1
|
Mao M, Zhou Y, Zhang H, Deng P, Yang J, Zhong J, Li N, Liu Q, Li X, Wu X, Cheng Y. Synthesis and identification of azocoumarin derivatives toward imaging of α-synuclein aggregates in the brain. Eur J Med Chem 2025; 290:117587. [PMID: 40168910 DOI: 10.1016/j.ejmech.2025.117587] [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: 01/25/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/03/2025]
Abstract
To identify α-synuclein aggregation in synucleinopathies is still challenging, due to the lack of specific probes for α-synuclein aggregates with efficient brain uptake. In this work, compact molecules based on coumarin scaffold were synthesized and evaluated for detection and bioimaging of α-synuclein aggregates in the brain. Among the developed compounds, azocoumarin 5 containing push-pull electronic architecture featured selective fluorescence enhancement towards α-synuclein aggregates in comparison to other β-sheet protein species (β-amyloid, tau). In addition, azocoumarin [18F]Cou-NNF was succesfully developed, and demonstrated its potential as radiotracer for imaging brain α-synuclein aggregates, owing to its favorable affinity for α-synuclein aggregates accompanied with efficient brain uptake and little defluorination in vivo. Overall, compact azocoumarin provides an effective lead structure for developing α-synuclein probes, and N=N bond shows promise in enhancing selective affinity for α-synuclein aggregates.
Collapse
Affiliation(s)
- Meiting Mao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yu Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Huihui Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Pengxin Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jie Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jing Zhong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Na Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Qiangqiang Liu
- Chengdu New Radiomedicine Technology Co., Ltd., Chengdu, 610200, China
| | - Xianghui Li
- Chengdu New Radiomedicine Technology Co., Ltd., Chengdu, 610200, China
| | - Xiaoai Wu
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yan Cheng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Sandhof CA, Murray HFB, Silva MC, Haggarty SJ. Targeted protein degradation with bifunctional molecules as a novel therapeutic modality for Alzheimer's disease & beyond. Neurotherapeutics 2025; 22:e00499. [PMID: 39638711 PMCID: PMC12047403 DOI: 10.1016/j.neurot.2024.e00499] [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: 09/16/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
Alzheimer's disease (AD) is associated with memory and cognitive impairment caused by progressive degeneration of neurons. The events leading to neuronal death are associated with the accumulation of aggregating proteins in neurons and glia of the affected brain regions, in particular extracellular deposition of amyloid plaques and intracellular formation of tau neurofibrillary tangles. Moreover, the accumulation of pathological tau proteoforms in the brain concurring with disease progression is a key feature of multiple neurodegenerative diseases, called tauopathies, like frontotemporal dementia (FTD) where autosomal dominant mutations in the tau encoding MAPT gene provide clear evidence of a causal role for tau dysfunction. Observations from disease models, post-mortem histology, and clinical evidence have demonstrated that pathological tau undergoes abnormal post-translational modifications, misfolding, oligomerization, changes in solubility, mislocalization, and intercellular spreading. Despite extensive research, there are few disease-modifying or preventative therapeutics for AD and none for other tauopathies. Challenges faced in tauopathy drug development include an insufficient understanding of pathogenic mechanisms of tau proteoforms, limited specificity of agents tested, and inadequate levels of brain exposure, altogether underscoring the need for innovative therapeutic modalities. In recent years, the development of experimental therapeutic modalities, such as targeted protein degradation (TPD) strategies, has shown significant and promising potential to promote the degradation of disease-causing proteins, thereby reducing accumulation and aggregation. Here, we review all modalities of TPD that have been developed to target tau in the context of AD and FTD, as well as other approaches that with innovation could be adapted for tau-specific TPD.
Collapse
Affiliation(s)
- C Alexander Sandhof
- Department of Neurology, Precision Therapeutics Unit, Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Heide F B Murray
- Department of Neurology, Precision Therapeutics Unit, Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - M Catarina Silva
- Department of Neurology, Precision Therapeutics Unit, Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Stephen J Haggarty
- Department of Neurology, Precision Therapeutics Unit, Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
3
|
Uliassi E, Bolognesi ML, Milelli A. Targeting Tau Protein with Proximity Inducing Modulators: A New Frontier to Combat Tauopathies. ACS Pharmacol Transl Sci 2025; 8:654-672. [PMID: 40109749 PMCID: PMC11915046 DOI: 10.1021/acsptsci.4c00733] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 03/22/2025]
Abstract
Dysregulation of correct protein tau homeostasis represents the seed for the development of several devastating central nervous system disorders, known as tauopathies, that affect millions of people worldwide. Despite massive public and private support to research funding, these diseases still represent unmet medical needs. In fact, the tau-targeting tools developed to date have failed to translate into the clinic. Recently, taking advantage of the modes that nature uses to mediate the flow of information in cells, researchers have developed a new class of molecules, called proximity-inducing modulators, which exploit spatial proximity to modulate protein function(s) and redirect cellular processes. In this perspective, after a brief discussion about tau protein and the classic tau-targeting approaches, we will discuss the different classes of proximity-inducing modulators developed so far and highlight the applications to modulate tau protein's function and tau-induced toxicity.
Collapse
Affiliation(s)
- Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Andrea Milelli
- Department for Life Quality Studies, Alma Mater Studiorum - University of Bologna, Corso d'Augusto 237, Rimini 47921, Italy
| |
Collapse
|
4
|
Wang L, Sooram B, Kumar R, Schedin‐Weiss S, Tjernberg LO, Winblad B. Tau degradation in Alzheimer's disease: Mechanisms and therapeutic opportunities. Alzheimers Dement 2025; 21:e70048. [PMID: 40109019 PMCID: PMC11923393 DOI: 10.1002/alz.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 03/22/2025]
Abstract
In Alzheimer's disease (AD), tau undergoes abnormal post-translational modifications and aggregations. Impaired intracellular degradation pathways further exacerbate the accumulation of pathological tau. A new strategy - targeted protein degradation - recently emerged as a modality in drug discovery where bifunctional molecules bring the target protein close to the degradation machinery to promote clearance. Since 2016, this strategy has been applied to tau pathologies and attracted broad interest in academia and the pharmaceutical industry. However, a systematic review of recent studies on tau degradation mechanisms is lacking. Here we review tau degradation mechanisms (the ubiquitin-proteasome system and the autophagy-lysosome pathway), their dysfunction in AD, and tau-targeted degraders, such as proteolysis-targeting chimeras and autophagy-targeting chimeras. We emphasize the need for a continuous exploration of tau degradation mechanisms and provide a future perspective for developing tau-targeted degraders, encouraging researchers to work on new treatment options for AD patients. HIGHLIGHTS: Post-translational modifications, aggregation, and mutations affect tau degradation. A vicious circle exists between impaired degradation pathways and tau pathologies. Ubiquitin plays an important role in complex degradation pathways. Tau-targeted degraders provide promising strategies for novel AD treatment.
Collapse
Affiliation(s)
- Lisha Wang
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Banesh Sooram
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Rajnish Kumar
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
- Department of Pharmaceutical Engineering & TechnologyIndian Institute of Technology (BHU)VaranasiIndia
| | - Sophia Schedin‐Weiss
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Lars O. Tjernberg
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
| | - Bengt Winblad
- Division of NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetSolnaSweden
- Theme Inflammation and AgingKarolinska University HospitalHuddingeSweden
| |
Collapse
|
5
|
Yao D, Li T, Yu L, Hu M, He Y, Zhang R, Wu J, Li S, Kuang W, Yang X, Liu G, Xie Y. Selective degradation of hyperphosphorylated tau by proteolysis-targeting chimeras ameliorates cognitive function in Alzheimer's disease model mice. Front Pharmacol 2024; 15:1351792. [PMID: 38919259 PMCID: PMC11196765 DOI: 10.3389/fphar.2024.1351792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most common chronic neurodegenerative diseases. Hyperphosphorylated tau plays an indispensable role in neuronal dysfunction and synaptic damage in AD. Proteolysis-targeting chimeras (PROTACs) are a novel type of chimeric molecule that can degrade target proteins by inducing their polyubiquitination. This approach has shown promise for reducing tau protein levels, which is a potential therapeutic target for AD. Compared with traditional drug therapies, the use of PROTACs to reduce tau levels may offer a more specific and efficient strategy for treating AD, with fewer side effects. In the present study, we designed and synthesized a series of small-molecule PROTACs to knock down tau protein. Of these, compound C8 was able to lower both total and phosphorylated tau levels in HEK293 cells with stable expression of wild-type full-length human tau (termed HEK293-htau) and htau-overexpressed mice. Western blot findings indicated that C8 degraded tau protein through the ubiquitin-proteasome system in a time-dependent manner. In htau-overexpressed mice, the results of both the novel object recognition and Morris water maze tests revealed that C8 markedly improved cognitive function. Together, our findings suggest that the use of the small-molecule PROTAC C8 to degrade phosphorylated tau may be a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Dongping Yao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Ting Li
- Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, The Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Mingxing Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Ye He
- Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, The Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruiming Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Junjie Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Shuoyuan Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Weihong Kuang
- Department of Psychiatry and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xifei Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology (2020–2024), Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Gongping Liu
- Key Laboratory of Ministry of Education of China and Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, The Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yongmei Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| |
Collapse
|
6
|
Qiu J, Feng X, Chen H, Liu W, Liu W, Wu L, Gao X, Liu Y, Huang Y, Gong H, Qi Y, Xu Z, Zhao Q. Discovery of novel harmine derivatives as GSK-3β/DYRK1A dual inhibitors for Alzheimer's disease treatment. Arch Pharm (Weinheim) 2024; 357:e2300404. [PMID: 38010470 DOI: 10.1002/ardp.202300404] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/22/2023] [Accepted: 11/02/2023] [Indexed: 11/29/2023]
Abstract
Multitarget-directed ligands (MTDLs) have recently attracted significant interest due to their superior effectiveness in multifactorial Alzheimer's disease (AD). Combined inhibition of two important AD targets, glycogen synthase kinase-3β (GSK-3β) and dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A), may be a breakthrough in the treatment of AD. Based on our previous work, we have designed and synthesized a series of novel harmine derivatives, investigated their inhibition of GSK-3β and DYRK1A, and evaluated a variety of biological activities. The results of the experiments showed that most of these compounds exhibited good activity against GSK-3β and DYRK1A in vitro. ZLQH-5 was selected as the best compound due to the most potent inhibitory effect against GSK-3β and DYRK1A. Molecular docking studies demonstrated that ZLQH-5 could form stable interactions with the ATP binding pocket of GSK-3β and DYRK1A. In addition, ZLQH-5 showed low cytotoxicity against SH-SY5Y and HL-7702, good blood-brain barrier permeability, and favorable pharmacokinetic properties. More importantly, ZLQH-5 also attenuated the tau hyperphosphorylation in the okadaic acid SH-SY5Y cell model. These results indicated that ZLQH-5 could be a promising dual-target drug candidate for the treatment of AD.
Collapse
Affiliation(s)
- Jingsong Qiu
- Bei Fang Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang, China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiangling Feng
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Huanhua Chen
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Wenwu Liu
- Bei Fang Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang, China
| | - Wenjie Liu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Limeng Wu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Xudong Gao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, China
| | - Yanfang Liu
- Department of Clinical Trial Center, General Hospital of Northern Theater Command, Shenyang, China
| | - Yaoguang Huang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Hao Gong
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yiming Qi
- Department of Graduate School, Dalian Medical University, Dalian, China
| | - Zihua Xu
- Bei Fang Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Qingchun Zhao
- Bei Fang Hospital of Shenyang Pharmaceutical University, General Hospital of Northern Theater Command, Shenyang, China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
7
|
Madhav H, Abdel-Rahman SA, Hashmi MA, Rahman MA, Rehan M, Pal K, Nayeem SM, Gabr MT, Hoda N. Multicomponent Petasis reaction for the identification of pyrazine based multi-target directed anti-Alzheimer's agents: In-silico design, synthesis, and characterization. Eur J Med Chem 2023; 254:115354. [PMID: 37043996 DOI: 10.1016/j.ejmech.2023.115354] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/14/2023]
Abstract
Multi-target directed ligands (MTDLs) have recently attracted significant interest due to their exceptional effectiveness against multi-factorial Alzheimer's disease. The present work described the development of pyrazine-based MTDLs using multicomponent Petasis reaction for the dual inhibition of tau-aggregation and human acetylcholinesterase (hAChE). The molecular structure of synthesized ligands was validated by 1H & 13C NMR and mass spectrometry. The screened compounds were shown to have a strong inhibitory effect at 10 μM concentration against tau-oligomerization and hAChE, but only moderate inhibitory activity against Aβ42. Among all the compounds, the half-maximal inhibitory concentration (IC50) for 21 and 24 against hAChE were 0.71 μM and 1.09 μM, respectively, while they displayed half-maximal effective concentrations (EC50) values of 2.21 μM and 2.71 μM for cellular tau-oligomerization, respectively. Additionally, an MTT experiment using tau-expressing SH-SY5Y neuroblastoma cells revealed that 21 was more neuroprotective than the FDA-approved medication donepezil. Furthermore, an MD simulation study was performed to investigate the dynamics and stability of AChE-21 and AChE-24 complexes in an aqueous environment. The MM-PBSA calculations were performed to evaluate the binding of 21 and 24 with AChE, and the relative binding energy was calculated as -870.578 and -875.697 kJ mol-1, respectively. As a result, the study offered insight into the design of new MTDLs and highlighted 21 as a potential roadblock to the development of anti-AD medications.
Collapse
Affiliation(s)
- Hari Madhav
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India
| | - Somaya A Abdel-Rahman
- Molecular Imaging Innovations Institute (MI3), Department of Radiology, Weill Cornell Medicine, New York, NY, 10065, USA; Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Md Amiruddin Hashmi
- Interdisciplinary Biotechnology Unit, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Md Ataur Rahman
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Mohammad Rehan
- Max-Planck-Institute für Molekulare Physiologie, Abteilung Chemische Biologie, Otto-Hahn-Straße 11, 44227, Dortmund, Germany
| | - Kavita Pal
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India
| | - Shahid M Nayeem
- Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Moustafa T Gabr
- Molecular Imaging Innovations Institute (MI3), Department of Radiology, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Nasimul Hoda
- Drug Design and Synthesis Laboratory, Department of Chemistry, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
8
|
Liu X, Lai LY, Chen JX, Li X, Wang N, Zhou LJ, Jiang XW, Hu XL, Liu WW, Jiao XM, Qi ZT, Liu WJ, Wu LM, Huang YG, Xu ZH, Zhao QC. An inhibitor with GSK3β and DYRK1A dual inhibitory properties reduces Tau hyperphosphorylation and ameliorates disease in models of Alzheimer's disease. Neuropharmacology 2023; 232:109525. [PMID: 37004752 DOI: 10.1016/j.neuropharm.2023.109525] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/28/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023]
Abstract
Since Alzheimer's disease (AD) is a complex and multifactorial neuropathology, the discovery of multi-targeted inhibitors has gradually demonstrated greater therapeutic potential. Neurofibrillary tangles (NFTs), the main neuropathologic hallmarks of AD, are mainly associated with hyperphosphorylation of the microtubule-associated protein Tau. The overexpression of GSK3β and DYRK1A has been recognized as an important contributor to hyperphosphorylation of Tau, leading to the strategy of using dual-targets inhibitors for the treatment of this disorder. ZDWX-12 and ZDWX-25, as harmine derivatives, were found good inhibition on dual targets in our previous study. Here, we firstly evaluated the inhibition effect of Tau hyperphosphorylation using two compounds by HEK293-Tau P301L cell-based model and okadaic acid (OKA)-induced mouse model. We found that ZDWX-25 was more effective than ZDWX-12. Then, based on comprehensively investigations on ZDWX-25 in vitro and in vivo, 1) the capability of ZDWX-25 to show a reduction in phosphorylation of multiple Tau epitopes in OKA-induced neurodegeneration cell models, and 2) the effect of reduction on NFTs by 3xTg-AD mouse model under administration of ZDWX-25, an orally bioavailable, brain-penetrant dual-targets inhibitor with low toxicity. Our data highlight that ZDWX-25 is a promising drug for treating AD.
Collapse
|
9
|
Liu W, Tian L, Wu L, Chen H, Wang N, Liu X, Zhao C, Wu Z, Jiang X, Wu Q, Xu Z, Liu W, Zhao Q. Discovery of novel β-carboline-1,2,3-triazole hybrids as AChE/GSK-3β dual inhibitors for Alzheimer's disease treatment. Bioorg Chem 2022; 129:106168. [DOI: 10.1016/j.bioorg.2022.106168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/02/2022]
|
10
|
V. Bala Aakash, Ramalakshmi N, Bhuvaneswari S, Sankari E, Arunkumar S. Comprehensive Review on Versatile Pharmacology of Quinoxaline Derivative. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1068162022040069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
11
|
Rai H, Gupta S, Kumar S, Yang J, Singh SK, Ran C, Modi G. Near-Infrared Fluorescent Probes as Imaging and Theranostic Modalities for Amyloid-Beta and Tau Aggregates in Alzheimer's Disease. J Med Chem 2022; 65:8550-8595. [PMID: 35759679 DOI: 10.1021/acs.jmedchem.1c01619] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A person suspected of having Alzheimer's disease (AD) is clinically diagnosed for the presence of principal biomarkers, especially misfolded amyloid-beta (Aβ) and tau proteins in the brain regions. Existing radiotracer diagnostic tools, such as PET imaging, are expensive and have limited availability for primary patient screening and pre-clinical animal studies. To change the status quo, small-molecular near-infrared (NIR) probes have been rapidly developed, which may serve as an inexpensive, handy imaging tool to comprehend the dynamics of pathogenic progression in AD and assess therapeutic efficacy in vivo. This Perspective summarizes the biochemistry of Aβ and tau proteins and then focuses on structurally diverse NIR probes with coverages of their spectroscopic properties, binding affinity toward Aβ and tau species, and theranostic effectiveness. With the summarized information and perspective discussions, we hope that this paper may serve as a guiding tool for designing novel in vivo imaging fluoroprobes with theranostic capabilities in the future.
Collapse
Affiliation(s)
- Himanshu Rai
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P.-221005, India
| | - Sarika Gupta
- Molecular Science Laboratory, National Institute of Immunology, New Delhi-110067, India
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Jian Yang
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Sushil K Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P.-221005, India
| | - Chongzhao Ran
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Gyan Modi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P.-221005, India
| |
Collapse
|
12
|
Soloperto A, Quaglio D, Baiocco P, Romeo I, Mori M, Ardini M, Presutti C, Sannino I, Ghirga S, Iazzetti A, Ippoliti R, Ruocco G, Botta B, Ghirga F, Di Angelantonio S, Boffi A. Rational design and synthesis of a novel BODIPY-based probe for selective imaging of tau tangles in human iPSC-derived cortical neurons. Sci Rep 2022; 12:5257. [PMID: 35347170 PMCID: PMC8960764 DOI: 10.1038/s41598-022-09016-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/15/2022] [Indexed: 12/26/2022] Open
Abstract
Numerous studies have shown a strong correlation between the number of neurofibrillary tangles of the tau protein and Alzheimer's disease progression, making the quantitative detection of tau very promising from a clinical point of view. However, the lack of highly reliable fluorescent probes for selective imaging of tau neurofibrillary tangles is a major challenge due to sharing similar β–sheet motifs with homologous Amyloid-β fibrils. In the current work, we describe the rational design and the in silico evaluation of a small-size focused library of fluorescent probes, consisting of a BODIPY core (electron acceptor) featuring highly conjugated systems (electron donor) with a length in the range 13–19 Å at C3. Among the most promising probes in terms of binding mode, theoretical affinity and polarity, BT1 has been synthesized and tested in vitro onto human induced pluripotent stem cells derived neuronal cell cultures. The probe showed excellent photophysical properties and high selectivity allowing in vitro imaging of hyperphosphorylated tau protein filaments with minimal background noise. Our findings offer new insight into the structure-activity relationship of this class of tau selective fluorophores, paving the way for boosting tau tangle detection in patients possibly through retinal spectral scans.
Collapse
Affiliation(s)
- Alessandro Soloperto
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs, Department of Excellence 2018-2022, Sapienza University of Rome, 00185, Rome, Italy
| | - Paola Baiocco
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy.,Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, 00185, Rome, Italy
| | - Isabella Romeo
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy.,Department of Chemistry and Technology of Drugs, Department of Excellence 2018-2022, Sapienza University of Rome, 00185, Rome, Italy
| | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, 53100, Siena, Italy
| | - Matteo Ardini
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Caterina Presutti
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy.,Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, 00185, Rome, Italy
| | - Ida Sannino
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy
| | - Silvia Ghirga
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy
| | - Antonia Iazzetti
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of Sacred Heart, 00168, Rome, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Giancarlo Ruocco
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy
| | - Bruno Botta
- Department of Chemistry and Technology of Drugs, Department of Excellence 2018-2022, Sapienza University of Rome, 00185, Rome, Italy
| | - Francesca Ghirga
- Department of Chemistry and Technology of Drugs, Department of Excellence 2018-2022, Sapienza University of Rome, 00185, Rome, Italy.
| | - Silvia Di Angelantonio
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy. .,Department of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy.
| | - Alberto Boffi
- Center for Life Nano- & Neuro-Science, Istituto Italiano Di Tecnologia, 00161, Rome, Italy.,Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, 00185, Rome, Italy
| |
Collapse
|
13
|
A Novel Fluoro-Pyrazine-Bridged Donor-Accepter-Donor Fluorescent Probe for Lipid Droplet-Specific Imaging in Diverse Cells and Superoxide Anion Generation. Pharm Res 2022; 39:1205-1214. [PMID: 35237921 DOI: 10.1007/s11095-022-03216-y] [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: 10/19/2021] [Accepted: 02/23/2022] [Indexed: 10/19/2022]
Abstract
PURPOSE Lipid droplets (LDs) are dynamic organelles which associated with many metabolic processes. Reliable long-term imaging of LD is of great importance in LD-based therapy and research. Conventional fluorescent probes suffer from poor photostability and difficulty of preparation, which compromise their LD imaging ability. In this study, we aim to provide a novel and universal fluorescent probe for LD-specific imaging in both eukaryotic and prokaryotic cells. The versatile and potential applications of the probe were also evaluated. METHODS We used one-step Suzuki coupling reaction to synthesize a fluoro-pyrazine-bridged donor-acceptor-donor fluorescent probe (T-FP-T). The fluorescent properties and stability of T-FP-T were detected. Then, LD-specific imaging and dynamic movement tracking capabilities of T-FP-T were studied in fungus, bacteria, plant and animal tissues. The biosafety and photodynamic toxicity of the probe under different light irradiation were characterized. RESULTS T-FP-T showed large Stokes shift, superior brightness, excellent photostability, low toxicity. T-FP-T exhibited significant overlaps with adipophilin antibody or the commercial LD probe (LipidSpot™) in the cytoplasm, but not with Mitotracker red, Lysotracker red and Peroxisome Labeling dye. Moreover, T-FP-T also showed efficient superoxide anion generation capability under white LED light irradiation. The viability of Hela cells co-treated with T-FP-T and 1-h white LED light irradiation decreased to 62%. CONCLUSIONS All these outstanding capabilities make T-FP-T a new efficient LD-specific imaging probe. The generated superoxide anion from T-FP-T under white LED light irradiation could cause obvious cell death, which will inspire broad study in LD-targeted photodynamic therapy.
Collapse
|
14
|
Taheri M, Aslani S, Ghafouri H, Mohammadi A, Akbary Moghaddam V, Moradi N, Naeimi H. Synthesis, in vitro biological evaluation and molecular modelling of new 2-chloro-3-hydrazinopyrazine derivatives as potent acetylcholinesterase inhibitors on PC12 cells. BMC Chem 2022; 16:7. [PMID: 35193649 PMCID: PMC8864858 DOI: 10.1186/s13065-022-00799-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 02/14/2022] [Indexed: 11/30/2022] Open
Abstract
Background The loss of cholinergic neurotransmission in Alzheimer's disease (AD) patients' brain is accompanied by a reduced concentration of Acetylcholine (ACh) within synaptic clefts. Thus, the use of acetylcholinesterase inhibitors (AChEIs) to block the cholinergic degradation of ACh is a promising approach for AD treatment. In the present study, a series of 2-chloro-3-hydrazinopyrazine derivatives (CHP1-5) were designed, synthesized, and biologically evaluated as potential multifunctional anti-AD agents. Methods In addition, the chemical structures and purity of the synthesized compounds were elucidated through using IR, 1H and 13C NMR, and elemental analyses. Further, the intended compounds were assessed in vitro for their AChE inhibitory and neuroprotective effects. Furthermore, DPPH, FRAP and ABTS assays were utilized to determine their antioxidant activity. The statistical analysis was performed using one-way ANOVA. Results Based on the results, CHP4 and CHP5 exhibited strong AChE inhibitory effects with the IC50 values of 3.76 and 4.2 µM compared to the donepezil (0.53 µM), respectively. The study examined the effect and molecular mechanism of CHP4 on the Ab1–42-induced cytotoxicity in differentiated PC12 cells. At concentrations of 0–100 μM, CHP4 was non-toxic in PC12. Additionally, Ab1–42 significantly stimulated tau hyperphosphorylation and induced differentiated PC12 cell death. Further, CHP4 resulted in diminishing the Ab1–42-induced toxicity in PC12 cell significantly. CHP4 at 30 μM concentration significantly increased the Ab1–42-induced HSP70 expression and decreased tau hyperphosphorylation. Conclusions According to the results of our studies CHP4 can be considered as safe and efficient AChEI and employed as a potential multifunctional anti-AD agent. Supplementary Information The online version contains supplementary material available at 10.1186/s13065-022-00799-w.
Collapse
Affiliation(s)
- Maryam Taheri
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran
| | - Samira Aslani
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran
| | - Hossein Ghafouri
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran. .,Department of Marine Sciences, The Caspian Sea Basin Research Center, University of Guilan, Rasht, Iran.
| | - Asadollah Mohammadi
- Department of Chemistry, Faculty of Sciences, University of Guilan, Rasht, Iran
| | | | - Nastarn Moradi
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran
| | - Hananeh Naeimi
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran
| |
Collapse
|
15
|
Discovery of novel β-carboline derivatives as selective AChE inhibitors with GSK-3β inhibitory property for the treatment of Alzheimer's disease. Eur J Med Chem 2021; 229:114095. [PMID: 34995924 DOI: 10.1016/j.ejmech.2021.114095] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/27/2021] [Accepted: 12/27/2021] [Indexed: 12/12/2022]
Abstract
The natural product harmine, a representative β-carboline alkaloid from the seeds of Peganum harmala L. (Zygophyllaceae), possesses a broad spectrum of biological activities. In this study, a novel series of harmine derivatives containing N-benzylpiperidine moiety were identified for the treatment of Alzheimer's disease (AD). The results showed that all the derivatives possessed significant anti-acetylcholinesterase (AChE) activity and good selectivity over butyrylcholinesterase (BChE). In particular, compound ZLWH-23 exhibited potent anti-AChE activity (IC50 = 0.27 μM) and selective BChE inhibition (IC50 = 20.82 μM), as well as acceptable glycogen synthase kinase-3 (GSK-3β) inhibition (IC50 = 6.78 μM). Molecular docking studies and molecular dynamics simulations indicated that ZLWH-23 could form stable interaction with AChE and GSK-3β. Gratifyingly, ZLWH-23 exhibited good selectivity for GSK-3β over multi-kinases and very low cytotoxicity towards SH-SY5Y, HEK-293T, HL-7702, and HepG2 cell lines. Importantly, ZLWH-23 displayed efficient reduction against tau hyperphosphorylation on Ser-396 site in Tau (P301L) 293T cell model. Collectively, harmine-based derivatives could be considered as possible drug leads for the development of AD therapies.
Collapse
|
16
|
Amaya‐García F, Caldera M, Koren A, Kubicek S, Menche J, Unterlass MM. Green Hydrothermal Synthesis of Fluorescent 2,3-Diarylquinoxalines and Large-Scale Computational Comparison to Existing Alternatives. CHEMSUSCHEM 2021; 14:1853-1863. [PMID: 33662183 PMCID: PMC8252754 DOI: 10.1002/cssc.202100433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Indexed: 06/05/2023]
Abstract
Here, the hydrothermal synthesis (HTS) of 2,3-diarylquinoxalines from 1,2-diketones and o-phenylendiamines (o-PDAs) was achieved. The synthesis is simple, fast, and generates high yields, without requiring any organic solvents, strong acids or toxic catalysts. Reaction times down to <10 min without decrease in yield could be achieved through adding acetic acid as promoter, even for highly apolar biquinoxalines (yield >90 % in all cases). Moreover, it was shown that HTS has high compatibility: (i) hydrochlorides, a standard commercial form of amines, could be used directly as combined amine source and acidic catalyst, and (ii) Boc-diprotected o-PDA could be directly employed as substrate that underwent HT deprotection. A systematic large-scale computational comparison of all reported syntheses of the presented quinoxalines from the same starting compounds showed that this method is more environmentally friendly and less toxic than all existing methods and revealed generic synthetic routes for improving reaction yields. Finally, the application of the synthesized compounds as fluorescent dyes for cell staining was explored.
Collapse
Affiliation(s)
- Fabián Amaya‐García
- Institute of Applied Synthetic ChemistryTechnische Universität WienGetreidemarkt 9/1631060ViennaAustria
- Institute of Materials ChemistryTechnische Universität WienGetreidemarkt 9/1651060ViennaAustria
| | - Michael Caldera
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesLazarettgasse 141090ViennaAustria
- Max Perutz LabsCampus Vienna Biocenter 51030ViennaAustria
| | - Anna Koren
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesLazarettgasse 141090ViennaAustria
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesLazarettgasse 141090ViennaAustria
| | - Jörg Menche
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesLazarettgasse 141090ViennaAustria
- Max Perutz LabsCampus Vienna Biocenter 51030ViennaAustria
| | - Miriam M. Unterlass
- Institute of Applied Synthetic ChemistryTechnische Universität WienGetreidemarkt 9/1631060ViennaAustria
- Institute of Materials ChemistryTechnische Universität WienGetreidemarkt 9/1651060ViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesLazarettgasse 141090ViennaAustria
| |
Collapse
|
17
|
Zhou J, Jangili P, Son S, Ji MS, Won M, Kim JS. Fluorescent Diagnostic Probes in Neurodegenerative Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2001945. [PMID: 32902000 DOI: 10.1002/adma.202001945] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/19/2020] [Indexed: 05/22/2023]
Abstract
Neurodegenerative diseases are debilitating disorders that feature progressive and selective loss of function or structure of anatomically or physiologically associated neuronal systems. Both chronic and acute neurodegenerative diseases are associated with high morbidity and mortality along with the death of neurons in different areas of the brain; moreover, there are few or no effective curative therapy options for treating these disorders. There is an urgent need to diagnose neurodegenerative disease as early as possible, and to distinguish between different disorders with overlapping symptoms that will help to decide the best clinical treatment. Recently, in neurodegenerative disease research, fluorescent-probe-mediated biomarker visualization techniques have been gaining increasing attention for the early diagnosis of neurodegenerative diseases. A survey of fluorescent probes for sensing and imaging biomarkers of neurodegenerative diseases is provided. These imaging probes are categorized based on the different potential biomarkers of various neurodegenerative diseases, and their advantages and disadvantages are discussed. Guides to develop new sensing strategies, recognition mechanisms, as well as the ideal features to further improve neurodegenerative disease fluorescence imaging are also explored.
Collapse
Affiliation(s)
- Jin Zhou
- College of Pharmacy, Weifang Medical University, Weifang, 261053, China
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Paramesh Jangili
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Subin Son
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Myung Sun Ji
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Miae Won
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| |
Collapse
|
18
|
Zhang X, Zhou J, Gu Z, Zhang H, Gong Q, Luo K. Advances in nanomedicines for diagnosis of central nervous system disorders. Biomaterials 2020; 269:120492. [PMID: 33153757 DOI: 10.1016/j.biomaterials.2020.120492] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/18/2020] [Accepted: 10/23/2020] [Indexed: 02/08/2023]
Abstract
In spite of a great improvement in medical health services and an increase in lifespan, we have witnessed a skyrocket increase in the incidence of central nervous system (CNS) disorders including brain tumors, neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease), ischemic stroke, and epilepsy, which have seriously undermined the quality of life and substantially increased economic and societal burdens. Development of diagnostic methods for CNS disorders is still in the early stage, and the clinical outcomes suggest these methods are not ready for the challenges associated with diagnosis of CNS disorders, such as early detection, specific binding, sharp contrast, and continuous monitoring of therapeutic interventions. Another challenge is to overcome various barrier structures during delivery of diagnostic agents, especially the blood-brain barrier (BBB). Fortunately, utilization of nanomaterials has been pursued as a potential and promising strategy to address these challenges. This review will discuss anatomical and functional structures of BBB and transport mechanisms of nanomaterials across the BBB, and special emphases will be placed on the state-of-the-art advances in the development of nanomedicines from a variety of nanomaterials for diagnosis of CNS disorders. Meanwhile, current challenges and future perspectives in this field are also highlighted.
Collapse
Affiliation(s)
- Xun Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Zhou
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
19
|
Bautista‐Aguilera ÓM, Ismaili L, Chioua M, Iriepa I, Ángeles Martinez‐Grau M, Beadle CD, Vetman T, López‐Muñoz F, Marco‐Contelles J. Automated Synthesis of New Quinoxalinetacrines. ChemistrySelect 2020. [DOI: 10.1002/slct.202001593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Óscar M. Bautista‐Aguilera
- Department of Organic Chemistry and Inorganic ChemistryAlcalá University 28805-Alcalá de Henares Madrid Spain
- Institute of Chemical Research Andrés M. del RíoAlcalá University 28805-Alcalá de Henares Madrid Spain
| | - Lhassane Ismaili
- Laboratoire de Chimie Organique et ThérapeutiqueNeurosciences intégratives et cliniques EA 481, Univ. Bourgogne Franche-Comté, UFR Santé 19, rue Ambroise Paré F-25000 Besançon France
| | - Mourad Chioua
- Laboratory of Medicinal Chemistry (IQOG, CSIC) C/ Juan de la Cierva 3 28006- Madrid Spain
| | - Isabel Iriepa
- Department of Organic Chemistry and Inorganic ChemistryAlcalá University 28805-Alcalá de Henares Madrid Spain
- Institute of Chemical Research Andrés M. del RíoAlcalá University 28805-Alcalá de Henares Madrid Spain
| | | | - Christopher D. Beadle
- Lilly Research CentreEli Lilly & Company, Erl Wood Manor Windlesham, Surrey GU20 6PH UK
| | - Tatiana Vetman
- Lilly Research LaboratoriesEli Lilly & Company Indianapolis IN 46285 USA
| | - Francisco López‐Muñoz
- Faculty of HealthCamilo José Cela University of Madrid (UCJC) Spain
- Neuropsychopharmacology Unit, “Hospital 12 de Octubre”Research Institute Madrid Spain
| | - José Marco‐Contelles
- Laboratory of Medicinal Chemistry (IQOG, CSIC) C/ Juan de la Cierva 3 28006- Madrid Spain
| |
Collapse
|
20
|
Arora H, Ramesh M, Rajasekhar K, Govindaraju T. Molecular Tools to Detect Alloforms of Aβ and Tau: Implications for Multiplexing and Multimodal Diagnosis of Alzheimer’s Disease. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2020. [DOI: 10.1246/bcsj.20190356] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Harshit Arora
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
| | - Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
| | - Kolla Rajasekhar
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
- VNIR Biotechnologies Pvt. Ltd., Bangalore Bioinnovation Center, Helix Biotech Park, Electronic City Phase I, Bengaluru 560100, Karnataka, India
| |
Collapse
|
21
|
Chen W, Du Y, Wang M, Fang Y, Yu W, Chang J. Synthesis of benzo[4,5]imidazo[1,2-a]quinoxalines by I2-mediated sp3 C–H amination. Org Chem Front 2020. [DOI: 10.1039/d0qo01101k] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Benzo[4,5]imidazo[1,2-a]quinoxalines were synthesized by I2-mediated sp3 C–H amination under transition-metal free conditions in an efficient and scalable fashion.
Collapse
Affiliation(s)
- Wenjun Chen
- Green Catalysis Center
- and College of Chemistry
- Zhengzhou University
- Zhengzhou
- China
| | - Yangxu Du
- Green Catalysis Center
- and College of Chemistry
- Zhengzhou University
- Zhengzhou
- China
| | - Manman Wang
- Green Catalysis Center
- and College of Chemistry
- Zhengzhou University
- Zhengzhou
- China
| | - Yingchao Fang
- Green Catalysis Center
- and College of Chemistry
- Zhengzhou University
- Zhengzhou
- China
| | - Wenquan Yu
- Green Catalysis Center
- and College of Chemistry
- Zhengzhou University
- Zhengzhou
- China
| | - Junbiao Chang
- Green Catalysis Center
- and College of Chemistry
- Zhengzhou University
- Zhengzhou
- China
| |
Collapse
|