1
|
Medvedeva YV, Sharman E, Weiss JH. Mechanisms of delayed ischemia/reperfusion evoked ROS generation in the hippocampal CA1 zone of adult mouse brain slices. RESEARCH SQUARE 2025:rs.3.rs-5640324. [PMID: 40313775 PMCID: PMC12045377 DOI: 10.21203/rs.3.rs-5640324/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
ROS overproduction is an important contributor to delayed ischemia/reperfusion induced neuronal injury, but relevant mechanisms remain poorly understood. We used oxygen-glucose deprivation (OGD)/reperfusion in mouse hippocampal slices to investigate ROS production in the CA1 pyramidal cell layer during and after transient ischemia. OGD evoked a 2-stage increase in ROS production: 1st - an abrupt increase in ROS generation starting during OGD followed by a marked slowing; and 2nd - a sharp ROS burst starting ~ 40 min after reperfusion. We further found that a slight mitochondrial hyperpolarization occurs shortly after OGD termination. Consequently, we showed that administration of low dose FCCP or of FTY720 (both of which cause mild, ~ 10%, mitochondrial depolarization), markedly diminished the delayed ROS burst, suggesting that mitochondrial hyperpolarization contributes to ROS production after reperfusion. Zn2+ chelation after OGD withdrawal also substantially decreased the late surge of ROS generation- in line with our prior studies indicating a critical contribution of Zn2+ entry into mitochondria via the mitochondrial Ca2+ uniporter (MCU) to mitochondrial damage after OGD. Thus, reperfusion-induced mitochondria hyperpolarization and mitochondrial Zn2+ accumulation both contribute to mitochondrial ROS overproduction after ischemia. As these events occur after reperfusion, they may be amenable to therapeutic interventions.
Collapse
|
2
|
Obeng E, Shen B, Wang W, Xie Z, Zhang W, Li Z, Yao Q, Wu W. Engineered bio-functional material-based nerve guide conduits for optic nerve regeneration: a view from the cellular perspective, challenges and the future outlook. Regen Biomater 2024; 12:rbae133. [PMID: 39776856 PMCID: PMC11703557 DOI: 10.1093/rb/rbae133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/16/2024] [Accepted: 11/03/2024] [Indexed: 01/11/2025] Open
Abstract
Nerve injuries can be tantamount to severe impairment, standard treatment such as the use of autograft or surgery comes with complications and confers a shortened relief. The mechanism relevant to the regeneration of the optic nerve seems yet to be fully uncovered. The prevailing rate of vision loss as a result of direct or indirect insult on the optic nerve is alarming. Currently, the use of nerve guide conduits (NGC) to some extent has proven reliable especially in rodents and among the peripheral nervous system, a promising ground for regeneration and functional recovery, however in the optic nerve, this NGC function seems quite unfamous. The insufficient NGC application and the unabridged regeneration of the optic nerve could be a result of the limited information on cellular and molecular activities. This review seeks to tackle two major factors (i) the cellular and molecular activity involved in traumatic optic neuropathy and (ii) the NGC application for the optic nerve regeneration. The understanding of cellular and molecular concepts encompassed, ocular inflammation, extrinsic signaling and intrinsic signaling for axon growth, mobile zinc role, Ca2+ factor associated with the optic nerve, alternative therapies from nanotechnology based on the molecular information and finally the nanotechnological outlook encompassing applicable biomaterials and the use of NGC for regeneration. The challenges and future outlook regarding optic nerve regenerations are also discussed. Upon the many approaches used, the comprehensive role of the cellular and molecular mechanism may set grounds for the efficient application of the NGC for optic nerve regeneration.
Collapse
Affiliation(s)
- Enoch Obeng
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Baoguo Shen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Zhenyuan Xie
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Wenyi Zhang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Zhixing Li
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Qinqin Yao
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Wencan Wu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou 325027, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang 325000, China
| |
Collapse
|
3
|
Alves JL, Reis PM, Quinta-Ferreira RM, Quinta-Ferreira ME, Matias CM. Changes in reactive oxygen species and autofluorescence under hypoxia at the hippocampal CA3 area: Role of calcium and zinc influxes. Neurochem Int 2024; 180:105882. [PMID: 39413928 DOI: 10.1016/j.neuint.2024.105882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/12/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Reactive oxygen species (ROS) have an important role in cellular biology, being involved, in a way that depends on their levels, in cell signaling processes or in oxidative stress, probably associated with neurodegenerative and other diseases. Most of the studies about ROS formation were performed in ischemic conditions, and thus, there is limited knowledge about ROS formation in less severe hypoxic conditions. This study investigates neuronal ROS generation and autofluorescence changes in hypoxic conditions, focusing on the involvement of calcium and zinc. Using hippocampal slices from Wistar rats, ROS production was monitored by the permeant fluorescent indicator H2DCFDA under different oxygenation levels. Moderate hypoxia (40% O2) led to a small ROS increase, while severe hypoxia (0% O2) showed a more pronounced rise. KCl-induced depolarization significantly enhanced ROS formation, particularly under severe hypoxia. Inhibition of NMDA receptors reduced ROS generation without affecting autofluorescence, while chelation of zinc ions decreased ROS production and increased flavin adenine dinucleotide (FAD) autofluorescence. These findings suggest that, in hypoxic conditions, ROS formation is mediated by calcium entry through NMDA receptors and also by zinc influxes. Thus, these ions play a crucial role in oxidative stress, which may be related with neurodegenerative diseases associated with ROS dysregulation.
Collapse
Affiliation(s)
- João L Alves
- Department of Life Sciences, University of Coimbra, Portugal; CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal
| | - Patrícia M Reis
- CERES - Chemical Engineering and Renewable Resources for Sustainability, Department of Chemical Engineering, University of Coimbra, Portugal
| | - Rosa M Quinta-Ferreira
- CERES - Chemical Engineering and Renewable Resources for Sustainability, Department of Chemical Engineering, University of Coimbra, Portugal
| | - M Emília Quinta-Ferreira
- CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; Department of Physics, University of Coimbra, Portugal
| | - Carlos M Matias
- CNC-UC - Center for Neurosciences and Cell Biology, University of Coimbra, Portugal; Department of Physics, UTAD, Vila Real, Portugal.
| |
Collapse
|
4
|
Bennett MC, Reinhart KM, Weisend JE, Morton RA, Carlson AP, Shuttleworth CW. Synaptic Zn 2+ contributes to deleterious consequences of spreading depolarizations. Neurobiol Dis 2024; 191:106407. [PMID: 38199272 PMCID: PMC10869643 DOI: 10.1016/j.nbd.2024.106407] [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: 10/26/2023] [Revised: 12/23/2023] [Accepted: 01/07/2024] [Indexed: 01/12/2024] Open
Abstract
Spreading depolarizations (SDs) are profound waves of neuroglial depolarization that can propagate repetitively through injured brain. Recent clinical work has established SD as an important contributor to expansion of acute brain injuries and have begun to extend SD studies into other neurological disorders. A critical challenge is to determine how to selectively prevent deleterious consequences of SD. In the present study, we determined whether a wave of profound Zn2+ release is a key contributor to deleterious consequences of SD, and whether this can be targeted pharmacologically. Focal KCl microinjection was used to initiate SD in the CA1 region of the hippocampus in murine brain slices. An extracellular Zn2+ chelator with rapid kinetics (ZX1) increased SD propagation rates and improved recovery of extracellular DC potential shifts. Under conditions of metabolic compromise, tissues showed sustained impairment of functional and structural recovery following a single SD. ZX1 effectively improved recovery of synaptic potentials and intrinsic optical signals in these vulnerable conditions. Fluorescence imaging and genetic deletion of a presynaptic Zn2+ transporter confirmed synaptic release as the primary contributor to extracellular accumulation and deleterious consequences of Zn2+ during SD. These results demonstrate a role for synaptic Zn2+ release in deleterious consequences of SD and show that targeted extracellular chelation could be useful for disorders where repetitive SD enlarges infarcts in injured tissues.
Collapse
Affiliation(s)
- Michael C Bennett
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Katelyn M Reinhart
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Jordan E Weisend
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Russell A Morton
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Andrew P Carlson
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - C William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA.
| |
Collapse
|
5
|
Boyle BR, Berghella AP, Blanco-Suarez E. Astrocyte Regulation of Neuronal Function and Survival in Stroke Pathophysiology. ADVANCES IN NEUROBIOLOGY 2024; 39:233-267. [PMID: 39190078 DOI: 10.1007/978-3-031-64839-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The interactions between astrocytes and neurons in the context of stroke play crucial roles in the disease's progression and eventual outcomes. After a stroke, astrocytes undergo significant changes in their morphology, molecular profile, and function, together termed reactive astrogliosis. Many of these changes modulate how astrocytes relate to neurons, inducing mechanisms both beneficial and detrimental to stroke recovery. For example, excessive glutamate release and astrocytic malfunction contribute to excitotoxicity in stroke, eventually causing neuronal death. Astrocytes also provide essential metabolic support and neurotrophic signals to neurons after stroke, ensuring homeostatic stability and promoting neuronal survival. Furthermore, several astrocyte-secreted molecules regulate synaptic plasticity in response to stroke, allowing for the rewiring of neural circuits to compensate for damaged areas. In this chapter, we highlight the current understanding of the interactions between astrocytes and neurons in response to stroke, explaining the varied mechanisms contributing to injury progression and the potential implications for future therapeutic interventions.
Collapse
Affiliation(s)
- Bridget R Boyle
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrea P Berghella
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Elena Blanco-Suarez
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Neurological Surgery, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Yang F, Smith MJ. Metal profiling in coronary ischemia-reperfusion injury: Implications for KEAP1/NRF2 regulated redox signaling. Free Radic Biol Med 2024; 210:158-171. [PMID: 37989446 DOI: 10.1016/j.freeradbiomed.2023.11.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/18/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023]
Abstract
Coronary ischemia-reperfusion (IR) injury results from a blockage of blood supply to the heart followed by restoration of perfusion, leading to oxidative stress induced pathological processes. Nuclear factor erythroid 2-related factor 2 (NRF2), a master antioxidant transcription factor, plays a key role in regulating redox signaling. Over the past decades, the field of metallomics has provided novel insights into the mechanism of pro-oxidant and antioxidant pathological processes. Both redox-active (e.g. Fe and Cu) and redox-inert (e.g. Zn and Mg) metals play unique roles in establishing redox balance under IR injury. Notably, Zn protects against oxidative stress in coronary IR injury by serving as a cofactor of antioxidant enzymes such as superoxide dismutase [Cu-Zn] (SOD1) and proteins such as metallothionein (MT) and KEAP1/NRF2 mediated antioxidant defenses. An increase in labile Zn2+ inhibits proteasomal degradation and ubiquitination of NRF2 by modifying KEAP1 and glycogen synthase kinase 3β (GSK3β) conformations. Fe and Cu catalyse the formation of reactive oxygen species via the Fenton reaction and also serve as cofactors of antioxidant enzymes and can activate NRF2 antioxidant signaling. We review the evidence that Zn and redox-active metals Fe and Cu affect redox signaling in coronary cells during IR and the mechanisms by which oxidative stress influences cellular metal content. In view of the unique double-edged characteristics of metals, we aim to bridge the role of metals and NRF2 regulated redox signaling to antioxidant defenses in IR injury, with a long-term aim of informing the design and application of novel therapeutics.
Collapse
Affiliation(s)
- Fan Yang
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom.
| | - Matthew J Smith
- MSD R&D Innovation Centre, 120 Moorgate, London EC2M 6UR, United Kingdom.
| |
Collapse
|
7
|
Bennett MC, Morton RA, Carlson AP, Shuttleworth CW. Synaptic Zn 2+ contributes to deleterious consequences of spreading depolarizations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.27.564408. [PMID: 37961648 PMCID: PMC10634912 DOI: 10.1101/2023.10.27.564408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Spreading depolarizations (SDs) are profound waves of neuroglial depolarization that can propagate repetitively through injured brain. Recent clinical work has established SD as an important contributor to expansion of acute brain injuries and have begun to extend SD studies into other neurological disorders. A critical challenge is to determine how to selectively prevent deleterious consequences of SD. In the present study, we determined whether a wave of profound Zn2+ release is a key contributor to deleterious consequences of SD, and whether this can be targeted pharmacologically. Focal KCl microinjection was used to initiate SD in the CA1 region of the hippocampus in murine brain slices. An extracellular Zn2+ chelator with rapid kinetics (ZX-1) increased SD propagation rates and improved recovery of extracellular DC potential shifts. Under conditions of metabolic compromise, tissues showed sustained impairment of functional and structural recovery following a single SD. ZX-1 effectively improved recovery of synaptic potentials and intrinsic optical signals in these vulnerable conditions. Fluorescence imaging and genetic deletion of a presynaptic Zn2+ transporter confirmed synaptic release as the primary contributor to extracellular accumulation and deleterious consequences of Zn2+ during SD. These results demonstrate a role for synaptic Zn2+ release in deleterious consequences of SD and show that targeted extracellular chelation could be useful for disorders where repetitive SD enlarges infarcts in injured tissues.
Collapse
Affiliation(s)
- Michael C Bennett
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Russell A Morton
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Andrew P Carlson
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - C William Shuttleworth
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
8
|
Qi Z, Zhou X, Dong W, Timmins GS, Pan R, Shi W, Yuan S, Zhao Y, Ji X, Liu KJ. Neuronal Zinc Transporter ZnT3 Modulates Cerebral Ischemia-Induced Blood-Brain Barrier Disruption. Aging Dis 2023; 15:2727-2741. [PMID: 37962463 PMCID: PMC11567248 DOI: 10.14336/ad.2023.1011] [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: 07/28/2023] [Accepted: 10/11/2023] [Indexed: 11/15/2023] Open
Abstract
Zinc plays important roles in both physiological and pathological processes in the brain. Accumulation of free zinc in ischemic tissue is recognized to contribute to blood-brain barrier (BBB) disruption following cerebral ischemia, but little is known either about the source of free zinc in microvessels or the mechanism by which free zinc mediates ischemia-induced BBB damage. We utilized cellular and animal models of ischemic stroke to determine the source of high levels of free zinc and the mechanism of free zinc-mediated BBB damage after ischemia. We report that cerebral ischemia elevated the level of extracellular fluid (ECF-Zn) of ischemic brain, leading to exacerbated BBB damage in a rat stroke model. Specifically suppressing zinc release from neurons, utilizing neuronal-specific zinc transporter 3 (ZnT3) knockout mice, markedly reduced ECF-Zn and BBB permeability after ischemia. Intriguingly, the activity of zinc-dependent metalloproteinase-2 (MMP-2) was modulated by ECF-Zn levels. Elevated ECF-Zn during ischemia directly bound to MMP-2 in extracellular fluid, increased its zinc content and augmented MMP-2 activity, leading to the degradation of tight junction protein in cerebral microvessels and BBB disruption. These findings suggest the role of neuronal ZnT3 in modulating ischemia-induced BBB disruption and reveal a novel mechanism of MMP-2 activation in BBB disruption after stroke, demonstrating ZnT3 as an effective target for stroke treatment.
Collapse
Affiliation(s)
- Zhifeng Qi
- Department of Neurology, Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Xixi Zhou
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Wen Dong
- Department of Neurology, Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Graham S. Timmins
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| | - Rong Pan
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Wenjuan Shi
- Department of Neurology, Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Shuhua Yuan
- Department of Neurology, Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Yongmei Zhao
- Department of Neurology, Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Xunming Ji
- Department of Neurology, Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Ke Jian Liu
- Department of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
9
|
Çınar R, Nazıroğlu M. TRPM2 Channel Inhibition Attenuates Amyloid β42-Induced Apoptosis and Oxidative Stress in the Hippocampus of Mice. Cell Mol Neurobiol 2023; 43:1335-1353. [PMID: 35840808 PMCID: PMC11414446 DOI: 10.1007/s10571-022-01253-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/01/2022] [Indexed: 01/16/2023]
Abstract
Alzheimer's disease (AD) is characterized by the increase of hippocampal Ca2+ influx-induced apoptosis and mitochondrial oxidative stress (OS). The OS is a stimulator of TRPM2, although N-(p-amylcinnamoyl)anthranilic acid (ACA), 2-aminoethyl diphenylborinate (2/APB), and glutathione (GSH) are non-specific antagonists of TRPM2. In the present study, we investigated the protective roles of GSH and TRPM2 antagonist treatments on the amyloid β42 peptide (Aβ)-caused oxidative neurotoxicity and apoptosis in the hippocampus of mice with AD model. After the isolation of hippocampal neurons from the newborn mice, they were divided into five incubation groups as follows: control, ACA, Aβ, Aβ+ACA, and Aβ+GSH. The levels of apoptosis, hippocampus death, cytosolic ROS, cytosolic Zn2+, mitochondrial ROS, caspase-3, caspase-9, lipid peroxidation, and cytosolic Ca2+ were increased in the primary hippocampus cultures by treatments of Aβ, although their levels were decreased in the neurons by the treatments of GSH, PARP-1 inhibitors (PJ34 and DPQ), and TRPM2 blockers (ACA and 2/APB). The Aβ-induced decreases of cell viability, cytosolic GSH, reduced GSH, and GSH peroxidase levels were also increased in the groups of Aβ+ACA and Aβ+GSH by the treatments of ACA and GSH. However, the Aβ-caused changes were not observed in the hippocampus of TRPM2-knockout mice. In conclusion, the present data demonstrate that maintaining the activation of TRPM2 is not only important for the quenching OS and neurotoxicity in the hippocampal neurons of mice with experimental AD but also equally critical to the modulation of Aβ-induced apoptosis. The possible positive effects of GSH and TRPM2 antagonist treatments on the amyloid-beta (Aβ)-induced oxidative toxicity in the hippocampus of mice. The ADP-ribose (ADPR) is produced via the stimulation of PARP-1 in the nucleus of neurons. The NUT9 in the C terminus of TRPM2 channel acts as a key role for the activation of TRPM2. The antagonists of TRPM2 are glutathione (GSH), ACA, and 2/APB in the hippocampus. The Aβ incubation-mediated TRPM2 stimulation increases the concentration of cytosolic-free Ca2+ and Zn2+ in the hippocampus. In turn, the increased concentration causes the increase of mitochondrial membrane potential (ΔΨm), which causes the excessive generations of mitochondria ROS and the decrease of cytosolic GSH and GSH peroxidase (GSH-Px). The ROS production and GSH depletion are two main causes in the neurobiology of Alzheimer's disease. However, the effect of Aβ was not shown in the hippocampus of TRPM2-knockout mice. The Aβ and TRPM2 stimulation-caused overload Ca2+ entry cause apoptosis and cell death via the activations of caspase-3 (Casp/3) and caspase-9 (Casp/9) in the hippocampus. The actions of Aβ-induced oxidative toxicity were modulated in the primary hippocampus by the incubations of ACA, GSH, 2/APB, and PARP-1 inhibitors (PJ34 and DPQ). (↑) Increase. (↓) Decrease.
Collapse
Affiliation(s)
- Ramazan Çınar
- Department of Neuroscience, Health Science Institute, Suleyman Demirel University, Isparta, Turkey
| | - Mustafa Nazıroğlu
- Department of Neuroscience, Health Science Institute, Suleyman Demirel University, Isparta, Turkey.
- Neuroscience Research Center, Suleyman Demirel University, Isparta, Turkey.
- Drug Discovery Unit, BSN Health, Analyses, Innovation, Consultancy, Organization, Agriculture and Industry Ltd., Isparta, Turkey.
- Department of Biophysics, School of Medicine, University of Suleyman Demirel, 32260, Isparta, Turkey.
| |
Collapse
|
10
|
Amani H, Soltani Khaboushan A, Terwindt GM, Tafakhori A. Glia Signaling and Brain Microenvironment in Migraine. Mol Neurobiol 2023; 60:3911-3934. [PMID: 36995514 DOI: 10.1007/s12035-023-03300-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/27/2023] [Indexed: 03/31/2023]
Abstract
Migraine is a complicated neurological disorder affecting 6% of men and 18% of women worldwide. Various mechanisms, including neuroinflammation, oxidative stress, altered mitochondrial function, neurotransmitter disturbances, cortical hyperexcitability, genetic factors, and endocrine system problems, are responsible for migraine. However, these mechanisms have not completely delineated the pathophysiology behind migraine, and they should be further studied. The brain microenvironment comprises neurons, glial cells, and vascular structures with complex interactions. Disruption of the brain microenvironment is the main culprit behind various neurological disorders. Neuron-glia crosstalk contributes to hyperalgesia in migraine. In the brain, microenvironment and related peripheral regulatory circuits, microglia, astrocytes, and satellite cells are necessary for proper function. These are the most important cells that could induce migraine headaches by disturbing the balance of the neurotransmitters in the nervous system. Neuroinflammation and oxidative stress are the prominent reactions glial cells drive during migraine. Understanding the role of cellular and molecular components of the brain microenvironment on the major neurotransmitters engaged in migraine pathophysiology facilitates the development of new therapeutic approaches with higher effectiveness for migraine headaches. Investigating the role of the brain microenvironment and neuroinflammation in migraine may help decipher its pathophysiology and provide an opportunity to develop novel therapeutic approaches for its management. This review aims to discuss the neuron-glia interactions in the brain microenvironment during migraine and their potential role as a therapeutic target for the treatment of migraine.
Collapse
Affiliation(s)
- Hanieh Amani
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Soltani Khaboushan
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Abbas Tafakhori
- Iranian Center of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Neurology, Imam Khomeini Hospital, Keshavarz Blvd., Tehran, Iran.
| |
Collapse
|
11
|
Gu Y, Tang J, Zhang F, Qu Y, Zhao M, Li M, Xie Z, Wang X, Song L, Jiang Z, Wang Y, Shen X, Xu L. Manganese potentiates lipopolysaccharide-induced innate immune responses and septic shock. Int J Biol Macromol 2023; 230:123202. [PMID: 36639076 DOI: 10.1016/j.ijbiomac.2023.123202] [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: 10/20/2022] [Revised: 12/10/2022] [Accepted: 01/05/2023] [Indexed: 01/12/2023]
Abstract
Divalent metal ions such as magnesium (Mg2+), manganese (Mn2+), and zinc (Zn2+) play important roles in regulating innate immune responses. Lipopolysaccharide stimulation led to increased intracellular Mn and Zn in macrophages. However, the effect of those metal ions in regulating lipopolysaccharide-induced innate immune responses remains unclear. Here, we uncovered that both Mn2+ and Zn2+ have immunostimulatory effects, which could potentiate the lipopolysaccharide-induced expression of interferon-stimulated genes (ISGs), cytokines and pro-inflammatory genes in a dose-dependent manner. Enhancement of lipopolysaccharide-induced innate immune gene expression by Mn2+ varies between 10 % and 900 %. Conversely, the chelating of Mn2+ almost totally diminished Mn2+-enhanced lipopolysaccharide-induced gene expression. In addition, Mn2+ exerted its ability to potentiate LPS-induced innate immune gene expression regardless of slight pH changes. Importantly, we found that Mn2+ potentiates lipopolysaccharide-induced immune responses independent of TLR4 but partially relies on cGAS-STING pathway. Further in vivo study showed that colloidal Mn2+ salt (Mn jelly [MnJ]) pretreatment exacerbated lipopolysaccharide-induced septic shock and mice death. In conclusion, we demonstrated that Mn2+ plays an essential role in boosting lipopolysaccharide-induced innate immune responses. These findings greatly expand the current understanding of the immunomodulatory potential of divalent metal Mn2+ and may provide a potential therapeutic target to prevent excessive immune responses.
Collapse
Affiliation(s)
- Yanchao Gu
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jingjing Tang
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Fuhua Zhang
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yichen Qu
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China; Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Min Zhao
- Comprehensive Technology Services Center of Chifeng Customs, Chifeng, Inner Mongolia 024000, China
| | - Mengyuan Li
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Zhen Xie
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xiao Wang
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Li Song
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Zhengfan Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Yao Wang
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Xihui Shen
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Lei Xu
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
12
|
Tang J, Liu Z, Han J, Xue J, Liu L, Lin J, Wu C, Zhang Q, Wu S, Liu C, Huang H, Fu Y, Li M, Zhuo Y, Li Y. Increased Mobile Zinc Regulates Retinal Ganglion Cell Survival via Activating Mitochondrial OMA1 and Integrated Stress Response. Antioxidants (Basel) 2022; 11:antiox11102001. [PMID: 36290724 PMCID: PMC9598227 DOI: 10.3390/antiox11102001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/01/2022] [Accepted: 10/05/2022] [Indexed: 11/26/2022] Open
Abstract
Retinal ganglion cells (RGCs), the projection neurons of the eye, are irreversibly lost once the optic nerve is injured, which is a critical mechanism of glaucoma. Mobile zinc (Zn2+) levels rapidly increase in retinal interneuron amacrine cells and Zn2+ is then transferred to RGCs via the Zn2+ transporter protein ZnT-3, triggering RGC loss in optic nerve injury. Zn2+ chelation and ZnT-3 deletion promote long-term RGC survival. However, the downstream signaling pathways of Zn2+ in RGCs remains unknown. Here, we show that increased levels of Zn2+ upregulate the expression and activity of mitochondrial zinc metallopeptidase OMA1 in the retina, leading to the cleavage of DELE1 and activation of cytosolic eIF2α kinase PKR, triggering the integrated stress response (ISR) in RGCs. Our study identified OMA1 and ISR as the downstream molecular mechanisms of retinal Zn2+ and potential targets for preventing the progression of Zn2+-associated neuronal damage.
Collapse
Affiliation(s)
- Jiahui Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Zhe Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jiaxu Han
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jingfei Xue
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Liyan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jicheng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Caiqing Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Qi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Siting Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Canying Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Haishun Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yuanyuan Fu
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Min Li
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
- Correspondence: (Y.Z.); (Y.L.)
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
- Correspondence: (Y.Z.); (Y.L.)
| |
Collapse
|
13
|
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: 17] [Impact Index Per Article: 5.7] [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
|
14
|
Zinc in Cognitive Impairment and Aging. Biomolecules 2022; 12:biom12071000. [PMID: 35883555 PMCID: PMC9312494 DOI: 10.3390/biom12071000] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [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
|
15
|
Medvedeva YV, Yin HZ, Bazrafkan A, Yeromin A, Ji SG, Weiss-Hung EJ, Sharman E, Avilez AP, Maki N, Rafi MA, Tian G, Akbari Y, Weiss JH. Blocking Mitochondrial Zn 2+ Accumulation after Ischemia Reduces Mitochondrial Dysfunction and Neuronal Injury. J Neurosci 2022; 42:5281-5292. [PMID: 35623885 PMCID: PMC9236293 DOI: 10.1523/jneurosci.0874-21.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 04/22/2022] [Accepted: 05/18/2022] [Indexed: 12/31/2022] Open
Abstract
Zn2+ is an important contributor to ischemic brain injury, and recent studies support the hypothesis that mitochondria are key sites of its injurious effects. In murine hippocampal slices (both sexes) subjected to oxygen glucose deprivation (OGD), we found that Zn2+ accumulation and its entry into mitochondria precedes and contributes to the induction of acute neuronal death. In addition, if the ischemic episode is short (and sublethal), there is ongoing Zn2+ accumulation in CA1 mitochondria after OGD that may contribute to their delayed dysfunction. Using this slice model of sublethal OGD, we have examined Zn2+ contributions to the progression of changes evoked by OGD and occurring over 4-5 h. We detected progressive mitochondrial depolarization occurring from ∼2 h after ischemia, a large increase in spontaneous synaptic activity between 2 and 3 h, and mitochondrial swelling and fragmentation at 4 h. Blockade of the primary route for Zn2+ entry, the mitochondrial Ca2+ uniporter (with ruthenium red [RR]) or Zn2+ chelation shortly after OGD withdrawal substantially attenuated the mitochondrial depolarization and the changes in synaptic activity. RR also largely reversed the mitochondrial swelling. Finally, using an in vivo rat (male) asphyxial cardiac arrest model of transient global ischemia, we found that ∼8 min asphyxia induces considerable injury of CA1 neurons 4 h later that is associated with strong Zn2+ accumulation within many damaged mitochondria. These effects were substantially attenuated by infusion of RR on reperfusion. Our findings highlight mitochondrial Zn2+ accumulation after ischemia as a possible target for neuroprotective therapy.SIGNIFICANCE STATEMENT Brain ischemia is a leading cause of mortality and long-term disability that still lacks effective treatment. After transient ischemia, delayed death of neurons occurs in vulnerable brain regions. There is a critical need to understand mechanisms of this delayed neurodegeneration which can be targeted for neuroprotection. We found progressive and long-lasting mitochondrial Zn2+ accumulation to occur in highly vulnerable CA1 neurons after ischemia. Here we demonstrate that this Zn2+ accumulation contributes strongly to deleterious events occurring after ischemia, including mitochondrial dysfunction, swelling, and structural changes. We suggest that this mitochondrial Zn2+ entry may constitute a promising target for development of therapeutic interventions to be delivered after termination of an episode of transient global ischemia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Yama Akbari
- Departments of Neurology
- Anatomy & Neurobiology
- Neurological surgery
- Beckman Laser Institute & Medical Clinic, University of California Irvine, Irvine, California 92697
| | | |
Collapse
|
16
|
Wu S, Zhang K, Liang Y, Wei Y, An J, Wang Y, Yang J, Zhang H, Zhang Z, Liu J, Shi J. Nano-enabled Tumor Systematic Energy Exhaustion via Zinc (II) Interference Mediated Glycolysis Inhibition and Specific GLUT1 Depletion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103534. [PMID: 34913610 PMCID: PMC8895132 DOI: 10.1002/advs.202103534] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/15/2021] [Indexed: 05/19/2023]
Abstract
Despite the promise of tumor starvation therapies, they are often associated with nonspecific and incomplete energy blockade. Here, a novel paradigm of starvation therapy is proposed to synergize the "Zn2+ interference"-mediated glycolysis inhibition and Zn2+ -activating GLUT1 (Glucose transporter 1) tumor specific depletion for systematic energy exhaustion. It is discovered that ZIF-8 (zinc imidazolate metal-organic frameworks ) can induce abrupt intracellular Zn2+ elevation preferentially in melanoma cells, and then achieve effective glycolysis blockade through "Zn2+ interference"-triggered decrease of NAD+ and inactivation of GAPDH, making it a powerful tumor energy nanoinhibitor. Meanwhile, Zn2+ -activating DNAzymes for specifically cleaving GLUT1 mRNA is designed. This DNAzyme can only be activated under intracellular Zn2+ overloading, and then directionally cut off glucose supply, which further restrains the adaptive up-regulation of glycolytic flux after glycolysis inhibition in tumors. Afterward, DNAzymes are loaded in ZIF-8 concurrently tethered by hyaluronic acid (HA), constructing a "nanoenabled energy interrupter ". Such a rational design presents a preferential accumulation tendency to tumor sites due to the active CD44-targeting mechanisms, specifically achieves remarkable systematic energy exhaustion in melanoma cells, and affords 80.8% in tumor growth suppression without systemic toxicity in vivo. This work verifies a fascinating therapeutic platform enabling ion interference-inductive starvation strategy for effective tumor therapy.
Collapse
Affiliation(s)
- Sixuan Wu
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou450001P. R. China
| | - Kaixiang Zhang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou450001P. R. China
| | - Yan Liang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou450001P. R. China
| | - Yongbin Wei
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou450001P. R. China
| | - Jingyi An
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou450001P. R. China
| | - Yifei Wang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou450001P. R. China
| | - Jiali Yang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou450001P. R. China
| | - Hongling Zhang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou450001P. R. China
| | - Zhenzhong Zhang
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou450001P. R. China
- Key Laboratory of Advanced Drug Preparation TechnologiesMinistry of EducationZhengzhou450001P. R. China
- State Key Laboratory of Esophageal Cancer Prevention & TreatmentZhengzhou450001P. R. China
| | - Junjie Liu
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou450001P. R. China
| | - Jinjin Shi
- School of Pharmaceutical SciencesZhengzhou UniversityZhengzhou450001P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesZhengzhou450001P. R. China
| |
Collapse
|
17
|
Wang Q, Liu N, Ni YS, Yang JM, Ma L, Lan XB, Wu J, Niu JG, Yu JQ. TRPM2 in ischemic stroke: Structure, molecular mechanisms, and drug intervention. Channels (Austin) 2021; 15:136-154. [PMID: 33455532 PMCID: PMC7833771 DOI: 10.1080/19336950.2020.1870088] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 01/14/2023] Open
Abstract
Ischemic stroke has a high lethality rate worldwide, and novel treatments are limited. Calcium overload is considered to be one of the mechanisms of cerebral ischemia. Transient receptor potential melastatin 2 (TRPM2) is a reactive oxygen species (ROS)-sensitive calcium channel. Cerebral ischemia-induced TRPM2 activation triggers abnormal intracellular Ca2+ accumulation and cell death, which in turn causes irreversible brain damage. Thus, TRPM2 has emerged as a new therapeutic target for ischemic stroke. This review provides data on the expression, structure, and function of TRPM2 and illustrates its cellular and molecular mechanisms in ischemic stroke. Natural and synthetic TRPM2 inhibitors (both specific and nonspecific) are also summarized. The three-dimensional protein structure of TRPM2 has been identified, and we speculate that molecular simulation techniques will be essential for developing new drugs that block TRPM2 channels. These insights about TRPM2 may be the key to find potent therapeutic approaches for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Qing Wang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Ning Liu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Yuan-Shu Ni
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Jia-Mei Yang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Lin Ma
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Xiao-Bing Lan
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Jing Wu
- Laboratory Animal Center, Ningxia Medical University, Yinchuan, China
| | - Jian-Guo Niu
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| | - Jian-Qiang Yu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
- Ningxia Collaborative Innovation Center of Regional Characteristic Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
18
|
ZnT1 is a neuronal Zn 2+/Ca 2+ exchanger. Cell Calcium 2021; 101:102505. [PMID: 34871934 DOI: 10.1016/j.ceca.2021.102505] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/31/2021] [Accepted: 11/22/2021] [Indexed: 01/22/2023]
Abstract
Zinc transporter 1 (ZnT1; SLC30A1) is present in the neuronal plasma membrane, critically modulating NMDA receptor function and Zn2+ neurotoxicity. The mechanism mediating Zn2+ transport by ZnT1, however, has remained elusive. Here, we investigated ZnT1-dependent Zn2+ transport by measuring intracellular changes of this ion using the fluorescent indicator FluoZin-3. In primary mouse cortical neurons, which express ZnT1, transient addition of extracellular Zn2+ triggered a rise in cytosolic Zn2+, followed by its removal. Knockdown of ZnT1 by adeno associated viral (AAV)-short hairpin RNA (shZnT1) markedly increased rates of Zn2+ rise, and decreased rates of its removal, suggesting that ZnT1 is a primary route for Zn2+ efflux in neurons. Although Zn2+ transport by other members of the SLC30A family is dependent on pH gradients across cellular membranes, altered H+ gradients were not coupled to ZnT1-dependent transport. Removal of cytoplasmic Zn2+, against a large inward gradient during the initial loading phase, suggests that Zn2+ efflux requires a large driving force. We therefore asked if Ca2+ gradients across the membrane can facilitate Zn2+ efflux. Elimination of extracellular Ca2+ abolished Zn2+ efflux, while increased extracellular Ca2+ levels enhanced Zn2+ efflux. Intracellular Ca2+ rises, measured in GCaMP6 expressing neurons, closely paralleled cytoplasmic Zn2+ removal. Taken together, these results strongly suggest that ZnT1 functions as a Zn2+/Ca2+ exchanger, thereby regulating the transport of two ions of fundamental importance in neuronal signaling.
Collapse
|
19
|
Wang X, An P, Gu Z, Luo Y, Luo J. Mitochondrial Metal Ion Transport in Cell Metabolism and Disease. Int J Mol Sci 2021; 22:7525. [PMID: 34299144 PMCID: PMC8305404 DOI: 10.3390/ijms22147525] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/06/2021] [Accepted: 07/13/2021] [Indexed: 12/19/2022] Open
Abstract
Mitochondria are vital to life and provide biological energy for other organelles and cell physiological processes. On the mitochondrial double layer membrane, there are a variety of channels and transporters to transport different metal ions, such as Ca2+, K+, Na+, Mg2+, Zn2+ and Fe2+/Fe3+. Emerging evidence in recent years has shown that the metal ion transport is essential for mitochondrial function and cellular metabolism, including oxidative phosphorylation (OXPHOS), ATP production, mitochondrial integrity, mitochondrial volume, enzyme activity, signal transduction, proliferation and apoptosis. The homeostasis of mitochondrial metal ions plays an important role in maintaining mitochondria and cell functions and regulating multiple diseases. In particular, channels and transporters for transporting mitochondrial metal ions are very critical, which can be used as potential targets to treat neurodegeneration, cardiovascular diseases, cancer, diabetes and other metabolic diseases. This review summarizes the current research on several types of mitochondrial metal ion channels/transporters and their functions in cell metabolism and diseases, providing strong evidence and therapeutic strategies for further insights into related diseases.
Collapse
Affiliation(s)
- Xuan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (X.W.); (P.A.)
| | - Peng An
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (X.W.); (P.A.)
| | - Zhenglong Gu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA;
| | - Yongting Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (X.W.); (P.A.)
| | - Junjie Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (X.W.); (P.A.)
| |
Collapse
|
20
|
The Multifaceted Roles of Zinc in Neuronal Mitochondrial Dysfunction. Biomedicines 2021; 9:biomedicines9050489. [PMID: 33946782 PMCID: PMC8145363 DOI: 10.3390/biomedicines9050489] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 12/17/2022] Open
Abstract
Zinc is a highly abundant cation in the brain, essential for cellular functions, including transcription, enzymatic activity, and cell signaling. However, zinc can also trigger injurious cascades in neurons, contributing to the pathology of neurodegenerative diseases. Mitochondria, critical for meeting the high energy demands of the central nervous system (CNS), are a principal target of the deleterious actions of zinc. An increasing body of work suggests that intracellular zinc can, under certain circumstances, contribute to neuronal damage by inhibiting mitochondrial energy processes, including dissipation of the mitochondrial membrane potential (MMP), leading to ATP depletion. Additional consequences of zinc-mediated mitochondrial damage include reactive oxygen species (ROS) generation, mitochondrial permeability transition, and excitotoxic calcium deregulation. Zinc can also induce mitochondrial fission, resulting in mitochondrial fragmentation, as well as inhibition of mitochondrial motility. Here, we review the known mechanisms responsible for the deleterious actions of zinc on the organelle, within the context of neuronal injury associated with neurodegenerative processes. Elucidating the critical contributions of zinc-induced mitochondrial defects to neurotoxicity and neurodegeneration may provide insight into novel therapeutic targets in the clinical setting.
Collapse
|
21
|
Ludhiadch A, Sharma R, Muriki A, Munshi A. Role of Calcium Homeostasis in Ischemic Stroke: A Review. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:52-61. [DOI: 10.2174/1871527320666210212141232] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/10/2020] [Accepted: 12/01/2020] [Indexed: 11/22/2022]
Abstract
:
Stroke is the second most common cause of death worldwide. It occurs due to the insufficient supply of oxygen-rich blood to the brain. It is a complex disease with multiple associated risk factors including smoking, alcoholism, age, sex, ethnicity, etc. Calcium ions are known to play a vital role in cell death pathways, which is a ubiquitous intracellular messenger during and immediately after an ischemic period. Disruption in normal calcium hemostasis is known to be a major initiator and activator of the ischemic cell death pathway. Under Ischemic stroke conditions, glutamate is released from the neurons and glia which further activates the N-methyl-D-aspartate (NMDA) receptor and triggers the rapid translocation of Ca2+ from extracellular to intracellular spaces in cerebral tissues and vice versa. Various studies indicated that Ca2+ could have harmful effects on neurons under acute ischemic conditions. Mitochondrial dysfunction also contributes to delayed neuronal death, and it was established decades ago that massive calcium accumulation triggers mitochondrial damage. Elevated Ca2+ levels cause mitochondria to swell and release their contents. As a result oxidative stress and mitochondrial calcium accumulation activate mitochondrial permeability transition and lead to depolarization-coupled production of reactive oxygen species. This association between calcium levels and mitochondrial death suggests that elevated calcium levels might have a role in the neurological outcome in ischemic stroke. Previous studies have also reported that elevated Ca2+ levels play a role in the determination of infarct size, outcome, and recurrence of ischemic stroke. The current review has been compiled to understand the multidimensional role of altered Ca2+ levels in the initiation and alteration of neuronal death after ischemic attack. The underlying mechanisms understood to date have also been discussed.
Collapse
Affiliation(s)
- Abhilash Ludhiadch
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda-151001, India
| | - Rashmi Sharma
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda-151001, India
| | - Aishwarya Muriki
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda-151001, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda-151001, India
| |
Collapse
|
22
|
Granzotto A, Canzoniero LMT, Sensi SL. A Neurotoxic Ménage-à-trois: Glutamate, Calcium, and Zinc in the Excitotoxic Cascade. Front Mol Neurosci 2020; 13:600089. [PMID: 33324162 PMCID: PMC7725690 DOI: 10.3389/fnmol.2020.600089] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
Fifty years ago, the seminal work by John Olney provided the first evidence of the neurotoxic properties of the excitatory neurotransmitter glutamate. A process hereafter termed excitotoxicity. Since then, glutamate-driven neuronal death has been linked to several acute and chronic neurological conditions, like stroke, traumatic brain injury, Alzheimer’s, Parkinson’s, and Huntington’s diseases, and Amyotrophic Lateral Sclerosis. Mechanisms linked to the overactivation of glutamatergic receptors involve an aberrant cation influx, which produces the failure of the ionic neuronal milieu. In this context, zinc, the second most abundant metal ion in the brain, is a key but still somehow underappreciated player of the excitotoxic cascade. Zinc is an essential element for neuronal functioning, but when dysregulated acts as a potent neurotoxin. In this review, we discuss the ionic changes and downstream effects involved in the glutamate-driven neuronal loss, with a focus on the role exerted by zinc. Finally, we summarize our work on the fascinating distinct properties of NADPH-diaphorase neurons. This neuronal subpopulation is spared from excitotoxic insults and represents a powerful tool to understand mechanisms of resilience against excitotoxic processes.
Collapse
Affiliation(s)
- Alberto Granzotto
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States.,Center for Advanced Sciences and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy.,Department of Neuroscience, Imaging, and Clinical Sciences (DNISC), Laboratory of Molecular Neurology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | | | - Stefano L Sensi
- Center for Advanced Sciences and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy.,Department of Neuroscience, Imaging, and Clinical Sciences (DNISC), Laboratory of Molecular Neurology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy.,Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
23
|
The role of labile Zn 2+ and Zn 2+-transporters in the pathophysiology of mitochondria dysfunction in cardiomyocytes. Mol Cell Biochem 2020; 476:971-989. [PMID: 33225416 DOI: 10.1007/s11010-020-03964-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023]
Abstract
An important energy supplier of cardiomyocytes is mitochondria, similar to other mammalian cells. Studies have demonstrated that any defect in the normal processes controlled by mitochondria can lead to abnormal ROS production, thereby high oxidative stress as well as lack of ATP. Taken into consideration, the relationship between mitochondrial dysfunction and overproduction of ROS as well as the relation between increased ROS and high-level release of intracellular labile Zn2+, those bring into consideration the importance of the events related with those stimuli in cardiomyocytes responsible from cellular Zn2+-homeostasis and responsible Zn2+-transporters associated with the Zn2+-homeostasis and Zn2+-signaling. Zn2+-signaling, controlled by cellular Zn2+-homeostatic mechanisms, is regulated with intracellular labile Zn2+ levels, which are controlled, especially, with the two Zn2+-transporter families; ZIPs and ZnTs. Our experimental studies in mammalian cardiomyocytes and human heart tissue showed that Zn2+-transporters localizes to mitochondria besides sarco(endo)plasmic reticulum and Golgi under physiological condition. The protein levels as well as functions of those transporters can re-distribute under pathological conditions, therefore, they can interplay among organelles in cardiomyocytes to adjust a proper intracellular labile Zn2+ level. In the present review, we aimed to summarize the already known Zn2+-transporters localize to mitochondria and function to stabilize not only the cellular Zn2+ level but also cellular oxidative stress status. In conclusion, one can propose that a detailed understanding of cellular Zn2+-homeostasis and Zn2+-signaling through mitochondria may emphasize the importance of new mitochondria-targeting agents for prevention and/or therapy of cardiovascular dysfunction in humans.
Collapse
|
24
|
Choi DW. Excitotoxicity: Still Hammering the Ischemic Brain in 2020. Front Neurosci 2020; 14:579953. [PMID: 33192266 PMCID: PMC7649323 DOI: 10.3389/fnins.2020.579953] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Interest in excitotoxicity expanded following its implication in the pathogenesis of ischemic brain injury in the 1980s, but waned subsequent to the failure of N-methyl-D-aspartate (NMDA) antagonists in high profile clinical stroke trials. Nonetheless there has been steady progress in elucidating underlying mechanisms. This review will outline the historical path to current understandings of excitotoxicity in the ischemic brain, and suggest that this knowledge should be leveraged now to develop neuroprotective treatments for stroke.
Collapse
Affiliation(s)
- Dennis W Choi
- Department of Neurology, SUNY Stony Brook, Stony Brook, NY, United States
| |
Collapse
|
25
|
Yin HZ, Wang HL, Ji SG, Medvedeva YV, Tian G, Bazrafkan AK, Maki NZ, Akbari Y, Weiss JH. Rapid Intramitochondrial Zn2+ Accumulation in CA1 Hippocampal Pyramidal Neurons After Transient Global Ischemia: A Possible Contributor to Mitochondrial Disruption and Cell Death. J Neuropathol Exp Neurol 2020; 78:655-664. [PMID: 31150090 DOI: 10.1093/jnen/nlz042] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial Zn2+ accumulation, particularly in CA1 neurons, occurs after ischemia and likely contributes to mitochondrial dysfunction and subsequent neurodegeneration. However, the relationship between mitochondrial Zn2+ accumulation and their disruption has not been examined at the ultrastructural level in vivo. We employed a cardiac arrest model of transient global ischemia (TGI), combined with Timm's sulfide silver labeling, which inserts electron dense metallic silver granules at sites of labile Zn2+ accumulation, and used transmission electron microscopy (TEM) to examine subcellular loci of the Zn2+ accumulation. In line with prior studies, TGI-induced damage to CA1 was far greater than to CA3 pyramidal neurons, and was substantially progressive in the hours after reperfusion (being significantly greater after 4- than 1-hour recovery). Intriguingly, TEM examination of Timm's-stained sections revealed substantial Zn2+ accumulation in many postischemic CA1 mitochondria, which was strongly correlated with their swelling and disruption. Furthermore, paralleling the evolution of neuronal injury, both the number of mitochondria containing Zn2+ and the degree of their disruption were far greater at 4- than 1-hour recovery. These data provide the first direct characterization of Zn2+ accumulation in CA1 mitochondria after in vivo TGI, and support the idea that targeting these events could yield therapeutic benefits.
Collapse
Affiliation(s)
| | | | - Sung G Ji
- Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, California
| | | | | | | | | | | | - John H Weiss
- Department of Neurology
- Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, California
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| |
Collapse
|
26
|
MCL-1 Matrix maintains neuronal survival by enhancing mitochondrial integrity and bioenergetic capacity under stress conditions. Cell Death Dis 2020; 11:321. [PMID: 32371858 PMCID: PMC7200794 DOI: 10.1038/s41419-020-2498-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022]
Abstract
Mitochondria play a crucial role in neuronal survival through efficient energy metabolism. In pathological conditions, mitochondrial stress leads to neuronal death, which is regulated by the anti-apoptotic BCL-2 family of proteins. MCL-1 is an anti-apoptotic BCL-2 protein localized to mitochondria either in the outer membrane (OM) or inner membrane (Matrix), which have distinct roles in inhibiting apoptosis and promoting bioenergetics, respectively. While the anti-apoptotic role for Mcl1 is well characterized, the protective function of MCL-1 Matrix remains poorly understood. Here, we show MCL-1OM and MCL-1Matrix prevent neuronal death through distinct mechanisms. We report that MCL-1Matrix functions to preserve mitochondrial energy transduction and improves respiratory chain capacity by modulating mitochondrial oxygen consumption in response to mitochondrial stress. We show that MCL-1Matrix protects neurons from stress by enhancing respiratory function, and by inhibiting mitochondrial permeability transition pore opening. Taken together, our results provide novel insight into how MCL-1Matrix may confer neuroprotection under stress conditions involving loss of mitochondrial function.
Collapse
|
27
|
Hosseini M, Wilson RH, Crouzet C, Amirhekmat A, Wei KS, Akbari Y. Resuscitating the Globally Ischemic Brain: TTM and Beyond. Neurotherapeutics 2020; 17:539-562. [PMID: 32367476 PMCID: PMC7283450 DOI: 10.1007/s13311-020-00856-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiac arrest (CA) afflicts ~ 550,000 people each year in the USA. A small fraction of CA sufferers survive with a majority of these survivors emerging in a comatose state. Many CA survivors suffer devastating global brain injury with some remaining indefinitely in a comatose state. The pathogenesis of global brain injury secondary to CA is complex. Mechanisms of CA-induced brain injury include ischemia, hypoxia, cytotoxicity, inflammation, and ultimately, irreversible neuronal damage. Due to this complexity, it is critical for clinicians to have access as early as possible to quantitative metrics for diagnosing injury severity, accurately predicting outcome, and informing patient care. Current recommendations involve using multiple modalities including clinical exam, electrophysiology, brain imaging, and molecular biomarkers. This multi-faceted approach is designed to improve prognostication to avoid "self-fulfilling" prophecy and early withdrawal of life-sustaining treatments. Incorporation of emerging dynamic monitoring tools such as diffuse optical technologies may provide improved diagnosis and early prognostication to better inform treatment. Currently, targeted temperature management (TTM) is the leading treatment, with the number of patients needed to treat being ~ 6 in order to improve outcome for one patient. Future avenues of treatment, which may potentially be combined with TTM, include pharmacotherapy, perfusion/oxygenation targets, and pre/postconditioning. In this review, we provide a bench to bedside approach to delineate the pathophysiology, prognostication methods, current targeted therapies, and future directions of research surrounding hypoxic-ischemic brain injury (HIBI) secondary to CA.
Collapse
Affiliation(s)
- Melika Hosseini
- Department of Neurology, School of Medicine, University of California, Irvine, USA
| | - Robert H Wilson
- Department of Neurology, School of Medicine, University of California, Irvine, USA
- Beckman Laser Institute, University of California, Irvine, USA
| | - Christian Crouzet
- Department of Neurology, School of Medicine, University of California, Irvine, USA
- Beckman Laser Institute, University of California, Irvine, USA
| | - Arya Amirhekmat
- Department of Neurology, School of Medicine, University of California, Irvine, USA
| | - Kevin S Wei
- Department of Neurology, School of Medicine, University of California, Irvine, USA
| | - Yama Akbari
- Department of Neurology, School of Medicine, University of California, Irvine, USA.
- Beckman Laser Institute, University of California, Irvine, USA.
| |
Collapse
|
28
|
Ji SG, Medvedeva YV, Weiss JH. Zn 2+ entry through the mitochondrial calcium uniporter is a critical contributor to mitochondrial dysfunction and neurodegeneration. Exp Neurol 2019; 325:113161. [PMID: 31881218 DOI: 10.1016/j.expneurol.2019.113161] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/19/2019] [Accepted: 12/21/2019] [Indexed: 12/19/2022]
Abstract
Excitotoxic Ca2+ accumulation contributes to ischemic neurodegeneration, and Ca2+ can enter the mitochondria through the mitochondrial calcium uniporter (MCU) to promote mitochondrial dysfunction. Yet, Ca2+-targeted therapies have met limited success. A growing body of evidence has highlighted the underappreciated importance of Zn2+, which also accumulates in neurons after ischemia and can induce mitochondrial dysfunction and cell death. While studies have indicated that Zn2+ can also enter the mitochondria through the MCU, the specificity of the pore's role in Zn2+-triggered injury is still debated. Present studies use recently available MCU knockout mice to examine how the deletion of this channel impacts deleterious effects of cytosolic Zn2+ loading. In cultured cortical neurons from MCU knockout mice, we find significantly reduced mitochondrial Zn2+ accumulation. Correspondingly, these neurons were protected from both acute and delayed Zn2+-triggered mitochondrial dysfunction, including mitochondrial reactive oxygen species generation, depolarization, swelling and inhibition of respiration. Furthermore, when toxic extramitochondrial effects of Ca2+ entry were moderated, both cultured neurons (exposed to Zn2+) and CA1 neurons of hippocampal slices (subjected to prolonged oxygen glucose deprivation to model ischemia) from MCU knockout mice displayed decreased neurodegeneration. Finally, to examine the therapeutic applicability of these findings, we added an MCU blocker after toxic Zn2+ exposure in wildtype neurons (to induce post-insult MCU blockade). This significantly attenuated the delayed evolution of both mitochondrial dysfunction and neurotoxicity. These data-combining both genetic and pharmacologic tools-support the hypothesis that Zn2+ entry through the MCU is a critical contributor to ischemic neurodegeneration that could be targeted for neuroprotection.
Collapse
Affiliation(s)
- Sung G Ji
- Department of Anatomy & Neurobiology, University of California, Irvine, United States of America
| | - Yuliya V Medvedeva
- Department of Neurology, University of California, Irvine, United States of America
| | - John H Weiss
- Department of Anatomy & Neurobiology, University of California, Irvine, United States of America; Department of Neurology, University of California, Irvine, United States of America.
| |
Collapse
|
29
|
Transmembrane 163 (TMEM163) protein effluxes zinc. Arch Biochem Biophys 2019; 677:108166. [PMID: 31697912 PMCID: PMC6864316 DOI: 10.1016/j.abb.2019.108166] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/09/2019] [Accepted: 10/31/2019] [Indexed: 01/19/2023]
Abstract
Recent investigations of rodent Tmem163 suggest that it binds to and transports zinc as a dimer, and that alanine mutagenesis of its two species-conserved aspartate (D123A/D127A) residues proposed to bind zinc, perturbs protein function. Direct corroboration, however, is lacking whether it is an influx or efflux transporter in cells. We hypothesized that human TMEM163 is a zinc effluxer based on its predicted protein characteristics. We used cultured human cell lines that either stably or transiently expressed TMEM163, and pre-loaded the cells with zinc to determine transport activity. We found that TMEM163-expressing cells exhibited significant reduction of intracellular zinc levels as evidenced by two zinc-specific fluorescent dyes and radionuclide zinc-65. The specificity of the fluorescence signal was confirmed upon treatment with TPEN, a high-affinity zinc chelator. Multiple sequence alignment and phylogenetic analyses showed that TMEM163 is related to distinct members of the cation diffusion facilitator (CDF) protein family. To further characterize the efflux function of TMEM163, we substituted alanine in two homologous aspartate residues (D124A/D128A) and performed site-directed mutagenesis of several conserved amino acid residues identified as non-synonymous single nucleotide polymorphism (S61R, S95C, S193P, and E286K). We found a significant reduction of zinc efflux upon cellular expression of D124A/D128A or E286K protein variant when compared with wild-type, suggesting that these particular amino acids are important for normal protein function. Taken together, our findings demonstrate that TMEM163 effluxes zinc, and it should now be designated ZNT11 as a new member of the mammalian CDF family of zinc efflux transporters.
Collapse
|
30
|
Zhao Y, Yan F, Yin J, Pan R, Shi W, Qi Z, Fang Y, Huang Y, Li S, Luo Y, Ji X, Liu KJ. Synergistic Interaction Between Zinc and Reactive Oxygen Species Amplifies Ischemic Brain Injury in Rats. Stroke 2019; 49:2200-2210. [PMID: 30354980 DOI: 10.1161/strokeaha.118.021179] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Although intracellular zinc accumulation has been shown to contribute to neuronal death after cerebral ischemia, the mechanism by which zinc keeps on accumulating to cause severe brain damage remains unclear. Herein the dynamic cause-effect relationships between zinc accumulation and reactive oxygen species (ROS) production during cerebral ischemia/reperfusion are investigated. Methods- Rats were treated with zinc chelator, ROS scavenger, mitochondria-targeted ROS inhibitor, or NADPH oxidase inhibitor during a 90-minute middle cerebral artery occlusion. Cytosolic labile zinc, ROS level, cerebral infarct volume, and neurological functions were assessed after ischemia/reperfusion. Results- Zinc and ROS were colocalized in neurons, leading to neuronal apoptotic death. Chelating zinc reduced ROS production at 6 and 24 hours after reperfusion, whereas eliminating ROS reduced zinc accumulation only at 24 hours. Furthermore, suppression of mitochondrial ROS production reduced the total ROS level and brain damage at 6 hours after reperfusion but did not change zinc accumulation, indicating that ROS is produced mainly from mitochondria during early reperfusion and the initial zinc release is upstream of ROS generation after ischemia. Suppression of NADPH oxidase decreased ROS generation, zinc accumulation, and brain damage only at 24 hours after reperfusion, indicating that the majority of ROS is produced by NADPH oxidase at later reperfusion time. Conclusions- This study provides the direct evidence that there exists a positive feedback loop between zinc accumulation and NADPH oxidase-induced ROS production, which greatly amplifies the damaging effects of both. These findings reveal that different ROS-generating source contributes to ischemia-generated ROS at different time, underscoring the critical importance of spatial and temporal factors in the interaction between ROS and zinc accumulation, and the consequent brain injury, after cerebral ischemia/reperfusion.
Collapse
Affiliation(s)
- Yongmei Zhao
- From the Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China (Y.Z., F.Y., J.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.).,Beijing Geriatric Medical Research Center, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, China (Y.Z., F.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.)
| | - Feng Yan
- From the Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China (Y.Z., F.Y., J.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.).,Beijing Geriatric Medical Research Center, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, China (Y.Z., F.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.)
| | - Jie Yin
- From the Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China (Y.Z., F.Y., J.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.)
| | - Rong Pan
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque (R.P., K.J.L.)
| | - Wenjuan Shi
- From the Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China (Y.Z., F.Y., J.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.).,Beijing Geriatric Medical Research Center, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, China (Y.Z., F.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.)
| | - Zhifeng Qi
- From the Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China (Y.Z., F.Y., J.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.).,Beijing Geriatric Medical Research Center, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, China (Y.Z., F.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.)
| | - Yalan Fang
- From the Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China (Y.Z., F.Y., J.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.).,Beijing Geriatric Medical Research Center, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, China (Y.Z., F.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.)
| | - Yuyou Huang
- From the Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China (Y.Z., F.Y., J.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.).,Beijing Geriatric Medical Research Center, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, China (Y.Z., F.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.)
| | - Sen Li
- From the Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China (Y.Z., F.Y., J.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.).,Beijing Geriatric Medical Research Center, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, China (Y.Z., F.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.)
| | - Yumin Luo
- From the Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China (Y.Z., F.Y., J.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.).,Beijing Geriatric Medical Research Center, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, China (Y.Z., F.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.)
| | - Xunming Ji
- From the Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China (Y.Z., F.Y., J.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.).,Beijing Geriatric Medical Research Center, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, China (Y.Z., F.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.)
| | - Ke Jian Liu
- From the Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China (Y.Z., F.Y., J.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.).,Beijing Geriatric Medical Research Center, Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, China (Y.Z., F.Y., W.S., Z.Q., Y.F., Y.H., S.L., Y.L., X.J., K.J.L.).,Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque (R.P., K.J.L.)
| |
Collapse
|
31
|
Wang C, Wei Y, Yuan Y, Yu Y, Xie K, Dong B, Shi Y, Wang G. The role of PI3K-mediated AMPA receptor changes in post-conditioning of propofol in brain protection. BMC Neurosci 2019; 20:51. [PMID: 31570094 PMCID: PMC6771103 DOI: 10.1186/s12868-019-0532-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 09/13/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND We aimed to study the role of amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPAR) glutamate receptor 2 (GluR2) subunit trafficking, and activity changes in short-term neuroprotection provided by propofol post-conditioning. We also aimed to determine the role of phosphoinositide-3-kinase (PI3K) in the regulation of these processes. METHODS Rats underwent 1 h of focal cerebral ischemia followed by 23 h of reperfusion were randomly divided into 6 groups (n = 36 per group): sham- operation (S), ischemia-reperfusion (IR), propofol (P group, propofol 20 mg/kg/h at the onset of reperfusion for 2 h after 60 min of occlusion), and LY294002 (PI3K non-selective antagonist) + sham (L + S, LY294002 of 1.5 mg/kg was infused 30 min before sham operation), LY294002+ ischemia-reperfusion (L + IR, LY294002 of 1.5 mg/kg was infused 30 min before middle cerebral artery occlusion), LY294002 + IR + propofol (L + P, LY294002 of 1.5 mg/kg was infused 30 min before middle cerebral artery occlusion and propofol 20 mg/kg/h at the onset of reperfusion for 2 h after 60 min of occlusion). RESULTS Compared with group IR, rats in group P had significant lower neurologic defect scores and infarct volume. Additionally, consistent with enhanced expression of PI3K-AMPAR GluR2 subunit complex substances in ipsilateral hippocampus, GluR2 subunits showed increased levels in both the plasma and postsynaptic membranes of neurons, while pGluR2 expression was reduced in group P. Furthermore, LY294002, the PI3K non-selective antagonist, blocked those effects. CONCLUSION These observations demonstrated that propofol post-conditioning revealed acute neuroprotective role against transient MCAO in rats. The short-term neuroprotective effect was contributed by enhanced GluR2 subunits trafficking to membrane and postsynaptic membranes of neurons, as well as down-regulated the expression of pGluR2 in damaged hippocampus. Finally, the above-mentioned protective mechanism might be contributed by increased combination of PI3K to AMPAR GluR2 subunit, thus maintained the expression and activation of AMPAR GluR2 in the ipsilateral hippocampus.
Collapse
Affiliation(s)
- Chenxu Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Ying Wei
- Department of Anesthesiology, Tianjin People’s Hospital, Tianjin Union Medical Center, Tianjin, 300191 China
| | - Yuan Yuan
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Beibei Dong
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Yuan Shi
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Guolin Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| |
Collapse
|
32
|
Siracusa R, Fusco R, Cuzzocrea S. Astrocytes: Role and Functions in Brain Pathologies. Front Pharmacol 2019; 10:1114. [PMID: 31611796 PMCID: PMC6777416 DOI: 10.3389/fphar.2019.01114] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/30/2019] [Indexed: 12/16/2022] Open
Abstract
Astrocytes are a population of cells with distinctive morphological and functional characteristics that differ within specific areas of the brain. Postnatally, astrocyte progenitors migrate to reach their brain area and related properties. They have a regulatory role of brain functions that are implicated in neurogenesis and synaptogenesis, controlling blood-brain barrier permeability and maintaining extracellular homeostasis. Mature astrocytes also express some genes enriched in cell progenitors, suggesting they can retain proliferative potential. Considering heterogeneity of cell population, it is not surprising that their disorders are related to a wide range of different neuro-pathologies. Brain diseases are characterized by the active inflammatory state of the astrocytes, which is usually described as up-regulation of glial fibrillary acidic protein (GFAP). In particular, the loss of astrocytes function as a result of cellular senescence could have implications for the neurodegenerative disorders, such as Alzheimer disease and Huntington disease, and for the aging brain. Astrocytes can also drive the induction and the progression of the inflammatory state due to their Ca2+ signals and that it is strongly related to the disease severity/state. Moreover, they contribute to the altered neuronal activity in several frontal cortex pathologies such as ischemic stroke and epilepsy. There, we describe the current knowledge pertaining to astrocytes' role in brain pathologies and discuss the possibilities to target them as approach toward pharmacological therapies for neuro-pathologies.
Collapse
Affiliation(s)
- Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Messina, Italy.,Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
33
|
Melegari SP, Fuzinatto CF, Gonçalves RA, Oscar BV, Vicentini DS, Matias WG. Can the surface modification and/or morphology affect the ecotoxicity of zinc oxide nanomaterials? CHEMOSPHERE 2019; 224:237-246. [PMID: 30822730 DOI: 10.1016/j.chemosphere.2019.02.093] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 02/08/2019] [Accepted: 02/14/2019] [Indexed: 06/09/2023]
Abstract
Among nanomaterials, zinc oxide (ZnO) is notable for its excellent biocidal properties. In particular, it can be incorporated in mortars to prevent biofouling. However, the morphology of these nanomaterials (NMs) and their impact on the action against biofouling are still unknown. This study aimed to assess how the morphology and surface modification can affect the ecotoxicology of ZnO NMs. The morphologies evaluated were nanoparticles (NPs) and nanorods (NRs), and the ZnO NMs were tested pure and with surface modification through amine functionalization (@AF). The toxic effects of these NMs were evaluated by acute and chronic ecotoxicity tests with the well-established model microcrustacean Daphnia magna. The ZnO NMs were characterized by transmission electron microscopy, X-ray diffraction and infrared spectroscopy. The EC5048h to D. magna indicated higher acute toxicity of ZnO@AF NRs compared to all tested NMs. Regarding the chronic test with D. magna, high toxic effects on reproduction and longevity were observed with ZnO@AF NRs and effects on growth were observed with ZnO NRs. In general, all tested ZnO NMs presented high toxicity when compared to the positive control, and the NRs presented higher toxicity than NPs in all tested parameters, regardless of the form tested (pure or with surface modification). Additionally, the pathways of ecotoxicity of the tested ZnO NMs was found to be related to combined factors of Zn ion release, effective diameter of particles and NM internalization in the organism.
Collapse
Affiliation(s)
- Silvia Pedroso Melegari
- Department of Sanitation and Environmental Engineering, Federal University of Santa Catarina - UFSC, Campus Universitário, CEP: 88040-970, Florianópolis, SC, Brazil; Center for Marine Studies, Federal University of Paraná - UFPR, Campus Pontal do Paraná, Beira-mar Avenue, 83255-976, Pontal do Paraná, PR, Brazil
| | - Cristiane Funghetto Fuzinatto
- Department of Sanitation and Environmental Engineering, Federal University of Santa Catarina - UFSC, Campus Universitário, CEP: 88040-970, Florianópolis, SC, Brazil; UFFS - Universidade Federal da Fronteira Sul - UFFS, Campus Erechim, CEP: 99700-970, Erechim, RS, Brazil
| | - Renata Amanda Gonçalves
- Department of Sanitation and Environmental Engineering, Federal University of Santa Catarina - UFSC, Campus Universitário, CEP: 88040-970, Florianópolis, SC, Brazil
| | - Bianca Vicente Oscar
- Department of Sanitation and Environmental Engineering, Federal University of Santa Catarina - UFSC, Campus Universitário, CEP: 88040-970, Florianópolis, SC, Brazil
| | - Denice Schulz Vicentini
- Department of Sanitation and Environmental Engineering, Federal University of Santa Catarina - UFSC, Campus Universitário, CEP: 88040-970, Florianópolis, SC, Brazil
| | - William Gerson Matias
- Department of Sanitation and Environmental Engineering, Federal University of Santa Catarina - UFSC, Campus Universitário, CEP: 88040-970, Florianópolis, SC, Brazil.
| |
Collapse
|
34
|
Eom JW, Kim TY, Seo BR, Park H, Koh JY, Kim YH. Identifying New AMP-Activated Protein Kinase Inhibitors That Protect against Ischemic Brain Injury. ACS Chem Neurosci 2019; 10:2345-2354. [PMID: 30763060 DOI: 10.1021/acschemneuro.8b00654] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We recently reported that AMP-activated protein kinase (AMPK) contributes to zinc-induced neuronal death by inducing Bim, a pro-apoptotic Bcl-2 homology domain 3-only protein, in a liver kinase B1 (LKB1)-dependent manner. Current data suggest AMPK plays key roles in excitotoxicity and ischemic brain injury, with zinc neurotoxicity representing at least one mechanism of ischemic neuronal death. Inhibition of AMPK could be a viable therapeutic strategy to prevent ischemic brain injury following stroke. This prompted our search for novel inhibitors of AMPK activity and zinc-induced neuronal death using cultured mouse cortex and a rat model of brain injury after middle cerebral artery occlusion (MCAO). In structure-based virtual screening, 118 compounds were predicted to bind the active site of AMPK α2, and 40 showed in vitro AMPK α2 inhibitory activity comparable to compound C (a well-known, potent AMPK inhibitor). In mouse cortical neuronal cultures, 7 of 40 compound reduced zinc-induced neuronal death at levels comparable to compound C. Ultimately, only agents 2G11 and 1H10 significantly attenuated various types of neuronal death, including oxidative stress, excitotoxicity, and apoptosis. When administered as intracerebroventricular injections prior to permanent MCAO in rats, 2G11 and 1H10 reduced brain infarct volumes, whereas compound C did not. Therefore, these novel AMPK inhibitors could be drug development candidates to treat stroke.
Collapse
Affiliation(s)
- Jae-Won Eom
- Department of Molecular Biology, Sejong University, Seoul 05006, Republic of Korea
| | - Tae-Youn Kim
- Neural Injury Research Laboratory, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea
| | - Bo-Ra Seo
- Neural Injury Research Laboratory, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea
| | - Hwangseo Park
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Jae-Young Koh
- Neural Injury Research Laboratory, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea
- Department of Neurology, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea
| | - Yang-Hee Kim
- Department of Molecular Biology, Sejong University, Seoul 05006, Republic of Korea
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| |
Collapse
|
35
|
Elitt CM, Fahrni CJ, Rosenberg PA. Zinc homeostasis and zinc signaling in white matter development and injury. Neurosci Lett 2019; 707:134247. [PMID: 31059767 DOI: 10.1016/j.neulet.2019.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 02/08/2023]
Abstract
Zinc is an essential dietary micronutrient that is abundant in the brain with diverse roles in development, injury, and neurological diseases. With new imaging tools and chelators selectively targeting zinc, the field of zinc biology is rapidly expanding. The importance of zinc homeostasis is now well recognized in neurodegeneration, but there is emerging data that zinc may be equally important in white matter disorders. This review provides an overview of zinc biology, including a discussion of clinical disorders of zinc deficiency, different zinc pools, zinc biomarkers, and methods for measuring zinc. It emphasizes our limited understanding of how zinc is regulated in oligodendrocytes and white matter. Gaps in knowledge about zinc transporters and zinc signaling are discussed. Zinc-induced oligodendrocyte injury pathways relevant to white matter stroke, multiple sclerosis, and white matter injury of prematurity are reviewed and examples of zinc-dependent proteins relevant to myelination highlighted. Finally, a novel ratiometric zinc sensor is reviewed, revealing new information about mobile zinc during oligodendrocyte differentiation. With a better understanding of zinc biology in oligodendrocytes, new therapeutic targets for white matter disorders may be possible and the necessary tools to appropriately study zinc are finally available.
Collapse
Affiliation(s)
- Christopher M Elitt
- Boston Children's Hospital, Department of Neurology and the F.M. Kirby Neurobiology Center, 300 Longwood Avenue, Boston, MA, United States; Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA.
| | - Christoph J Fahrni
- School of Chemistry and Biochemistry and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Paul A Rosenberg
- Boston Children's Hospital, Department of Neurology and the F.M. Kirby Neurobiology Center, 300 Longwood Avenue, Boston, MA, United States; Program in Neuroscience, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
36
|
Engin AB, Engin A. Nanoparticles and neurotoxicity: Dual response of glutamatergic receptors. PROGRESS IN BRAIN RESEARCH 2019; 245:281-303. [PMID: 30961871 DOI: 10.1016/bs.pbr.2019.03.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although the use of nanoparticles for neuro-diagnostic and neurotherapeutic purposes provides superior benefits than the conventional approaches, it may be potentially toxic in central nervous system. In this respect, nanotechnological research focuses on nanoneurotoxicity-nanoneurosafety concepts. Despite these efforts, nanoparticles (NPs) may cause neurotoxicity, neuroinflammation, and neurodegeneration by penetrating the brain-olfactory route and blood-brain barrier (BBB). Indeed, due to their unique structures nanomaterials can easily cross biological barriers, thus avoid drug delivery problems. Despite the advancement of nanotechnology for designing therapeutic agents, toxicity of these nanomaterials is still a concern. Activation of neurons by astrocytic glutamate is a result of NPs-mediated astrocyte-neuron crosstalk. Increased extracellular glutamate levels due to enhanced synthesis and reduced reuptake may induce neuronal damage by abnormal activation of extrasynaptic N-methyl d-aspartate receptor (NMDAR) subunits. NMDAR is the key factor that mediates the disturbances in intracellular calcium homeostasis, mitochondrial dysfunction and generation of reactive oxygen species in NPs exposed neurons. While some NPs cause neuronal death by inducing NMDARs, others may be neurotoxic through the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors or protect the neurons via blocking NMDARs. However, mechanisms of dual effects of NPs, neurotoxicity or neuroprotection are not precisely known. Some NPs present neuroprotective effect either by selectively inhibiting extrasynaptic subunit of NMDARs or by attenuating oxidative stress. NPs-related proinflammatory activation of microglia contributes to the dysfunction and cytotoxicity in neurons. Therefore, investigation of the interaction of NPs with the neuronal signaling molecules and neuronal receptors is necessary for the better understanding of the neurotoxicity or neurosafety of nanomaterials.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
37
|
Ortiz-Pérez A, Limón-Morales O, Rojas-Castañeda J, Cerbón M, Picazo O. Prolactin prevents the kainic acid-induced neuronal loss in the rat hippocampus by inducing prolactin receptor and putatively increasing the VGLUT1 overexpression. Neurosci Lett 2019; 694:116-123. [DOI: 10.1016/j.neulet.2018.11.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/24/2018] [Accepted: 11/26/2018] [Indexed: 01/08/2023]
|
38
|
Chabosseau P, Woodier J, Cheung R, Rutter GA. Sensors for measuring subcellular zinc pools. Metallomics 2019; 10:229-239. [PMID: 29431830 DOI: 10.1039/c7mt00336f] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Zinc homeostasis is essential for normal cellular function, and defects in this process are associated with a number of diseases including type 2 diabetes (T2D), neurological disorders and cardiovascular disease. Thus, variants in the SLC30A8 gene, encoding the vesicular/granular zinc transporter ZnT8, are associated with altered insulin release and increased T2D risk while the zinc importer ZIP12 is implicated in pulmonary hypertension. In light of these, and findings in other diseases, recent efforts have focused on the development of refined sensors for intracellular free zinc ions that can be targeted to subcellular regions including the cytosol, endoplasmic reticulum (ER), secretory granules, Golgi apparatus, nucleus and the mitochondria. Here, we discuss recent advances in Zn2+ probe engineering and their applications to the measurement of labile subcellular zinc pools in different cell types.
Collapse
Affiliation(s)
- Pauline Chabosseau
- Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| | | | | | | |
Collapse
|
39
|
Ni H, Chen SH, Li LL, Jin MF. Alterations in the Neurobehavioral Phenotype and ZnT3/CB-D28k Expression in the Cerebral Cortex Following Lithium-Pilocarpine-Induced Status Epilepticus: the Ameliorative Effect of Leptin. Biol Trace Elem Res 2019; 187:100-106. [PMID: 29687372 DOI: 10.1007/s12011-018-1343-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 04/10/2018] [Indexed: 12/31/2022]
Abstract
Zinc transporter 3 (ZnT3)-dependent "zincergic" vesicular zinc accounts for approximately 20% of the total zinc content of the mammalian telencephalon. Elevated hippocampal ZnT3 expression is acknowledged to be associated with mossy fiber sprouting and cognitive deficits. However, no studies have compared the long-term neurobehavioral phenotype with the expression of ZnT3 in the cerebral cortex following status epilepticus (SE). The aim of this study was to investigate changes in the long-term neurobehavioral phenotype as well as the expression of ZnT3 and calcium homeostasis-related CB-D28k in the cerebral cortex of rats subjected to neonatal SE and to determine the effects of leptin treatment immediately after neonatal SE. Fifty Sprague-Dawley rats (postnatal day 6, P6) were randomly assigned to two groups: the pilocarpine hydrochloride-induced status epilepticus group (RS, n = 30) and control group (n = 20). Rats were further divided into the control group without leptin (Control), control-plus-leptin treatment group (Leptin), RS group without leptin treatment (RS), and RS-plus-leptin treatment group (RS + Leptin). On P6, all rats in the RS group and RS + Leptin group were injected intraperitoneally (i.p.) with lithium chloride (5 mEq/kg). Pilocarpine (320 mg/kg, i.p.) was administered 30 min after the scopolamine methyl chloride (1 mg/kg) injection on P7. From P8 to P14, animals of the Leptin group and RS + Leptin group were given leptin (4 mg/kg/day, i.p.). The neurological behavioral parameters (negative geotaxis reaction reflex, righting reflex, cliff avoidance reflex, forelimb suspension reflex, and open field test) were observed from P23 to P30. The protein levels of ZnT3 and CB-D28k in the cerebral cortex were detected subsequently by the western blot method. Pilocarpine-treated neonatal rats showed long-term abnormal neurobehavioral parameters. In parallel, there was a significantly downregulated protein level of CB-D28k and upregulated protein level of ZnT3 in the cerebral cortex of the RS group. Leptin treatment soon after epilepticus for 7 consecutive days counteracted these abnormal changes. Taken together with the results from our previous reports on another neonatal seizure model, which showed a significant positive inter-relationship between ZnT3 and calcium/calmodulin-dependent protein kinase IIα (CaMKIIα), the data here suggest that ZnT3/CB-D28k-associated Zn (2+)/Ca(2+) signaling might be involved in neonatal SE-induced long-term brain damage in the aspects of neurobehavioral impairment. Moreover, consecutive leptin treatment is effect at counteracting these hyperexcitability-related changes, suggesting a potential clinical significance.
Collapse
Affiliation(s)
- Hong Ni
- Neurology Laboratory, Institute of Pediatric Research, Children's Hospital of Soochow University, No.303, Jingde Road, 215003, Suzhou, People's Republic of China.
| | - Su-Hong Chen
- Neurology Laboratory, Institute of Pediatric Research, Children's Hospital of Soochow University, No.303, Jingde Road, 215003, Suzhou, People's Republic of China
| | - Li-Li Li
- Neurology Laboratory, Institute of Pediatric Research, Children's Hospital of Soochow University, No.303, Jingde Road, 215003, Suzhou, People's Republic of China
| | - Mei-Fang Jin
- Neurology Laboratory, Institute of Pediatric Research, Children's Hospital of Soochow University, No.303, Jingde Road, 215003, Suzhou, People's Republic of China
| |
Collapse
|
40
|
Zyśk M, Gapys B, Ronowska A, Gul-Hinc S, Erlandsson A, Iwanicki A, Sakowicz-Burkiewicz M, Szutowicz A, Bielarczyk H. Protective effects of voltage-gated calcium channel antagonists against zinc toxicity in SN56 neuroblastoma cholinergic cells. PLoS One 2018; 13:e0209363. [PMID: 30571745 PMCID: PMC6301650 DOI: 10.1371/journal.pone.0209363] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 12/04/2018] [Indexed: 12/13/2022] Open
Abstract
One of the pathological site effects in excitotoxic activation is Zn2+ overload to postsynaptic neurons. Such an effect is considered to be equivalent to the glutamate component of excitotoxicity. Excessive uptake of Zn2+ by active voltage-dependent transport systems in these neurons may lead to significant neurotoxicity. The aim of this study was to investigate whether and which antagonists of the voltage gated calcium channels (VGCC) might modify this Zn2+-induced neurotoxicity in neuronal cells. Our data demonstrates that depolarized SN56 neuronal cells may take up large amounts of Zn2+ and store these in cytoplasmic and mitochondrial sub-fractions. The mitochondrial Zn2+ excess suppressed pyruvate uptake and oxidation. Such suppression was caused by inhibition of pyruvate dehydrogenase complex, aconitase and NADP-isocitrate dehydrogenase activities, resulting in the yielding of acetyl-CoA and ATP shortages. Moreover, incoming Zn2+ increased both oxidized glutathione and malondialdehyde levels, known parameters of oxidative stress. In depolarized SN56 cells, nifedipine treatment (L-type VGCC antagonist) reduced Zn2+ uptake and oxidative stress. The treatment applied prevented the activities of PDHC, aconitase and NADP-IDH enzymes, and also yielded the maintenance of acetyl-CoA and ATP levels. Apart from suppression of oxidative stress, N- and P/Q-type VGCCs presented a similar, but weaker protective influence. In conclusion, our data shows that in the course of excitotoxity, impairment to calcium homeostasis is tightly linked with an excessive neuronal Zn2+ uptake. Hence, the VGCCs types L, N and P/Q share responsibility for neuronal Zn2+ overload followed by significant energy-dependent neurotoxicity. Moreover, Zn2+ affects the target tricarboxylic acid cycle enzymes, yields acetyl-CoA and energy deficits as well.
Collapse
Affiliation(s)
- Marlena Zyśk
- Department of Laboratory Medicine, Medical University of Gdansk, Gdansk, Poland
- * E-mail:
| | - Beata Gapys
- Department of Laboratory Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Anna Ronowska
- Department of Laboratory Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Sylwia Gul-Hinc
- Department of Laboratory Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Anna Erlandsson
- Department of Public Health & Caring Sciences/Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Adam Iwanicki
- Department of Molecular Bacteriology, University of Gdańsk & Medical University of Gdańsk, Gdansk, Poland
| | | | - Andrzej Szutowicz
- Department of Laboratory Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Hanna Bielarczyk
- Department of Laboratory Medicine, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
41
|
Tuo QZ, Liuyang ZY, Lei P, Yan X, Shentu YP, Liang JW, Zhou H, Pei L, Xiong Y, Hou TY, Zhou XW, Wang Q, Wang JZ, Wang XC, Liu R. Zinc induces CDK5 activation and neuronal death through CDK5-Tyr15 phosphorylation in ischemic stroke. Cell Death Dis 2018; 9:870. [PMID: 30158515 PMCID: PMC6115431 DOI: 10.1038/s41419-018-0929-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 02/05/2023]
Abstract
CDK5 activation promotes ischemic neuronal death in stroke, with the recognized activation mechanism being calpain-dependent p35 cleavage to p25. Here we reported that CDK5-Tyr15 phosphorylation by zinc induced CDK5 activation in brain ischemic injury. CDK5 activation and CDK5-Tyr15 phosphorylation were observed in the hippocampus of the rats that had been subjected to middle cerebral artery occlusion, both of which were reversed by pretreatment with zinc chelator; while p35 cleavage and calpain activation in ischemia were not reversed. Zinc incubation resulted in CDK5-Tyr15 phosphorylation and CDK5 activation, without increasing p35 cleavage in cultured cells. Site mutation experiment confirmed that zinc-induced CDK5 activation was dependent on Tyr15 phosphorylation. Further exploration showed that Src kinase contributed to zinc-induced Tyr15 phosphorylation and CDK5 activation. Src kinase inhibition or expression of an unphosphorylable mutant Y15F-CDK5 abolished Tyr15 phosphorylation, prevented CDK5 activation and protected hippocampal neurons from ischemic insult in rats. We conclude that zinc-induced CDK5-Tyr15 phosphorylation underlies CDK5 activation and promotes ischemic neuronal death in stroke.
Collapse
Affiliation(s)
- Qing-Zhang Tuo
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, China
| | - Zhen-Yu Liuyang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, China
| | - Xiong Yan
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang-Ping Shentu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Wei Liang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan Zhou
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Pei
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Xiong
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong-Yao Hou
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin-Wen Zhou
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qun Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Chuan Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
42
|
Savelieff MG, Nam G, Kang J, Lee HJ, Lee M, Lim MH. Development of Multifunctional Molecules as Potential Therapeutic Candidates for Alzheimer’s Disease, Parkinson’s Disease, and Amyotrophic Lateral Sclerosis in the Last Decade. Chem Rev 2018; 119:1221-1322. [DOI: 10.1021/acs.chemrev.8b00138] [Citation(s) in RCA: 270] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Masha G. Savelieff
- SciGency Science Communications, Ann Arbor, Michigan 48104, United States
| | - Geewoo Nam
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Juhye Kang
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Hyuck Jin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Misun Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
43
|
Ji SG, Medvedeva YV, Wang HL, Yin HZ, Weiss JH. Mitochondrial Zn 2+ Accumulation: A Potential Trigger of Hippocampal Ischemic Injury. Neuroscientist 2018; 25:126-138. [PMID: 29742958 DOI: 10.1177/1073858418772548] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Ischemic stroke is a major cause of death and disabilities worldwide, and it has been long hoped that improved understanding of relevant injury mechanisms would yield targeted neuroprotective therapies. While Ca2+ overload during ischemia-induced glutamate excitotoxicity has been identified as a major contributor, failures of glutamate targeted therapies to achieve desired clinical efficacy have dampened early hopes for the development of new treatments. However, additional studies examining possible contributions of Zn2+, a highly prevalent cation in the brain, have provided new insights that may help to rekindle the enthusiasm. In this review, we discuss both old and new findings yielding clues as to sources of the Zn2+ that accumulates in many forebrain neurons after ischemia, and mechanisms through which it mediates injury. Specifically, we highlight the growing evidence of important Zn2+ effects on mitochondria in promoting neuronal injury. A key focus has been to examine Zn2+ contributions to the degeneration of highly susceptible hippocampal pyramidal neurons. Recent studies provide evidence of differences in sources of Zn2+ and its interactions with mitochondria in CA1 versus CA3 neurons that may pertain to their differential vulnerabilities in disease. We propose that Zn2+-induced mitochondrial dysfunction is a critical and potentially targetable early event in the ischemic neuronal injury cascade, providing opportunities for the development of novel neuroprotective strategies to be delivered after transient ischemia.
Collapse
Affiliation(s)
- Sung G Ji
- 1 Department of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | | | - Hwai-Lee Wang
- 2 Department of Neurology, University of California, Irvine, CA, USA.,3 Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Hong Z Yin
- 2 Department of Neurology, University of California, Irvine, CA, USA
| | - John H Weiss
- 1 Department of Anatomy & Neurobiology, University of California, Irvine, CA, USA.,2 Department of Neurology, University of California, Irvine, CA, USA
| |
Collapse
|
44
|
Slepchenko KG, Holub JM, Li YV. Intracellular zinc increase affects phosphorylation state and subcellular localization of protein kinase C delta (δ). Cell Signal 2018; 44:148-157. [DOI: 10.1016/j.cellsig.2018.01.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/14/2018] [Indexed: 10/18/2022]
|
45
|
Intermittent hypoxia-generated ROS contributes to intracellular zinc regulation that limits ischemia/reperfusion injury in adult rat cardiomyocyte. J Mol Cell Cardiol 2018; 118:122-132. [PMID: 29577873 DOI: 10.1016/j.yjmcc.2018.03.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/05/2018] [Accepted: 03/21/2018] [Indexed: 12/20/2022]
Abstract
Intermittent hypoxia (IH) has been shown to exert cardioprotective effects against ischemia/reperfusion (I/R) injury through the preservation of ion homeostasis. I/R dramatically elevated cytosolic Zn2+ and caused cardiomyocyte death. However, the role of IH exposure in the relationship between Zn2+ regulation and cardioprotection is still unclear. The aim of the present study was to study whether IH exposure could help in intracellular Zn2+ regulation, hence contributing to cardioprotection against I/R injury. Adult rat cardiomyocytes were exposed to IH (5% O2, 5% CO2 and balanced N2) for 30 min followed by 30 min of normoxia (21% O2, 5% CO2 and balanced N2). Changes in intracellular Zn2+ concentration were determined using a Zn2+-specific fluorescent dye, FluoZin-3 or RhodZin-3. Fluorescence was monitored under an inverted fluorescent or confocal microscope. The results demonstrated that I/R or 2,2'-dithiodipyridine (DTDP), a reactive disulphide compound, induced Zn2+ release from metallothioneins (MTs), subsequently causing cytosolic Zn2+ overload, which in turn increased intracellular Zn2+ entry into the mitochondria via a Ca2+ uniporter, hence inducing mitochondrial membrane potential loss, and eventually led to cell death. However, the cytosolic Zn2+ overload and cell death caused by I/R or DTDP was significantly reduced by treatment of cardiomyocytes with IH. The findings from this study suggest that IH might exert its cardioprotective effect through reducing the I/R-induced cytosolic Zn2+ overload and cell death in cardiomyocytes.
Collapse
|
46
|
Ji SG, Weiss JH. Zn 2+-induced disruption of neuronal mitochondrial function: Synergism with Ca 2+, critical dependence upon cytosolic Zn 2+ buffering, and contributions to neuronal injury. Exp Neurol 2018; 302:181-195. [PMID: 29355498 DOI: 10.1016/j.expneurol.2018.01.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 12/15/2017] [Accepted: 01/15/2018] [Indexed: 10/18/2022]
Abstract
Excitotoxic Zn2+ and Ca2+ accumulation contributes to neuronal injury after ischemia or prolonged seizures. Synaptically released Zn2+ can enter postsynaptic neurons via routes including voltage sensitive Ca2+ channels (VSCC), and, more rapidly, through Ca2+ permeable AMPA channels. There are also intracellular Zn2+ binding proteins which can either buffer neuronal Zn2+ influx or release bound Zn2+ into the cytosol during pathologic conditions. Studies in culture highlight mitochondria as possible targets of Zn2+; cytosolic Zn2+ can enter mitochondria and induce effects including loss of mitochondrial membrane potential (ΔΨm), mitochondrial swelling, and reactive oxygen species (ROS) generation. While brief (5 min) neuronal depolarization (to activate VSCC) in the presence of 300 μM Zn2+ causes substantial delayed neurodegeneration, it only mildly impacts acute mitochondrial function, raising questions as to contributions of Zn2+-induced mitochondrial dysfunction to neuronal injury. Using brief high (90 mM) K+/Zn2+ exposures to mimic neuronal depolarization and extracellular Zn2+ accumulation as may accompany ischemia in vivo, we examined effects of disrupted cytosolic Zn2+ buffering and/or the presence of Ca2+, and made several observations: 1. Mild disruption of cytosolic Zn2+ buffering-while having little effects alone-markedly enhanced mitochondrial Zn2+ accumulation and dysfunction (including loss of ∆Ψm, ROS generation, swelling and respiratory inhibition) caused by relatively low (10-50 μM) Zn2+ with high K+. 2. The presence of Ca2+ during the Zn2+ exposure decreased cytosolic and mitochondrial Zn2+ accumulation, but markedly exacerbated the consequent dysfunction. 3. Paralleling effects on mitochondria, disruption of buffering and presence of Ca2+ enhanced Zn2+-induced neurodegeneration. 4. Zn2+ chelation after the high K+/Zn2+ exposure attenuated both ROS production and neurodegeneration, supporting the potential utility of delayed interventions. Taken together, these data lend credence to the idea that in pathologic states that impair cytosolic Zn2+ buffering, slow uptake of Zn2+ along with Ca2+ into neurons via VSCC can disrupt the mitochondria and induce neurodegeneration.
Collapse
Affiliation(s)
- Sung G Ji
- Department of Anatomy & Neurobiology, University of California, Irvine, USA
| | - John H Weiss
- Department of Anatomy & Neurobiology, University of California, Irvine, USA; Department of Neurology, University of California, Irvine, USA.
| |
Collapse
|
47
|
Li X, Yang W, Jiang LH. Alteration in Intracellular Zn 2+ Homeostasis as a Result of TRPM2 Channel Activation Contributes to ROS-Induced Hippocampal Neuronal Death. Front Mol Neurosci 2017; 10:414. [PMID: 29311807 PMCID: PMC5732979 DOI: 10.3389/fnmol.2017.00414] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 11/29/2017] [Indexed: 12/23/2022] Open
Abstract
Transient receptor potential melastatin-related 2 (TRPM2) channel, a molecular sensor for reactive oxygen species (ROS), plays an important role in cognitive dysfunction associated with post-ischemia brain damage thought to result from ROS-induced TRPM2-dependent neuronal death during reperfusion. Emerging evidence further suggests that an alteration in the Zn2+ homeostasis is critical in ROS-induced TRPM2-dependent neuronal death. Here we applied genetic and pharmacological interventions to define the role of TRPM2 channel in ROS-induced neuronal death and explore the mechanisms contributing in the alteration in intracellular Zn2+ homeostasis in mouse hippocampal neurons. Exposure of neurons to 30–300 μM H2O2 for 2–24 h caused concentration/duration-dependent neuronal death, which was significantly suppressed, but not completely prevented, by TRPM2-knockout (TRPM2-KO) and pharmacological inhibition of the TRPM2 channel. H2O2-induced neuronal death was also attenuated by treatment with TPEN acting as a Zn2+ selective chelator. Single cell imaging demonstrated that H2O2 evoked a prominent increase in the intracellular Zn2+ concentration, which was completely prevented by TPEN as well as TRPM2-KO and inhibition of the TRPM2 channel. Furthermore, H2O2 induced lysosomal Zn2+ release and lysosomal dysfunction, and subsequent mitochondrial Zn2+ accumulation that provokes mitochondrial dysfunction and ROS generation. These H2O2-induced lysosomal/mitochondrial effects were prevented by TRPM2-KO or TPEN. Taken together, our results provide evidence to show that a dynamic alteration in the intracellular Zn2+ homeostasis as a result of activation of the TRPM2 channel contributes to ROS-induced hippocampal neuronal death.
Collapse
Affiliation(s)
- Xin Li
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Wei Yang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.,Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin-Hua Jiang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom.,Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
48
|
Wang JC, Bindokas VP, Skinner M, Emrick T, Marks JD. Mitochondrial mechanisms of neuronal rescue by F-68, a hydrophilic Pluronic block co-polymer, following acute substrate deprivation. Neurochem Int 2017; 109:126-140. [PMID: 28433663 PMCID: PMC5641222 DOI: 10.1016/j.neuint.2017.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/29/2017] [Accepted: 04/10/2017] [Indexed: 01/09/2023]
Abstract
Global brain ischemia can lead to widespread neuronal death and poor neurologic outcomes in patients. Despite detailed understanding of the cellular and molecular mechanisms mediating neuronal death following focal and global brain hypoxia-ischemia, treatments to reduce ischemia-induced brain injury remain elusive. One pathway central to neuronal death following global brain ischemia is mitochondrial dysfunction, one consequence of which is the cascade of intracellular events leading to mitochondrial outer membrane permeabilization. A novel approach to rescuing injured neurons from death involves targeting cellular membranes using a class of synthetic molecules called Pluronics. Pluronics are triblock copolymers of hydrophilic poly[ethylene oxide] (PEO) and hydrophobic poly[propylene oxide] (PPO). Evidence is accumulating to suggest that hydrophilic Pluronics rescue injured neurons from death following substrate deprivation by preventing mitochondrial dysfunction. Here, we will review current understanding of the nature of interaction of Pluronic molecules with biological membranes and the efficacy of F-68, an 80% hydrophilic Pluronic, in rescuing neurons from injury. We will review data indicating that F-68 reduces mitochondrial dysfunction and mitochondria-dependent death pathways in a model of neuronal injury in vitro, and present new evidence that F-68 acts directly on mitochondria to inhibit mitochondrial outer membrane permeabilization. Finally, we will present results of a pilot, proof-of-principle study suggesting that F-68 is effective in reducing hippocampal injury induced by transient global ischemia in vivo. By targeting mitochondrial dysfunction, F-68 and other Pluronic molecules constitute an exciting new approach to rescuing neurons from acute injury.
Collapse
Affiliation(s)
- Janice C Wang
- Department of Pediatrics, University of Chicago, Chicago, IL, United States
| | - Vytautas P Bindokas
- Department of Pharmacological, Physiological Sciences, University of Chicago, IL, United States
| | - Matthew Skinner
- Department of Polymer Science and Engineering, University of Massachusetts, Amherst, MA, United States
| | - Todd Emrick
- Department of Polymer Science and Engineering, University of Massachusetts, Amherst, MA, United States
| | - Jeremy D Marks
- Department of Pediatrics, University of Chicago, Chicago, IL, United States; Grossman Institute for Neuroscience, Quantitative Biology and Human Behavior, University of Chicago, Chicago, IL, United States.
| |
Collapse
|
49
|
Zieminska E, Ruszczynska A, Lazarewicz JW. Tetrabromobisphenol A disturbs zinc homeostasis in cultured cerebellar granule cells: A dual role in neurotoxicity. Food Chem Toxicol 2017; 109:363-375. [PMID: 28919410 DOI: 10.1016/j.fct.2017.09.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 01/15/2023]
Abstract
The brominated flame retardant tetrabromobisphenol A (TBBPA) has recognized neurotoxic properties mediated by intracellular Ca2+ imbalance and oxidative stress. Although these factors are known to trigger the release of Zn2+ from intracellular stores, the effects of TBBPA on Zn2+ homeostasis in neurons and the role of Zn2+in TBBPA neurotoxicity have not yet been studied. Therefore, we investigated zinc transients in primary cultures of rat cerebellar granule cells and assessed their involvement in TBBPA neurotoxicity. The results demonstrate that TBBPA releases Zn2+ from the intracellular stores and increases its intracellular concentration, followed by Zn2+ displacement from the cells. TBBPA-evoked Zn2+ transients are partially mediated by Ca2+ and ROS. Application of TPEN, Zn2+ chelator, potentiates TBBPA- and glutamate-induced 45Ca uptake, enhances TBBPA-induced ROS production and potentiates decreases in the ΔΨm in cells treated with 25 μM TBBPA, revealing the potential neuroprotective capacity of endogenous Zn2+. However, the administration of TPEN does not aggravate TBBPA neurotoxicity, and even slightly decreases neuronal death induced by 25 μM TBBPA. In summary, it was shown for the first time that TBBPA interferes with the cellular Zn2+ homeostasis in neuronal cultures, and we revealed complex roles for endogenous Zn2+ in cytoprotection and TBBPA toxicity in cultured neurons.
Collapse
Affiliation(s)
- Elzbieta Zieminska
- Department of Neurochemistry, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.
| | - Anna Ruszczynska
- University of Warsaw, Faculty of Chemistry, Biological and Chemical Research Centre, Warsaw, Poland
| | - Jerzy W Lazarewicz
- Department of Neurochemistry, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
50
|
Schulien AJ, Justice JA, Di Maio R, Wills ZP, Shah NH, Aizenman E. Zn(2+) -induced Ca(2+) release via ryanodine receptors triggers calcineurin-dependent redistribution of cortical neuronal Kv2.1 K(+) channels. J Physiol 2017; 594:2647-59. [PMID: 26939666 DOI: 10.1113/jp272117] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/14/2016] [Indexed: 12/30/2022] Open
Abstract
KEY POINTS Increases in intracellular Zn(2+) concentrations are an early, necessary signal for the modulation of Kv2.1 K(+) channel localization and physiological function. Intracellular Zn(2+) -mediated Kv2.1 channel modulation is dependent on calcineurin, a Ca(2+) -activated phosphatase. We show that intracellular Zn(2+) induces a significant increase in ryanodine receptor-dependent cytosolic Ca(2+) transients, which leads to a calcineurin-dependent redistribution of Kv2.1 channels from pre-existing membrane clusters to diffuse localization. As such, the link between Zn(2+) and Ca(2+) signalling in this Kv2.1 modulatory pathway is established. We observe that a sublethal ischaemic preconditioning insult also leads to Kv2.1 redistribution in a ryanodine receptor-dependent fashion. We suggest that Zn(2+) may be an early and ubiquitous signalling molecule mediating Ca(2+) release from the cortical endoplasmic reticulum via ryanodine receptor activation. ABSTRACT Sublethal injurious stimuli in neurons induce transient increases in free intracellular Zn(2+) that are associated with regulating adaptive responses to subsequent lethal injury, including alterations in the function and localization of the delayed-rectifier potassium channel, Kv2.1. However, the link between intracellular Zn(2+) signalling and the observed changes in Kv2.1 remain undefined. In the present study, utilizing exogenous Zn(2+) treatment, along with a selective Zn(2+) ionophore, we show that transient elevations in intracellular Zn(2+) concentrations are sufficient to induce calcineurin-dependent Kv2.1 channel dispersal in rat cortical neurons in vitro, which is accompanied by a relatively small but significant hyperpolarizing shift in the voltage-gated activation kinetics of the channel. Critically, using a molecularly encoded calcium sensor, we found that the calcineurin-dependent changes in Kv2.1 probably occur as a result of Zn(2+) -induced cytosolic Ca(2+) release via activation of neuronal ryanodine receptors. Finally, we couple this mechanism with an established model for in vitro ischaemic preconditioning and show that Kv2.1 channel modulation in this process is also ryanodine receptor-sensitive. Our results strongly suggest that intracellular Zn(2+) -initiated signalling may represent an early and possibly widespread component of Ca(2+) -dependent processes in neurons.
Collapse
Affiliation(s)
- Anthony J Schulien
- Department of Neurobiology.,Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, PA, USA
| | - Jason A Justice
- Department of Neurobiology.,Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, PA, USA
| | - Roberto Di Maio
- Department of Neurology.,Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, PA, USA
| | | | | | - Elias Aizenman
- Department of Neurobiology.,Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, PA, USA
| |
Collapse
|