1
|
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 2024:112744. [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] [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
|
2
|
Gui W, Wang WX. Copper redox state in cells and aquatic organisms: Implication for toxicity. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135039. [PMID: 38941830 DOI: 10.1016/j.jhazmat.2024.135039] [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: 04/21/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
Copper (Cu) redox state has been an important issue in biology and toxicology research, but many research gaps remain to be explored due to the limitations in the detecting techniques. Herein, the regulation of Cu homeostasis, including absorption, translocation, utilization, storage, and elimination behavior is discussed. Cuproptosis, a newly identified type of cell death caused by excessive Cu accumulation, which results in the aggregation of DLAT protein or the loss of Fe-S cluster and finally proteotoxic stress, is reviewed. Several longstanding mysteries of diseases such as Wilson disease and toxic effects, may be attributed to cuproptosis. Furthermore, we review the advanced detection methods and application of Cu(I) and Cu(II), especially the in-situ imaging techniques such as XANES, and chemosensors. Most of the existing studies using these detection techniques focus on the bioaccumulation and toxicity of Cu(I) and Cu(II) in cells and aquatic organisms. Finally, it will be important to identify the roles of Cu(I) and Cu(II) in the growth, development, and diseases of organisms, as well as the relationship between bioaccumulation and toxicity of Cu(I) and Cu(II) in cellular and aquatic toxicology.
Collapse
Affiliation(s)
- Wanying Gui
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| | - Wen-Xiong Wang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China.
| |
Collapse
|
3
|
Suryana E, Rowlands BD, Bishop DP, Finkelstein DI, Double KL. Empirically derived formulae for calculation of age- and region-related levels of iron, copper and zinc in the adult C57BL/6 mouse brain. Neurobiol Aging 2024; 136:34-43. [PMID: 38301453 DOI: 10.1016/j.neurobiolaging.2024.01.003] [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: 05/18/2023] [Revised: 12/05/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024]
Abstract
Metal dyshomeostasis is associated with neurodegenerative disorders, cancers and vascular disease. We report the effects of age (range: 3 to 18 months) on regional copper, iron and zinc levels in the brain of the C57BL/6 mouse, a widely used inbred strain with a permissive background allowing maximal expression of mutations in models that recapitulate these disorders. We present formulae that can be used to determine regional brain metal concentrations in the C57BL/6 mouse at any age in the range of three to eighteen months of life. Copper levels in the C57BL/6 mouse adult brain were highest in the striatum and cerebellum and increased with age, excepting the cortex and hippocampus. Regional iron levels increased linearly with age in all brain regions, while regional zinc concentrations became more homogeneous with age. Knockdown of the copper transporter Ctr1 reduced brain copper, but not iron or zinc, concentrations in a regionally-dependent manner. These findings demonstrate biometals in the brain change with age in a regionally-dependent manner. These data and associated formulae have implications for improving design and interpretation of a wide variety of studies in the C57BL/6 mouse.
Collapse
Affiliation(s)
- E Suryana
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - B D Rowlands
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - D P Bishop
- School of Mathematical and Physical Sciences, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - D I Finkelstein
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - K L Double
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia.
| |
Collapse
|
4
|
Bhoite R, Han Y, Chaitanya AK, Varshney RK, Sharma DL. Genomic approaches to enhance adaptive plasticity to cope with soil constraints amidst climate change in wheat. THE PLANT GENOME 2024; 17:e20358. [PMID: 37265088 DOI: 10.1002/tpg2.20358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/09/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023]
Abstract
Climate change is varying the availability of resources, soil physicochemical properties, and rainfall events, which collectively determines soil physical and chemical properties. Soil constraints-acidity (pH < 6), salinity (pH ≤ 8.5), sodicity, and dispersion (pH > 8.5)-are major causes of wheat yield loss in arid and semiarid cropping systems. To cope with changing environments, plants employ adaptive strategies such as phenotypic plasticity, a key multifaceted trait, to promote shifts in phenotypes. Adaptive strategies for constrained soils are complex, determined by key functional traits and genotype × environment × management interactions. The understanding of the molecular basis of stress tolerance is particularly challenging for plasticity traits. Advances in sequencing and high-throughput genomics technologies have identified functional alleles in gene-rich regions, haplotypes, candidate genes, mechanisms, and in silico gene expression profiles at various growth developmental stages. Our review focuses on favorable alleles for enhanced gene expression, quantitative trait loci, and epigenetic regulation of plant responses to soil constraints, including heavy metal stress and nutrient limitations. A strategy is then described for quantitative traits in wheat by investigating significant alleles and functional characterization of variants, followed by gene validation using advanced genomic tools, and marker development for molecular breeding and genome editing. Moreover, the review highlights the progress of gene editing in wheat, multiplex gene editing, and novel alleles for smart control of gene expression. Application of these advanced genomic technologies to enhance plasticity traits along with soil management practices will be an effective tool to build yield, stability, and sustainability on constrained soils in the face of climate change.
Collapse
Affiliation(s)
- Roopali Bhoite
- Department of Primary Industries and Regional Development, South Perth, Western Australia, Australia
- The UWA Institute of Agriculture, The University of Western Australia, Perth, Western Australia, Australia
| | - Yong Han
- Department of Primary Industries and Regional Development, South Perth, Western Australia, Australia
- Centre for Crop & Food Innovation, State Agricultural Biotechnology Centre, Murdoch University, Perth, Western Australia, Australia
| | - Alamuru Krishna Chaitanya
- Grains Genetics Portfolio, University of Southern Queensland, Centre for Crop Health, Toowoomba, Queensland, Australia
| | - Rajeev K Varshney
- Centre for Crop & Food Innovation, State Agricultural Biotechnology Centre, Murdoch University, Perth, Western Australia, Australia
| | - Darshan Lal Sharma
- Department of Primary Industries and Regional Development, South Perth, Western Australia, Australia
- Centre for Crop & Food Innovation, State Agricultural Biotechnology Centre, Murdoch University, Perth, Western Australia, Australia
| |
Collapse
|
5
|
Schmued L, Maloney B, Schmued C, Lahiri DK. Treatment with 1, 10 Phenanthroline-5-Amine Reduced Amyloid Burden in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2024; 97:239-247. [PMID: 38073385 PMCID: PMC10789349 DOI: 10.3233/jad-221285] [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] [Accepted: 10/16/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent age-related dementia, and, despite numerous attempts to halt or reverse its devastating progression, no effective therapeutics have yet been confirmed clinically. However, one class of agents that has shown promise is certain metal chelators. OBJECTIVE For the novel assessment of the effect of oral administration of 1,10-phenanthroline-5-amine (PAA) on the severity of amyloid plaque load, we used a transgenic (Tg) mouse model with inserted human autosomally dominant (familial) AD genes: amyloid-β protein precursor (AβPP) and tau. METHODS AβPP/Tau transgenic mice that model AD were allotted into one of two groups. The control group received no treatment while the experimental group received PAA in their drinking water starting at 4 months of age. All animals were sacrificed at 1 year of age and their brains were stained with two different markers of amyloid plaques, Amylo-Glo+ and HQ-O. RESULTS The control animals exhibited numerous dense core plaques throughout the neo- and allo- cortical brain regions. The experimental group treated with PAA, however, showed 62% of the amyloid plaque burden seen in the control group. CONCLUSIONS Oral daily dosing with PAA will significantly reduce the amyloid plaque burden in transgenic mice that model AD. The underlying mechanism for this protection is not fully known; however, one proposed mechanism involves inhibiting the "metal-seeding" of Aβ.
Collapse
Affiliation(s)
| | - Bryan Maloney
- Department of Psychiatry, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Debomoy K. Lahiri
- Department of Psychiatry, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Departments of Medical & Molecular Genetics, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
6
|
Chourrout M, Sandt C, Weitkamp T, Dučić T, Meyronet D, Baron T, Klohs J, Rama N, Boutin H, Singh S, Olivier C, Wiart M, Brun E, Bohic S, Chauveau F. Virtual histology of Alzheimer's disease: Biometal entrapment within amyloid-β plaques allows for detection via X-ray phase-contrast imaging. Acta Biomater 2023; 170:260-272. [PMID: 37574159 DOI: 10.1016/j.actbio.2023.07.046] [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: 03/30/2023] [Revised: 07/21/2023] [Accepted: 07/25/2023] [Indexed: 08/15/2023]
Abstract
Amyloid-β (Aβ) plaques from Alzheimer's Disease (AD) can be visualized ex vivo in label-free brain samples using synchrotron X-ray phase-contrast tomography (XPCT). However, for XPCT to be useful as a screening method for amyloid pathology, it is essential to understand which factors drive the detection of Aβ plaques. The current study was designed to test the hypothesis that Aβ-related contrast in XPCT could be caused by Aβ fibrils and/or by metals trapped in the plaques. Fibrillar and elemental compositions of Aβ plaques were probed in brain samples from different types of AD patients and AD models to establish a relationship between XPCT contrast and Aβ plaque characteristics. XPCT, micro-Fourier-Transform Infrared spectroscopy and micro-X-Ray Fluorescence spectroscopy were conducted on human samples (one genetic and one sporadic case) and on four transgenic rodent strains (mouse: APPPS1, ArcAβ, J20; rat: TgF344). Aβ plaques from the genetic AD patient were visible using XPCT, and had higher β-sheet content and higher metal levels than those from the sporadic AD patient, which remained undetected by XPCT. Aβ plaques in J20 mice and TgF344 rats appeared hyperdense on XPCT images, while they were hypodense with a hyperdense core in the case of APPPS1 and ArcAβ mice. In all four transgenic strains, β-sheet content was similar, while metal levels were highly variable: J20 (zinc and iron) and TgF344 (copper) strains showed greater metal accumulation than APPPS1 and ArcAβ mice. Hence, a hyperdense contrast formation of Aβ plaques in XPCT images was associated with biometal entrapment within plaques. STATEMENT OF SIGNIFICANCE: The role of metals in Alzheimer's disease (AD) has been a subject of continuous interest. It was already known that amyloid-β plaques (Aβ), the earliest hallmark of AD, tend to trap endogenous biometals like zinc, iron and copper. Here we show that this metal accumulation is the main reason why Aβ plaques are detected with a new technique called X-ray phase contrast tomography (XPCT). XPCT enables to map the distribution of Aβ plaques in the whole excised brain without labeling. In this work we describe a unique collection of four transgenic models of AD, together with a human sporadic and a rare genetic case of AD, thus exploring the full spectrum of amyloid contrast in XPCT.
Collapse
Affiliation(s)
- Matthieu Chourrout
- Univ. Lyon, Lyon Neuroscience Research Center (CRNL); CNRS UMR5292; INSERM U1028, Univ. Lyon 1, Lyon, France
| | | | | | - Tanja Dučić
- ALBA-CELLS Synchrotron, MIRAS Beamline, Cerdanyola del Vallès, Spain
| | - David Meyronet
- Hospices Civils de Lyon, Neuropathology Department, Lyon, France; Univ. Lyon, Cancer Research Center of Lyon (CRCL); INSERM U1052; CNRS UMR5286, Univ. Lyon 1; Centre Léon Bérard, Lyon, France
| | | | - Jan Klohs
- ETH Zurich, Institute for Biomedical Engineering, Zurich, Switzerland
| | - Nicolas Rama
- Univ. Lyon, Cancer Research Center of Lyon (CRCL); INSERM U1052; CNRS UMR5286, Univ. Lyon 1; Centre Léon Bérard, Lyon, France
| | - Hervé Boutin
- Univ. Manchester, Faculty of Biology Medicine and Health, Wolfson Molecular Imaging Centre, Manchester, United Kingdom
| | - Shifali Singh
- Univ. Grenoble Alpes, Synchrotron Radiation for Biomedicine (STROBE); Inserm UA7, Grenoble, France
| | - Cécile Olivier
- Univ. Grenoble Alpes, Synchrotron Radiation for Biomedicine (STROBE); Inserm UA7, Grenoble, France
| | - Marlène Wiart
- Univ. Lyon, CarMeN Laboratory; INSERM U1060, INRA U1397, INSA Lyon, Univ. Lyon 1, Lyon, France; CNRS, France
| | - Emmanuel Brun
- Univ. Grenoble Alpes, Synchrotron Radiation for Biomedicine (STROBE); Inserm UA7, Grenoble, France
| | - Sylvain Bohic
- Univ. Grenoble Alpes, Synchrotron Radiation for Biomedicine (STROBE); Inserm UA7, Grenoble, France
| | - Fabien Chauveau
- Univ. Lyon, Lyon Neuroscience Research Center (CRNL); CNRS UMR5292; INSERM U1028, Univ. Lyon 1, Lyon, France; CNRS, France.
| |
Collapse
|
7
|
Hosseinpour Mashkani SM, Bishop DP, Raoufi-Rad N, Adlard PA, Shimoni O, Golzan SM. Distribution of Copper, Iron, and Zinc in the Retina, Hippocampus, and Cortex of the Transgenic APP/PS1 Mouse Model of Alzheimer's Disease. Cells 2023; 12:cells12081144. [PMID: 37190053 DOI: 10.3390/cells12081144] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 05/17/2023] Open
Abstract
A mis-metabolism of transition metals (i.e., copper, iron, and zinc) in the brain has been recognised as a precursor event for aggregation of Amyloid-β plaques, a pathological hallmark of Alzheimer's disease (AD). However, imaging cerebral transition metals in vivo can be extremely challenging. As the retina is a known accessible extension of the central nervous system, we examined whether changes in the hippocampus and cortex metal load are also mirrored in the retina. Laser ablation inductively coupled plasma-mass spectrometry (LA-ICP-MS) was used to visualise and quantify the anatomical distribution and load of Cu, Fe, and Zn in the hippocampus, cortex, and retina of 9-month-old Amyloid Precursor Protein/Presenilin 1 (APP/PS1, n = 10) and Wild Type (WT, n = 10) mice. Our results show a similar metal load trend between the retina and the brain, with the WT mice displaying significantly higher concentrations of Cu, Fe, and Zn in the hippocampus (p < 0.05, p < 0.0001, p < 0.01), cortex (p < 0.05, p = 0.18, p < 0.0001) and the retina (p < 0.001, p = 0.01, p < 0.01) compared with the APP/PS1 mice. Our findings demonstrate that dysfunction of the cerebral transition metals in AD is also extended to the retina. This could lay the groundwork for future studies on the assessment of transition metal load in the retina in the context of early AD.
Collapse
Affiliation(s)
- Seyed Mostafa Hosseinpour Mashkani
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| | - David P Bishop
- Hyphenated Mass Spectrometry Laboratory (HyMaS), School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| | - Newsha Raoufi-Rad
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| | - Paul A Adlard
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Olga Shimoni
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| | - S Mojtaba Golzan
- Vision Science Group, Graduate School of Health (GSH), University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| |
Collapse
|
8
|
Foley PB, Hare DJ, Double KL. A brief history of brain iron accumulation in Parkinson disease and related disorders. J Neural Transm (Vienna) 2022; 129:505-520. [PMID: 35534717 PMCID: PMC9188502 DOI: 10.1007/s00702-022-02505-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 04/22/2022] [Indexed: 12/21/2022]
Abstract
Iron has a long and storied history in Parkinson disease and related disorders. This essential micronutrient is critical for normal brain function, but abnormal brain iron accumulation has been associated with extrapyramidal disease for a century. Precisely why, how, and when iron is implicated in neuronal death remains the subject of investigation. In this article, we review the history of iron in movement disorders, from the first observations in the early twentieth century to recent efforts that view extrapyramidal iron as a novel therapeutic target and diagnostic indicator.
Collapse
Affiliation(s)
| | - Dominic J. Hare
- Atomic Medicine Initiative, University of Technology, Sydney, Australia
| | - Kay L. Double
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| |
Collapse
|
9
|
Foley PB, Hare DJ, Double KL. A brief history of brain iron accumulation in Parkinson disease and related disorders. J Neural Transm (Vienna) 2022; 129:505-520. [PMID: 35534717 DOI: 10.1007/s00702-022-025055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 04/22/2022] [Indexed: 05/26/2023]
Abstract
Iron has a long and storied history in Parkinson disease and related disorders. This essential micronutrient is critical for normal brain function, but abnormal brain iron accumulation has been associated with extrapyramidal disease for a century. Precisely why, how, and when iron is implicated in neuronal death remains the subject of investigation. In this article, we review the history of iron in movement disorders, from the first observations in the early twentieth century to recent efforts that view extrapyramidal iron as a novel therapeutic target and diagnostic indicator.
Collapse
Affiliation(s)
| | - Dominic J Hare
- Atomic Medicine Initiative, University of Technology, Sydney, Australia
| | - Kay L Double
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, University of Sydney, Sydney, Australia.
| |
Collapse
|
10
|
Zhao C, Cai Z. Three-dimensional quantitative mass spectrometry imaging in complex system: From subcellular to whole organism. MASS SPECTROMETRY REVIEWS 2022; 41:469-487. [PMID: 33300181 DOI: 10.1002/mas.21674] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 10/13/2020] [Accepted: 10/22/2020] [Indexed: 06/12/2023]
Abstract
Mass spectrometry imaging (MSI) has been applied for label-free three-dimensional (3D) imaging from position array across the whole organism, which provides high-dimensional quantitative data of inorganic or organic compounds that may play an important role in the regulation of cellular signaling, including metals, metabolites, lipids, drugs, peptides, and proteins. While MSI is suitable for investigation of the spatial distribution of molecules, it has a limitation with visualization and quantification of multiple molecules. 3D-MSI, however, can be applied toward exploring metabolic pathway as well as the interactions of lipid-protein, protein-protein, and metal-protein in complex systems from subcellular to the whole organism through an untargeted methodology. In this review, we highlight the methods and applications of MS-based 3D imaging to address the complexity of molecular interaction from nano- to micrometer lateral resolution, with particular focus on: (a) common and hybrid 3D-MSI techniques; (b) quantitative MSI methodology, including the methods using a stable isotope labeling internal standard (SILIS) and SILIS-free approaches with tissue extinction coefficient or virtual calibration; (c) reconstruction of the 3D organ; (d) application of 3D-MSI for biomarker screening and environmental toxicological research. 3D-MSI quantitative analysis provides accurate spatial information and quantitative variation of biomolecules, which may be valuable for the exploration of the molecular mechanism of the disease progresses and toxicological assessment of environmental pollutants in the whole organism. Additionally, we also discuss the challenges and perspectives on the future of 3D quantitative MSI.
Collapse
Affiliation(s)
- Chao Zhao
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
11
|
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: 2.0] [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
|
12
|
Folarin OR, Olopade FE, Olopade JO. Essential Metals in the Brain and the Application of Laser Ablation-Inductively Coupled Plasma-Mass Spectrometry for their Detection. Niger J Physiol Sci 2021; 36:123-147. [PMID: 35947740 DOI: 10.54548/njps.v36i2.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 06/15/2023]
Abstract
Metals are natural component of the ecosystem present throughout the layers of atmosphere; their abundant expression in the brain indicates their importance in the central nervous system (CNS). Within the brain tissue, their distribution is highly compartmentalized, the pattern of which is determined by their primary roles. Bio-imaging of the brain to reveal spatial distribution of metals within specific regions has provided a unique understanding of brain biochemistry and architecture, linking both the structures and the functions through several metal mediated activities. Bioavailability of essential trace metal is needed for normal brain function. However, disrupted metal homeostasis can influence several biochemical pathways in different fields of metabolism and cause characteristic neurological disorders with a typical disease process usually linked with aberrant metal accumulations. In this review we give a brief overview of roles of key essential metals (Iron, Copper and Zinc) including their molecular mechanisms and bio-distribution in the brain as well as their possible involvement in the pathogenesis of related neurodegenerative diseases. In addition, we also reviewed recent applications of Laser Ablation Inductively Couple Plasma Mass Spectrophotometry (LA-ICP-MS) in the detection of both toxic and essential metal dyshomeostasis in neuroscience research and other related brain diseases.
Collapse
|
13
|
Yuan Z, Zhou Q, Cai L, Pan L, Sun W, Qumu S, Yu S, Feng J, Zhao H, Zheng Y, Shi M, Li S, Chen Y, Zhang X, Zhang MQ. SEAM is a spatial single nuclear metabolomics method for dissecting tissue microenvironment. Nat Methods 2021; 18:1223-1232. [PMID: 34608315 DOI: 10.1038/s41592-021-01276-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 08/18/2021] [Indexed: 02/08/2023]
Abstract
Spatial metabolomics can reveal intercellular heterogeneity and tissue organization. Here we report on the spatial single nuclear metabolomics (SEAM) method, a flexible platform combining high-spatial-resolution imaging mass spectrometry and a set of computational algorithms that can display multiscale and multicolor tissue tomography together with identification and clustering of single nuclei by their in situ metabolic fingerprints. We first applied SEAM to a range of wild-type mouse tissues, then delineated a consistent pattern of metabolic zonation in mouse liver. We further studied the spatial metabolic profile in the human fibrotic liver. We discovered subpopulations of hepatocytes with special metabolic features associated with their proximity to the fibrotic niche, and validated this finding by spatial transcriptomics with Geo-seq. These demonstrations highlighted SEAM's ability to explore the spatial metabolic profile and tissue histology at the single-cell level, leading to a deeper understanding of tissue metabolic organization.
Collapse
Affiliation(s)
- Zhiyuan Yuan
- MOE Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist, Institute of TCM-X, Department of Automation, Tsinghua University, Beijing, China
| | - Qiming Zhou
- MOE Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist, School of Life Sciences, Tsinghua University, Beijing, China
| | - Lesi Cai
- Department of Chemistry, Tsinghua University, Beijing, China
| | - Lin Pan
- Institute of Clinical Medicine, China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Weiliang Sun
- Institute of Clinical Medicine, China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Science, Beijing, China
| | - Shiwei Qumu
- Department of Pulmonary and Critical Care Medicine, China-Japan Friend Hospital, National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Si Yu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaxin Feng
- Department of Chemistry, Tsinghua University, Beijing, China
| | - Hansen Zhao
- Department of Chemistry, Tsinghua University, Beijing, China
| | - Yongchang Zheng
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minglei Shi
- MOE Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Shao Li
- MOE Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist, Institute of TCM-X, Department of Automation, Tsinghua University, Beijing, China
| | - Yang Chen
- MOE Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist, Institute of TCM-X, Department of Automation, Tsinghua University, Beijing, China. .,The State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.
| | - Xinrong Zhang
- Department of Chemistry, Tsinghua University, Beijing, China.
| | - Michael Q Zhang
- MOE Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist, Institute of TCM-X, Department of Automation, Tsinghua University, Beijing, China. .,MOE Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China. .,Department of Biological Sciences, Center for Systems Biology, The University of Texas, Richardson, TX, USA.
| |
Collapse
|
14
|
Doble PA, de Vega RG, Bishop DP, Hare DJ, Clases D. Laser Ablation-Inductively Coupled Plasma-Mass Spectrometry Imaging in Biology. Chem Rev 2021; 121:11769-11822. [PMID: 34019411 DOI: 10.1021/acs.chemrev.0c01219] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Elemental imaging gives insight into the fundamental chemical makeup of living organisms. Every cell on Earth is comprised of a complex and dynamic mixture of the chemical elements that define structure and function. Many disease states feature a disturbance in elemental homeostasis, and understanding how, and most importantly where, has driven the development of laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) as the principal elemental imaging technique for biologists. This review provides an outline of ICP-MS technology, laser ablation cell designs, imaging workflows, and methods of quantification. Detailed examples of imaging applications including analyses of cancers, elemental uptake and accumulation, plant bioimaging, nanomaterials in the environment, and exposure science and neuroscience are presented and discussed. Recent incorporation of immunohistochemical workflows for imaging biomolecules, complementary and multimodal imaging techniques, and image processing methods is also reviewed.
Collapse
Affiliation(s)
- Philip A Doble
- Atomic Medicine Initiative, University of Technology Sydney, Broadway, New South Wales 2007, Australia
| | - Raquel Gonzalez de Vega
- Atomic Medicine Initiative, University of Technology Sydney, Broadway, New South Wales 2007, Australia
| | - David P Bishop
- Atomic Medicine Initiative, University of Technology Sydney, Broadway, New South Wales 2007, Australia
| | - Dominic J Hare
- Atomic Medicine Initiative, University of Technology Sydney, Broadway, New South Wales 2007, Australia.,School of BioSciences, University of Melbourne, Parkville, Victoria 3052, Australia
| | - David Clases
- Atomic Medicine Initiative, University of Technology Sydney, Broadway, New South Wales 2007, Australia
| |
Collapse
|
15
|
Dales JP, Desplat-Jégo S. Metal Imbalance in Neurodegenerative Diseases with a Specific Concern to the Brain of Multiple Sclerosis Patients. Int J Mol Sci 2020; 21:E9105. [PMID: 33266021 PMCID: PMC7730295 DOI: 10.3390/ijms21239105] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/29/2020] [Accepted: 11/19/2020] [Indexed: 12/16/2022] Open
Abstract
There is increasing evidence that deregulation of metals contributes to a vast range of neurodegenerative diseases including multiple sclerosis (MS). MS is a chronic inflammatory disease of the central nervous system (CNS) manifesting disability and neurological symptoms. The precise origin of MS is unknown, but the disease is characterized by focal inflammatory lesions in the CNS associated with an autoimmune reaction against myelin. The treatment of this disease has mainly been based on the prescription of immunosuppressive and immune-modulating agents. However, the rate of progressive disability and early mortality is still worrisome. Metals may represent new diagnostic and predictive markers of severity and disability as well as innovative candidate drug targets for future therapies. In this review, we describe the recent advances in our understanding on the role of metals in brain disorders of neurodegenerative diseases and MS patients.
Collapse
Affiliation(s)
- Jean-Philippe Dales
- Institute of Neurophysiopathology, CNRS, INP, Aix-Marseille University, 13005 Marseille, France;
- Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Pavillon Etoile, Pôle de Biologie, Service d’anatomie-pathologie, CEDEX 20, 13915 Marseille, France
| | - Sophie Desplat-Jégo
- Institute of Neurophysiopathology, CNRS, INP, Aix-Marseille University, 13005 Marseille, France;
- Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, Pôle de Biologie, Service d’Immunologie, 13005 Marseille, France
| |
Collapse
|
16
|
Pushie M, Hollings A, Reinhardt J, Webb S, Lam V, Takechi R, Mamo J, Paterson P, Kelly M, George G, Pickering I, Hackett M. Sample preparation with sucrose cryoprotection dramatically alters Zn distribution in the rodent hippocampus, as revealed by elemental mapping. JOURNAL OF ANALYTICAL ATOMIC SPECTROMETRY 2020; 35:2498-2508. [PMID: 33795908 PMCID: PMC8009441 DOI: 10.1039/d0ja00323a] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Transition metal ions (Fe, Mn, Cu, Zn) are essential for healthy brain function, but altered concentration, distribution, or chemical form of the metal ions has been implicated in numerous brain pathologies. Currently, it is not possible to image the cellular or sub-cellular distribution of metal ions in vivo and therefore, studying brain-metal homeostasis largely relies on ex vivo in situ elemental mapping. Sample preparation methods that accurately preserve the in vivo elemental distribution are essential if one wishes to translate the knowledge of elemental distributions measured ex vivo toward increased understanding of chemical and physiological pathways of brain disease. The choice of sample preparation is particularly important for metal ions that exist in a labile or mobile form, for which the in vivo distribution could be easily distorted by inappropriate sample preparation. One of the most widely studied brain structures, the hippocampus, contains a large pool of labile and mobile Zn. Herein, we describe how sucrose cryoprotection, the gold standard method of preparing tissues for immuno-histochemistry or immuno-fluorescence, which is also often used as a sample preparation method for elemental mapping studies, drastically alters hippocampal Zn distribution. Based on the results of this study, in combination with a comparison against the strong body of published literature that has used either rapid plunge freezing of brain tissue, or sucrose cryo-protection, we strongly urge investigators in the future to cease using sucrose cryoprotection as a method of sample preparation for elemental mapping, especially if Zn is an analyte of interest.
Collapse
Affiliation(s)
- M.J. Pushie
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - A. Hollings
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, AUS
- School of Molecular and Life Sciences, Curtin University, Perth, WA 6845, AUS
| | - J. Reinhardt
- Australian Nuclear Science and Technology Organisation, 800 Blackburn Road, Clayton, VIC, AUS 3168
| | - S.M. Webb
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, California, USA 94025
| | - V. Lam
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, AUS
- School of Public Health, Faculty of Health Sciences, Curtin University, WA, Australia
| | - R Takechi
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, AUS
- School of Public Health, Faculty of Health Sciences, Curtin University, WA, Australia
| | - J.C. Mamo
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, AUS
- School of Public Health, Faculty of Health Sciences, Curtin University, WA, Australia
| | - P.G. Paterson
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, Saskatchewan, S7N 5E5, Canada
| | - M.E. Kelly
- Department of Surgery, Division of Neurosurgery, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - G.N. George
- School Molecular and Environmental Sciences Group, Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - I.J. Pickering
- School Molecular and Environmental Sciences Group, Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - M.J. Hackett
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, AUS
- School of Molecular and Life Sciences, Curtin University, Perth, WA 6845, AUS
| |
Collapse
|
17
|
Perry WJ, Weiss A, Van de Plas R, Spraggins JM, Caprioli RM, Skaar EP. Integrated molecular imaging technologies for investigation of metals in biological systems: A brief review. Curr Opin Chem Biol 2020; 55:127-135. [PMID: 32087551 PMCID: PMC7237308 DOI: 10.1016/j.cbpa.2020.01.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/25/2019] [Accepted: 01/14/2020] [Indexed: 02/08/2023]
Abstract
Metals play an essential role in biological systems and are required as structural or catalytic co-factors in many proteins. Disruption of the homeostatic control and/or spatial distributions of metals can lead to disease. Imaging technologies have been developed to visualize elemental distributions across a biological sample. Measurement of elemental distributions by imaging mass spectrometry and imaging X-ray fluorescence are increasingly employed with technologies that can assess histological features and molecular compositions. Data from several modalities can be interrogated as multimodal images to correlate morphological, elemental, and molecular properties. Elemental and molecular distributions have also been axially resolved to achieve three-dimensional volumes, dramatically increasing the biological information. In this review, we provide an overview of recent developments in the field of metal imaging with an emphasis on multimodal studies in two and three dimensions. We specifically highlight studies that present technological advancements and biological applications of how metal homeostasis affects human health.
Collapse
Affiliation(s)
- William J Perry
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN, 37232, USA; Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Andy Weiss
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Raf Van de Plas
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, 37232, USA; Delft Center for Systems and Control, Delft University of Technology, Delft, Netherlands; Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - Jeffrey M Spraggins
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN, 37232, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - Richard M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, TN, 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN, 37232, USA; Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37232, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA; Department of Medicine, Vanderbilt University, Nashville, TN, 37232, USA
| | - Eric P Skaar
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
18
|
Rao SS, Lago L, Gonzalez de Vega R, Bray L, Hare DJ, Clases D, Doble PA, Adlard PA. Characterising the spatial and temporal brain metal profile in a mouse model of tauopathy. Metallomics 2020; 12:301-313. [PMID: 31904058 DOI: 10.1039/c9mt00267g] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A dysregulation in the homeostasis of metals such as copper, iron and zinc is speculated to be involved in the pathogenesis of tauopathies, which includes Alzheimer's disease (AD). In particular, there is a growing body of evidence to support a role for iron in facilitating the hyperphosphorylation and aggregation of the tau protein into neurofibrillary tangles (NFTs) - a primary neuropathological hallmark of tauopathies. Therefore, the aim of this study was to characterize the spatial and temporal brain metallomic profile in a mouse model of tauopathy (rTg(tauP301L)4510), so as to provide some insight into the potential interaction between tau pathology and iron. Using laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS), our results revealed an age-dependent increase in brain iron levels in both WT and rTg(tauP301L)4510 mice. In addition, size exclusion chromatography-ICP-MS (SEC-ICP-MS) revealed significant age-related changes in iron bound to metalloproteins such as ferritin. The outcomes from this study may provide valuable insight into the inter-relationship between iron and tau in ageing and neurodegeneration.
Collapse
Affiliation(s)
- Shalini S Rao
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, The Melbourne Dementia Research Centre, Parkville, Victoria, Australia.
| | - Larissa Lago
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, The Melbourne Dementia Research Centre, Parkville, Victoria, Australia.
| | | | - Lisa Bray
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, The Melbourne Dementia Research Centre, Parkville, Victoria, Australia.
| | - Dominic J Hare
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, The Melbourne Dementia Research Centre, Parkville, Victoria, Australia.
| | - David Clases
- The Atomic Medicine Initiative, University of Technology Sydney, Sydney, NSW, Australia
| | - Philip A Doble
- The Atomic Medicine Initiative, University of Technology Sydney, Sydney, NSW, Australia
| | - Paul A Adlard
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, The Melbourne Dementia Research Centre, Parkville, Victoria, Australia.
| |
Collapse
|
19
|
Peacey L, Elphick MR, Jones CE. Roles of copper in neurokinin B and gonadotropin-releasing hormone structure and function and the endocrinology of reproduction. Gen Comp Endocrinol 2020; 287:113342. [PMID: 31783025 DOI: 10.1016/j.ygcen.2019.113342] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/24/2019] [Accepted: 11/25/2019] [Indexed: 11/30/2022]
Abstract
Copper is a metal ion present in all organisms, where it has well-known roles in association with proteins and enzymes essential for cellular processes. In the early decades of the twentieth century copper was shown to influence mammalian reproductive biology, and it was subsequently shown to exert effects primarily at the level of the pituitary gland and/or hypothalamic regions of the brain. Furthermore, it has been reported that copper can interact with key neuropeptides in the hypothalamic-pituitary-gonadal axis, notably gonadotropin-releasing hormone (GnRH) and neurokinin B. Interestingly, recent phylogenetic analysis of the sequences of GnRH-related peptides indicates that copper binding is an evolutionarily ancient property of this neuropeptide family, which has been variously retained, modified or lost in the different taxa. In this mini-review the metal-binding properties of neuropeptides in the vertebrate reproductive pathway are reviewed and the evolutionary and functional significance of copper binding by GnRH-related neuropeptides in vertebrates and invertebrates are discussed.
Collapse
Affiliation(s)
- Lorraine Peacey
- School of Science and Health, The University of Western Sydney, Locked Bag 1797, Penrith, New South Wales, Australia
| | - Maurice R Elphick
- Queen Mary University of London, School of Biological and Chemical Sciences, Mile End Road, London E14NS, UK
| | - Christopher E Jones
- School of Science and Health, The University of Western Sydney, Locked Bag 1797, Penrith, New South Wales, Australia.
| |
Collapse
|
20
|
Westerhausen MT, Bishop DP, Dowd A, Wanagat J, Cole N, Doble PA. Super-Resolution Reconstruction for Two- and Three-Dimensional LA-ICP-MS Bioimaging. Anal Chem 2019; 91:14879-14886. [PMID: 31640341 PMCID: PMC7232986 DOI: 10.1021/acs.analchem.9b02380] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The resolution of laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) elemental bioimaging is usually constrained by the diameter of the laser spot size and is often not adequate to explore in situ subcellular distributions of elements and proteins in biological tissue sections. Super-resolution reconstruction is a method typically used for many imaging modalities and combines multiple lower resolution images to create a higher resolution image. Here, we present a super-resolution reconstruction method for LA-ICP-MS imaging by ablating consecutive layers of a biological specimen with offset orthogonal scans, resulting in a 10× improvement in resolution for quantitative measurement of dystrophin in murine muscle fibers. Layer-by-layer image reconstruction was also extended to the third dimension without the requirement of image registration across multiple thin section specimens. Quantitative super-resolution reconstruction, combined with Gaussian filtering and application of the Richardson-Lucy total variation algorithm, provided superior image clarity and fidelity in two- and three-dimensions.
Collapse
Affiliation(s)
- Mika T. Westerhausen
- The Atomic Medicine Initiative, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway, NSW 2007, Australia
| | - David P. Bishop
- The Atomic Medicine Initiative, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway, NSW 2007, Australia
| | - Annette Dowd
- School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway NSW 2007, Australia
| | - Jonathan Wanagat
- Department of Medicine, Division of Geriatrics, David Geffen School of Medicine, University of California, Los Angeles, California, United States
| | - Nerida Cole
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, John Street, Hawthorn, Victoria 3122, Australia
| | - Philip A. Doble
- The Atomic Medicine Initiative, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway, NSW 2007, Australia
| |
Collapse
|
21
|
Jayawardena BM, Jones MR, Hong Y, Jones CE. Copper ions trigger disassembly of neurokinin B functional amyloid and inhibit de novo assembly. J Struct Biol 2019; 208:107394. [DOI: 10.1016/j.jsb.2019.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/23/2019] [Accepted: 09/23/2019] [Indexed: 01/24/2023]
|
22
|
Samal J, Rebelo AL, Pandit A. A window into the brain: Tools to assess pre-clinical efficacy of biomaterials-based therapies on central nervous system disorders. Adv Drug Deliv Rev 2019; 148:68-145. [PMID: 30710594 DOI: 10.1016/j.addr.2019.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/04/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
Abstract
Therapeutic conveyance into the brain is a cardinal requirement for treatment of diverse central nervous system (CNS) disorders and associated pathophysiology. Effectual shielding of the brain by the blood-brain barrier (BBB) sieves out major proportion of therapeutics with the exception of small lipophilic molecules. Various nano-delivery systems (NDS) provide an effective solution around this obstacle owing to their small size and targeting properties. To date, these systems have been used for several pre-clinical disease models including glioma, neurodegenerative diseases and psychotic disorders. An efficacy screen for these systems involves a test battery designed to probe into the multiple facets of therapeutic delivery. Despite their wide application in redressing various disease targets, the efficacy evaluation strategies for all can be broadly grouped into four modalities, namely: histological, bio-imaging, molecular and behavioural. This review presents a comprehensive insight into all of these modalities along with their strengths and weaknesses as well as perspectives on an ideal design for a panel of tests to screen brain nano-delivery systems.
Collapse
Affiliation(s)
- Juhi Samal
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Ana Lucia Rebelo
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
23
|
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: 17] [Impact Index Per Article: 3.4] [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
|
24
|
Billings JL, Gordon SL, Rawling T, Doble PA, Bush AI, Adlard PA, Finkelstein DI, Hare DJ. l
‐3,4‐dihydroxyphenylalanine (
l
‐DOPA) modulates brain iron, dopaminergic neurodegeneration and motor dysfunction in iron overload and mutant alpha‐synuclein mouse models of Parkinson's disease. J Neurochem 2019; 150:88-106. [DOI: 10.1111/jnc.14676] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/15/2018] [Accepted: 01/16/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Jessica L. Billings
- Melbourne Dementia Research Centre at The Florey Institute of Neuroscience and Mental Health and The University of Melbourne Parkville Victoria Australia
| | - Sarah L. Gordon
- Melbourne Dementia Research Centre at The Florey Institute of Neuroscience and Mental Health and The University of Melbourne Parkville Victoria Australia
| | - Tristan Rawling
- School of Mathematical and Physical Sciences Faculty of Science University of Technology Sydney Broadway New South Wales Australia
| | - Philip A. Doble
- Elemental Bio‐imaging Facility University of Technology Sydney Broadway New South Wales Australia
| | - Ashley I. Bush
- Melbourne Dementia Research Centre at The Florey Institute of Neuroscience and Mental Health and The University of Melbourne Parkville Victoria Australia
| | - Paul A. Adlard
- Melbourne Dementia Research Centre at The Florey Institute of Neuroscience and Mental Health and The University of Melbourne Parkville Victoria Australia
| | - David I. Finkelstein
- Melbourne Dementia Research Centre at The Florey Institute of Neuroscience and Mental Health and The University of Melbourne Parkville Victoria Australia
| | - Dominic J. Hare
- Melbourne Dementia Research Centre at The Florey Institute of Neuroscience and Mental Health and The University of Melbourne Parkville Victoria Australia
- Elemental Bio‐imaging Facility University of Technology Sydney Broadway New South Wales Australia
- Department of Clinical Pathology The University of Melbourne Parkville Victoria Australia
| |
Collapse
|
25
|
Van Malderen SJM, Van Acker T, Laforce B, De Bruyne M, de Rycke R, Asaoka T, Vincze L, Vanhaecke F. Three-dimensional reconstruction of the distribution of elemental tags in single cells using laser ablation ICP-mass spectrometry via registration approaches. Anal Bioanal Chem 2019; 411:4849-4859. [PMID: 30790022 DOI: 10.1007/s00216-019-01677-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 01/22/2019] [Accepted: 02/06/2019] [Indexed: 12/15/2022]
Abstract
This paper describes a workflow towards the reconstruction of the three-dimensional elemental distribution profile within human cervical carcinoma cells (HeLa), at a spatial resolution down to 1 μm, employing state-of-the-art laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) instrumentation. The suspended cells underwent a series of fixation/embedding protocols and were stained with uranyl acetate and an Ir-based DNA intercalator. A priori, laboratory-based absorption micro-computed tomography (μ-CT) was applied to acquire a reference frame of the morphology of the cells and their spatial distribution before sectioning. After CT analysis, a trimmed 300 × 300 × 300 μm3 block was sectioned into a sequential series of 132 sections with a thickness of 2 μm, which were subjected to LA-ICP-MS imaging. A pixel acquisition rate of 250 pixels s-1 was achieved, through a bidirectional scanning strategy. After acquisition, the two-dimensional elemental images were reconstructed using the timestamps in the laser log file. The synchronization of the data required an improved optimization algorithm, which forces the pixels of scans in different ablation directions to be spatially coherent in the direction orthogonal to the scan direction. The volume was reconstructed using multiple registration approaches. Registration using the section outline itself as a fiducial marker resulted into a volume which was in good agreement with the morphology visualized in the μ-CT volume. The 3D μ-CT volume could be registered to the LA-ICP-MS volume, consisting of 2.9 × 107 voxels, and the nucleus dimensions in 3D space could be derived.
Collapse
Affiliation(s)
- Stijn J M Van Malderen
- Atomic & Mass Spectrometry (A&MS) Research Unit, Department of Chemistry, Ghent University, Campus Sterre, Krijgslaan 281 - S12, 9000, Ghent, Belgium
| | - Thibaut Van Acker
- Atomic & Mass Spectrometry (A&MS) Research Unit, Department of Chemistry, Ghent University, Campus Sterre, Krijgslaan 281 - S12, 9000, Ghent, Belgium
| | - Brecht Laforce
- X-ray Microspectroscopy and Imaging (XMI) Research Unit, Department of Chemistry, Ghent University, Campus Sterre, Krijgslaan 281 - S12, 9000, Ghent, Belgium
| | - Michiel De Bruyne
- Department of Biomedical Molecular Biology and VIB Center for Inflammation Research, Ghent University, Technologiepark 71, 9052, Ghent, Belgium
- Ghent University Expertise Centre for Transmission Electron Microscopy and VIB BioImaging Core, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
| | - Riet de Rycke
- Department of Biomedical Molecular Biology and VIB Center for Inflammation Research, Ghent University, Technologiepark 71, 9052, Ghent, Belgium
- Ghent University Expertise Centre for Transmission Electron Microscopy and VIB BioImaging Core, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
| | - Tomoko Asaoka
- Department of Biomedical Molecular Biology and VIB Center for Inflammation Research, Ghent University, Technologiepark 71, 9052, Ghent, Belgium
| | - Laszlo Vincze
- X-ray Microspectroscopy and Imaging (XMI) Research Unit, Department of Chemistry, Ghent University, Campus Sterre, Krijgslaan 281 - S12, 9000, Ghent, Belgium
| | - Frank Vanhaecke
- Atomic & Mass Spectrometry (A&MS) Research Unit, Department of Chemistry, Ghent University, Campus Sterre, Krijgslaan 281 - S12, 9000, Ghent, Belgium.
| |
Collapse
|
26
|
Weiskirchen R, Weiskirchen S, Kim P, Winkler R. Software solutions for evaluation and visualization of laser ablation inductively coupled plasma mass spectrometry imaging (LA-ICP-MSI) data: a short overview. J Cheminform 2019; 11:16. [PMID: 30778692 PMCID: PMC6690067 DOI: 10.1186/s13321-019-0338-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/09/2019] [Indexed: 12/19/2022] Open
Abstract
Mass spectrometry imaging (MSI) using laser ablation (LA) inductively coupled plasma (ICP) is an innovative and exciting methodology to perform highly sensitive elemental analyses. LA-ICP-MSI of metals, trace elements or isotopes in tissues has been applied to a range of biological samples. Several LA-ICP-MSI studies have shown that metals have a highly compartmentalized distribution in some organs, which might be altered in consequence of genetic diseases, intoxication, or malnutrition. Although metal imaging by LA-ICP-MSI is an established methodology, potential pitfalls in the determination of metal concentrations might result from erroneous calibration, standardization, and normalization. In addition, for simple display of final imaging results, most LA-ICP-MSI users prefer to process their measurements by commercial processing software. Such programs typically visualize the regional metal differences in colorful and vivid imaging maps, but might not represent the actual signal densities correctly. There is a great abundance of such MSI data processing programs available differing in quality, usability, integrated features, workflow, reliability, system requirements, speed of data processing, and price. Some software packages contain a multitude of features which are superfluous for most users. In contrast, often only few data formats are used, in case of commercial programs even only the instrument provider’s own raw data format. Therefore, first time and average users are often confused and helpless in choosing the correct software for processing their data. Here we have briefly summarized software packages, data routines, macros, programming tools, scripts, algorithms, or self-written patches and updates for existing programs presently in use for mining LA-ICP-MSI data.![]()
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074, Aachen, Germany.
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074, Aachen, Germany
| | - Philipp Kim
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074, Aachen, Germany
| | - Robert Winkler
- Department of Biochemistry and Biotechnology, Center for Research and Advanced Studies (CINVESTAV) Irapuato, Km. 9.6 Libramiento Norte Carr. Irapuato-León, 36824, Irapuato, Gto., Mexico. .,Mass Spectrometry Group, Max Planck Institute for Chemical Ecology, Hans-Knöll-Straße 8, 07745, Jena, Germany.
| |
Collapse
|
27
|
Laser ablation ICP-MS for simultaneous quantitative imaging of iron and ferroportin in hippocampus of human brain tissues with Alzheimer's disease. Talanta 2019; 197:413-421. [PMID: 30771955 DOI: 10.1016/j.talanta.2019.01.056] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 11/23/2022]
Abstract
Laser ablation inductively coupled plasma - mass spectrometry (LA-ICP-MS) is proposed for a better understanding of metals and proteins distribution in micrometre structures of human brain tissues. Simultaneous absolute quantitative imaging of Fe and ferroportin (FPN), in 5 µm thick tissue sections of the stratum pyramidale of hippocampus CA1 region, was carried out for Alzheimer disease (AD) patients and healthy controls (HC). For the imaging of FPN by LA-ICP-MS, antibodies were labelled via carbodiimide crosslinking with fluorescent gold nanoclusters (AuNCs) of 2.2 nm diameter, enabling a high amplification (314 gold atoms per NC). Laboratory made gelatin standards containing Fe and Au were used for LA-ICP-MS calibration. Results showed that iron presents an increased concentration in AD donors compared with HC donors, whereas similar concentrations of FPN in AD donors with respect to HC donors were obtained. The average absolute FPN concentrations in selected areas obtained with the proposed AuNCs method were compared with the levels obtained by densitometric analysis with a traditional IHC approach, observing a similar trend in all cases.
Collapse
|
28
|
Yamada Y, Prosser RA. Copper in the suprachiasmatic circadian clock: A possible link between multiple circadian oscillators. Eur J Neurosci 2018; 51:47-70. [PMID: 30269387 DOI: 10.1111/ejn.14181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 09/05/2018] [Accepted: 09/17/2018] [Indexed: 01/07/2023]
Abstract
The mammalian circadian clock in the suprachiasmatic nucleus (SCN) is very robust, able to coordinate our daily physiological and behavioral rhythms with exquisite accuracy. Simultaneously, the SCN clock is highly sensitive to environmental timing cues such as the solar cycle. This duality of resiliency and sensitivity may be sustained in part by a complex intertwining of three cellular oscillators: transcription/translation, metabolic/redox, and membrane excitability. We suggest here that one of the links connecting these oscillators may be forged from copper (Cu). Cellular Cu levels are highly regulated in the brain and peripherally, and Cu affects cellular metabolism, redox state, cell signaling, and transcription. We have shown that both Cu chelation and application induce nighttime phase shifts of the SCN clock in vitro and that these treatments affect glutamate, N-methyl-D-aspartate receptor, and associated signaling processes differently. More recently we found that Cu induces mitogen-activated protein kinase-dependent phase shifts, while the mechanisms by which Cu removal induces phase shifts remain unclear. Lastly, we have found that two Cu transporters are expressed in the SCN, and that one of these transporters (ATP7A) exhibits a day/night rhythm. Our results suggest that Cu homeostasis is tightly regulated in the SCN, and that changes in Cu levels may serve as a time cue for the circadian clock. We discuss these findings in light of the existing literature and current models of multiple coupled circadian oscillators in the SCN.
Collapse
Affiliation(s)
- Yukihiro Yamada
- Department of Biochemistry & Cellular and Molecular Biology, NeuroNET Research Center, University of Tennessee, Knoxville, Tennessee
| | - Rebecca A Prosser
- Department of Biochemistry & Cellular and Molecular Biology, NeuroNET Research Center, University of Tennessee, Knoxville, Tennessee
| |
Collapse
|
29
|
Tran VT, Tran NHT, Nguyen TT, Yoon WJ, Ju H. Liquid Cladding Mediated Optical Fiber Sensors for Copper Ion Detection. MICROMACHINES 2018; 9:E471. [PMID: 30424404 PMCID: PMC6187453 DOI: 10.3390/mi9090471] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/10/2018] [Accepted: 09/13/2018] [Indexed: 01/25/2023]
Abstract
We present a label-free optical fiber based sensor device to detect copper ions (Cu2+) in water. A multimode optical fiber, with its polymer cladding removed along a 1-cm length, is used for the optical sensor head, where the injected Cu2+ in the liquid phase acts as a liquid cladding for the optical mode. The various Cu2+ concentrations modulate the numerical aperture (NA) of the liquid cladding waveguide part. The degree of NA mismatch between the liquid cladding and solid cladding guided parts gives rise to an optical power transmittance change, forming the sensing principle. The presented liquid cladding fiber sensor exhibits a minimum resolvable refractive index of 2.48 × 10-6. For Cu2+ detection, we functionalize the sensor head surface (fiber core) using chitosan conjugated ethylenediaminetetraacetic acid (EDTA) which captures Cu2+ effectively due to the enhanced chelating effects. We obtain a limit of detection of Cu2+ of 1.62 nM (104 ppt), which is significantly lower than the tolerable level in drinking water (~30 µM), and achieve a dynamic range of 1 mM. The simple structure of the sensor head and the sensing system ensures the potential capability of being miniaturized. This may allow for in-situ, highly-sensitive, heavy metal sensors in a compact format.
Collapse
Affiliation(s)
- Vien Thi Tran
- Department of Nano-Physics, Gachon University, Seongnam-si 461-701, Korea.
- Gachon Bionano Research Institute, Gachon University, Seongnam-si 461-701, Korea.
| | - Nhu Hoa Thi Tran
- Department of Nano-Physics, Gachon University, Seongnam-si 461-701, Korea.
- Gachon Bionano Research Institute, Gachon University, Seongnam-si 461-701, Korea.
| | - Than Thi Nguyen
- Department of Nano-Physics, Gachon University, Seongnam-si 461-701, Korea.
- Gachon Bionano Research Institute, Gachon University, Seongnam-si 461-701, Korea.
| | - Won Jung Yoon
- Department of Chemical and Bio Engineering, Gachon University, Seongnam-si 461-701, Korea.
| | - Heongkyu Ju
- Department of Nano-Physics, Gachon University, Seongnam-si 461-701, Korea.
- Gachon Bionano Research Institute, Gachon University, Seongnam-si 461-701, Korea.
- Neuroscience Institute, Gil Hospital, Incheon 405-760, Korea.
| |
Collapse
|
30
|
Garza-Lombó C, Posadas Y, Quintanar L, Gonsebatt ME, Franco R. Neurotoxicity Linked to Dysfunctional Metal Ion Homeostasis and Xenobiotic Metal Exposure: Redox Signaling and Oxidative Stress. Antioxid Redox Signal 2018; 28:1669-1703. [PMID: 29402131 PMCID: PMC5962337 DOI: 10.1089/ars.2017.7272] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Essential metals such as copper, iron, manganese, and zinc play a role as cofactors in the activity of a wide range of processes involved in cellular homeostasis and survival, as well as during organ and tissue development. Throughout our life span, humans are also exposed to xenobiotic metals from natural and anthropogenic sources, including aluminum, arsenic, cadmium, lead, and mercury. It is well recognized that alterations in the homeostasis of essential metals and an increased environmental/occupational exposure to xenobiotic metals are linked to several neurological disorders, including neurodegeneration and neurodevelopmental alterations. Recent Advances: The redox activity of essential metals is key for neuronal homeostasis and brain function. Alterations in redox homeostasis and signaling are central to the pathological consequences of dysfunctional metal ion homeostasis and increased exposure to xenobiotic metals. Both redox-active and redox-inactive metals trigger oxidative stress and damage in the central nervous system, and the exact mechanisms involved are starting to become delineated. CRITICAL ISSUES In this review, we aim to appraise the role of essential metals in determining the redox balance in the brain and the mechanisms by which alterations in the homeostasis of essential metals and exposure to xenobiotic metals disturb the cellular redox balance and signaling. We focus on recent literature regarding their transport, metabolism, and mechanisms of toxicity in neural systems. FUTURE DIRECTIONS Delineating the specific mechanisms by which metals alter redox homeostasis is key to understand the pathological processes that convey chronic neuronal dysfunction in neurodegenerative and neurodevelopmental disorders. Antioxid. Redox Signal. 28, 1669-1703.
Collapse
Affiliation(s)
- Carla Garza-Lombó
- 1 Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln , Lincoln, Nebraska.,2 Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas , Universidad Nacional Autónoma de México, Mexico City, México
| | - Yanahi Posadas
- 3 Departamentos de Farmacología y de, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México .,4 Departamentos de Química, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México
| | - Liliana Quintanar
- 4 Departamentos de Química, Centro de Investigación y de Estudios Avanzados (CINVESTAV) , Mexico City, México
| | - María E Gonsebatt
- 2 Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas , Universidad Nacional Autónoma de México, Mexico City, México
| | - Rodrigo Franco
- 1 Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln , Lincoln, Nebraska
| |
Collapse
|
31
|
Portbury SD, Hare DJ, Sgambelloni C, Perronnes K, Portbury AJ, Finkelstein DI, Adlard PA. Trehalose Improves Cognition in the Transgenic Tg2576 Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2018; 60:549-560. [PMID: 28869469 PMCID: PMC5611803 DOI: 10.3233/jad-170322] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study assessed the therapeutic utility of the autophagy enhancing stable disaccharide trehalose in the Tg2576 transgenic mouse model of Alzheimer’s disease (AD) via an oral gavage of a 2% trehalose solution for 31 days. Furthermore, as AD is a neurodegenerative condition in which the transition metals, iron, copper, and zinc, are understood to be intricately involved in the cellular cascades leading to the defining pathologies of the disease, we sought to determine any parallel impact of trehalose treatment on metal levels. Trehalose treatment significantly improved performance in the Morris water maze, consistent with enhanced learning and memory. The improvement was not associated with significant modulation of full length amyloid-β protein precursor or other amyloid-β fragments. Trehalose had no effect on autophagy as assessed by western blot of the LC3-1 to LC3-2 protein ratio, and no alteration in biometals that might account for the improved cognition was observed. Biochemical analysis revealed a significant increase in the hippocampus of both synaptophysin, a synaptic vesicle protein and surrogate marker of synapses, and doublecortin, a reliable marker of neurogenesis. The growth factor progranulin was also significantly increased in the hippocampus and cortex with trehalose treatment. This study suggests that trehalose might invoke a suite of neuroprotective mechanisms that can contribute to improved cognitive performance in AD that are independent of more classical trehalose-mediated pathways, such as Aβ reduction and activation of autophagy. Thus, trehalose may have utility as a potential AD therapeutic, with conceivable implications for the treatment of other neurodegenerative disorders.
Collapse
Affiliation(s)
- Stuart D Portbury
- The Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, The University of Melbourne, Parkville, VIC, Australia
| | - Dominic J Hare
- The Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, The University of Melbourne, Parkville, VIC, Australia.,University of Technology Sydney, Elemental Bio-imaging, Broadway, Australia
| | - Charlotte Sgambelloni
- The Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, The University of Melbourne, Parkville, VIC, Australia
| | - Kali Perronnes
- The Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, The University of Melbourne, Parkville, VIC, Australia
| | - Ashley J Portbury
- The Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, The University of Melbourne, Parkville, VIC, Australia
| | - David I Finkelstein
- The Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, The University of Melbourne, Parkville, VIC, Australia
| | - Paul A Adlard
- The Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
32
|
Helfrich KK, Saini N, Kling PJ, Smith SM. Maternal iron nutriture as a critical modulator of fetal alcohol spectrum disorder risk in alcohol-exposed pregnancies. Biochem Cell Biol 2018; 96:204-212. [PMID: 29017023 PMCID: PMC5914169 DOI: 10.1139/bcb-2017-0206] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Alcohol consumption during pregnancy places the fetus at risk for permanent physical, cognitive, and behavioral impairments, collectively termed fetal alcohol spectrum disorder (FASD). However, prenatal alcohol exposure (PAE) outcomes vary widely, and growing evidence suggests that maternal nutrition is a modifying factor. Certain nutrients, such as iron, may modulate FASD outcomes. Untreated gestational iron deficiency (ID) causes persistent neurodevelopmental deficits in the offspring that affect many of the same domains damaged by PAE. Although chronic alcohol consumption enhances iron uptake and elevates liver iron stores in adult alcoholics, alcohol-abusing premenopausal women often have low iron reserves due to menstruation, childbirth, and poor diet. Recent investigations show that low iron reserves during pregnancy are strongly associated with a worsening of several hallmark features in FASD including reduced growth and impaired associative learning. This review discusses recent clinical and animal model findings that maternal ID worsens fetal outcomes in response to PAE. It also discusses underlying mechanisms by which PAE disrupts maternal and fetal iron homeostasis. We suggest that alcohol-exposed ID pregnancies contribute to the severe end of the FASD spectrum.
Collapse
Affiliation(s)
- Kaylee K Helfrich
- a UNC Nutrition Research Institute and Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nipun Saini
- a UNC Nutrition Research Institute and Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Pamela J Kling
- b Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Susan M Smith
- a UNC Nutrition Research Institute and Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
33
|
Peters DG, Pollack AN, Cheng KC, Sun D, Saido T, Haaf MP, Yang QX, Connor JR, Meadowcroft MD. Dietary lipophilic iron alters amyloidogenesis and microglial morphology in Alzheimer's disease knock-in APP mice. Metallomics 2018; 10:426-443. [PMID: 29424844 DOI: 10.1039/c8mt00004b] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized pathologically by amyloid beta (Aβ) deposition, microgliosis, and iron dyshomeostasis. Increased labile iron due to homeostatic dysregulation is believed to facilitate amyloidogenesis. Free iron is incorporated into aggregating amyloid peptides during Aβ plaque formation and increases potential for oxidative stress surrounding plaques. The goal of this work was to observe how brain iron levels temporally influence Aβ plaque formation, plaque iron concentration, and microgliosis. We fed humanized APPNL-F and APPNL-G-F knock-in mice lipophilic iron compound 3,5,5-trimethylhexanoyl ferrocene (TMHF) and iron deficient diets for twelve months. TMHF elevated brain iron by 22% and iron deficiency decreased brain iron 21% relative to control diet. Increasing brain iron with TMHF accelerated plaque formation, increased Aβ staining, and increased senile morphology of amyloid plaques. Increased brain iron was associated with increased plaque-iron loading and microglial iron inclusions. TMHF decreased IBA1+ microglia branch length while increasing roundness indicative of microglial activation. This body of work suggests that increasing mouse brain iron with TMHF potentiates a more human-like Alzheimer's disease phenotype with iron integration into Aβ plaques and associated microgliosis.
Collapse
Affiliation(s)
- Douglas G Peters
- Department of Neurosurgery, The Pennsylvania State University - College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, USA and Department of Neural and Behavioral Science, The Pennsylvania State University - College of Medicine, Hershey, Pennsylvania, USA
| | - Alexis N Pollack
- Department of Neurosurgery, The Pennsylvania State University - College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, USA
| | - Keith C Cheng
- Department of Pathology (Gittlen Cancer Research Institute), The Pennsylvania State University - College of Medicine, Hershey, Pennsylvania, USA
| | - Dongxiao Sun
- Department of Pharmacology, The Pennsylvania State University - College of Medicine, Hershey, Pennsylvania, USA
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wakō-shi, Saitama-ken, Japan
| | - Michael P Haaf
- Department of Chemistry, Ithaca College, Ithaca, New York, USA
| | - Qing X Yang
- Department of Radiology (Center for NMR Research), The Pennsylvania State University - College of Medicine, Hershey, Pennsylvania, USA
| | - James R Connor
- Department of Neurosurgery, The Pennsylvania State University - College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, USA
| | - Mark D Meadowcroft
- Department of Neurosurgery, The Pennsylvania State University - College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, USA and Department of Radiology (Center for NMR Research), The Pennsylvania State University - College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
34
|
Moraleja I, Mena M, Lázaro A, Neumann B, Tejedor A, Jakubowski N, Gómez-Gómez M, Esteban-Fernández D. An approach for quantification of platinum distribution in tissues by LA-ICP-MS imaging using isotope dilution analysis. Talanta 2018; 178:166-171. [DOI: 10.1016/j.talanta.2017.09.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/06/2017] [Accepted: 09/10/2017] [Indexed: 11/27/2022]
|
35
|
Dressler VL, Müller EI, Pozebon D. Bioimaging Metallomics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1055:139-181. [DOI: 10.1007/978-3-319-90143-5_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
3D Quantitative Chemical Imaging of Tissues by Spectromics. Trends Biotechnol 2017; 35:1194-1207. [DOI: 10.1016/j.tibtech.2017.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 07/31/2017] [Accepted: 08/04/2017] [Indexed: 12/14/2022]
|
37
|
New EJ, Wimmer VC, Hare DJ. Promises and Pitfalls of Metal Imaging in Biology. Cell Chem Biol 2017; 25:7-18. [PMID: 29153850 DOI: 10.1016/j.chembiol.2017.10.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/02/2017] [Accepted: 10/18/2017] [Indexed: 10/18/2022]
Abstract
A picture may speak a thousand words, but if those words fail to form a coherent sentence there is little to be learned. As cutting-edge imaging technology now provides us the tools to decipher the multitude of roles played by metals and metalloids in molecular, cellular, and developmental biology, as well as health and disease, it is time to reflect on the advances made in imaging, the limitations discovered, and the future of a burgeoning field. In this Perspective, the current state of the art is discussed from a self-imposed contrarian position, as we not only highlight the major advances made over the years but use them as teachable moments to zoom in on challenges that remain to be overcome. We also describe the steps being taken toward being able to paint a completely undisturbed picture of cellular metal metabolism, which is, metaphorically speaking, the Holy Grail of the discipline.
Collapse
Affiliation(s)
- Elizabeth J New
- School of Chemistry, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Verena C Wimmer
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Dominic J Hare
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia; Elemental Bio-imaging Facility, University of Technology Sydney, Broadway, NSW 2007, Australia; Department of Pathology, The University of Melbourne, Parkville, VIC 3052, Australia.
| |
Collapse
|
38
|
Andreozzi EM, Torres JB, Sunassee K, Dunn J, Walker-Samuel S, Szanda I, Blower PJ. Studies of copper trafficking in a mouse model of Alzheimer's disease by positron emission tomography: comparison of 64Cu acetate and 64CuGTSM. Metallomics 2017; 9:1622-1633. [PMID: 29063080 PMCID: PMC6205627 DOI: 10.1039/c7mt00227k] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease can involve brain copper dyshomeostasis. We aimed to determine the effect of AD-like pathology on 64Cu trafficking in mice, using positron emission tomography (PET imaging), during 24 hours after intravenous administration of ionic 64Cu (Cu(ii) acetate) and 64Cu-GTSM (GTSMH2 = glyoxalbis(thiosemicarbazone)). Copper trafficking was evaluated in 6-8-month-old and 13-15 month-old TASTPM transgenic and wild-type mice, by imaging 0-30 min and 24-25 h after intravenous administration of 64Cu tracer. Regional 64Cu distribution in brains was compared by ex vivo autoradiography to that of amyloid-β plaque. 64Cu-acetate showed uptake in, and excretion through, liver and kidneys. There was minimal uptake in other tissues by 30 minutes, and little further change after 24 h. Radioactivity within brain was focussed in and around the ventricles and was significantly greater in younger mice. 64CuGTSM was taken up in all tissues by 30 min, remaining high in brain but clearing substantially from other tissues by 24 h. Distribution in brain was not localised to specific regions. TASTPM mice showed no major changes in global or regional 64Cu brain uptake compared to wildtype after administration of 64Cu acetate (unlike 64Cu-GTSM) but efflux of 64Cu from brain by 24 h was slightly greater in 6-8 month-old TASTPM mice than in wildtype controls. Changes in copper trafficking associated with Alzheimer's-like pathology after administration of ionic 64Cu are minor compared to those observed after administration of 64Cu-GTSM. PET imaging with 64Cu could help understand changes in brain copper dynamics in AD and underpin new clinical diagnostic imaging methods.
Collapse
Affiliation(s)
- Erica M Andreozzi
- Division of Imaging Sciences, Kings College London, St. Thomas Hospital, London, UK.
| | - Julia Baguña Torres
- Division of Imaging Sciences, Kings College London, St. Thomas Hospital, London, UK.
| | - Kavitha Sunassee
- Division of Imaging Sciences, Kings College London, St. Thomas Hospital, London, UK.
| | - Joel Dunn
- Division of Imaging Sciences, Kings College London, St. Thomas Hospital, London, UK.
| | - Simon Walker-Samuel
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, London, UK
| | - Istvan Szanda
- Division of Imaging Sciences, Kings College London, St. Thomas Hospital, London, UK.
| | - Philip J Blower
- Division of Imaging Sciences, Kings College London, St. Thomas Hospital, London, UK.
| |
Collapse
|
39
|
Gardner B, Dieriks BV, Cameron S, Mendis LHS, Turner C, Faull RLM, Curtis MA. Metal concentrations and distributions in the human olfactory bulb in Parkinson's disease. Sci Rep 2017; 7:10454. [PMID: 28874699 PMCID: PMC5585381 DOI: 10.1038/s41598-017-10659-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 08/14/2017] [Indexed: 01/10/2023] Open
Abstract
In Parkinson's disease (PD), the olfactory bulb is typically the first region in the body to accumulate alpha-synuclein aggregates. This pathology is linked to decreased olfactory ability, which becomes apparent before any motor symptoms occur, and may be due to a local metal imbalance. Metal concentrations were investigated in post-mortem olfactory bulbs and tracts from 17 human subjects. Iron (p < 0.05) and sodium (p < 0.01) concentrations were elevated in the PD olfactory bulb. Combining laser ablation inductively coupled plasma mass spectrometry and immunohistochemistry, iron and copper were evident at very low levels in regions of alpha-synuclein aggregation. Zinc was high in these regions, and free zinc was detected in Lewy bodies, mitochondria, and lipofuscin of cells in the anterior olfactory nucleus. Increased iron and sodium in the human PD olfactory bulb may relate to the loss of olfactory function. In contrast, colocalization of free zinc and alpha-synuclein in the anterior olfactory nucleus implicate zinc in PD pathogenesis.
Collapse
Affiliation(s)
- Bronwen Gardner
- Centre for Brain Research and Department of Anatomy with Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Birger V Dieriks
- Centre for Brain Research and Department of Anatomy with Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Steve Cameron
- Waikato Mass Spectrometry Facility, University of Waikato, Hamilton, New Zealand
| | - Lakshini H S Mendis
- Centre for Brain Research and Department of Anatomy with Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Clinton Turner
- Centre for Brain Research and Department of Anatomy with Medical Imaging, University of Auckland, Auckland, New Zealand
- Department of Anatomical Pathology, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research and Department of Anatomy with Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research and Department of Anatomy with Medical Imaging, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
40
|
|
41
|
Folarin OR, Snyder AM, Peters DG, Olopade F, Connor JR, Olopade JO. Brain Metal Distribution and Neuro-Inflammatory Profiles after Chronic Vanadium Administration and Withdrawal in Mice. Front Neuroanat 2017; 11:58. [PMID: 28790895 PMCID: PMC5524677 DOI: 10.3389/fnana.2017.00058] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/28/2017] [Indexed: 01/23/2023] Open
Abstract
Vanadium is a potentially toxic environmental pollutant and induces oxidative damage in biological systems including the central nervous system (CNS). Its deposition in brain tissue may be involved in the pathogenesis of certain neurological disorders which after prolonged exposure can culminate into more severe pathology. Most studies on vanadium neurotoxicity have been done after acute exposure but in reality some populations are exposed for a lifetime. This work was designed to ascertain neurodegenerative consequences of chronic vanadium administration and to investigate the progressive changes in the brain after withdrawal from vanadium treatment. A total of 85 male BALB/c mice were used for the experiment and divided into three major groups of vanadium treated (intraperitoneally (i.p.) injected with 3 mg/kg body weight of sodium metavanadate and sacrificed every 3 months till 18 months); matched controls; and animals that were exposed to vanadium for 3 months and thereafter the metal was withdrawn. Brain tissues were obtained after animal sacrifice. Sagittal cut sections of paraffin embedded tissue (5 μm) were analyzed by the Laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) to show the absorption and distribution of vanadium metal. Also, Haematoxylin and Eosin (H&E) staining of brain sections, and immunohistochemistry for Microglia (Iba-1), Astrocytes (GFAP), Neurons (Neu-N) and Neu-N + 4',6-diamidine-2'-pheynylindole dihydrochloride (Dapi) Immunofluorescent labeling were observed for morphological and morphometric parameters. The LA-ICP-MS results showed progressive increase in vanadium uptake with time in different brain regions with prediction for regions like the olfactory bulb, brain stem and cerebellum. The withdrawal brains still show presence of vanadium metal in the brain slightly more than the controls. There were morphological alterations (of the layering profile, nuclear shrinkage) in the prefrontal cortex, cellular degeneration (loss of dendritic arborization) and cell death in the Hippocampal CA1 pyramidal cells and Purkinje cells of the cerebellum, including astrocytic and microglial activation in vanadium exposed brains which were all attenuated in the withdrawal group. With exposure into old age, the evident neuropathology was microgliosis, while progressive astrogliosis became more attenuated. We have shown that chronic administration of vanadium over a lifetime in mice resulted in metal accumulation which showed regional variabilities with time. The metal profile and pathological effects were not completely eliminated from the brain even after a long time withdrawal from vanadium metal.
Collapse
Affiliation(s)
- Oluwabusayo R Folarin
- Department of Medical Laboratory Science, Ladoke Akintola University of TechnologyOsogbo, Nigeria
| | - Amanda M Snyder
- Department of Neurosurgery, Pennsylvania State College of MedicineHershey, PA, United States
| | - Douglas G Peters
- Department of Neurosurgery, Pennsylvania State College of MedicineHershey, PA, United States
| | | | - James R Connor
- Department of Neurosurgery, Pennsylvania State College of MedicineHershey, PA, United States
| | - James O Olopade
- Department of Veterinary Anatomy, University of IbadanIbadan, Nigeria
| |
Collapse
|
42
|
The novel compound PBT434 prevents iron mediated neurodegeneration and alpha-synuclein toxicity in multiple models of Parkinson's disease. Acta Neuropathol Commun 2017; 5:53. [PMID: 28659169 PMCID: PMC5490188 DOI: 10.1186/s40478-017-0456-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/14/2017] [Indexed: 12/18/2022] Open
Abstract
Elevated iron in the SNpc may play a key role in Parkinson's disease (PD) neurodegeneration since drug candidates with high iron affinity rescue PD animal models, and one candidate, deferirpone, has shown efficacy recently in a phase two clinical trial. However, strong iron chelators may perturb essential iron metabolism, and it is not yet known whether the damage associated with iron is mediated by a tightly bound (eg ferritin) or lower-affinity, labile, iron pool. Here we report the preclinical characterization of PBT434, a novel quinazolinone compound bearing a moderate affinity metal-binding motif, which is in development for Parkinsonian conditions. In vitro, PBT434 was far less potent than deferiprone or deferoxamine at lowering cellular iron levels, yet was found to inhibit iron-mediated redox activity and iron-mediated aggregation of α-synuclein, a protein that aggregates in the neuropathology. In vivo, PBT434 did not deplete tissue iron stores in normal rodents, yet prevented loss of substantia nigra pars compacta neurons (SNpc), lowered nigral α-synuclein accumulation, and rescued motor performance in mice exposed to the Parkinsonian toxins 6-OHDA and MPTP, and in a transgenic animal model (hA53T α-synuclein) of PD. These improvements were associated with reduced markers of oxidative damage, and increased levels of ferroportin (an iron exporter) and DJ-1. We conclude that compounds designed to target a pool of pathological iron that is not held in high-affinity complexes in the tissue can maintain the survival of SNpc neurons and could be disease-modifying in PD.
Collapse
|
43
|
Commentary: Comments regarding Becker et al. (Analytica Chimica Acta, 835, 2014, 1–18). Anal Chim Acta 2017; 972:12-15. [DOI: 10.1016/j.aca.2017.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 03/20/2017] [Accepted: 03/21/2017] [Indexed: 11/20/2022]
|
44
|
Van Malderen SJM, Laforce B, Van Acker T, Nys C, De Rijcke M, de Rycke R, De Bruyne M, Boone MN, De Schamphelaere K, Borovinskaya O, De Samber B, Vincze L, Vanhaecke F. Three-Dimensional Reconstruction of the Tissue-Specific Multielemental Distribution within Ceriodaphnia dubia via Multimodal Registration Using Laser Ablation ICP-Mass Spectrometry and X-ray Spectroscopic Techniques. Anal Chem 2017; 89:4161-4168. [DOI: 10.1021/acs.analchem.7b00111] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Stijn J. M. Van Malderen
- Department of Analytical
Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S12, 9000 Ghent, Belgium
| | - Brecht Laforce
- Department of Analytical
Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S12, 9000 Ghent, Belgium
| | - Thibaut Van Acker
- Department of Analytical
Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S12, 9000 Ghent, Belgium
| | - Charlotte Nys
- Department of Applied Ecology and Environmental Biology, Ghent University, Jozef Plateaustraat 22, 9000 Ghent, Belgium
| | - Maarten De Rijcke
- Department of Applied Ecology and Environmental Biology, Ghent University, Jozef Plateaustraat 22, 9000 Ghent, Belgium
| | | | | | - Matthieu N. Boone
- Department of Physics and Astronomy, Ghent University, Proeftuinstraat 86, 9000 Ghent, Belgium
| | - Karel De Schamphelaere
- Department of Applied Ecology and Environmental Biology, Ghent University, Jozef Plateaustraat 22, 9000 Ghent, Belgium
| | | | - Björn De Samber
- Department of Analytical
Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S12, 9000 Ghent, Belgium
| | - Laszlo Vincze
- Department of Analytical
Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S12, 9000 Ghent, Belgium
| | - Frank Vanhaecke
- Department of Analytical
Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S12, 9000 Ghent, Belgium
| |
Collapse
|
45
|
James SA, Churches QI, de Jonge MD, Birchall IE, Streltsov V, McColl G, Adlard PA, Hare DJ. Iron, Copper, and Zinc Concentration in Aβ Plaques in the APP/PS1 Mouse Model of Alzheimer's Disease Correlates with Metal Levels in the Surrounding Neuropil. ACS Chem Neurosci 2017; 8:629-637. [PMID: 27958708 DOI: 10.1021/acschemneuro.6b00362] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The metal ions of iron, copper, and zinc have long been associated with the aggregation of β-amyloid (Aβ) plaques in Alzheimer's disease; an interaction that has been suggested to promote increased oxidative stress and neuronal dysfunction. We examined plaque metal load in the hippocampus of APP/PS1 mice using X-ray fluorescence microscopy to assess how the anatomical location of Aβ plaques was influenced by the metal content of surrounding tissue. Immunohistochemical staining of Aβ plaques colocalized with areas of increased X-ray scattering power in unstained tissue sections, allowing direct X-ray based-assessment of plaque metal levels in sections subjected to minimal chemical fixation. We identified and mapped 48 individual plaques in four subregions of the hippocampus from four biological replicates. Iron, Cu, and Zn areal concentrations (ng cm-2) were increased in plaques compared to the surrounding neuropil. However, this elevation in metal load reflected the local metal makeup of the surrounding neuropil, where different brain regions are enriched for different metal ions. After correcting for tissue density, only Zn levels remained elevated in plaques. This study suggests that the in vivo binding of Zn to plaques is not simply due to increased protein deposition.
Collapse
Affiliation(s)
- Simon A. James
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
- Australian Synchrotron, Clayton, Victoria 3168, Australia
| | - Quentin I. Churches
- Biomedical
Manufacturing,
CSIRO Manufacturing, Clayton South, Victoria 3169, Australia
| | | | - Ian E. Birchall
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Victor Streltsov
- Biomedical
Manufacturing,
CSIRO Manufacturing, Clayton South, Victoria 3169, Australia
| | - Gawain McColl
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Paul A. Adlard
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Dominic J. Hare
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
- Elemental
Bio-imaging Facility, University of Technology Sydney, Broadway, Sydney, New South Wales 2007, Australia
| |
Collapse
|
46
|
Hare DJ, Kysenius K, Paul B, Knauer B, Hutchinson RW, O'Connor C, Fryer F, Hennessey TP, Bush AI, Crouch PJ, Doble PA. Imaging Metals in Brain Tissue by Laser Ablation - Inductively Coupled Plasma - Mass Spectrometry (LA-ICP-MS). J Vis Exp 2017. [PMID: 28190025 PMCID: PMC5352277 DOI: 10.3791/55042] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Metals are found ubiquitously throughout an organism, with their biological role dictated by both their chemical reactivity and abundance within a specific anatomical region. Within the brain, metals have a highly compartmentalized distribution, depending on the primary function they play within the central nervous system. Imaging the spatial distribution of metals has provided unique insight into the biochemical architecture of the brain, allowing direct correlation between neuroanatomical regions and their known function with regard to metal-dependent processes. In addition, several age-related neurological disorders feature disrupted metal homeostasis, which is often confined to small regions of the brain that are otherwise difficult to analyze. Here, we describe a comprehensive method for quantitatively imaging metals in the mouse brain, using laser ablation - inductively coupled plasma - mass spectrometry (LA-ICP-MS) and specially designed image processing software. Focusing on iron, copper and zinc, which are three of the most abundant and disease-relevant metals within the brain, we describe the essential steps in sample preparation, analysis, quantitative measurements and image processing to produce maps of metal distribution within the low micrometer resolution range. This technique, applicable to any cut tissue section, is capable of demonstrating the highly variable distribution of metals within an organ or system, and can be used to identify changes in metal homeostasis and absolute levels within fine anatomical structures.
Collapse
Affiliation(s)
- Dominic J Hare
- Elemental Bio-imaging Facility, University of Technology Sydney; Florey Institute of Neuroscience and Mental Health, The University of Melbourne;
| | - Kai Kysenius
- Department of Pathology, The University of Melbourne
| | - Bence Paul
- School of Earth Sciences, The University of Melbourne
| | - Beate Knauer
- Research School, Ruhr University; Department of Physiology, Monash University
| | | | | | | | | | - Ashley I Bush
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne
| | | | - Philip A Doble
- Elemental Bio-imaging Facility, University of Technology Sydney
| |
Collapse
|
47
|
Portbury SD, Hare DJ, Sgambelloni CJ, Bishop DP, Finkelstein DI, Doble PA, Adlard PA. Age modulates the injury-induced metallomic profile in the brain. Metallomics 2017; 9:402-410. [DOI: 10.1039/c6mt00260a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
48
|
A comparison of sample preparation strategies for biological tissues and subsequent trace element analysis using LA-ICP-MS. Anal Bioanal Chem 2016; 409:1805-1814. [PMID: 27966170 PMCID: PMC5591616 DOI: 10.1007/s00216-016-0124-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 02/08/2023]
Abstract
Laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) is one of the most commonly applied methods for lateral trace element distribution analysis in medical studies. Many improvements of the technique regarding quantification and achievable lateral resolution have been achieved in the last years. Nevertheless, sample preparation is also of major importance and the optimal sample preparation strategy still has not been defined. While conventional histology knows a number of sample pre-treatment strategies, little is known about the effect of these approaches on the lateral distributions of elements and/or their quantities in tissues. The technique of formalin fixation and paraffin embedding (FFPE) has emerged as the gold standard in tissue preparation. However, the potential use for elemental distribution studies is questionable due to a large number of sample preparation steps. In this work, LA-ICP-MS was used to examine the applicability of the FFPE sample preparation approach for elemental distribution studies. Qualitative elemental distributions as well as quantitative concentrations in cryo-cut tissues as well as FFPE samples were compared. Results showed that some metals (especially Na and K) are severely affected by the FFPE process, whereas others (e.g., Mn, Ni) are less influenced. Based on these results, a general recommendation can be given: FFPE samples are completely unsuitable for the analysis of alkaline metals. When analyzing transition metals, FFPE samples can give comparable results to snap-frozen tissues. Graphical abstract Sample preparation strategies for biological tissues are compared with regard to the elemental distributions and average trace element concentrations.
Collapse
|
49
|
Colvin RA, Jin Q, Lai B, Kiedrowski L. Visualizing Metal Content and Intracellular Distribution in Primary Hippocampal Neurons with Synchrotron X-Ray Fluorescence. PLoS One 2016; 11:e0159582. [PMID: 27434052 PMCID: PMC4951041 DOI: 10.1371/journal.pone.0159582] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/04/2016] [Indexed: 02/03/2023] Open
Abstract
Increasing evidence suggests that metal dyshomeostasis plays an important role in human neurodegenerative diseases. Although distinctive metal distributions are described for mature hippocampus and cortex, much less is known about metal levels and intracellular distribution in individual hippocampal neuronal somata. To solve this problem, we conducted quantitative metal analyses utilizing synchrotron radiation X-Ray fluorescence on frozen hydrated primary cultured neurons derived from rat embryonic cortex (CTX) and two regions of the hippocampus: dentate gyrus (DG) and CA1. Comparing average metal contents showed that the most abundant metals were calcium, iron, and zinc, whereas metals such as copper and manganese were less than 10% of zinc. Average metal contents were generally similar when compared across neurons cultured from CTX, DG, and CA1, except for manganese that was larger in CA1. However, each metal showed a characteristic spatial distribution in individual neuronal somata. Zinc was uniformly distributed throughout the cytosol, with no evidence for the existence of previously identified zinc-enriched organelles, zincosomes. Calcium showed a peri-nuclear distribution consistent with accumulation in endoplasmic reticulum and/or mitochondria. Iron showed 2-3 distinct highly concentrated puncta only in peri-nuclear locations. Notwithstanding the small sample size, these analyses demonstrate that primary cultured neurons show characteristic metal signatures. The iron puncta probably represent iron-accumulating organelles, siderosomes. Thus, the metal distributions observed in mature brain structures are likely the result of both intrinsic neuronal factors that control cellular metal content and extrinsic factors related to the synaptic organization, function, and contacts formed and maintained in each region.
Collapse
Affiliation(s)
- Robert A. Colvin
- Department of Biological Sciences, Interdisciplinary Graduate Program in Molecular and Cellular Biology, Neuroscience Program, Ohio University, Athens, Ohio, United States of America
| | - Qiaoling Jin
- Department of Physics & Astronomy, Northwestern University, Evanston, Illinois, United States of America
| | - Barry Lai
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, Illinois, United States of America
| | - Lech Kiedrowski
- The Psychiatric Institute, Departments of Psychiatry and Pharmacology, the University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
50
|
Billings JL, Hare DJ, Nurjono M, Volitakis I, Cherny RA, Bush AI, Adlard PA, Finkelstein DI. Effects of Neonatal Iron Feeding and Chronic Clioquinol Administration on the Parkinsonian Human A53T Transgenic Mouse. ACS Chem Neurosci 2016; 7:360-6. [PMID: 26712118 DOI: 10.1021/acschemneuro.5b00305] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Increased nigral iron (Fe) is a cardinal feature of Parkinson's disease, as is the accumulation of aggregates comprising α-synuclein. We used wild-type mice and transgenic mice overexpressing the human A53T mutation to α-synuclein to examine the influence of increased Fe (days 10-17 postpartum) on the parkinsonian development phenotype of these animals (including abnormal nigral Fe levels and deficits in both cell numbers and locomotor activity), and to explore the impact of the Fe chelator clioquinol in the model. Both untreated and Fe-loaded A53T mice showed similar levels of nigral cell loss, though 5 months of clioquinol treatment was only able to prevent the loss in the non-Fe-loaded A53T group. Iron levels in the Fe-loaded A53T mice returned to normal at 8 months, though effects of dopamine denervation remained, demonstrated by limited locomotor activity and sustained neuron loss. These data suggest that Fe exposure during a critical developmental window, combined with the overexpression mutant α-synuclein, presents a disease phenotype resistant to intervention using clioquinol later in life.
Collapse
Affiliation(s)
- Jessica L. Billings
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Dominic J. Hare
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
- Elemental
Bio-imaging Facility, University of Technology Sydney, Broadway, New South Wales 2007, Australia
| | - Milawaty Nurjono
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Irene Volitakis
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Robert A. Cherny
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Ashley I. Bush
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Paul A. Adlard
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - David I. Finkelstein
- The
Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|