1
|
Zaitseva NV, Zemlyanova МА, Gekht AB, Dedaev SI, Kol'dibekova YV, Peskova ЕV, Stepankov МS, Tinkov AA, Martins AC, Skalny AV, Aschner M. Neurotoxic effects of aluminum and manganese: From molecular to clinical effects. J Neurol Sci 2025; 473:123480. [PMID: 40233648 DOI: 10.1016/j.jns.2025.123480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 02/25/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025]
Abstract
The existing data demonstrate that aluminum (Al) and manganese (Mn) possess neurotoxic effects upon overexposure due to induction of neuronal oxidative stress and apoptosis, synaptic dysfunction and neurotransmitter metabolism, neuroinflammation, and cytoskeletal pathology. However, systematic evidence regarding contribution of these metals to development of neurological diseases are lacking. Therefore, in this review we provide a summary of the existing data on contribution of Al and Mn exposure to brain diseases and its symptoms. Causal relations were demonstrated for development of parkinsonism upon exposure to high doses of Mn, whereas Al overload is considered the key contributor to dialysis encephalopathy. Certain studies demonstrate that Al and Mn overexposure is associated with neurodegenerative diseases including Alzheimer's and Parkinson's diseases, as well as neurodevelopmental disorders like autism spectrum disorder (ASD) and attention-deficit/hyperactivity disorder (ADHD). Although laboratory studies demonstrate the potential contribution of Al and Mn to molecular pathogenesis of these diseases, clinical findings supporting the causal role of metals is these pathologies are yet insufficient. Therefore, estimation of the contribution of these metals to neurological disorders is essential for development of more effective early diagnostics and prevention of diseases under exposure to adverse neurological effects of Al and Mn compounds.
Collapse
Affiliation(s)
- N V Zaitseva
- Federal Scientific Center for Medical and Preventive Health Risk Management Technologies, Perm, Russian Federation.
| | - М А Zemlyanova
- Federal Scientific Center for Medical and Preventive Health Risk Management Technologies, Perm, Russian Federation
| | - A B Gekht
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russian Federation; Pirogov Russian Medical Research University, Moscow, Russian Federation
| | - S I Dedaev
- Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russian Federation
| | - Yu V Kol'dibekova
- Federal Scientific Center for Medical and Preventive Health Risk Management Technologies, Perm, Russian Federation
| | - Е V Peskova
- Federal Scientific Center for Medical and Preventive Health Risk Management Technologies, Perm, Russian Federation
| | - М S Stepankov
- Federal Scientific Center for Medical and Preventive Health Risk Management Technologies, Perm, Russian Federation
| | - Alexey A Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl 150003, Russian Federation; Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russian Federation
| | - Airton C Martins
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Anatoly V Skalny
- Center of Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow 119435, Russian Federation; Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow 117198, Russian Federation
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
2
|
Cavaleri F. Reevaluating the role of amyloid β-peptides in Alzheimer's disease: from pathogenic agents to protective chelation mechanisms. Front Neurol 2025; 16:1550709. [PMID: 40224312 PMCID: PMC11987711 DOI: 10.3389/fneur.2025.1550709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/11/2025] [Indexed: 04/15/2025] Open
Abstract
Alzheimer's disease (AD) is a multifaceted neurodegenerative disorder with complex etiology, often associated with histological markers of oxidative stress, inflammation, and disturbances in calcium homeostasis. Traditionally, amyloid β-peptides (Aβ) have been considered key contributors to these pathological processes. However, emerging evidence suggests a protective role for Aβ and the enzymes involved in its production. This article further explores the hypothesis published by us a decade before that posits amyloid β-peptides and the β-secretase enzyme (BACE1) are part of an intentionally designed cellular defense mechanism against metal toxicity. This challenges the conventional understanding of their roles in AD pathogenesis. It is not until this BACE1 system, primarily the associated amyloid plaque deposit sites, are saturated with heavy and other metals and the exposure to these cations continues to influx oxidative ions into the brain, do the indications of neurodegeneration begin to become symptomatic. Until this metal oversaturation takes place, the system - Aβ and the enzymes involved in its production and conveyance - keeps the oxidative potential of the metal toxins sequestered extracellularly and out of the way of the neuron's intracellular activities.
Collapse
|
3
|
Duță C, Muscurel C, Dogaru CB, Stoian I. Selenoproteins: Zoom-In to Their Metal-Binding Properties in Neurodegenerative Diseases. Int J Mol Sci 2025; 26:1305. [PMID: 39941073 PMCID: PMC11818150 DOI: 10.3390/ijms26031305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/30/2025] [Accepted: 02/02/2025] [Indexed: 02/16/2025] Open
Abstract
Selenoproteins contain selenium (Se), which is included in the 21st proteinogenic amino acid selenocysteine (Sec). Selenium (Se) is an essential trace element that exerts its biological actions mainly through selenoproteins. Selenoproteins have crucial roles in maintaining healthy brain activity. At the same time, brain-function-associated selenoproteins may also be involved in neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD). The selenoproteins GPx4 (glutathione peroxidase 4), GPx1 (glutathione peroxidase 1), SELENOP (selenoprotein P), SELENOK (selenoprotein K), SELENOS (selenoprotein S), SELENOW (selenoprotein W), and SELENOT (selenoprotein T) are highly expressed, specifically in AD-related brain regions being closely correlated to brain function. Only a few selenoproteins, mentioned above (especially SELENOP), can bind transition and heavy metals. Metal ion homeostasis accomplishes the vital physiological function of the brain. Dyshomeostasis of these metals induces and entertains neurodegenerative diseases. In this review, we described some of the proposed and established mechanisms underlying the actions and properties of the above-mentioned selenoproteins having the characteristic feature of binding transition or heavy metals.
Collapse
Affiliation(s)
| | | | - Carmen Beatrice Dogaru
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (C.D.); (C.M.); (I.S.)
| | | |
Collapse
|
4
|
Streit WJ, Phan L, Bechmann I. Ferroptosis and pathogenesis of neuritic plaques in Alzheimer disease. Pharmacol Rev 2025; 77:100005. [PMID: 39952690 DOI: 10.1124/pharmrev.123.000823] [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: 12/14/2023] [Revised: 07/25/2024] [Accepted: 09/13/2024] [Indexed: 10/09/2024] Open
Abstract
Neuritic plaques are pathognomonic and terminal lesions of Alzheimer disease (AD). They embody AD pathogenesis because they harbor in one space critical pathologic features of the disease: amyloid deposits, neurofibrillary degeneration, neuroinflammation, and iron accumulation. Neuritic plaques are thought to arise from the conversion of diffuse extracellular deposits of amyloid-β protein (Aβ), and it is believed that during conversion, amyloid toxicity creates the dystrophic neurites of neuritic plaques, as well as neurofibrillary tangles However, recent evidence from human postmortem studies suggests a much different mechanism of neuritic plaque formation, where the first step in their creation is neuronal degeneration driven by iron overload and ferroptosis. Similarly, neurofibrillary tangles represent the corpses of iron-laden neurons that develop independently of Aβ deposits. In this review, we will focus on the role of free redox-active iron in the development of typical AD pathology, as determined largely by evidence obtained in the human temporal lobe during early, preclinical stages of AD. The findings have allowed the construction of a scheme of AD pathogenesis where brain iron is center stage and is involved in every step of the sequence of events that produce characteristic AD pathology. We will discuss how the study of preclinical AD has produced a fresh and revised assessment of AD pathogenesis that may be important for reconsidering current therapeutic efforts and guiding future ones. SIGNIFICANCE STATEMENT: This review offers a novel perspective on Alzheimer disease pathogenesis where elevated brain iron plays a central role and is involved throughout the development of lesions. Herein, we review arguments against the amyloid cascade theory and explain how recent findings in humans during early preclinical disease support iron-mediated cell death and endogenous iron containment mechanisms as critical components of neuritic plaque formation and ensuing dementia.
Collapse
Affiliation(s)
- Wolfgang J Streit
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida.
| | - Leah Phan
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| |
Collapse
|
5
|
Cendron A, Chianese M, Zarzycki K, Ruzza P, Honisch C, Brasuń J, Carraro M. Chelating Properties of N 6O-Donors Toward Cu(II) Ions: Speciation in Aqueous Environments and Catalytic Activity of the Dinuclear Complexes. Molecules 2024; 29:5708. [PMID: 39683868 DOI: 10.3390/molecules29235708] [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: 10/30/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
This study focuses on the use of three isostructural N6O donor ligands, specifically known to form complexes with copper ions, to chelate Cu(II) from aqueous solutions. The corresponding Cu(II) complexes feature a dinuclear copper core mimicking the active site of natural superoxide dismutase (SOD) enzymes while also creating a coordination environment favorable for catalase (CAT) activity, being thus appealing as catalytic antioxidant systems. Given the critical role of copper dysregulation in the pathophysiology of Alzheimer's disease (AD), these complexes may help mitigate the harmful effects of free Cu(II) ions: the goal is to transform copper's reactive oxygen species (ROS)-generating properties into beneficial ROS-scavenging action. This study investigates the speciation, chelating efficiency, and metal selectivity of these ligands, as well as the antioxidant activity of the resulting complexes under aqueous and physiologically relevant conditions. Additionally, the ligands, equipped with functional groups for attaching targeting moieties, are conjugated with a small peptide that may act as an anti-aggregating agent of β-amyloid peptides, aiming to develop a multifunctional therapeutic strategy against Alzheimer's disease.
Collapse
Affiliation(s)
- Andrea Cendron
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Martina Chianese
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Kamil Zarzycki
- Biomolecule Student Science Club, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Paolo Ruzza
- Institute of Biomolecular Chemistry of CNR (ICB-CNR), Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Claudia Honisch
- Institute of Biomolecular Chemistry of CNR (ICB-CNR), Padova, Via Marzolo 1, 35131 Padova, Italy
| | - Justyna Brasuń
- Department of the Basic Chemical Sciences, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Mauro Carraro
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, 35131 Padova, Italy
- Institute on Membrane Technology of CNR (ITM-CNR), Unit of Padova, Via Marzolo 1, 35131 Padova, Italy
| |
Collapse
|
6
|
Das V, Miller JH, Alladi CG, Annadurai N, De Sanctis JB, Hrubá L, Hajdúch M. Antineoplastics for treating Alzheimer's disease and dementia: Evidence from preclinical and observational studies. Med Res Rev 2024; 44:2078-2111. [PMID: 38530106 DOI: 10.1002/med.22033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024]
Abstract
As the world population ages, there will be an increasing need for effective therapies for aging-associated neurodegenerative disorders, which remain untreatable. Dementia due to Alzheimer's disease (AD) is one of the leading neurological diseases in the aging population. Current therapeutic approaches to treat this disorder are solely symptomatic, making the need for new molecular entities acting on the causes of the disease extremely urgent. One of the potential solutions is to use compounds that are already in the market. The structures have known pharmacokinetics, pharmacodynamics, toxicity profiles, and patient data available in several countries. Several drugs have been used successfully to treat diseases different from their original purposes, such as autoimmunity and peripheral inflammation. Herein, we divulge the repurposing of drugs in the area of neurodegenerative diseases, focusing on the therapeutic potential of antineoplastics to treat dementia due to AD and dementia. We briefly touch upon the shared pathological mechanism between AD and cancer and drug repurposing strategies, with a focus on artificial intelligence. Next, we bring out the current status of research on the development of drugs, provide supporting evidence from retrospective, clinical, and preclinical studies on antineoplastic use, and bring in new areas, such as repurposing drugs for the prion-like spreading of pathologies in treating AD.
Collapse
Affiliation(s)
- Viswanath Das
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - John H Miller
- School of Biological Sciences and Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Charanraj Goud Alladi
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Narendran Annadurai
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - Lenka Hrubá
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| |
Collapse
|
7
|
Mirdha L. Aggregation Behavior of Amyloid Beta Peptide Depends Upon the Membrane Lipid Composition. J Membr Biol 2024; 257:151-164. [PMID: 38888760 DOI: 10.1007/s00232-024-00314-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024]
Abstract
Protein aggregation plays a crucial role in the development of several neurodegenerative diseases. It is important to understand the aggregation process for the detection of the onset of these diseases. Alzheimer's Disease (AD) is one of the most prevalent neurodegenerative diseases caused by the aggregation of Aβ-40 and Aβ-42 peptides. The smaller oligomers lead to the formation of protein plaque at the neural membranes leading to memory loss and other disorders. Interestingly, aggregation takes place at the neural membranes, therefore the membrane composition seems to play an important role in the aggregation process. Despite a large number of literatures on the effect of lipid composition on protein aggregation, there are very few concise reviews that highlight the role of membrane composition in protein aggregation. In this review, we have discussed the implication of membrane composition on the aggregation of amyloid beta peptide with a special emphasis on cholesterol. We have further discussed the role of the degree of unsaturation of fatty acids and the participation of apolipoprotein E4 (ApoE4) in the onset of AD.
Collapse
Affiliation(s)
- Lipika Mirdha
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla, Odisha, 768 019, India.
| |
Collapse
|
8
|
Martín LR, Santiago LR, Korendovych IV, Sodupe M, Maréchal JD. Computational modelling of supramolecular metallopeptide assemblies. Methods Enzymol 2024; 697:211-245. [PMID: 38816124 DOI: 10.1016/bs.mie.2024.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Among the important questions in supramolecular peptide self-assemblies are their interactions with metallic compounds and ions. In the last decade, intensive efforts have been devoted to understanding the structural properties of these interactions including their dynamical and catalytic impact in natural and de novo systems. Since structural insights from experimental approaches could be particularly challenging, computational chemistry methods are interesting complementary tools. Here, we present the general multiscale strategies we developed and applied for the study of metallopeptide assemblies. These strategies include prediction of metal binding site, docking of metallic moieties, classical and accelerated molecular dynamics and finally QM/MM calculations. The systems of choice for this chapter are, on one side, peptides involved in neurodegenerative diseases and, on the other, de novo fibrillar systems with catalytic properties. Both successes and remaining challenges are highlighted so that the protocol could be apply to other system of this kind.
Collapse
Affiliation(s)
| | | | - Ivan V Korendovych
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, United States
| | - Mariona Sodupe
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| | | |
Collapse
|
9
|
Rodak K, Bęben D, Birska M, Siwiela O, Kokot I, Moreira H, Radajewska A, Szyjka A, Kratz EM. Evaluating the Neuroprotective Potential of Caffeinated Coffee in the Context of Aluminum-Induced Neurotoxicity: Insights from a PC12 Cell Culture Model. Antioxidants (Basel) 2024; 13:342. [PMID: 38539875 PMCID: PMC10968250 DOI: 10.3390/antiox13030342] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/26/2024] [Accepted: 03/11/2024] [Indexed: 05/02/2025] Open
Abstract
Exposure to aluminum (Al) and its compounds is an environmental factor that induces neurotoxicity, partially through oxidative stress, potentially leading to the development of neurodegenerative diseases. Components of the diet, such as caffeinated coffee, may play a significant role in preventing these diseases. In the present study, an experimental model of PC12 cells (rat pheochromocytoma tumor cells) was developed to investigate the influence of caffeine and caffeinated coffee on neurotoxicity induced by Al compounds and/or oxidative stress. For the induction of neurotoxicity, aluminum maltolate (Almal) and H2O2 were used. The present study demonstrates that 100 μM Almal reduced cell survival, while caffeinated coffee with caffeine concentrations of 5 μg/mL and 80 μg/mL reversed this effect, resulting in a higher than fivefold increase in PC12 cell survival. However, despite the observed antioxidant properties typical for caffeine and caffeinated coffee, it is unlikely that they are the key factors contributing to cell protection against neurotoxicity induced by both oxidative stress and Al exposure. Moreover, the present study reveals that for coffee to exert its effects, it is possible that Al must first activate certain mechanisms within the cell. Therefore, various signaling pathways are discussed, and modifications of these pathways might significantly decrease the risk of Al-induced neurotoxicity.
Collapse
Affiliation(s)
- Kamil Rodak
- Department of Laboratory Diagnostics, Division of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wroclaw, Poland;
- Student Research Club, “Biomarkers in Medical Diagnostics”, Department of Laboratory Diagnostics, Division of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wroclaw, Poland
| | - Dorota Bęben
- Student Research Club of Flow Cytometry and Biomedical Research, Department of Basic Medical Sciences and Immunology, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211, 50-556 Wroclaw, Poland; (D.B.); (M.B.); (O.S.)
| | - Monika Birska
- Student Research Club of Flow Cytometry and Biomedical Research, Department of Basic Medical Sciences and Immunology, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211, 50-556 Wroclaw, Poland; (D.B.); (M.B.); (O.S.)
| | - Oliwia Siwiela
- Student Research Club of Flow Cytometry and Biomedical Research, Department of Basic Medical Sciences and Immunology, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211, 50-556 Wroclaw, Poland; (D.B.); (M.B.); (O.S.)
| | - Izabela Kokot
- Department of Laboratory Diagnostics, Division of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wroclaw, Poland;
| | - Helena Moreira
- Department of Basic Medical Sciences and Immunology, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211, 50-556 Wroclaw, Poland; (H.M.); (A.S.)
| | - Anna Radajewska
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Hematology, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wroclaw, Poland;
| | - Anna Szyjka
- Department of Basic Medical Sciences and Immunology, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211, 50-556 Wroclaw, Poland; (H.M.); (A.S.)
| | - Ewa Maria Kratz
- Department of Laboratory Diagnostics, Division of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wroclaw, Poland;
| |
Collapse
|
10
|
Roldán-Martín L, Sodupe M, Maréchal JD. Computational Study of Amyloidβ 42 Familial Mutations and Metal Interaction: Impact on Monomers and Aggregates Dynamical Behaviors. Inorg Chem 2024; 63:4725-4737. [PMID: 38408469 PMCID: PMC10934806 DOI: 10.1021/acs.inorgchem.3c04555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/02/2024] [Accepted: 02/02/2024] [Indexed: 02/28/2024]
Abstract
One of the main hallmarks of Alzheimer's Disease is the formation of β-amyloid plaques, whose formation may be enhanced by metal binding or the appearance of familial mutations. In the present study, the simultaneous effect of familial mutations (E22Q, E22G, E22K, and D23N) and binding to metal ions (Cu(II) or Al(III)) is studied at the Aβ42 monomeric and fibrillar levels. With the application of GaMD and MD simulations, it is observed that the effects of metal binding and mutations differ in the monomeric and fibrillar forms. In the monomeric structures, without metal binding, all mutations reduce the amount of α-helix and increase, in some cases, the β-sheet content. In the presence of Cu(II) and Al(III) metal ions, the peptide becomes less flexible, and the β-sheet content decreases in favor of forming α-helix motifs that stabilize the system through interhelical contacts. Regarding the fibrillar structures, mutations decrease the opening of the fiber in the vertical axis, thereby stabilizing the S-shaped structure of the fiber. This effect is, in general, enhanced upon metal binding. These results may explain the different Aβ42 aggregation patterns observed in familial mutations.
Collapse
Affiliation(s)
- Lorena Roldán-Martín
- Departament de Química, Universitat Autònoma de Barcelona, 08193 Cerdanyola
del Vallès, Spain
| | - Mariona Sodupe
- Departament de Química, Universitat Autònoma de Barcelona, 08193 Cerdanyola
del Vallès, Spain
| | - Jean-Didier Maréchal
- Departament de Química, Universitat Autònoma de Barcelona, 08193 Cerdanyola
del Vallès, Spain
| |
Collapse
|
11
|
Kazmi I, Afzal M, Imam F, Alzarea SI, Patil S, Mhaiskar A, Shah U, Almalki WH. Barbaloin's Chemical Intervention in Aluminum Chloride Induced Cognitive Deficits and Changes in Rats through Modulation of Oxidative Stress, Cytokines, and BDNF Expression. ACS OMEGA 2024; 9:6976-6985. [PMID: 38371830 PMCID: PMC10870395 DOI: 10.1021/acsomega.3c08791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/16/2023] [Accepted: 01/05/2024] [Indexed: 02/20/2024]
Abstract
Alzheimer's disease (AD) is a long-term neurodegenerative condition characterized by impaired cognitive functions, particularly in the domains of learning and memory. Finding promising options for AD can be successful with a medication repurposing strategy. The goal of the research was to examine the neuroprotective characteristics of barbaloin in aluminum chloride (AlCl3)-induced cognitive deficits and changes in rats through modulation of oxidative stress, cytokines, and brain-derived neurotrophic factor (BDNF) expression. Thirty male Wistar rats were subjected to AlCl3 at a dosage of 100 mg/kg via the per oral route (p.o.), which induced cognitive decline. Morris water maze (MWM) is used to assess behavioral metrics. Assays for catalase (CAT), malondialdehyde (MDA), reduced glutathione (GSH), acetylcholinesterase (AChE), choline-acetyltransferase (ChAT), interleukins-1β (IL-1β), superoxide dismutase (SOD), tumor necrosis factor-α (TNF-α), nuclear factor kappa-B (NF-κB), interleukins-6 (IL-6), BDNF, and neurotransmitter levels [dopamine (DA), acetylcholine (Ach), and γ-aminobutyric acid (GABA)] were performed. Results: The transfer latency time was notably decreased, and substantial modifications in the concentrations of GSH, MDA, CAT, SOD, AChE, ChAT and observed modulations in the formation of interleukins-6 (IL-6), TNF-α, IL-1β, BDNF, and NF-κB were also evidenced after the treatment of rats with barbaloin in comparison to AlCl3-induced control groups. Significant alterations in neurotransmitter levels (DA, Ach, and GABA) were also seen in barbaloin-treated groups in comparison to AlCl3-induced groups. The current investigation has provided evidence that the administration of barbaloin yielded notable enhancements in cognitive function in rats through the inhibition of MDA, enhancing endogenous antioxidant enzymes, reduction of cytokine levels, and enhancement of neurotransmitter contents in the brain. These effects were observed in comparison to a control group treated with AlCl3 and can be attributable to barbaloin's strong anti-inflammatory and antioxidant properties, and metal chelating properties may contribute to its neuroprotective effects. Barbaloin may also promote neuronal survival and enhance learning and memory by upregulating the expression of BDNF.
Collapse
Affiliation(s)
- Imran Kazmi
- Department
of Biochemistry, Faculty of Sciences, King
Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Muhammad Afzal
- Department
of Pharmaceutical Sciences, Pharmacy Program,
Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Faisal Imam
- Department
of Pharmacology and Toxicology, College
of Pharmacy, King Saud University, P.O.
Box 2457, Riyadh 11451, Saudi Arabia
| | - Sami I. Alzarea
- Department
of Pharmacology, College of Pharmacy, Jouf
University, Aljouf, Sakaka 72341, Saudi Arabia
| | - Shaktipal Patil
- Department
of Pharmacology, H. R. Patel Institute of
Pharmaceutical Education and Research, Karwand naka, Shirpur 425405, Maharashtra, India
- Department
of Chemistry, Nootan Pharmacy College, Sankalchand
Patel University, Visnagar 384315, Gujarat, India
| | - Amrapali Mhaiskar
- Department
of Pharmacology, R. C. Patel Institute of
Pharmaceutical Education and Research, Karwand naka, Shirpur 425405, Maharashtra, India
| | - Ujashkumar Shah
- Department
of Chemistry, Nootan Pharmacy College, Sankalchand
Patel University, Visnagar 384315, Gujarat, India
| | - Waleed Hassan Almalki
- Department
of Pharmacology, College of Pharmacy, Umm
Al-Qura University, Makkah 21955, Saudi Arabia
| |
Collapse
|
12
|
Korde DS, Humpel C. A Combination of Heavy Metals and Intracellular Pathway Modulators Induces Alzheimer Disease-like Pathologies in Organotypic Brain Slices. Biomolecules 2024; 14:165. [PMID: 38397402 PMCID: PMC10887098 DOI: 10.3390/biom14020165] [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: 12/06/2023] [Revised: 01/17/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by amyloid-beta (Aβ) plaques and tau neurofibrillary tangles (NFT). Modelling aspects of AD is challenging due to its complex multifactorial etiology and pathology. The present study aims to establish a cost-effective and rapid method to model the two primary pathologies in organotypic brain slices. Coronal hippocampal brain slices (150 µm) were generated from postnatal (day 8-10) C57BL6 wild-type mice and cultured for 9 weeks. Collagen hydrogels containing either an empty load or a mixture of human Aβ42 and P301S aggregated tau were applied to the slices. The media was further supplemented with various intracellular pathway modulators or heavy metals to augment the appearance of Aβ plaques and tau NFTs, as assessed by immunohistochemistry. Immunoreactivity for Aβ and tau was significantly increased in the ventral areas in slices with a mixture of human Aβ42 and P301S aggregated tau compared to slices with empty hydrogels. Aβ plaque- and tau NFT-like pathologies could be induced independently in slices. Heavy metals (aluminum, lead, cadmium) potently augmented Aβ plaque-like pathology, which developed intracellularly prior to cell death. Intracellular pathway modulators (scopolamine, wortmannin, MHY1485) significantly boosted tau NFT-like pathologies. A combination of nanomolar concentrations of scopolamine, wortmannin, MHY1485, lead, and cadmium in the media strongly increased Aβ plaque- and tau NFT-like immunoreactivity in ventral areas compared to the slices with non-supplemented media. The results highlight that we could harness the potential of the collagen hydrogel-based spreading of human Aβ42 and P301S aggregated tau, along with pharmacological manipulation, to produce pathologies relevant to AD. The results offer a novel ex vivo organotypic slice model to investigate AD pathologies with potential applications for screening drugs or therapies in the future.
Collapse
Affiliation(s)
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
13
|
Kosmachevskaya OV, Novikova NN, Yakunin SN, Topunov AF. Formation of Supplementary Metal-Binding Centers in Proteins under Stress Conditions. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:S180-S204. [PMID: 38621750 DOI: 10.1134/s0006297924140104] [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: 08/16/2023] [Revised: 09/21/2023] [Accepted: 10/29/2023] [Indexed: 04/17/2024]
Abstract
In many proteins, supplementary metal-binding centers appear under stress conditions. They are known as aberrant or atypical sites. Physico-chemical properties of proteins are significantly changed after such metal binding, and very stable protein aggregates are formed, in which metals act as "cross-linking" agents. Supplementary metal-binding centers in proteins often arise as a result of posttranslational modifications caused by reactive oxygen and nitrogen species and reactive carbonyl compounds. New chemical groups formed as a result of these modifications can act as ligands for binding metal ions. Special attention is paid to the role of cysteine SH-groups in the formation of supplementary metal-binding centers, since these groups are the main target for the action of reactive species. Supplementary metal binding centers may also appear due to unmasking of amino acid residues when protein conformation changing. Appearance of such centers is usually considered as a pathological process. Such unilateral approach does not allow to obtain an integral view of the phenomenon, ignoring cases when formation of metal complexes with altered proteins is a way to adjust protein properties, activity, and stability under the changed redox conditions. The role of metals in protein aggregation is being studied actively, since it leads to formation of non-membranous organelles, liquid condensates, and solid conglomerates. Some proteins found in such aggregates are typical for various diseases, such as Alzheimer's and Huntington's diseases, amyotrophic lateral sclerosis, and some types of cancer.
Collapse
Affiliation(s)
- Olga V Kosmachevskaya
- Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | | | - Sergey N Yakunin
- National Research Center "Kurchatov Institute", Moscow, 123182, Russia
| | - Alexey F Topunov
- Bach Institute of Biochemistry, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia.
| |
Collapse
|
14
|
Chowdhury S, Sarkar N. Exploring the potential of amyloids in biomedical applications: A review. Biotechnol Bioeng 2024; 121:26-38. [PMID: 37822225 DOI: 10.1002/bit.28569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/31/2023] [Accepted: 09/24/2023] [Indexed: 10/13/2023]
Abstract
Amyloid is defined as a fibrous quaternary structure formed by assembling protein or peptide monomers into intermolecularly hydrogen linked β-sheets. There is a prevalent issue with protein aggregation and the buildup of amyloid molecules, which results in human neurological illnesses including Alzheimer's and Parkinson's. But it is now evident that many organisms, like bacteria, fungi as well as humans, use the same fibrillar structure to carry out a variety of biological functions, such as structure and protection supporting interface transitions and cell-cell recognition, protein control and storage, epigenetic inheritance, and memory. Recent discoveries of self-assembling amyloidogenic peptides and proteins, based on the amyloid core structure, give rise to interesting biomaterials with potential uses in numerous industries. These functions dramatically diverge from the initial conception of amyloid fibrils as intrinsically diseased entities. Apart from the natural ability of amyloids to spontaneously arrange themselves and their exceptional material characteristics, this aspect has prompted extensive research into engineering artificial amyloids for generating various nanostructures, molecular substances, and combined materials. Here, we discuss significant developments in the artificial design of useful amyloids as well as how amyloid materials serve as examples of how function emerges from protein self-assembly at various length scales.
Collapse
Affiliation(s)
- Srijita Chowdhury
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela, Odisha, India
| | - Nandini Sarkar
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela, Odisha, India
| |
Collapse
|
15
|
Hussain R, Graham U, Elder A, Nedergaard M. Air pollution, glymphatic impairment, and Alzheimer's disease. Trends Neurosci 2023; 46:901-911. [PMID: 37777345 PMCID: PMC11934145 DOI: 10.1016/j.tins.2023.08.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/12/2023] [Accepted: 08/29/2023] [Indexed: 10/02/2023]
Abstract
Epidemiological evidence demonstrates a link between air pollution exposure and the onset and progression of cognitive impairment and Alzheimer's disease (AD). However, current understanding of the underlying pathophysiological mechanisms is limited. This opinion article examines the hypothesis that air pollution-induced impairment of glymphatic clearance represents a crucial etiological event in the development of AD. Exposure to airborne particulate matter (PM) leads to systemic inflammation and neuroinflammation, increased metal load, respiratory and cardiovascular dysfunction, and sleep abnormalities. All these factors are known to reduce the efficiency of glymphatic clearance. Rescuing glymphatic function by restricting the impact of causative agents, and improving sleep and cardiovascular system health, may increase the efficiency of waste metabolite clearance and subsequently slow the progression of AD. In sum, we introduce air pollution-mediated glymphatic impairment as an important mechanistic factor to be considered when interpreting the etiology and progression of AD as well as its responsiveness to therapeutic interventions.
Collapse
Affiliation(s)
- Rashad Hussain
- Center for Translational Neuromedicine, University of Rochester, Rochester, NY 14642, USA.
| | | | - Alison Elder
- Department of Environmental Medicine, University of Rochester, Rochester, NY 14642, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester, Rochester, NY 14642, USA; Center for Translational Neuroscience, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
16
|
Silva-Brea D, de Sancho D, Lopez X. Influence of metal binding on the conformational landscape of neurofilament peptides. Phys Chem Chem Phys 2023; 25:26429-26442. [PMID: 37551731 DOI: 10.1039/d3cp03179a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
In order to understand the preferred modes of chelation in metal-binding peptides, quantum mechanical calculations can be used to compute energies, resulting in a hierarchy of binding affinities. These calculations often produce increasing stabilization energies the higher the coordination of the complex. However, as the coordination of a metal increases, the conformational freedom of the polypeptide chain is inevitably reduced, resulting in an entropic penalty. Estimating the magnitude of this penalty from the many different degrees of freedom of biomolecular systems is very challenging, and as a result this contribution to the free energy is often ignored. Here we explore this problem focusing on a family of phosphorylated neuropeptides that bind to aluminum. We find that there is a general negative correlation between both stabilization energy and entropy. Our results suggest that a subtle interplay between enthalpic and entropic forces will determine the population of the most favourable species. Additionally, we discuss the requirements for a possible "Metal Ion Hypothesis" based on our findings.
Collapse
Affiliation(s)
- David Silva-Brea
- Donostia International Physics Center (DIPC), PK 1072, 20080 Donostia San-Sebastian, Spain.
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, UPV/EHU, Spain
| | - David de Sancho
- Donostia International Physics Center (DIPC), PK 1072, 20080 Donostia San-Sebastian, Spain.
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, UPV/EHU, Spain
| | - Xabier Lopez
- Donostia International Physics Center (DIPC), PK 1072, 20080 Donostia San-Sebastian, Spain.
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, UPV/EHU, Spain
| |
Collapse
|
17
|
Rani S, Dhar SB, Khajuria A, Gupta D, Jaiswal PK, Singla N, Kaur M, Singh G, Barnwal RP. Advanced Overview of Biomarkers and Techniques for Early Diagnosis of Alzheimer's Disease. Cell Mol Neurobiol 2023; 43:2491-2523. [PMID: 36847930 PMCID: PMC11410160 DOI: 10.1007/s10571-023-01330-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/15/2023] [Indexed: 03/01/2023]
Abstract
The development of early non-invasive diagnosis methods and identification of novel biomarkers are necessary for managing Alzheimer's disease (AD) and facilitating effective prognosis and treatment. AD has multi-factorial nature and involves complex molecular mechanism, which causes neuronal degeneration. The primary challenges in early AD detection include patient heterogeneity and lack of precise diagnosis at the preclinical stage. Several cerebrospinal fluid (CSF) and blood biomarkers have been proposed to show excellent diagnosis ability by identifying tau pathology and cerebral amyloid beta (Aβ) for AD. Intense research endeavors are being made to develop ultrasensitive detection techniques and find potent biomarkers for early AD diagnosis. To mitigate AD worldwide, understanding various CSF biomarkers, blood biomarkers, and techniques that can be used for early diagnosis is imperative. This review attempts to provide information regarding AD pathophysiology, genetic and non-genetic factors associated with AD, several potential blood and CSF biomarkers, like neurofilament light, neurogranin, Aβ, and tau, along with biomarkers under development for AD detection. Besides, numerous techniques, such as neuroimaging, spectroscopic techniques, biosensors, and neuroproteomics, which are being explored to aid early AD detection, have been discussed. The insights thus gained would help in finding potential biomarkers and suitable techniques for the accurate diagnosis of early AD before cognitive dysfunction.
Collapse
Affiliation(s)
- Shital Rani
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Sudhrita Basu Dhar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Akhil Khajuria
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Dikshi Gupta
- JoyScore Inc., 2440 Cerritos Ave, Signal Hill, CA, 90755, USA
| | - Pradeep Kumar Jaiswal
- Department of Biochemistry and Biophysics, Texas A & M University, College Station, TX, 77843, USA
| | - Neha Singla
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Mandeep Kaur
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India.
| | | |
Collapse
|
18
|
Hnath B, Chen J, Reynolds J, Choi E, Wang J, Zhang D, Sha CM, Dokholyan NV. Big versus small: The impact of aggregate size in disease. Protein Sci 2023; 32:e4686. [PMID: 37243896 PMCID: PMC10273386 DOI: 10.1002/pro.4686] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/17/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023]
Abstract
Protein aggregation results in an array of different size soluble oligomers and larger insoluble fibrils. Insoluble fibrils were originally thought to cause neuronal cell deaths in neurodegenerative diseases due to their prevalence in tissue samples and disease models. Despite recent studies demonstrating the toxicity associated with soluble oligomers, many therapeutic strategies still focus on fibrils or consider all types of aggregates as one group. Oligomers and fibrils require different modeling and therapeutic strategies, targeting the toxic species is crucial for successful study and therapeutic development. Here, we review the role of different-size aggregates in disease, and how factors contributing to aggregation (mutations, metals, post-translational modifications, and lipid interactions) may promote oligomers opposed to fibrils. We review two different computational modeling strategies (molecular dynamics and kinetic modeling) and how they are used to model both oligomers and fibrils. Finally, we outline the current therapeutic strategies targeting aggregating proteins and their strengths and weaknesses for targeting oligomers versus fibrils. Altogether, we aim to highlight the importance of distinguishing the difference between oligomers and fibrils and determining which species is toxic when modeling and creating therapeutics for protein aggregation in disease.
Collapse
Affiliation(s)
- Brianna Hnath
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Jiaxing Chen
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Joshua Reynolds
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Esther Choi
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Medical Scientist Training ProgramPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Jian Wang
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Dongyan Zhang
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
| | - Congzhou M. Sha
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Medical Scientist Training ProgramPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of Engineering Science and MechanicsPenn State UniversityUniversity ParkPennsylvaniaUSA
| | - Nikolay V. Dokholyan
- Department of Biomedical EngineeringPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of PharmacologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of Engineering Science and MechanicsPenn State UniversityUniversity ParkPennsylvaniaUSA
- Department of Biochemistry & Molecular BiologyPenn State College of MedicineHersheyPennsylvaniaUSA
- Department of ChemistryPenn State UniversityUniversity ParkPennsylvaniaUSA
| |
Collapse
|
19
|
Sajjad H, Sajjad A, Haya RT, Khan MM, Zia M. Copper oxide nanoparticles: In vitro and in vivo toxicity, mechanisms of action and factors influencing their toxicology. Comp Biochem Physiol C Toxicol Pharmacol 2023; 271:109682. [PMID: 37328134 DOI: 10.1016/j.cbpc.2023.109682] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/21/2023] [Accepted: 06/11/2023] [Indexed: 06/18/2023]
Abstract
Copper oxide nanoparticles (CuO NPs) have received increasing interest due to their distinctive properties, including small particle size, high surface area, and reactivity. Due to these properties, their applications have been expanded rapidly in various areas such as biomedical properties, industrial catalysts, gas sensors, electronic materials, and environmental remediation. However, because of these widespread uses, there is now an increased risk of human exposure, which could lead to short- and long-term toxicity. This review addresses the underlying toxicity mechanisms of CuO NPs in cells which include reactive oxygen species generation, leaching of Cu ion, coordination effects, non-homeostasis effect, autophagy, and inflammation. In addition, different key factors responsible for toxicity, characterization, surface modification, dissolution, NPs dose, exposure pathways and environment are discussed to understand the toxicological impact of CuO NPs. In vitro and in vivo studies have shown that CuO NPs cause oxidative stress, cytotoxicity, genotoxicity, immunotoxicity, neurotoxicity, and inflammation in bacterial, algal, fish, rodents, and human cell lines. Therefore, to make CuO NPs a more suitable candidate for various applications, it is essential to address their potential toxic effects, and hence, more studies should be done on the long-term and chronic impacts of CuO NPs at different concentrations to assure the safe usage of CuO NPs.
Collapse
Affiliation(s)
- Humna Sajjad
- Department of Biotechnology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Anila Sajjad
- Department of Biotechnology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Rida Tul Haya
- Department of Biotechnology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | | | - Muhammad Zia
- Department of Biotechnology, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|
20
|
Bryliński Ł, Kostelecka K, Woliński F, Duda P, Góra J, Granat M, Flieger J, Teresiński G, Buszewicz G, Sitarz R, Baj J. Aluminium in the Human Brain: Routes of Penetration, Toxicity, and Resulting Complications. Int J Mol Sci 2023; 24:7228. [PMID: 37108392 PMCID: PMC10139039 DOI: 10.3390/ijms24087228] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Aluminium (Al) is the most ubiquitous metal in the Earth's crust. Even though its toxicity is well-documented, the role of Al in the pathogenesis of several neurological diseases remains debatable. To establish the basic framework for future studies, we review literature reports on Al toxicokinetics and its role in Alzheimer's disease (AD), autism spectrum disorder (ASD), alcohol use disorder (AUD), multiple sclerosis (MS), Parkinson's disease (PD), and dialysis encephalopathy (DE) from 1976 to 2022. Despite poor absorption via mucosa, the biggest amount of Al comes with food, drinking water, and inhalation. Vaccines introduce negligible amounts of Al, while the data on skin absorption (which might be linked with carcinogenesis) is limited and requires further investigation. In the above-mentioned diseases, the literature shows excessive Al accumulation in the central nervous system (AD, AUD, MS, PD, DE) and epidemiological links between greater Al exposition and their increased prevalence (AD, PD, DE). Moreover, the literature suggests that Al has the potential as a marker of disease (AD, PD) and beneficial results of Al chelator use (such as cognitive improvement in AD, AUD, MS, and DE cases).
Collapse
Affiliation(s)
- Łukasz Bryliński
- Student Scientific Group, Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Katarzyna Kostelecka
- Student Scientific Group, Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Filip Woliński
- Student Scientific Group, Department of Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland
| | - Piotr Duda
- Student Scientific Group, Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Joanna Góra
- Student Scientific Group, Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Michał Granat
- Student Scientific Group, Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland
| | - Grzegorz Teresiński
- Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Grzegorz Buszewicz
- Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Ryszard Sitarz
- I Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, 20-059 Lublin, Poland
| | - Jacek Baj
- Department of Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland
| |
Collapse
|
21
|
Ge K, Li Z, Wang A, Bai Z, Zhang X, Zheng X, Liu Z, Gao F. An NIR-Driven Upconversion/C 3N 4/CoP Photocatalyst for Efficient Hydrogen Production by Inhibiting Electron-Hole Pair Recombination for Alzheimer's Disease Therapy. ACS NANO 2023; 17:2222-2234. [PMID: 36688477 DOI: 10.1021/acsnano.2c08499] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Redox imbalance and abnormal amyloid protein (Aβ) buildup are key factors in the etiology of Alzheimer's disease (AD). As an antioxidant, the hydrogen molecule (H2) has the potential to cure AD by specifically scavenging highly harmful reactive oxygen species (ROS) such as •OH. However, due to the low solubility of H2 (1.6 ppm), the traditional H2 administration pathway cannot easily achieve long-term and effective accumulation of H2 in the foci. Therefore, how to achieve the continuous release of H2 in situ is the key to improve the therapeutic effect on AD. As a corollary, we designed a rare earth ion doped g-C3N4 upconversion photocatalyst, which can respond to NIR and realize the continuous production of H2 by photocatalytic decomposition of H2O in biological tissue, which avoids the problem of the poor penetration of visible light. The introduction of CoP cocatalyst accelerates the separation and transfer of photogenerated electrons in g-C3N4, thus improving the photocatalytic activity of hydrogen evolution reaction. The morphology of the composite photocatalyst was shown by transmission electron microscopy, and the crystal structure was studied by X-ray diffractometry and Raman analysis. In addition, the ability of g-C3N4 to chelate metal ions and the photothermal properties of CoP can inhibit Aβ and reduce the deposition of Aβ in the brain. Efficient in situ hydrogen production therapy combined with multitarget synergism solves the problem of a poor therapeutic effect of a single target. In vivo studies have shown that UCNP@CoP@g-C3N4 can reduce Aβ deposition, improve memory impairment, and reduce neuroinflammation in AD mice.
Collapse
Affiliation(s)
- Kezhen Ge
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Zheng Li
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Ali Wang
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Zetai Bai
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Xing Zhang
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Xin Zheng
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Zhao Liu
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| | - Fenglei Gao
- Jiangsu Key Laboratory of Brain Disease and Bioinformation, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| |
Collapse
|
22
|
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: 2.5] [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
|
23
|
Cerón R, Peimbert M, Rojo-Domínguez A, Nájera H. Hen lysozyme fibrillogenesis, molten globule intermediate and effect of copper salts. J Biomol Struct Dyn 2023; 41:423-434. [PMID: 34821199 DOI: 10.1080/07391102.2021.2006090] [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: 01/04/2023]
Abstract
The amyloid fibres have been related to many diseases. The molten globule intermediate has been proposed to form part of the folding pathway of many proteins. In the present study, we investigated the mechanism of amyloid-fibres formation of hen egg-white lysozyme (HEWL) incubated in a potassium phosphate buffer, pH 11.8, 100 mM, at 37 °C for 30 h, and evaluated the influence of Cu(II) present in two salts (CuSO4 and CuCl2) during fibrillogenesis. Co-incubation and post-incubation of lysozyme with copper salts reduced the fluorescence signal of thioflavin T with an increment in the intrinsic fluorescence of the protein. The ANS fluorescence test showed that incubation of HEWL for 6 h generated a molten globule intermediate state that formed amyloid fibres when incubation was carried out for a 30-h timespan. Dynamic light scattering showed a heterogeneous population of states in samples incubated in the absence or the presence of salts during the fibrillation process. The existence of a reducing potential was verified during the formation of HEWL amyloid fibres with the bathocuproine disulphonate test. Transmission electron microscopy confirmed the presence and absence of fibres in solutions incubated with and without Cu(II). This work demonstrated that lysozyme formed amyloid fibres at 37 °C and copper inhibited its formation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ricardo Cerón
- Posgrado en Ciencias Naturales e Ingeniería, Universidad Autónoma Metropolitana-Cuajimalpa, Mexico City, Mexico.,Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana-Cuajimalpa, Mexico City, Mexico
| | - Mariana Peimbert
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana-Cuajimalpa, Mexico City, Mexico
| | - Arturo Rojo-Domínguez
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana-Cuajimalpa, Mexico City, Mexico
| | - Hugo Nájera
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana-Cuajimalpa, Mexico City, Mexico
| |
Collapse
|
24
|
Oliyaei N, Moosavi-Nasab M, Tanideh N, Iraji A. Multiple roles of fucoxanthin and astaxanthin against Alzheimer's disease: Their pharmacological potential and therapeutic insights. Brain Res Bull 2023; 193:11-21. [PMID: 36435362 DOI: 10.1016/j.brainresbull.2022.11.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/14/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022]
Abstract
Alzheimer's disease (AD) is the most devastating neurodegenerative disorder affecting the elderly. The exact pathology of AD is not yet fully understood and several hallmarks such as the deposition of amyloid-β, tau hyperphosphorylation, and neuroinflammation, as well as mitochondrial, metal ions, autophagy, and cholinergic dysfunctions are known as pathologic features of AD. Since no definitive treatment has been proposed to target AD to date, many natural products have shown promising preventive potentials and contributed to slowing down the disease progression. Algae is a promising source of novel bioactive substances known to prevent neurodegenerative disorders including AD. In this context, fucoxanthin and astaxanthin, natural carotenoids abundant in algae, has shown to possess neuroprotective properties through antioxidant, and anti-inflammatory characteristics in modulating the symptoms of AD. Fucoxanthin and astaxanthin exhibit anti-AD activities by inhibition of AChE, BuChE, BACE-1, and MAO, suppression of Aβ accumulation. Also, fucoxanthin and astaxanthin inhibit apoptosis induced by Aβ1-42 and H2O2-induced cytotoxicity, and modulate the antioxidant enzymes (SOD and CAT), through inhibition of the ERK pathway. Moreover, cellular and animal studies on the beneficial effects of fucoxanthin and astaxanthin against AD were also reviewed. The potential role of fucoxanthin and astaxanthin exhibits great efficacy for the management of AD by acting on multiple targets.
Collapse
Affiliation(s)
- Najmeh Oliyaei
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Seafood Processing Research Center, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Marzieh Moosavi-Nasab
- Seafood Processing Research Center, School of Agriculture, Shiraz University, Shiraz, Iran; Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran.
| | - Nader Tanideh
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Iraji
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Central Research laboratory, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
25
|
Soliman TN, Mohammed DM, El-Messery TM, Elaaser M, Zaky AA, Eun JB, Shim JH, El-Said MM. Microencapsulation of Plant Phenolic Extracts Using Complex Coacervation Incorporated in Ultrafiltered Cheese Against AlCl 3-Induced Neuroinflammation in Rats. Front Nutr 2022; 9:929977. [PMID: 35845781 PMCID: PMC9278961 DOI: 10.3389/fnut.2022.929977] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/01/2022] [Indexed: 11/19/2022] Open
Abstract
Plant-derived phenolic compounds have numerous biological effects, including antioxidant, anti-inflammatory, and neuroprotective effects. However, their application is limited because they are degraded under environmental conditions. The aim of this study was to microencapsulate plant phenolic extracts using a complex coacervation method to mitigate this problem. Red beet (RB), broccoli (BR), and spinach leaf (SL) phenolic extracts were encapsulated by complex coacervation. The characteristics of complex coacervates [zeta potential, encapsulation efficiency (EE), FTIR, and morphology] were evaluated. The RB, BR, and SL complex coacervates were incorporated into an ultrafiltered (UF) cheese system. The chemical properties, pH, texture profile, microstructure, and sensory properties of UF cheese with coacervates were determined. In total, 54 male Sprague-Dawley rats were used, among which 48 rats were administered an oral dose of AlCl3 (100 mg/kg body weight/d). Nutritional and biochemical parameters, including malondialdehyde, superoxide dismutase, catalase, reduced glutathione, nitric oxide, acetylcholinesterase, butyrylcholinesterase, dopamine, 5-hydroxytryptamine, brain-derived neurotrophic factor, and glial fibrillary acidic protein, were assessed. The RB, BR, and SL phenolic extracts were successfully encapsulated. The RB, BR, and SL complex coacervates had no impact on the chemical composition of UF cheese. The structure of the RB, BR, and SL complex coacervates in UF cheese was the most stable. The hardness of UF cheese was progressively enhanced by using the RB, BR, and SL complex coacervates. The sensory characteristics of the UF cheese samples achieved good scores and were viable for inclusion in food systems. Additionally, these microcapsules improved metabolic strategies and neurobehavioral systems and enhanced the protein biosynthesis of rat brains. Both forms failed to induce any severe side effects in any experimental group. It can be concluded that the microencapsulation of plant phenolic extracts using a complex coacervation technique protected rats against AlCl3-induced neuroinflammation. This finding might be of interest to food producers and researchers aiming to deliver natural bioactive compounds in the most acceptable manner (i.e., food).
Collapse
Affiliation(s)
- Tarek N. Soliman
- Dairy Department, Food Industries and Nutrition Research Institute, National Research Centre, Cairo, Egypt
| | - Dina Mostafa Mohammed
- Department of Nutrition and Food Sciences, Food Industries and Nutrition Research Institute, National Research Centre, Cairo, Egypt
| | - Tamer M. El-Messery
- Dairy Department, Food Industries and Nutrition Research Institute, National Research Centre, Cairo, Egypt
| | - Mostafa Elaaser
- Dairy Department, Food Industries and Nutrition Research Institute, National Research Centre, Cairo, Egypt
| | - Ahmed A. Zaky
- Department of Food Technology, Food Industries and Nutrition Research Institute, National Research Centre, Cairo, Egypt
| | - Jong-Bang Eun
- Department of Food Science and Technology, Chonnam National University, Gwangju, South Korea
| | - Jae-Han Shim
- Natural Products Chemistry Laboratory, Biotechnology Research Institute, Chonnam National University, Gwangju, South Korea
| | - Marwa M. El-Said
- Dairy Department, Food Industries and Nutrition Research Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
26
|
Whitmore CA, Haynes JR, Behof WJ, Rosenberg AJ, Tantawy MN, Hachey BC, Wadzinski BE, Spiller BW, Peterson TE, Paffenroth KC, Harrison FE, Beelman RB, Wijesinghe P, Matsubara JA, Pham W. Longitudinal Consumption of Ergothioneine Reduces Oxidative Stress and Amyloid Plaques and Restores Glucose Metabolism in the 5XFAD Mouse Model of Alzheimer's Disease. Pharmaceuticals (Basel) 2022; 15:742. [PMID: 35745661 PMCID: PMC9228400 DOI: 10.3390/ph15060742] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 01/27/2023] Open
Abstract
Background: Ergothioneine (ERGO) is a unique antioxidant and a rare amino acid available in fungi and various bacteria but not in higher plants or animals. Substantial research data indicate that ERGO is a physiological antioxidant cytoprotectant. Different from other antioxidants that need to breach the blood-brain barrier to enter the brain parenchyma, a specialized transporter called OCTN1 has been identified for transporting ERGO to the brain. Purpose: To assess whether consumption of ERGO can prevent the progress of Alzheimer's disease (AD) on young (4-month-old) 5XFAD mice. Methods and materials: Three cohorts of mice were tested in this study, including ERGO-treated 5XFAD, non-treated 5XFAD, and WT mice. After the therapy, the animals went through various behavioral experiments to assess cognition. Then, mice were scanned with PET imaging to evaluate the biomarkers associated with AD using [11C]PIB, [11C]ERGO, and [18F]FDG radioligands. At the end of imaging, the animals went through cardiac perfusion, and the brains were isolated for immunohistology. Results: Young (4-month-old) 5XFAD mice did not show a cognitive deficit, and thus, we observed modest improvement in the treated counterparts. In contrast, the response to therapy was clearly detected at the molecular level. Treating 5XFAD mice with ERGO resulted in reduced amyloid plaques, oxidative stress, and rescued glucose metabolism. Conclusions: Consumption of high amounts of ERGO benefits the brain. ERGO has the potential to prevent AD. This work also demonstrates the power of imaging technology to assess response during therapy.
Collapse
Affiliation(s)
- Clayton A. Whitmore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (C.A.W.); (J.R.H.); (W.J.B.); (A.J.R.); (M.N.T.); (T.E.P.)
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Justin R. Haynes
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (C.A.W.); (J.R.H.); (W.J.B.); (A.J.R.); (M.N.T.); (T.E.P.)
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - William J. Behof
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (C.A.W.); (J.R.H.); (W.J.B.); (A.J.R.); (M.N.T.); (T.E.P.)
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Adam J. Rosenberg
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (C.A.W.); (J.R.H.); (W.J.B.); (A.J.R.); (M.N.T.); (T.E.P.)
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mohammed N. Tantawy
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (C.A.W.); (J.R.H.); (W.J.B.); (A.J.R.); (M.N.T.); (T.E.P.)
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Brian C. Hachey
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA;
| | - Brian E. Wadzinski
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37233, USA; (B.E.W.); (B.W.S.); (K.C.P.)
| | - Benjamin W. Spiller
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37233, USA; (B.E.W.); (B.W.S.); (K.C.P.)
| | - Todd E. Peterson
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (C.A.W.); (J.R.H.); (W.J.B.); (A.J.R.); (M.N.T.); (T.E.P.)
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Krista C. Paffenroth
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37233, USA; (B.E.W.); (B.W.S.); (K.C.P.)
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA;
| | - Fiona E. Harrison
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA;
- Department of Medicine, Diabetes, Endocrinology & Metabolism, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Robert B. Beelman
- Department of Food Science, Center for Plant and Mushroom Foods for Health, Penn State University, University Park, PA 16802, USA;
| | - Printha Wijesinghe
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC V5Z 3N9, Canada; (P.W.); (J.A.M.)
| | - Joanne A. Matsubara
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC V5Z 3N9, Canada; (P.W.); (J.A.M.)
| | - Wellington Pham
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (C.A.W.); (J.R.H.); (W.J.B.); (A.J.R.); (M.N.T.); (T.E.P.)
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA;
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN 37212, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Ingram Cancer Center, Nashville, TN 37232, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Institute of Nanoscale Science and Engineering, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
27
|
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
|
28
|
Petersingham G, Zaman MS, Johnson AJ, Reddy N, Torres AM, Wu MJ. Molecular details of aluminium-amyloid β peptide interaction by nuclear magnetic resonance. Biometals 2022; 35:759-769. [PMID: 35639270 DOI: 10.1007/s10534-022-00399-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 05/16/2022] [Indexed: 11/24/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative condition that poses major challenges to human health. Both amyloid β (Aβ) and metal ions such as aluminium are implicated in the development of AD. By the means of NMR, the interactions of Al3+ with Aβ1-28 peptide as well as the Aβ1-28 analogues were studied, and the key binding sites of Al3+ in Aβ determined. NMR data showed Al3+ interacts with Aβ1-28 at the NH and αH of numerous residues by exhibiting upfield shifts. Using Aβ analogues where His6, His13 and His14 were individually replaced by alanine residue(s), including Aβ H6A, Aβ H13A, Aβ H14A, and Aβ H6,13,14A, the results demonstrated that the histidine residues (His6, His13 and His14) and N-terminal Asp1 were involved in the Al3+ coordination. These findings provide, for the first time, the details of the molecular interaction between Al3+ and Aβ, which points to the potential role of Al3+ in Aβ aggregation, hence in AD development.
Collapse
Affiliation(s)
- Gayani Petersingham
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW, 2751, Australia
| | - Mohammad S Zaman
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW, 2751, Australia
| | - Adam J Johnson
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW, 2751, Australia
| | - Narsimha Reddy
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW, 2751, Australia
| | - Allan M Torres
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW, 2751, Australia.,Nanoscale Organisation and Dynamics Group, Western Sydney University, Locked Bag 1797, Penrith, NSW, 2751, Australia
| | - Ming J Wu
- School of Science, Western Sydney University, Locked Bag 1797, Penrith, NSW, 2751, Australia.
| |
Collapse
|
29
|
Abstract
Amyloids are protein aggregates bearing a highly ordered cross β structural motif, which may be functional but are mostly pathogenic. Their formation, deposition in tissues and consequent organ dysfunction is the central event in amyloidogenic diseases. Such protein aggregation may be brought about by conformational changes, and much attention has been directed toward factors like metal binding, post-translational modifications, mutations of protein etc., which eventually affect the reactivity and cytotoxicity of the associated proteins. Over the past decade, a global effort from different groups working on these misfolded/unfolded proteins/peptides has revealed that the amino acid residues in the second coordination sphere of the active sites of amyloidogenic proteins/peptides cause changes in H-bonding pattern or protein-protein interactions, which dramatically alter the structure and reactivity of these proteins/peptides. These second sphere effects not only determine the binding of transition metals and cofactors, which define the pathology of some of these diseases, but also change the mechanism of redox reactions catalyzed by these proteins/peptides and form the basis of oxidative damage associated with these amyloidogenic diseases. The present review seeks to discuss such second sphere modifications and their ramifications in the etiopathology of some representative amyloidogenic diseases like Alzheimer's disease (AD), type 2 diabetes mellitus (T2Dm), Parkinson's disease (PD), Huntington's disease (HD), and prion diseases.
Collapse
Affiliation(s)
- Madhuparna Roy
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Arnab Kumar Nath
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Ishita Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Somdatta Ghosh Dey
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
30
|
Neurotoxic effects of aluminium exposure as a potential risk factor for Alzheimer's disease. Pharmacol Rep 2022; 74:439-450. [PMID: 35088386 DOI: 10.1007/s43440-022-00353-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 01/02/2023]
Abstract
Aluminium is one of the most widely distributed elements of the Earth's crust. Its routine use has resulted in excessive human exposure and due to the potential neurotoxic effects has attained a huge interest in recent years. Despite its ubiquitous abundance, aluminium has no crucial biological functions in the human body. Oxidative stress and neuroinflammatory effects are attributed to its neurotoxic manifestations implicated in Alzheimer's disease. In this review, we have discussed the neuroinflammatory and neurodegenerative events in the brain induced by aluminium exposure. We have highlighted the neurotoxic events caused by aluminium, such as oxidative stress, apoptosis, inflammatory events, calcium dyshomeostasis, Aβ deposition, and neurofibrillary tangle formation in the brain. In addition, the protective measures needed for prevention of aluminium-induced neuronal dysregulations have also been discussed.
Collapse
|
31
|
Devi S, Chaturvedi M, Fatima S, Priya S. Environmental factors modulating protein conformations and their role in protein aggregation diseases. Toxicology 2022; 465:153049. [PMID: 34818560 DOI: 10.1016/j.tox.2021.153049] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/12/2021] [Accepted: 11/20/2021] [Indexed: 12/13/2022]
Abstract
The adverse physiological conditions have been long known to impact protein synthesis, folding and functionality. Major physiological factors such as the effect of pH, temperature, salt and pressure are extensively studied for their impact on protein structure and homeostasis. However, in the current scenario, the environmental risk factors (pollutants) have gained impetus in research because of their increasing concentrations in the environment and strong epidemiologic link with protein aggregation disorders. Here, we review the physiological and environmental risk factors for their impact on protein conformational changes, misfolding, aggregation, and associated pathological conditions, especially environmental risk factors associated pathologies.
Collapse
Affiliation(s)
- Shweta Devi
- Systems Toxicology and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Minal Chaturvedi
- Systems Toxicology and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Siraj Fatima
- Systems Toxicology and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Smriti Priya
- Systems Toxicology and Health Risk Assessment Group, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, CSIR-Indian Institute of Toxicology Research, Lucknow-226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
32
|
Folarin OR, Olopade FE, Olopade JO. Essential Metals in the Brain and the Application of Laser Ablation-Inductively Coupled Plasma-Mass Spectrometry for their Detection. Niger J Physiol Sci 2021; 36:123-147. [PMID: 35947740 DOI: 10.54548/njps.v36i2.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 06/15/2023]
Abstract
Metals are natural component of the ecosystem present throughout the layers of atmosphere; their abundant expression in the brain indicates their importance in the central nervous system (CNS). Within the brain tissue, their distribution is highly compartmentalized, the pattern of which is determined by their primary roles. Bio-imaging of the brain to reveal spatial distribution of metals within specific regions has provided a unique understanding of brain biochemistry and architecture, linking both the structures and the functions through several metal mediated activities. Bioavailability of essential trace metal is needed for normal brain function. However, disrupted metal homeostasis can influence several biochemical pathways in different fields of metabolism and cause characteristic neurological disorders with a typical disease process usually linked with aberrant metal accumulations. In this review we give a brief overview of roles of key essential metals (Iron, Copper and Zinc) including their molecular mechanisms and bio-distribution in the brain as well as their possible involvement in the pathogenesis of related neurodegenerative diseases. In addition, we also reviewed recent applications of Laser Ablation Inductively Couple Plasma Mass Spectrophotometry (LA-ICP-MS) in the detection of both toxic and essential metal dyshomeostasis in neuroscience research and other related brain diseases.
Collapse
|
33
|
Streit WJ, Rotter J, Winter K, Müller W, Khoshbouei H, Bechmann I. Droplet Degeneration of Hippocampal and Cortical Neurons Signifies the Beginning of Neuritic Plaque Formation. J Alzheimers Dis 2021; 85:1701-1720. [PMID: 34958037 DOI: 10.3233/jad-215334] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Neuritic plaques contain neural and microglial elements, and amyloid-β protein (Aβ), but their pathogenesis remains unknown. OBJECTIVE Elucidate neuritic plaque pathogenesis. METHODS Histochemical visualization of hyperphosphorylated-tau positive (p-tau+) structures, microglia, Aβ, and iron. RESULTS Disintegration of large projection neurons in human hippocampus and neocortex presents as droplet degeneration: pretangle neurons break up into spheres of numerous p-tau+ droplets of various sizes, which marks the beginning of neuritic plaques. These droplet spheres develop in the absence of colocalized Aβ deposits but once formed become encased in diffuse Aβ with great specificity. In contrast, neurofibrillary tangles often do not colocalize with Aβ. Double-labelling for p-tau and microglia showed a lack of microglial activation or phagocytosis of p-tau+ degeneration droplets but revealed massive upregulation of ferritin in microglia suggesting presence of high levels of free iron. Perl's Prussian blue produced positive staining of microglia, droplet spheres, and Aβ plaque cores supporting the suggestion that droplet degeneration of pretangle neurons in the hippocampus and cortex represents ferroptosis, which is accompanied by the release of neuronal iron extracellularly. CONCLUSION Age-related iron accumulation and ferroptosis in the CNS likely trigger at least two endogenous mechanisms of neuroprotective iron sequestration and chelation, microglial ferritin expression and Aβ deposition, respectively, both contributing to the formation of neuritic plaques. Since neurofibrillary tangles and Aβ deposits colocalize infrequently, tangle formation likely does not involve release of neuronal iron extracellularly. In human brain, targeted deposition of Aβ occurs specifically in response to ongoing ferroptotic droplet degeneration thereby producing neuritic plaques.
Collapse
Affiliation(s)
- Wolfgang J Streit
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
| | - Jonas Rotter
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Karsten Winter
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Wolf Müller
- Department of Neuropathology, Leipzig University, Leipzig, Germany
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| |
Collapse
|
34
|
Lippi SLP, Neely CLC, Amaya AL. Trace concentrations, heavy implications: Influences of biometals on major brain pathologies of Alzheimer's disease. Int J Biochem Cell Biol 2021; 143:106136. [PMID: 34906694 DOI: 10.1016/j.biocel.2021.106136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that involves accumulation of toxic protein species, notably amyloid-β (Aβ)plaques and neurofibrillary tau tangles that are associated with cognitive decline. These proteins can bind metal ions, ultimately affecting their structure and function. In this review, we discuss key biometals such as zinc, copper, and iron that interact with protein species involved in AD, mainly Aβ, tau, and the late-onset AD risk factor Apolipoprotein E (APOE). These metals interact with Aβ and tau proteins, affecting their aggregation and toxicity. The allele variants of APOE also have different interactions with these metals, affecting APOE protein expression and aggregation of AD protein species.
Collapse
Affiliation(s)
- Stephen L P Lippi
- Angelo State University, Department of Psychology, San Angelo, TX, USA.
| | - Caroline L C Neely
- Massachusetts General Hospital, Department of Neurosurgery, Boston, MA, USA
| | - Anthony L Amaya
- University of Texas at San Antonio, Department of Chemistry, San Antonio, TX, USA
| |
Collapse
|
35
|
Shardlow E, Khan B, Exley C. Monitoring the early aggregatory behaviour and size of Aβ 1-42 in the absence & presence of metal ions using dynamic light scattering. J Trace Elem Med Biol 2021; 67:126766. [PMID: 33964808 DOI: 10.1016/j.jtemb.2021.126766] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIM Aβ1-42 is an amyloidogenic peptide found within senile plaques extracted from those who died with a diagnosis of Alzheimer's disease. The potent neurotoxicity of this peptide is related to its propensity to form aggregated conformations in vivo, a process that is influenced by the species and concentration of metal ions present within the local environment. This study examines the impact of different metals upon the early aggregatory behaviour and size of Aβ1-42 under simulated physiological conditions. METHODS The size and aggregatory behaviour of Aβ1-42 in the presence and absence of metal ions was monitored during the initial 30 min of fibril formation in real-time using dynamic light scattering. RESULTS Intensity scattering measurements showed a clear tendency towards aggregation with regards to Aβ1-42 only solutions (10 μM). Both equimolar Al3+ & Cu2+ lowered and stabilised the dimensions of Aβ1-42 aggregates; however, a diminutive but significant increase in size was still observed over a 30-min period. While excess Al3+ continued to supress the size of Aβ1-42, a 10-fold increase in the concentration of Cu2+ accelerated peptide aggregation relative to that observed for equimolar metal but not compared to Aβ1-42 alone. CONCLUSION These results infer that Al3+ ions stabilise and aid in the maintenance of smaller, toxic intermediates while excess Cu2+ facilitates the formation of larger, more inert, amorphous species exceeding 1 μm in size. Furthermore, we propose that metal-induced toxicity of Aβ1-42 is reflective of their ability to preserve smaller oligomeric species in vitro.
Collapse
Affiliation(s)
- Emma Shardlow
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| | - Bakhtbilland Khan
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - Christopher Exley
- The Birchall Centre, Lennard-Jones Laboratories, Keele University, Keele, Staffordshire, ST5 5BG, UK
| |
Collapse
|
36
|
Tuzlakoğlu Öztürk M, Güllülü Ö. Dimerization underlies the aggregation propensity of intrinsically disordered coiled-coil domain-containing 124. Proteins 2021; 90:218-228. [PMID: 34369007 DOI: 10.1002/prot.26210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 11/10/2022]
Abstract
Coiled-coil domain-containing 124 (CCDC124) is a recently discovered ribosome-binding protein conserved in eukaryotes. CCDC124 has regulatory functions on the mediation of reversible ribosomal hibernation and translational recovery by direct attachment to large subunit ribosomal protein uL5, 25S rRNA backbone, and tRNA-binding P/A-site major groove. Moreover, it independently mediates cell division and cellular stress response by facilitating cytokinetic abscission and disulfide stress-dependent transcriptional regulation, respectively. However, the structural characterization and intracellular physiological status of CCDC124 remain unknown. In this study, we employed advanced in silico protein modeling and characterization tools to generate a native-like tertiary structure of CCDC124 and examine the disorder, low sequence complexity, and aggregation propensities, as well as high-order dimeric/oligomeric states. Subsequently, dimerization of CCDC124 was investigated with co-immunoprecipitation (CO-IP) analysis, immunostaining, and a recent live-cell protein-protein interaction method, bimolecular fluorescence complementation (BiFC). Results revealed CCDC124 as a highly disordered protein consisting of low complexity regions at the N-terminus and an aggregation sequence (151-IAVLSV-156) located in the middle region. Molecular docking and post-docking binding free energy analyses highlighted a potential involvement of V153 residue on the generation of high-order dimeric/oligomeric structures. Co-IP, immunostaining, and BiFC analyses were used to further confirm the dimeric state of CCDC124 predominantly localized at the cytoplasm. In conclusion, our findings revealed in silico structural characterization and in vivo subcellular physiological state of CCDC124, suggesting low-complexity regions located at the N-terminus of disordered CCDC124 may regulate the formation of aggregates or high-order dimeric/oligomeric states.
Collapse
Affiliation(s)
| | - Ömer Güllülü
- Department of Radiotherapy and Oncology, University Hospital Frankfurt, Frankfurt am Main, Germany.,Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
37
|
Jamal R, Dihmis OW, Carroll LS, Pengas G. Hypocupraemia-induced anaemia, sensory ataxia and cognitive impairment secondary to zinc-containing dental adhesive. BMJ Case Rep 2021; 14:14/7/e239375. [PMID: 34289999 DOI: 10.1136/bcr-2020-239375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
A 67-year-old man presented with 5 months of worsening memory impairment and sensory gait ataxia on the background of symptomatic anaemia. He experienced falls, agitation and became socially withdrawn over 3 weeks, resulting in hospital admission. On examination, he had sensory gait ataxia consistent with a dorsal column syndrome. He scored 13/30 on the Montreal Cognitive Assessment. Serum analysis showed normocytic anaemia and leucopenia, severe hypocupraemia, reduced caeruloplasmin and normal zinc levels. Overuse of zinc-containing denture cream was the cause of excess zinc ingestion and resultant copper deficiency, leading to blood dyscrasia and myelopathy. The cream was withdrawn and intravenous and then oral copper supplementation was implemented. Direct questions with regard to excess zinc in the diet and serological testing of copper and zinc should be considered in any patient with a dorsal column syndrome, particularly with concurrent anaemia. Copper deficiency may also have a role in exacerbating pre-existing cognitive impairment.
Collapse
Affiliation(s)
- Rayyan Jamal
- Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Liam Stuart Carroll
- Neurology, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, UK
| | - George Pengas
- Neurology, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, UK
| |
Collapse
|
38
|
Al-Shammari N, Savva L, Kennedy-Britten O, Platts JA. Forcefield evaluation and accelerated molecular dynamics simulation of Zn(II) binding to N-terminus of amyloid-β. Comput Biol Chem 2021; 93:107540. [PMID: 34271422 DOI: 10.1016/j.compbiolchem.2021.107540] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/11/2021] [Accepted: 06/21/2021] [Indexed: 01/06/2023]
Abstract
We report conventional and accelerated molecular dynamics simulation of Zn(II) bound to the N-terminus of amyloid-β. By comparison against NMR data for the experimentally determined binding mode, we find that certain combinations of forcefield and solvent model perform acceptably in describing the size, shape and secondary structure, and that there is no appreciable difference between implicit and explicit solvent models. We therefore used the combination of ff14SB forcefield and GBSA solvent model to compare the result of different binding modes of Zn(II) to the same peptide, using accelerated MD to enhance sampling and comparing the free peptide simulated in the same way. We show that Zn(II) imparts significant rigidity to the peptide, disrupts the secondary structure and pattern of salt bridges seen in the free peptide, and induces closer contact between residues. Free energy surfaces in 1 or 2 dimensions further highlight the effect of metal coordination on peptide's spatial extent. We also provide evidence that accelerated MD provides improved sampling over conventional MD by visiting as many or more configurations in much shorter simulation times.
Collapse
Affiliation(s)
| | - Loizos Savva
- School of Chemistry, Cardiff University, Park Place, Cardiff, CF10 3AT, UK
| | | | - James A Platts
- School of Chemistry, Cardiff University, Park Place, Cardiff, CF10 3AT, UK.
| |
Collapse
|
39
|
Roldán-Martín L, Peccati F, Sciortino G, Sodupe M, Maréchal JD. Impact of Cu(II) and Al(III) on the conformational landscape of amyloidβ 1-42. Phys Chem Chem Phys 2021; 23:13023-13032. [PMID: 34095932 DOI: 10.1039/d1cp01561c] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Metal ions have been found to play an important role in the formation of extracellular β-amyloid plaques, a major hallmark of Alzheimer's disease. In the present study, the conformational landscape of Aβ42 with Al(iii) and Cu(ii) has been explored using Gaussian accelerated molecular dynamics. Both metals reduce the flexibility of the peptide and entail a higher structural organization, although to different degrees. As a general trend, Cu(ii) binding leads to an increased α-helix content and to the formation of two α-helices that tend to organize in a U-shape. By contrast, most Al(iii) complexes induce a decrease in helical content, leading to more extended structures that favor the appearance of transitory β-strands.
Collapse
Affiliation(s)
- Lorena Roldán-Martín
- Departament de Química, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.
| | - Francesca Peccati
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain
| | - Giuseppe Sciortino
- Departament de Química, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain. and Institut of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology (BIST), 43007 Tarragona, Catalonia, Spain
| | - Mariona Sodupe
- Departament de Química, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.
| | - Jean-Didier Maréchal
- Departament de Química, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.
| |
Collapse
|
40
|
Salkov VN, Khudoerkov RM. [The role of aluminum and lead in the development of Alzheimer's and Parkinson's diseases]. Arkh Patol 2021; 83:56-61. [PMID: 34041898 DOI: 10.17116/patol20218303156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The article summarizes the data available in the literature on the toxic effects of aluminum and lead on the human brain and assesses the relationship of these effects to the etiopathogenesis of the most common neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases. The accumulation of ions of these metals in the brain structures leads to chronic intoxication that is manifested by the morphological signs that are typical for Alzheimer's disease, such as deposits of β-amyloid and τ-protein mainly in the frontal and temporal regions of the cortex, and for Parkinson's disease, such as degeneration of dopamine neurons in the substantia nigra and their accumulation of α-synuclein. The most likely forms of participation of aluminum and lead ions in the mechanisms of neurodegeneration are the replacement of bivalent metal ions necessary for brain functioning, oxidative stress initiation, epigenetic modifications of histones, and increased expression of noncoding ribonucleic acids.
Collapse
Affiliation(s)
- V N Salkov
- Research Center of Neurology, Moscow, Russia
| | | |
Collapse
|
41
|
Shunan D, Yu M, Guan H, Zhou Y. Neuroprotective effect of Betalain against AlCl 3-induced Alzheimer's disease in Sprague Dawley Rats via putative modulation of oxidative stress and nuclear factor kappa B (NF-κB) signaling pathway. Biomed Pharmacother 2021; 137:111369. [PMID: 33582452 DOI: 10.1016/j.biopha.2021.111369] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/30/2021] [Accepted: 02/03/2021] [Indexed: 12/26/2022] Open
Abstract
Alzheimer's disease (AD) is the most progressive form of neurodegenerative disease, which severely impairs cognitive function. Oxidative stress is identified to contribute to the mechanisms responsible for the pathogenesis of such neurodegenerative diseases. Aluminum is a potent neurotoxin for inducing oxidative stress associated with neurodegenerative diseases. The treatment for AD is limited; hence more treatment options are the need of the day. Betalain is known for its multitude of medicinal assets, including anti-inflammatory activity. Hence, this study was intended to investigate the possible protective effect of betalain against aluminum chloride (AlCl3) induced AD on Sprague Dawley (SD) rats. AlCl3 (100 mg/kg) was administrated orally to induce the AD in SD rats. The rats were supplemented with low and high betalain doses (10 mg/kg and 20 mg/kg) for four weeks. At the end of the experiment, the rats were subjected to behavioral examination and sacrificed to study the biochemical and histological parameters. The results showed attenuation of memory and learning capacity, suppression of lipid oxidation (MDA) through regulation of antioxidant content (SOD, CAT, and GSH) and inhibition of lactate dehydrogenase (LDH), nitric oxide (NO), acetylcholinesterase (AChE), and transmembrane protein (Na+K+ATPase) activity. In addition, the NF-ƙB associated mRNA expression (TNF-α IL-6, Il-1β, iNOS, COX-2) was decreased, as evidenced in histopathological results. The present investigation established that the betalain treatment ameliorated the AlCl3 induced AD by modulating NF-κB pathway activation.
Collapse
Affiliation(s)
- Di Shunan
- Basic Theory of Traditional Chinese Medicine, School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Miao Yu
- Basic Theory of Traditional Chinese Medicine, School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Huibo Guan
- Chinese medicine diagnostics, School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150010, China
| | - Yanyan Zhou
- Basic Theory of Traditional Chinese Medicine, School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
42
|
Cai M, Zhang X, He W, Zhang J. The Involvement of Metals in Alzheimer's Disease Through Epigenetic Mechanisms. Front Genet 2020; 11:614666. [PMID: 33363576 PMCID: PMC7753070 DOI: 10.3389/fgene.2020.614666] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/18/2020] [Indexed: 12/03/2022] Open
Abstract
Alzheimer’s disease (AD) is the most frequent cause of dementia among neurodegenerative diseases. Two factors were hypothesized to be involved in the pathogenesis of AD, namely beta-amyloid cascade and tauopathy. At present, accumulating evidence suggest that epigenetics may be the missing linkage between genes and environment factors, providing possible clues to understand the etiology of the development of AD. In this article, we focus on DNA methylation and histone modification involved in AD and the environment factor of heavy metals’ contribution to AD, especially epigenetic mechanisms. If we can integrate information together, and that may find new potential targets for the treatment.
Collapse
Affiliation(s)
- Menghua Cai
- State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiangjin Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei He
- State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Jianmin Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
43
|
Yawson GK, Huffman SE, Fisher SS, Bothwell PJ, Platt DC, Jones MA, Ferrence GM, Hamaker CG, Webb MI. Ruthenium(III) complexes with imidazole ligands that modulate the aggregation of the amyloid-β peptide via hydrophobic interactions. J Inorg Biochem 2020; 214:111303. [PMID: 33166864 DOI: 10.1016/j.jinorgbio.2020.111303] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by extracellular protein deposits, comprised primarily of the peptide amyloid-beta (Aβ), are a pathological indicator of the disease. Commonly known as Aβ plaques, these deposits contain a relatively high concentration of metals, making metallotherapeutics uniquely suited to target soluble Aβ, thereby limiting its aggregation and cytotoxicity. Ruthenium-based complexes are promising candidates for advancement, as the complex PMRU20 (2-aminothiazolium [trans-RuCl4(2-aminothiazole)2]) and several thiazole-based derivatives were found to prevent the aggregation of Aβ, with hydrogen-bonding functional groups improving their performance. Further investigation into the impact of the heteroatom in the azole ring on the activity of Ru complexes was achieved through the synthesis and evaluation of a small set of imidazole-based compounds. The ability of the complexes to prevent the aggregation of Aβ was determined where the same sample was subjected to analysis by three complementary methods: ThT fluorescence, dynamic light scattering (DLS), and transmission electron microscopy (TEM). It was found that hydrophobic interactions, along with hydrogen-bonding via the imidazole nitrogen heteroatom, promoted interactions with the Aβ peptide, thereby limiting its aggregation. Furthermore, it was found that having rapid and sequential exchange proved detrimental as it resulted in a decreased association with Aβ. These results highlight important considerations between a balance of intermolecular interactions and ligand exchange kinetics in the design of further therapeutic candidates.
Collapse
Affiliation(s)
- Gideon K Yawson
- Department of Chemistry, Illinois State University, Normal, IL 61790-4160, United States of America
| | - Samantha E Huffman
- Department of Chemistry, Illinois State University, Normal, IL 61790-4160, United States of America
| | - Samuel S Fisher
- Department of Chemistry, Illinois State University, Normal, IL 61790-4160, United States of America
| | - Paige J Bothwell
- Core Microscope Facility, Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115, United States of America
| | - David C Platt
- Department of Chemistry, Illinois State University, Normal, IL 61790-4160, United States of America
| | - Marjorie A Jones
- Department of Chemistry, Illinois State University, Normal, IL 61790-4160, United States of America
| | - Gregory M Ferrence
- Department of Chemistry, Illinois State University, Normal, IL 61790-4160, United States of America
| | - Christopher G Hamaker
- Department of Chemistry, Illinois State University, Normal, IL 61790-4160, United States of America
| | - Michael I Webb
- Department of Chemistry, Illinois State University, Normal, IL 61790-4160, United States of America.
| |
Collapse
|
44
|
Evidence of cadmium and mercury involvement in the Aβ42 aggregation process. Biophys Chem 2020; 266:106453. [DOI: 10.1016/j.bpc.2020.106453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/30/2020] [Accepted: 08/02/2020] [Indexed: 12/11/2022]
|
45
|
Klenner MA, Fraser BH, Moon V, Evans BJ, Massi M, Pascali G. Telescoping the Synthesis of the [
18
F]CABS13 Alzheimer's Disease Radiopharmaceutical via Flow Microfluidic Rhenium(I) Complexations. Eur J Inorg Chem 2020. [DOI: 10.1002/ejic.202000433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Mitchell A. Klenner
- National Deuteration Facility (NDF) & Human Health Australian Nuclear Science and Technology Organisation (ANSTO) 2234 Lucas Heights NSW Australia
- School of Molecular and Life Sciences Curtin University 6102 Bentley WA. Australia
| | - Benjamin H. Fraser
- National Deuteration Facility (NDF) & Human Health Australian Nuclear Science and Technology Organisation (ANSTO) 2234 Lucas Heights NSW Australia
| | - Vaughan Moon
- National Deuteration Facility (NDF) & Human Health Australian Nuclear Science and Technology Organisation (ANSTO) 2234 Lucas Heights NSW Australia
- Department of Molecular Sciences Macquarie University 2109 Macquarie Park NSW Australia
| | - Brendan J. Evans
- National Deuteration Facility (NDF) & Human Health Australian Nuclear Science and Technology Organisation (ANSTO) 2234 Lucas Heights NSW Australia
- Department of Molecular Sciences Macquarie University 2109 Macquarie Park NSW Australia
| | - Massimiliano Massi
- School of Molecular and Life Sciences Curtin University 6102 Bentley WA. Australia
| | - Giancarlo Pascali
- National Deuteration Facility (NDF) & Human Health Australian Nuclear Science and Technology Organisation (ANSTO) 2234 Lucas Heights NSW Australia
- Prince of Wales Hospital 2031 Randwick NSW Australia
- School of Chemistry University of New South Wales (UNSW) 2052 Kensington NSW Australia
| |
Collapse
|
46
|
Maia MT, Sena DN, Calais GB, Luna FMT, Beppu MM, Vieira RS. Effects of histidine modification of chitosan microparticles on metal ion adsorption. REACT FUNCT POLYM 2020. [DOI: 10.1016/j.reactfunctpolym.2020.104694] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
47
|
Li Y, Yang C, Wang S, Yang D, Zhang Y, Xu L, Ma L, Zheng J, Petersen RB, Zheng L, Chen H, Huang K. Copper and iron ions accelerate the prion-like propagation of α-synuclein: A vicious cycle in Parkinson's disease. Int J Biol Macromol 2020; 163:562-573. [PMID: 32629061 DOI: 10.1016/j.ijbiomac.2020.06.274] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022]
Abstract
Protein fibrils drive the onset and progression of many diseases in a prion-like manner, i.e. they transcellular propagate through the extracellular space to health cells to initiate toxic aggregation as seeds. The conversion of native α-synuclein into filamentous aggregates in Lewy bodies is a hallmark of Parkinson's disease (PD). Copper and iron ions accumulate in PD brains, however, whether they influence the prion-like propagation of α-synuclein remain unclear. Here, we reported that copper/iron ions accelerate prion-like propagation of α-synuclein fibrils by promoting cellular internalization of α-synuclein fibrils, intracellular α-synuclein aggregation and the subsequent release of mature fibrils to the extracellular space to induce further propagation. Mechanistically, copper/iron ions enhanced α-synuclein fibrils internalization was mediated by negatively charged membrane heparan sulfate proteoglycans (HSPGs). α-Synuclein fibrils formed in the presence of copper/iron ions were more cytotoxic, causing increased ROS production, cell apoptosis, and shortened the lifespan of a C. elegans PD model overexpressing human α-synuclein. Notably, these deleterious effects were ameliorated by two clinically used chelators, triethylenetetramine and deferiprone. Together, our results suggest a new role for heavy metal ions, e.g. copper and iron, in the pathogenesis of PD through accelerating prion-like propagation of α-synuclein fibrils.
Collapse
Affiliation(s)
- Yang Li
- School of Pharmacy, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Chen Yang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Shilin Wang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Dong Yang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Yu Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Li Xu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Liang Ma
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430012, China
| | - Jiaojiao Zheng
- School of Pharmacy, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Robert B Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant 48858, MI, USA
| | - Ling Zheng
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Hong Chen
- School of Pharmacy, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | - Kun Huang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China.
| |
Collapse
|
48
|
Sasanian N, Bernson D, Horvath I, Wittung-Stafshede P, Esbjörner EK. Redox-Dependent Copper Ion Modulation of Amyloid-β (1-42) Aggregation In Vitro. Biomolecules 2020; 10:E924. [PMID: 32570820 PMCID: PMC7355640 DOI: 10.3390/biom10060924] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/12/2020] [Accepted: 06/16/2020] [Indexed: 12/20/2022] Open
Abstract
Plaque deposits composed of amyloid-β (Aβ) fibrils are pathological hallmarks of Alzheimer's disease (AD). Although copper ion dyshomeostasis is apparent in AD brains and copper ions are found co-deposited with Aβ peptides in patients' plaques, the molecular effects of copper ion interactions and redox-state dependence on Aβ aggregation remain elusive. By combining biophysical and theoretical approaches, we here show that Cu2+ (oxidized) and Cu+ (reduced) ions have opposite effects on the assembly kinetics of recombinant Aβ(1-42) into amyloid fibrils in vitro. Cu2+ inhibits both the unseeded and seeded aggregation of Aβ(1-42) at pH 8.0. Using mathematical models to fit the kinetic data, we find that Cu2+ prevents fibril elongation. The Cu2+-mediated inhibition of Aβ aggregation shows the largest effect around pH 6.0 but is lost at pH 5.0, which corresponds to the pH in lysosomes. In contrast to Cu2+, Cu+ ion binding mildly catalyzes the Aβ(1-42) aggregation via a mechanism that accelerates primary nucleation, possibly via the formation of Cu+-bridged Aβ(1-42) dimers. Taken together, our study emphasizes redox-dependent copper ion effects on Aβ(1-42) aggregation and thereby provides further knowledge of putative copper-dependent mechanisms resulting in AD.
Collapse
Affiliation(s)
| | | | | | | | - Elin K. Esbjörner
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Gothenburg, Sweden; (N.S.); (D.B.); (I.H.); (P.W.-S.)
| |
Collapse
|
49
|
Van Dyke N, Yenugadhati N, Birkett NJ, Lindsay J, Turner MC, Willhite CC, Krewski D. Association between aluminum in drinking water and incident Alzheimer's disease in the Canadian Study of Health and Aging cohort. Neurotoxicology 2020; 83:157-165. [PMID: 32360354 DOI: 10.1016/j.neuro.2020.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 03/03/2020] [Accepted: 04/15/2020] [Indexed: 01/17/2023]
Abstract
Epidemiological evidence linking aluminum in drinking water and Alzheimer's disease (AD) has been inconsistent, with previous studies often limited by small sample sizes. The present study addresses this issue using data from the Canadian Study of Health and Aging (CSHA), a prospective cohort of 10,263 subjects followed-up from 1991-1992 through 2001-2002. Participants' residential histories were linked to municipal drinking water sources in 35 Canadian municipalities to obtain ecologic pH, aluminum, fluoride, iron and silica concentrations in drinking water. Cox proportional hazards models were used to examine associations between aluminum and incident AD [Hazard Ratios (HRs), 95% confidence intervals (CIs)], adjusting for age, gender, history of stroke, education, and high blood pressure. A total of 240 incident AD cases were identified during follow-up of 3, 638 subjects derived from the CSHA cohort with complete data on all covariates. With categorical aluminum measurements, there was an increasing, but not statistically significant, exposure-response relationship (HR = 1.34, 95% CI 0.88-2.04, in the highest aluminum exposure category; p = 0.13 for linear trend). Similar results were observed using continuous aluminum measurements (HR=1.21, 95% CI 0.97-1.52, at the interquartile range of 333.8 μg/L; p = 0.09 for linear trend). In a subsample genotyped for ApoE-ε4, there was some evidence of an association between aluminum and AD (p = 0.03 for linear trend). Although a clear association between aluminum in drinking water and AD was not found, the linear trend observed in ApoE-ε4 subsample warrants further examination.
Collapse
Affiliation(s)
- Nicole Van Dyke
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada; McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Ontario, Canada
| | - Nagarajkumar Yenugadhati
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Ontario, Canada; Risk Sciences International, Ottawa, Canada; Department of Epidemiology and Biostatistics, College of Public Health and Health Informatics, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| | - Nicholas J Birkett
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada; McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Ontario, Canada
| | - Joan Lindsay
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Michelle C Turner
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Ontario, Canada; Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | | | - Daniel Krewski
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada; McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, Ontario, Canada; Risk Sciences International, Ottawa, Canada
| |
Collapse
|
50
|
Roy M, Pal I, Nath AK, Dey SG. Peroxidase activity of heme bound amyloid β peptides associated with Alzheimer's disease. Chem Commun (Camb) 2020; 56:4505-4518. [PMID: 32297620 DOI: 10.1039/c9cc09758a] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The amyloid cascade hypothesis attributes the neurodegeneration observed in Alzheimer's disease (AD) to the deposition of the amyloid β (Aβ) peptide into plaques and fibrils in the AD brain. The metal ion hypothesis which implicates several metal ions, viz. Zn2+, Cu2+ and Fe3+, in the AD pathology on account of their abnormal accumulation in the Aβ plaques along with an overall dyshomeostasis of these metals in the AD brain was proposed a while back. Metal ion chelators and ionophores, put forward as possible drug candidates for AD, are yet to succeed in clinical trials. Heme, which is widely distributed in the mammalian body as the prosthetic group of several important proteins and enzymes, has been thought to be associated with AD by virtue of its colocalization in the Aβ plaques along with the similarity of several heme deficiency symptoms with those of AD and most importantly, due to its ability to bind Aβ. This feature article illustrates the active site environment of heme-Aβ which resembles those of peroxidases. It also discusses the peroxidase activity of heme-Aβ, its ability to effect oxidative degradation of neurotransmitters like serotonin and also the identification of the highly reactive high-valent intermediate, compound I. The effect of second sphere residues on the formation and peroxidase activity of heme-Aβ along with the generation and decay of compound I is highlighted throughout the article. The reactivities of heme bound Aβ peptides give an alternative theory to understand the possible cause of this disease.
Collapse
Affiliation(s)
- Madhuparna Roy
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India.
| | | | | | | |
Collapse
|