1
|
Behar AE, Maayan G. A cocktail of Cu 2+- and Zn 2+-peptoid-based chelators can stop ROS formation for Alzheimer's disease therapy. Chem Sci 2024:d4sc04313h. [PMID: 39464602 PMCID: PMC11503657 DOI: 10.1039/d4sc04313h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
The formation of reactive oxygen species (ROS) in the brain is a major cause of neuropathologic degradation associated with Alzheimer's Disease (AD). It has been suggested that the copper (Cu)-amyloid-β (Aβ) peptide complex can lead to ROS formation in the brain. An external chelator for Cu that can extract Cu from the CuAβ complex should inhibit the formation of ROS, making Cu chelation an excellent therapeutic approach for AD. Such a chelator should possess high selectivity for Cu over zinc (Zn), which is also present within the synaptic cleft. However, such selectivity is generally hard to achieve in one molecule due to the similarities in the binding preferences of these two metal ions. As an alternative to monotherapy (where Cu extraction is performed using a single chelator), herein we describe a variation of combination therapy - a novel cocktail approach, which is based on the co-administration of two structurally different peptidomimetic chelators, aiming to target both Cu2+ and Zn2+ ions simultaneously but independently from each other. Based on rigorous spectroscopic experiments, we demonstrate that our peptidomimetic cocktail allows, for the first time, the complete and immediate inhibition of ROS production by the CuAβ complex in the presence of Zn2+. In addition, we further demonstrate the high stability of the cocktail under simulated physiological conditions and its resistance to proteolytic degradation by trypsin and report the water/n-octanol partition coefficient, initially assessing the blood-brain barrier (BBB) permeability potential of the chelators.
Collapse
Affiliation(s)
- Anastasia E Behar
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Technion City 3200008 Haifa Israel
| | - Galia Maayan
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Technion City 3200008 Haifa Israel
| |
Collapse
|
2
|
Shi W, Zhou Q, Lu L, Zhang Y, Zhang H, Pu Y, Yin L. Copper induced cytosolic escape of mitochondrial DNA and activation of cGAS-STING-NLRP3 pathway-dependent pyroptosis in C8-D1A cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117085. [PMID: 39321529 DOI: 10.1016/j.ecoenv.2024.117085] [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: 06/29/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Copper, a vital mineral nutrient, possesses redox qualities that make it both beneficial and toxic to organisms. Excessive environmental copper exposure can result in neurological damage and cognitive decline in humans. Astrocytes, the predominant glial cells in the brain, are particularly vulnerable to pollutants, but the mechanism of copper-induced damage to astrocytes remains elusive. The aim of this study was to determine the role of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway in initiating NLRP3 inflammasome-induced astrocyte pyroptosis and chronic inflammation under conditions of copper overload. Our findings indicated that copper exposure elevated mitochondrial ROS (mtROS) levels, resulting in mitochondrial damage in astrocytes. This damage caused the release of mitochondrial DNA (mtDNA) into the cytoplasm, which subsequently activated the cGAS-STING pathway. This activation resulted in interactions between STING and NLRP3 proteins, facilitating the assembly of the NLRP3 inflammasome and inducing pyroptosis. Furthermore, depletion of mtROS mitigated copper-induced mitochondrial damage in astrocytes and reduced mtDNA leakage. Pharmacological inhibition of STING or STING transfection further reversed copper-induced pyroptosis and the inflammatory response. In conclusion, this study demonstrated that the leakage of mtDNA into the cytoplasm and the subsequent activation of the cGAS-STING-NLRP3 pathway may be potential mechanisms underlying copper-induced pyroptosis in astrocytes. These findings provided new insights into the toxicity of copper.
Collapse
Affiliation(s)
- Wei Shi
- School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Qian Zhou
- School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Lu Lu
- School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Ying Zhang
- School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Hu Zhang
- School of Public Health, Yangzhou University, Yangzhou 225000, China.
| | - Yuepu Pu
- School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Lihong Yin
- School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
3
|
Mondal T, Kalita S, Dutta R, Mandal B. A smart adaptable metal sequestering peptide conjugate to modulate Aβ fibrillar aggregation. J Mater Chem B 2024. [PMID: 39382176 DOI: 10.1039/d4tb01093k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
The aggregation of amyloid β peptide (Aβ) in the presence of elevated levels of transition-metal ions, e.g., Fe3+, Cu2+, Zn2+, is accountable for enhanced cellular toxicity in Alzheimer's disease. Many strategies are reported to inhibit either Cu2+, Zn2+, or Fe3+-induced Aβ fibrillation, focused on one metal. Herein, a taurine-containing adaptable metal sequestering peptide (AMSP) has been developed as the modulator of any of the cited metal-induced Aβ-aggregation in vitro. We designed the peptide conjugate comprising VFFA as a recognition motif and a taurine moiety coupled with a pendant chain of glutamic acid such that the -SO3H groups of taurine lie nearby 13His and 14His of Aβ, and sequester such metal ions that construct the salt bridge preponderantly via13His-metal-14His composition as well as bridges with 6His of Aβ. We checked the modulation of fibrillar aggregates of Aβ in the presence of metal ions by monitoring the fibril accumulation using several biophysical methods. The results established that non-aggregating AMSP sequesters Zn2+ preferably, along with Fe3+ and Cu2+ ions from the metal-Aβ complex at the physiological condition, efficiently inhibiting Aβ aggregation. While such adaptable metal binders that can chelate various metals are new, AMSP inhibits aggregation through selective recognition and metal scavenging.
Collapse
Affiliation(s)
- Tanmay Mondal
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam-781039, India.
| | - Sujan Kalita
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam-781039, India.
- Department of Chemistry, Kamrup College Chamata, Nalbari 781306, India
| | - Rinku Dutta
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam-781039, India.
| | - Bhubaneswar Mandal
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam-781039, India.
| |
Collapse
|
4
|
Zhang J, Phetsanthad A, Li L. Investigating Anion Effects on Metal Ion Binding Interactions With Amyloid β Peptide by Ion Mobility Mass Spectrometry. JOURNAL OF MASS SPECTROMETRY : JMS 2024; 59:e5090. [PMID: 39328006 PMCID: PMC11446473 DOI: 10.1002/jms.5090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 09/28/2024]
Abstract
The study of metal ion's role in the biological processes of Alzheimer's disease has spurred investigations into the coordination chemistry of amyloid beta peptide and its fragments. Nano-electrospray ionization mass spectrometry (nESI-MS) has been utilized to examine the stabilization of bound anions on multiprotein complexes without bulk solvent. However, the effects of anions on metal ion binding interactions with amyloid beta peptide have not been explored. This study directly examined metal-peptide complexes using nESI-MS and investigated the effects of various anions on the binding ratio and stability of these complexes from ammonium salt solutions. The results indicate that different anions have distinct effects on the binding ratio and stability of various metal-peptide complexes. Of these, the bicarbonate ion exhibits the highest binding ratios for metal-peptide complexes, while binding ratios for these complexes in phosphate are comparatively low. Our results suggest that acetate, formate, bicarbonate, and phosphate have weak affinities and act as weak stabilizers of the metal-peptide complex structure in the gas phase. Intriguingly, chloride and sulfate act as stabilizers of the metal-peptide complex in the gas phase. The rank order determined from these data is substantially different from the Hofmeister salt series in solution. Although this outcome was anticipated due to the reduced influence of anions and water solvation, our findings correlate well with expected anion binding in solution and emphasize the importance of both hydration layer and anion-metal-peptide binding effects for Hofmeister-type stabilization in solution. This approach proved useful in examining the interactions between metal ions and amyloid beta peptide, which are relevant to Alzheimer's disease, using direct ESI-MS.
Collapse
Affiliation(s)
- Jingwei Zhang
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin 53705, United States
| | - Ashley Phetsanthad
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin 53705, United States
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin 53705, United States
- School of Pharmacy, University of Wisconsin–Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
5
|
Carrazoni GS, Garces NB, Cadore CR, Sosa PM, Cattaneo R, Mello-Carpes PB. Supplementation with Manihot esculenta Crantz (Cassava) leaves' extract prevents recognition memory deficits and hippocampal antioxidant dysfunction induced by Amyloid-β. Nutr Neurosci 2024; 27:942-950. [PMID: 37948133 DOI: 10.1080/1028415x.2023.2280815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
OBJECTIVE The Manihot esculenta Crantz (Cassava) is a typical South American plant rich in nutrients and energetic compounds. Lately, our group has shown that non-pharmacological interventions with natural antioxidants present different neuroprotective effects on oxidative balance and memory deficits in AD-like animal models. Here, our objective was to evaluate the neuroprotective effects of Cassava leaves' extract (CAS) in an AD-like model induced by amyloid-beta (Aβ) 25-35 peptide. METHODS Male Wistar rats (n = 40; 60 days old) were subjected to 10 days of CAS supplementation; then, we injected 2 μL Aβ 25-35 in the hippocampus by stereotaxic surgery. Ten days later, we evaluated object recognition (OR) memory. Cassavas' total polyphenols, flavonoids, and condensed tannins content were measured, as well as hippocampal lipid peroxidation and total antioxidant capacity. RESULTS CAS protected against Aβ-induced OR memory deficits. In addition, Aβ promoted antioxidant capacity decrease, while CAS was able to prevent it, in addition to diminishing lipoperoxidation compared to Aβ. DISCUSSION We show that treatment with Cassava leaves' extract before AD induction prevents recognition memory deficits related to Aβ hippocampal injection. At least part of these effects can be related to the Cassava leaves' extract supplementation effects on diminishing lipid peroxidation and preventing a decrease in the hippocampal total antioxidant capacity in the hippocampus of AD-like animals without adverse effects. Once cassavais a plant of warm and dry ground that can adapt to growon various soil types and seems to resist several insects, our results enable Cassava to be considered asa potential preventive intervention to avoid or minimizeAD-induced memory deficits worldwide.
Collapse
Affiliation(s)
- Guilherme Salgado Carrazoni
- Physiology Research Group, Stress, Memory and Behavior Lab, Universidade Federal do Pampa, Uruguaiana, Brazil
| | | | - Caroline Ramires Cadore
- Physiology Research Group, Stress, Memory and Behavior Lab, Universidade Federal do Pampa, Uruguaiana, Brazil
| | - Priscila Marques Sosa
- Physiology Research Group, Stress, Memory and Behavior Lab, Universidade Federal do Pampa, Uruguaiana, Brazil
| | | | - Pâmela Billig Mello-Carpes
- Physiology Research Group, Stress, Memory and Behavior Lab, Universidade Federal do Pampa, Uruguaiana, Brazil
| |
Collapse
|
6
|
Shastri D, Raj V, Lee S. Revolutionizing Alzheimer's treatment: Harnessing human serum albumin for targeted drug delivery and therapy advancements. Ageing Res Rev 2024; 99:102379. [PMID: 38901740 DOI: 10.1016/j.arr.2024.102379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder initiated by amyloid-beta (Aβ) accumulation, leading to impaired cognitive function. Several delivery approaches have been improved for AD management. Among them, human serum albumin (HSA) is broadly employed for drug delivery and targeting the Aβ in AD owing to its biocompatibility, Aβ inhibitory effect, and nanoform, which showed blood-brain barrier (BBB) crossing ability via glycoprotein 60 (gp60) receptor and secreted protein acidic and rich in cysteine (SPARC) protein to transfer the drug molecules in the brain. Thus far, there is no previous review focusing on HSA and its drug delivery system in AD. Hence, the reviewed article aimed to critically compile the HSA therapeutic as well as drug delivery role in AD management. It also delivers information on how HSA-incorporated nanoparticles with surfaced embedded ligands such as TAT, GM1, and so on, not only improve BBB permeability but also increase neuron cell targetability in AD brain. Additionally, Aβ and tau pathology, including various metabolic markers likely BACE1 and BACE2, etc., are discussed. Besides, the molecular interaction of HSA with Aβ and its distinctive forms are critically reviewed that HSA can segregate Zn(II) and Cu(II) metal ions from Aβ owing to high affinity. Furthermore, the BBB drug delivery challenges in AD are addressed. Finally, the clinical formulation of HSA for the management of AD is critically discussed on how the HSA inhibits Aβ oligomer and fibril, while glycated HSA participates in amyloid plaque formation, i.e., β-structure sheet formation. This review report provides theoretical background on HSA-based AD drug delivery and makes suggestions for future prospect-related work.
Collapse
Affiliation(s)
- Divya Shastri
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, the Republic of Korea; College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, the Republic of Korea
| | - Vinit Raj
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, the Republic of Korea.
| | - Sangkil Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, the Republic of Korea.
| |
Collapse
|
7
|
Puello-Silva J, Alí-Torres J. Computational 3D Models of Fe 2+/3+-Aβ 1-42 Complexes Associated with Alzheimer's Disease. J Phys Chem B 2024; 128:7022-7032. [PMID: 39016210 DOI: 10.1021/acs.jpcb.4c01173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
The interaction between iron and amyloid-beta (Aβ) peptides has received significant attention in Alzheimer's disease (AD) research due to its potential implications in developing this pathology. However, the coordination preferences of iron and Aβ1-42 have not been thoroughly investigated or remain unknown. This study employs a computational protocol that combines homology modeling techniques with quantum mechanics (DTF-xTB) calculations to build and evaluate several 3D models of Fe2+/3+-Aβ1-42. Our results reveal well-defined complexes for both the metal and peptide moieties, and we discuss the molecular interactions stabilizing these complexes by elucidating the coordinating environments and binding preferences. These proposed models offer valuable insights into the role of iron in Alzheimer's disease (AD) pathology.
Collapse
Affiliation(s)
- Jorge Puello-Silva
- Departamento de Química, Universidad Nacional de Colombia - Sede Bogotá, Bogotá 110111, Colombia
| | - Jorge Alí-Torres
- Departamento de Química, Universidad Nacional de Colombia - Sede Bogotá, Bogotá 110111, Colombia
| |
Collapse
|
8
|
De Santis E, Alleva S, Minicozzi V, Morante S, Stellato F. Probing the Dynamic Landscape: From Static to Time-Resolved X-Ray Absorption Spectroscopy to Investigate Copper Redox Chemistry in Neurodegenerative Disorders. Chempluschem 2024; 89:e202300712. [PMID: 38526934 DOI: 10.1002/cplu.202300712] [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: 12/01/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 03/27/2024]
Abstract
Copper (Cu), with its ability to exist in various oxidation states, notably Cu(I) and Cu(II), plays a crucial role in diverse biological redox reactions. This includes its involvement in pathways associated with oxidative stress in neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and Transmissible Spongiform Encephalopathies. This paper offers an overview of X-ray Absorption Spectroscopy (XAS) studies designed to elucidate the interactions between Cu ions and proteins or peptides associated with these neurodegenerative diseases. The emphasis lies on XAS specificity, revealing the local coordination environment, and on its sensitivity to Cu oxidation states. Furthermore, the paper focuses on XAS applications targeting the characterization of intermediate reaction states and explores the opportunities arising from recent advancements in time-resolved XAS at ultrabright synchrotron and Free Electron Laser radiation sources.
Collapse
Affiliation(s)
- Emiliano De Santis
- Department of Chemistry-BMC, Uppsala University, Box 576, SE-751 23, Uppsala, Sweden
| | - Stefania Alleva
- Department of Physics, University of Rome, Tor Vergata, Rome, 00133, Italy
- INFN, Rome, Tor Vergata, Rome, 00133, Italy
| | - Velia Minicozzi
- Department of Physics, University of Rome, Tor Vergata, Rome, 00133, Italy
- INFN, Rome, Tor Vergata, Rome, 00133, Italy
| | - Silvia Morante
- Department of Physics, University of Rome, Tor Vergata, Rome, 00133, Italy
- INFN, Rome, Tor Vergata, Rome, 00133, Italy
| | - Francesco Stellato
- Department of Physics, University of Rome, Tor Vergata, Rome, 00133, Italy
- INFN, Rome, Tor Vergata, Rome, 00133, Italy
| |
Collapse
|
9
|
Tan RX, Li WH, Pang JM, Zhong SM, Huang XY, Deng JZ, Zhou LY, Wu JQ, Wang XQ. Design, synthesis, and evaluation of 2,2'-bipyridyl derivatives as bifunctional agents against Alzheimer's disease. Mol Divers 2024; 28:1225-1238. [PMID: 37119457 DOI: 10.1007/s11030-023-10651-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/14/2023] [Indexed: 05/01/2023]
Abstract
Alzheimer's disease (AD) is a complex multifactorial neurodegenerative disease. Metal ion dyshomeostasis and Aβ aggregation have been proposed to contribute to AD progression. Metal ions can bind to Aβ and promote Aβ aggregation, and ultimately lead to neuronal death. Bifunctional (metal chelation and Aβ interaction) compounds are showing promise against AD. In this work, eleven new 3,3'-diamino-2,2'-bipyridine derivatives 4a-4k were synthesized, and evaluated as bifunctional agents for AD treatment. In vitro Aβ aggregation inhibition assay confirmed that most of the synthesized compounds exhibited significant self-induced Aβ1-42 aggregation inhibition. Among them, compound 4d displayed the best inhibitory potency of self-induced Aβ1-42 aggregation with IC50 value of 9.4 µM, and it could selectively chelate with Cu2+ and exhibited 66.2% inhibition of Cu2+-induced Aβ1-42 aggregation. Meanwhile, compound 4d showed strong neuroprotective activity against Aβ1-42 and Cu2+-treated Aβ1-42 induced cell damage. Moreover, compound 4d in high dose significantly reversed Aβ-induced memory impairment in mice.
Collapse
Affiliation(s)
- Ren-Xian Tan
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Wei-Hao Li
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Jia-Min Pang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Si-Min Zhong
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Xin-Yi Huang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Jun-Ze Deng
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Lu-Yi Zhou
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Jia-Qiang Wu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, China
| | - Xiao-Qin Wang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| |
Collapse
|
10
|
Miyamoto E, Hayashi H, Murayama S, Yanagisawa K, Sato T, Matsubara T. Prevention of amyloid β fibril deposition on the synaptic membrane in the precuneus by ganglioside nanocluster-targeting inhibitors. RSC Chem Biol 2024; 5:459-466. [PMID: 38725912 PMCID: PMC11078214 DOI: 10.1039/d4cb00038b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/16/2024] [Indexed: 05/12/2024] Open
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative condition, is one of the most common causes of dementia. Senile plaques, a hallmark of AD, are formed by the accumulation of amyloid β protein (Aβ), which starts to aggregate before the onset of the disease. Gangliosides, sialic acid-containing glycosphingolipids, play a key role in the formation of toxic Aβ aggregates. In membrane rafts, ganglioside-bound complexes (GAβ) act as nuclei for Aβ assembly, suggesting that GAβ is a promising target for AD therapy. The formation of GAβ-induced Aβ assemblies has been evaluated using reconstituted planar lipid membranes composed of synaptosomal plasma membrane (SPM) lipids extracted from human and mouse brains. Although the effects of gangliosides on Aβ accumulation in the precuneus have been established, effects on Aβ fibrils have not been determined. In this study, Aβ42 fibrils on reconstituted membranes composed of SPM lipids prepared from the precuneus cortex of human autopsied brains were evaluated by atomic force microscopy. In particular, Aβ42 accumulation, as well as the fibril number and size were higher for membranes with precuneus lipids than for membranes with calcarine cortex lipids. In addition, artificial peptide inhibitors targeting Aβ-sensitive ganglioside nanoclusters cleared Aβ assemblies on synaptic membranes in the brain, providing a novel therapeutic strategy for AD.
Collapse
Affiliation(s)
- Erika Miyamoto
- Department of Biosciences and Informatics, Keio University 3-14-1 Hiyoshi, Kouhoku-ku Yokohama 223-8522 Japan
| | - Hideki Hayashi
- Department of Applied Biochemistry, Tokyo University of Pharmacy and Life Sciences 1432-1 Horinouchi Hachioji Tokyo 192-0392 Japan
| | - Shigeo Murayama
- Brain Bank for Aging Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology 35-2 Sakae-cho Itabashi-ku Tokyo 173-0015 Japan
- Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University 2-2 Yamadaoka, Suita Osaka 565-0871 Japan
| | - Katsuhiko Yanagisawa
- Research and Development Center for Precision Medicine, University of Tsukuba 1-2 Kasuga Tsukuba Ibaraki 305-8550 Japan
| | - Toshinori Sato
- Department of Biosciences and Informatics, Keio University 3-14-1 Hiyoshi, Kouhoku-ku Yokohama 223-8522 Japan
| | - Teruhiko Matsubara
- Department of Biosciences and Informatics, Keio University 3-14-1 Hiyoshi, Kouhoku-ku Yokohama 223-8522 Japan
| |
Collapse
|
11
|
Kuramochi M, Nakamura M, Takahashi H, Komoriya T, Takita T, Pham NTK, Yasukawa K, Yoshimune K. Adenosine triphosphate induces amorphous aggregation of amyloid β by increasing Aβ dynamics. Sci Rep 2024; 14:8134. [PMID: 38584155 PMCID: PMC10999452 DOI: 10.1038/s41598-024-58773-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/03/2024] [Indexed: 04/09/2024] Open
Abstract
Amyloid β (Aβ) aggregates into two distinct fibril and amorphous forms in the brains of patients with Alzheimer's disease. Adenosine triphosphate (ATP) is a biological hydrotrope that causes Aβ to form amorphous aggregates and inhibit fibril formation at physiological concentrations. Based on diffracted X-ray blinking (DXB) analysis, the dynamics of Aβ significantly increased immediately after ATP was added compared to those in the absence and presence of ADP and AMP, and the effect diminished after 30 min as the aggregates formed. In the presence of ATP, the β-sheet content of Aβ gradually increased from the beginning, and in the absence of ATP, the content increased rapidly after 180 min incubation, as revealed by a time-dependent thioflavin T fluorescence assay. Images of an atomic force microscope revealed that ATP induces the formation of amorphous aggregates with an average diameter of less than 100 nm, preventing fibrillar formation during 4 days of incubation at 37 °C. ATP may induce amorphous aggregation by increasing the dynamics of Aβ, and as a result, the other aggregation pathway is omitted. Our results also suggest that DXB analysis is a useful method to evaluate the inhibitory effect of fibrillar formation.
Collapse
Affiliation(s)
- Masahiro Kuramochi
- Graduate School of Science and Engineering, Ibaraki University, Hitachi, 316-8511, Japan
| | - Momoka Nakamura
- Department of Applied Molecular Chemistry, Graduate School of Industrial Technology, Nihon University, 1-2-1, Izumichou, Narashino, Chiba, 275-8575, Japan
| | - Hiroto Takahashi
- Graduate School of Science and Engineering, Ibaraki University, Hitachi, 316-8511, Japan
| | - Tomoe Komoriya
- Department of Sustainable Engineering, College of Industrial Technology, Nihon University, 1-2-1, Izumichou, Narashino, Chiba, 275-8575, Japan
| | - Teisuke Takita
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Ngan Thi Kim Pham
- Department of Applied Molecular Chemistry, Graduate School of Industrial Technology, Nihon University, 1-2-1, Izumichou, Narashino, Chiba, 275-8575, Japan
| | - Kiyoshi Yasukawa
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Kazuaki Yoshimune
- Department of Applied Molecular Chemistry, Graduate School of Industrial Technology, Nihon University, 1-2-1, Izumichou, Narashino, Chiba, 275-8575, Japan.
| |
Collapse
|
12
|
Nam E, Lin Y, Park J, Do H, Han J, Jeong B, Park S, Lee DY, Kim M, Han J, Baik M, Lee Y, Lim MH. APP-C31: An Intracellular Promoter of Both Metal-Free and Metal-Bound Amyloid-β 40 Aggregation and Toxicity in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307182. [PMID: 37949680 PMCID: PMC10811509 DOI: 10.1002/advs.202307182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/18/2023] [Indexed: 11/12/2023]
Abstract
Intracellular C-terminal cleavage of the amyloid precursor protein (APP) is elevated in the brains of Alzheimer's disease (AD) patients and produces a peptide labeled APP-C31 that is suspected to be involved in the pathology of AD. But details about the role of APP-C31 in the development of the disease are not known. Here, this work reports that APP-C31 directly interacts with the N-terminal and self-recognition regions of amyloid-β40 (Aβ40 ) to form transient adducts, which facilitates the aggregation of both metal-free and metal-bound Aβ40 peptides and aggravates their toxicity. Specifically, APP-C31 increases the perinuclear and intranuclear generation of large Aβ40 deposits and, consequently, damages the nucleus leading to apoptosis. The Aβ40 -induced degeneration of neurites and inflammation are also intensified by APP-C31 in human neurons and murine brains. This study demonstrates a new function of APP-C31 as an intracellular promoter of Aβ40 amyloidogenesis in both metal-free and metal-present environments, and may offer an interesting alternative target for developing treatments for AD that have not been considered thus far.
Collapse
Affiliation(s)
- Eunju Nam
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Yuxi Lin
- Research Center for Bioconvergence AnalysisKorea Basic Science Institute (KBSI)OchangChungbuk28119Republic of Korea
| | - Jiyong Park
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
- Center for Catalytic Hydrocarbon FunctionalizationsInstitute for Basic Science (IBS)Daejeon34141Republic of Korea
| | - Hyunsu Do
- Graduate School of Medical Science and EngineeringKAISTDaejeon34141Republic of Korea
| | - Jiyeon Han
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Bohyeon Jeong
- Rare Disease Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)Daejeon34141Republic of Korea
| | - Subin Park
- Rare Disease Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)Daejeon34141Republic of Korea
- Department of BiochemistryDepartment of Medical ScienceChungnam National University School of MedicineDaejeon35015Republic of Korea
| | - Da Yong Lee
- Rare Disease Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)Daejeon34141Republic of Korea
| | - Mingeun Kim
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Jinju Han
- Graduate School of Medical Science and EngineeringKAISTDaejeon34141Republic of Korea
| | - Mu‐Hyun Baik
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
- Center for Catalytic Hydrocarbon FunctionalizationsInstitute for Basic Science (IBS)Daejeon34141Republic of Korea
| | - Young‐Ho Lee
- Research Center for Bioconvergence AnalysisKorea Basic Science Institute (KBSI)OchangChungbuk28119Republic of Korea
- Bio‐Analytical ScienceUniversity of Science and Technology (UST)Daejeon34113Republic of Korea
- Graduate School of Analytical Science and TechnologyChungnam National UniversityDaejeon34134Republic of Korea
- Department of Systems BiotechnologyChung‐Ang UniversityGyeonggi17546Republic of Korea
- Frontier Research Institute for Interdisciplinary SciencesTohoku UniversityMiyagi980‐8578Japan
| | - Mi Hee Lim
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| |
Collapse
|
13
|
Studzińska-Sroka E, Bulicz M, Henkel M, Rosiak N, Paczkowska-Walendowska M, Szwajgier D, Baranowska-Wójcik E, Korybalska K, Cielecka-Piontek J. Pleiotropic Potential of Evernia prunastri Extracts and Their Main Compounds Evernic Acid and Atranorin: In Vitro and In Silico Studies. Molecules 2023; 29:233. [PMID: 38202817 PMCID: PMC10780513 DOI: 10.3390/molecules29010233] [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: 12/01/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Evernia prunastri is a lichen widely distributed in the Northern Hemisphere. Its biological properties still need to be discovered. Therefore, our paper focuses on studies of E. prunastri extracts, including its main metabolites evernic acid (EA) or atranorin (ATR). Phytochemical profiles using chromatographic analysis were confirmed. The antioxidant activity was evaluated using in vitro chemical tests and in vitro enzymatic cells-free tests, namely superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione reductase (GR), and catalase (CAT). The anti-inflammatory potential using cyclooxygenase-2 (COX-2) and hyaluronidase were determined. The neuroprotective potential using acetylcholinesterase, (AChE), butyrylcholinesterase (BChE), and tyrosinase (Tyr) was estimated. The hypoglycemic activity was also confirmed (α-glucosidase). Principal component analysis was performed to determine the relationship between the biological activity of extracts. The inhibitory effect of EA and ATR on COX-2 AChE, BChE, Tyr, and α-glucosidase was evaluated using molecular docking techniques and confirmed for EA and ATR (besides α-glucosidase). The penetration of EA and ATR from extracts through the blood-brain barrier was confirmed using the parallel artificial membrane permeability assay blood-brain barrier test. In conclusion, depending on chemical surroundings and the concentration, the E. prunastri extracts, EA or ATR, showed attractive pleiotropic properties, which should be further investigated.
Collapse
Affiliation(s)
- Elżbieta Studzińska-Sroka
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznań, Poland; (M.B.); (M.H.); (N.R.); (M.P.-W.); (J.C.-P.)
| | - Magdalena Bulicz
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznań, Poland; (M.B.); (M.H.); (N.R.); (M.P.-W.); (J.C.-P.)
| | - Marika Henkel
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznań, Poland; (M.B.); (M.H.); (N.R.); (M.P.-W.); (J.C.-P.)
| | - Natalia Rosiak
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznań, Poland; (M.B.); (M.H.); (N.R.); (M.P.-W.); (J.C.-P.)
| | - Magdalena Paczkowska-Walendowska
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznań, Poland; (M.B.); (M.H.); (N.R.); (M.P.-W.); (J.C.-P.)
| | - Dominik Szwajgier
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, Skromna 8 Str., 20-704 Lublin, Poland; (D.S.); (E.B.-W.)
| | - Ewa Baranowska-Wójcik
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, Skromna 8 Str., 20-704 Lublin, Poland; (D.S.); (E.B.-W.)
| | - Katarzyna Korybalska
- Department of Patophysiology, Poznan University of Medical Science, Rokietnicka 8 Str., 60-806 Poznań, Poland;
| | - Judyta Cielecka-Piontek
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznań, Poland; (M.B.); (M.H.); (N.R.); (M.P.-W.); (J.C.-P.)
| |
Collapse
|
14
|
Miyamoto E, Sato T, Matsubara T. Cyclization of Peptides Enhances the Inhibitory Activity against Ganglioside-Induced Aβ Fibril Formation. ACS Chem Neurosci 2023; 14:4199-4207. [PMID: 37971427 DOI: 10.1021/acschemneuro.3c00589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disease and is the most common cause of dementia. It has been reported that the assembly of amyloid β-protein (Aβ) on the cell membrane is induced by the interaction of the Aβ monomer with gangliosides such as GM1. The ganglioside-bound Aβ (GAβ) complex acts as a seed to promote the toxic assembly of the Aβ fibrils. In a previous study, we found that a GM1 cluster-binding peptide (GCBP) specifically recognizes Aβ-sensitive ganglioside nanoclusters and inhibits the assembly of Aβ on a GM1-containing lipid membrane. In this study, cysteine-substituted double mutants of GCBP were designed and cyclized by intramolecular disulfide bond formation. Affinity assays indicated that one of the cyclic peptides had a higher affinity to a GM1-containing membrane compared to that of GCBP. Furthermore, surface topography analysis indicated that this peptide recognizes GM1 nanoclusters on the lipid membrane. An evaluation of the inhibitory kinetics indicated that the cyclic peptide could inhibit the formation of Aβ fibrils with an IC50 value of 1.2 fM, which is 10,000-fold higher than that of GCBP. The cyclic peptide was also shown to have a clearance effect on Aβ fibrils deposited on the lipid membrane and suppressed the formation of toxic Aβ assemblies. Our results indicate that the cyclic peptide that binds to the Aβ-sensitive ganglioside nanocluster is a potential novel inhibitor of ganglioside-induced Aβ assembly.
Collapse
Affiliation(s)
- Erika Miyamoto
- Department of Biosciences and Informatics, Keio University, 3-14-1 Hiyoshi, Kouhoku-ku, Yokohama 223-8522, Japan
| | - Toshinori Sato
- Department of Biosciences and Informatics, Keio University, 3-14-1 Hiyoshi, Kouhoku-ku, Yokohama 223-8522, Japan
| | - Teruhiko Matsubara
- Department of Biosciences and Informatics, Keio University, 3-14-1 Hiyoshi, Kouhoku-ku, Yokohama 223-8522, Japan
| |
Collapse
|
15
|
Posadas Y, Sánchez-López C, Quintanar L. Copper binding and protein aggregation: a journey from the brain to the human lens. RSC Chem Biol 2023; 4:974-985. [PMID: 38033729 PMCID: PMC10685798 DOI: 10.1039/d3cb00145h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/11/2023] [Indexed: 12/02/2023] Open
Abstract
Metal ions have been implicated in several proteinopathies associated to degenerative and neurodegenerative diseases. While the molecular mechanisms for protein aggregation are still under investigation, recent findings from Cryo-EM point out to polymorphisms in aggregates obtained from patients, as compared to those formed in vitro, suggesting that several factors may impact aggregation in vivo. One of these factors could be the direct binding of metal ions to the proteins engaged in aggregate formation. In this opinion article, three case studies are discussed to address the question of how metal ion binding to a peptide or protein may impact its conformation, folding, and aggregation, and how this may be relevant in understanding the polymorphic nature of the aggregates related to disease. Specifically, the impact of Cu2+ ions in the amyloid aggregation of amyloid-β and amylin (or IAPP- islet amyloid polypeptide) are discussed and then contrasted to the case of Cu2+-induced non-amyloid aggregation of human lens γ-crystallin proteins. For the intrinsically disordered peptides amyloid-β and IAPP, the impact of Cu2+ ion binding is highly dependent on the relative location of the metal binding site and the hydrophobic regions involved in β-sheet folding and amyloid formation. Further structural studies of how Cu2+ binding impacts amyloid aggregation pathways and the molecular structure of the final amyloid fibril, both, in vitro and in vivo, will certainly shed light into the molecular origins of the polymorphisms observed in diseased tissue. Finally, contrasting these cases to that of Cu2+-induced non-amyloid aggregation of γ-crystallins, it is evident that, although the impact in aggregation - and the nature of the aggregate - may differ in each system, at the molecular level there is a competition between metal ion coordination and the stability of β-sheet structures. Considering the importance of the β-sheet fold in biology, it is fundamental to understand the energetics and molecular details behind such competition. This opinion article aims to highlight future research directions in the field that can help tackle the important question of how metal ion binding may impact protein folding and aggregation and how this relates to disease.
Collapse
Affiliation(s)
- Yanahi Posadas
- Center for Research in Aging, Center for Research and Advanced Studies (Cinvestav) Mexico City 14330 Mexico
| | - Carolina Sánchez-López
- Center for Research in Aging, Center for Research and Advanced Studies (Cinvestav) Mexico City 14330 Mexico
| | - Liliana Quintanar
- Center for Research in Aging, Center for Research and Advanced Studies (Cinvestav) Mexico City 14330 Mexico
- Department of Chemistry, Center for Research and Advanced Studies (Cinvestav) Mexico City 07350 Mexico
| |
Collapse
|
16
|
Yu TG, Lee J, Yoon J, Choi JM, Kim DG, Heo WD, Song JJ, Kim HS. Engineering of a Fluorescent Protein for a Sensing of an Intrinsically Disordered Protein through Transition in the Chromophore State. JACS AU 2023; 3:3055-3065. [PMID: 38034956 PMCID: PMC10685427 DOI: 10.1021/jacsau.3c00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/18/2023] [Accepted: 10/02/2023] [Indexed: 12/02/2023]
Abstract
Intrinsically disordered proteins (IDPs) not only play important roles in biological processes but are also linked with the pathogenesis of various human diseases. Specific and reliable sensing of IDPs is crucial for exploring their roles but remains elusive due to structural plasticity. Here, we present the development of a new type of fluorescent protein for the ratiometric sensing and tracking of an IDP. A β-strand of green fluorescent protein (GFP) was truncated, and the resulting GFP was further engineered to undergo the transition in the absorption maximum upon binding of a target motif within amyloid-β (Aβ) as a model IDP through rational design and directed evolution. Spectroscopic and structural analyses of the engineered truncated GFP demonstrated that a shift in the absorption maximum is driven by the change in the chromophore state from an anionic (460 nm) state into a neutral (390 nm) state as the Aβ binds, allowing a ratiometric detection of Aβ. The utility of the developed GFP was shown by the efficient and specific detection of an Aβ and the tracking of its conformational change and localization in astrocytes.
Collapse
Affiliation(s)
- Tae-Geun Yu
- Departement
of Biological Sciences, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jinsu Lee
- Departement
of Biological Sciences, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jungmin Yoon
- Departement
of Biological Sciences, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jung Min Choi
- School
of Food Biotechnology and Nutrition, Kyungsung
University, Busan 48434, Korea
| | - Dong-Gun Kim
- Departement
of Biological Sciences, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Won Do Heo
- Departement
of Biological Sciences, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Ji-Joon Song
- Departement
of Biological Sciences, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Hak-Sung Kim
- Departement
of Biological Sciences, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
17
|
Wang J, Fu J, Zhao Y, Liu Q, Yan X, Su J. Iron and Targeted Iron Therapy in Alzheimer's Disease. Int J Mol Sci 2023; 24:16353. [PMID: 38003544 PMCID: PMC10671546 DOI: 10.3390/ijms242216353] [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: 09/17/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide. β-amyloid plaque (Aβ) deposition and hyperphosphorylated tau, as well as dysregulated energy metabolism in the brain, are key factors in the progression of AD. Many studies have observed abnormal iron accumulation in different regions of the AD brain, which is closely correlated with the clinical symptoms of AD; therefore, understanding the role of brain iron accumulation in the major pathological aspects of AD is critical for its treatment. This review discusses the main mechanisms and recent advances in the involvement of iron in the above pathological processes, including in iron-induced oxidative stress-dependent and non-dependent directions, summarizes the hypothesis that the iron-induced dysregulation of energy metabolism may be an initiating factor for AD, based on the available evidence, and further discusses the therapeutic perspectives of targeting iron.
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Su
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130012, China; (J.W.); (J.F.); (Y.Z.); (Q.L.); (X.Y.)
| |
Collapse
|
18
|
Abelein A. Metal Binding of Alzheimer's Amyloid-β and Its Effect on Peptide Self-Assembly. Acc Chem Res 2023; 56:2653-2663. [PMID: 37733746 PMCID: PMC10552549 DOI: 10.1021/acs.accounts.3c00370] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Indexed: 09/23/2023]
Abstract
Metal ions have been identified as key factors modulating the aggregation of amyloid-β peptide (Aβ) implicated in Alzheimer's disease (AD). The presence of elevated levels of metal ions in the amyloid plaques in AD patients supports the notion that the dysfunction of metal homeostasis is connected to the development of AD pathology. Here, recent findings from high- and low-resolution biophysical techniques are put into perspective, providing detailed insights into the molecular structures and dynamics of metal-bound Aβ complexes and the effect of metal ions on the Aβ aggregation process. In particular, the development of theoretical kinetic models deducing different microscopic nucleation events from the macroscopic aggregation behavior has enabled deciphering of the effect of metal ions on specific nucleation processes. In addition to these macroscopic measurements of bulk aggregation to quantify microscopic rates, recent NMR studies have revealed details about the structures and dynamics of metal-Aβ complexes, thereby linking structural events to bulk aggregation. Interestingly, transition-metal ions, such as copper, zinc, and silver ions, form a compact complex with the N-terminal part of monomeric Aβ, respectively, where the metal-bound "folded" state is in dynamic equilibrium with an "unfolded" state. The rates and thermodynamic features of these exchange dynamics have been determined by using NMR relaxation dispersion experiments. Additionally, the application of specifically tailored paramagnetic NMR experiments on the Cu(II)-Aβ complex has been fruitful in obtaining structural constraints within the blind sphere of conventional NMR experiments. This enables the determination of molecular structures of the "folded" Cu(II)-coordinated N-terminal region of Aβ. Furthermore, the discussed transition-metal ions modulate Aβ self-assembly in a concentration-dependent manner, where low metal ion concentrations inhibit Aβ fibril formation, while at high metal ion concentrations other processes occur, resulting in amorphous aggregate formation. Remarkably, the metal-Aβ interaction predominately reduces one specific nucleation step, the fibril-end elongation, whereas primary and surface-catalyzed secondary nucleation mechanisms are less affected. Specific inhibition of fibril-end elongation theoretically predicts an enhanced generation of Aβ oligomers, which is an interesting contribution to understanding metal-Aβ-associated neurotoxic effects. Taken together, the metal binding process creates a metal-bound Aβ complex, which is seemingly inert to aggregation. This process hence efficiently reduces the aggregation-prone peptide pool, which on the macroscopic level is reflected as slower aggregation kinetics. Thus, the specific binding of metals to the Aβ monomer can be linked to the macroscopic inhibitory effect on Aβ bulk aggregation, providing a molecular understanding of the Aβ aggregation mechanism in the presence of metal ions, where the metal ion can be seen as a minimalist agent against Aβ self-assembly. These insights can help to target Aβ aggregation in vivo, where metal ions are key factors modulating the Aβ self-assembly and Aβ-associated neurotoxicity.
Collapse
Affiliation(s)
- Axel Abelein
- Department of Biosciences
and Nutrition, Karolinska Institutet, 141 52 Huddinge, Sweden
| |
Collapse
|
19
|
Park S, Kim M, Lin Y, Hong M, Nam G, Mieczkowski A, Kardos J, Lee YH, Lim MH. Designing multi-target-directed flavonoids: a strategic approach to Alzheimer's disease. Chem Sci 2023; 14:9293-9305. [PMID: 37712013 PMCID: PMC10498667 DOI: 10.1039/d3sc00752a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 08/02/2023] [Indexed: 09/16/2023] Open
Abstract
The underlying causes of Alzheimer's disease (AD) remain a mystery, with multiple pathological components, including oxidative stress, acetylcholinesterase, amyloid-β, and metal ions, all playing a role. Here we report a strategic approach to designing flavonoids that can effectively tackle multiple pathological elements involved in AD. Our systematic investigations revealed key structural features for flavonoids to simultaneously target and regulate pathogenic targets. Our findings led to the development of a highly promising flavonoid that exhibits a range of functions, based on a complete structure-activity relationship analysis. Furthermore, our mechanistic studies confirmed that this flavonoid's versatile reactivities are driven by its redox potential and direct interactions with pathogenic factors. This work highlights the potential of multi-target-directed flavonoids as a novel solution in the fight against AD.
Collapse
Affiliation(s)
- Seongmin Park
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Mingeun Kim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Yuxi Lin
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI) Ochang Chungbuk 28119 Republic of Korea
| | - Mannkyu Hong
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
- Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS) Daejeon 34141 Republic of Korea
| | - Geewoo Nam
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Adam Mieczkowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences Pawińskiego 5a 02-106 Warsaw Poland
| | - József Kardos
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University Budapest 1117 Hungary
| | - Young-Ho Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI) Ochang Chungbuk 28119 Republic of Korea
- Bio-Analytical Science, University of Science and Technology (UST) Daejeon 34113 Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University Daejeon 34134 Republic of Korea
- Department of Systems Biotechnology, Chung-Ang University (CAU) Gyeonggi 17546 Republic of Korea
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University Sendai Miyagi 980-8578 Japan
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
20
|
Clemente-Suárez VJ, Redondo-Flórez L, Beltrán-Velasco AI, Ramos-Campo DJ, Belinchón-deMiguel P, Martinez-Guardado I, Dalamitros AA, Yáñez-Sepúlveda R, Martín-Rodríguez A, Tornero-Aguilera JF. Mitochondria and Brain Disease: A Comprehensive Review of Pathological Mechanisms and Therapeutic Opportunities. Biomedicines 2023; 11:2488. [PMID: 37760929 PMCID: PMC10526226 DOI: 10.3390/biomedicines11092488] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Mitochondria play a vital role in maintaining cellular energy homeostasis, regulating apoptosis, and controlling redox signaling. Dysfunction of mitochondria has been implicated in the pathogenesis of various brain diseases, including neurodegenerative disorders, stroke, and psychiatric illnesses. This review paper provides a comprehensive overview of the intricate relationship between mitochondria and brain disease, focusing on the underlying pathological mechanisms and exploring potential therapeutic opportunities. The review covers key topics such as mitochondrial DNA mutations, impaired oxidative phosphorylation, mitochondrial dynamics, calcium dysregulation, and reactive oxygen species generation in the context of brain disease. Additionally, it discusses emerging strategies targeting mitochondrial dysfunction, including mitochondrial protective agents, metabolic modulators, and gene therapy approaches. By critically analysing the existing literature and recent advancements, this review aims to enhance our understanding of the multifaceted role of mitochondria in brain disease and shed light on novel therapeutic interventions.
Collapse
Affiliation(s)
- Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (V.J.C.-S.); (J.F.T.-A.)
- Group de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| | - Laura Redondo-Flórez
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, C/Tajo s/n, Villaviciosa de Odón, 28670 Madrid, Spain
| | - Ana Isabel Beltrán-Velasco
- Psychology Department, Facultad de Ciencias de la Vida y la Naturaleza, Universidad Antonio de Nebrija, 28240 Madrid, Spain
| | - Domingo Jesús Ramos-Campo
- LFE Research Group, Department of Health and Human Performance, Faculty of Physical Activity and Sport Science-INEF, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Pedro Belinchón-deMiguel
- Department of Nursing and Nutrition, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain;
| | | | - Athanasios A. Dalamitros
- Laboratory of Evaluation of Human Biological Performance, School of Physical Education and Sport Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Rodrigo Yáñez-Sepúlveda
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2520000, Chile;
| | - Alexandra Martín-Rodríguez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (V.J.C.-S.); (J.F.T.-A.)
| | | |
Collapse
|
21
|
Liu L, Li X, Chen N, Chen X, Xing L, Zhou X, Liu S. Influence of cadmium ion on denaturation kinetics of hen egg white-lysozyme under thermal and acidic conditions. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 296:122650. [PMID: 36989696 DOI: 10.1016/j.saa.2023.122650] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/11/2023] [Accepted: 03/17/2023] [Indexed: 06/19/2023]
Abstract
To study the influence of Cd(II) ions on denaturation kinetics of hen egg white lysozyme (HEWL) under thermal and acidic conditions, spontaneous Raman spectroscopy in conjunction with Thioflavin-T fluorescence, AFM imaging, far-UV circular dichroism spectroscopy, and transmittance assays was conducted. Four distinctive Raman spectral markers for protein tertiary and secondary structures were recorded to follow the kinetics of conformational transformation. Through comparing variations of these markers in the presence or absence of Cd(II) ions, Cd(II) ions show an ability to efficiently accelerate the disruption of tertiary structure, and meanwhile, to promote the direct formation of organized β-sheets from the uncoiling of α-helices by skipping intermediate random coils. More significantly, with the action of Cd(II) ions, the initially resulting oligomers with disordered structures tend to assemble into aggregates with random structures like gels more than amyloid fibrils, along with a so-called "off-pathway" denaturation pathway. Our results advance the in-depth understanding of corresponding ion-specific effects.
Collapse
Affiliation(s)
- Liming Liu
- Department of Chemical Physics, University of Science and Technology of China, Hefei 230026, China
| | - Xinfei Li
- Department of Chemical Physics, University of Science and Technology of China, Hefei 230026, China
| | - Ning Chen
- Department of Chemical Physics, University of Science and Technology of China, Hefei 230026, China
| | - Xiaodong Chen
- Department of Chemical Physics, University of Science and Technology of China, Hefei 230026, China
| | - Lei Xing
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Xiaoguo Zhou
- Department of Chemical Physics, University of Science and Technology of China, Hefei 230026, China.
| | - Shilin Liu
- Department of Chemical Physics, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
22
|
Berntsson E, Vosough F, Noormägi A, Padari K, Asplund F, Gielnik M, Paul S, Jarvet J, Tõugu V, Roos PM, Kozak M, Gräslund A, Barth A, Pooga M, Palumaa P, Wärmländer SKTS. Characterization of Uranyl (UO 22+) Ion Binding to Amyloid Beta (Aβ) Peptides: Effects on Aβ Structure and Aggregation. ACS Chem Neurosci 2023; 14:2618-2633. [PMID: 37487115 PMCID: PMC10401651 DOI: 10.1021/acschemneuro.3c00130] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/06/2023] [Indexed: 07/26/2023] Open
Abstract
Uranium (U) is naturally present in ambient air, water, and soil, and depleted uranium (DU) is released into the environment via industrial and military activities. While the radiological damage from U is rather well understood, less is known about the chemical damage mechanisms, which dominate in DU. Heavy metal exposure is associated with numerous health conditions, including Alzheimer's disease (AD), the most prevalent age-related cause of dementia. The pathological hallmark of AD is the deposition of amyloid plaques, consisting mainly of amyloid-β (Aβ) peptides aggregated into amyloid fibrils in the brain. However, the toxic species in AD are likely oligomeric Aβ aggregates. Exposure to heavy metals such as Cd, Hg, Mn, and Pb is known to increase Aβ production, and these metals bind to Aβ peptides and modulate their aggregation. The possible effects of U in AD pathology have been sparsely studied. Here, we use biophysical techniques to study in vitro interactions between Aβ peptides and uranyl ions, UO22+, of DU. We show for the first time that uranyl ions bind to Aβ peptides with affinities in the micromolar range, induce structural changes in Aβ monomers and oligomers, and inhibit Aβ fibrillization. This suggests a possible link between AD and U exposure, which could be further explored by cell, animal, and epidemiological studies. General toxic mechanisms of uranyl ions could be modulation of protein folding, misfolding, and aggregation.
Collapse
Affiliation(s)
- Elina Berntsson
- Chemistry
Section, Arrhenius Laboratories, Stockholm
University, 106 91 Stockholm, Sweden
- Department
of Chemistry and Biotechnology, Tallinn
University of Technology, 19086 Tallinn, Estonia
| | - Faraz Vosough
- Chemistry
Section, Arrhenius Laboratories, Stockholm
University, 106 91 Stockholm, Sweden
| | - Andra Noormägi
- Department
of Chemistry and Biotechnology, Tallinn
University of Technology, 19086 Tallinn, Estonia
| | - Kärt Padari
- Institute
of Molecular and Cell Biology, University
of Tartu, 50090 Tartu, Estonia
| | - Fanny Asplund
- Chemistry
Section, Arrhenius Laboratories, Stockholm
University, 106 91 Stockholm, Sweden
| | - Maciej Gielnik
- Department
of Molecular Biology and Genetics, Aarhus
University, 8000 Aarhus, Denmark
| | - Suman Paul
- Chemistry
Section, Arrhenius Laboratories, Stockholm
University, 106 91 Stockholm, Sweden
| | - Jüri Jarvet
- Chemistry
Section, Arrhenius Laboratories, Stockholm
University, 106 91 Stockholm, Sweden
- CellPept
Sweden AB, Kvarngatan
10B, 118 47 Stockholm, Sweden
| | - Vello Tõugu
- Department
of Chemistry and Biotechnology, Tallinn
University of Technology, 19086 Tallinn, Estonia
| | - Per M. Roos
- Institute
of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- University
Healthcare Unit of Capio St. Göran Hospital, 112 81 Stockholm, Sweden
| | - Maciej Kozak
- Department
of Biomedical Physics, Institute of Physics, Faculty of Physics, Adam Mickiewicz University, 61-712 Poznań, Poland
- SOLARIS
National Synchrotron Radiation Centre, Jagiellonian
University, 31-007 Kraków, Poland
| | - Astrid Gräslund
- Chemistry
Section, Arrhenius Laboratories, Stockholm
University, 106 91 Stockholm, Sweden
- CellPept
Sweden AB, Kvarngatan
10B, 118 47 Stockholm, Sweden
| | - Andreas Barth
- Chemistry
Section, Arrhenius Laboratories, Stockholm
University, 106 91 Stockholm, Sweden
| | - Margus Pooga
- Institute
of Technology, University of Tartu, 50090 Tartu, Estonia
| | - Peep Palumaa
- Department
of Chemistry and Biotechnology, Tallinn
University of Technology, 19086 Tallinn, Estonia
| | - Sebastian K. T. S. Wärmländer
- Chemistry
Section, Arrhenius Laboratories, Stockholm
University, 106 91 Stockholm, Sweden
- CellPept
Sweden AB, Kvarngatan
10B, 118 47 Stockholm, Sweden
| |
Collapse
|
23
|
Wegermann CA, Pirota V, Monzani E, Casella L, Costa LAS, Novato WTG, Machini MT, da Costa Ferreira AM. Interaction studies of oxindole-derivatives with β-amyloid peptides inhibiting its aggregation induced by metal ions. J Inorg Biochem 2023; 245:112227. [PMID: 37156056 DOI: 10.1016/j.jinorgbio.2023.112227] [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: 12/19/2022] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 05/10/2023]
Abstract
Some hydrazones and Schiff bases derived from isatin, an endogenous oxindole formed in the metabolism of tryptophan, were obtained to investigate their effects on in vitro aggregation of β-amyloid peptides (Aβ), macromolecules implicated in Alzheimer's disease. Some hydrazone ligands, prepared by condensation reactions of isatin with hydrazine derivatives, showed a large affinity binding to the synthetic peptides Aβ, particularly to Aβ1-16. Measurements by NMR spectroscopy indicated that those interactions occur mainly at the metal binding site of the peptide, involving His6, His13, and His14 residues, and that hydrazone E-diastereoisomer interacts preferentially with the amyloid peptides. Experimental results were consistent with simulations using a docking approach, where it is demonstrated that the amino acid residues Glu3, His6, His13, and His14 are those that mostly interact with the ligands. Further, these oxindole-derived ligands can efficiently chelate copper(II) and zinc(II) ions, forming moderate stable [ML] 1:1 species. The corresponding formation constants were determined by UV/Vis spectroscopy, by titrations of the ligands with increasing amounts of metal salts, and the obtained log K values were in the range 2.74 to 5.11. Both properties, good affinity for amyloid peptides, and reasonably good capacity of chelating biometal ions, like copper and zinc, can explain the efficient inhibition of Aβ fragments aggregation, as shown by experiments carried out with the oxindole derivatives in the presence of metal ions.
Collapse
Affiliation(s)
- Camila Anchau Wegermann
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Valentina Pirota
- Dipartimento di Chimica, Università degli Studi di Pavia, Pavia, Italy.
| | - Enrico Monzani
- Dipartimento di Chimica, Università degli Studi di Pavia, Pavia, Italy.
| | - Luigi Casella
- Dipartimento di Chimica, Università degli Studi di Pavia, Pavia, Italy.
| | - Luiz Antônio Sodré Costa
- NEQC - Núcleo de Estudos em Química Computacional, Departamento de Química, ICE, Universidade Federal de Juiz de Fora, MG, Brazil.
| | - Willian Tássio Gomes Novato
- NQTCM, Núcleo de Química Teórica e Computacional de Macaé, Instituto Multidisciplinar de Química, CM UFRJ Macaé, Universidade Federal do Rio de Janeiro, RJ, Brazil.
| | - M Teresa Machini
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Ana Maria da Costa Ferreira
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
24
|
Baek JH, Kang JS, Song M, Lee DK, Kim HJ. Glutamine Supplementation Preserves Glutamatergic Neuronal Activity in the Infralimbic Cortex, Which Delays the Onset of Mild Cognitive Impairment in 3×Tg-AD Female Mice. Nutrients 2023; 15:2794. [PMID: 37375700 DOI: 10.3390/nu15122794] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/16/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
It was recently found that glutamine (Gln) supplementation activates glutamatergic neurotransmission and prevents chronic-stress-induced mild cognitive impairment (MCI). In this study, we evaluated the effects of Gln on glutamatergic activity in the medial prefrontal cortex and the onset of cognitive impairment in a triple-transgenic Alzheimer's disease mouse model (3×Tg-AD). Female 3×Tg-AD mice were fed a normal diet (3×Tg) or a Gln-supplemented diet (3×Tg+Gln) from 2 to 6 months of age. Glutamatergic neuronal activity was analyzed at 6 months, and cognitive function was examined at 2, 4, and 6 months. 3×Tg mice exhibited a decrease in glutamatergic neurotransmission in the infralimbic cortex, but 3×Tg+Gln mice did not. The 3×Tg group showed MCI at 6 months of age, but the 3×Tg+Gln group did not. The expressions of amyloid peptide, inducible nitric oxide synthase, and IBA-1 were not elevated in the infralimbic cortex in the 3×Tg+Gln group. Therefore, a Gln-supplemented diet could delay the onset of MCI even in a mouse model predisposed to cognitive impairment and dementia through genetic modification.
Collapse
Affiliation(s)
- Ji Hyeong Baek
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Tyrosine Peptide Multiuse Research Group, Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University Medical School, 15 Jinju-daero 816 Beongil, Jinju 52727, Gyeongnam, Republic of Korea
| | - Jae Soon Kang
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Tyrosine Peptide Multiuse Research Group, Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University Medical School, 15 Jinju-daero 816 Beongil, Jinju 52727, Gyeongnam, Republic of Korea
| | - Miyoung Song
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Tyrosine Peptide Multiuse Research Group, Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University Medical School, 15 Jinju-daero 816 Beongil, Jinju 52727, Gyeongnam, Republic of Korea
| | - Dong Kun Lee
- Department of Physiology, Institute of Health Sciences, Gyeongsang National University Medical School, 15 Jinju-daero 816 Beongil, Jinju 52727, Gyeongnam, Republic of Korea
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Sciences, Institute of Health Sciences, Tyrosine Peptide Multiuse Research Group, Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University Medical School, 15 Jinju-daero 816 Beongil, Jinju 52727, Gyeongnam, Republic of Korea
| |
Collapse
|
25
|
Du Z, Nam E, Lin Y, Hong M, Molnár T, Kondo I, Ishimori K, Baik MH, Lee YH, Lim MH. Unveiling the impact of oxidation-driven endogenous protein interactions on the dynamics of amyloid-β aggregation and toxicity. Chem Sci 2023; 14:5340-5349. [PMID: 37234895 PMCID: PMC10208028 DOI: 10.1039/d3sc00881a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Cytochrome c (Cyt c), a multifunctional protein with a crucial role in controlling cell fate, has been implicated in the amyloid pathology associated with Alzheimer's disease (AD); however, the interaction between Cyt c and amyloid-β (Aβ) with the consequent impact on the aggregation and toxicity of Aβ is not known. Here we report that Cyt c can directly bind to Aβ and alter the aggregation and toxicity profiles of Aβ in a manner that is dependent on the presence of a peroxide. When combined with hydrogen peroxide (H2O2), Cyt c redirects Aβ peptides into less toxic, off-pathway amorphous aggregates, whereas without H2O2, it promotes Aβ fibrillization. The mechanisms behind these effects may involve a combination of the complexation between Cyt c and Aβ, the oxidation of Aβ by Cyt c and H2O2, and the modification of Cyt c by H2O2. Our findings demonstrate a new function of Cyt c as a modulator against Aβ amyloidogenesis.
Collapse
Affiliation(s)
- Zhi Du
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
- Department of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology Taiyuan 030024 PR China
| | - Eunju Nam
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Yuxi Lin
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI) Ochang Chungbuk 28119 Republic of Korea
| | - Mannkyu Hong
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
- Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS) Daejeon 34141 Republic of Korea
| | - Tamás Molnár
- Department of Biochemistry, Institute of Biology, Eötvös Loránd University H-1117 Budapest Hungary
| | - Ikufumi Kondo
- Graduate School of Chemical Sciences and Engineering, Hokkaido University Kita 13, Nishi 8, Kita-ku Sapporo 060-8628 Japan
| | - Koichiro Ishimori
- Graduate School of Chemical Sciences and Engineering, Hokkaido University Kita 13, Nishi 8, Kita-ku Sapporo 060-8628 Japan
- Department of Chemistry, Faculty of Science, Hokkaido University Kita 10, Nishi 8, Kita-ku Sapporo 060-0810 Japan
| | - Mu-Hyun Baik
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
- Center for Catalytic Hydrocarbon Functionalizations, Institute for Basic Science (IBS) Daejeon 34141 Republic of Korea
| | - Young-Ho Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI) Ochang Chungbuk 28119 Republic of Korea
- Bio-Analytical Science, University of Science and Technology (UST) Daejeon 34113 Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University Daejeon 34134 Republic of Korea
- Research Headquarters, Korea Brain Research Institute (KBRI) Daegu 41068 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
26
|
Mehta PK, Lee J, Oh ET, Park HJ, Lee KH. Ratiometric Fluorescence Sensing System for Lead Ions Based on Self-Assembly of Bioprobes Triggered by Specific Pb 2+-Peptide Interactions. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 36883859 DOI: 10.1021/acsami.3c00567] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Lead is one of the most toxic substances. However, there are few ratiometric fluorescent probes for sensing Pb2+ in aqueous solution as well as living cells because specific ligands for Pb2+ ions have not been well characterized. Considering the interactions between Pb2+ and peptides, we developed ratiometric fluorescent probes for Pb2+ based on the peptide receptor in two steps. First, we synthesized fluorescent probes (1-3) based on the tetrapeptide receptor (ECEE-NH2) containing hard and soft ligands by conjugation with diverse fluorophores that showed excimer emission when they aggregated. After investigation of fluorescent responses to metal ions, benzothiazolyl-cyanovinylene was evaluated as an appropriate fluorophore for ratiometric detection of Pb2+. Next, we modified the peptide receptor to decrease the number of hard ligands and/or to replace Cys with disulfide bond and methylated Cys for improving selectivity and cell permeability. From this process, we developed two fluorescent probes (3 and 8) among the probes (1-8) that exhibited remarkable ratiometric sensing properties for Pb2+ including high water solubility (≤2% DMF), visible light excitation, high sensitivity, selectivity for Pb2+, low detection limits (<10 nM), and fast response (<6 min). The binding mode study revealed that specific Pb2+-peptide interactions of the probes caused nanosized aggregates in which the fluorophores of the probes came close each other, exhibiting excimer emission. In particular, 8 based on tetrapeptide bearing a disulfide bond and two carboxyl groups with a good permeability successfully quantified intracellular uptake of Pb2+ in live cells through ratiometric fluorescent signals. The ratiometric sensing system based on specific metal-peptide interactions and excimer emission process could provide a valuable tool to quantify Pb2+ in live cells and pure aqueous solutions.
Collapse
Affiliation(s)
- Pramod Kumar Mehta
- Education and Research Center for Smart Energy Materials and Process, Department of Chemistry and Chemical Engineering, Inha University, Incheon 402-751, South Korea
| | - JaeYoon Lee
- Education and Research Center for Smart Energy Materials and Process, Department of Chemistry and Chemical Engineering, Inha University, Incheon 402-751, South Korea
| | - Eun-Taex Oh
- Department in Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, South Korea
| | - Heon Joo Park
- Program in Biomedical Science & Engineering, Inha University, Incheon 22212, South Korea
| | - Keun-Hyeung Lee
- Education and Research Center for Smart Energy Materials and Process, Department of Chemistry and Chemical Engineering, Inha University, Incheon 402-751, South Korea
| |
Collapse
|
27
|
Suh JM, Kim M, Yoo J, Han J, Paulina C, Lim MH. Intercommunication between metal ions and amyloidogenic peptides or proteins in protein misfolding disorders. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
28
|
Berntsson E, Vosough F, Svantesson T, Pansieri J, Iashchishyn IA, Ostojić L, Dong X, Paul S, Jarvet J, Roos PM, Barth A, Morozova-Roche LA, Gräslund A, Wärmländer SKTS. Residue-specific binding of Ni(II) ions influences the structure and aggregation of amyloid beta (Aβ) peptides. Sci Rep 2023; 13:3341. [PMID: 36849796 PMCID: PMC9971182 DOI: 10.1038/s41598-023-29901-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. AD brains display deposits of insoluble amyloid plaques consisting mainly of aggregated amyloid-β (Aβ) peptides, and Aβ oligomers are likely a toxic species in AD pathology. AD patients display altered metal homeostasis, and AD plaques show elevated concentrations of metals such as Cu, Fe, and Zn. Yet, the metal chemistry in AD pathology remains unclear. Ni(II) ions are known to interact with Aβ peptides, but the nature and effects of such interactions are unknown. Here, we use numerous biophysical methods-mainly spectroscopy and imaging techniques-to characterize Aβ/Ni(II) interactions in vitro, for different Aβ variants: Aβ(1-40), Aβ(1-40)(H6A, H13A, H14A), Aβ(4-40), and Aβ(1-42). We show for the first time that Ni(II) ions display specific binding to the N-terminal segment of full-length Aβ monomers. Equimolar amounts of Ni(II) ions retard Aβ aggregation and direct it towards non-structured aggregates. The His6, His13, and His14 residues are implicated as binding ligands, and the Ni(II)·Aβ binding affinity is in the low µM range. The redox-active Ni(II) ions induce formation of dityrosine cross-links via redox chemistry, thereby creating covalent Aβ dimers. In aqueous buffer Ni(II) ions promote formation of beta sheet structure in Aβ monomers, while in a membrane-mimicking environment (SDS micelles) coil-coil helix interactions appear to be induced. For SDS-stabilized Aβ oligomers, Ni(II) ions direct the oligomers towards larger sizes and more diverse (heterogeneous) populations. All of these structural rearrangements may be relevant for the Aβ aggregation processes that are involved in AD brain pathology.
Collapse
Affiliation(s)
- Elina Berntsson
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden.
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia.
| | - Faraz Vosough
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Teodor Svantesson
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Jonathan Pansieri
- Department of Medical Biochemistry and Biophysics, Umeå University, 901 87, Umeå, Sweden
| | - Igor A Iashchishyn
- Department of Medical Biochemistry and Biophysics, Umeå University, 901 87, Umeå, Sweden
| | - Lucija Ostojić
- Department of Medical Biochemistry and Biophysics, Umeå University, 901 87, Umeå, Sweden
| | - Xiaolin Dong
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Suman Paul
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | - Jüri Jarvet
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
- The National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Per M Roos
- Institute of Environmental Medicine, Karolinska Institutet, Nobels Väg 13, 171 77, Stockholm, Sweden
- Department of Clinical Physiology, Capio St. Göran Hospital, St. Göransplan 1, 112 19, Stockholm, Sweden
| | - Andreas Barth
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | | | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, 106 91, Stockholm, Sweden
| | | |
Collapse
|
29
|
Yi Y, Lim MH. Current understanding of metal-dependent amyloid-β aggregation and toxicity. RSC Chem Biol 2023; 4:121-131. [PMID: 36794021 PMCID: PMC9906324 DOI: 10.1039/d2cb00208f] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/22/2022] [Indexed: 11/23/2022] Open
Abstract
The discovery of effective therapeutics targeting amyloid-β (Aβ) aggregates for Alzheimer's disease (AD) has been very challenging, which suggests its complicated etiology associated with multiple pathogenic elements. In AD-affected brains, highly concentrated metals, such as copper and zinc, are found in senile plaques mainly composed of Aβ aggregates. These metal ions are coordinated to Aβ and affect its aggregation and toxicity profiles. In this review, we illustrate the current view on molecular insights into the assembly of Aβ peptides in the absence and presence of metal ions as well as the effect of metal ions on their toxicity.
Collapse
Affiliation(s)
- Yelim Yi
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
30
|
Roldán-Martín L, Sodupe M, Maréchal JD. Computational assessment of the impact of Cu(II) and Al(III) on β-amyloid 42 fibrils: Binding sites, structural stability, and possible physiological implications. Front Neurosci 2023; 17:1110311. [PMID: 36814794 PMCID: PMC9940836 DOI: 10.3389/fnins.2023.1110311] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/10/2023] [Indexed: 02/09/2023] Open
Abstract
One of Alzheimer's disease major hallmarks is the aggregation of β-amyloid peptide, a process in which metal ions play an important role. In the present work, an integrative computational study has been performed to identify the metal-binding regions and determine the conformational impact of Cu(II) and Al(III) ion binding to the β-amyloid (Aβ42) fibrillary structure. Through classical and Gaussian accelerated molecular dynamics, it has been observed that the metal-free fiber shows a hinge fan-like motion of the S-shaped structure, maintaining the general conformation. Upon metal coordination, distinctive patterns are observed depending on the metal. Cu(II) binds to the flexible N-terminal region and induces structural changes that could ultimately disrupt the fibrillary structure. In contrast, Al(III) binding takes place with the residues Glu22 and Asp23, and its binding reinforces the core stability of the system. These results give clues on the molecular impact of the interaction of metal ions with the aggregates and sustain their non-innocent roles in the evolution of the illness.
Collapse
|
31
|
Suprun EV, Radko SP, Kozin SA, Mitkevich VA, Makarov AA. Electrochemical Analysis in Studying β-Amyloid Aggregation. BIOCHEMISTRY (MOSCOW) 2023; 88:S88-S104. [PMID: 37069116 DOI: 10.1134/s0006297923140067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
β-amyloid (Aβ) is comprised of a group of peptides formed as a result of cleavage of the amyloid precursor protein by secretases. Aβ aggregation is considered as a central event in pathogenesis of Alzheimer's disease, the most common human neurodegenerative disorder. Molecular mechanisms of Aβ aggregation have intensively being investigated using synthetic Aβ peptides by methods based on monitoring of aggregates, including determination of their size and structure. In this review, an orthogonal approach to the study of Aβ aggregation is considered, which relies on electrochemical registration of the loss of peptide monomers. Electrochemical analysis of Aβ (by voltammetry and amperometric flow injection analysis) is based on registration of the oxidation signal of electroactive amino acid residues of the peptide on an electrode surface. The Aβ oxidation signal disappears, when the peptide is included in the aggregate. The advantages and disadvantages of electrochemical analysis for the study of spontaneous and metal-induced aggregation of Aβ, comparative analysis of various peptide isoforms, and study of the process of complexation of metal ions with the metal-binding domain of Aβ are discussed. It is concluded that the combined use of the electrochemical method and the methods based on detection of Aβ aggregates makes it possible to obtain more complete information about the mechanisms of peptide aggregation.
Collapse
Affiliation(s)
- Elena V Suprun
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia.
- Orekhovich Institute of Biomedical Chemistry, Moscow, 119121, Russia
| | - Sergey P Radko
- Orekhovich Institute of Biomedical Chemistry, Moscow, 119121, Russia
| | - Sergey A Kozin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Vladimir A Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Alexander A Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| |
Collapse
|
32
|
Abelein A, Ciofi-Baffoni S, Mörman C, Kumar R, Giachetti A, Piccioli M, Biverstål H. Molecular Structure of Cu(II)-Bound Amyloid-β Monomer Implicated in Inhibition of Peptide Self-Assembly in Alzheimer's Disease. JACS AU 2022; 2:2571-2584. [PMID: 36465548 PMCID: PMC9709942 DOI: 10.1021/jacsau.2c00438] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 05/22/2023]
Abstract
Metal ions, such as copper and zinc ions, have been shown to strongly modulate the self-assembly of the amyloid-β (Aβ) peptide into insoluble fibrils, and elevated concentrations of metal ions have been found in amyloid plaques of Alzheimer's patients. Among the physiological transition metal ions, Cu(II) ions play an outstanding role since they can trigger production of neurotoxic reactive oxygen species. In contrast, structural insights into Cu(II) coordination of Aβ have been challenging due to the paramagnetic nature of Cu(II). Here, we employed specifically tailored paramagnetic NMR experiments to determine NMR structures of Cu(II) bound to monomeric Aβ. We found that monomeric Aβ binds Cu(II) in the N-terminus and combined with molecular dynamics simulations, we could identify two prevalent coordination modes of Cu(II). For these, we report here the NMR structures of the Cu(II)-bound Aβ complex, exhibiting heavy backbone RMSD values of 1.9 and 2.1 Å, respectively. Further, applying aggregation kinetics assays, we identified the specific effect of Cu(II) binding on the Aβ nucleation process. Our results show that Cu(II) efficiently retards Aβ fibrillization by predominately reducing the rate of fibril-end elongation at substoichiometric ratios. A detailed kinetic analysis suggests that this specific effect results in enhanced Aβ oligomer generation promoted by Cu(II). These results can quantitatively be understood by Cu(II) interaction with the Aβ monomer, forming an aggregation inert complex. In fact, this mechanism is strikingly similar to other transition metal ions, suggesting a common mechanism of action of retarding Aβ self-assembly, where the metal ion binding to monomeric Aβ is a key determinant.
Collapse
Affiliation(s)
- Axel Abelein
- Department
of Biosciences and Nutrition, Karolinska
Institutet, Huddinge141 83, Sweden
| | - Simone Ciofi-Baffoni
- Magnetic
Resonance Center and Department of Chemistry, University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino50019 , Florence, Italy
| | - Cecilia Mörman
- Department
of Biosciences and Nutrition, Karolinska
Institutet, Huddinge141 83, Sweden
- Department
of Biochemistry and Biophysics, The Arrhenius Laboratories, Stockholm University, Stockholm106 91, Sweden
| | - Rakesh Kumar
- Department
of Biosciences and Nutrition, Karolinska
Institutet, Huddinge141 83, Sweden
| | - Andrea Giachetti
- Magnetic
Resonance Center and Department of Chemistry, University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino50019 , Florence, Italy
| | - Mario Piccioli
- Magnetic
Resonance Center and Department of Chemistry, University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino50019 , Florence, Italy
| | - Henrik Biverstål
- Department
of Biosciences and Nutrition, Karolinska
Institutet, Huddinge141 83, Sweden
- Department
of Physical Organic Chemistry, Latvian Institute
of Organic Synthesis, RigaLV-1006, Latvia
| |
Collapse
|
33
|
Diaz-Espinoza R. Catalytically Active Amyloids as Future Bionanomaterials. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3802. [PMID: 36364578 PMCID: PMC9656882 DOI: 10.3390/nano12213802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 06/16/2023]
Abstract
Peptides and proteins can aggregate into highly ordered and structured conformations called amyloids. These supramolecular structures generally have convergent features, such as the formation of intermolecular beta sheets, that lead to fibrillary architectures. The resulting fibrils have unique mechanical properties that can be exploited to develop novel nanomaterials. In recent years, sequences of small peptides have been rationally designed to self-assemble into amyloids that catalyze several chemical reactions. These amyloids exhibit reactive surfaces that can mimic the active sites of enzymes. In this review, I provide a state-of-the-art summary of the development of catalytically active amyloids. I will focus especially on catalytic activities mediated by hydrolysis, which are the most studied examples to date, as well as novel types of recently reported activities that promise to expand the possible repertoires. The combination of mechanical properties with catalytic activity in an amyloid scaffold has great potential for the development of future bionanomaterials aimed at specific applications.
Collapse
Affiliation(s)
- Rodrigo Diaz-Espinoza
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago 3363, Chile
| |
Collapse
|
34
|
Mocanu CS, Darie-Ion L, Petre BA, Gradinaru VR, Drochioiu G. A computational study of metal ions interaction with amyloid-β 1-42 peptide structure in hyperpyrexia: Implications for Alzheimer disease. JOURNAL OF KING SAUD UNIVERSITY. SCIENCE 2022; 34:102184. [PMID: 35783243 PMCID: PMC9238029 DOI: 10.1016/j.jksus.2022.102184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 05/28/2023]
Abstract
Given the current context of the SARS-CoV-19 pandemic, among the interfering risky factors with the Aβ peptide aggregation in the brains of Alzheimer's disease (AD) patients can be hyperpyrexia and increased intracranial pressure (ICP). According to our hypothesis on the relationship between hyperpyrexia and cognitive decline in AD, two models of Aβ peptides were used in this study: the structure of AD amyloid beta-peptide and near-atomic resolution fibril structures of the Aβ peptide. Therefore, the binding templates were constructed for Aβ peptide regions able to bind 9 different metal ions. The fragment transformation method was used for the structural comparison between Aβ chains. Molecular dynamics simulation (MDS) was applied using the Nose-Poincare-Anderson equation to generate a theoretically correct NPT (isothermal-isobaric ensemble). The smallest dissimilarities were observed in the case of Cu+ binding potential followed by Co2+, both with similar variation. Structural changes have also occurred as a result of the dynamic simulation. All these changes suggest an aggravating factor in both hyperpyretic and AD conditions. Our findings suggest that elevated temperature and increased intracranial pressure rise the effect of peptide aggregation, by converting α-helix motif to β-sheet and random coil conformation, which are related to the formation of senile plaques in AD brains.
Collapse
Affiliation(s)
- Cosmin Stefan Mocanu
- Faculty of Chemistry, "Al. I. Cuza" University of Iasi, 11 Carol I, Iasi 70605, Romania
| | - Laura Darie-Ion
- Faculty of Chemistry, "Al. I. Cuza" University of Iasi, 11 Carol I, Iasi 70605, Romania
| | - Brindusa Alina Petre
- Faculty of Chemistry, "Al. I. Cuza" University of Iasi, 11 Carol I, Iasi 70605, Romania
- Center for Fundamental Research and Experimental Development in Translation Medicine, Regional Institute of Oncology, 2-4 General Henri Mathias Berthelot Str., 700483 Iasi, Romania
| | | | - Gabi Drochioiu
- Faculty of Chemistry, "Al. I. Cuza" University of Iasi, 11 Carol I, Iasi 70605, Romania
| |
Collapse
|
35
|
Kim C, Park J, Kim W, Lee W, Na S, Park J. Detection of Cd 2+ and Pb 2+ using amyloid oligomer-reduced graphene oxide composite. Bioelectrochemistry 2022; 147:108214. [PMID: 35901626 DOI: 10.1016/j.bioelechem.2022.108214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/02/2022]
Abstract
Heavy metal ions are toxic to humans and can further interact with amyloid in the human body to produce amyloid plaques, which disrupt neurotransmitter function and are linked to Alzheimer's and Parkinson's diseases. In this study, we developed an amyloid oligomer-reduced graphene oxide composite (AOrGOC) electrochemical sensor for effective heavy metal ion detection based on square-wave anodic stripping voltammetry. The reactivity between amyloids and heavy metal ions was studied by analyzing the stripping current for different amyloids (lysozyme, bovine serum albumin, and β-lactoglobulin) and amyloid growth types (monomers, oligomers, and fibrils). Reduced graphene oxide was used to improve the sensitivity of the sensor. The AOrGOC sensor exhibited the detection limits of 86.0 and 9.5 nM for Cd2+ and Pb2+, respectively, and selectively detected Cd2+ and Pb2+ over other heavy metal ions. The AOrGOC sensor also detected Cd2+ and Pb2+ in human plasma, thus exhibiting its potential as a biosensor. This study not only promoted our fundamental understanding of amyloids and the detection of heavy metal ions using amyloids, but also provided valuable insights into amyloid-based electrochemical sensors.
Collapse
Affiliation(s)
- Chihyun Kim
- Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, South Korea
| | - Joohyung Park
- Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, South Korea
| | - Woochang Kim
- Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, South Korea
| | - Wonseok Lee
- Department of Electrical Engineering, Korea National University of Transportation, Chungju 27469, South Korea.
| | - Sungsoo Na
- Department of Mechanical Engineering, Korea University, Seoul 02841, South Korea.
| | - Jinsung Park
- Department of Biomechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
36
|
Han J, Yoon J, Shin J, Nam E, Qian T, Li Y, Park K, Lee SH, Lim MH. Conformational and functional changes of the native neuropeptide somatostatin occur in the presence of copper and amyloid-β. Nat Chem 2022; 14:1021-1030. [PMID: 35817963 DOI: 10.1038/s41557-022-00984-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 05/26/2022] [Indexed: 11/09/2022]
Abstract
The progression of neurodegenerative disorders can lead to impaired neurotransmission; however, the role of pathogenic factors associated with these diseases and their impact on the structures and functions of neurotransmitters have not been clearly established. Here we report the discovery that conformational and functional changes of a native neuropeptide, somatostatin (SST), occur in the presence of copper ions, metal-free amyloid-β (Aβ) and metal-bound Aβ (metal-Aβ) found as pathological factors in the brains of patients with Alzheimer's disease. These pathological elements induce the self-assembly of SST and, consequently, prevent it from binding to the receptor. In the reverse direction, SST notably modifies the aggregation profiles of Aβ species in the presence of metal ions, attenuating their cytotoxicity and interactions with cell membranes. Our work demonstrates a loss of normal function of SST as a neurotransmitter and a gain of its modulative function against metal-Aβ under pathological conditions.
Collapse
Affiliation(s)
- Jiyeon Han
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jiwon Yoon
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
| | - Jeongcheol Shin
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Eunju Nam
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Tongrui Qian
- State Key Laboratory Membrane Biology, Peking University School of Life Sciences, Beijing, China
| | - Yulong Li
- State Key Laboratory Membrane Biology, Peking University School of Life Sciences, Beijing, China.,PKU-IDG/McGovern Institute for Brain Research, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Kiyoung Park
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| | - Seung-Hee Lee
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea.
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| |
Collapse
|
37
|
Rana M, Cho HJ, Arya H, Bhatt TK, Bhar K, Bhatt S, Mirica LM, Sharma AK. Azo-Stilbene and Pyridine-Amine Hybrid Multifunctional Molecules to Target Metal-Mediated Neurotoxicity and Amyloid-β Aggregation in Alzheimer's Disease. Inorg Chem 2022; 61:10294-10309. [PMID: 35768324 DOI: 10.1021/acs.inorgchem.2c00502] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD) are associated with progressive neuronal cell death, and they are commonly correlated with aberrant protein misfolding and aggregation of Aβ peptides. Transition metal ions (Cu, Fe, and Zn) have been shown to promote aggregation and oxidative stress through formation of Aβ-metal complexes. In this context, integrating molecular scaffolds rationally is used here to generate multifunctional molecules as modulators for metal-induced abnormalities. This work encompasses two azo-stilbene (AS)-derived compounds (AS-HL1 and AS-HL2), the rationale behind the design, their synthesis, characterization, and metal chelation ability [Cu(II) and Zn(II)]. The molecular frameworks of the designed compounds consist of stilbene as an Aβ-interacting moiety, whereas N,N,O and N,N,N,O donor atoms are linked to generate the metal chelation moiety. Furthermore, we went on exploring their multifunctionality with respect to (w.r.t.) (i) their metal chelating capacities and (ii) their utility to modulate the aggregation pathways of both metal-free and metal-bound amyloid-β, (iii) scavenge free radicals, and (iv) inhibit the activity of acetylcholinesterase and (v) cytotoxicity. Moreover, the compounds were able to sequester Cu2+ from the Aβ-Cu complex as studied by the UV-visible spectroscopic assay. Molecular docking studies were also performed with Aβ and acetylcholinesterase enzyme. Overall, the studies presented here qualify these molecules as promising candidates for further investigation in the quest for finding a treatment for Alzheimer's disease.
Collapse
Affiliation(s)
- Monika Rana
- Department of Chemistry, Central University of Rajasthan, Bandarsindri, Ajmer 305817, India
| | - Hong-Jun Cho
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Hemant Arya
- Department of Biotechnology, Central University of Rajasthan, Bandarsindri, Ajmer 305817, India
| | - Tarun Kumar Bhatt
- Department of Biotechnology, Central University of Rajasthan, Bandarsindri, Ajmer 305817, India
| | - Kishalay Bhar
- Department of Chemistry, Central University of Rajasthan, Bandarsindri, Ajmer 305817, India
| | - Surabhi Bhatt
- Department of Chemistry, Central University of Rajasthan, Bandarsindri, Ajmer 305817, India
| | - Liviu M Mirica
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Anuj Kumar Sharma
- Department of Chemistry, Central University of Rajasthan, Bandarsindri, Ajmer 305817, India
| |
Collapse
|
38
|
Ramesh M, Balachandra C, Andhare P, Govindaraju T. Rationally Designed Molecules Synergistically Modulate Multifaceted Aβ Toxicity, Microglial Activation, and Neuroinflammation. ACS Chem Neurosci 2022; 13:2209-2221. [PMID: 35759686 DOI: 10.1021/acschemneuro.2c00276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Synergistic modulation of multifaceted toxicity is the key to tackle multifactorial Alzheimer's disease (AD). The etiology of AD includes amyloid β (Aβ) amyloidosis, metal ion dyshomeostasis, reactive oxygen species (ROS), oxidative stress, mitochondrial damage, and neuroinflammation. We rationally designed multifunctional modulators by integrating pharmacophores for metal chelation, antioxidant and anti-inflammatory properties, and modulation of Aβ42 aggregation on the naphthalene monoimide (NMI) scaffold. The in vitro and cellular studies of NMIs revealed that M3 synergistically modulates metal-independent and -dependent amyloid toxicity, scavenges ROS, alleviates oxidative stress, and emulates Nrf2-mediated stress response in neuronal cells. M3 effectively reduced structural and functional damage of mitochondria, reduced Cyt c levels, and rescued cells from apoptosis. The biological atomic force microscopy and Western blot analysis revealed the ability of M3 to suppress microglial activation and neuroinflammation through inhibition of the NF-κβ pathway. The synergistic action of M3 is in agreement with our design strategy to develop a multifunctional therapeutic candidate by integrating multiple pharmacophores with distinct structural and functional elements to ameliorate the multifaceted toxicity of AD.
Collapse
Affiliation(s)
- Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru, Karnataka 560064, India
| | - Chenikkayala Balachandra
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru, Karnataka 560064, India
| | - Pradhnesh Andhare
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru, Karnataka 560064, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru, Karnataka 560064, India
| |
Collapse
|
39
|
Novel Rivastigmine Derivatives as Promising Multi-Target Compounds for Potential Treatment of Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10071510. [PMID: 35884815 PMCID: PMC9313321 DOI: 10.3390/biomedicines10071510] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 01/03/2023] Open
Abstract
Alzheimer’s disease (AD) is the most serious and prevalent neurodegenerative disorder still without cure. Since its aetiology is diverse, recent research on anti-AD drugs has been focused on multi-target compounds. In this work, seven novel hybrids (RIV–BIM) conjugating the active moiety of the drug rivastigmine (RIV) with 2 isomeric hydroxyphenylbenzimidazole (BIM) units were developed and studied. While RIV assures the inhibition of cholinesterases, BIM provides further appropriate properties, such as inhibition of amyloid β-peptide (Aβ) aggregation, antioxidation and metal chelation. The evaluated biological properties of these hybrids included antioxidant activity; inhibition of acetylcholinesterase (AChE), butyrylcholinesterase (BChE) and Aβ42 aggregation; as well as promotion of cell viability and neuroprotection. All the compounds are better inhibitors of AChE than rivastigmine (IC50 = 32.1 µM), but compounds of series 5 are better inhibitors of BChE (IC50 = 0.9−1.7 µM) than those of series 4. Series 5 also showed good capacity to inhibit self- (42.1−58.7%) and Cu(II)-induced (40.3−60.8%) Aβ aggregation and also to narrow (22.4−42.6%) amyloid fibrils, the relevant compounds being 5b and 5d. Some of these compounds can also prevent the toxicity induced in SH-SY5Y cells by Aβ42 and oxidative stress. Therefore, RIV–BIM hybrids seem to be potential drug candidates for AD with multi-target abilities.
Collapse
|
40
|
Assembly of redox active metallo-enzymes and metallo-peptides on electrodes: Abiological constructs to probe natural processes. Curr Opin Chem Biol 2022; 68:102142. [DOI: 10.1016/j.cbpa.2022.102142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 11/23/2022]
|
41
|
Abstract
Amyloid-β (Aβ) peptides are involved in Alzheimer's disease (AD) development. The interactions of these peptides with copper and zinc ions also seem to be crucial for this pathology. Although Cu(II) and Zn(II) ions binding by Aβ peptides has been scrupulously investigated, surprisingly, this phenomenon has not been so thoroughly elucidated for N-truncated Aβ4-x-probably the most common version of this biomolecule. This negligence also applies to mixed Cu-Zn complexes. From the structural in silico analysis presented in this work, it appears that there are two possible mixed Cu-Zn(Aβ4-x) complexes with different stoichiometries and, consequently, distinct properties. The Cu-Zn(Aβ4-x) complex with 1:1:1 stoichiometry may have a neuroprotective superoxide dismutase-like activity. On the other hand, another mixed 2:1:2 Cu-Zn(Aβ4-x) complex is perhaps a seed for toxic oligomers. Hence, this work proposes a novel research direction for our better understanding of AD development.
Collapse
|
42
|
Arrigoni F, Di Carlo C, Rovetta A, De Gioia L, Zampella G, Bertini L. Superoxide reduction by Cu‐Amyloid Beta peptide complexes. A Density Functional Theory study. Eur J Inorg Chem 2022. [DOI: 10.1002/ejic.202200245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Federica Arrigoni
- Università degli Studi di Milano-Bicocca: Universita degli Studi di Milano-Bicocca Biotecnology and Biosciences ITALY
| | - Chiara Di Carlo
- Università degli Studi di Milano-Bicocca: Universita degli Studi di Milano-Bicocca Biotecnology and Biosciences ITALY
| | - Alberto Rovetta
- University of Milano–Bicocca University Library: Universita degli Studi di Milano-Bicocca Biotecnology and Biosciences ITALY
| | - Luca De Gioia
- University of Milan–Bicocca: Universita degli Studi di Milano-Bicocca Biotecnology and Biosciences ITALY
| | - Giuseppe Zampella
- University of Milan–Bicocca: Universita degli Studi di Milano-Bicocca Biotecnology and Biosciences ITALY
| | - Luca Bertini
- Universita' degli studi di MIlano-Bicocca Biotecnologie e Bioscienze Piazza della Scienza 2 20127 Milano ITALY
| |
Collapse
|
43
|
Ghosh D, Konar M, Mondal T, Govindaraju T. Differential copper-guided architectures of amyloid β peptidomimetics modulate oxidation states and catalysis. NANOSCALE ADVANCES 2022; 4:2196-2200. [PMID: 36133442 PMCID: PMC9419866 DOI: 10.1039/d2na00161f] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/30/2022] [Indexed: 06/16/2023]
Abstract
Orchestration of differential architectures of designer peptidomimetics that modulate metal oxidation states to perform multiple chemical transformations remains a challenge. Cu-chelation and self-assembly properties of amyloid β (Aβ14-23) peptide were tuned by the incorporation of cyclic dipeptide (CDP) and pyrene (Py) as the assembly directing and reporting units, respectively. We explore the molecular architectonics of Aβ14-23 derived peptidomimetics (AkdNMCPy) to form differential architectures that stabilize distinct Cu oxidation states. The fibrillar self-assembly of AkdNMCPy is modulated to form nanosheets by the one-off addition of CuII. Notably, the serial addition of CuII resulted in the formation of micelle-like core-shell architectures. The micelle-like and nanosheet architectures were found to differentially stabilize CuII and CuI states and catalyze tandem oxidative-hydrolysis and alkyne-azide cycloaddition reactions, respectively.
Collapse
Affiliation(s)
- Debasis Ghosh
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru 560064 Karnataka India
| | - Mouli Konar
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru 560064 Karnataka India
| | - Tanmay Mondal
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru 560064 Karnataka India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru 560064 Karnataka India
| |
Collapse
|
44
|
The kinetic mechanism of cations induced protein nanotubes self-assembly and their application as delivery system. Biomaterials 2022; 286:121600. [DOI: 10.1016/j.biomaterials.2022.121600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/23/2022] [Accepted: 05/20/2022] [Indexed: 11/23/2022]
|
45
|
Bioactive Compounds and Their Derivatives: An Insight into Prospective Phytotherapeutic Approach against Alzheimer’s Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5100904. [PMID: 35450410 PMCID: PMC9017558 DOI: 10.1155/2022/5100904] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/24/2022] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative brain disorder that causes cellular response alterations, such as impaired cholinergic mechanism, amyloid-beta (Aβ) AD aggregation, neuroinflammation, and several other pathways. AD is still the most prevalent form of dementia and affects many individuals across the globe. The exact cause of the disorder is obscure. There are yet no effective medications for halting, preventing, or curing AD's progress. Plenty of natural products are isolated from several sources and analyzed in preclinical and clinical settings for neuroprotective effects in preventing and treating AD. In addition, natural products and their derivatives have been promising in treating and preventing AD. Natural bioactive compounds play an active modulatory role in the pathological molecular mechanisms of AD development. This review focuses on natural products from plant sources and their derivatives that have demonstrated neuroprotective activities and maybe promising to treat and prevent AD. In addition, this article summarizes the literature pertaining to natural products as agents in the treatment of AD. Rapid metabolism, nonspecific targeting, low solubility, lack of BBB permeability, and limited bioavailability are shortcomings of most bioactive molecules in treating AD. We can use nanotechnology and nanocarriers based on different types of approaches.
Collapse
|
46
|
Dey C, Roy M, Dey SG. Insights from Self-Assembled Aggregates of Amyloid β Peptides on Gold Surfaces. ACS OMEGA 2022; 7:9973-9983. [PMID: 35382274 PMCID: PMC8973063 DOI: 10.1021/acsomega.1c06056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/24/2022] [Indexed: 06/12/2023]
Abstract
Amyloid β (Aβ) peptides mutated at different positions using a cysteine moiety assemble on Au electrodes using the thiol functionality of cysteine. Self-assembled monolayers (SAMs) of Aβ on Au surfaces can act as abiological platforms that allow the mimicking of fibrils and oligomeric Aβ via the formation of controlled large and small peptide aggregates. These Aβ constructs bind with heme and Cu and exhibit different reactivities. These abiological platforms can also be used to investigate potential drugs that can interact with heme and Cu-Aβ. SAM formation of Aβ mutants allows the study of different morphology and structure as well as behavior changes on binding with different metals and cytochrome c (Cyt c). This review provides a detailed insight into the structure and reactivities of various Aβ aggregated on Au electrodes mimicking the cell membrane.
Collapse
|
47
|
Sánchez JM, Carratalá JV, Serna N, Unzueta U, Nolan V, Sánchez-Chardi A, Voltà-Durán E, López-Laguna H, Ferrer-Miralles N, Villaverde A, Vazquez E. The Poly-Histidine Tag H6 Mediates Structural and Functional Properties of Disintegrating, Protein-Releasing Inclusion Bodies. Pharmaceutics 2022; 14:pharmaceutics14030602. [PMID: 35335976 PMCID: PMC8955739 DOI: 10.3390/pharmaceutics14030602] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022] Open
Abstract
The coordination between histidine-rich peptides and divalent cations supports the formation of nano- and micro-scale protein biomaterials, including toxic and non-toxic functional amyloids, which can be adapted as drug delivery systems. Among them, inclusion bodies (IBs) formed in recombinant bacteria have shown promise as protein depots for time-sustained protein release. We have demonstrated here that the hexahistidine (H6) tag, fused to recombinant proteins, impacts both on the formation of bacterial IBs and on the conformation of the IB-forming protein, which shows a higher content of cross-beta intermolecular interactions in H6-tagged versions. Additionally, the addition of EDTA during the spontaneous disintegration of isolated IBs largely affects the protein leakage rate, again protein release being stimulated in His-tagged materials. This event depends on the number of His residues but irrespective of the location of the tag in the protein, as it occurs in either C-tagged or N-tagged proteins. The architectonic role of H6 in the formation of bacterial IBs, probably through coordination with divalent cations, offers an easy approach to manipulate protein leakage and to tailor the applicability of this material as a secretory amyloidal depot in different biomedical interfaces. In addition, the findings also offer a model to finely investigate, in a simple set-up, the mechanics of protein release from functional secretory amyloids.
Collapse
Affiliation(s)
- Julieta María Sánchez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain; (J.M.S.); (J.V.C.); (N.S.); (E.V.-D.); (H.L.-L.); (N.F.-M.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain
- Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT), CONICET-Universidad Nacional de Córdoba, ICTA & Cátedra de Química Biológica, Departamento de Química, FCEFyN, UNC. Av. Velez Sarsfield 1611, Córdoba X 5016GCA, Argentina;
| | - José Vicente Carratalá
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain; (J.M.S.); (J.V.C.); (N.S.); (E.V.-D.); (H.L.-L.); (N.F.-M.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Naroa Serna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain; (J.M.S.); (J.V.C.); (N.S.); (E.V.-D.); (H.L.-L.); (N.F.-M.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Ugutz Unzueta
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain
- Biomedical Research Institute Sant Pau (IIB Sant Pau), Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, 08025 Barcelona, Spain
| | - Verónica Nolan
- Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT), CONICET-Universidad Nacional de Córdoba, ICTA & Cátedra de Química Biológica, Departamento de Química, FCEFyN, UNC. Av. Velez Sarsfield 1611, Córdoba X 5016GCA, Argentina;
| | - Alejandro Sánchez-Chardi
- Servei de Microscòpia, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain;
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain
| | - Eric Voltà-Durán
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain; (J.M.S.); (J.V.C.); (N.S.); (E.V.-D.); (H.L.-L.); (N.F.-M.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Hèctor López-Laguna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain; (J.M.S.); (J.V.C.); (N.S.); (E.V.-D.); (H.L.-L.); (N.F.-M.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Neus Ferrer-Miralles
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain; (J.M.S.); (J.V.C.); (N.S.); (E.V.-D.); (H.L.-L.); (N.F.-M.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain; (J.M.S.); (J.V.C.); (N.S.); (E.V.-D.); (H.L.-L.); (N.F.-M.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain
- Correspondence: (A.V.); (E.V.)
| | - Esther Vazquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain; (J.M.S.); (J.V.C.); (N.S.); (E.V.-D.); (H.L.-L.); (N.F.-M.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Plaça Cívica s/n, Bellaterra, 08193 Barcelona, Spain;
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), C/ Monforte de Lemos 3-5, 28029 Madrid, Spain
- Correspondence: (A.V.); (E.V.)
| |
Collapse
|
48
|
Sciacca MF, Naletova I, Giuffrida ML, Attanasio F. Semax, a Synthetic Regulatory Peptide, Affects Copper-Induced Abeta Aggregation and Amyloid Formation in Artificial Membrane Models. ACS Chem Neurosci 2022; 13:486-496. [PMID: 35080861 PMCID: PMC8855339 DOI: 10.1021/acschemneuro.1c00707] [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] [Indexed: 12/02/2022] Open
Abstract
![]()
Alzheimer’s
disease, the most common form of dementia, is
characterized by the aggregation of amyloid beta protein (Aβ).
The aggregation and toxicity of Aβ are strongly modulated by
metal ions and phospholipidic membranes. In particular, Cu2+ ions play a pivotal role in modulating Aβ aggregation. Although
in the last decades several natural or synthetic compounds were evaluated
as candidate drugs, to date, no treatments are available for the pathology.
Multifunctional compounds able to both inhibit fibrillogenesis, and
in particular the formation of oligomeric species, and prevent the
formation of the Aβ:Cu2+ complex are of particular
interest. Here we tested the anti-aggregating properties of a heptapeptide,
Semax, an ACTH-like peptide, which is known to form a stable complex
with Cu2+ ions and has been proven to have neuroprotective
and nootropic effects. We demonstrated through a combination of spectrofluorometric,
calorimetric, and MTT assays that Semax not only is able to prevent
the formation of Aβ:Cu2+ complexes but also has anti-aggregating
and protective properties especially in the presence of Cu2+. The results suggest that Semax inhibits fiber formation by interfering
with the fibrillogenesis of Aβ:Cu2+ complexes.
Collapse
Affiliation(s)
- Michele F.M. Sciacca
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Irina Naletova
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Maria Laura Giuffrida
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Francesco Attanasio
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| |
Collapse
|
49
|
Imidazolylacetophenone oxime-based multifunctional neuroprotective agents: Discovery and structure-activity relationships. Eur J Med Chem 2022; 228:114031. [PMID: 34875520 DOI: 10.1016/j.ejmech.2021.114031] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/25/2021] [Accepted: 11/27/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) possesses a complex pathogenetic mechanism. Nowadays, multitarget agents are considered to have potential in effectively treating AD via triggering molecules in functionally complementary pathways at the same time. Here, based on the screening (∼1400 compounds) against neuroinflammation, an imidazolylacetophenone oxime ether (IOE) was discovered as a novel hit. In order to obtain SARs, a series of imidazolylacetophenone oxime derivatives were constructed, and their C=N bonds were confirmed as the Z configuration by single crystals. These derivatives exhibited potential multifunctional neuroprotective effects including anti-neuroinflammatory, antioxidative damage, metal-chelating, inhibition of acetylcholinesterase (AChE) properties. Among these derivatives, compound 12i displayed the most potent inhibitory activity against nitric oxide (NO) production with EC50 value of 0.57 μM 12i can dose-dependently suppress the expression of iNOS and COX-2 but not change the expression of HO-1 protein. Moreover, 12i exhibited evidently neuroprotective effects on H2O2-induced PC12 cells damage and ferroptosis without cytotoxicity at 10 μM, as well as selectively metal chelating properties via chelating Cu2+. In addition, 12i showed a mixed-type inhibitory effect on AChE in vitro. The structure-activity relationships (SARs) analysis indicated that dioxolane groups on benzene ring and rigid oxime ester can improve the activity. Parallel artificial membrane permeation assay (PAMPA) also verified that 12i can overcome the blood-brain barrier (BBB). Overall, this is the first report on imidazolylacetophenone oxime-based multifunctional neuroprotective effects, suggesting that this type of compounds might be novel multifunctional agents against AD.
Collapse
|
50
|
Probable Reasons for Neuron Copper Deficiency in the Brain of Patients with Alzheimer’s Disease: The Complex Role of Amyloid. INORGANICS 2022. [DOI: 10.3390/inorganics10010006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease is a progressive neurodegenerative disorder that eventually leads the affected patients to die. The appearance of senile plaques in the brains of Alzheimer’s patients is known as a main symptom of this disease. The plaques consist of different components, and according to numerous reports, their main components include beta-amyloid peptide and transition metals such as copper. In this disease, metal dyshomeostasis leads the number of copper ions to simultaneously increase in the plaques and decrease in neurons. Copper ions are essential for proper brain functioning, and one of the possible mechanisms of neuronal death in Alzheimer’s disease is the copper depletion of neurons. However, the reason for the copper depletion is as yet unknown. Based on the available evidence, we suggest two possible reasons: the first is copper released from neurons (along with beta-amyloid peptides), which is deposited outside the neurons, and the second is the uptake of copper ions by activated microglia.
Collapse
|