1
|
Zhang Y, Liang Y, Gu Y. The dopaminergic system and Alzheimer's disease. Neural Regen Res 2025; 20:2495-2512. [PMID: 39314145 PMCID: PMC11801300 DOI: 10.4103/nrr.nrr-d-24-00230] [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: 02/25/2024] [Revised: 06/21/2024] [Accepted: 07/31/2024] [Indexed: 09/25/2024] Open
Abstract
Alzheimer's disease is a common neurodegenerative disorder in older adults. Despite its prevalence, its pathogenesis remains unclear. In addition to the most widely accepted causes, which include excessive amyloid-beta aggregation, tau hyperphosphorylation, and deficiency of the neurotransmitter acetylcholine, numerous studies have shown that the dopaminergic system is also closely associated with the occurrence and development of this condition. Dopamine is a crucial catecholaminergic neurotransmitter in the human body. Dopamine-associated treatments, such as drugs that target dopamine receptor D and dopamine analogs, can improve cognitive function and alleviate psychiatric symptoms as well as ameliorate other clinical manifestations. However, therapeutics targeting the dopaminergic system are associated with various adverse reactions, such as addiction and exacerbation of cognitive impairment. This review summarizes the role of the dopaminergic system in the pathology of Alzheimer's disease, focusing on currently available dopamine-based therapies for this disorder and the common side effects associated with dopamine-related drugs. The aim of this review is to provide insights into the potential connections between the dopaminergic system and Alzheimer's disease, thus helping to clarify the mechanisms underlying the condition and exploring more effective therapeutic options.
Collapse
Affiliation(s)
- Yuhan Zhang
- International Medical College, Chongqing Medical University, Chongqing, China
| | - Yuan Liang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yixue Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| |
Collapse
|
2
|
Bacchella C, Capucciati A, Monzani E. A Focus on the Link Between Metal Dyshomeostasis, Norepinephrine, and Protein Aggregation. Antioxidants (Basel) 2025; 14:347. [PMID: 40227404 PMCID: PMC11939683 DOI: 10.3390/antiox14030347] [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: 02/04/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Neurodegenerative disorders are one of the main public health problems worldwide and, for this reason, they have attracted the attention of several researchers who aim to better understand the molecular processes linked to the etiology of these disorders, including Alzheimer's and Parkinson's diseases. In this review, we describe both the beneficial and toxic effect of norepinephrine (NE) and its connected ROS/metal-mediated pathways, which end in neuromelanin (NM) formation and protein aggregation. In particular, we emphasize the importance of stabilizing the delicate homeostatic balance that regulates (i) the metal/ROS-promoted oxidation of catecholamines, as NE, and (ii) the generation of oxidative by-products capable of covalently and non-covalently modifying neuroproteins, thus altering their stability and their oligomerization; these processes may end in (iii) the incorporation of protein conjugates into vesicles, which then evolve into neuromelanin (NM) organelles. In general, we aim to provide an up-to-date overview of the challenges and controversies emerging from the current literature to delineate a direction for future research.
Collapse
Affiliation(s)
- Chiara Bacchella
- Dipartimento di Chimica, Università di Pavia, Via Taramelli 12, 27100 Pavia, Italy;
| | - Andrea Capucciati
- Dipartimento di Chimica, Università di Pavia, Via Taramelli 12, 27100 Pavia, Italy;
- Fondazione Grigioni per il Morbo di Parkinson, Via Gianfranco Zuretti 35, 20125 Milano, Italy
| | - Enrico Monzani
- Dipartimento di Chimica, Università di Pavia, Via Taramelli 12, 27100 Pavia, Italy;
| |
Collapse
|
3
|
Atanasova M. Small-Molecule Inhibitors of Amyloid Beta: Insights from Molecular Dynamics-Part A: Endogenous Compounds and Repurposed Drugs. Pharmaceuticals (Basel) 2025; 18:306. [PMID: 40143085 PMCID: PMC11944459 DOI: 10.3390/ph18030306] [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: 02/10/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
The amyloid hypothesis is the predominant model of Alzheimer's disease (AD) pathogenesis, suggesting that amyloid beta (Aβ) peptide is the primary driver of neurotoxicity and a cascade of pathological events in the central nervous system. Aβ aggregation into oligomers and deposits triggers various processes, such as vascular damage, inflammation-induced astrocyte and microglia activation, disrupted neuronal ionic homeostasis, oxidative stress, abnormal kinase and phosphatase activity, tau phosphorylation, neurofibrillary tangle formation, cognitive dysfunction, synaptic loss, cell death, and, ultimately, dementia. Molecular dynamics (MD) is a powerful structure-based drug design (SBDD) approach that aids in understanding the properties, functions, and mechanisms of action or inhibition of biomolecules. As the only method capable of simulating atomic-level internal motions, MD provides unique insights that cannot be obtained through other techniques. Integrating experimental data with MD simulations allows for a more comprehensive understanding of biological processes and molecular interactions. This review summarizes and evaluates MD studies from the past decade on small molecules, including endogenous compounds and repurposed drugs, that inhibit amyloid beta. Furthermore, it outlines key considerations for future MD simulations of amyloid inhibitors, offering a potential framework for studies aimed at elucidating the mechanisms of amyloid beta inhibition by small molecules.
Collapse
|
4
|
Niu C, Dong M, Niu Y. Natural polyphenol: Their pathogenesis-targeting therapeutic potential in Alzheimer's disease. Eur J Med Chem 2024; 269:116359. [PMID: 38537514 DOI: 10.1016/j.ejmech.2024.116359] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 03/17/2024] [Accepted: 03/24/2024] [Indexed: 04/07/2024]
Abstract
Alzheimer's disease (AD) is a detrimental neurodegenerative disease affecting the elderly. Clinically, it is characterized by progressive memory decline and subsequent loss of broader cognitive functions. Current drugs provide only symptomatic relief but do not have profound disease-modifying effects. There is an unmet need to identify novel pharmacological agents for AD therapy. Neuropathologically, the characteristic hallmarks of the disease are extracellular senile plaques containing amyloid β-peptides and intracellular neurofibrillary tangles containing hyperphosphorylated microtubule-associated protein tau. Simultaneously, oxidative stress, neuroinflammation and mitochondrial dysfunction in specific brain regions are early events during the process of AD pathologic changes and are associated with Aβ/tau toxicity. Here, we first summarized probable pathogenic mechanisms leading to neurodegeneration and hopefully identify pathways that serve as specific targets to improve therapy for AD. We then reviewed the mechanisms that underlie disease-modifying effects of natural polyphenols, with a focus on nuclear factor erythroid 2-related factor 2 activators for AD treatment. Lastly, we discussed challenges in the preclinical to clinical translation of natural polyphenols. In conclusion, there is evidence that natural polyphenols can be therapeutically useful in AD through their multifaceted mechanism of action. However, more clinical studies are needed to confirm these effects.
Collapse
Affiliation(s)
- Chengu Niu
- Internal Medicine Residency Program, Rochester General Hospital, Rochester, NY, 14621, USA
| | - Miaoxian Dong
- The Institute of Medicine, Qiqihar Medical University, Qiqihar, 161006, China
| | - Yingcai Niu
- The Institute of Medicine, Qiqihar Medical University, Qiqihar, 161006, China.
| |
Collapse
|
5
|
Wicks SL, Roberts JA, Hurtt MJ, Hernandez BP, Jones JJ, Taylor AL, Logan JK, Schreiber WJ, Murray MG, Crenshaw BL, Stevens CB, Lammi RK, Hanna JM. Synthesis of symmetrical and unsymmetrical tetrahydroxybiphenyls and their evaluation as amyloid-β aggregation inhibitors. LETT ORG CHEM 2024; 21:964-972. [PMID: 40191153 PMCID: PMC11970624 DOI: 10.2174/0115701786286700240322065602] [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: 12/05/2023] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 04/09/2025]
Abstract
Our group recently reported that the polyhydroxy aromatic compound 3,3',4,4'-biphenyltetrol (2a) is a successful inhibitor of amyloid-β peptide (Aβ) aggregation, decreasing Aβ aggregation by 50 % when present in equimolar concentrations. In the present study, several additional biphenyltetrols were prepared and examined for their in vitro activity against aggregation of Aβ, to investigate the effect of the relative positions of hydrogen-bond donors on the aggregation process. Congo red spectral shift assays have shown that, of the eight (8) additional biphenyltetrol compounds prepared, three (3) successfully inhibit association of Aβ monomers - two symmetrical isomers, 2,2',5,5'-biphenyltetrol (2c), and 2,2',3,3'-biphenyltetrol (2d), along with one unsymmetrical isomer, 2,3',4',5-biphenyltetrol (2g). These results, along with previously reported results of 2a, strongly suggest that hydroxyl group position affects the ability of the inhibitor to bind to Aβ assemblies, thus impacting inhibitory efficacy.
Collapse
Affiliation(s)
- Sarah L. Wicks
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Jake A. Roberts
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Matthew J. Hurtt
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Benjamin P. Hernandez
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Jason J. Jones
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Andrea L. Taylor
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Jessica K. Logan
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - William J. Schreiber
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Mouskudah G. Murray
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Brandy L. Crenshaw
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Craig B. Stevens
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Robin K. Lammi
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - James M. Hanna
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| |
Collapse
|
6
|
Gou X, Fu Y, Li J, Xiang J, Yang M, Zhang Y. Impact of nanoplastics on Alzheimer 's disease: Enhanced amyloid-β peptide aggregation and augmented neurotoxicity. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133518. [PMID: 38228001 DOI: 10.1016/j.jhazmat.2024.133518] [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: 10/17/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/18/2024]
Abstract
Nanoplastics, widely existing in the environment and organisms, have been proven to cross the blood-brain barrier, increasing the incidence of neurodegenerative diseases like Alzheimer's disease (AD). However, current studies mainly focus on the neurotoxicity of nanoplastics themselves, neglecting their synergistic effects with other biomolecules and the resulting neurotoxicity. Amyloid β peptide (Aβ), which triggers neurotoxicity through its self-aggregation, is the paramount pathogenic protein in AD. Here, employing polystyrene nanoparticles (PS) as a model for nanoplastics, we reveal that 100 pM PS nanoparticles significantly accelerate the nucleation rate of two Aβ subtypes (Aβ40 and Aβ42) at low concentrations, promoting the formation of more Aβ oligomers and leading to evident neurotoxicity. The hydrophobic surface of PS facilitates the interaction of hydrophobic fragments between Aβ monomers, responsible for the augmented neurotoxicity. This work provides consequential insights into the modulatory impact of low-dose PS on Aβ aggregation and the ensuing neurotoxicity, presenting a valuable foundation for future research on the intricate interplay between environmental toxins and brain diseases.
Collapse
Affiliation(s)
- Xiaoli Gou
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yongchun Fu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Juan Li
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Juan Xiang
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China.
| | - Minghui Yang
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China.
| | - Yi Zhang
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China.
| |
Collapse
|
7
|
Allnutt MA, Matera KM. Stabilization and Reduced Cytotoxicity of Amyloid Beta Aggregates in the Presence of Catechol Neurotransmitters. Neurochem Res 2024; 49:379-387. [PMID: 37847330 DOI: 10.1007/s11064-023-04036-1] [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: 07/21/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 10/18/2023]
Abstract
Oligomeric aggregates of the amyloid-beta (Aβ) peptide have been implicated as the toxic species for Alzheimer's disease by contributing to oxidative cytotoxicity and physical disruption in cell membranes in the brain. Recent evidence points to the ability of the catecholamine neurotransmitter dopamine in the presence of copper ions to both stabilize oligomers and decrease the toxic effects of these oligomers. Based on these results, physical characterization of aggregates and subsequent cell studies with a neuroblastoma line were performed that show both dopamine and the related neurotransmitter, norepinephrine, can stabilize oligomers and decrease toxicity of Aβ aggregates without copper present. To investigate this reduction of toxicity, structural characterization of oligomers in the presence of neurotransmitters was compared to aggregates formed with Aβ alone. Gel electrophoresis and transmission electron microscopy show higher levels of oligomers in the presence of dopamine and norepinephrine, yet the oligomer structure is largely amorphous. Aβ aggregated alone forms the predicted highly organized fibrillar species, with increased levels of dityrosine covalent linkages, which are largely absent in the presence of the neurotransmitters. A proposed mechanism for the observed decrease in cell death by Aβ in the presence of dopamine and norepinephrine suggests the neurotransmitters both block the formation of organized oligomer structures and dityrosine stabilizing linkages while also behaving as antioxidants, providing a dual mechanism for increased cell viability.
Collapse
|
8
|
Le NTK, Kang EJ, Park JH, Kang K. Catechol-Amyloid Interactions. Chembiochem 2023; 24:e202300628. [PMID: 37850717 DOI: 10.1002/cbic.202300628] [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: 09/13/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/19/2023]
Abstract
This review introduces multifaceted mutual interactions between molecules containing a catechol moiety and aggregation-prone proteins. The complex relationships between these two molecular species have previously been elucidated primarily in a unidirectional manner, as demonstrated in cases involving the development of catechol-based inhibitors for amyloid aggregation and the elucidation of the role of functional amyloid fibers in melanin biosynthesis. This review aims to consolidate scattered clues pertaining to catechol-based amyloid inhibitors, functional amyloid scaffold of melanin biosynthesis, and chemically designed peptide fibers for providing chemical insights into the role of the local three-dimensional orientation of functional groups in manifesting such interactions. These orientations may play crucial, yet undiscovered, roles in various supramolecular structures.
Collapse
Affiliation(s)
- Nghia T K Le
- Department of Applied Chemistry, Kyung Hee University, Yongin, Gyeonggi, 17104, South Korea
| | - Eun Joo Kang
- Department of Applied Chemistry, Kyung Hee University, Yongin, Gyeonggi, 17104, South Korea
| | - Ji Hun Park
- Department of Science Education, Ewha Womans University, Seoul, 03760, South Korea
| | - Kyungtae Kang
- Department of Applied Chemistry, Kyung Hee University, Yongin, Gyeonggi, 17104, South Korea
| |
Collapse
|
9
|
Krohn F, Lancini E, Ludwig M, Leiman M, Guruprasath G, Haag L, Panczyszyn J, Düzel E, Hämmerer D, Betts M. Noradrenergic neuromodulation in ageing and disease. Neurosci Biobehav Rev 2023; 152:105311. [PMID: 37437752 DOI: 10.1016/j.neubiorev.2023.105311] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
The locus coeruleus (LC) is a small brainstem structure located in the lower pons and is the main source of noradrenaline (NA) in the brain. Via its phasic and tonic firing, it modulates cognition and autonomic functions and is involved in the brain's immune response. The extent of degeneration to the LC in healthy ageing remains unclear, however, noradrenergic dysfunction may contribute to the pathogenesis of Alzheimer's (AD) and Parkinson's disease (PD). Despite their differences in progression at later disease stages, the early involvement of the LC may lead to comparable behavioural symptoms such as preclinical sleep problems and neuropsychiatric symptoms as a result of AD and PD pathology. In this review, we draw attention to the mechanisms that underlie LC degeneration in ageing, AD and PD. We aim to motivate future research to investigate how early degeneration of the noradrenergic system may play a pivotal role in the pathogenesis of AD and PD which may also be relevant to other neurodegenerative diseases.
Collapse
Affiliation(s)
- F Krohn
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - E Lancini
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany.
| | - M Ludwig
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - M Leiman
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - G Guruprasath
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - L Haag
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - J Panczyszyn
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - E Düzel
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neuroscience, University College London, London UK-WC1E 6BT, UK; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - D Hämmerer
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neuroscience, University College London, London UK-WC1E 6BT, UK; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany; Department of Psychology, University of Innsbruck, A-6020 Innsbruck, Austria
| | - M Betts
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| |
Collapse
|
10
|
Zheng Y, Zheng C, Tu W, Jiang Y, Lin H, Chen W, Lee Q, Zheng W. Danshensu inhibits Aβ aggregation and neurotoxicity as one of the main prominent features of Alzheimer's disease. Int J Biol Macromol 2023:125294. [PMID: 37315666 DOI: 10.1016/j.ijbiomac.2023.125294] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
It has been found that the main cause of neurodegenerative proteinopathies, especially Alzheimer's disease (AD) is the formation of Aβ amyloid plaques, which can be regulated by application of potential small molecules. In the present study, we aimed to investigate the inhibitory effect of danshensu on Aβ(1-42) aggregation and relevant apoptotic pathway in neurons. A broad range of spectroscopic, theoretical, and cellular assays were done to investigate the anti-amyloidogenic characteristics of danshensu. It was found that danshensu triggers its inhibitory effect against Aβ(1-42) aggregation through modulation of hydrophobic patches as well as structural and morphological changes through a stacking interaction. Furthermore, it was observed that incubation of Aβ(1-42) samples with danshensu during aggregation process recovered the cell viability and mitigated the expression of caspase-3 mRNA and protein as well caspase-3 activity deregulated by Aβ(1-42) amyloid fibrils alone. In general, obtained data showed that danshensu potentially inhibits Aβ(1-42) aggregation and associated proteinopathies through regulation of apoptotic pathway in a concentration-dependent manner. Therefore, danshensu may be used as a promising biomolecule against the Aβ aggregation and associated proteinopathies, which can be further analyzed in the future studies for the treatment of AD.
Collapse
Affiliation(s)
- Yuyin Zheng
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Integrative & Optimized Medicine Research center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Cheng Zheng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Wenzhan Tu
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Integrative & Optimized Medicine Research center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yiwei Jiang
- Alberta Institute, Wenzhou Medical University, Wenzhou 325000, China
| | - Haiyan Lin
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Integrative & Optimized Medicine Research center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Wangchao Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Qian Lee
- The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wu Zheng
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Integrative & Optimized Medicine Research center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
11
|
Dimitrova YN, Gutierrez JA, Huard K. It's ok to be outnumbered - sub-stoichiometric modulation of homomeric protein complexes. RSC Med Chem 2023; 14:22-46. [PMID: 36760737 PMCID: PMC9890894 DOI: 10.1039/d2md00212d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
An arsenal of molecular tools with increasingly diversified mechanisms of action is being developed by the scientific community to enable biological interrogation and pharmaceutical modulation of targets and pathways of ever increasing complexity. While most small molecules interact with the target of interest in a 1 : 1 relationship, a noteworthy number of recent examples were reported to bind in a sub-stoichiometric manner to a homomeric protein complex. This approach requires molecular understanding of the physiologically relevant protein assemblies and in-depth characterization of the compound's mechanism of action. The recent literature examples summarized here were selected to illustrate methods used to identify and characterize molecules with such mechanisms. The concept of one small molecule targeting a homomeric protein assembly is not new but the subject deserves renewed inspection in light of emerging technologies and increasingly diverse target biology, to ensure relevant in vitro systems are used and valuable compounds with potentially novel sub-stoichiometric mechanisms of action aren't overlooked.
Collapse
Affiliation(s)
| | | | - Kim Huard
- Genentech 1 DNA Way South San Francisco CA 94080 USA
| |
Collapse
|
12
|
Cirsiliol mitigates Aβ fibrillation and underlying membrane-leakage associated neurotoxicity: A possible implication in the treatment of neurodegenerative disease. Int J Biol Macromol 2022; 213:915-922. [PMID: 35688279 DOI: 10.1016/j.ijbiomac.2022.06.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 05/30/2022] [Accepted: 06/05/2022] [Indexed: 12/06/2022]
Abstract
Protein aggregating is known as a leading pathogenic characteristic of a wide range of neurodegenerative diseases (NDs). Preventing amyloid-β (Aβ) aggregation and uncovering the associated mechanism through the application of small bioactive compounds can be considered as a useful strategy in hampering the onset of ND. In this study, we analyzed the inhibitory effects of cirsiliol, a trihydroxy-dimethoxyflavone, against human Αβ42 fibrillization. Also, we explored the probable neurotoxicity of Αβ42 oligomers grown with cirsiliol at different molar ratios on PC-12 cells after 24 h. The results showed that significant changes in ThT and ANS fluorescence intensities, Congo red absorbance, and ellipticity changes were modulated by co-incubation of cirsiliol with Αβ42, in a concentration-dependent manner. The spectroscopy outcomes were also supported by imaging analysis, where a few Αβ42 fibrillar conformations were detected with cirsiliol. In addition, cellular assays demonstrated that co-incubated Αβ42 samples with cirsiliol regulated the cell mortality, LDH release, and caspase-3 activation relative to the PC-12 exposed to Aβ42 oligomers alone. In conclusion, it can suggest that cirsiliol can be used as a potential candidate in the development of small molecules-based drugs for the advancement of therapeutic platforms against ND.
Collapse
|
13
|
Wang Y, Hu T, Wei J, Yin X, Gao Z, Li H. Inhibitory activities of flavonoids from Scutellaria baicalensis Georgi on amyloid aggregation related to type 2 diabetes and the possible structural requirements for polyphenol in inhibiting the nucleation phase of hIAPP aggregation. Int J Biol Macromol 2022; 215:531-540. [PMID: 35724902 DOI: 10.1016/j.ijbiomac.2022.06.107] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/01/2022] [Accepted: 06/13/2022] [Indexed: 11/05/2022]
Abstract
Human islet amyloid polypeptide (hIAPP)-mediated cytotoxicity is identified as a potential target for developing new anti-diabetic molecules. Herein, we investigated the effect of the major bioactive compounds of Scutellaria baicalensis Georgi (S. baicalensis), including baicalein, baicalin, wogonin and oroxylin A, on hIAPP aggregation. We found that all of these compounds inhibited hIAPP fibril formation in a dose-dependent manner. But baicalein and baicalin, especially baicalein are more effective than wogonin and oroxylin A in stabilizing hIAPP monomers and eliminating toxic hIAPP assembly, suggesting that flavonoids with ortho-hydroxyl group on the A-ring exhibited higher anti-hIAPP nucleation potential than those without this structure. This stimulated our interest in further studying the possible structure-activity relationship between polyphenol and hIAPP aggregation inhibition. Our results demonstrated that flavonoids with ortho-hydroxyl group on the B-ring are also more effective against hIAPP nucleation than those without this structure. These results suggest that the ortho-hydroxybenzene structure is a key structural feature required for polyphenols to effectively inhibit hIAPP nucleation. This was further confirmed by the effect of polyphenoland phenols in inhibiting hIAPP nucleation. The conclusion that pyrogallol-type polyphenols are potential lead inhibitors may provide a valuable structural template for the further development of polyphenol-based inhibitor of amyloid peptides.
Collapse
Affiliation(s)
- Ying Wang
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China
| | - Ting Hu
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China
| | - Jingjing Wei
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China
| | - Xiaoying Yin
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China
| | - Zhonghong Gao
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China.
| | - Hailing Li
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China.
| |
Collapse
|
14
|
Zong B, Yu F, Zhang X, Zhao W, Sun P, Li S, Li L. Understanding How Physical Exercise Improves Alzheimer’s Disease: Cholinergic and Monoaminergic Systems. Front Aging Neurosci 2022; 14:869507. [PMID: 35663578 PMCID: PMC9158463 DOI: 10.3389/fnagi.2022.869507] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/14/2022] [Indexed: 01/11/2023] Open
Abstract
Alzheimer’s disease (AD) is an age-related neurodegenerative disorder, characterized by the accumulation of proteinaceous aggregates and neurofibrillary lesions composed of β-amyloid (Aβ) peptide and hyperphosphorylated microtubule-associated protein tau, respectively. It has long been known that dysregulation of cholinergic and monoaminergic (i.e., dopaminergic, serotoninergic, and noradrenergic) systems is involved in the pathogenesis of AD. Abnormalities in neuronal activity, neurotransmitter signaling input, and receptor function exaggerate Aβ deposition and tau hyperphosphorylation. Maintenance of normal neurotransmission is essential to halt AD progression. Most neurotransmitters and neurotransmitter-related drugs modulate the pathology of AD and improve cognitive function through G protein-coupled receptors (GPCRs). Exercise therapies provide an important alternative or adjunctive intervention for AD. Cumulative evidence indicates that exercise can prevent multiple pathological features found in AD and improve cognitive function through delaying the degeneration of cholinergic and monoaminergic neurons; increasing levels of acetylcholine, norepinephrine, serotonin, and dopamine; and modulating the activity of certain neurotransmitter-related GPCRs. Emerging insights into the mechanistic links among exercise, the neurotransmitter system, and AD highlight the potential of this intervention as a therapeutic approach for AD.
Collapse
Affiliation(s)
- Boyi Zong
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Fengzhi Yu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Xiaoyou Zhang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Wenrui Zhao
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Peng Sun
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Shichang Li
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
| | - Lin Li
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- College of Physical Education and Health, East China Normal University, Shanghai, China
- *Correspondence: Lin Li,
| |
Collapse
|
15
|
Hong SW, Teesdale-Spittle P, Page R, Ellenbroek B, Truman P. Biologically Active Compounds Present in Tobacco Smoke: Potential Interactions Between Smoking and Mental Health. Front Neurosci 2022; 16:885489. [PMID: 35557609 PMCID: PMC9087043 DOI: 10.3389/fnins.2022.885489] [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: 02/28/2022] [Accepted: 03/28/2022] [Indexed: 12/01/2022] Open
Abstract
Tobacco dependence remains one of the major preventable causes of premature morbidity and mortality worldwide. There are well over 8,000 compounds present in tobacco and tobacco smoke, but we do not know what effect, if any, many of them have on smokers. Major interest has been on nicotine, as well as on toxic and carcinogenic effects and several major and minor components of tobacco smoke responsible for the negative health effects of smoking have been elucidated. Smokers themselves report a variety of positive effects from smoking, including effects on depression, anxiety and mental acuity. Smoking has also been shown to have protective effects in Parkinson’s Disease. Are the subjective reports of a positive effect of smoking due to nicotine, of some other components of tobacco smoke, or are they a manifestation of the relief from nicotine withdrawal symptoms that smoking provides? This mini-review summarises what is currently known about the components of tobacco smoke with potential to have positive effects on smokers.
Collapse
Affiliation(s)
- Sa Weon Hong
- School of Health Sciences, Massey University, Wellington, New Zealand
| | - Paul Teesdale-Spittle
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Rachel Page
- School of Health Sciences, Massey University, Wellington, New Zealand
| | - Bart Ellenbroek
- Department of Psychology, Victoria University of Wellington, Wellington, New Zealand
| | - Penelope Truman
- School of Health Sciences, Massey University, Wellington, New Zealand
| |
Collapse
|
16
|
Administration of mucuna beans (Mucuna pruriences (L.) DC. var. utilis) improves cognition and neuropathology of 3 × Tg-AD mice. Sci Rep 2022; 12:996. [PMID: 35046433 PMCID: PMC8770455 DOI: 10.1038/s41598-022-04777-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/17/2021] [Indexed: 11/09/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by the accumulation of extracellular amyloid-beta peptides (Aβ) resulting in senile plaques and intracellular hyperphosphorylated tau protein resulting in neurofibrillary tangles (NFTs). Mucuna beans (Mucuna pruriences (L.) DC. var. utilis) are unique plants containing 3–9% L-3,4-dihydroxyphenylalanine (L-DOPA). Here we investigated the effect of the administration of Mucuna beans on AD prevention by feeding triple-transgenic mice (3 × Tg-AD mice) with a diet containing Mucuna beans for 13 months. The levels of Aβ oligomers and detergent-insoluble phosphorylated tau decreased in the brain of mice fed with Mucuna beans (Mucuna group) compared to those of the Control group. Aβ accumulation and phosphorylated tau accumulation in the brain in the Mucuna group were also reduced. In addition, administration of Mucuna beans improved cognitive function. These results suggest that administration of Mucuna beans may have a preventive effect on AD development in 3 × Tg-AD mice.
Collapse
|
17
|
Gao D, Wan J, Zou Y, Gong Y, Dong X, Xu Z, Tang J, Wei G, Zhang Q. Destructive Mechanism of Aβ 1-42 Protofibril by Norepinephrine revealed via Molecular Dynamics Simulations. Phys Chem Chem Phys 2022; 24:19827-19836. [DOI: 10.1039/d2cp01754g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Amyloid-β (Aβ) fibrillary plaques represent the main hallmarks of Alzheimer’s disease (AD), in addition to tau neurofibrillary tangles. Disrupting early-formed Aβ protofibril is considered as one of the primary therapeutic...
Collapse
|
18
|
Nie T, Cooper GJS. Mechanisms Underlying the Antidiabetic Activities of Polyphenolic Compounds: A Review. Front Pharmacol 2021; 12:798329. [PMID: 34970150 PMCID: PMC8712966 DOI: 10.3389/fphar.2021.798329] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/18/2021] [Indexed: 12/16/2022] Open
Abstract
Polyphenolic compounds are thought to show considerable promise for the treatment of various metabolic disorders, including type 2 diabetes mellitus (T2DM). This review addresses evidence from in vitro, in vivo, and clinical studies for the antidiabetic effects of certain polyphenolic compounds. We focus on the role of cytotoxic human amylin (hA) aggregates in the pathogenesis of T2DM, and how polyphenols can ameliorate this process by suppressing or modifying their formation. Small, soluble amylin oligomers elicit cytotoxicity in pancreatic islet β-cells and may thus cause β-cell disruption in T2DM. Amylin oligomers may also contribute to oxidative stress and inflammation that lead to the triggering of β-cell apoptosis. Polyphenols may exert antidiabetic effects via their ability to inhibit hA aggregation, and to modulate oxidative stress, inflammation, and other pathways that are β-cell-protective or insulin-sensitizing. There is evidence that their ability to inhibit and destabilize self-assembly by hA requires aromatic molecular structures that bind to misfolding monomers or oligomers, coupled with adjacent hydroxyl groups present on single phenyl rings. Thus, these multifunctional compounds have the potential to be effective against the pleiotropic mechanisms of T2DM. However, substantial further research will be required before it can be determined whether a polyphenol-based molecular entity can be used as a therapeutic for type 2 diabetes.
Collapse
Affiliation(s)
- Tina Nie
- School of Biological Sciences, Faculty of Science, the University of Auckland, Auckland, New Zealand
| | - Garth J. S. Cooper
- School of Biological Sciences, Faculty of Science, the University of Auckland, Auckland, New Zealand
- The Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, the University of Auckland, Auckland, New Zealand
- Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, Faculty of Biology Medicine & Health, School of Medical Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
19
|
Pathak BK, Dey S, Mozumder S, Sengupta J. The role of membranes in function and dysfunction of intrinsically disordered amyloidogenic proteins. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:397-434. [PMID: 35034725 DOI: 10.1016/bs.apcsb.2021.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Membrane-protein interactions play a major role in human physiology as well as in diseases pathology. Interaction of a protein with the membrane was previously thought to be dependent on well-defined three-dimensional structure of the protein. In recent decades, however, it has become evident that a large fraction of the proteome, particularly in eukaryotes, stays disordered in solution and these proteins are termed as intrinsically disordered proteins (IDPs). Also, a vast majority of human proteomes have been reported to contain substantially long disordered regions, called intrinsically disordered regions (IDRs), in addition to the structurally ordered regions. IDPs exist in an ensemble of conformations and the conformational flexibility enables IDPs to achieve functional diversity. IDPs (and IDRs) are found to be important players in cell signaling, where biological membranes act as anchors for signaling cascades. Therefore, IDPs modulate the membrane architectures, at the same time membrane composition also affects the binding of IDPs. Because of intrinsic disorders, misfolding of IDPs often leads to formation of oligomers, protofibrils and mature fibrils through progressive self-association. Accumulation of amyloid-like aggregates of some of the IDPs is a known causative agent for numerous diseases. In this chapter we highlight recent advances in understanding membrane interactions of some of the intrinsically disordered proteins involved in the pathogenesis of human diseases.
Collapse
Affiliation(s)
- Bani Kumar Pathak
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Sandip Dey
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Sukanya Mozumder
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Jayati Sengupta
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
20
|
Chen Y, Li X, Zhan C, Lao Z, Li F, Dong X, Wei G. A Comprehensive Insight into the Mechanisms of Dopamine in Disrupting Aβ Protofibrils and Inhibiting Aβ Aggregation. ACS Chem Neurosci 2021; 12:4007-4019. [PMID: 34472835 DOI: 10.1021/acschemneuro.1c00306] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Fibrillary aggregates of amyloid-β (Aβ) are the pathological hallmark of Alzheimer's disease (AD). Clearing Aβ deposition or inhibiting Aβ aggregation is a promising approach to treat AD. Experimental studies reported that dopamine (DA), an important neurotransmitter, can inhibit Aβ aggregation and disrupt Aβ fibrils in a dose-dependent manner. However, the underlying molecular mechanisms still remain mostly elusive. Herein, we investigated the effect of DA on Aβ42 protofibrils at three different DA-to-Aβ molar ratios (1:1, 2:1, and 10:1) using all-atom molecular dynamics simulations. Our simulations demonstrate that protonated DA at a DA-to-Aβ ratio of 2:1 exhibits stronger Aβ protofibril disruptive capacity than that at a molar-ratio of 1:1 by mostly disrupting the F4-L34-V36 hydrophobic core. When the ratio of DA-to-Aβ increases to 10:1, DA has a high probability to bind to the outer surface of protofibril and has negligible effect on the protofibril structure. Interestingly, at the same DA-to-Aβ ratio (10:1), a mixture of protonated (DA+) and deprotonated (DA0) DA molecules significantly disrupts Aβ protofibrils by the binding of DA0 to the F4-L34-V36 hydrophobic core. Replica-exchange molecular dynamics simulations of Aβ42 dimer show that DA+ inhibits the formation of β-sheets, K28-A42/K28-D23 salt-bridges, and interpeptide hydrophobic interactions and results in disordered coil-rich Aβ dimers, which would inhibit the subsequent fibrillization of Aβ. Further analyses reveal that DA disrupts Aβ protofibril and prevents Aβ dimerization mostly through π-π stacking interactions with residues F4, H6, and H13, hydrogen bonding interactions with negatively charged residues D7, E11, E22 and D23, and cation-π interactions with residues R5. This study provides a complete picture of the molecular mechanisms of DA in disrupting Aβ protofibril and inhibiting Aβ aggregation, which could be helpful for the design of potent drug candidates for the treatment/intervention of AD.
Collapse
Affiliation(s)
- Yujie Chen
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Sciences (Ministry of Education), Fudan University, Shanghai 200438, People’s Republic of China
| | - Xuhua Li
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, People’s Republic of China
| | - Chendi Zhan
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Sciences (Ministry of Education), Fudan University, Shanghai 200438, People’s Republic of China
| | - Zenghui Lao
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Sciences (Ministry of Education), Fudan University, Shanghai 200438, People’s Republic of China
| | - Fangying Li
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Sciences (Ministry of Education), Fudan University, Shanghai 200438, People’s Republic of China
| | - Xuewei Dong
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Sciences (Ministry of Education), Fudan University, Shanghai 200438, People’s Republic of China
| | - Guanghong Wei
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Sciences (Ministry of Education), Fudan University, Shanghai 200438, People’s Republic of China
| |
Collapse
|
21
|
Solomatina AI, Slobodina AD, Ryabova EV, Bolshakova OI, Chelushkin PS, Sarantseva SV, Tunik SP. Blood-Brain Barrier Penetrating Luminescent Conjugates Based on Cyclometalated Platinum(II) Complexes. Bioconjug Chem 2020; 31:2628-2637. [DOI: 10.1021/acs.bioconjchem.0c00542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
| | - Aleksandra D. Slobodina
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre ≪Kurchatov Institute≫, Gatchina 188300, Russia
| | - Elena V. Ryabova
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre ≪Kurchatov Institute≫, Gatchina 188300, Russia
| | - Olga I. Bolshakova
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre ≪Kurchatov Institute≫, Gatchina 188300, Russia
| | - Pavel S. Chelushkin
- Institute of Chemistry, St. Petersburg State University, St. Petersburg 198504, Russia
| | - Svetlana V. Sarantseva
- Petersburg Nuclear Physics Institute named by B.P. Konstantinov of National Research Centre ≪Kurchatov Institute≫, Gatchina 188300, Russia
| | - Sergey P. Tunik
- Institute of Chemistry, St. Petersburg State University, St. Petersburg 198504, Russia
| |
Collapse
|
22
|
Sharma M, Tiwari V, Shukla S, Panda JJ. Fluorescent Dopamine-Tryptophan Nanocomposites as Dual-Imaging and Antiaggregation Agents: New Generation of Amyloid Theranostics with Trimeric Effects. ACS APPLIED MATERIALS & INTERFACES 2020; 12:44180-44194. [PMID: 32870652 DOI: 10.1021/acsami.0c13223] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The aggregation of neurotoxic amyloid-β (Aβ) polypeptides into aberrant extracellular senile plaques is the major neuropathological hallmark of Alzheimer's disease (AD). Inhibiting aggregation of these peptides to control the progression of this deadly disease can serve as a viable therapeutic option. In the current work, inherently fluorescent theranostic dopamine-tryptophan nanocomposites (DTNPs) were developed and investigated for their amyloid inhibition propensity along with their ability to act as a cellular bioimaging agent in neuronal cells. The antiaggregation potency of the nanocomposites was further investigated against an in vitro established reductionist amyloid aggregation model consisting of a mere dipeptide, phenylalanine-phenylalanine (FF). As opposed to large peptide/protein-derived robust and high-molecular-weight amyloid aggregation models of Alzheimer's disease, our dipeptide-based amyloid model provides an edge over others in terms of the ease of handling, synthesis, and cost-effectiveness. Results demonstrated positive antiaggregation behavior of the DTNPs toward both FF-derived amyloid fibrils and preformed Aβ-peptide fibers by means of electron microscopic and circular dichroism-based studies. Our results further pointed toward the neuroprotective effects of the DTNPs in neuroblastoma cells against FF amyloid fibril-induced toxicity and also that they significantly suppressed the accumulation of Aβ42 oligomers in both cortex and hippocampus regions and improved cognitive impairment in an intracerebroventricular streptozotocin (ICV-STZ)-induced animal model of dementia. Besides, DTNPs also exhibited excellent fluorescent properties and light up the cytoplasm of neuroblastoma cells when being coincubated with cells, confirming their ability to serve as an intracellular bioimaging agent. Overall, these results signify the potency of the DTNPs as promising multifunctional theranostic agents for treating AD.
Collapse
Affiliation(s)
- Manju Sharma
- Institute of Nano Science and Technology, Mohali, Punjab 160062, India
| | - Virendra Tiwari
- CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Shubha Shukla
- CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031, India
| | - Jiban Jyoti Panda
- Institute of Nano Science and Technology, Mohali, Punjab 160062, India
| |
Collapse
|
23
|
Mezeiova E, Chalupova K, Nepovimova E, Gorecki L, Prchal L, Malinak D, Kuca K, Soukup O, Korabecny J. Donepezil Derivatives Targeting Amyloid-β Cascade in Alzheimer's Disease. Curr Alzheimer Res 2020; 16:772-800. [PMID: 30819078 DOI: 10.2174/1567205016666190228122956] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/04/2019] [Accepted: 01/31/2019] [Indexed: 11/22/2022]
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder with an increasing impact on society. Because currently available therapy has only a short-term effect, a huge number of novel compounds are developed every year exploiting knowledge of the various aspects of AD pathophysiology. To better address the pathological complexity of AD, one of the most extensively pursued strategies by medicinal chemists is based on Multi-target-directed Ligands (MTDLs). Donepezil is one of the currently approved drugs for AD therapy acting as an acetylcholinesterase inhibitor. In this review, we have made an extensive literature survey focusing on donepezil-derived MTDL hybrids primarily targeting on different levels cholinesterases and amyloid beta (Aβ) peptide. The targeting includes direct interaction of the compounds with Aβ, AChE-induced Aβ aggregation, inhibition of BACE-1 enzyme, and modulation of biometal balance thus impeding Aβ assembly.
Collapse
Affiliation(s)
- Eva Mezeiova
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic.,National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic.,Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Katarina Chalupova
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic.,National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Lukas Gorecki
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic.,Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Lukas Prchal
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - David Malinak
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic.,National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic.,Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Jan Korabecny
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic.,National Institute of Mental Health, Topolova 748, 250 67 Klecany, Czech Republic.,Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| |
Collapse
|
24
|
Notaro A, Frei A, Rubbiani R, Jakubaszek M, Basu U, Koch S, Mari C, Dotou M, Blacque O, Gouyon J, Bedioui F, Rotthowe N, Winter RF, Goud B, Ferrari S, Tharaud M, Řezáčová M, Humajová J, Tomšík P, Gasser G. Ruthenium(II) Complex Containing a Redox-Active Semiquinonate Ligand as a Potential Chemotherapeutic Agent: From Synthesis to In Vivo Studies. J Med Chem 2020; 63:5568-5584. [PMID: 32319768 DOI: 10.1021/acs.jmedchem.0c00431] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chemotherapy remains one of the dominant treatments to cure cancer. However, due to the many inherent drawbacks, there is a search for new chemotherapeutic drugs. Many classes of compounds have been investigated over the years to discover new targets and synergistic mechanisms of action including multicellular targets. In this work, we designed a new chemotherapeutic drug candidate against cancer, namely, [Ru(DIP)2(sq)](PF6) (Ru-sq) (DIP = 4,7-diphenyl-1,10-phenanthroline; sq = semiquinonate ligand). The aim was to combine the great potential expressed by Ru(II) polypyridyl complexes and the singular redox and biological properties associated with the catecholate moiety. Experimental evidence (e.g., X-ray crystallography, electron paramagnetic resonance, electrochemistry) demonstrates that the semiquinonate is the preferred oxidation state of the dioxo ligand in this complex. The biological activity of Ru-sq was then scrutinized in vitro and in vivo, and the results highlight the promising potential of this complex as a chemotherapeutic agent against cancer.
Collapse
Affiliation(s)
- Anna Notaro
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, F-75005 Paris, France
| | - Angelo Frei
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Riccardo Rubbiani
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Marta Jakubaszek
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, F-75005 Paris, France.,Institut Curie, PSL University, CNRS UMR 144, F-75005 Paris, France
| | - Uttara Basu
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, F-75005 Paris, France
| | - Severin Koch
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Cristina Mari
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Mazzarine Dotou
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, F-75005 Paris, France
| | - Olivier Blacque
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Jérémie Gouyon
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Team Synthèse, Electrochimie, Imagerie et Systèmes Analytiques pour le Diagnostic, F-75005 Paris, France
| | - Fethi Bedioui
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Team Synthèse, Electrochimie, Imagerie et Systèmes Analytiques pour le Diagnostic, F-75005 Paris, France
| | - Nils Rotthowe
- Department of Chemistry, University of Konstanz, Universitätsstrasse 10, D-78457 Konstanz, Germany
| | - Rainer F Winter
- Department of Chemistry, University of Konstanz, Universitätsstrasse 10, D-78457 Konstanz, Germany
| | - Bruno Goud
- Institut Curie, PSL University, CNRS UMR 144, F-75005 Paris, France
| | - Stefano Ferrari
- Institute of Molecular Cancer Research, University of Zurich, CH-8057 Zurich, Switzerland.,Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 143 00 Prague, Czech Republic
| | - Mickaël Tharaud
- Université de Paris, Institut de physique du Globe de Paris, CNRS, F-75005 Paris, France
| | - Martina Řezáčová
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Šimkova 870, 500 03 Hradec Kralove, Czech Republic
| | - Jana Humajová
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University in Prague, 150 06 Prague, Czech Republic
| | - Pavel Tomšík
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Šimkova 870, 500 03 Hradec Kralove, Czech Republic
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, F-75005 Paris, France
| |
Collapse
|
25
|
Shin JH, Le NTK, Jang H, Lee T, Kang K. Supramolecular Regulation of Polydopamine Formation by Amyloid Fibers. Chemistry 2020; 26:5500-5507. [PMID: 32092201 DOI: 10.1002/chem.202000437] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/19/2020] [Indexed: 11/10/2022]
Abstract
Polydopamine (PD) and melanin species are chemically complex systems, the formation and properties of which are incompletely understood. Inspired by the role of functional amyloids in melanin biosynthesis, this paper examines the influences of the supramolecular structure of amyloids on oxidative polymerization of dopamine. Kinetic analyses on the formation of PD species in the presence of hen egg white lysozyme (HEWL) fibers or soluble HEWL revealed that both forms gave rise to the total quantity of PD species, but the rate of their formation could be accelerated only by the amyloid form. PD species formed with HEWL fibers showed a morphology of bundled fibers, whereas those with soluble HEWL had a mesh-like structure. Amyloid fibers of recombinant Pmel17 had properties similar to those of HEWL fibers in modulating PD formation. The results presented here suggest how nature designs functionality with an amyloid structure and can help understand and engineer chemistries of other functional amyloids.
Collapse
Affiliation(s)
- J H Shin
- Department of Applied Chemistry, Kyung Hee University, 1732 Deogyoung-daero, Yongin, Gyeonggi, 17104, Republic of Korea
| | - Nghia T K Le
- Department of Applied Chemistry, Kyung Hee University, 1732 Deogyoung-daero, Yongin, Gyeonggi, 17104, Republic of Korea
| | - Hongje Jang
- Department of Chemistry, Kwangwoon University, 20 Gwangwoon-ro, Nowon-gu, Seoul, 01897, Republic of Korea
| | - Taehoon Lee
- Department of Applied Chemistry, Kyung Hee University, 1732 Deogyoung-daero, Yongin, Gyeonggi, 17104, Republic of Korea
| | - Kyungtae Kang
- Department of Applied Chemistry, Kyung Hee University, 1732 Deogyoung-daero, Yongin, Gyeonggi, 17104, Republic of Korea
| |
Collapse
|
26
|
Zou Y, Qian Z, Chen Y, Qian H, Wei G, Zhang Q. Norepinephrine Inhibits Alzheimer's Amyloid-β Peptide Aggregation and Destabilizes Amyloid-β Protofibrils: A Molecular Dynamics Simulation Study. ACS Chem Neurosci 2019; 10:1585-1594. [PMID: 30605312 DOI: 10.1021/acschemneuro.8b00537] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The abnormal self-assembly of amyloid-β (Aβ) peptides into toxic fibrillar aggregates is associated with the pathogenesis of Alzheimer's disease (AD). The inhibition of β-sheet-rich oligomer formation is considered as the primary therapeutic strategy for AD. Previous experimental studies reported that norepinephrine (NE), one of the neurotransmitters, is able to inhibit Aβ aggregation and disaggregate the preformed fibrils. Moreover, exercise can markedly increase the level of NE. However, the underlying inhibitory and disruptive mechanisms remain elusive. In this work, we performed extensive replica-exchange molecular dynamic (REMD) simulations to investigate the conformational ensemble of Aβ1-42 dimer with and without NE molecules. Our results show that without NE molecules, Aβ1-42 dimer transiently adopts a β-hairpin-containing structure, and the β-strand regions of this β-hairpin (residues 15QKLVFFA21 and 33GLMVGGVV40) strongly resemble those of the Aβ fibril structure (residues 15QKLVFFA21 and 30AIIGLMVG37) reported in an electron paramagnetic resonance spectroscopy study. NE molecules greatly reduce the interpeptide β-sheet content and suppress the formation of the above-mentioned β-hairpin, leading to a more disordered coil-rich Aβ dimer. Five dominant binding sites are identified, and the central hydrophobic core 16KLVFFA21 site and C-terminal 31IIGLMV36 hydrophobic site are the two most favorable ones. Our data reveal that hydrophobic, aromatic stacking, hydrogen-bonding and cation-π interactions synergistically contribute to the binding of NE molecules to Aβ peptides. MD simulations of Aβ1-42 protofibril show that NE molecules destabilize Aβ protofibril by forming H-bonds with residues D1, A2, D23, and A42. This work reveals the molecular mechanism by which NE molecules inhibit Aβ1-42 aggregation and disaggregate Aβ protofibrils, providing valuable information for developing new drug candidates and exercise therapy against AD.
Collapse
Affiliation(s)
- Yu Zou
- College of Physical Education and Training, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People’s Republic of China
| | - Zhenyu Qian
- Key Laboratory of Exercise and Health Sciences (Ministry of Education) and School of Kinesiology, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People’s Republic of China
| | - Yujie Chen
- State Key Laboratory of Surface Physics, Key Laboratory for Computational Physical Science (Ministry of Education), and Department of Physics, Fudan University, Shanghai 200433, People’s Republic of China
| | - Hongsheng Qian
- College of Physical Education and Training, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People’s Republic of China
| | - Guanghong Wei
- State Key Laboratory of Surface Physics, Key Laboratory for Computational Physical Science (Ministry of Education), and Department of Physics, Fudan University, Shanghai 200433, People’s Republic of China
| | - Qingwen Zhang
- College of Physical Education and Training, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People’s Republic of China
| |
Collapse
|
27
|
Snow AD, Castillo GM, Nguyen BP, Choi PY, Cummings JA, Cam J, Hu Q, Lake T, Pan W, Kastin AJ, Kirschner DA, Wood SG, Rockenstein E, Masliah E, Lorimer S, Tanzi RE, Larsen L. The Amazon rain forest plant Uncaria tomentosa (cat's claw) and its specific proanthocyanidin constituents are potent inhibitors and reducers of both brain plaques and tangles. Sci Rep 2019; 9:561. [PMID: 30728442 PMCID: PMC6365538 DOI: 10.1038/s41598-019-38645-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 01/04/2019] [Indexed: 01/15/2023] Open
Abstract
Brain aging and Alzheimer's disease both demonstrate the accumulation of beta-amyloid protein containing "plaques" and tau protein containing "tangles" that contribute to accelerated memory loss and cognitive decline. In the present investigation we identified a specific plant extract and its constituents as a potential alternative natural solution for preventing and reducing both brain "plaques and tangles". PTI-00703 cat's claw (Uncaria tomentosa from a specific Peruvian source), a specific and natural plant extract from the Amazon rain forest, was identified as a potent inhibitor and reducer of both beta-amyloid fibrils (the main component of "plaques") and tau protein paired helical filaments/fibrils (the main component of "tangles"). PTI-00703 cat's claw demonstrated both the ability to prevent formation/aggregation and disaggregate preformed Aβ fibrils (1-42 and 1-40) and tau protein tangles/filaments. The disaggregation/dissolution of Aβ fibrils occurred nearly instantly when PTI-00703 cat's claw and Aβ fibrils were mixed together as shown by a variety of methods including Thioflavin T fluorometry, Congo red staining, Thioflavin S fluorescence and electron microscopy. Sophisticated structural elucidation studies identified the major fractions and specific constituents within PTI-00703 cat's claw responsible for both the observed "plaque" and "tangle" inhibitory and reducing activity. Specific proanthocyanidins (i.e. epicatechin dimers and variants thereof) are newly identified polyphenolic components within Uncaria tomentosa that possess both "plaque and tangle" reducing and inhibitory activity. One major identified specific polyphenol within PTI-00703 cat's claw was epicatechin-4β-8-epicatechin (i.e. an epicatechin dimer known as proanthocyanidin B2) that markedly reduced brain plaque load and improved short-term memory in younger and older APP "plaque-producing" (TASD-41) transgenic mice (bearing London and Swedish mutations). Proanthocyanidin B2 was also a potent inhibitor of brain inflammation as shown by reduction in astrocytosis and gliosis in TASD-41 transgenic mice. Blood-brain-barrier studies in Sprague-Dawley rats and CD-1 mice indicated that the major components of PTI-00703 cat's claw crossed the blood-brain-barrier and entered the brain parenchyma within 2 minutes of being in the blood. The discovery of a natural plant extract from the Amazon rain forest plant (i.e. Uncaria tomentosa or cat's claw) as both a potent "plaque and tangle" inhibitor and disaggregator is postulated to represent a potential breakthrough for the natural treatment of both normal brain aging and Alzheimer's disease.
Collapse
Affiliation(s)
- Alan D Snow
- Cognitive Clarity Inc., Edmonds, WA, USA.
- ProteoTech Inc., Kirkland, WA, USA.
| | | | - Beth P Nguyen
- ProteoTech Inc., Kirkland, WA, USA
- Healthcare Legacy Consulting LLC, Dallas, TX, USA
| | | | - Joel A Cummings
- Cognitive Clarity Inc., Edmonds, WA, USA
- ProteoTech Inc., Kirkland, WA, USA
| | - Judy Cam
- ProteoTech Inc., Kirkland, WA, USA
- Preclinical GPS, Washington University, St. Louis, MO, USA
| | - Qubai Hu
- ProteoTech Inc., Kirkland, WA, USA
| | - Thomas Lake
- Cognitive Clarity Inc., Edmonds, WA, USA
- ProteoTech Inc., Kirkland, WA, USA
| | - Weihong Pan
- Blood-Brain Barrier Laboratory, Pennington Biomedical Research Center at Louisiana State University, Baton Rouge, Louisiana, USA
- Biopotentials Sleep Center, Baton Rouge, LA, USA
| | - Abba J Kastin
- Blood-Brain Barrier Laboratory, Pennington Biomedical Research Center at Louisiana State University, Baton Rouge, Louisiana, USA
| | | | - Steven G Wood
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Edward Rockenstein
- Departments of Neurosciences and Pathology, University of California- San Diego, La Jolla, CA, USA
| | - Eliezer Masliah
- Departments of Neurosciences and Pathology, University of California- San Diego, La Jolla, CA, USA
- Division of Neurosciences, National Institute on Aging, Bethesda, MD, USA
| | - Stephen Lorimer
- Department of Chemistry, University of Otago, Dunedin, New Zealand
- VicLink Ltd., Wellington, New Zealand
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Lesley Larsen
- Department of Chemistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
28
|
Nam E, Derrick JS, Lee S, Kang J, Han J, Lee SJC, Chung SW, Lim MH. Regulatory Activities of Dopamine and Its Derivatives toward Metal-Free and Metal-Induced Amyloid-β Aggregation, Oxidative Stress, and Inflammation in Alzheimer's Disease. ACS Chem Neurosci 2018; 9:2655-2666. [PMID: 29782798 DOI: 10.1021/acschemneuro.8b00122] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A catecholamine neurotransmitter, dopamine (DA), is suggested to be linked to the pathology of dementia; however, the involvement of DA and its structural analogues in the pathogenesis of Alzheimer's disease (AD), the most common form of dementia, composed of multiple pathogenic factors has not been clear. Herein, we report that DA and its rationally designed structural derivatives (1-6) based on DA's oxidative transformation are able to modulate multiple pathological elements found in AD [i.e., metal ions, metal-free amyloid-β (Aβ), metal-bound Aβ (metal-Aβ), and reactive oxygen species (ROS)], with demonstration of detailed molecular-level mechanisms. Our multidisciplinary studies validate that the protective effects of DA and its derivatives on Aβ aggregation and Aβ-mediated toxicity are induced by their oxidative transformation with concomitant ROS generation under aerobic conditions. In particular, DA and the derivatives (i.e., 3 and 4) show their noticeable anti-amyloidogenic ability toward metal-free Aβ and/or metal-Aβ, verified to occur via their oxidative transformation that facilitates Aβ oxidation. Moreover, in primary pan-microglial marker (CD11b)-positive cells, the major producers of inflammatory mediators in the brain, DA and its derivatives significantly diminish inflammation and oxidative stress triggered by lipopolysaccharides and Aβ through the reduced induction of inflammatory mediators as well as upregulated expression of heme oxygenase-1, the enzyme responsible for production of antioxidants. Collectively, we illuminate how DA and its derivatives could prevent multiple pathological features found in AD. The overall studies could advance our understanding regarding distinct roles of neurotransmitters in AD and identify key interactions for alleviation of AD pathology.
Collapse
Affiliation(s)
- Eunju Nam
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jeffrey S. Derrick
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seunghee Lee
- School of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Juhye Kang
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jiyeon Han
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Shin Jung C. Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Su Wol Chung
- School of Biological Sciences, College of Natural Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
29
|
Liu M, Wan L, Bin Y, Xiang J. Role of norepinephrine in Aβ-related neurotoxicity: dual interactions with Tyr10 and SNK(26-28) of Aβ. Acta Biochim Biophys Sin (Shanghai) 2017; 49:170-178. [PMID: 28069584 DOI: 10.1093/abbs/gmw126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/27/2016] [Indexed: 01/05/2023] Open
Abstract
With their capability to inhibit the formation of amyloid-β peptide (Aβ) fibril, norepinephrine (NE), and other catechol derivatives have been considered for the potential treatment of Alzheimer's disease (AD). Such treatment, however, remains debatable because of the diverse functions of Aβ and NE in AD pathology. Moreover, the complicated oxidation accompanying NE has caused the majority of the previous research to focus on the binding of NE oxides onto Aβ. The molecular mechanism by which Aβ interacts with the reduction state of NE, which is correlated with the brain function, should be urgently explored. In this work, by controlling rigorous anaerobic experimental conditions, the molecular mechanism of the Aβ/NE interaction was investigated, and two binding sites were revealed. Tyr10 was identified as the strong binding site of NE, and SNK(26-28) segment was the weak binding segment. Furthermore, thioflavin T fluorescence confirmed NE's positive function of inhibiting Aβ aggregation through its weak binding with SNK(26-28) segment. Meanwhile, 7-OHCCA fluorescence exhibited NE's negative function of enhancing ·OH generation through inhibiting the Aβ/Cu2+ coordination. The viability tests of the neuroblastoma SH-SY5Y cells displayed that the coexistence of NE, Cu2+, and Aβ induced lower cell viability than free Cu2+, indicating the significant negative effect of excessive NE on AD progression. These data revealed the possible pathway of NE-induced damage in AD brain, which is significant for understanding the function of NE in Aβ-involved AD neuropathology and for designing an NE-related therapeutic strategy for AD.
Collapse
Affiliation(s)
- Mengmeng Liu
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Liping Wan
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yannan Bin
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Juan Xiang
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
30
|
Liu M, Kou L, Bin Y, Wan L, Xiang J. Complicated function of dopamine in Aβ-related neurotoxicity: Dual interactions with Tyr10 and SNK(26–28) of Aβ. J Inorg Biochem 2016; 164:119-128. [DOI: 10.1016/j.jinorgbio.2016.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/31/2016] [Accepted: 09/13/2016] [Indexed: 12/29/2022]
|
31
|
Mahmoodabadi N, Ajloo D. QSAR, docking, and Molecular dynamic studies on the polyphenolic as inhibitors of β-amyloid aggregation. Med Chem Res 2016. [DOI: 10.1007/s00044-016-1620-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
32
|
Quinopeptide formation associated with the disruptive effect of epigallocatechin-gallate on lysozyme fibrils. Int J Biol Macromol 2015; 78:389-95. [DOI: 10.1016/j.ijbiomac.2015.04.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 04/02/2015] [Accepted: 04/20/2015] [Indexed: 02/05/2023]
|
33
|
Ogi H, Fukukshima M, Hamada H, Noi K, Hirao M, Yagi H, Goto Y. Ultrafast propagation of β-amyloid fibrils in oligomeric cloud. Sci Rep 2014; 4:6960. [PMID: 25376301 PMCID: PMC4223663 DOI: 10.1038/srep06960] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 10/22/2014] [Indexed: 11/13/2022] Open
Abstract
Interaction between monomer peptides and seeds is essential for clarifying the fibrillation mechanism of amyloid β (Aβ) peptides. We monitored the deposition reaction of Aβ1–40 peptides on immobilized seeds grown from Aβ1–42, which caused formation of oligomers in the early stage. The deposition reaction and fibrillation procedure were monitored throughout by novel total-internal-reflection-fluorescence microscopy with a quartz-crystal microbalance (TIRFM-QCM) system. This system allows simultaneous evaluation of the amount of deposited peptides on the surface seeds by QCM and fibril nucleation and elongation by TIRFM. Most fibrils reached other nuclei, forming the fibril network across the nucleus hubs in a short time. We found a fibril-elongation rate two-orders-of-magnitude higher in an oligomeric cloud than reported values, indicating ultrafast transition of oligomers into fibrils.
Collapse
Affiliation(s)
- Hirotsugu Ogi
- Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Masahiko Fukukshima
- Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Hiroki Hamada
- Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Kentaro Noi
- Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Masahiko Hirao
- Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Hisashi Yagi
- Institute for Protein Research, Osaka University, Yamadaoka 3-2, Suita, Osaka 565-0871, Japan
| | - Yuji Goto
- Institute for Protein Research, Osaka University, Yamadaoka 3-2, Suita, Osaka 565-0871, Japan
| |
Collapse
|
34
|
Takai E, Uda K, Yoshida T, Zako T, Maeda M, Shiraki K. Cysteine inhibits the fibrillisation and cytotoxicity of amyloid-β 40 and 42: implications for the contribution of the thiophilic interaction. Phys Chem Chem Phys 2014; 16:3566-72. [PMID: 24413447 DOI: 10.1039/c3cp54245a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Inhibitors of amyloid fibril formation have been at the centre of intense research efforts for the prevention of amyloidosis. Here, we hypothesise that a specific non-covalent interaction, the thiophilic interaction between the side chain of an aromatic residue in a polypeptide and a sulphur atom of the compound, effectively inhibits amyloid fibril formation. Fluorescence spectroscopy and transmission electron microscopy revealed that sulphur compounds, particularly Cys, inhibit the fibrillisation of amyloid-β 1-40 (Aβ40) and 1-42 (Aβ42). Interestingly, aggregates of Aβ40 and Aβ42 induced by Cys were less cytotoxic than those induced by catechin, which is the most typical inhibitor of amyloid fibril formation. Because the essential amino acid, Cys, is an abundant molecule in the blood and cytosol, our data provide a new basis for the prevention of amyloid-related diseases and the elucidation of the mechanism of these diseases.
Collapse
Affiliation(s)
- Eisuke Takai
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8573, Japan.
| | | | | | | | | | | |
Collapse
|
35
|
Tarozzi A, Bartolini M, Piazzi L, Valgimigli L, Amorati R, Bolondi C, Djemil A, Mancini F, Andrisano V, Rampa A. From the dual function lead AP2238 to AP2469, a multi-target-directed ligand for the treatment of Alzheimer's disease. Pharmacol Res Perspect 2014; 2:e00023. [PMID: 25505579 PMCID: PMC4184701 DOI: 10.1002/prp2.23] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/31/2013] [Accepted: 12/03/2013] [Indexed: 12/21/2022] Open
Abstract
The development of drugs with different pharmacological properties appears to be an innovative therapeutic approach for Alzheimer's disease. In this article, we describe a simple structural modification of AP2238, a first dual function lead, in particular the introduction of the catechol moiety performed in order to search for multi-target ligands. The new compound AP2469 retains anti-acetylcholinesterase (AChE) and beta-site amyloid precursor protein cleaving enzyme (BACE)1 activities compared to the reference, and is also able to inhibit Aβ42 self-aggregation, Aβ42 oligomer-binding to cell membrane and subsequently reactive oxygen species formation in both neuronal and microglial cells. The ability of AP2469 to interfere with Aβ42 oligomer-binding to neuron and microglial cell membrane gives this molecule both neuroprotective and anti-inflammatory properties. These findings, together with its strong chain-breaking antioxidant performance, make AP2469 a potential drug able to modify the course of the disease.
Collapse
Affiliation(s)
- Andrea Tarozzi
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna Corso d'Augusto 237, 47921, Rimini, Italy
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna Via Belmeloro 6, 40126, Bologna, Italy
| | - Lorna Piazzi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna Via Belmeloro 6, 40126, Bologna, Italy ; ICIQ - Institute of Chemical Research of Catalonia Avenida Països Catalans 16, 43007, Tarragona, Spain
| | - Luca Valgimigli
- Department of Chemistry "Giacomo Ciamician", Alma Mater Studiorum-University of Bologna Via S. Giacomo 11, 40126, Bologna, Italy
| | - Riccardo Amorati
- Department of Chemistry "Giacomo Ciamician", Alma Mater Studiorum-University of Bologna Via S. Giacomo 11, 40126, Bologna, Italy
| | - Cecilia Bolondi
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna Corso d'Augusto 237, 47921, Rimini, Italy
| | - Alice Djemil
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna Corso d'Augusto 237, 47921, Rimini, Italy
| | - Francesca Mancini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna Via Belmeloro 6, 40126, Bologna, Italy
| | - Vincenza Andrisano
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna Corso d'Augusto 237, 47921, Rimini, Italy
| | - Angela Rampa
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna Via Belmeloro 6, 40126, Bologna, Italy
| |
Collapse
|
36
|
Takai E, Uda K, Matsushita S, Shikiya Y, Yamada Y, Shiraki K, Zako T, Maeda M. Cysteine inhibits amyloid fibrillation of lysozyme and directs the formation of small worm-like aggregates through non-covalent interactions. Biotechnol Prog 2014; 30:470-8. [DOI: 10.1002/btpr.1866] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/30/2013] [Indexed: 12/27/2022]
Affiliation(s)
- Eisuke Takai
- Faculty of Pure and Applied Sciences; University of Tsukuba; Tsukuba Ibaraki 305-8573 Japan
| | - Ken Uda
- Faculty of Pure and Applied Sciences; University of Tsukuba; Tsukuba Ibaraki 305-8573 Japan
| | - Shuhei Matsushita
- Faculty of Pure and Applied Sciences; University of Tsukuba; Tsukuba Ibaraki 305-8573 Japan
| | - Yui Shikiya
- Faculty of Pure and Applied Sciences; University of Tsukuba; Tsukuba Ibaraki 305-8573 Japan
| | - Yoichi Yamada
- Faculty of Pure and Applied Sciences; University of Tsukuba; Tsukuba Ibaraki 305-8573 Japan
| | - Kentaro Shiraki
- Faculty of Pure and Applied Sciences; University of Tsukuba; Tsukuba Ibaraki 305-8573 Japan
| | - Tamotsu Zako
- Bioengineering Laboratory; RIKEN, 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Mizuo Maeda
- Bioengineering Laboratory; RIKEN, 2-1 Hirosawa Wako Saitama 351-0198 Japan
| |
Collapse
|
37
|
Sato M, Murakami K, Uno M, Nakagawa Y, Katayama S, Akagi KI, Masuda Y, Takegoshi K, Irie K. Site-specific inhibitory mechanism for amyloid β42 aggregation by catechol-type flavonoids targeting the Lys residues. J Biol Chem 2013; 288:23212-24. [PMID: 23792961 PMCID: PMC3743493 DOI: 10.1074/jbc.m113.464222] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 06/15/2013] [Indexed: 11/06/2022] Open
Abstract
The aggregation of the 42-residue amyloid β-protein (Aβ42) is involved in the pathogenesis of Alzheimer disease (AD). Numerous flavonoids exhibit inhibitory activity against Aβ42 aggregation, but their mechanism remains unclear in the molecular level. Here we propose the site-specific inhibitory mechanism of (+)-taxifolin, a catechol-type flavonoid, whose 3',4'-dihydroxyl groups of the B-ring plays a critical role. Addition of sodium periodate, an oxidant, strengthened suppression of Aβ42 aggregation by (+)-taxifolin, whereas no inhibition was observed under anaerobic conditions, suggesting the inhibition to be associated with the oxidation to form o-quinone. Because formation of the Aβ42-taxifolin adduct was suggested by mass spectrometry, Aβ42 mutants substituted at Arg(5), Lys(16), and/or Lys(28) with norleucine (Nle) were prepared to identify the residues involved in the conjugate formation. (+)-Taxifolin did not suppress the aggregation of Aβ42 mutants at Lys(16) and/or Lys(28) except for the mutant at Arg(5). In addition, the aggregation of Aβ42 was inhibited by other catechol-type flavonoids, whereas that of K16Nle-Aβ42 was not. In contrast, some non-catechol-type flavonoids suppressed the aggregation of K16Nle-Aβ42 as well as Aβ42. Furthermore, interaction of (+)-taxifolin with the β-sheet region in Aβ42 was not observed using solid-state NMR unlike curcumin of the non-catechol-type. These results demonstrate that catechol-type flavonoids could specifically suppress Aβ42 aggregation by targeting Lys residues. Although the anti-AD activity of flavonoids has been ascribed to their antioxidative activity, the mechanism that the o-quinone reacts with Lys residues of Aβ42 might be more intrinsic. The Lys residues could be targets for Alzheimer disease therapy.
Collapse
Affiliation(s)
- Mizuho Sato
- From the Division of Food Science and Biotechnology, Graduate School of Agriculture, and
| | - Kazuma Murakami
- From the Division of Food Science and Biotechnology, Graduate School of Agriculture, and
| | - Mayumi Uno
- From the Division of Food Science and Biotechnology, Graduate School of Agriculture, and
| | - Yu Nakagawa
- From the Division of Food Science and Biotechnology, Graduate School of Agriculture, and
- the Synthetic Cellular Chemistry Laboratory, RIKEN Advanced Science Institute, Saitama 351-0198
| | - Sumie Katayama
- the National Institute of Biomedical Innovation, Osaka 567-0085, and
| | - Ken-ichi Akagi
- the National Institute of Biomedical Innovation, Osaka 567-0085, and
| | - Yuichi Masuda
- the Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8502
- the Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Kiyonori Takegoshi
- the Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto 606-8502
| | - Kazuhiro Irie
- From the Division of Food Science and Biotechnology, Graduate School of Agriculture, and
| |
Collapse
|
38
|
Stefani M, Rigacci S. Protein folding and aggregation into amyloid: the interference by natural phenolic compounds. Int J Mol Sci 2013; 14:12411-57. [PMID: 23765219 PMCID: PMC3709793 DOI: 10.3390/ijms140612411] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 05/29/2013] [Accepted: 06/04/2013] [Indexed: 01/17/2023] Open
Abstract
Amyloid aggregation is a hallmark of several degenerative diseases affecting the brain or peripheral tissues, whose intermediates (oligomers, protofibrils) and final mature fibrils display different toxicity. Consequently, compounds counteracting amyloid aggregation have been investigated for their ability (i) to stabilize toxic amyloid precursors; (ii) to prevent the growth of toxic oligomers or speed that of fibrils; (iii) to inhibit fibril growth and deposition; (iv) to disassemble preformed fibrils; and (v) to favor amyloid clearance. Natural phenols, a wide panel of plant molecules, are one of the most actively investigated categories of potential amyloid inhibitors. They are considered responsible for the beneficial effects of several traditional diets being present in green tea, extra virgin olive oil, red wine, spices, berries and aromatic herbs. Accordingly, it has been proposed that some natural phenols could be exploited to prevent and to treat amyloid diseases, and recent studies have provided significant information on their ability to inhibit peptide/protein aggregation in various ways and to stimulate cell defenses, leading to identify shared or specific mechanisms. In the first part of this review, we will overview the significance and mechanisms of amyloid aggregation and aggregate toxicity; then, we will summarize the recent achievements on protection against amyloid diseases by many natural phenols.
Collapse
Affiliation(s)
- Massimo Stefani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, Florence 50134, Italy; E-Mail:
- Research Centre on the Molecular Basis of Neurodegeneration, Viale Morgagni 50, Florence 50134, Italy
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +39-55-275-8307; Fax: +39-55-275-8905
| | - Stefania Rigacci
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, Florence 50134, Italy; E-Mail:
| |
Collapse
|
39
|
Stevens CB, Hanna JM, Lammi RK. Synthesis of tetrahydroxybiphenyls and tetrahydroxyterphenyls and their evaluation as amyloid-β aggregation inhibitors. Bioorg Med Chem Lett 2013; 23:1703-6. [PMID: 23403086 PMCID: PMC3594554 DOI: 10.1016/j.bmcl.2013.01.076] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/04/2013] [Accepted: 01/16/2013] [Indexed: 10/27/2022]
Abstract
3,3',4,4'-Tetrahydroxybiphenyl and three isomeric 3,3″,4,4″-tetrahydroxyterphenyls with varying geometries around the central phenyl ring have been synthesized and evaluated for their in vitro activity against aggregation of Alzheimer's amyloid-β peptide (Aβ). Results from Congo red spectral-shift assays reveal that all four compounds successfully inhibit association of Aβ monomers. For the tetrahydroxyterphenyls, efficacy varies with linker geometry: the ortho-arrangement affords the most successful inhibition and the para-geometry the least, perhaps due to differing abilities of these compounds to bind Aβ. Of the four small molecules studied, 3,3',4,4'-tetrahydroxybiphenyl is the most effective inhibitor, reducing Aβ aggregation by 50% when present in stoichiometric concentrations.
Collapse
Affiliation(s)
- Craig B. Stevens
- Department of Chemistry, Physics, and Geology, Winthrop University, 101 Sims Science Building, Rock Hill, SC 29733
| | - James M. Hanna
- Department of Chemistry, Physics, and Geology, Winthrop University, 101 Sims Science Building, Rock Hill, SC 29733
| | - Robin K. Lammi
- Department of Chemistry, Physics, and Geology, Winthrop University, 101 Sims Science Building, Rock Hill, SC 29733
| |
Collapse
|
40
|
Interaction of selegiline-loaded PLGA-b-PEG nanoparticles with beta-amyloid fibrils. J Neural Transm (Vienna) 2013; 120:903-10. [DOI: 10.1007/s00702-013-0992-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Accepted: 02/05/2013] [Indexed: 11/26/2022]
|
41
|
|
42
|
Shimizu T, Yoshimune K, Komoriya T, Akiyama T, Ye X, Kohno H. Monoclonal antibodies against large oval aggregates of Aβ1-42. J Biosci Bioeng 2012; 115:216-20. [PMID: 23041139 DOI: 10.1016/j.jbiosc.2012.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 09/06/2012] [Accepted: 09/12/2012] [Indexed: 11/19/2022]
Abstract
Abnormal cerebral accumulation of amyloid beta protein(1-42) (Aβ(1-42)) is one of the hallmarks of Alzheimer's disease (AD). Aβ(1-42) aggregates exist in two distinct forms: fibrils that are composed of highly ordered β-sheets and amorphous aggregates that differ in size and toxicity. Here, we generated large oval aggregates (LOA) 369 ± 81 nm and 224 ± 92 nm in size on their major and minor axes, respectively, as measured by tapping-mode atomic force microscopy. LOA were produced by slow rotation of high concentrations (0.22 mM, 1.0 mg/mL) of Aβ(1-42) for 16 h at 37°C in the presence of 2.2 mM Aβ(16-20), which prevents the fibril formation, and purified with 0.22-μm filters. Analysis with thioflavin T showed that LOA have little β-sheet structure on their surfaces. Monoclonal antibodies that react with LOA, but not the fibril forms, were screened from 960 mouse hybridoma cell lines, and seven antibodies consisting of four IgG and three IgM antibodies were obtained. Four IgG monoclonal antibodies showed cross-reactivity of <10% against the monomer and fibril forms and amorphous aggregates that passed through 0.22-μm filters. Among the four antibodies, the antibody that was designated as 31-2 exhibited the highest reactivity against LOA and showed the lowest reactivity against the fibril forms. On the basis of these results, a unique epitope on the surface of LOA was suggested. The 31-2 antibody may be useful for future basic research and therapeutic applications for AD.
Collapse
Affiliation(s)
- Takenori Shimizu
- Department of Applied Molecular Chemistry, Graduate School of Industrial Technology, Nihon University, 1-2-1 Izumichou, Narashino, Chiba 275-8575, Japan
| | | | | | | | | | | |
Collapse
|
43
|
Shimanouchi T, Onishi R, Kitaura N, Umakoshi H, Kuboi R. Effect of copper (II) ion against elongation behavior of amyloid β fibrils on liposome membranes. CRYSTAL RESEARCH AND TECHNOLOGY 2011. [DOI: 10.1002/crat.201100344] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
44
|
Yang W, Wong Y, Ng OTW, Bai LP, Kwong DWJ, Ke Y, Jiang ZH, Li HW, Yung KKL, Wong MS. Inhibition of Beta-Amyloid Peptide Aggregation by Multifunctional Carbazole-Based Fluorophores. Angew Chem Int Ed Engl 2011; 51:1804-10. [DOI: 10.1002/anie.201104150] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 08/31/2011] [Indexed: 01/18/2023]
|
45
|
Yang W, Wong Y, Ng OTW, Bai LP, Kwong DWJ, Ke Y, Jiang ZH, Li HW, Yung KKL, Wong MS. Inhibition of Beta-Amyloid Peptide Aggregation by Multifunctional Carbazole-Based Fluorophores. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201104150] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
46
|
Shimanouchi T, Onishi R, Kitaura N, Umakoshi H, Kuboi R. Copper-mediated growth of amyloid β fibrils in the presence of oxidized and negatively charged liposomes. J Biosci Bioeng 2011; 112:611-5. [PMID: 21917513 DOI: 10.1016/j.jbiosc.2011.08.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 08/03/2011] [Accepted: 08/12/2011] [Indexed: 10/17/2022]
Abstract
Amyloid β protein (Aβ) from Alzheimer's disease formed fibrillar aggregates and their morphology depended on oxidized and negatively charged liposomes. The morphology of fibrillar aggregates was affected by Cu(2+), together with their growth kinetics. This is because Cu(2+) inhibited the nucleation step in the formation of amyloid Aβ fibrillar aggregates by forming Aβ/Cu complex inactive to the growth of fibrillar aggregates. In addition, this is probably because Cu(2+) affected the fibrillar aggregate formed on the surface of liposomes. These findings would give a better understanding of the formation mechanism of amyloid fibrils on biomembranes.
Collapse
Affiliation(s)
- Toshinori Shimanouchi
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1–3 Machikaneyama-cho, Toyonaka, Osaka 560–8531, Japan
| | | | | | | | | |
Collapse
|