1
|
Yang X, Gao X, Jiang X, Yue K, Luo P. Targeting capabilities of engineered extracellular vesicles for the treatment of neurological diseases. Neural Regen Res 2025; 20:3076-3094. [PMID: 39435635 DOI: 10.4103/nrr.nrr-d-24-00462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/07/2024] [Indexed: 10/23/2024] Open
Abstract
Recent advances in research on extracellular vesicles have significantly enhanced their potential as therapeutic agents for neurological diseases. Owing to their therapeutic properties and ability to cross the blood-brain barrier, extracellular vesicles are recognized as promising drug delivery vehicles for various neurological conditions, including ischemic stroke, traumatic brain injury, neurodegenerative diseases, glioma, and psychosis. However, the clinical application of natural extracellular vesicles is hindered by their limited targeting ability and short clearance from the body. To address these limitations, multiple engineering strategies have been developed to enhance the targeting capabilities of extracellular vesicles, thereby enabling the delivery of therapeutic contents to specific tissues or cells. Therefore, this review aims to highlight the latest advancements in natural and targeting-engineered extracellular vesicles, exploring their applications in treating traumatic brain injury, ischemic stroke, Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, glioma, and psychosis. Additionally, we summarized recent clinical trials involving extracellular vesicles and discussed the challenges and future prospects of using targeting-engineered extracellular vesicles for drug delivery in treating neurological diseases. This review offers new insights for developing highly targeted therapies in this field.
Collapse
Affiliation(s)
- Xinyu Yang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | | | | | | | | |
Collapse
|
2
|
Yoo J, Lee J, Ahn B, Han J, Lim MH. Multi-target-directed therapeutic strategies for Alzheimer's disease: controlling amyloid-β aggregation, metal ion homeostasis, and enzyme inhibition. Chem Sci 2025:d4sc06762b. [PMID: 39810997 PMCID: PMC11726323 DOI: 10.1039/d4sc06762b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia, marked by progressive cognitive decline and memory impairment. Despite advances in therapeutic research, single-target-directed treatments often fall short in addressing the complex, multifactorial nature of AD. This arises from various pathological features, including amyloid-β (Aβ) aggregate deposition, metal ion dysregulation, oxidative stress, impaired neurotransmission, neuroinflammation, mitochondrial dysfunction, and neuronal cell death. This review illustrates their interrelationships, with a particular emphasis on the interplay among Aβ, metal ions, and AD-related enzymes, such as β-site amyloid precursor protein cleaving enzyme 1 (BACE1), matrix metalloproteinase 9 (MMP9), lysyl oxidase-like 2 (LOXL2), acetylcholinesterase (AChE), and monoamine oxidase B (MAOB). We further underscore the potential of therapeutic strategies that simultaneously inhibit Aβ aggregation and address other pathogenic mechanisms. These approaches offer a more comprehensive and effective method for combating AD, overcoming the limitations of conventional therapies.
Collapse
Affiliation(s)
- Jeasang Yoo
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jimin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Byeongha Ahn
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jiyeon Han
- Department of Applied Chemistry, University of Seoul Seoul 02504 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
3
|
Kwak J, Yi Y, Park S, Lim MH. Multi-target macrocycles: pyrogallol derivatives to control multiple pathological factors associated with Alzheimer's disease. Chem Sci 2025; 16:889-900. [PMID: 39660289 PMCID: PMC11626466 DOI: 10.1039/d4sc06417h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024] Open
Abstract
Designing multi-target chemical tools is a vital approach to understanding the pathology of Alzheimer's disease (AD), which involves a complex network of pathological factors, such as free organic radicals, amyloid-β (Aβ), and metal-bound Aβ (metal-Aβ). The pyrogallol moiety, known for its ability to lower redox potentials and interact with both Aβ and metal ions, presents a promising framework for this molecular design. Here we show how simple structural variations of pyrogallol can be used to enhance its ability to scavenge free organic radicals and regulate the aggregation of both metal-free Aβ and metal-Aβ. By incorporating multiple pyrogllol units into a macrocyclic scaffold via methylene bridges, we achieve synergistic reactivity against several pathological targets. Our structure-reactivity relationship studies also reveal that the macrocyclic structure noticeably improves antioxidant activity as well as interactions with both Aβ and metal ions, leading to oxidation of Aβ peptides and influencing their conformation and aggregation in both the absence and presence of metal ions. This work demonstrates the potential of simple redox-active structural entities in developing multifunctional chemical reagents that effectively manage the pathological components associated with AD.
Collapse
Affiliation(s)
- Jimin Kwak
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Yelim Yi
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Seongmin Park
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
4
|
Das V, Miller JH, Alladi CG, Annadurai N, De Sanctis JB, Hrubá L, Hajdúch M. Antineoplastics for treating Alzheimer's disease and dementia: Evidence from preclinical and observational studies. Med Res Rev 2024; 44:2078-2111. [PMID: 38530106 DOI: 10.1002/med.22033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024]
Abstract
As the world population ages, there will be an increasing need for effective therapies for aging-associated neurodegenerative disorders, which remain untreatable. Dementia due to Alzheimer's disease (AD) is one of the leading neurological diseases in the aging population. Current therapeutic approaches to treat this disorder are solely symptomatic, making the need for new molecular entities acting on the causes of the disease extremely urgent. One of the potential solutions is to use compounds that are already in the market. The structures have known pharmacokinetics, pharmacodynamics, toxicity profiles, and patient data available in several countries. Several drugs have been used successfully to treat diseases different from their original purposes, such as autoimmunity and peripheral inflammation. Herein, we divulge the repurposing of drugs in the area of neurodegenerative diseases, focusing on the therapeutic potential of antineoplastics to treat dementia due to AD and dementia. We briefly touch upon the shared pathological mechanism between AD and cancer and drug repurposing strategies, with a focus on artificial intelligence. Next, we bring out the current status of research on the development of drugs, provide supporting evidence from retrospective, clinical, and preclinical studies on antineoplastic use, and bring in new areas, such as repurposing drugs for the prion-like spreading of pathologies in treating AD.
Collapse
Affiliation(s)
- Viswanath Das
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - John H Miller
- School of Biological Sciences and Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Charanraj Goud Alladi
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Narendran Annadurai
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - Lenka Hrubá
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| |
Collapse
|
5
|
Fang S, Zhang K, Liu D, Yang Y, Xi H, Xie W, Diao K, Rao Z, Wang D, Yang W. Polyphenol-based polymer nanoparticles for inhibiting amyloid protein aggregation: recent advances and perspectives. Front Nutr 2024; 11:1408620. [PMID: 39135555 PMCID: PMC11317421 DOI: 10.3389/fnut.2024.1408620] [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: 04/03/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
Polyphenols are a group of naturally occurring compounds that possess a range of biological properties capable of potentially mitigating or preventing the progression of age-related cognitive decline and Alzheimer's disease (AD). AD is a chronic neurodegenerative disease known as one of the fast-growing diseases, especially in the elderly population. Moreover, as the primary etiology of dementia, it poses challenges for both familial and societal structures, while also imposing a significant economic strain. There is currently no pharmacological intervention that has demonstrated efficacy in treating AD. While polyphenols have exhibited potential in inhibiting the pathological hallmarks of AD, their limited bioavailability poses a significant challenge in their therapeutic application. Furthermore, in order to address the therapeutic constraints, several polymer nanoparticles are being explored as improved therapeutic delivery systems to optimize the pharmacokinetic characteristics of polyphenols. Polymer nanoparticles have demonstrated advantageous characteristics in facilitating the delivery of polyphenols across the blood-brain barrier, resulting in their efficient distribution within the brain. This review focuses on amyloid-related diseases and the role of polyphenols in them, in addition to discussing the anti-amyloid effects and applications of polyphenol-based polymer nanoparticles.
Collapse
Affiliation(s)
- Shuzhen Fang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Kangyi Zhang
- State Key Laboratory of Tea Plant Biology and Utilization, Key Laboratory of Food Nutrition and Safety, School of Tea, Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Danqing Liu
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Yulong Yang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Hu Xi
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Wenting Xie
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Ke Diao
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhihong Rao
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Wenming Yang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, Anhui, China
| |
Collapse
|
6
|
Gharai PK, Khan J, Pradhan K, Mallesh R, Garg S, Arshi MU, Barman S, Ghosh S. Power of Dopamine: Multifunctional Compound Assisted Conversion of the Most Risk Factor into Therapeutics of Alzheimer's Disease. ACS Chem Neurosci 2024; 15:2470-2483. [PMID: 38874606 DOI: 10.1021/acschemneuro.3c00777] [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] [Indexed: 06/15/2024] Open
Abstract
In Alzheimer's disease (AD), reactive oxygen species (ROS) plays a crucial role, which is produced from molecular oxygen with extracellular deposited amyloid-β (Aβ) aggregates through the reduction of a Cu2+ ion. In the presence of a small amount of redox-active Cu2+ ion, ROS is produced by the Aβ-Cu2+ complex as Aβ peptide alone is unable to generate excess ROS. Therefore, Cu2+ ion chelators are considered promising therapeutics against AD. Here, we have designed and synthesized a series of Schiff base derivatives (SB) based on 2-hydroxy aromatic aldehyde derivatives and dopamine. These SB compounds contain one copper chelating core, which captures the Cu2+ ions from the Aβ-Cu2+ complex. Thereby, it inhibits copper-induced amyloid aggregation as well as amyloid self-aggregation. It also inhibits copper-catalyzed ROS production through sequestering of Cu2+ ions. The uniqueness of our designed ligands has the dual property of dopamine, which not only acts as a ROS scavenger but also chelates the copper ion. The crystallographic analysis proves the power of the dopamine unit. Therefore, dual exploration of dopamine core can be considered as potential therapeutics for future AD treatment.
Collapse
Affiliation(s)
- Prabir Kumar Gharai
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR─Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar 342037, Rajasthan, India
| | - Juhee Khan
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR─Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar 342037, Rajasthan, India
| | - Krishnangsu Pradhan
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR─Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
| | - Rathnam Mallesh
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR─Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar 342037, Rajasthan, India
- National Institute of Pharmaceutical Education and Research, Kolkata, Chunilal Bhawan 168, Maniktala Main Road, Kolkata 700054, India
| | - Shubham Garg
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar 342037, Rajasthan, India
| | - Mohammad Umar Arshi
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar 342037, Rajasthan, India
| | - Surajit Barman
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR─Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
| | - Surajit Ghosh
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR─Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar 342037, Rajasthan, India
- National Institute of Pharmaceutical Education and Research, Kolkata, Chunilal Bhawan 168, Maniktala Main Road, Kolkata 700054, India
| |
Collapse
|
7
|
Sun Y, Zhang H, Liu R, Huang R, Gao Z, Tian L, Zhu Y, Liu Y, Lu C, Wu L. Lancao decoction alleviates cognitive dysfunction: A new therapeutic drug and its therapeutic mechanism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155531. [PMID: 38492366 DOI: 10.1016/j.phymed.2024.155531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 02/22/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Cognitive dysfunction (CD) is a neurodegenerative disease characterized primarily by the decline of learning and memory abilities. The physiological and pathological mechanisms of CD are very complex, which is mainly related to normal function of the hippocampus. Lancao decoction (LC) is a Chinese medicine formula, which has been used to treat neurodegenerative disorders. However, the potential of LC for the treatment of CD, as well as its underlying mechanisms, is unclear. PURPOSE In the study, we aimed to reveal the functional and neuronal mechanisms of LC's treatments for CD in scopolamine-induced mice. METHODS Gas chromatography (GC) was used to determine the stability of LC's extraction. CD model was established by the chronic induction of scopolamine (Scop, 1 mg/kg/day) for 1 week. Behavioral tests including morris water maze (MWM) and y-maze were used to evaluate learning and memory abilities of mice after LC's treatments. Immunofluorescence was used to detected the expressions of cFOS, Brdu and Ki67 after LC's treatments. Pharmacological blockade experiments explored the role of α-Amino-3‑hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) in LC's treatments for CD and its relationships with regeneration, activities and differentiation of neurons. RESULTS The results showed that LC was capable of improving spatial learning and memory and spontaneous alternating abilities in Scop-induced mice, which was similar to donepezil. LC could increase the number of cFOS positive cells, which was used as a marker of neuronal activity to upregulate by neuronal activities in hippocampus, but donepezil did not. Moreover, LC could strengthen neurogenesis and neuro-differentiation by increasing the number of Brdu and Ki67 positive cells in hippocampal dentate gyrus (DG), meanwhile, donepezil could only enhance the number of Ki67 positive cells. Transient inhibition of AMPAR by NBQX blunted the function of LC's treatment for CD and inhibited the enhanced effect of LC on Scop-induced hippocampal neuronal excitability and neurogenesis in mice. CONCLUSION To sum up, our study demonstrated that LC had the function of treating CD by enhancing content of acetylcholine (ACh) to activate AMPAR, which further up-regulated neurogenesis and neuronal differentiation to strengthen neuroactivities in hippocampus.
Collapse
Affiliation(s)
- Yan Sun
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China; College of Chinese Medicine & College of Integrated Chinese and Western Medicine, Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Hailou Zhang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders and School of Chinese Medicine, Jinan University, Guangzhou 510632, PR China.
| | - Ruiyi Liu
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders and School of Chinese Medicine, Jinan University, Guangzhou 510632, PR China
| | - Rumin Huang
- College of Chinese Medicine & College of Integrated Chinese and Western Medicine, Key Laboratory of Integrative Biomedicine for Brain Diseases, College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Ziwei Gao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China
| | - Liyuan Tian
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China
| | - Yaping Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China
| | - Yuxin Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China
| | - Chao Lu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China
| | - Lei Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Department of Pharmacy, Nanjing 210029, PR China.
| |
Collapse
|
8
|
Liu X, Su X, Chen M, Xie Y, Li M. Self-calibrating surface-enhanced Raman scattering-lateral flow immunoassay for determination of amyloid-β biomarker of Alzheimer's disease. Biosens Bioelectron 2024; 245:115840. [PMID: 37988777 DOI: 10.1016/j.bios.2023.115840] [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: 07/11/2023] [Revised: 10/24/2023] [Accepted: 11/12/2023] [Indexed: 11/23/2023]
Abstract
Rapid early diagnosis of Alzheimer's disease (AD) is critical for its effective and prompt treatment since the clinically available treatments can only relieve the symptoms or slow the disease progression. However, it is still a grand challenge to accurately diagnose AD at its early stage because of the indiscernible early symptoms and the lack of sensitive detection tools. Here, we develop a self-calibrating surface-enhanced Raman scattering (SERS)-lateral flow immunoassay (LFIA) biosensor for quantitative analysis of amyloid-β1-42 (Aβ1-42) biomarker in biofluids, enabling accurate AD diagnosis. The designed SERS-LFIA biosensor makes full use of the unique aspects of the LFIA format and the SERS technique to quantify the Aβ1-42 level in complex biofluids with high sensitivity, excellent anti-interference capability, low-cost, and operation simplicity. The key aspect of the design of this biosensor is that internal standard (IS)-SERS nanoparticles are embedded in the test line of the test strip as a self-calibration unit for correction of fluctuations of SERS signals caused by various external factors such as test parameters and sample heterogeneity. We demonstrate significant improvement of the detection performance of the SERS-LFIA biosensor for ratiometric quantification of Aβ1-42 owing to the built-in IS in the test line. We expect that the present IS-based biosensing strategy provides a promising tool for accurate AD diagnosis and longitudinal monitoring of therapeutic response with great promises for clinical translation.
Collapse
Affiliation(s)
- Xinyu Liu
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Xiaoming Su
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Mingyang Chen
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Yangcenzi Xie
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Ming Li
- School of Materials Science and Engineering, Central South University, Changsha, Hunan, 410083, China.
| |
Collapse
|
9
|
Zhu Y, Hu Z, Liu Y, Yan T, Liu L, Wang Y, Bai B. AChE activity self-breathing control mechanisms regulated by H 2S n and GSH: Persulfidation and glutathionylation on sulfhydryl after disulfide bonds cleavage. Int J Biol Macromol 2024; 259:129117. [PMID: 38211930 DOI: 10.1016/j.ijbiomac.2023.129117] [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: 07/13/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/13/2024]
Abstract
Hydrogen sulfide (H2S), or dihydrogen sulfane (H2Sn), acts as a signal molecule through the beneficial mechanism of persulfidation, known as the post-translational transformation of cysteine residues to persulfides. We previously reported that Glutathione (GSH) could regulate enzyme activity through S-desulfurization or glutathionylation of residues to generate protein-SG or protein-SSG, releasing H2S. However, little is known about the mechanisms by which H2Sn and GSH affect the disulfide bonds. In this study, we provide direct evidences that H2Sn and GSH modify the sulfhydryl group on Cys272, which forms disulfide bonds in acetylcholinesterase (AChE), to generate Cys-SSH and Cys-SSG, respectively. Glutathionylation of disulfide is a two-step reaction based on nucleophilic substitution, in which the first CS bond is broken, then the SS bond is broken to release H2S. H2Sn and GSH controlled self-breathing motion in enzyme catalysis by disconnecting specific disulfide bonds and modifying cysteine residues, thereby regulating AChE activity. Here, we elucidated H2Sn and GSH mechanisms on disulfide in the AChE system and proposed a self-breathing control theory induced by H2Sn and GSH. These theoretical findings shed light on the biological functions of H2Sn and GSH on sulfhydryl and disulfide bonds and enrich the theory of enzyme activity regulation.
Collapse
Affiliation(s)
- Yanwen Zhu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Zhaoliang Hu
- Department of Surgical Oncology, First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Yunen Liu
- Shenyang Medical College, Shenyang 110034, China
| | - Tingcai Yan
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Ling Liu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Yanqun Wang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Bing Bai
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China.
| |
Collapse
|
10
|
Long J, Qin F, Luo J, Zhong G, Huang S, Jing L, Yi T, Liu J, Jiang N. Design, synthesis, and biological evaluation of novel capsaicin-tacrine hybrids as multi-target agents for the treatment of Alzheimer's disease. Bioorg Chem 2024; 143:107026. [PMID: 38103330 DOI: 10.1016/j.bioorg.2023.107026] [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: 09/13/2023] [Revised: 11/18/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
A series of novel hybrid compounds were designed, synthesized, and utilized as multi-target drugs to treat Alzheimer's disease (AD) by connecting capsaicin and tacrine moieties. The biological assays indicated that most of these compounds demonstrated strong inhibition of acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) activities with IC50 values in the nanomolar, as well as good blood-brain barrier permeability. Among the synthesized hybrids, compound 5s displayed the most balanced inhibitory effect on hAChE (IC50 = 69.8 nM) and hBuChE (IC50 = 68.0 nM), and exhibited promising inhibitory activity against β-secretase-1 (BACE-1) (IC50 = 3.6 µM). Combining inhibition kinetics and molecular model analysis, compound 5s was shown to be a mixed inhibitor affecting both the catalytic active site (CAS) and peripheral anionic site (PAS) of hAChE. Additionally, compound 5s showed low toxicity in PC12 and BV2 cell assays. Moreover, compound 5s demonstrated good tolerance at the dose of up to 2500 mg/kg and exhibited no hepatotoxicity at the dose of 3 mg/kg in mice, and it could effectively improve memory ability in mice. Taken together, these findings suggest that compound 5s is a promising and effective multi-target agent for the potential treatment of AD.
Collapse
Affiliation(s)
- Juanyue Long
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, PR China
| | - Fengxue Qin
- Blood Transfusion Department, Affiliated Hospital of Youjiang Medical University For Nationalities, Baise, Guangxi 533000, PR China
| | - Jinchong Luo
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330006, PR China
| | - Guohui Zhong
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, PR China
| | - Shutong Huang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, PR China
| | - Lin Jing
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, PR China
| | - Tingzhuang Yi
- Department of Oncology, Affiliated Hospital of Youjiang Medical University For Nationalities, Baise, Guangxi 533000, PR China.
| | - Jing Liu
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, PR China; School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330006, PR China.
| | - Neng Jiang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, PR China.
| |
Collapse
|
11
|
Grcic L, Leech G, Kwan K, Storr T. Targeting misfolding and aggregation of the amyloid-β peptide and mutant p53 protein using multifunctional molecules. Chem Commun (Camb) 2024; 60:1372-1388. [PMID: 38204416 DOI: 10.1039/d3cc05834d] [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: 01/12/2024]
Abstract
Biomolecule misfolding and aggregation play a major role in human disease, spanning from neurodegeneration to cancer. Inhibition of these processes is of considerable interest, and due to the multifactorial nature of these diseases, the development of drugs that act on multiple pathways simultaneously is a promising approach. This Feature Article focuses on the development of multifunctional molecules designed to inhibit the misfolding and aggregation of the amyloid-β (Aβ) peptide in Alzheimer's disease (AD), and the mutant p53 protein in cancer. While for the former, the goal is to accelerate the removal of the Aβ peptide and associated aggregates, for the latter, the goal is reactivation via stabilization of the active folded form of mutant p53 protein and/or aggregation inhibition. Due to the similar aggregation pathway of the Aβ peptide and mutant p53 protein, a common therapeutic approach may be applicable.
Collapse
Affiliation(s)
- Lauryn Grcic
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada.
| | - Grace Leech
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada.
| | - Kalvin Kwan
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada.
| | - Tim Storr
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada.
| |
Collapse
|
12
|
Nam E, Lin Y, Park J, Do H, Han J, Jeong B, Park S, Lee DY, Kim M, Han J, Baik M, Lee Y, Lim MH. APP-C31: An Intracellular Promoter of Both Metal-Free and Metal-Bound Amyloid-β 40 Aggregation and Toxicity in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307182. [PMID: 37949680 PMCID: PMC10811509 DOI: 10.1002/advs.202307182] [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: 09/28/2023] [Revised: 10/18/2023] [Indexed: 11/12/2023]
Abstract
Intracellular C-terminal cleavage of the amyloid precursor protein (APP) is elevated in the brains of Alzheimer's disease (AD) patients and produces a peptide labeled APP-C31 that is suspected to be involved in the pathology of AD. But details about the role of APP-C31 in the development of the disease are not known. Here, this work reports that APP-C31 directly interacts with the N-terminal and self-recognition regions of amyloid-β40 (Aβ40 ) to form transient adducts, which facilitates the aggregation of both metal-free and metal-bound Aβ40 peptides and aggravates their toxicity. Specifically, APP-C31 increases the perinuclear and intranuclear generation of large Aβ40 deposits and, consequently, damages the nucleus leading to apoptosis. The Aβ40 -induced degeneration of neurites and inflammation are also intensified by APP-C31 in human neurons and murine brains. This study demonstrates a new function of APP-C31 as an intracellular promoter of Aβ40 amyloidogenesis in both metal-free and metal-present environments, and may offer an interesting alternative target for developing treatments for AD that have not been considered thus far.
Collapse
Affiliation(s)
- Eunju Nam
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Yuxi Lin
- Research Center for Bioconvergence AnalysisKorea Basic Science Institute (KBSI)OchangChungbuk28119Republic of Korea
| | - Jiyong Park
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
- Center for Catalytic Hydrocarbon FunctionalizationsInstitute for Basic Science (IBS)Daejeon34141Republic of Korea
| | - Hyunsu Do
- Graduate School of Medical Science and EngineeringKAISTDaejeon34141Republic of Korea
| | - Jiyeon Han
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Bohyeon Jeong
- Rare Disease Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)Daejeon34141Republic of Korea
| | - Subin Park
- Rare Disease Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)Daejeon34141Republic of Korea
- Department of BiochemistryDepartment of Medical ScienceChungnam National University School of MedicineDaejeon35015Republic of Korea
| | - Da Yong Lee
- Rare Disease Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)Daejeon34141Republic of Korea
| | - Mingeun Kim
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Jinju Han
- Graduate School of Medical Science and EngineeringKAISTDaejeon34141Republic of Korea
| | - Mu‐Hyun Baik
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
- Center for Catalytic Hydrocarbon FunctionalizationsInstitute for Basic Science (IBS)Daejeon34141Republic of Korea
| | - Young‐Ho Lee
- Research Center for Bioconvergence AnalysisKorea Basic Science Institute (KBSI)OchangChungbuk28119Republic of Korea
- Bio‐Analytical ScienceUniversity of Science and Technology (UST)Daejeon34113Republic of Korea
- Graduate School of Analytical Science and TechnologyChungnam National UniversityDaejeon34134Republic of Korea
- Department of Systems BiotechnologyChung‐Ang UniversityGyeonggi17546Republic of Korea
- Frontier Research Institute for Interdisciplinary SciencesTohoku UniversityMiyagi980‐8578Japan
| | - Mi Hee Lim
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| |
Collapse
|
13
|
Zhang M, Niu H, Li Q, Jiao L, Li H, Wu W. Active Compounds of Panax ginseng in the Improvement of Alzheimer's Disease and Application of Spatial Metabolomics. Pharmaceuticals (Basel) 2023; 17:38. [PMID: 38256872 PMCID: PMC10818864 DOI: 10.3390/ph17010038] [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: 11/13/2023] [Revised: 12/14/2023] [Accepted: 12/24/2023] [Indexed: 01/24/2024] Open
Abstract
Panax ginseng C.A. Meyer (P. ginseng) is one of the more common traditional Chinese medicines (TCMs). It contains numerous chemical components and exhibits a range of pharmacological effects. An enormous burden is placed on people's health and life by Alzheimer's disease (AD), a neurodegenerative condition. Recent research has shown that P. ginseng's chemical constituents, particularly ginsenosides, have a significant beneficial impact on the prevention and management of neurological disorders. To understand the current status of research on P. ginseng to improve AD, this paper discusses the composition of P. ginseng, its mechanism of action, and its clinical application. The pathogenesis of AD includes amyloid beta protein (Aβ) generation and aggregation, tau protein hyperphosphorylation, oxidant stress, neuroinflammation, mitochondrial damage, and neurotransmitter and gut microbiota disorders. This review presents the key molecular mechanisms and signaling pathways of the active ingredients in P. ginseng involved in improving AD from the perspective of AD pathogenesis. A P. ginseng-related signaling pathway network was constructed to provide effective targets for the treatment of AD. In addition, the application of spatial metabolomics techniques in studying P. ginseng and AD is discussed. In summary, this paper discusses research perspectives for the study of P. ginseng in the treatment of AD, including a systematic and in-depth review of the mechanisms of action of the active substances in P. ginseng, and evaluates the feasibility of applying spatial metabolomics in the study of AD pathogenesis and pharmacological treatment.
Collapse
Affiliation(s)
| | | | | | | | - Hui Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.Z.); (H.N.); (Q.L.); (L.J.)
| | - Wei Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (M.Z.); (H.N.); (Q.L.); (L.J.)
| |
Collapse
|
14
|
Li QY, Yu X, Li X, Bao LN, Zhang Y, Xie MJ, Jiang M, Wang YQ, Huang K, Xu L. Silicon-Carbon Dots-Loaded Mesoporous Silica Nanocomposites (mSiO 2@SiCDs): An Efficient Dual Inhibitor of Cu 2+-Mediated Oxidative Stress and Aβ Aggregation for Alzheimer's Disease. ACS APPLIED MATERIALS & INTERFACES 2023; 15:54221-54233. [PMID: 37962427 DOI: 10.1021/acsami.3c10053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The redox-active metal ions, especially Cu2+, are highly correlated to Alzheimer's disease (AD) by causing metal ion-mediated oxidative stress and toxic metal-bound β-amyloid (Aβ) aggregates. Numerous pieces of evidence have revealed that the regulation of metal homeostasis could be an effective therapeutic strategy for AD. Herein, in virtue of the interaction of both amino-containing silane and ethylenediaminetetraacetic acid disodium salt for Cu2+, the silicon-carbon dots (SiCDs) are deliberately prepared using these two raw materials as the cocarbon source; meanwhile, to realize the local enrichment of SiCDs and further maximize the chelating ability to Cu2+, the SiCDs are feasibly loaded to the biocompatible mesoporous silica nanoparticles (mSiO2) with the interaction between residual silane groups on SiCDs and silanol groups of mSiO2. Thus-obtained nanocomposites (i.e., mSiO2@SiCDs) could serve as an efficient Cu2+ chelator with satisfactory metal selectivity and further modulate the enzymic activity of free Cu2+ and the Aβ42-Cu2+ complex to alleviate the pathological oxidative stress with an anti-inflammatory effect. Besides, mSiO2@SiCDs show an inspiring inhibitory effect on Cu2+-mediated Aβ aggregation and further protect the neural cells against the toxic Aβ42-Cu2+ complex. Moreover, the transgenic Caenorhabditis elegans CL2120 assay demonstrates the protective efficacy of mSiO2@SiCDs on Cu2+-mediated Aβ toxicity in vivo, indicating its potential for AD treatment.
Collapse
Affiliation(s)
- Qin-Ying Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xu Yu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Jiangxia Laboratory, Wuhan 430200, China
| | - Xi Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li-Na Bao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Min-Jie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Ming Jiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ya Qian Wang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Xu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
15
|
Park S, Kim M, Lin Y, Hong M, Nam G, Mieczkowski A, Kardos J, Lee YH, Lim MH. Designing multi-target-directed flavonoids: a strategic approach to Alzheimer's disease. Chem Sci 2023; 14:9293-9305. [PMID: 37712013 PMCID: PMC10498667 DOI: 10.1039/d3sc00752a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 08/02/2023] [Indexed: 09/16/2023] Open
Abstract
The underlying causes of Alzheimer's disease (AD) remain a mystery, with multiple pathological components, including oxidative stress, acetylcholinesterase, amyloid-β, and metal ions, all playing a role. Here we report a strategic approach to designing flavonoids that can effectively tackle multiple pathological elements involved in AD. Our systematic investigations revealed key structural features for flavonoids to simultaneously target and regulate pathogenic targets. Our findings led to the development of a highly promising flavonoid that exhibits a range of functions, based on a complete structure-activity relationship analysis. Furthermore, our mechanistic studies confirmed that this flavonoid's versatile reactivities are driven by its redox potential and direct interactions with pathogenic factors. This work highlights the potential of multi-target-directed flavonoids as a novel solution in the fight against AD.
Collapse
Affiliation(s)
- Seongmin Park
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Mingeun Kim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Yuxi Lin
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI) Ochang Chungbuk 28119 Republic of Korea
| | - Mannkyu Hong
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
- Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS) Daejeon 34141 Republic of Korea
| | - Geewoo Nam
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Adam Mieczkowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences Pawińskiego 5a 02-106 Warsaw Poland
| | - József Kardos
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University Budapest 1117 Hungary
| | - Young-Ho Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI) Ochang Chungbuk 28119 Republic of Korea
- Bio-Analytical Science, University of Science and Technology (UST) Daejeon 34113 Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University Daejeon 34134 Republic of Korea
- Department of Systems Biotechnology, Chung-Ang University (CAU) Gyeonggi 17546 Republic of Korea
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University Sendai Miyagi 980-8578 Japan
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
16
|
Oliveri V. Unveiling the Effects of Copper Ions in the Aggregation of Amyloidogenic Proteins. Molecules 2023; 28:6446. [PMID: 37764220 PMCID: PMC10537474 DOI: 10.3390/molecules28186446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Amyloid diseases have become a global concern due to their increasing prevalence. Transition metals, including copper, can affect the aggregation of the pathological proteins involved in these diseases. Copper ions play vital roles in organisms, but the disruption of their homeostasis can negatively impact neuronal function and contribute to amyloid diseases with toxic protein aggregates, oxidative stress, mitochondrial dysfunction, impaired cellular signaling, inflammation, and cell death. Gaining insight into the imbalance of copper ions and its impact on protein folding and aggregation is crucial for developing focused therapies. This review examines the influence of copper ions on significant amyloid proteins/peptides, offering a comprehensive overview of the current understanding in this field.
Collapse
Affiliation(s)
- Valentina Oliveri
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A Doria 6, 95125 Catania, Italy
| |
Collapse
|
17
|
Maiuolo J, Costanzo P, Masullo M, D’Errico A, Nasso R, Bonacci S, Mollace V, Oliverio M, Arcone R. Hydroxytyrosol-Donepezil Hybrids Play a Protective Role in an In Vitro Induced Alzheimer's Disease Model and in Neuronal Differentiated Human SH-SY5Y Neuroblastoma Cells. Int J Mol Sci 2023; 24:13461. [PMID: 37686262 PMCID: PMC10488223 DOI: 10.3390/ijms241713461] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative pathology among progressive dementias, and it is characterized by the accumulation in the brain of extracellular aggregates of beta-amyloid proteins and neurofibrillary intracellular tangles consisting of τ-hyperphosphorylated proteins. Under normal conditions, beta-amyloid peptides exert important trophic and antioxidant roles, while their massive presence leads to a cascade of events culminating in the onset of AD. The fibrils of beta-amyloid proteins are formed by the process of fibrillogenesis that, starting from individual monomers of beta-amyloid, can generate polymers of this protein, constituting the hypothesis of the "amyloid cascade". To date, due to the lack of pharmacological treatment for AD without toxic side effects, chemical research is directed towards the realization of hybrid compounds that can act as an adjuvant in the treatment of this neurodegenerative pathology. The hybrid compounds used in this work include moieties of a hydroxytyrosol, a nitrohydroxytyrosol, a tyrosol, and a homovanillyl alcohol bound to the N-benzylpiperidine moiety of donepezil, the main drug used in AD. Previous experiments have shown different properties of these hybrids, including low toxicity and antioxidant and chelating activities. The purpose of this work was to test the effects of hybrid compounds mixed with Aβ1-40 to induce fibrillogenesis and mimic AD pathogenesis. This condition has been studied both in test tubes and by an in vitro model of neuronal differentiated human SH-SY5Y neuroblastoma cells. The results obtained from test tube experiments showed that some hybrids inhibit the activity of the enzymes AChE, BuChE, and BACE-1. Cell experiments suggested that hybrids could inhibit fibrillogenesis, negatively modulating caspase-3. They were also shown to exert antioxidant effects, and the acetylated hybrids were found to be more functional and efficient than nonacetylated forms.
Collapse
Affiliation(s)
- Jessica Maiuolo
- Department of Health Science, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Græcia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Paola Costanzo
- Department of Chemistry and Chemical Technologies, University of Calabria, Via P. Bucci, Cubo 12C, 87036 Rende, Italy
| | - Mariorosario Masullo
- Department of Movement Science and Well-Being, University “Parthenope” of Naples, Via Medina, 40, 80133 Napoli, Italy; (M.M.); (R.A.)
| | - Antonio D’Errico
- Department of Movement Science and Well-Being, University “Parthenope” of Naples, Via Medina, 40, 80133 Napoli, Italy; (M.M.); (R.A.)
| | - Rosarita Nasso
- Department of Movement Science and Well-Being, University “Parthenope” of Naples, Via Medina, 40, 80133 Napoli, Italy; (M.M.); (R.A.)
| | - Sonia Bonacci
- Department of Health Sciences, University Magna Græcia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy (M.O.)
| | - Vincenzo Mollace
- Department of Health Science, Institute of Research for Food Safety & Health (IRC-FSH), University Magna Græcia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Manuela Oliverio
- Department of Health Sciences, University Magna Græcia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy (M.O.)
| | - Rosaria Arcone
- Department of Movement Science and Well-Being, University “Parthenope” of Naples, Via Medina, 40, 80133 Napoli, Italy; (M.M.); (R.A.)
| |
Collapse
|
18
|
Du Z, Nam E, Lin Y, Hong M, Molnár T, Kondo I, Ishimori K, Baik MH, Lee YH, Lim MH. Unveiling the impact of oxidation-driven endogenous protein interactions on the dynamics of amyloid-β aggregation and toxicity. Chem Sci 2023; 14:5340-5349. [PMID: 37234895 PMCID: PMC10208028 DOI: 10.1039/d3sc00881a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Cytochrome c (Cyt c), a multifunctional protein with a crucial role in controlling cell fate, has been implicated in the amyloid pathology associated with Alzheimer's disease (AD); however, the interaction between Cyt c and amyloid-β (Aβ) with the consequent impact on the aggregation and toxicity of Aβ is not known. Here we report that Cyt c can directly bind to Aβ and alter the aggregation and toxicity profiles of Aβ in a manner that is dependent on the presence of a peroxide. When combined with hydrogen peroxide (H2O2), Cyt c redirects Aβ peptides into less toxic, off-pathway amorphous aggregates, whereas without H2O2, it promotes Aβ fibrillization. The mechanisms behind these effects may involve a combination of the complexation between Cyt c and Aβ, the oxidation of Aβ by Cyt c and H2O2, and the modification of Cyt c by H2O2. Our findings demonstrate a new function of Cyt c as a modulator against Aβ amyloidogenesis.
Collapse
Affiliation(s)
- Zhi Du
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
- Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology Taiyuan 030024 PR China
| | - Eunju Nam
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Yuxi Lin
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI) Ochang Chungbuk 28119 Republic of Korea
| | - Mannkyu Hong
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
- Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS) Daejeon 34141 Republic of Korea
| | - Tamás Molnár
- Department of Biochemistry, Institute of Biology, Eötvös Loránd University H-1117 Budapest Hungary
| | - Ikufumi Kondo
- Graduate School of Chemical Sciences and Engineering, Hokkaido University Kita 13, Nishi 8, Kita-ku Sapporo 060-8628 Japan
| | - Koichiro Ishimori
- Graduate School of Chemical Sciences and Engineering, Hokkaido University Kita 13, Nishi 8, Kita-ku Sapporo 060-8628 Japan
- Department of Chemistry, Faculty of Science, Hokkaido University Kita 10, Nishi 8, Kita-ku Sapporo 060-0810 Japan
| | - Mu-Hyun Baik
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
- Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS) Daejeon 34141 Republic of Korea
| | - Young-Ho Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI) Ochang Chungbuk 28119 Republic of Korea
- Bio-Analytical Science, University of Science and Technology (UST) Daejeon 34113 Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University Daejeon 34134 Republic of Korea
- Research Headquarters, Korea Brain Research Institute (KBRI) Daegu 41068 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
19
|
Fonseca-Santos B, Cazarin CA, da Silva PB, Dos Santos KP, da Rocha MCO, Báo SN, De-Souza MM, Chorilli M. Intranasal in situ gelling liquid crystal for delivery of resveratrol ameliorates memory and neuroinflammation in Alzheimer's disease. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023:102689. [PMID: 37156330 DOI: 10.1016/j.nano.2023.102689] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/03/2023] [Accepted: 04/26/2023] [Indexed: 05/10/2023]
Abstract
Alzheimer's disease (AD) is an illness that affects people aged 65 or older and affects around 6.5 million in the United States. Resveratrol is a chemical obtained from natural products and it exhibits biological activity based on inhibiting the formation, depolymerization of the amyloid, and decreasing neuroinflammation. Due to the insolubility of this compound; its incorporation in surfactant-based systems was proposed to design an intranasal formulation. A range of systems has been produced by mixing oleic acid, CETETH-20 and water. Polarised light microscopy (PLM), small angle x-ray scattering (SAXS) and transmission electron microscopy (TEM) confirm the initial liquid formulation (F) presented as microemulsion (ME). After dilution, the gelled systems were characterized as hexagonal mesophase and they showed feasibility proprieties. Pharmacological assays performed after intranasal administration showed the ability to improve learning and memory in animals, as well as remission of neuroinflammation via inhibition of interleukin.
Collapse
Affiliation(s)
- Bruno Fonseca-Santos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, São Paulo 14801-902, Brazil; Federal University of Bahia (UFBA), Health Sciences Institute, Department of Biotechnology, Salvador, Bahia 40170-115, Brazil.
| | - Camila André Cazarin
- University of Vale do Itajaí (UNIVALI), Postgraduate in Pharmaceutical Sciences, Itajaí, Santa Catarina 88302-901, Brazil
| | - Patrícia Bento da Silva
- Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia (UnB), Brasilia, Federal District 70910-900, Brazil
| | - Kaio Pini Dos Santos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, São Paulo 14801-902, Brazil
| | - Márcia Cristina Oliveira da Rocha
- Microscopy and Microanalysis Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia (UnB), Brasilia, Federal District 70910-900, Brazil
| | - Sônia Nair Báo
- Microscopy and Microanalysis Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia (UnB), Brasilia, Federal District 70910-900, Brazil
| | - Márcia Maria De-Souza
- University of Vale do Itajaí (UNIVALI), Postgraduate in Pharmaceutical Sciences, Itajaí, Santa Catarina 88302-901, Brazil
| | - Marlus Chorilli
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, São Paulo 14801-902, Brazil.
| |
Collapse
|
20
|
Ailioaie LM, Ailioaie C, Litscher G. Photobiomodulation in Alzheimer's Disease-A Complementary Method to State-of-the-Art Pharmaceutical Formulations and Nanomedicine? Pharmaceutics 2023; 15:916. [PMID: 36986776 PMCID: PMC10054386 DOI: 10.3390/pharmaceutics15030916] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Alzheimer's disease (AD), as a neurodegenerative disorder, usually develops slowly but gradually worsens. It accounts for approximately 70% of dementia cases worldwide, and is recognized by WHO as a public health priority. Being a multifactorial disease, the origins of AD are not satisfactorily understood. Despite huge medical expenditures and attempts to discover new pharmaceuticals or nanomedicines in recent years, there is no cure for AD and not many successful treatments are available. The current review supports introspection on the latest scientific results from the specialized literature regarding the molecular and cellular mechanisms of brain photobiomodulation, as a complementary method with implications in AD. State-of-the-art pharmaceutical formulations, development of new nanoscale materials, bionanoformulations in current applications and perspectives in AD are highlighted. Another goal of this review was to discover and to speed transition to completely new paradigms for the multi-target management of AD, to facilitate brain remodeling through new therapeutic models and high-tech medical applications with light or lasers in the integrative nanomedicine of the future. In conclusion, new insights from this interdisciplinary approach, including the latest results from photobiomodulation (PBM) applied in human clinical trials, combined with the latest nanoscale drug delivery systems to easily overcome protective brain barriers, could open new avenues to rejuvenate our central nervous system, the most fascinating and complex organ. Picosecond transcranial laser stimulation could be successfully used to cross the blood-brain barrier together with the latest nanotechnologies, nanomedicines and drug delivery systems in AD therapy. Original, smart and targeted multifunctional solutions and new nanodrugs may soon be developed to treat AD.
Collapse
Affiliation(s)
- Laura Marinela Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania
| | - Constantin Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania
| | - Gerhard Litscher
- President of ISLA (International Society for Medical Laser Applications), Research Unit of Biomedical Engineering in Anesthesia and Intensive Care Medicine, Research Unit for Complementary and Integrative Laser Medicine, Traditional Chinese Medicine (TCM) Research Center Graz, Department of Anesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 39, 8036 Graz, Austria
| |
Collapse
|
21
|
Suh JM, Kim M, Yoo J, Han J, Paulina C, Lim MH. Intercommunication between metal ions and amyloidogenic peptides or proteins in protein misfolding disorders. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
22
|
Oxidative Damages on the Alzheimer's Related-Aβ Peptide Alters Its Ability to Assemble. Antioxidants (Basel) 2023; 12:antiox12020472. [PMID: 36830030 PMCID: PMC9951946 DOI: 10.3390/antiox12020472] [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: 12/28/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
Oxidative stress that can lead to oxidation of the amyloid-β (Aβ) peptide is considered a key feature in Alzheimer's disease (AD), influencing the ability of Aβ to assemble into β-sheet rich fibrils that are commonly found in senile plaques of AD patients. The present study aims at investigating the fallouts of Aβ oxidation on the assembly properties of the Aβ peptide. To accomplish this, we performed kinetics and analysis on an oxidized Aβ (oxAβ) peptide, resulting from the attack of reactive oxygen species (ROS) that are formed by the biologically relevant Cu/Aβ/dioxygen/ascorbate system. oxAβ was still able to assemble but displayed ill-defined and small oligomeric assemblies compared to the long and thick β-sheet rich fibrils from the non-oxidized counterpart. In addition, oxAβ does affect the assembly of the parent Aβ peptide. In a mixture of the two peptides, oxAβ has a mainly kinetic effect on the assembly of the Aβ peptide and was able to slow down the formation of Aβ fibril in a wide pH range [6.0-7.4]. However, oxAβ does not change the quantity and morphology of the Aβ fibrils formed to a significant extent. In the presence of copper or zinc di-cations, oxAβ assembled into weakly-structured aggregates rather than short, untangled Cu-Aβ fibrils and long untangled Zn-Aβ fibrils. The delaying effect of oxAβ on metal altered Aβ assembly was also observed. Hence, our results obtained here bring new insights regarding the tight interconnection between (i) ROS production leading to Aβ oxidation and (ii) Aβ assembly, in particular via the modulation of the Aβ assembly by oxAβ. It is the first time that co-assembly of oxAβ and Aβ under various environmental conditions (pH, metal ions …) are reported.
Collapse
|
23
|
Yi Y, Lim MH. Current understanding of metal-dependent amyloid-β aggregation and toxicity. RSC Chem Biol 2023; 4:121-131. [PMID: 36794021 PMCID: PMC9906324 DOI: 10.1039/d2cb00208f] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/22/2022] [Indexed: 11/23/2022] Open
Abstract
The discovery of effective therapeutics targeting amyloid-β (Aβ) aggregates for Alzheimer's disease (AD) has been very challenging, which suggests its complicated etiology associated with multiple pathogenic elements. In AD-affected brains, highly concentrated metals, such as copper and zinc, are found in senile plaques mainly composed of Aβ aggregates. These metal ions are coordinated to Aβ and affect its aggregation and toxicity profiles. In this review, we illustrate the current view on molecular insights into the assembly of Aβ peptides in the absence and presence of metal ions as well as the effect of metal ions on their toxicity.
Collapse
Affiliation(s)
- Yelim Yi
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
24
|
Tang J, Li Y, Liu X, Yu G, Zheng F, Guo Z, Zhang Y, Shao W, Wu S, Li H. Cobalt induces neurodegenerative damages through impairing autophagic flux by activating hypoxia-inducible factor-1α triggered ROS overproduction. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 857:159432. [PMID: 36243078 DOI: 10.1016/j.scitotenv.2022.159432] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/25/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
Cobalt is an environmental toxicant, and excessive bodily exposure can damage the nervous system. Particularly, our previous study reported that low-dose cobalt (significantly less than the safety threshold) is still able to induce neurodegenerative changes. However, the underlying molecular mechanism is still insufficient revealed. Herein, we further investigate the molecular mechanism between cobalt-induced neurodegeneration and autophagy, as well as explore the interplay between hypoxia-inducible factor-1α (HIF-1α), reactive oxygen species (ROS), and autophagy in cobalt-exposed mice and human neuroglioma cells. We first reveal cobalt as an environmental toxicant to severely induce β amyloid (Aβ) deposition, tau hyperphosphorylation, and dysregulated autophagy in the hippocampus and cortex of mice. In particular, we further identify that cobalt-induced neurotoxicity is triggered by the impairment of autophagic flux in vitro experiments. Moreover, the mechanistic study reveals that cobalt exposure extremely activates HIF-1α expression to facilitate the overproduction of ROS. Then, elevated ROS can target the amino-threonine kinase (AKT)-mammalian target of rapamycin (mTOR)-Unc-51 like autophagy activating kinase 1 (ULK1) signaling pathway to participate in cobalt-induced impairment of autophagic flux. Subsequently, defected autophagy further exacerbates cobalt-induced neurotoxicity for its unable to eliminate the deposition of pathological protein. Therefore, our data provide scientific evidence for cobalt safety evaluation and risk assessment and propose a breakthrough for understanding the regulatory relationship between HIF-1α, ROS, and autophagy in cobalt-induced neurodegeneration.
Collapse
Affiliation(s)
- Jianping Tang
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yanjun Li
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Xu Liu
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Guangxia Yu
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Fuli Zheng
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Zhenkun Guo
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yating Zhang
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Wenya Shao
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Siying Wu
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| | - Huangyuan Li
- Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
25
|
Park S, Na C, Han J, Lim MH. Methods for analyzing the coordination and aggregation of metal-amyloid-β. METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2023; 15:6973210. [PMID: 36617236 DOI: 10.1093/mtomcs/mfac102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/28/2022] [Indexed: 01/09/2023]
Abstract
The misfolding and aggregation of amyloid-β (Aβ) peptides are histopathological features found in the brains of Alzheimer's disease (AD). To discover effective therapeutics for AD, numerous efforts have been made to control the aggregation of Aβ species and their interactions with other pathological factors, including metal ions. Metal ions, such as Cu(II) and Zn(II), can bind to Aβ peptides forming metal-bound Aβ (metal-Aβ) complexes and, subsequently, alter their aggregation pathways. In particular, redox-active metal ions bound to Aβ species can produce reactive oxygen species leading to oxidative stress. In this review, we briefly illustrate some experimental approaches for characterizing the coordination and aggregation properties of metal-Aβ complexes.
Collapse
Affiliation(s)
- Seongmin Park
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Chanju Na
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jiyeon Han
- Department of Applied Chemistry, University of Seoul, Seoul 02504, Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
26
|
Ramesh M, Govindaraju T. Multipronged diagnostic and therapeutic strategies for Alzheimer's disease. Chem Sci 2022; 13:13657-13689. [PMID: 36544728 PMCID: PMC9710308 DOI: 10.1039/d2sc03932j] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/13/2022] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and a major contributor to dementia cases worldwide. AD is clinically characterized by learning, memory, and cognitive deficits. The accumulation of extracellular amyloid β (Aβ) plaques and neurofibrillary tangles (NFTs) of tau are the pathological hallmarks of AD and are explored as targets for clinical diagnosis and therapy. AD pathology is poorly understood and there are no fully approved diagnosis and treatments. Notwithstanding the gap, decades of research in understanding disease mechanisms have revealed the multifactorial nature of AD. As a result, multipronged and holistic approaches are pertinent to targeting multiple biomarkers and targets for developing effective diagnosis and therapeutics. In this perspective, recent developments in Aβ and tau targeted diagnostic and therapeutic tools are discussed. Novel indirect, combination, and circulating biomarkers as potential diagnostic targets are highlighted. We underline the importance of multiplexing and multimodal detection of multiple biomarkers to generate biomarker fingerprints as a reliable diagnostic strategy. The classical therapeutics targeting Aβ and tau aggregation pathways are described with bottlenecks in the strategy. Drug discovery efforts targeting multifaceted toxicity involving protein aggregation, metal toxicity, oxidative stress, mitochondrial damage, and neuroinflammation are highlighted. Recent efforts focused on multipronged strategies to rationally design multifunctional modulators targeting multiple pathological factors are presented as future drug development strategies to discover potential therapeutics for AD.
Collapse
Affiliation(s)
- Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| |
Collapse
|
27
|
Hong M, Kim M, Yoon J, Lee SH, Baik MH, Lim MH. Excited-State Intramolecular Hydrogen Transfer of Compact Molecules Controls Amyloid Aggregation Profiles. JACS AU 2022; 2:2001-2012. [PMID: 36186552 PMCID: PMC9516708 DOI: 10.1021/jacsau.2c00281] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/11/2022] [Accepted: 07/27/2022] [Indexed: 06/16/2023]
Abstract
Developing chemical methodologies to directly modify harmful biomolecules affords the mitigation of their toxicity by persistent changes in their properties and structures. Here we report compact photosensitizers composed of the anthraquinone (AQ) backbone that undergo excited-state intramolecular hydrogen transfer, effectively oxidize amyloidogenic peptides, and, subsequently, alter their aggregation pathways. Density functional theory calculations showed that the appropriate position of the hydroxyl groups in the AQ backbone and the consequent intramolecular hydrogen transfer can facilitate the energy transfer to triplet oxygen. Biochemical and biophysical investigations confirmed that these photoactive chemical reagents can oxidatively vary both metal-free amyloid-β (Aβ) and metal-bound Aβ, thereby redirecting their on-pathway aggregation into off-pathway as well as disassembling their preformed aggregates. Moreover, the in vivo histochemical analysis of Aβ species produced upon photoactivation of the most promising candidate demonstrated that they do not aggregate into oligomeric or fibrillar aggregates in the brain. Overall, our combined computational and experimental studies validate a light-based approach for designing small molecules, with minimum structural complexity, as chemical reagents targeting and controlling amyloidogenic peptides associated with neurodegenerative disorders.
Collapse
Affiliation(s)
- Mannkyu Hong
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Mingeun Kim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jiwon Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Seung-Hee Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Mu-Hyun Baik
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
28
|
Nascimento LA, Nascimento ÉCM, Martins JBL. In silico study of tacrine and acetylcholine binding profile with human acetylcholinesterase: docking and electronic structure. J Mol Model 2022; 28:252. [PMID: 35947248 DOI: 10.1007/s00894-022-05252-2] [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: 03/14/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer disease (AD) is a neurodegenerative process, one of the most common and incident dementia in the population over 60 years. AD manifests the presence of complex biochemical processes involved in neuronal degeneration, such as the formation of senile plaques containing amyloid-β peptides, the development of intracellular neurofibrillary tangles, and the suppression of the acetylcholine neurotransmitter. In this way, we performed a set of theoretical tests of tacrine ligand and acetylcholine neurotransmitter against the human acetylcholinesterase enzyme. Molecular docking was used to understand the most important interactions of these molecules with the enzyme. Computational chemistry calculation was carried out using MP2, DFT, and semi-empirical methods, starting from molecular docking structures. We have also performed studies regarding the non-covalent interactions, electron localization function, molecular electrostatic potential and explicit water molecule influence. For Trp86 residue, we show two main interactions in accordance to the results of the literature for TcAChE. First, intermolecular interactions of the cation-π and sigma-π type were found. Second, close stacking interactions were stablished between THA+ and Trp86 residue on one side and with Tyr337 residue on the other side.
Collapse
Affiliation(s)
- Letícia A Nascimento
- Computational Chemistry Laboratory, Institute of Chemistry, University of Brasilia, Brasilia, DF, 70910-900, Brazil
| | - Érica C M Nascimento
- Computational Chemistry Laboratory, Institute of Chemistry, University of Brasilia, Brasilia, DF, 70910-900, Brazil
| | - João B L Martins
- Computational Chemistry Laboratory, Institute of Chemistry, University of Brasilia, Brasilia, DF, 70910-900, Brazil.
| |
Collapse
|
29
|
Han J, Yoon J, Shin J, Nam E, Qian T, Li Y, Park K, Lee SH, Lim MH. Conformational and functional changes of the native neuropeptide somatostatin occur in the presence of copper and amyloid-β. Nat Chem 2022; 14:1021-1030. [PMID: 35817963 DOI: 10.1038/s41557-022-00984-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 05/26/2022] [Indexed: 11/09/2022]
Abstract
The progression of neurodegenerative disorders can lead to impaired neurotransmission; however, the role of pathogenic factors associated with these diseases and their impact on the structures and functions of neurotransmitters have not been clearly established. Here we report the discovery that conformational and functional changes of a native neuropeptide, somatostatin (SST), occur in the presence of copper ions, metal-free amyloid-β (Aβ) and metal-bound Aβ (metal-Aβ) found as pathological factors in the brains of patients with Alzheimer's disease. These pathological elements induce the self-assembly of SST and, consequently, prevent it from binding to the receptor. In the reverse direction, SST notably modifies the aggregation profiles of Aβ species in the presence of metal ions, attenuating their cytotoxicity and interactions with cell membranes. Our work demonstrates a loss of normal function of SST as a neurotransmitter and a gain of its modulative function against metal-Aβ under pathological conditions.
Collapse
Affiliation(s)
- Jiyeon Han
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jiwon Yoon
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
| | - Jeongcheol Shin
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Eunju Nam
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Tongrui Qian
- State Key Laboratory Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Yulong Li
- State Key Laboratory Membrane Biology, Peking University School of Life Sciences, Beijing, China.,PKU-IDG/McGovern Institute for Brain Research, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Kiyoung Park
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| | - Seung-Hee Lee
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea.
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| |
Collapse
|
30
|
Advancements in the development of multi-target directed ligands for the treatment of Alzheimer's disease. Bioorg Med Chem 2022; 61:116742. [PMID: 35398739 DOI: 10.1016/j.bmc.2022.116742] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/01/2022] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial irreversible neurological disorder which results in cognitive impairment, loss of cholinergic neurons in synapses of the basal forebrain and neuronal death. Exact pathology of the disease is not yet known however, many hypotheses have been proposed for its treatment. The available treatments including monotherapies and combination therapies are not able to combat the disease effectively because of its complex pathological mechanism. A multipotent drug for AD has the potential to bind or inhibit multiple targets responsible for the progression of the disease like aggregated Aβ, hyperphosphorylated tau proteins, cholinergic and adrenergic receptors, MAO enzymes, overactivated N-methyl-d-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor etc. The traditional approach of one disease-one target-one drug has been rationalized to one drug-multi targets for the chronic diseases like AD and cancer. Thus, over the last decade research focus has been shifted towards the development of multi target directed ligands (MTDLs) which can simultaneously inhibit multiple targets and stop or slow the progression of the disease. The MTDLs can be more effective against AD and eliminate any possibility of drug-drug interactions. Many important active pharmacophore units have been fused, merged or incorporated into different scaffolds to synthesize new potent drugs. In the current article, we have described various hypothesis for AD and effectiveness of the MTDLs treatment strategy is discussed in detail. Different chemical scaffolds and their synthetic strategies have been described and important functionalities are identified in the chemical scaffold that have the potential to bind to the multiple targets. The important leads identified in this study with MTDL characteristics have the potential to be developed as drug candidates for the effective treatment of AD.
Collapse
|
31
|
Probing the Influence of Single-Site Mutations in the Central Cross-β Region of Amyloid β (1-40) Peptides. Biomolecules 2021; 11:biom11121848. [PMID: 34944492 PMCID: PMC8699037 DOI: 10.3390/biom11121848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022] Open
Abstract
Amyloid β (Aβ) is a peptide known to form amyloid fibrils in the brain of patients suffering from Alzheimer’s disease. A complete mechanistic understanding how Aβ peptides form neurotoxic assemblies and how they kill neurons has not yet been achieved. Previous analysis of various Aβ40 mutants could reveal the significant importance of the hydrophobic contact between the residues Phe19 and Leu34 for cell toxicity. For some mutations at Phe19, toxicity was completely abolished. In the current study, we assessed if perturbations introduced by mutations in the direct proximity of the Phe19/Leu34 contact would have similar relevance for the fibrillation kinetics, structure, dynamics and toxicity of the Aβ assemblies. To this end, we rationally modified positions Phe20 or Gly33. A small library of Aβ40 peptides with Phe20 mutated to Lys, Tyr or the non-proteinogenic cyclohexylalanine (Cha) or Gly33 mutated to Ala was synthesized. We used electron microscopy, circular dichroism, X-ray diffraction, solid-state NMR spectroscopy, ThT fluorescence and MTT cell toxicity assays to comprehensively investigate the physicochemical properties of the Aβ fibrils formed by the modified peptides as well as toxicity to a neuronal cell line. Single mutations of either Phe20 or Gly33 led to relatively drastic alterations in the Aβ fibrillation kinetics but left the global, as well as the local structure, of the fibrils largely unchanged. Furthermore, the introduced perturbations caused a severe decrease or loss of cell toxicity compared to wildtype Aβ40. We suggest that perturbations at position Phe20 and Gly33 affect the fibrillation pathway of Aβ40 and, thereby, influence the especially toxic oligomeric species manifesting so that the region around the Phe19/Leu34 hydrophobic contact provides a promising site for the design of small molecules interfering with the Aβ fibrillation pathway.
Collapse
|