| | - Susi Zara
- Department of Pharmacy, University “G. d'Annunzio” Chieti-Pescara, Chieti, 66100, Italy
| |
Collapse
|
47
|
Riemma MA, Mele E, Donniacuo M, Telesca M, Bellocchio G, Castaldo G, Rossi F, De Angelis A, Cappetta D, Urbanek K, Berrino L. Glucagon-like peptide-1 receptor agonists and sodium-glucose cotransporter 2 inhibitors, anti-diabetic drugs in heart failure and cognitive impairment: potential mechanisms of the protective effects. Front Pharmacol 2024; 15:1422740. [PMID: 38948473 PMCID: PMC11212466 DOI: 10.3389/fphar.2024.1422740] [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: 04/24/2024] [Accepted: 05/21/2024] [Indexed: 07/02/2024] Open
Abstract
Heart failure and cognitive impairment emerge as public health problems that need to be addressed due to the aging global population. The conditions that often coexist are strongly related to advancing age and multimorbidity. Epidemiological evidence indicates that cardiovascular disease and neurodegenerative processes shares similar aspects, in term of prevalence, age distribution, and mortality. Type 2 diabetes increasingly represents a risk factor associated not only to cardiometabolic pathologies but also to neurological conditions. The pathophysiological features of type 2 diabetes and its metabolic complications (hyperglycemia, hyperinsulinemia, and insulin resistance) play a crucial role in the development and progression of both heart failure and cognitive dysfunction. This connection has opened to a potential new strategy, in which new classes of anti-diabetic medications, such as glucagon-like peptide-1 receptor (GLP-1R) agonists and sodium-glucose cotransporter 2 (SGLT2) inhibitors, are able to reduce the overall risk of cardiovascular events and neuronal damage, showing additional protective effects beyond glycemic control. The pleiotropic effects of GLP-1R agonists and SGLT2 inhibitors have been extensively investigated. They exert direct and indirect cardioprotective and neuroprotective actions, by reducing inflammation, oxidative stress, ions overload, and restoring insulin signaling. Nonetheless, the specificity of pathways and their contribution has not been fully elucidated, and this underlines the urgency for more comprehensive research.
Collapse
Affiliation(s)
- Maria Antonietta Riemma
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Elena Mele
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Maria Donniacuo
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Marialucia Telesca
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Gabriella Bellocchio
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giuseppe Castaldo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
- CEINGE-Advanced Biotechnologies, Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Donato Cappetta
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Konrad Urbanek
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, Naples, Italy
- CEINGE-Advanced Biotechnologies, Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
48
|
Mohl GA, Dixon G, Marzette E, McKetney J, Samelson AJ, Serras CP, Jin J, Li A, Boggess SC, Swaney DL, Kampmann M. The disease-causing tau V337M mutation induces tau hypophosphorylation and perturbs axon morphology pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597496. [PMID: 38895329 PMCID: PMC11185762 DOI: 10.1101/2024.06.04.597496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Tau aggregation is a hallmark of several neurodegenerative diseases, including Alzheimer's disease and frontotemporal dementia. There are disease-causing variants of the tau-encoding gene, MAPT, and the presence of tau aggregates is highly correlated with disease progression. However, the molecular mechanisms linking pathological tau to neuronal dysfunction are not well understood due to our incomplete understanding of the normal functions of tau in development and aging and how these processes change in the context of causal disease variants of tau. To address these questions in an unbiased manner, we conducted multi-omic characterization of iPSC-derived neurons harboring the MAPT V337M mutation. RNA-seq and phosphoproteomics revealed that both V337M tau and tau knockdown consistently perturbed levels of transcripts and phosphorylation of proteins related to axonogenesis or axon morphology. Surprisingly, we found that neurons with V337M tau had much lower tau phosphorylation than neurons with WT tau. We conducted functional genomics screens to uncover regulators of tau phosphorylation in neurons and found that factors involved in axonogenesis modified tau phosphorylation in both MAPT WT and MAPT V337M neurons. Intriguingly, the p38 MAPK pathway specifically modified tau phosphorylation in MAPT V337M neurons. We propose that V337M tau might perturb axon morphology pathways and tau hypophosphorylation via a "loss of function" mechanism, which could contribute to previously reported cognitive changes in preclinical MAPT gene carriers.
Collapse
Affiliation(s)
- Gregory A Mohl
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Gary Dixon
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Emily Marzette
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Justin McKetney
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, CA, USA
- Quantitative Bioscience Institute, University of California San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Avi J Samelson
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Carlota Pereda Serras
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA USA
| | - Julianne Jin
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Andrew Li
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Steven C Boggess
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
| | - Danielle L Swaney
- Gladstone Data Science and Biotechnology Institute, The J. David Gladstone Institutes, San Francisco, CA, USA
- Quantitative Bioscience Institute, University of California San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
49
|
Sindi G, Ismael S, Uddin R, Slepchenko KG, Colvin RA, Lee D. Endogenous tau released from human ReNCell VM cultures by neuronal activity is phosphorylated at multiple sites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.597022. [PMID: 38854111 PMCID: PMC11160771 DOI: 10.1101/2024.06.02.597022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Tau is an intracellular protein but also known to be released into the extracellular fluid. Tau release mechanisms have drawn intense attention as these are known to play a key role in Alzheimer's disease (AD) pathology. However, tau can also be released under physiological conditions although its physiological function and release mechanisms have been poorly characterized, especially in human neuronal cells. We investigated endogenous tau release in ReNCell VM, a human neuroprogenitor cell line, under physiological conditions and found that tau is spontaneously released from cells. To study activity-dependent release of endogenous tau, human ReNCell VM culture was stimulated by 100μM AMPA or 50mM KCl for one-hour, tau was actively released to the culture medium. The released tau was highly phosphorylated at nine phosphorylation sites (pSites) detected by phospho-specific tau antibodies including AT270 (T175/T181), AT8 (S202/T205), AT100 (T212/S214), AT180 (T231), and PHF-1 (S396/S404), showing that these pSites are important for activity-dependent tau release from human ReNCell VM. Intracellular tau showed various phosphorylation status across these sites, with AT270 and PHF-1 highly phosphorylated while AT8 and AT180 were minimally phosphorylated, suggesting that AT8 and AT180 pSites exhibit a propensity for secretion rather than being retained intracellularly. This activity-dependent tau release was significantly decreased by inhibition of GSK-3β, demonstrating that GSK3β-dependent phosphorylation of tau plays an important role in its release by neuronal activity. In this study, we showed that ReNCell VM serves as a valuable model for studying endogenous physiological tau release. Further, ReNCell model can be also used to study pathological release of human tau that will contribute to our understanding of the progression of AD and related dementias.
Collapse
Affiliation(s)
| | - Sazan Ismael
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Reaz Uddin
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Kira G. Slepchenko
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Robert A. Colvin
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Daewoo Lee
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
50
|
Arico DS, Burachik NB, Wengier DL, Mazzella MA. Arabidopsis hypocotyl growth in darkness requires the phosphorylation of a microtubule-associated protein. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2024; 118:1815-1831. [PMID: 38494883 DOI: 10.1111/tpj.16711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 02/23/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024]
Abstract
Rapid hypocotyl elongation allows buried seedlings to emerge, where light triggers de-etiolation and inhibits hypocotyl growth mainly by photoreceptors. Phosphorylation/dephosphorylation events regulate many aspects of plant development. Only recently we have begun to uncover the earliest phospho-signaling responders to light. Here, we reported a large-scale phosphoproteomic analysis and identified 20 proteins that changed their phosphorylation pattern following a 20 min light pulse compared to darkness. Microtubule-associated proteins were highly overrepresented in this group. Among them, we studied CIP7 (COP1-INTERACTING-PROTEIN 7), which presented microtubule (MT) localization in contrast to the previous description. An isoform of CIP7 phosphorylated at Serine915 was detected in etiolated seedlings but was undetectable after a light pulse in the presence of photoreceptors, while CIP7 transcript expression decays with long light exposure. The short hypocotyl phenotype and rearrangement of MTs in etiolated cip7 mutants are complemented by CIP7-YFP and the phospho-mimetic CIP7S915D-YFP, but not the phospho-null CIP7S915A-YFP suggesting that the phosphorylated S915CIP7 isoform promotes hypocotyl elongation through MT reorganization in darkness. Our evidence on Serine915 of CIP7 unveils phospho-regulation of MT-based processes during skotomorphogenic hypocotyl growth.
Collapse
Affiliation(s)
- Denise Soledad Arico
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular-Héctor Torres, Vuelta de obligado, 2490, Caba, Argentina
| | - Natalia B Burachik
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular-Héctor Torres, Vuelta de obligado, 2490, Caba, Argentina
| | - Diego Leonardo Wengier
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular-Héctor Torres, Vuelta de obligado, 2490, Caba, Argentina
| | - María Agustina Mazzella
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular-Héctor Torres, Vuelta de obligado, 2490, Caba, Argentina
| |
Collapse
|