1
|
Schwehr BJ, Hartnell D, Ellison G, Hindes MT, Milford B, Dallerba E, Hickey SM, Pfeffer FM, Brooks DA, Massi M, Hackett MJ. Fluorescent probes for neuroscience: imaging ex vivo brain tissue sections. Analyst 2024; 149:4536-4552. [PMID: 39171617 DOI: 10.1039/d4an00663a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Neurobiological research relies heavily on imaging techniques, such as fluorescence microscopy, to understand neurological function and disease processes. However, the number and variety of fluorescent probes available for ex vivo tissue section imaging limits the advance of research in the field. In this review, we outline the current range of fluorescent probes that are available to researchers for ex vivo brain section imaging, including their physical and chemical characteristics, staining targets, and examples of discoveries for which they have been used. This review is organised into sections based on the biological target of the probe, including subcellular organelles, chemical species (e.g., labile metal ions), and pathological phenomenon (e.g., degenerating cells, aggregated proteins). We hope to inspire further development in this field, given the considerable benefits to be gained by the greater availability of suitably sensitive probes that have specificity for important brain tissue targets.
Collapse
Affiliation(s)
- Bradley J Schwehr
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
| | - David Hartnell
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
- Curtin University, Curtin Health Innovation Research Institute, Perth, WA, Australia 6102
| | - Gaewyn Ellison
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
- Curtin University, Curtin Health Innovation Research Institute, Perth, WA, Australia 6102
| | - Madison T Hindes
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000
| | - Breah Milford
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
| | - Elena Dallerba
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
| | - Shane M Hickey
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000
| | - Frederick M Pfeffer
- School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - Doug A Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000
| | - Massimiliano Massi
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
| | - Mark J Hackett
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
- Curtin University, Curtin Health Innovation Research Institute, Perth, WA, Australia 6102
| |
Collapse
|
2
|
Syed RA, Hayat M, Qaiser H, Uzair M, Al-Regaiey K, Khallaf R, Kaleem I, Bashir S. Aging-Related Protein Alterations in the Brain. J Alzheimers Dis 2024; 99:S5-S22. [PMID: 38339930 DOI: 10.3233/jad-230801] [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: 02/12/2024]
Abstract
Aging is an intrinsic aspect of an organism's life cycle and is characterized by progressive physiological decline and increased susceptibility to mortality. Many age-associated disorders, including neurological disorders, are most commonly linked with the aging process, such as Alzheimer's disease (AD). This review aims to provide a comprehensive overview of the effects of aging and AD on the molecular pathways and levels of different proteins in the brain, including metalloproteins, neurotrophic factors, amyloid proteins, and tau proteins. AD is caused by the aggregation of amyloid proteins in the brain. Factors such as metal ions, protein ligands, and the oligomerization state of amyloid precursor protein significantly influence the proteolytic processing of amyloid-β protein precursor (AβPP). Tau, a disordered cytosolic protein, serves as the principal microtubule-associated protein in mature neurons. AD patients exhibit decreased levels of nerve growth factor within their nervous systems and cerebrospinal fluid. Furthermore, a significant increase in brain-derived neurotrophic factor resulting from the neuroprotective effect of glial cell line-derived neurotrophic factor suggests that the synergistic action of these proteins plays a role in inhibiting neuronal degeneration and atrophy. The mechanism through which Aβ and AβPP govern Cu2+ transport and their influence on Cu2+ and other metal ion pools requires elucidation in future studies. A comprehensive understanding of the influence of aging and AD on molecular pathways and varying protein levels may hold the potential for the development of novel diagnostic and therapeutic methods for the treatment of AD.
Collapse
Affiliation(s)
- Rafay Ali Syed
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Mahnoor Hayat
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Hammad Qaiser
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
| | - Mohammad Uzair
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University Islamabad, Pakistan
| | - Khalid Al-Regaiey
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Roaa Khallaf
- Department of Neurology, Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Imdad Kaleem
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam, Saudi Arabia
| |
Collapse
|
3
|
Feng J, She Y, Li C, Shen L. Metal ion mediated aggregation of Alzheimer's disease peptides and proteins in solutions and at surfaces. Adv Colloid Interface Sci 2023; 320:103009. [PMID: 37776735 DOI: 10.1016/j.cis.2023.103009] [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: 06/20/2023] [Revised: 08/29/2023] [Accepted: 09/24/2023] [Indexed: 10/02/2023]
Abstract
Although the pathogenesis of Alzheimer's disease (AD) is still unclear, abnormally high concentrations of metal ions, like copper, iron and zinc, were found in senile plaques of AD brain, which inspires extensive studies on the fundamental molecular interactions of metal ions with the pathogenic hallmarks, amyloid-β (Aβ) peptides and tau proteins, respectively forming senile plaques and neurofibrillary tangles (NFTs) in AD brains. Early works concern the concentration effect of the metal ions on Aβ and tau aggregation. Yet, it is obvious that the surrounding environment of the metal ions must also be considered, not just the metal ions as free accessible forms in the solution phase. The most important surrounding environment in vivo is a very large surface area from cell membranes and other macromolecular surfaces. These bio-interfaces make the kinetic pathways of metal ion mediated Aβ and tau aggregation radically different from those in the solution phase. To better understand the role of metal ions in AD peptide and protein aggregation, we summarize and discuss the recent achievements in the research of metal ion mediated Aβ and tau aggregation, particularly the corresponding mechanism differences between the solution phase and the surface environment. The metal ion chelation therapy for AD is also discussed from the point of the surface pool of metal ions.
Collapse
Affiliation(s)
- Jiahao Feng
- Key Laboratory for Neurodegenerative Diseases Nanomedicine of Hubei Province, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Yifei She
- Key Laboratory for Neurodegenerative Diseases Nanomedicine of Hubei Province, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Chongjia Li
- Key Laboratory for Neurodegenerative Diseases Nanomedicine of Hubei Province, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Lei Shen
- Key Laboratory for Neurodegenerative Diseases Nanomedicine of Hubei Province, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China.
| |
Collapse
|
4
|
Zhang C, Dischler A, Glover K, Qin Y. Neuronal signalling of zinc: from detection and modulation to function. Open Biol 2022; 12:220188. [PMID: 36067793 PMCID: PMC9448499 DOI: 10.1098/rsob.220188] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Zinc is an essential trace element that stabilizes protein structures and allosterically modulates a plethora of enzymes, ion channels and neurotransmitter receptors. Labile zinc (Zn2+) acts as an intracellular and intercellular signalling molecule in response to various stimuli, which is especially important in the central nervous system. Zincergic neurons, characterized by Zn2+ deposits in synaptic vesicles and presynaptic Zn2+ release, are found in the cortex, hippocampus, amygdala, olfactory bulb and spinal cord. To provide an overview of synaptic Zn2+ and intracellular Zn2+ signalling in neurons, the present paper summarizes the fluorescent sensors used to detect Zn2+ signals, the cellular mechanisms regulating the generation and buffering of Zn2+ signals, as well as the current perspectives on their pleiotropic effects on phosphorylation signalling, synapse formation, synaptic plasticity, as well as sensory and cognitive function.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Anna Dischler
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Kaitlyn Glover
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Yan Qin
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| |
Collapse
|
5
|
Kim YM, Park S, Choi SY, Oh SB, Jung M, Pack CG, Hwang JJ, Tak E, Lee JY. Clusterin Binding Modulates the Aggregation and Neurotoxicity of Amyloid-β(1-42). Mol Neurobiol 2022; 59:6228-6244. [PMID: 35904715 DOI: 10.1007/s12035-022-02973-6] [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: 04/18/2022] [Accepted: 07/20/2022] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by the accumulation of amyloid-β (Aβ) aggregates in the brain. Clusterin (CLU), also known as apolipoprotein J, is a potent risk factor associated with AD pathogenesis, in which Aβ aggregation is essentially involved. We observed close colocalization of CLU and Aβ(1-42) (Aβ42) in parenchymal amyloid plaques or vascular amyloid deposits in the brains of human amyloid precursor protein (hAPP)-transgenic Tg2576 mice. Therefore, to elucidate the binding interaction between CLU and Aβ42 and its impact on amyloid aggregation and toxicity, the two synthetic proteins were incubated together under physiological conditions, and their structural and morphological variations were investigated using biochemical, biophysical, and microscopic analyses. Synthetic CLU spontaneously bound to different possible variants of Aβ42 aggregates with very high affinity (Kd = 2.647 nM) in vitro to form solid CLU-Aβ42 complexes. This CLU binding prevented further aggregation of Aβ42 into larger oligomers or fibrils, enriching the population of smaller Aβ42 oligomers and protofibrils and monomers. CLU either alleviated or augmented Aβ42-induced cytotoxicity and apoptosis in the neuroblastoma-derived SH-SY5Y and N2a cells, depending on the incubation period and the molar ratio of CLU:Aβ42 involved in the reaction before addition to the cells. Thus, the effects of CLU on Aβ42-induced cytotoxicity were likely determined by the extent to which it bound and sequestered toxic Aβ42 oligomers or protofibrils. These findings suggest that CLU could influence amyloid neurotoxicity and pathogenesis by modulating Aβ aggregation.
Collapse
Affiliation(s)
- Yun-Mi Kim
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.,Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - SuJi Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Su Yeon Choi
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.,Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Shin Bi Oh
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - MinKyo Jung
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Chan-Gi Pack
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Jung Jin Hwang
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Eunyoung Tak
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Joo-Yong Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea. .,Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| |
Collapse
|
6
|
Zinc in Cognitive Impairment and Aging. Biomolecules 2022; 12:biom12071000. [PMID: 35883555 PMCID: PMC9312494 DOI: 10.3390/biom12071000] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/05/2023] Open
Abstract
Zinc, an essential micronutrient for life, was first discovered in 1869 and later found to be indispensable for the normal development of plants and for the normal growth of rats and birds. Zinc plays an important role in many physiological and pathological processes in normal mammalian brain development, especially in the development of the central nervous system. Zinc deficiency can lead to neurodegenerative diseases, mental abnormalities, sleep disorders, tumors, vascular diseases, and other pathological conditions, which can cause cognitive impairment and premature aging. This study aimed to review the important effects of zinc and zinc-associated proteins in cognitive impairment and aging, to reveal its molecular mechanism, and to highlight potential interventions for zinc-associated aging and cognitive impairments.
Collapse
|
7
|
Chin-Chan M, Montes S, Blanco-Álvarez VM, Aguirre-Alarcón HA, Hernández-Rodríguez I, Bautista E. Relevance of biometals during neuronal differentiation and myelination: in vitro and in vivo studies. Biometals 2022; 35:395-427. [DOI: 10.1007/s10534-022-00380-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 02/27/2022] [Indexed: 12/20/2022]
|
8
|
Kowalczyk A, Gbadamosi O, Kolor K, Sosa J, Andrzejczuk L, Gibson G, Croix C, Chikina M, Aizenman E, Clark N, Kiselyov K. Evolutionary rate covariation identifies SLC30A9 (ZnT9) as a mitochondrial zinc transporter. Biochem J 2021; 478:3205-3220. [PMID: 34397090 PMCID: PMC10491466 DOI: 10.1042/bcj20210342] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022]
Abstract
Recent advances in genome sequencing have led to the identification of new ion and metabolite transporters, many of which have not been characterized. Due to the variety of subcellular localizations, cargo and transport mechanisms, such characterization is a daunting task, and predictive approaches focused on the functional context of transporters are very much needed. Here we present a case for identifying a transporter localization using evolutionary rate covariation (ERC), a computational approach based on pairwise correlations of amino acid sequence evolutionary rates across the mammalian phylogeny. As a case study, we find that poorly characterized transporter SLC30A9 (ZnT9) coevolves with several components of the mitochondrial oxidative phosphorylation chain, suggesting mitochondrial localization. We confirmed this computational finding experimentally using recombinant human SLC30A9. SLC30A9 loss caused zinc mishandling in the mitochondria, suggesting that under normal conditions it acts as a zinc exporter. We therefore propose that ERC can be used to predict the functional context of novel transporters and other poorly characterized proteins.
Collapse
Affiliation(s)
- Amanda Kowalczyk
- Joint Carnegie Mellon University-University of Pittsburgh PhD Program in Computational Biology, Pittsburgh, PA 15213, U.S.A
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, U.S.A
| | - Omotola Gbadamosi
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| | - Kathryn Kolor
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| | - Jahree Sosa
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| | - Livia Andrzejczuk
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| | - Gregory Gibson
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| | - Claudette Croix
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| | - Maria Chikina
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15213, U.S.A
| | - Elias Aizenman
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, U.S.A
| | - Nathan Clark
- Department of Human Genetics, University of Utah, Utah 84112, U.S.A
| | - Kirill Kiselyov
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA 15260, U.S.A
| |
Collapse
|
9
|
Kiyohara ACP, Torres DJ, Hagiwara A, Pak J, Rueli RHLH, Shuttleworth CWR, Bellinger FP. Selenoprotein P Regulates Synaptic Zinc and Reduces Tau Phosphorylation. Front Nutr 2021; 8:683154. [PMID: 34277682 PMCID: PMC8280497 DOI: 10.3389/fnut.2021.683154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 05/26/2021] [Indexed: 01/11/2023] Open
Abstract
Selenoprotein P (SELENOP1) is a selenium-rich antioxidant protein involved in extracellular transport of selenium (Se). SELENOP1 also has metal binding properties. The trace element Zinc (Zn2+) is a neuromodulator that can be released from synaptic terminals in the brain, primarily from a subset of glutamatergic terminals. Both Zn2+ and Se are necessary for normal brain function. Although these ions can bind together with high affinity, the biological significance of an interaction of SELENOP1 with Zn2+ has not been investigated. We examined changes in brain Zn2+ in SELENOP1 knockout (KO) animals. Timm-Danscher and N-(6-methoxy-8-quinolyl)-p-toluenesulphonamide (TSQ) staining revealed increased levels of intracellular Zn2+ in the SELENOP1-/- hippocampus compared to wildtype (WT) mice. Mass spectrometry analysis of frozen whole brain samples demonstrated that total Zn2+ was not increased in the SELENOP1-/- mice, suggesting only local changes in Zn2+ distribution. Unexpectedly, live Zn2+ imaging of hippocampal slices with a selective extracellular fluorescent Zn2+ indicator (FluoZin-3) showed that SELENOP1-/- mice have impaired Zn2+ release in response to KCl-induced neuron depolarization. The zinc/metal storage protein metallothionein 3 (MT-3) was increased in SELENOP1-/- hippocampus relative to wildtype, possibly in response to an elevated Zn2+ content. We found that depriving cultured cells of selenium resulted in increased intracellular Zn2+, as did inhibition of selenoprotein GPX4 but not GPX1, suggesting the increased Zn2+ in SELENOP1-/- mice is due to a downregulation of antioxidant selenoproteins and subsequent release of Zn2+ from intracellular stores. Surprisingly, we found increased tau phosphorylation in the hippocampus of SELENOP1-/- mice, possibly resulting from intracellular zinc changes. Our findings reveal important roles for SELENOP1 in the maintenance of synaptic Zn2+ physiology and preventing tau hyperphosphorylation.
Collapse
Affiliation(s)
- Arlene C. P. Kiyohara
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Daniel J. Torres
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
- Pacific Biosciences Research Center, School of Ocean and Earth Science and Technology, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Ayaka Hagiwara
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Jenna Pak
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Rachel H. L. H. Rueli
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | | | - Frederick P. Bellinger
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| |
Collapse
|
10
|
Abstract
Since the discovery of manifest Zn deficiency in 1961, the increasing number of studies demonstrated the association between altered Zn status and multiple diseases. In this chapter, we provide a review of the most recent advances on the role of Zn in health and disease (2010-20), with a special focus on the role of Zn in neurodegenerative and neurodevelopmental disorders, diabetes and obesity, male and female reproduction, as well as COVID-19. In parallel with the revealed tight association between ASD risk and severity and Zn status, the particular mechanisms linking Zn2+ and ASD pathogenesis like modulation of synaptic plasticity through ProSAP/Shank scaffold, neurotransmitter metabolism, and gut microbiota, have been elucidated. The increasing body of data indicate the potential involvement of Zn2+ metabolism in neurodegeneration. Systemic Zn levels in Alzheimer's and Parkinson's disease were found to be reduced, whereas its sequestration in brain may result in modulation of amyloid β and α-synuclein processing with subsequent toxic effects. Zn2+ was shown to possess adipotropic effects through the role of zinc transporters, zinc finger proteins, and Zn-α2-glycoprotein in adipose tissue physiology, underlying its particular role in pathogenesis of obesity and diabetes mellitus type 2. Recent findings also contribute to further understanding of the role of Zn2+ in spermatogenesis and sperm functioning, as well as oocyte development and fertilization. Finally, Zn2+ was shown to be the potential adjuvant therapy in management of novel coronavirus infection (COVID-19), underlining the perspectives of zinc in management of old and new threats.
Collapse
Affiliation(s)
- Anatoly V Skalny
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia
| | - Michael Aschner
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; Yaroslavl State University, Yaroslavl, Russia.
| |
Collapse
|
11
|
Synaptic Zinc: An Emerging Player in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22094724. [PMID: 33946908 PMCID: PMC8125092 DOI: 10.3390/ijms22094724] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 02/03/2023] Open
Abstract
Alterations of zinc homeostasis have long been implicated in Parkinson's disease (PD). Zinc plays a complex role as both deficiency and excess of intracellular zinc levels have been incriminated in the pathophysiology of the disease. Besides its role in multiple cellular functions, Zn2+ also acts as a synaptic transmitter in the brain. In the forebrain, subset of glutamatergic neurons, namely cortical neurons projecting to the striatum, use Zn2+ as a messenger alongside glutamate. Overactivation of the cortico-striatal glutamatergic system is a key feature contributing to the development of PD symptoms and dopaminergic neurotoxicity. Here, we will cover recent evidence implicating synaptic Zn2+ in the pathophysiology of PD and discuss its potential mechanisms of actions. Emphasis will be placed on the functional interaction between Zn2+ and glutamatergic NMDA receptors, the most extensively studied synaptic target of Zn2+.
Collapse
|
12
|
Datki Z, Galik-Olah Z, Janosi-Mozes E, Szegedi V, Kalman J, Hunya ÁG, Fulop L, Tamano H, Takeda A, Adlard PA, Bush AI. Alzheimer risk factors age and female sex induce cortical Aβ aggregation by raising extracellular zinc. Mol Psychiatry 2020; 25:2728-2741. [PMID: 32518388 DOI: 10.1038/s41380-020-0800-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 05/16/2020] [Accepted: 05/21/2020] [Indexed: 12/20/2022]
Abstract
Aging and female sex are the major risk factors for Alzheimer's disease and its associated brain amyloid-β (Aβ) neuropathology, but the mechanisms mediating these risk factors remain uncertain. Evidence indicates that Aβ aggregation by Zn2+ released from glutamatergic neurons contributes to amyloid neuropathology, so we tested whether aging and sex adversely influences this neurophysiology. Using acute hippocampal slices, we found that extracellular Zn2+-elevation induced by high K+ stimulation was significantly greater with older (65 weeks vs 10 weeks old) rats, and was exaggerated in females. This was driven by slower reuptake of extracellular Zn2+, which could be recapitulated by mitochondrial intoxication. Zn2+:Aβ aggregates were toxic to the slices, but Aβ alone was not. Accordingly, high K+ caused synthetic human Aβ added to the slices to form soluble oligomers as detected by bis-ANS, attaching to neurons and inducing toxicity, with older slices being more vulnerable. Age-dependent energy failure impairing Zn2+ reuptake, and a higher maximal capacity for Zn2+ release by females, could contribute to age and sex being major risk factors for Alzheimer's disease.
Collapse
Affiliation(s)
- Zsolt Datki
- Department of Psychiatry, University of Szeged, Szeged, 6725, Hungary.
| | - Zita Galik-Olah
- Department of Psychiatry, University of Szeged, Szeged, 6725, Hungary
| | | | - Viktor Szegedi
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, 6726, Hungary
| | - Janos Kalman
- Department of Psychiatry, University of Szeged, Szeged, 6725, Hungary
| | - Ákos Gábor Hunya
- Department of Medical Chemistry, University of Szeged, Szeged, 6726, Hungary
| | - Livia Fulop
- Department of Medical Chemistry, University of Szeged, Szeged, 6726, Hungary
| | - Haruna Tamano
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Atsushi Takeda
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Paul A Adlard
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience & Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience & Mental Health, and The University of Melbourne, Parkville, VIC, 3052, Australia.
| |
Collapse
|
13
|
Harris CJ, Gray NE, Caruso M, Hunter M, Ralle M, Quinn JF. Copper Modulation and Memory Impairment due to Hippocampal Tau Pathology. J Alzheimers Dis 2020; 78:49-60. [PMID: 32250309 DOI: 10.3233/jad-200002] [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: 11/15/2022]
Abstract
Background:Environmental copper has been implicated in the pathogenesis of Alzheimer’s disease based on evidence that: 1) brain copper levels increase with age, 2) copper promotes misfolding and toxicity of amyloid-β in vitro, 3) copper-modulating interventions reduce amyloid pathology in animal models. However, the effect of copper upon non-amyloid Alzheimer’s pathology is relatively under-explored.Objective:To determine if modulation of brain copper level affects brain tau pathology and/or associated cognitive impairment.Methods:We tested the hypothesis that brain copper modulates tau pathology by manipulating brain levels of copper in the PS19 transgenic mouse model of tau pathology. We treated PS19 and wild-type mice with oral zinc acetate, an established therapy for long term control of excess brain copper, and examined treatment effects upon brain copper, brain tau, NFT-like pathology, and spatial memory. We treated a second cohort of mice with exogenous dietary copper in order to evaluate whether excess environmental copper promotes brain tau pathology.Results:Copper-lowering with oral zinc attenuated spatial memory impairment in female but not male PS19 mice, without a significant effect upon tau pathology. Copper loading increased brain copper, but did not have an effect on brain tau pathology or spatial memory function.Conclusion:These findings suggest that a strategy to lower brain copper may be viable for symptomatic benefit in the setting of tau neuropathology, but unlikely to have robust effects on the underlying pathology. These findings are consistent with dietary or other exogenous copper being unlikely to promote tau pathology.
Collapse
Affiliation(s)
- Christopher J Harris
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Nora E Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Maya Caruso
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Marguex Hunter
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Martina Ralle
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA.,Parkinsons Disease Research, Education, and Clinical Center, Portland Veterans Affairs Medical Center, Portland, OR, USA
| |
Collapse
|
14
|
Oh SB, Kim JA, Park S, Lee JY. Associative Interactions among Zinc, Apolipoprotein E, and Amyloid-β in the Amyloid Pathology. Int J Mol Sci 2020; 21:ijms21030802. [PMID: 31991844 PMCID: PMC7037199 DOI: 10.3390/ijms21030802] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 01/06/2023] Open
Abstract
Zinc and apolipoprotein E (apoE) are reportedly involved in the pathology of Alzheimer's disease. To investigate the associative interaction among zinc, apoE, and amyloid-β (Aβ) and its role in amyloid pathogenesis, we performed various biochemical and immunoreactive analyses using brain tissues of Tg2576 mice and synthetic Aβ and apoE peptides. On amyloid plaques or in brain lysates of Tg2576 mice, apoE and Aβ immunoreactivities increased after zinc chelation and were restored by its subsequent replacement. Zinc depletion dissociated apoE/Aβ complexes or larger-molecular sizes of Aβ oligomers/aggregates into smaller-molecular sizes of apoE and/or Aβ monomers/complexes. In the presence of zinc, synthetic apoE and/or Aβ peptides aggregated into larger-molecular sizes of oligomers or complexes. Endogenous proteases or plasmin in brain lysates degraded apoE and/or Aβ complexes, and their proteolytic activity increased with zinc depletion. These biochemical findings suggest that zinc associates with apoE and Aβ to encourage the formation of apoE/Aβ complexes or large aggregates, raising the deposition of zinc-rich amyloid plaques. In turn, the presence of abundant zinc around and within apoE/Aβ complexes may block the access or activity of Aβ-degrading antibodies or proteases. These results support the plausibility of chelation strategy aiming at reducing amyloid pathology in Alzheimer's disease.
Collapse
Affiliation(s)
- Shin Bi Oh
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (S.B.O.); (J.A.K.); (S.P.)
| | - Jung Ah Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (S.B.O.); (J.A.K.); (S.P.)
| | - SuJi Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (S.B.O.); (J.A.K.); (S.P.)
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Joo-Yong Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (S.B.O.); (J.A.K.); (S.P.)
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Korea
- Correspondence: ; Tel.: +82-2-3010-4143; Fax: +82-2-3010-4680
| |
Collapse
|
15
|
Wijasa TS, Sylvester M, Brocke-Ahmadinejad N, Schwartz S, Santarelli F, Gieselmann V, Klockgether T, Brosseron F, Heneka MT. Quantitative proteomics of synaptosome S-nitrosylation in Alzheimer's disease. J Neurochem 2019; 152:710-726. [PMID: 31520481 DOI: 10.1111/jnc.14870] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/23/2019] [Accepted: 09/04/2019] [Indexed: 12/20/2022]
Abstract
Increasing evidence suggests that both synaptic loss and neuroinflammation constitute early pathologic hallmarks of Alzheimer's disease. A downstream event during inflammatory activation of microglia and astrocytes is the induction of nitric oxide synthase type 2, resulting in an increased release of nitric oxide and the post-translational S-nitrosylation of protein cysteine residues. Both early events, inflammation and synaptic dysfunction, could be connected if this excess nitrosylation occurs on synaptic proteins. In the long term, such changes could provide new insight into patho-mechanisms as well as biomarker candidates from the early stages of disease progression. This study investigated S-nitrosylation in synaptosomal proteins isolated from APP/PS1 model mice in comparison to wild type and NOS2-/- mice, as well as human control, mild cognitive impairment and Alzheimer's disease brain tissues. Proteomics data were obtained using an established protocol utilizing an isobaric mass tag method, followed by nanocapillary high performance liquid chromatography tandem mass spectrometry. Statistical analysis identified the S-nitrosylation sites most likely derived from an increase in nitric oxide (NO) in dependence of presence of AD pathology, age and the key enzyme NOS2. The resulting list of candidate proteins is discussed considering function, previous findings in the context of neurodegeneration, and the potential for further validation studies.
Collapse
Affiliation(s)
| | - Marc Sylvester
- Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany
| | | | - Stephanie Schwartz
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | | | - Volkmar Gieselmann
- Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany
| | - Thomas Klockgether
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurology, University of Bonn, Bonn, Germany
| | | | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
16
|
Tian ZY, Wang CY, Wang T, Li YC, Wang ZY. Glial S100A6 Degrades β-amyloid Aggregation through Targeting Competition with Zinc Ions. Aging Dis 2019; 10:756-769. [PMID: 31440382 PMCID: PMC6675528 DOI: 10.14336/ad.2018.0912] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/12/2018] [Indexed: 01/02/2023] Open
Abstract
Evidence has been accumulating that zinc ions can trigger β-amyloid (Aβ) deposition and senile plaque formation in the brain, a pathological hallmark of Alzheimer's disease (AD). Chelating zinc inhibits Aβ aggregation and may hold promise as a therapeutic strategy for AD. S100A6 is an acidic Ca2+/Zn2+-binding protein found only in a small number of astrocytes in the normal brain. However, in the AD brain, S100A6 is highly expressed in astrocytes around Aβ plaques. The role of the astrocytic S100A6 upregulation in AD is unknown. In the present study, we examined the effects of S100A6 on Aβ plaques and intracellular zinc levels in a mouse model of AD. Chronic exposure to zinc increased Aβ deposition and S100A6 expression, both reversible by the zinc chelator clioquinol, in the brains of amyloid precursor protein/presenilin 1 (APP/PS1) transgenic mice. To examine whether exogenous S100A6 could induce Aβ plaque disaggregation through competition for zinc in vitro, we incubated APP/PS1 mouse brain sections with recombinant human S100A6 protein or co-incubated them with human S100A6-expressing cells. Both treatments efficiently reduced the Aβ plaque burden in situ. In addition, treatment with exogenous S100A6 protected cultured COS-7 cells against zinc toxicity. Our results show for the first time that increased S100A6 levels correlate with both Aβ disaggregation and decrease of Aβ plaque-associated zinc contents in brain sections with AD-like pathology. Astrocytic S100A6 in AD may protect from Aβ deposition through zinc sequestration.
Collapse
Affiliation(s)
- Zhi-Ying Tian
- 1Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang 110122, China
| | - Chun-Yan Wang
- 1Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang 110122, China
| | - Tao Wang
- 1Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang 110122, China
| | - Yan-Chun Li
- 2Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Zhan-You Wang
- 1Institute of Health Sciences, Key Laboratory of Medical Cell Biology of Ministry of Education, China Medical University, Shenyang 110122, China
| |
Collapse
|
17
|
Oh SB, Kim MS, Park S, Son H, Kim SY, Kim MS, Jo DG, Tak E, Lee JY. Clusterin contributes to early stage of Alzheimer's disease pathogenesis. Brain Pathol 2018; 29:217-231. [PMID: 30295351 DOI: 10.1111/bpa.12660] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/28/2018] [Indexed: 01/27/2023] Open
Abstract
While clusterin is reportedly involved in Alzheimer's disease (AD) pathogenesis, how clusterin interacts with amyloid-β (Aß) to cause Aß neurotoxicity remains unclear in vivo. Using 5×FAD transgenic mice, which develop robust AD pathology and memory deficits when very young, we detected interactions between clusterin and Aß in the mouse brains. The two proteins were concurrently upregulated and bound or colocalized with each other in the same complexes or in amyloid plaques. Neuropathology and cognitive performance were assessed in the progeny of clusterin-null mice crossed with 5×FAD mice, yielding clu-/- ;5×FAD and clu+/+ ;5×FAD. We found far less of the various pools of Aß proteins, most strikingly soluble Aß oligomers and amyloid plaques in clu-/- ;5×FAD mice at 5 months of age. At that age, those mice also had higher levels of neuronal and synaptic proteins and better motor coordination, spatial learning and memory than age-matched clu+/+ ;5×FAD mice. However, at 10 months of age, these differences disappeared, with Aß and plaque deposition, neuronal and synaptic proteins and impairment of behavioral and cognitive performance similar in both groups. These findings demonstrate that clusterin is necessarily involved in early stages of AD pathogenesis by enhancing toxic Aß pools to cause Aß-directed neurodegeneration and behavioral and cognitive impairments, but not in late stage.
Collapse
Affiliation(s)
- Shin-Bi Oh
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Min Sun Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - SuJi Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - HyunJu Son
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Seog-Young Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min-Seon Kim
- Department of Endocrinology and Metabolism, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Gyu Jo
- The School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Eunyoung Tak
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joo-Yong Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
18
|
Essential Role of Zinc and Zinc Transporters in Myeloid Cell Function and Host Defense against Infection. J Immunol Res 2018; 2018:4315140. [PMID: 30417019 PMCID: PMC6207864 DOI: 10.1155/2018/4315140] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/27/2018] [Indexed: 12/19/2022] Open
Abstract
Zinc is an essential micronutrient known to play a vital role in host defense against pathogens. Diets that are deficient in zinc lead to impaired immunity and delayed recovery from and worse outcomes following infection. Sustained insufficient zinc intake leads to dysregulation of the innate immune response and increases susceptibility to infection whereas zinc supplementation in at-risk populations has been shown to restore host defense and reduce pathogen-related morbidity and mortality. Upon infection, zinc deficiency leads to increased pathology due to imbalance in key signaling networks that result in excessive inflammation and collateral tissue damage. In particular, zinc impacts macrophage function, a critical front-line cell in host defense, in addition to other immune cells. Deficits in zinc adversely impact macrophage function resulting in dysregulation of phagocytosis, intracellular killing, and cytokine production. An additional work in this field has revealed a vital role for several zinc transporter proteins that are required for proper bioredistribution of zinc within mononuclear cells to achieve an optimal immune response against invading microorganisms. In this review, we will discuss the most recent developments regarding zinc's role in innate immunity and protection against pathogen invasion.
Collapse
|
19
|
Aratake T, Higashi Y, Ueba Y, Hamada T, Shimizu T, Shimizu S, Yawata T, Ueba T, Saito M. The inhibitory role of intracellular free zinc in the regulation of Arg-1 expression in interleukin-4-induced activation of M2 microglia. Metallomics 2018; 10:1501-1509. [PMID: 30206632 DOI: 10.1039/c8mt00248g] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Microglia, the resident immune cells of the central nervous system, can display a pro-inflammatory M1 phenotype or an anti-inflammatory M2 phenotype. Arginase (Arg)-1 expressed in interleukin-4 (IL-4)-induced M2 microglia reduces nitric oxide (NO) production by competing with inducible NO synthase for l-arginine, which contributes to the attenuation of brain inflammation. Although previous studies have indicated that brain zinc promotes M1 activation, the effect of zinc on M2 microglial activation remains to be determined. In the present study, murine primary microglia treated with 10 ng mL-1 IL-4 exhibited increased Arg-1 mRNA expression and levels of intracellular free zinc. Chelation of this increased intracellular free zinc by the cell permeable zinc chelator N,N,N',N'-tetrakis-(2-pyridylmethyl)ethylenediamine (TPEN) aggravated the IL-4-induced mRNA expression and enzymatic activity of Arg-1. However, the cell impermeable zinc chelator CaEDTA had no effect on Arg-1 expression or cytosolic levels of free zinc in IL-4-induced M2-polarized microglia. Furthermore, treatment with IL-4 resulted in upregulation of phagocytic activity in microglia, while administration of TPEN abolished IL-4-induced phagocytic activity. Moreover, this effect was reversed vial-arginine supplementation. These findings suggest that IL-4 induces an increase in intracellular free zinc in microglia, which may act as a negative regulator of IL-4-induced Arg-1 expression, and that such negative regulation is essential for microglial phagocytic activity.
Collapse
Affiliation(s)
- Takaaki Aratake
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Youichirou Higashi
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Yusuke Ueba
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Tomoya Hamada
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Takahiro Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Shogo Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Toshio Yawata
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Tetsuya Ueba
- Department of Neurosurgery, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| | - Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University, Kohasu, Okoh-cho, Nankoku 783-8505, Japan.
| |
Collapse
|
20
|
McAllister BB, Dyck RH. Zinc transporter 3 (ZnT3) and vesicular zinc in central nervous system function. Neurosci Biobehav Rev 2017. [DOI: 10.1016/j.neubiorev.2017.06.006] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
21
|
Bielarczyk H, Jankowska-Kulawy A, Höfling C, Ronowska A, Gul-Hinc S, Roßner S, Schliebs R, Pawelczyk T, Szutowicz A. AβPP-Transgenic 2576 Mice Mimic Cell Type-Specific Aspects of Acetyl-CoA-Linked Metabolic Deficits in Alzheimer's Disease. J Alzheimers Dis 2016; 48:1083-94. [PMID: 26402099 DOI: 10.3233/jad-150327] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The pyruvate-derived acetyl-CoA is a principal direct precursor substrate for bulk energy synthesis in the brain. Deficits of pyruvate dehydrogenase in the neocortex are common features of Alzheimer's disease and other age-related encephalopathies in humans. Therefore, amyloid-β overload in brains of diverse transgenic Alzheimer's disease model animals was investigated as one of neurotoxic compounds responsible for pyruvate dehydrogenase inhibition yielding deficits of cholinergic neurotransmission and cognitive functions. Brains of aged, 14-16-month-old Tg2576 mice contained 0.6 μmol/kg levels of amyloid-β1 - 42. Activities of pyruvate dehydrogenase complex, choline acetyltransferase, and several enzymes of acetyl-CoA and energy metabolism were found to be unchanged in both forebrain mitochondria and synaptosomes of Tg2576 mice, indicating preservation of structural integrity at least in cholinergic neuronal cells. However, in transgenic brain synaptosomes, pyruvate utilization, mitochondrial levels, and cytoplasmic acetyl-CoA levels, as well as acetylcholine content and its quantal release, were all found to be decreased by 25-40% . On the contrary, activation of pyruvate utilization was detected and no alterations in acetyl-CoA content and citrate or α-ketoglutarate accumulation were observed in transgenic whole brain mitochondria. These data indicate that amyloid-β evoked deficits in acetyl-CoA are confined to mitochondrial and cytoplasmic compartments of Tg2576 nerve terminals, becoming early primary signals paving the path for further stages of neurodegeneration. On the other hand, acetyl-CoA synthesis in mitochondrial compartments of glial cells seems to be activated despite amyloid-β accumulated in transgenic brains.
Collapse
Affiliation(s)
- Hanna Bielarczyk
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Corinna Höfling
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Anna Ronowska
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Sylwia Gul-Hinc
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Reinhard Schliebs
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Tadeusz Pawelczyk
- Department of Molecular Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Andrzej Szutowicz
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
22
|
Metals and Neuronal Metal Binding Proteins Implicated in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9812178. [PMID: 26881049 PMCID: PMC4736980 DOI: 10.1155/2016/9812178] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/17/2015] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent age-related dementia affecting millions of people worldwide. Its main pathological hallmark feature is the formation of insoluble protein deposits of amyloid-β and hyperphosphorylated tau protein into extracellular plaques and intracellular neurofibrillary tangles, respectively. Many of the mechanistic details of this process remain unknown, but a well-established consequence of protein aggregation is synapse dysfunction and neuronal loss in the AD brain. Different pathways including mitochondrial dysfunction, oxidative stress, inflammation, and metal metabolism have been suggested to be implicated in this process. In particular, a body of evidence suggests that neuronal metal ions such as copper, zinc, and iron play important roles in brain function in health and disease states and altered homeostasis and distribution as a common feature across different neurodegenerative diseases and aging. In this focused review, we overview neuronal proteins that are involved in AD and whose metal binding properties may underlie important biochemical and regulatory processes occurring in the brain during the AD pathophysiological process.
Collapse
|
23
|
Impacts of aging and amyloid-β deposition on plasminogen activators and plasminogen activator inhibitor-1 in the Tg2576 mouse model of Alzheimer's disease. Brain Res 2014; 1597:159-67. [PMID: 25454795 DOI: 10.1016/j.brainres.2014.11.042] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 11/19/2014] [Accepted: 11/20/2014] [Indexed: 11/21/2022]
Abstract
Plasminogen activators (PAs), which convert plasminogen into the fibrinolytic protease plasmin, may initiate the degradation of amyloid-β (Aβ) to suppress the amyloid pathogenesis. In that way, tissue plasminogen activator (tPA)-mediated plasmin activation could maintain a low level of Aβ deposition to delay the pathogenesis of Alzheimer's disease (AD). In a previous study, we reported that tPA/plasmin proteolytic activity is attenuated throughout the brain during aging or with Aβ accumulation but clustered intense around the amyloid plaques in AD brain. The present study demonstrates that the altered proteolytic activity primarily results from the competition between the expressions of tPA and plasminogen activator inhibitor-1 (PAI-1) in the brains of Tg2576 Aβ-transgenic mice, as revealed by immunohistochemistry and immunoblot assays. Compared with that in the brains of younger Tg2576 mice, tPA protein is generally reduced throughout the brain in older Tg2576 mice but elevated near amyloid plaques. In contrary, PAI-1 expression increases during aging or Aβ deposition with its clusters surrounding amyloid plaques. No significant alteration in the expression of urokinase plasminogen activator (uPA) is detected. These results suggest reciprocal feedback influences between tPA, PAI-1 and Aβ during aging and amyloid pathogenesis in AD brain; tPA-mediated plasmin activity is declined throughout the brain causing Aβ deposition during aging, and the Aβ deposits locally attract the cluster of tPA and/or PAI-1 around their deposits to competitively determine tPA/plasmin-mediated Aβ proteolysis.
Collapse
|
24
|
Flinn JM, Bozzelli PL, Adlard PA, Railey AM. Spatial memory deficits in a mouse model of late-onset Alzheimer's disease are caused by zinc supplementation and correlate with amyloid-beta levels. Front Aging Neurosci 2014; 6:174. [PMID: 25374537 PMCID: PMC4205817 DOI: 10.3389/fnagi.2014.00174] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 07/01/2014] [Indexed: 11/23/2022] Open
Abstract
Much of the research in Alzheimer’s disease (AD) that uses mouse models focuses on the early-onset form of the disease, which accounts for less than 5% of cases. In contrast, this study used a late-onset AD model to examine the interaction between increased dietary zinc (Zn) and the apolipoprotein E (ApoE) gene. ApoE ε4 is overrepresented in late-onset AD and enhances Zn binding to amyloid-β (Aβ). This study sought to determine if elevated dietary Zn would impair spatial memory in CRND8 mice (CRND8), as well as mice who carry both the mutated human amyloid precursor protein (APP) and ApoE ε4 genes (CRND8/E4). Mice were provided with either lab tap water or water enhanced with 10 ppm Zn (ZnCO3) for 4 months. At 6 months of age, spatial memory was measured by the Barnes maze. CRND8 mice exhibited significant memory deficits compared to WT mice, as shown by an increased latency to reach the escape box. For the CRND8/E4, but not the CRND8 mice, those given Zn water made significantly more errors than those on lab water. During the probe trial for the WT group, those on Zn water spent significantly less time in the target quadrant than those on lab water. These data suggest that increased dietary Zn can significantly impair spatial memory in CRND8/E4. WT mice given Zn water were also impaired on the 24-h probe trial when compared to lab water WTs. Within the CRND8/E4 group only, levels of soluble Aβ were significantly correlated with average primary latencies. Within the Zn-treated CRND8/E4 group, there was a significant correlation between insoluble Aβ and average primary errors. Levels of the zinc transporter 3, ZnT3, were negatively correlated with soluble Aβ (p < 0.01). These findings are particularly relevant because increased intake of dietary supplements, such as Zn, are common in the elderly—a population already at risk for AD. Given the effects observed in the CRND8/E4 mice, ApoE status should be taken into consideration when evaluating the efficacy of therapies targeting metals.
Collapse
Affiliation(s)
- Jane M Flinn
- Department of Psychology, George Mason University Fairfax, VA, USA
| | | | - Paul A Adlard
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia
| | - Angela M Railey
- Department of Psychology, George Mason University Fairfax, VA, USA
| |
Collapse
|
25
|
Hancock SM, Finkelstein DI, Adlard PA. Glia and zinc in ageing and Alzheimer's disease: a mechanism for cognitive decline? Front Aging Neurosci 2014; 6:137. [PMID: 25009495 PMCID: PMC4069481 DOI: 10.3389/fnagi.2014.00137] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 06/09/2014] [Indexed: 11/13/2022] Open
Abstract
Normal ageing is characterized by cognitive decline across a range of neurological functions, which are further impaired in Alzheimer’s disease (AD). Recently, alterations in zinc (Zn) concentrations, particularly at the synapse, have emerged as a potential mechanism underlying the cognitive changes that occur in both ageing and AD. Zn is now accepted as a potent neuromodulator, affecting a variety of signaling pathways at the synapse that are critical to normal cognition. While the focus has principally been on the neuron: Zn interaction, there is a growing literature suggesting that glia may also play a modulatory role in maintaining both Zn ion homeostasis and the normal function of the synapse. Indeed, zinc transporters (ZnT’s) have been demonstrated in glial cells where Zn has also been shown to have a role in signaling. Furthermore, there is increasing evidence that the pathogenesis of AD critically involves glial cells (such as astrocytes), which have been reported to contribute to amyloid-beta (Aβ) neurotoxicity. This review discusses the current evidence supporting a complex interplay of glia, Zn dyshomeostasis and synaptic function in ageing and AD.
Collapse
Affiliation(s)
- Sara M Hancock
- Synaptic Neurobiology Laboratory, Florey Institute of Neuroscience and Mental Health Parkville, VIC, Australia
| | - David I Finkelstein
- Parkinson's Disease Laboratory, Florey Institute of Neuroscience and Mental Health Parkville, VIC, Australia
| | - Paul A Adlard
- Synaptic Neurobiology Laboratory, Florey Institute of Neuroscience and Mental Health Parkville, VIC, Australia
| |
Collapse
|
26
|
Takeda A, Fujii H, Minamino T, Tamano H. Intracellular Zn(2+) signaling in cognition. J Neurosci Res 2014; 92:819-24. [PMID: 24723300 DOI: 10.1002/jnr.23385] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/30/2014] [Accepted: 02/21/2014] [Indexed: 12/22/2022]
Abstract
Brain zinc homeostasis is strictly controlled under healthy conditions, indicating the importance of zinc for physiological function in the brain. A part of zinc in the brain exists in the synaptic vesicles, is released from a subclass of glutamatergic neurons (i.e., zincergic neurons), and serves as a signal factor (Zn(2+) signal) in the intracellular (cytosol) compartment as well as in the extracellular compartment. Zn(2+) signaling is dynamically linked to glutamate signaling and may be involved in synaptic plasticity, such as long-term potentiaion and cognitive activity. In zincergic synapses, intracellular Zn(2+) signaling in the postsynaptic neurons, which is linked to Zn(2+) release from zincergic neuron terminals, plays a role in cognitive activity. When nonzincergic synapses participate in cognition, on the other hand, it is possible that intracellular Zn(2+) signaling, which is due mainly to Zn(2+) release from the internal stores and/or metallothioneins, also is involved in cognitive activity, because zinc-dependent system such as zinc-binding proteins is usually required for cognitive process. Intracellular Zn(2+) dynamics may be modified via an endocrine system activity, glucocorticoid secretion in both zincergic and nonzincergic neurons, which is linked to a long-lasting change in synaptic efficacy. On the basis of the evidence of cognitive decline caused by the lack and/or the blockade of synaptic Zn(2+) signaling, this article summarizes the involvement of intracellular Zn(2+) signaling in zincergic synapses in cognition and a hypothetical involvement of that in nonzincergic synapses.
Collapse
Affiliation(s)
- Atsushi Takeda
- Department of Bioorganic Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan; Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | | | | | | |
Collapse
|
27
|
Oh SB, Byun CJ, Yun JH, Jo DG, Carmeliet P, Koh JY, Lee JY. Tissue plasminogen activator arrests Alzheimer's disease pathogenesis. Neurobiol Aging 2014; 35:511-9. [DOI: 10.1016/j.neurobiolaging.2013.09.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 10/26/2022]
|
28
|
Takeda A, Tamano H. Cognitive decline due to excess synaptic Zn(2+) signaling in the hippocampus. Front Aging Neurosci 2014; 6:26. [PMID: 24578691 PMCID: PMC3936311 DOI: 10.3389/fnagi.2014.00026] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/13/2014] [Indexed: 12/02/2022] Open
Abstract
Zinc is an essential component of physiological brain function. Vesicular zinc is released from glutamatergic (zincergic) neuron terminals and serves as a signal factor (Zn2+ signal) in both the intracellular (cytosol) compartment and the extracellular compartment. Synaptic Zn2+ signaling is dynamically linked to neurotransmission and is involved in processes of synaptic plasticity such as long-term potentiation and cognitive activity. On the other hand, the activity of the hypothalamic–pituitary–adrenal (HPA) axis, i.e., glucocorticoid secretion, which can potentiate glutamatergic neuron activity, is linked to cognitive function. HPA axis activity modifies synaptic Zn2+ dynamics at zincergic synapses. An increase in HPA axis activity, which occurs after exposure to stress, may induce excess intracellular Zn2+ signaling in the hippocampus, followed by hippocampus-dependent memory deficit. Excessive excitation of zincergic neurons in the hippocampus can contribute to cognitive decline under stressful and/or pathological conditions. This paper provides an overview of the ``Hypothesis and Theory'' of Zn2+-mediated modification of cognitive activity.
Collapse
Affiliation(s)
- Atsushi Takeda
- Department of Bioorganic Chemistry, School of Pharmaceutical Sciences, University of Shizuoka Shizuoka, Japan
| | - Haruna Tamano
- Department of Bioorganic Chemistry, School of Pharmaceutical Sciences, University of Shizuoka Shizuoka, Japan
| |
Collapse
|
29
|
Greenough MA, Camakaris J, Bush AI. Metal dyshomeostasis and oxidative stress in Alzheimer’s disease. Neurochem Int 2013; 62:540-55. [DOI: 10.1016/j.neuint.2012.08.014] [Citation(s) in RCA: 266] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/13/2012] [Accepted: 08/30/2012] [Indexed: 01/21/2023]
|
30
|
Takeda A, Nakamura M, Fujii H, Tamano H. Synaptic Zn2+ homeostasis and its significance. Metallomics 2013; 5:417-23. [DOI: 10.1039/c3mt20269k] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
31
|
Jellinger KA. The relevance of metals in the pathophysiology of neurodegeneration, pathological considerations. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2013; 110:1-47. [PMID: 24209432 DOI: 10.1016/b978-0-12-410502-7.00002-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurodegenerative disorders are featured by a variety of pathological conditions that share similar critical processes, such as oxidative stress, free radical activity, proteinaceous aggregations, mitochondrial dysfunctions, and energy failure. They are mediated or triggered by an imbalance of metal ions leading to changes of critical biological systems and initiating a cascade of events finally leading to neurodegeneration and cell death. Their causes are multifactorial, and although the source of the shift in oxidative homeostasis is still unclear, current evidence points to changes in the balance of redox transition metals, especially iron, copper, and other trace metals. They are present at elevated levels in Alzheimer disease, Parkinson disease, multisystem atrophy, etc., while in other neurodegenerative disorders, copper, zinc, aluminum, and manganese are involved. This chapter will review the recent advances of the role of metals in the pathogenesis and pathophysiology of major neurodegenerative diseases and discuss the use of chelating agents as potential therapies for metal-related disorders.
Collapse
|