1
|
Lane AR, Roberts BR, Fahrni CJ, Faundez V. A primer on copper biology in the brain. Neurobiol Dis 2025:106974. [PMID: 40414313 DOI: 10.1016/j.nbd.2025.106974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2025] [Revised: 05/14/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025] Open
Abstract
This primer aims to expose scientists who study the brain to the field of copper biology. We briefly discuss key copper homeostasis mechanisms and proteins and place these functions in the context of the brain and neurodevelopment. A small number of key copper genes are explored as representative examples of the importance of this metal to the brain. We show that these genes are expressed throughout the brain and their defects are linked to a diverse array of neurological phenotypes, which we discuss further in the context of several neurological and neurodegenerative diseases associated with dysregulation of copper. This review aims to expose interested scientists to the fundamental roles for copper in the brain, the primary proteins responsible for maintaining copper homeostasis in the brain, and the classic neurological diseases associated with this metal.
Collapse
Affiliation(s)
- Alicia R Lane
- Department of Cell Biology, Emory University, 615 Michael St, Atlanta, GA 30322, USA.
| | - Blaine R Roberts
- Department of Biochemistry, Emory University, 1510 Clifton Rd, Atlanta, GA 30322, USA; Department of Neurology, Emory University, 12 Executive Park Dr NE, Atlanta, GA 30322, USA.
| | - Christoph J Fahrni
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - Victor Faundez
- Department of Cell Biology, Emory University, 615 Michael St, Atlanta, GA 30322, USA.
| |
Collapse
|
2
|
Locatelli M, Farina C. Role of copper in central nervous system physiology and pathology. Neural Regen Res 2025; 20:1058-1068. [PMID: 38989937 PMCID: PMC11438321 DOI: 10.4103/nrr.nrr-d-24-00110] [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: 01/26/2024] [Revised: 03/21/2024] [Accepted: 04/23/2024] [Indexed: 07/12/2024] Open
Abstract
Copper is a transition metal and an essential element for the organism, as alterations in its homeostasis leading to metal accumulation or deficiency have pathological effects in several organs, including the central nervous system. Central copper dysregulations have been evidenced in two genetic disorders characterized by mutations in the copper-ATPases ATP7A and ATP7B, Menkes disease and Wilson's disease, respectively, and also in multifactorial neurological disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. This review summarizes current knowledge about the role of copper in central nervous system physiology and pathology, reports about unbalances in copper levels and/or distribution under disease, describes relevant animal models for human disorders where copper metabolism genes are dysregulated, and discusses relevant therapeutic approaches modulating copper availability. Overall, alterations in copper metabolism may contribute to the etiology of central nervous system disorders and represent relevant therapeutic targets to restore tissue homeostasis.
Collapse
Affiliation(s)
- Martina Locatelli
- Institute of Experimental Neurology, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Cinthia Farina
- Institute of Experimental Neurology, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
3
|
Mamsa SSA, Ellison G, Koehn J, Inder-Smith K, Evans CW, Graham RM, Howard DL, Hackett MJ. Correlative analysis of metallomic gene expression and metal ion content within the mouse hippocampus. Metallomics 2025; 17:mfaf009. [PMID: 40175292 PMCID: PMC12086694 DOI: 10.1093/mtomcs/mfaf009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Indexed: 04/04/2025]
Abstract
Brain metal homeostasis is essential for healthy neurological function, and disturbed brain metal homeostasis has deleterious consequences for neurodevelopment or cognitive outcome following injury or during disease. Specific regions of the brain (e.g. the hippocampus and subregions within) are known to be enriched with transition metals (i.e. ions of iron, copper, and zinc). Neither the physiological need for localized enrichment, nor the mechanisms driving the enrichment, however, are well understood. In this study we have applied a multimodal template, incorporating elemental mapping using X-ray fluorescence microscopy with spatial transcriptomics, to help reveal a molecular basis for metallomic heterogeneity across key subregions of the hippocampus. Our results reveal that significant differences in iron, zinc, and copper enrichment are associated with regional enrichment of specific transcripts related to metal transport, metal storage, and metal regulatory proteins. In addition to providing novel biological insight into the neurometallomic profile of the hippocampus, this study also provides an important template for others to integrate transcriptomics into multimodal workflows investigating the neurometallome.
Collapse
Affiliation(s)
- Somayra S A Mamsa
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6102, Australia
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Gaewyn Ellison
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6102, Australia
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA 6102, Australia
| | - Julia Koehn
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6102, Australia
| | - Keea Inder-Smith
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6102, Australia
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Cameron W Evans
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Ross M Graham
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6102, Australia
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Daryl L Howard
- Australian Synchrotron, ANSTO, Clayton, VIC 3168, Australia
| | - Mark J Hackett
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6102, Australia
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA 6102, Australia
| |
Collapse
|
4
|
Hollings AL, Ellison GC, Willans M, Lam V, Munyard T, Remy AR, Takechi R, Mamo JCL, Webb S, New EJ, James SA, Glover C, Klein A, Vongsvivut J, Howard D, Hackett MJ. Subventricular Accumulation of Cu in the Aging Mouse Brain Does Not Associate with Anticipated Increases in Markers of Oxidative Stress. ACS Chem Neurosci 2025; 16:292-302. [PMID: 39873122 DOI: 10.1021/acschemneuro.4c00320] [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] [Indexed: 01/30/2025] Open
Abstract
Natural aging is associated with mild memory loss and cognitive decline, and age is the greatest risk factor for neurodegenerative diseases, such as Alzheimer's disease. There is substantial evidence that oxidative stress is a major contributor to both natural aging and neurodegenerative disease, and coincidently, levels of redox active metals such as Fe and Cu are known to be elevated later in life. Recently, a pronounced age-related increase in Cu content has been reported to occur in mice and rats around a vital regulatory brain region, the subventricular zone of lateral ventricles. In our study herein, we have characterized lateral ventricle Cu content in a unique murine model of accelerated aging, senescence accelerated mouse-prone 8 (SAMP8) mice. Our results confirm an age-related increase in ventricle Cu content, consistent with the studies by others in wild-type mice and rats. Specifically, we observed Cu content to increase over the time frame 1 to 5 months and 5 to 9 months, but interestingly, no significant increase occurred between 9 and 12 months (although brain Cu content at 12 months was significantly elevated relative to 1 and 5 month-old animals). Despite the magnitude of Cu increase observed within the cells that comprise the subventricular zone of lateral ventricles (average 3 mM Cu, with isolated subcellular concentrations of 17 mM), we did not detect spectroscopic markers of thiol oxidation, protein aggregation, or lipid oxidation. The lack of evidence for oxidative stress in ex vivo animal tissue is in contrast to in vitro studies demonstrating that thiol, protein, and lipid oxidation is pronounced at these Cu concentrations. We suggest that our findings most likely indicate that the Cu ions in this brain region are sequestered in an unreactive form, possibly extended chains of Cu-thiolate complexes, which do not readily redox cycle in the aqueous cytosol. These results also appear to partially challenge the long-held view that age-related increases in brain metal content drive oxidative stress as we did not observe a concomitant association between age-related Cu increase and markers of oxidative stress, nor did we observe a net increase in Cu content between mice aged 9 and 12 months.
Collapse
Affiliation(s)
- Ashley L Hollings
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA 6845, Australia
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6845, Australia
| | - Gaewyn C Ellison
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA 6845, Australia
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6845, Australia
| | - Meg Willans
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA 6845, Australia
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6845, Australia
| | - Virginie Lam
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6845, Australia
- School of Population Health, Faculty of Health Sciences, Curtin University, Bentley, WA 6845, Australia
- Perron Institute for Neurological and Translational Sciences, 8 Verdun Street, Nedlands, WA 6009, Australia
| | - Thomas Munyard
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA 6845, Australia
| | - Aedena-Raquel Remy
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW 2006, Australia
| | - Ryu Takechi
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6845, Australia
- School of Population Health, Faculty of Health Sciences, Curtin University, Bentley, WA 6845, Australia
- Perron Institute for Neurological and Translational Sciences, 8 Verdun Street, Nedlands, WA 6009, Australia
| | - John C L Mamo
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6845, Australia
- School of Population Health, Faculty of Health Sciences, Curtin University, Bentley, WA 6845, Australia
- Perron Institute for Neurological and Translational Sciences, 8 Verdun Street, Nedlands, WA 6009, Australia
| | - Sam Webb
- SLAC National Accelerator Laboratory, Stanford Synchrotron Radiation Lightsource, Menlo Park, California 94025, United States
| | - Elizabeth J New
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW 2006, Australia
- Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Simon A James
- Medium Energy X-ray Absorption Spectroscopy (MEX) Beamline, ANSTO-Australian Synchrotron, 800 Blackburn Road, Clayton, VIC 3168, Australia
| | - Chris Glover
- Medium Energy X-ray Absorption Spectroscopy (MEX) Beamline, ANSTO-Australian Synchrotron, 800 Blackburn Road, Clayton, VIC 3168, Australia
| | - Annaleise Klein
- Infrared Microspectroscopy (IRM) Beamline, ANSTO-Australian Synchrotron, 800 Blackburn Road, Clayton, VIC 3168, Australia
| | - Jitraporn Vongsvivut
- Infrared Microspectroscopy (IRM) Beamline, ANSTO-Australian Synchrotron, 800 Blackburn Road, Clayton, VIC 3168, Australia
| | - Daryl Howard
- X-ray Fluorescence Microscopy (XFM) Beamline, ANSTO-Australian Synchrotron, 800 Blackburn Road, Clayton, VIC3168Australia
| | - Mark J Hackett
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA 6845, Australia
- Curtin Medical Research Institute, Curtin University, Bentley, WA 6845, Australia
| |
Collapse
|
5
|
Hadrian K, Szczerbowska-Boruchowska M, Surówka A, Ciepiela O, Litwin T, Przybyłkowski A. Effect of primary copper metabolism disturbance on elemental, protein, and lipid composition of the organs in Jackson toxic milk mouse. Biometals 2025; 38:103-121. [PMID: 39365499 PMCID: PMC11754380 DOI: 10.1007/s10534-024-00640-y] [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: 07/17/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024]
Abstract
Toxic milk (txJ) is an autosomal recessive mutation in the Atp7b gene in the C3H/HeJ strain, observed at The Jackson Laboratory in Maine, USA. TxJ mice exhibit symptoms similar to those of human Wilson's disease (WD). The study aimed to verify organ involvement in a mouse model of WD. TxJ mice and control animals were sacrificed at 2, 4, 8, and 14 months of age. Total X-ray Fluorescence Spectroscopy (TXRF) was used to determine the elemental concentration in organs. Tissue chemical composition was measured by Fourier Transform Infrared Spectroscopy (FTIR). Additionally, hybrid mapping of FTIR and microXRF was performed. Elevated concentrations of Cu were observed in the liver, striatum, eye, heart, and duodenum of txJ mice across age groups. In the striatum of the oldest txJ mice, there was lower lipid content and a higher fraction of saturated fats. The secondary structure of striatum proteins was disturbed in txJ mice. In the livers of txJ mice, higher concentrations of saturated fats and disturbances in the secondary structure of proteins were observed. The concentration of neurofilaments was significantly higher in txJ serum. The distribution of Cu deposits in brains was uniform with no prevalence in any anatomic structure in either group, but significant protein structure changes were observed exclusively in the striatum of txJ. In this txJ animal model of WD, pathologic copper accumulation occurs in the duodenum, heart, and eye tissues. Increased copper concentration in the liver and brain results in increased saturated fat content and disturbances in secondary protein structure, leading to hepatic injury and neurodegeneration.
Collapse
Affiliation(s)
- Krzysztof Hadrian
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | | | - Artur Surówka
- Department of Medical Physics and Biophysics, AGH University of Science and Technology, Cracow, Poland
| | - Olga Ciepiela
- Department of Laboratory Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
6
|
Willans M, Hollings A, Boseley RE, Munyard T, Ellison GC, Hackett MJ. The application of X-ray fluorescence microscopy and micro-XANES spectroscopy to study neuro-metallomics. J Inorg Biochem 2025; 262:112744. [PMID: 39341704 DOI: 10.1016/j.jinorgbio.2024.112744] [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: 06/04/2024] [Revised: 09/02/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
This early career research highlight provides a review of my own research program over the last decade, a time frame that encompasses my transition from postdoctoral fellowships to independent researcher. As an analytical chemist and applied spectroscopist, the central theme of my research program over this time has been protocol development at synchrotron facilities, with the main objective to investigate brain metal homeostasis during both brain health and brain disease. I will begin my review with an overview of brain metal homeostasis, before introducing analytical challenges associated with its study. I will then provide a brief summary of the two main X-ray techniques I have used to study brain metal homeostasis, X-ray fluorescence microscopy (XFM) and X-ray absorption near edge structure spectroscopy (XANES). The review then finishes with a summary of my main research contributions using these two techniques, put in the context of the results from others in the field.
Collapse
Affiliation(s)
- Meg Willans
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA, Australia
| | - Ashley Hollings
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Rhiannon E Boseley
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA, Australia
| | - Thomas Munyard
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA, Australia
| | - Gaewyn C Ellison
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Mark J Hackett
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia.
| |
Collapse
|
7
|
Lutsenko S, Roy S, Tsvetkov P. Mammalian copper homeostasis: physiological roles and molecular mechanisms. Physiol Rev 2025; 105:441-491. [PMID: 39172219 PMCID: PMC11918410 DOI: 10.1152/physrev.00011.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/15/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024] Open
Abstract
In the past decade, evidence for the numerous roles of copper (Cu) in mammalian physiology has grown exponentially. The discoveries of Cu involvement in cell signaling, autophagy, cell motility, differentiation, and regulated cell death (cuproptosis) have markedly extended the list of already known functions of Cu, such as a cofactor of essential metabolic enzymes, a protein structural component, and a regulator of protein trafficking. Novel and unexpected functions of Cu transporting proteins and enzymes have been identified, and new disorders of Cu homeostasis have been described. Significant progress has been made in the mechanistic studies of two classic disorders of Cu metabolism, Menkes disease and Wilson's disease, which paved the way for novel approaches to their treatment. The discovery of cuproptosis and the role of Cu in cell metastatic growth have markedly increased interest in targeting Cu homeostatic pathways to treat cancer. In this review, we summarize the established concepts in the field of mammalian Cu physiology and discuss how new discoveries of the past decade expand and modify these concepts. The roles of Cu in brain metabolism and in cell functional speciation and a recently discovered regulated cell death have attracted significant attention and are highlighted in this review.
Collapse
Affiliation(s)
- Svetlana Lutsenko
- Department of Physiology, Johns Hopkins Medical Institutes, Baltimore, Maryland, United States
| | - Shubhrajit Roy
- Department of Physiology, Johns Hopkins Medical Institutes, Baltimore, Maryland, United States
| | - Peter Tsvetkov
- Department of Pathology, Cancer Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| |
Collapse
|
8
|
Roy S, Lutsenko S. Mechanism of Cu entry into the brain: many unanswered questions. Neural Regen Res 2024; 19:2421-2429. [PMID: 38526278 PMCID: PMC11090436 DOI: 10.4103/1673-5374.393107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/10/2023] [Accepted: 12/09/2023] [Indexed: 03/26/2024] Open
Abstract
Brain tissue requires high amounts of copper (Cu) for its key physiological processes, such as energy production, neurotransmitter synthesis, maturation of neuropeptides, myelination, synaptic plasticity, and radical scavenging. The requirements for Cu in the brain vary depending on specific brain regions, cell types, organism age, and nutritional status. Cu imbalances cause or contribute to several life-threatening neurologic disorders including Menkes disease, Wilson disease, Alzheimer's disease, Parkinson's disease, and others. Despite the well-established role of Cu homeostasis in brain development and function, the mechanisms that govern Cu delivery to the brain are not well defined. This review summarizes available information on Cu transfer through the brain barriers and discusses issues that require further research.
Collapse
Affiliation(s)
- Shubhrajit Roy
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Svetlana Lutsenko
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
Shi W, Zhou Q, Lu L, Zhang Y, Zhang H, Pu Y, Yin L. Copper induced cytosolic escape of mitochondrial DNA and activation of cGAS-STING-NLRP3 pathway-dependent pyroptosis in C8-D1A cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117085. [PMID: 39321529 DOI: 10.1016/j.ecoenv.2024.117085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Copper, a vital mineral nutrient, possesses redox qualities that make it both beneficial and toxic to organisms. Excessive environmental copper exposure can result in neurological damage and cognitive decline in humans. Astrocytes, the predominant glial cells in the brain, are particularly vulnerable to pollutants, but the mechanism of copper-induced damage to astrocytes remains elusive. The aim of this study was to determine the role of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway in initiating NLRP3 inflammasome-induced astrocyte pyroptosis and chronic inflammation under conditions of copper overload. Our findings indicated that copper exposure elevated mitochondrial ROS (mtROS) levels, resulting in mitochondrial damage in astrocytes. This damage caused the release of mitochondrial DNA (mtDNA) into the cytoplasm, which subsequently activated the cGAS-STING pathway. This activation resulted in interactions between STING and NLRP3 proteins, facilitating the assembly of the NLRP3 inflammasome and inducing pyroptosis. Furthermore, depletion of mtROS mitigated copper-induced mitochondrial damage in astrocytes and reduced mtDNA leakage. Pharmacological inhibition of STING or STING transfection further reversed copper-induced pyroptosis and the inflammatory response. In conclusion, this study demonstrated that the leakage of mtDNA into the cytoplasm and the subsequent activation of the cGAS-STING-NLRP3 pathway may be potential mechanisms underlying copper-induced pyroptosis in astrocytes. These findings provided new insights into the toxicity of copper.
Collapse
Affiliation(s)
- Wei Shi
- School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Qian Zhou
- School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Lu Lu
- School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Ying Zhang
- School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Hu Zhang
- School of Public Health, Yangzhou University, Yangzhou 225000, China.
| | - Yuepu Pu
- School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Lihong Yin
- School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education of China, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
10
|
Evans CW, Egid A, Mamsa SSA, Paterson DJ, Ho D, Bartlett CA, Fehily B, Lins BR, Fitzgerald M, Hackett MJ, Smith NM. Elemental Mapping in a Preclinical Animal Model Reveals White Matter Copper Elevation in the Acute Phase of Central Nervous System Trauma. ACS Chem Neurosci 2023; 14:3518-3527. [PMID: 37695072 DOI: 10.1021/acschemneuro.3c00421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023] Open
Abstract
Understanding the chemical events following trauma to the central nervous system could assist in identifying causative mechanisms and potential interventions to protect neural tissue. Here, we apply a partial optic nerve transection model of injury in rats and use synchrotron X-ray fluorescence microscopy (XFM) to perform elemental mapping of metals (K, Ca, Fe, Cu, Zn) and other related elements (P, S, Cl) in white matter tracts. The partial optic nerve injury model and spatial precision of microscopy allow us to obtain previously unattained resolution in mapping elemental changes in response to a primary injury and subsequent secondary effects. We observed significant elevation of Cu levels at multiple time points following the injury, both at the primary injury site and in neural tissue near the injury site vulnerable to secondary damage, as well as significant changes in Cl, K, P, S, and Ca. Our results suggest widespread metal dyshomeostasis in response to central nervous system trauma and that altered Cu homeostasis may be a specific secondary event in response to white matter injury. The findings highlight metal homeostasis as a potential point of intervention in limiting damage following nervous system injury.
Collapse
Affiliation(s)
- Cameron W Evans
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Abigail Egid
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia
- University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
| | - Somayra S A Mamsa
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | | | - Diwei Ho
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Carole A Bartlett
- Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia
| | - Brooke Fehily
- Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia
- Perron Institute for Neurological and Translational Sciences, 8 Verdun Street, Nedlands, WA 6009, Australia
| | - Brittney R Lins
- Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia
- Perron Institute for Neurological and Translational Sciences, 8 Verdun Street, Nedlands, WA 6009, Australia
| | - Melinda Fitzgerald
- Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia
- Perron Institute for Neurological and Translational Sciences, 8 Verdun Street, Nedlands, WA 6009, Australia
| | - Mark J Hackett
- Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, WA 6102, Australia
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA 6102, Australia
| | - Nicole M Smith
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| |
Collapse
|
11
|
Li X, Bai Y, Huo H, Wu H, Liao J, Han Q, Zhang H, Hu L, Li Y, Pan J, Tang Z, Guo J. Long-term Copper Exposure Induces Mitochondrial Dynamics Disorder and Mitophagy in the Cerebrum of Pigs. Biol Trace Elem Res 2023; 201:1197-1204. [PMID: 35616827 DOI: 10.1007/s12011-022-03224-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/28/2022] [Indexed: 02/07/2023]
Abstract
Copper (Cu) is an essential trace element for growth and development in most organisms. However, environmental exposure to high doses of Cu can damage multiple organs. To investigate the underlying mechanism of Cu toxicity on mitochondrial dynamics and mitophagy in the cerebrum of pigs, 60 30-day-old pigs were randomly divided into three groups and treated with different contents of anhydrous Cu sulfate in the diets (Cu 10 mg/kg, control group; Cu 125 mg/kg, group I; Cu 250 mg/kg, group II) for 80 days. The Cu levels and histological changes in the cerebrum were measured. Moreover, the protein and mRNA expression levels related to mitophagy and mitochondrial dynamics were determined. The results showed that the contents of Cu were increased in the cerebrum with increasing dietary Cu. Vacuolar degeneration was found in group I and group II compared to the control group. Additionally, the protein and mRNA expression levels of PINK1, Parkin, and Drp1 and the protein level of LC3-II were remarkably upregulated with increasing levels of dietary Cu. Nevertheless, the protein and mRNA expression levels of MFN1 and MFN2 and the mRNA expression of P62 were obviously downregulated in a Cu dose-dependent manner. Overall, these results suggested that excess Cu could trigger mitochondrial dynamics disorder and mitophagy in the pig cerebrum, which provided a novel insight into Cu-induced toxicology.
Collapse
Affiliation(s)
- Xinrun Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China
| | - Yuman Bai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China
| | - Haihua Huo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China
| | - Haitong Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China
| | - Qingyue Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China
| | - Jiaqiang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China.
| | - Jianying Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, People's Republic of China.
| |
Collapse
|
12
|
Liu LL, van Rijn RM, Zheng W. Copper Modulates Adult Neurogenesis in Brain Subventricular Zone. Int J Mol Sci 2022; 23:ijms23179888. [PMID: 36077284 PMCID: PMC9456150 DOI: 10.3390/ijms23179888] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/18/2022] Open
Abstract
The subventricular zone (SVZ) in lateral ventricles is the largest neurogenic region in adult brain containing high amounts of copper (Cu). This study aims to define the role of Cu in adult neurogenesis by chelating labile Cu ions using a well-established Cu chelator D-Penicillamine (D-Pen). A neurosphere model derived from adult mouse SVZ tissues was established and characterized for its functionality with regards to neural stem/progenitor cells (NSPCs). Applying D-Pen in cultured neurospheres significantly reduced intracellular Cu levels and reversed the Cu-induced suppression of NSPC’s differentiation and migration. An in vivo intracerebroventricular (ICV) infusion model was subsequently established to infuse D-Pen directly into the lateral ventricle. Metal analyses revealed a selective reduction of Cu in SVZ by 13.1% (p = 0.19) and 21.4% (p < 0.05) following D-Pen infusions at low (0.075 μg/h) and high (0.75 μg/h) doses for 28 days, respectively, compared to saline-infused controls. Immunohistochemical studies revealed that the 7-day, low-dose D-Pen infusion significantly increased Ki67(+)/Nestin(+) cell counts in SVZ by 28% (p < 0.05). Quantification of BrdU(+)/doublecortin (DCX)(+) newborn neuroblasts in the rostral migration stream (RMS) and olfactory bulb (OB) further revealed that the short-term, low-dose D-Pen infusion, as compared with saline-infused controls, resulted in more newborn neuroblasts in OB, while the high-dose D-Pen infusion showed fewer newborn neuroblasts in OB but with more arrested in the RMS. Long-term (28-day) infusion revealed similar outcomes. The qPCR data from neurosphere experiments revealed altered expressions of mRNAs encoding key proteins known to regulate SVZ adult neurogenesis, including, but not limited to, Shh, Dlx2, and Slit1, in response to the changed Cu level in neurospheres. Further immunohistochemical data indicated that Cu chelation also altered the expression of high-affinity copper uptake protein 1 (CTR1) and metallothionein-3 (MT3) in the SVZ as well as CTR1 in the choroid plexus, a tissue regulating brain Cu homeostasis. Taken together, this study provides first-hand evidence that a high Cu level in SVZ appears likely to maintain the stability of adult neurogenesis in this neurogenic zone.
Collapse
Affiliation(s)
- Luke L. Liu
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Richard M. van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, West Lafayette, IN 47907, USA
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
- Correspondence:
| |
Collapse
|
13
|
Pushie MJ, Sylvain NJ, Hou H, Hackett MJ, Kelly ME, Webb SM. X-ray fluorescence microscopy methods for biological tissues. Metallomics 2022; 14:mfac032. [PMID: 35512669 PMCID: PMC9226457 DOI: 10.1093/mtomcs/mfac032] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 05/05/2022] [Indexed: 11/14/2022]
Abstract
Synchrotron-based X-ray fluorescence microscopy is a flexible tool for identifying the distribution of trace elements in biological specimens across a broad range of sample sizes. The technique is not particularly limited by sample type and can be performed on ancient fossils, fixed or fresh tissue specimens, and in some cases even live tissue and live cells can be studied. The technique can also be expanded to provide chemical specificity to elemental maps, either at individual points of interest in a map or across a large field of view. While virtually any sample type can be characterized with X-ray fluorescence microscopy, common biological sample preparation methods (often borrowed from other fields, such as histology) can lead to unforeseen pitfalls, resulting in altered element distributions and concentrations. A general overview of sample preparation and data-acquisition methods for X-ray fluorescence microscopy is presented, along with outlining the general approach for applying this technique to a new field of investigation for prospective new users. Considerations for improving data acquisition and quality are reviewed as well as the effects of sample preparation, with a particular focus on soft tissues. The effects of common sample pretreatment steps as well as the underlying factors that govern which, and to what extent, specific elements are likely to be altered are reviewed along with common artifacts observed in X-ray fluorescence microscopy data.
Collapse
Affiliation(s)
- M Jake Pushie
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Nicole J Sylvain
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
- Clinical Trial Support Unit, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 0W8 Canada
| | - Huishu Hou
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Mark J Hackett
- Curtin Health Innovation Research Institute, Curtin University, Perth, Western Austrailia 6102, Australia
- School of Molecular and Life Sciences, Curtin University, Perth, Western Austrailia 6845, Australia
| | - Michael E Kelly
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5 Canada
| | - Samuel M Webb
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, CA 94025, USA
| |
Collapse
|
14
|
Mehlenbacher MR, Elsiesy R, Lakha R, Villones RLE, Orman M, Vizcarra CL, Meloni G, Wilcox DE, Austin RN. Metal binding and interdomain thermodynamics of mammalian metallothionein-3: enthalpically favoured Cu + supplants entropically favoured Zn 2+ to form Cu 4 + clusters under physiological conditions. Chem Sci 2022; 13:5289-5304. [PMID: 35655557 PMCID: PMC9093145 DOI: 10.1039/d2sc00676f] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/01/2022] [Indexed: 01/02/2023] Open
Abstract
Metallothioneins (MTs) are a ubiquitous class of small metal-binding proteins involved in metal homeostasis and detoxification. While known for their high affinity for d10 metal ions, there is a surprising dearth of thermodynamic data on metals binding to MTs. In this study, Zn2+ and Cu+ binding to mammalian metallothionein-3 (MT-3) were quantified at pH 7.4 by isothermal titration calorimetry (ITC). Zn2+ binding was measured by chelation titrations of Zn7MT-3, while Cu+ binding was measured by Zn2+ displacement from Zn7MT-3 with competition from glutathione (GSH). Titrations in multiple buffers enabled a detailed analysis that yielded condition-independent values for the association constant (K) and the change in enthalpy (ΔH) and entropy (ΔS) for these metal ions binding to MT-3. Zn2+ was also chelated from the individual α and β domains of MT-3 to quantify the thermodynamics of inter-domain interactions in metal binding. Comparative titrations of Zn7MT-2 with Cu+ revealed that both MT isoforms have similar Cu+ affinities and binding thermodynamics, indicating that ΔH and ΔS are determined primarily by the conserved Cys residues. Inductively coupled plasma mass spectrometry (ICP-MS) analysis and low temperature luminescence measurements of Cu-replete samples showed that both proteins form two Cu4 +-thiolate clusters when Cu+ displaces Zn2+ under physiological conditions. Comparison of the Zn2+ and Cu+ binding thermodynamics reveal that enthalpically-favoured Cu+, which forms Cu4 +-thiolate clusters, displaces the entropically-favoured Zn2+. These results provide a detailed thermodynamic analysis of d10 metal binding to these thiolate-rich proteins and quantitative support for, as well as molecular insight into, the role that MT-3 plays in the neuronal chemistry of copper.
Collapse
Affiliation(s)
| | - Rahma Elsiesy
- Department of Chemistry, Barnard College of Columbia University New York NY 10027 USA
| | - Rabina Lakha
- Department of Chemistry, Barnard College of Columbia University New York NY 10027 USA
| | - Rhiza Lyne E Villones
- Department of Chemistry and Biochemistry, University of Texas at Dallas Richardson TX 75080 USA
| | - Marina Orman
- Department of Chemistry, Barnard College of Columbia University New York NY 10027 USA
| | - Christina L Vizcarra
- Department of Chemistry, Barnard College of Columbia University New York NY 10027 USA
| | - Gabriele Meloni
- Department of Chemistry and Biochemistry, University of Texas at Dallas Richardson TX 75080 USA
| | - Dean E Wilcox
- Department of Chemistry, Dartmouth College Hanover NH 03755 USA
| | - Rachel N Austin
- Department of Chemistry, Barnard College of Columbia University New York NY 10027 USA
| |
Collapse
|
15
|
Tao L, Kong Y, Xiang Y, Cao Y, Ye X, Liu Z. Implantable optical fiber microelectrode with anti-biofouling ability for in vivo photoelectrochemical analysis. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.04.079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
16
|
Ellison G, Hollings AL, Hackett MJ. A review of the “metallome” within neurons and glia, as revealed by elemental mapping of brain tissue. BBA ADVANCES 2022; 2:100038. [PMID: 37082604 PMCID: PMC10074908 DOI: 10.1016/j.bbadva.2021.100038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 01/01/2023] Open
Abstract
It is now well established that transition metals, such as Iron (Fe), Copper (Cu), and Zinc (Zn) are necessary for healthy brain function. Although Fe, Cu, and Zn are essential to the brain, imbalances in the amount, distribution, or chemical form ("metallome") of these metals is linked to the pathology of numerous brain diseases or disorders. Despite the known importance of metal ions for both brain health and disease, the metallome that exists within specific types of brain cells is yet to be fully characterised. The aim of this mini-review is to present an overview of the current knowledge of the metallome found within specific brain cells (oligodendrocytes, astrocytes, microglia, and neurons), as revealed by direct elemental mapping techniques. It is hoped this review will foster continued research using direct elemental mapping techniques to fully characterise the brain cell metallome.
Collapse
Affiliation(s)
- Gaewyn Ellison
- School of Molecular and Life Sciences, Curtin University, Perth, WA 6845, Australia
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Ashley L. Hollings
- School of Molecular and Life Sciences, Curtin University, Perth, WA 6845, Australia
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Mark J. Hackett
- School of Molecular and Life Sciences, Curtin University, Perth, WA 6845, Australia
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
- Corresponding author.
| |
Collapse
|
17
|
Anorexia Nervosa-What Has Changed in the State of Knowledge about Nutritional Rehabilitation for Patients over the Past 10 Years? A Review of Literature. Nutrients 2021; 13:nu13113819. [PMID: 34836075 PMCID: PMC8619053 DOI: 10.3390/nu13113819] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/19/2021] [Accepted: 10/24/2021] [Indexed: 11/16/2022] Open
Abstract
Anorexia nervosa (AN) is a psycho-metabolic disorder with a high risk of somatic complications such as refeeding syndrome (RFS) and carries the highest mortality rate of all psychiatric illnesses. To date, the consensus on the care for patients with AN has been based on recommendations for a combination of alimentation and psychotherapy. It is important to establish an initial caloric intake that will provide weight gain and minimize the risk of complications in the treatment of undernourished patients. Research over the past few years suggests that current treatment recommendations may be too stringent and should be updated. The aim of this paper is to systematize the current reports on nutritional rehabilitation in AN, to present the results of studies on the safe supplementation of patients and its potential impact on improving prognosis and the healing process. This review of literature, from 2011-2021, describes the changing trend in the nutritional protocols used and the research on their efficacy, safety, and long-term effects. In addition, it presents previous reports on the potential benefits of introducing vitamin, pro-and prebiotic and fatty acid supplementation.
Collapse
|
18
|
Adamson SXF, Zheng W, Agim ZS, Du S, Fleming S, Shannahan J, Cannon J. Systemic Copper Disorders Influence the Olfactory Function in Adult Rats: Roles of Altered Adult Neurogenesis and Neurochemical Imbalance. Biomolecules 2021; 11:1315. [PMID: 34572528 PMCID: PMC8471899 DOI: 10.3390/biom11091315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/18/2022] Open
Abstract
Disrupted systemic copper (Cu) homeostasis underlies neurodegenerative diseases with early symptoms including olfactory dysfunction. This study investigated the impact of Cu dyshomeostasis on olfactory function, adult neurogenesis, and neurochemical balance. Models of Cu deficiency (CuD) and Cu overload (CuO) were established by feeding adult rats with Cu-restricted diets plus ip. injection of a Cu chelator (ammonium tetrathiomolybdate) and excess Cu, respectively. CuD reduced Cu levels in the olfactory bulb (OB), subventricular zone (SVZ), rostral migratory stream (RMS), and striatum, while CuO increased Cu levels in these areas. The buried pellet test revealed both CuD and CuO prolonged the latency to uncover food. CuD increased neural proliferation and stem cells in the SVZ and newly differentiated neurons in the OB, whereas CuO caused opposite alterations, suggesting a "switch"-type function of Cu in regulating adult neurogenesis. CuO increased GABA in the OB, while both CuD and CuO reduced DOPAC, HVA, 5-HT and the DA turnover rate in olfactory-associated brain regions. Altered mRNA expression of Cu transport and storage proteins in tested brain areas were observed under both conditions. Together, results support an association between systemic Cu dyshomeostasis and olfactory dysfunction. Specifically, altered adult neurogenesis along the SVZ-RMS-OB pathway and neurochemical imbalance could be the factors that may contribute to olfactory dysfunction.
Collapse
Affiliation(s)
- Sherleen Xue-Fu Adamson
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (S.X.-F.A.); (Z.S.A.); (S.D.); (J.S.)
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (S.X.-F.A.); (Z.S.A.); (S.D.); (J.S.)
- Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, IN 47907, USA
| | - Zeynep Sena Agim
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (S.X.-F.A.); (Z.S.A.); (S.D.); (J.S.)
| | - Sarah Du
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (S.X.-F.A.); (Z.S.A.); (S.D.); (J.S.)
| | - Sheila Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA;
| | - Jonathan Shannahan
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (S.X.-F.A.); (Z.S.A.); (S.D.); (J.S.)
| | - Jason Cannon
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (S.X.-F.A.); (Z.S.A.); (S.D.); (J.S.)
- Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
19
|
Hackett MJ, Hollings AL, Lam V, Takechi R, Mamo JCL, de Jonge MD, Paterson D, Okuyama S. [Mapping the Metallo-maze to Memory Loss: Does Neuronal Metal Ion Deficiency Contribute to Dementia?]. YAKUGAKU ZASSHI 2021; 141:835-842. [PMID: 34078791 DOI: 10.1248/yakushi.20-00251-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Dementia has no cure and is an international health crisis. In addition to the immeasurable loss of QOL caused by dementia, the global economic cost is predicted to reach $2 trillion (USD) by 2030. Although much remains unknown about the biochemical pathways driving cognitive decline and memory loss during dementia, metals have been implicated in neurodegenerative disease. For example, total levels of Fe and Cu increase, which has been proposed to drive oxidative stress; and Fe, Cu, and Zn can bind amyloid-β, catalysing aggregation and formation of amyloid plaques. Unfortunately, despite these known facets through which metal ions may induce pathology, studies in greater detail have been hampered by a lack of microscopy methods to directly visualise metal ions, and their chemical form, within brain cells. Herein we report the use of synchrotron X-ray fluorescence microscopy to simultaneously image Fe, Cu, and Zn within neurons in ex vivo brain tissue sections. Using animal models of dementia, we now demonstrate for the first time that despite global increases in brain metal content and metal ion accumulation within amyloid plaques, key brain regions may also become metal ion deficient. Such deficiency could contribute to cognitive decline because of the essential roles metal ions play in neurotransmitter synthesis and energy metabolism. These recent findings are discussed in the context of memory loss, and the impact that metal ion dis-homeostasis may have on diagnostic and therapeutic development.
Collapse
Affiliation(s)
- Mark J Hackett
- School of Molecular and Life Sciences, Curtin University.,Curtin Health Innovation Research Institute, Curtin University.,Curtin Institute of Functional Molecules and Interfaces, Curtin University
| | - Ashley L Hollings
- School of Molecular and Life Sciences, Curtin University.,Curtin Health Innovation Research Institute, Curtin University.,Curtin Institute of Functional Molecules and Interfaces, Curtin University
| | - Virginie Lam
- Curtin Health Innovation Research Institute, Curtin University
| | - Ryusuke Takechi
- Curtin Health Innovation Research Institute, Curtin University
| | - John C L Mamo
- Curtin Health Innovation Research Institute, Curtin University
| | | | | | - Satoshi Okuyama
- Department of Pharmaceutical Pharmacology, College of Pharmaceutical Sciences, Matsuyama University
| |
Collapse
|
20
|
Witt B, Stiboller M, Raschke S, Friese S, Ebert F, Schwerdtle T. Characterizing effects of excess copper levels in a human astrocytic cell line with focus on oxidative stress markers. J Trace Elem Med Biol 2021; 65:126711. [PMID: 33486291 DOI: 10.1016/j.jtemb.2021.126711] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/02/2020] [Accepted: 01/02/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Being an essential trace element, copper is involved in diverse physiological processes. However, excess levels might lead to adverse effects. Disrupted copper homeostasis, particularly in the brain, has been associated with human diseases including the neurodegenerative disorders Wilson and Alzheimer's disease. In this context, astrocytes play an important role in the regulation of the copper homeostasis in the brain and likely in the prevention against neuronal toxicity, consequently pointing them out as a potential target for the neurotoxicity of copper. Major toxic mechanisms are discussed to be directed against mitochondria probably via oxidative stress. However, the toxic potential and mode of action of copper in astrocytes is poorly understood, so far. METHODS In this study, excess copper levels affecting human astrocytic cell model and their involvement in the neurotoxic mode of action of copper, as well as, effects on the homeostasis of other trace elements (Mn, Fe, Ca and Mg) were investigated. RESULTS Copper induced substantial cytotoxic effects in the human astrocytic cell line following 48 h incubation (EC30: 250 μM) and affected mitochondrial function, as observed via reduction of mitochondrial membrane potential and increased ROS production, likely originating from mitochondria. Moreover, cellular GSH metabolism was altered as well. Interestingly, not only cellular copper levels were affected, but also the homeostasis of other elements (Ca, Fe and Mn) were disrupted. CONCLUSION One potential toxic mode of action of copper seems to be effects on the mitochondria along with induction of oxidative stress in the human astrocytic cell model. Moreover, excess copper levels seem to interact with the homeostasis of other essential elements such as Ca, Fe and Mn. Disrupted element homeostasis might also contribute to the induction of oxidative stress, likely involved in the onset and progression of neurodegenerative disorders. These insights in the toxic mechanisms will help to develop ideas and approaches for therapeutic strategies against copper-mediated diseases.
Collapse
Affiliation(s)
- Barbara Witt
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.
| | - Michael Stiboller
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Stefanie Raschke
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Sharleen Friese
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Franziska Ebert
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Tanja Schwerdtle
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena, Germany; German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589, Berlin, Germany
| |
Collapse
|
21
|
Liu Y, Kang T, He Q, Hu Y, Zuo Z, Cao Z, Ke B, Zhang W, Qi Q. A selective and sensitive near-infrared fluorescent probe for real-time detection of Cu(i). RSC Adv 2021; 11:14824-14828. [PMID: 35423960 PMCID: PMC8697812 DOI: 10.1039/d1ra00725d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/26/2021] [Indexed: 02/05/2023] Open
Abstract
The disruption of copper homeostasis (Cu+/Cu2+) may cause neurodegenerative disorders. Thus, the need for understanding the role of Cu+ in physiological and pathological processes prompted the development of improved methods of Cu+ analysis. Herein, a new near-infrared (NIR) fluorescent turn-on probe (NPCu) for the detection of Cu+ was developed based on a Cu+-mediated benzylic ether bond cleavage mechanism. The probe showed high selectivity and sensitivity toward Cu+, and was successfully applied for bioimaging of Cu+ in living cells.
Collapse
Affiliation(s)
- Yiqing Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug, SichuanResearch Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University Chengdu 610041 P. R. China
| | - Ting Kang
- Department of Anaesthesiology, West China Hospital, Sichuan University China
| | - Qian He
- Department of Emergency, West China Hospital, Sichuan University Chengdu 610000 Sichuan China
| | - Yuefu Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug, SichuanResearch Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University Chengdu 610041 P. R. China
| | - Zeping Zuo
- Department of Anaesthesiology, West China Hospital, Sichuan University China
| | - Zhihua Cao
- Department of Anaesthesiology, West China Hospital, Sichuan University China
| | - Bowen Ke
- Department of Anaesthesiology, West China Hospital, Sichuan University China
| | - Weiyi Zhang
- Department of Anaesthesiology, West China Hospital, Sichuan University China
| | - Qingrong Qi
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug, SichuanResearch Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University Chengdu 610041 P. R. China
| |
Collapse
|
22
|
Chen Z, Tsytsarev V, Finfrock YZ, Antipova OA, Cai Z, Arakawa H, Lischka FW, Hooks BM, Wilton R, Wang D, Liu Y, Gaitan B, Tao Y, Chen Y, Erzurumlu RS, Yang H, Rozhkova EA. Wireless Optogenetic Modulation of Cortical Neurons Enabled by Radioluminescent Nanoparticles. ACS NANO 2021; 15:5201-5208. [PMID: 33625219 DOI: 10.1021/acsnano.0c10436] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
While offering high-precision control of neural circuits, optogenetics is hampered by the necessity to implant fiber-optic waveguides in order to deliver photons to genetically engineered light-gated neurons in the brain. Unlike laser light, X-rays freely pass biological barriers. Here we show that radioluminescent Gd2(WO4)3:Eu nanoparticles, which absorb external X-rays energy and then downconvert it into optical photons with wavelengths of ∼610 nm, can be used for the transcranial stimulation of cortical neurons expressing red-shifted, ∼590-630 nm, channelrhodopsin ReaChR, thereby promoting optogenetic neural control to the practical implementation of minimally invasive wireless deep brain stimulation.
Collapse
Affiliation(s)
- Zhaowei Chen
- Center for Nanoscale Materials, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
- Institute of Food Safety and Environment Monitoring, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Vassiliy Tsytsarev
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Y Zou Finfrock
- Advanced Photon Source, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
- Science Division, Canadian Light Source Inc., 44 Innovation Boulevard, Saskatoon, Saskatchewan S7N 2 V3, Canada
| | - Olga A Antipova
- Advanced Photon Source, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
| | - Zhonghou Cai
- Advanced Photon Source, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
| | - Hiroyuki Arakawa
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Fritz W Lischka
- Biomedical Instrumentation Center, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, Maryland 20814-4799, United States
| | - Bryan M Hooks
- Department of Neurobiology, University of Pittsburgh, 3500 Terrace Street, Suite W1458, Pittsburgh, Pennsylvania 15213-2500, United States
| | - Rosemarie Wilton
- Biosciences, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
| | - Dongyi Wang
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Yi Liu
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Brandon Gaitan
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Yang Tao
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Yu Chen
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, Maryland 20742, United States
| | - Reha S Erzurumlu
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Huanghao Yang
- Institute of Food Safety and Environment Monitoring, MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Elena A Rozhkova
- Center for Nanoscale Materials, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, Illinois 60439, United States
| |
Collapse
|
23
|
Hollings AL, Lam V, Takechi R, Mamo JCL, Reinhardt J, de Jonge MD, Kappen P, Hackett MJ. Revealing differences in the chemical form of zinc in brain tissue using K-edge X-ray absorption near-edge structure spectroscopy. Metallomics 2020; 12:2134-2144. [PMID: 33300524 DOI: 10.1039/d0mt00198h] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Zinc is a prominent trace metal required for normal memory function. Memory loss and cognitive decline during natural ageing and neurodegenerative disease have been associated with altered brain-Zn homeostasis. Yet, the exact chemical pathways through which Zn influences memory function during health, natural ageing, or neurodegenerative disease remain unknown. The gap in the literature may in part be due to the difficulty to simultaneously image, and therefore, study the different chemical forms of Zn within the brain (or biological samples in general). To this extent, we have begun developing and optimising protocols that incorporate X-ray absorption near-edge structure (XANES) spectroscopic analysis of tissue at the Zn K-edge as an analytical tool to study Zn speciation in the brain. XANES is ideally suited for this task as all chemical forms of Zn are detected, the technique requires minimal sample preparation that may otherwise redistribute or alter the chemical form of Zn, and the Zn K-edge has known sensitivity to coordination geometry and ligand type. Herein, we report our initial results where we fit K-edge spectra collected from micro-dissected flash-frozen brain tissue, to a spectral library prepared from standard solutions, to demonstrate differences in the chemical form of Zn that exist between two brain regions, the hippocampus and cerebellum. Lastly, we have used an X-ray microprobe to demonstrate differences in Zn speciation within sub-regions of thin air-dried sections of the murine hippocampus; but, the corresponding results highlight that the chemical form of Zn is easily perturbed by sample preparation such as tissue sectioning or air-drying, which must be a critical consideration for future work.
Collapse
Affiliation(s)
- Ashley L Hollings
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
de Jesus JR, Arruda MAZ. Unravelling neurological disorders through metallomics-based approaches. Metallomics 2020; 12:1878-1896. [PMID: 33237082 DOI: 10.1039/d0mt00234h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Understanding the biological process involving metals and biomolecules in the brain is essential for establishing the origin of neurological disorders, such as neurodegenerative and psychiatric diseases. From this perspective, this critical review presents recent advances in this topic, showing possible mechanisms involving the disruption of metal homeostasis and the pathogenesis of neurological disorders. We also discuss the main challenges observed in metallomics studies associated with neurological disorders, including those related to sample preparation and analyte quantification.
Collapse
|
25
|
Hackett MJ, Hollings A, Caine S, Bewer BE, Alaverdashvili M, Takechi R, Mamo JCL, Jones MWM, de Jonge MD, Paterson PG, Pickering IJ, George GN. Elemental characterisation of the pyramidal neuron layer within the rat and mouse hippocampus. Metallomics 2020; 11:151-165. [PMID: 30398510 DOI: 10.1039/c8mt00230d] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A unique combination of sensitivity, resolution, and penetration make X-ray fluorescence imaging (XFI) ideally suited to investigate trace elemental distributions in the biological context. XFI has gained widespread use as an analytical technique in the biological sciences, and in particular enables exciting new avenues of research in the field of neuroscience. In this study, elemental mapping by XFI was applied to characterise the elemental content within neuronal cell layers of hippocampal sub-regions of mice and rats. Although classical histochemical methods for metal detection exist, such approaches are typically limited to qualitative analysis. Specifically, histochemical methods are not uniformly sensitive to all chemical forms of a metal, often displaying variable sensitivity to specific "pools" or chemical forms of a metal. In addition, histochemical methods require fixation and extensive chemical treatment of samples, creating the strong likelihood for metal redistribution, leaching, or contamination. Direct quantitative elemental mapping of total elemental pools, in situ within ex vivo tissue sections, without the need for chemical fixation or addition of staining reagents is not possible with traditional histochemical methods; however, such a capability, which is provided by XFI, can offer an enormous analytical advantage. The results we report herein demonstrate the analytical advantage of XFI elemental mapping for direct, label-free metal quantification, in situ within ex vivo brain tissue sections. Specifically, we definitively characterise for the first time, the abundance of Fe within the pyramidal cell layers of the hippocampus. Localisation of Fe to this cell layer is not reproducibly achieved with classical Perls histochemical Fe stains. The ability of XFI to directly quantify neuronal elemental (P, S, Cl, K, Ca, Fe, Cu, Zn) distributions, revealed unique profiles of Fe and Zn within anatomical sub-regions of the hippocampus i.e., cornu ammonis 1, 2 or 3 (CA1, CA2 or CA3) sub-regions. Interestingly, our study reveals a unique Fe gradient across neuron populations within the non-degenerating and pathology free rat hippocampus, which curiously mirrors the pattern of region-specific vulnerability of the hippocampus that has previously been established to occur in various neurodegenerative diseases.
Collapse
Affiliation(s)
- M J Hackett
- Curtin Institute for Functional Molecules and Interfaces, School of Molecular and Life Sciences, Curtin University, GPOBox U1987, Bentley, WA 6845, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Witt B, Schaumlöffel D, Schwerdtle T. Subcellular Localization of Copper-Cellular Bioimaging with Focus on Neurological Disorders. Int J Mol Sci 2020; 21:ijms21072341. [PMID: 32231018 PMCID: PMC7178132 DOI: 10.3390/ijms21072341] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/17/2022] Open
Abstract
As an essential trace element, copper plays a pivotal role in physiological body functions. In fact, dysregulated copper homeostasis has been clearly linked to neurological disorders including Wilson and Alzheimer’s disease. Such neurodegenerative diseases are associated with progressive loss of neurons and thus impaired brain functions. However, the underlying mechanisms are not fully understood. Characterization of the element species and their subcellular localization is of great importance to uncover cellular mechanisms. Recent research activities focus on the question of how copper contributes to the pathological findings. Cellular bioimaging of copper is an essential key to accomplish this objective. Besides information on the spatial distribution and chemical properties of copper, other essential trace elements can be localized in parallel. Highly sensitive and high spatial resolution techniques such as LA-ICP-MS, TEM-EDS, S-XRF and NanoSIMS are required for elemental mapping on subcellular level. This review summarizes state-of-the-art techniques in the field of bioimaging. Their strengths and limitations will be discussed with particular focus on potential applications for the elucidation of copper-related diseases. Based on such investigations, further information on cellular processes and mechanisms can be derived under physiological and pathological conditions. Bioimaging studies might enable the clarification of the role of copper in the context of neurodegenerative diseases and provide an important basis to develop therapeutic strategies for reduction or even prevention of copper-related disorders and their pathological consequences.
Collapse
Affiliation(s)
- Barbara Witt
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114–116, 14558 Nuthetal, Germany;
- Correspondence: ; Tel.: +49-3320-088-5241
| | - Dirk Schaumlöffel
- Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux (IPREM), UMR 5254, CNRS/Université de Pau et des Pays de l’Adour/E2S UPPA, 64000 Pau, France;
| | - Tanja Schwerdtle
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114–116, 14558 Nuthetal, Germany;
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Potsdam-Berlin-Jena, Germany
| |
Collapse
|
27
|
Leary SC, Ralle M. Advances in visualization of copper in mammalian systems using X-ray fluorescence microscopy. Curr Opin Chem Biol 2020; 55:19-25. [PMID: 31911338 DOI: 10.1016/j.cbpa.2019.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/23/2022]
Abstract
Synchrotron-based X-ray fluorescence microscopy (XFM) has become an important imaging technique to investigate elemental concentrations and distributions in biological specimens. Advances in technology now permit imaging at resolutions rivaling that of electron microscopy, and researchers can now visualize elemental concentrations in subcellular organelles when using appropriate correlative methods. XFM is an especially valuable tool to determine the distribution of endogenous trace metals that are involved in neurodegenerative diseases. Here, we discuss the latest research on the unusual copper (Cu) storage vesicles that were originally identified in mouse brains and the involvement of Cu in Alzheimer's disease. Finally, we provide an outlook of how future improvements to XFM will drive current trace element research forward.
Collapse
Affiliation(s)
- Scot C Leary
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, S7N 5E5, Canada
| | - Martina Ralle
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, United States.
| |
Collapse
|
28
|
Copper and the brain noradrenergic system. J Biol Inorg Chem 2019; 24:1179-1188. [PMID: 31691104 DOI: 10.1007/s00775-019-01737-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/21/2019] [Indexed: 02/08/2023]
Abstract
Copper (Cu) plays an essential role in the development and function of the brain. In humans, genetic disorders of Cu metabolism may cause either severe Cu deficiency (Menkes disease) or excessive Cu accumulation (Wilson disease) in the brain tissue. In either case, the loss of Cu homeostasis results in catecholamine misbalance, abnormal myelination of neurons, loss of normal brain architecture, and a spectrum of neurologic and/or psychiatric manifestations. Several metabolic processes have been identified as particularly sensitive to Cu dis-homeostasis. This review focuses on the role of Cu in noradrenergic neurons and summarizes the current knowledge of mechanisms that maintain Cu homeostasis in these cells. The impact of Cu misbalance on catecholamine metabolism and functioning of noradrenergic system is discussed.
Collapse
|
29
|
Kardos J, Héja L, Simon Á, Jablonkai I, Kovács R, Jemnitz K. Copper signalling: causes and consequences. Cell Commun Signal 2018; 16:71. [PMID: 30348177 PMCID: PMC6198518 DOI: 10.1186/s12964-018-0277-3] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/24/2018] [Indexed: 12/18/2022] Open
Abstract
Copper-containing enzymes perform fundamental functions by activating dioxygen (O2) and therefore allowing chemical energy-transfer for aerobic metabolism. The copper-dependence of O2 transport, metabolism and production of signalling molecules are supported by molecular systems that regulate and preserve tightly-bound static and weakly-bound dynamic cellular copper pools. Disruption of the reducing intracellular environment, characterized by glutathione shortage and ambient Cu(II) abundance drives oxidative stress and interferes with the bidirectional, copper-dependent communication between neurons and astrocytes, eventually leading to various brain disease forms. A deeper understanding of of the regulatory effects of copper on neuro-glia coupling via polyamine metabolism may reveal novel copper signalling functions and new directions for therapeutic intervention in brain disorders associated with aberrant copper metabolism.
Collapse
Affiliation(s)
- Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - Ágnes Simon
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - István Jablonkai
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - Richard Kovács
- Institute of Neurophysiology, Charité-Universitätsmedizin, Berlin, Germany
| | - Katalin Jemnitz
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| |
Collapse
|
30
|
Porcaro F, Roudeau S, Carmona A, Ortega R. Advances in element speciation analysis of biomedical samples using synchrotron-based techniques. Trends Analyt Chem 2018. [DOI: 10.1016/j.trac.2017.09.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
31
|
Ziomber A, Surowka AD, Antkiewicz-Michaluk L, Romanska I, Wrobel P, Szczerbowska-Boruchowska M. Combined brain Fe, Cu, Zn and neurometabolite analysis - a new methodology for unraveling the efficacy of transcranial direct current stimulation (tDCS) in appetite control. Metallomics 2018; 10:397-405. [PMID: 29384550 DOI: 10.1039/c7mt00329c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Obesity is a chronic, multifactorial origin disease that has recently become one of the most frequent lifestyle disorders. Unfortunately, current obesity treatments seem to be ineffective. At present, transcranial direct current brain stimulation (tDCS) represents a promising novel treatment methodology that seems to be efficient, well-tolerated and safe for a patient. Unfortunately, the biochemical action of tDCS remains unknown, which prevents its widespread use in the clinical arena, although neurobiochemical changes in brain signaling and metal metabolism are frequently reported. Therefore, our research aimed at exploring the biochemical response to tDCS in situ, in the brain areas triggering feeding behavior in obese animals. The objective was to propose a novel neurochemical (serotoninergic and dopaminergic signaling) and trace metal analysis of Fe, Cu and Zn. In doing so, we used energy-dispersive X-ray fluorescence (EDXRF) and high-performance liquid chromatography (HPLC). Anodal-type stimulation (atDCS) of the right frontal cortex was utilized to down-regulate food intake and body weight gain in obese rats. EDXRF was coupled with the external standard method in order to quantify the chemical elements within appetite-triggering brain areas. Major dopamine metabolites were assessed in the brains, based on the HPLC assay utilizing the external standard assay. Our study confirms that elemental analysis by EDXRF and brain metabolite assay by HPLC can be considered as a useful tool for the in situ investigation of the interplay between neurochemical and Fe/Cu/Zn metabolism in the brain upon atDCS. With this methodology, an increase in both Cu and Zn in the satiety center of the stimulated group could be reported. In turn, the most significant neurochemical changes involved dopaminergic and serotoninergic signaling in the brain reward system.
Collapse
Affiliation(s)
- Agata Ziomber
- Jagiellonian University, Chair of Pathophysiology, Faculty of Medicine, Krakow, Poland
| | - Artur Dawid Surowka
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, al. A. Mickiewicza 30, 30-059 Krakow, Poland.
| | - Lucyna Antkiewicz-Michaluk
- Department of Neurochemistry, Institute of Pharmacology Polish Academy of Sciences, ul. Smetna 12, 31-343 Kraków, Poland
| | - Irena Romanska
- Department of Neurochemistry, Institute of Pharmacology Polish Academy of Sciences, ul. Smetna 12, 31-343 Kraków, Poland
| | - Pawel Wrobel
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, al. A. Mickiewicza 30, 30-059 Krakow, Poland.
| | - Magdalena Szczerbowska-Boruchowska
- AGH University of Science and Technology, Faculty of Physics and Applied Computer Science, al. A. Mickiewicza 30, 30-059 Krakow, Poland.
| |
Collapse
|