1
|
Franklin ME, Grant JL, Lee GM, Alvarez-Ciara A, Bennett C, Mattis S, Gallardo N, Corrales N, Cui XT, Capadona JR, Streit WJ, Olivier JH, Keane RW, Dietrich WD, de Rivero Vaccari JP, Prasad A. Effects of iron accumulation and its chelation on oxidative stress in intracortical implants. Acta Biomater 2025:S1742-7061(25)00349-6. [PMID: 40355018 DOI: 10.1016/j.actbio.2025.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Long-term reliability of microelectrodes implanted in the cortex is hindered due to the foreign body response that occurs at the electrode-tissue interface. Following implantation, there is disruption of the blood-brain-barrier and vasculature, resulting in activation of immune cells and release of erythrocytes. As a result of hemolysis, erythrocytes degrade to heme and then to free iron. Excess free iron can participate in the Fenton Reaction, producing reactive oxygen species (ROS). Iron-mediated ROS production can contribute to oxidation of lipids, proteins, and DNA, facilitating a hostile environment of oxidative stress leading to oxidative cellular damage, cytotoxicity, and cell death. The objective of this study was to show the iron accumulation and the downstream effects of oxidative stress at the injury site. A 16-channel microelectrode array (MEA) was implanted in the rat somatosensory cortex. Our results indicated significant elevation of NOX complex subunits across timepoints, suggesting sustained oxidative stress. In a separate group of animals, we administered an iron chelator, deferoxamine mesylate (DFX), to evaluate the effects of chelation on iron accumulation, oxidative stress and damage, and neuronal survival. Results indicate that animals with iron chelation showed reduced ferric iron and markers of oxidative stress and damage corresponding with increased expression of neuronal cell bodies and electrophysiological functional performance. In summary, the study reveals the role of iron in mediating oxidative stress and the effects of modulating iron levels using iron chelation at the electrode-tissue interface. STATEMENT OF SIGNIFICANCE: Iron accumulation has been observed in central nervous system injuries and in neurodegenerative diseases such as Alzheimer's and Parkinson's disease. While the role of iron is studied in various neurodegenerative diseases and traumatic brain injury, iron accumulation and its effect on oxidative stress is not known for intracortical implants where there is a persistent injury due to the presence of a foreign device in the brain tissue. The study seeks to understand the effects of iron accumulation on oxidative stress and damage at the electrode-tissue interface in intracortical implants by using iron chelation as a method of modulating iron levels at the interface.
Collapse
Affiliation(s)
- Melissa E Franklin
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Jordan L Grant
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Grant M Lee
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | | | - Cassie Bennett
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Serene Mattis
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Nicolas Gallardo
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Natalie Corrales
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Wolfgang J Streit
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | | | - Robert W Keane
- Department of Cellular Physiology and Molecular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - W Dalton Dietrich
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Cellular Physiology and Molecular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
2
|
Sharma V, Sharma P, Singh TG. Ferroptosis and Alzheimer's: unveiling new avenues for the treatment and prevention. Metab Brain Dis 2025; 40:167. [PMID: 40167846 DOI: 10.1007/s11011-025-01587-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD), one of the most prevalent neurodegenerative illnesses worldwide, has a devastating effect on individual, families and society. Despite the extensive research and effort, various clinical trials aimed against amyloid-β, which is suspected to have a causative role in the illness, have not yet shown any clinically significant success to date. Emerging evidence suggests that ferroptosis, a kind of programmed cell death triggered by lipid peroxidation and dependent on iron, plays a role in AD. There is a complex relationship between AD and ferroptosis. In both the processes iron dysregulation, altered anti-oxidant mechanisms and lipid peroxidation is involved. Ferroptotic processes contributes to the neuro-inflammation, oxidative stress and damage to the neurons as observed in AD. Additionally, amyloid-β, a hallmark of AD, may influence the ferroptosis, further linked the two pathways. Numerous signalling pathways such as Phospho inositide 3-kinase, Glycogen synthase kinase-3β, 5'-AMP-activated protein kinase, nuclear factor erythroid 2-related factor-2 and Sirtuin pathway plays a part in the pathophysiology of AD. Through a comprehensive review of current research and experimentation, this investigation elucidates the interactions between novel pharmacological agents (ferroptotic inhibitors) and AD-related pathways. Furthermore, this review highlights the various ferroptotic inhibitors as the therapeutic agents for the slowing down the progression of AD. The crosstalk between these processes could unveil the potential therapeutic targets for the AD treatment.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Prateek Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
3
|
Willans M, Hollings A, Boseley RE, Munyard T, Ellison GC, Hackett MJ. The application of X-ray fluorescence microscopy and micro-XANES spectroscopy to study neuro-metallomics. J Inorg Biochem 2025; 262:112744. [PMID: 39341704 DOI: 10.1016/j.jinorgbio.2024.112744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/02/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
This early career research highlight provides a review of my own research program over the last decade, a time frame that encompasses my transition from postdoctoral fellowships to independent researcher. As an analytical chemist and applied spectroscopist, the central theme of my research program over this time has been protocol development at synchrotron facilities, with the main objective to investigate brain metal homeostasis during both brain health and brain disease. I will begin my review with an overview of brain metal homeostasis, before introducing analytical challenges associated with its study. I will then provide a brief summary of the two main X-ray techniques I have used to study brain metal homeostasis, X-ray fluorescence microscopy (XFM) and X-ray absorption near edge structure spectroscopy (XANES). The review then finishes with a summary of my main research contributions using these two techniques, put in the context of the results from others in the field.
Collapse
Affiliation(s)
- Meg Willans
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA, Australia
| | - Ashley Hollings
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Rhiannon E Boseley
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA, Australia
| | - Thomas Munyard
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA, Australia
| | - Gaewyn C Ellison
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Mark J Hackett
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Bentley, WA, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia.
| |
Collapse
|
4
|
LeVine SM. The Azalea Hypothesis of Alzheimer Disease: A Functional Iron Deficiency Promotes Neurodegeneration. Neuroscientist 2024; 30:525-544. [PMID: 37599439 PMCID: PMC10876915 DOI: 10.1177/10738584231191743] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Chlorosis in azaleas is characterized by an interveinal yellowing of leaves that is typically caused by a deficiency of iron. This condition is usually due to the inability of cells to properly acquire iron as a consequence of unfavorable conditions, such as an elevated pH, rather than insufficient iron levels. The causes and effects of chlorosis were found to have similarities with those pertaining to a recently presented hypothesis that describes a pathogenic process in Alzheimer disease. This hypothesis states that iron becomes sequestered (e.g., by amyloid β and tau), causing a functional deficiency of iron that disrupts biochemical processes leading to neurodegeneration. Additional mechanisms that contribute to iron becoming unavailable include iron-containing structures not undergoing proper recycling (e.g., disrupted mitophagy and altered ferritinophagy) and failure to successfully translocate iron from one compartment to another (e.g., due to impaired lysosomal acidification). Other contributors to a functional deficiency of iron in patients with Alzheimer disease include altered metabolism of heme or altered production of iron-containing proteins and their partners (e.g., subunits, upstream proteins). A review of the evidence supporting this hypothesis is presented. Also, parallels between the mechanisms underlying a functional iron-deficient state in Alzheimer disease and those occurring for chlorosis in plants are discussed. Finally, a model describing the generation of a functional iron deficiency in Alzheimer disease is put forward.
Collapse
Affiliation(s)
- Steven M. LeVine
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, US
| |
Collapse
|
5
|
Schwehr BJ, Hartnell D, Ellison G, Hindes MT, Milford B, Dallerba E, Hickey SM, Pfeffer FM, Brooks DA, Massi M, Hackett MJ. Fluorescent probes for neuroscience: imaging ex vivo brain tissue sections. Analyst 2024; 149:4536-4552. [PMID: 39171617 DOI: 10.1039/d4an00663a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Neurobiological research relies heavily on imaging techniques, such as fluorescence microscopy, to understand neurological function and disease processes. However, the number and variety of fluorescent probes available for ex vivo tissue section imaging limits the advance of research in the field. In this review, we outline the current range of fluorescent probes that are available to researchers for ex vivo brain section imaging, including their physical and chemical characteristics, staining targets, and examples of discoveries for which they have been used. This review is organised into sections based on the biological target of the probe, including subcellular organelles, chemical species (e.g., labile metal ions), and pathological phenomenon (e.g., degenerating cells, aggregated proteins). We hope to inspire further development in this field, given the considerable benefits to be gained by the greater availability of suitably sensitive probes that have specificity for important brain tissue targets.
Collapse
Affiliation(s)
- Bradley J Schwehr
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
| | - David Hartnell
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
- Curtin University, Curtin Health Innovation Research Institute, Perth, WA, Australia 6102
| | - Gaewyn Ellison
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
- Curtin University, Curtin Health Innovation Research Institute, Perth, WA, Australia 6102
| | - Madison T Hindes
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000
| | - Breah Milford
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
| | - Elena Dallerba
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
| | - Shane M Hickey
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000
| | - Frederick M Pfeffer
- School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - Doug A Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000
| | - Massimiliano Massi
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
| | - Mark J Hackett
- Curtin University, School of Molecular and Life Sciences, Perth, WA, Australia 6845.
- Curtin University, Curtin Health Innovation Research Institute, Perth, WA, Australia 6102
| |
Collapse
|
6
|
Hackett MJ. A commentary on studies of brain iron accumulation during ageing. J Biol Inorg Chem 2024; 29:385-394. [PMID: 38735007 PMCID: PMC11186910 DOI: 10.1007/s00775-024-02060-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
Brain iron content is widely reported to increase during "ageing", across multiple species from nematodes, rodents (mice and rats) and humans. Given the redox-active properties of iron, there has been a large research focus on iron-mediated oxidative stress as a contributor to tissue damage during natural ageing, and also as a risk factor for neurodegenerative disease. Surprisingly, however, the majority of published studies have not investigated brain iron homeostasis during the biological time period of senescence, and thus knowledge of how brain homeostasis changes during this critical stage of life largely remains unknown. This commentary examines the literature published on the topic of brain iron homeostasis during ageing, providing a critique on limitations of currently used experimental designs. The commentary also aims to highlight that although much research attention has been given to iron accumulation or iron overload as a pathological feature of ageing, there is evidence to support functional iron deficiency may exist, and this should not be overlooked in studies of ageing or neurodegenerative disease.
Collapse
Affiliation(s)
- 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.
| |
Collapse
|
7
|
Nwafor DC, Benkovic SA, Clary BL, Brichacek AL, Lambert HW, Zdilla MJ, Brown CM. A Scalable Histological Method to Embed and Section Multiple Brains Simultaneously. Cells 2024; 13:860. [PMID: 38786081 PMCID: PMC11119749 DOI: 10.3390/cells13100860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
The preparation and processing of rodent brains for evaluation by immunohistochemistry is time-consuming. A large number of mouse brains are routinely used in experiments in neuroscience laboratories to evaluate several models of human diseases. Thus, methods are needed to reduce the time associated with processing brains for histology. A scalable method was developed to embed, section, and stain multiple mouse brains using supplies found in any common histology laboratory. Section collection schemes can be scaled to provide identical bregma locations between adjacent sections for immunohistochemistry, facilitating comprehensive, high-quality immunohistochemistry. As a result, sectioning and staining times are considerably reduced as sections from multiple blocks are stained simultaneously. This method improves on previous procedures and allows multiple embedding and subsequent immunostaining of brains easily with a dramatically reduced time requirement. Furthermore, we expand this method for use in numerous mouse tissues, rat brain tissue, and post-mortem human brain and arterial tissues. In summary, this procedure allows the processing of many rodent or human tissues from perfusion through microscopy in 10 days or less.
Collapse
Affiliation(s)
- Divine C. Nwafor
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 22903, USA;
| | - Stanley A. Benkovic
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV 26506, USA (B.L.C.)
| | - Briana L. Clary
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV 26506, USA (B.L.C.)
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Allison L. Brichacek
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV 26506, USA;
| | - H. Wayne Lambert
- Department of Pathology, Anatomy and Laboratory Medicine, School of Medicine, West Virginia University, Morgantown, WV 26506, USA; (H.W.L.); (M.J.Z.)
| | - Matthew J. Zdilla
- Department of Pathology, Anatomy and Laboratory Medicine, School of Medicine, West Virginia University, Morgantown, WV 26506, USA; (H.W.L.); (M.J.Z.)
| | - Candice M. Brown
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV 26506, USA (B.L.C.)
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV 26506, USA;
| |
Collapse
|
8
|
Joshi J, Yao M, Kakazu A, Ouyang Y, Duan W, Aggarwal M. Distinguishing microgliosis and tau deposition in the mouse brain using paramagnetic and diamagnetic susceptibility source separation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.11.588962. [PMID: 38659855 PMCID: PMC11042227 DOI: 10.1101/2024.04.11.588962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Tauopathies, including Alzheimer's disease (AD), are neurodegenerative disorders characterized by hyperphosphorylated tau protein aggregates in the brain. In addition to protein aggregates, microglia-mediated inflammation and iron dyshomeostasis are other pathological features observed in AD and other tauopathies. It is known that these alterations at the subcellular level occur much before the onset of macroscopic tissue atrophy or cognitive deficits. The ability to detect these microstructural changes with MRI therefore has substantive importance for improved characterization of disease pathogenesis. In this study, we demonstrate that quantitative susceptibility mapping (QSM) with paramagnetic and diamagnetic susceptibility source separation has the potential to distinguish neuropathological alterations in a transgenic mouse model of tauopathy. 3D multi-echo gradient echo data were acquired from fixed brains of PS19 (Tau) transgenic mice and age-matched wild-type (WT) mice (n = 5 each) at 11.7 T. The multi-echo data were fit to a 3-pool complex signal model to derive maps of paramagnetic component susceptibility (PCS) and diamagnetic component susceptibility (DCS). Group-averaged signal fraction and composite susceptibility maps showed significant region-specific differences between the WT and Tau mouse brains. Significant bilateral increases in PCS and |DCS| were observed in specific hippocampal and cortical sub-regions of the Tau mice relative to WT controls. Comparison with immunohistological staining for microglia (Iba1) and phosphorylated-tau (AT8) further indicated that the PCS and DCS differences corresponded to regional microgliosis and tau deposition in the PS19 mouse brains, respectively. The results demonstrate that quantitative susceptibility source separation may provide sensitive imaging markers to detect distinct pathological alterations in tauopathies.
Collapse
Affiliation(s)
- Jayvik Joshi
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Minmin Yao
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aaron Kakazu
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuxiao Ouyang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Manisha Aggarwal
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
Kong R, Ji L, Pang Y, Zhao D, Gao J. Exosomes from osteoarthritic fibroblast-like synoviocytes promote cartilage ferroptosis and damage via delivering microRNA-19b-3p to target SLC7A11 in osteoarthritis. Front Immunol 2023; 14:1181156. [PMID: 37691947 PMCID: PMC10484587 DOI: 10.3389/fimmu.2023.1181156] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/10/2023] [Indexed: 09/12/2023] Open
Abstract
Objective Our previous studies revealed that normal synovial exosomes promoted chondrogenesis, and microRNA (miR)-19b-3p independently related to osteoarthritis (OA) risk. Subsequently, this study intended to further explore the effect of OA fibroblast-like synoviocyte (OA-FLS) exosomal miR-19b-3p on OA ferroptosis and its potential mechanisms. Methods Interleukin (IL)-1β-stimulated chondrocytes and medial meniscus surgery were used to construct the OA cellular model and the OA rat model, respectively. OA-FLS exosomes with/without miR-19b-3p modification were added to the IL-1β-stimulated chondrocytes and OA rat models, followed by direct miR-19b-3p mimic/inhibitor transfection with/without SLC7A11 overexpression plasmids. miR-19b-3p, ferroptosis-related markers (malondialdehyde (MDA), glutathione (GSH)/oxidized glutathione (GSSG), ferrous ion (Fe2+), glutathione peroxidase 4 (GPX4), solute carrier family 7 member 11 (SLC7A11), and acyl-CoA synthetase long-chain family member 4 (ACSL4)), mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) levels were detected. Results Enhanced ferroptosis reflected by dysregulated ferroptosis-related markers, a reduced MMP, and an increased ROS was observed in cartilage tissues from OA patients vs. controls, IL-1β-stimulated chondrocytes vs. normal ones, and OA rat models vs. sham, so did miR-19b-3p. OA-FLS exosomes promoted MDA, Fe2+, ACSL4, and ROS but reduced cell viability, GSH/GSSG, GPX4, SLC7A11, and MMP in IL-1β-stimulated chondrocytes, whose effect was enhanced by miR-19b-3p mimics and attenuated by miR-19b-3p inhibitors. miR-19b-3p negatively regulated SLC7A11 and directly bound to SLC7A11 via luciferase reporter gene assay. Furthermore, SLC7A11 overexpression weakened miR-19b-3p mimics' effect on ferroptosis-related markers, MMP, or ROS in IL-1β-stimulated chondrocytes. OA-FLS exosomes also induced cartilage damage and ferroptosis in OA rats whose influence was tempered by miR-19b-3p inhibitors. Conclusion OA-FLS exosomal miR-19b-3p enhances cartilage ferroptosis and damage by sponging SLC7A11 in OA, indicating a potential linkage among synovium, cartilage, and ferroptosis during the OA process.
Collapse
Affiliation(s)
- Ruina Kong
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lianmei Ji
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yafei Pang
- Department of Rheumatology, Shanghai Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Dongbao Zhao
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jie Gao
- Department of Rheumatology and Immunology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
10
|
Apostolopoulou EP, Raikos N, Vlemmas I, Michaelidis E, Brellou GD. Metallothionein I/II Expression and Metal Ion Levels in Correlation with Amyloid Beta Deposits in the Aged Feline Brain. Brain Sci 2023; 13:1115. [PMID: 37509045 PMCID: PMC10377600 DOI: 10.3390/brainsci13071115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/13/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Brain aging has been correlated with high metallothionein I-II (MT-I/II) expression, iron and zinc dyshomeostasis, and Aβ deposition in humans and experimental animals. In the present study, iron and zinc accumulation, the expression of MT-I/II and Aβ42, and their potential association with aging in the feline brain were assessed. Tissue sections from the temporal and frontal grey (GM) and white (WM) matter, hippocampus, thalamus, striatum, cerebellum, and dentate nucleus were examined histochemically for the presence of age-related histopathological lesions and iron deposits and distribution. We found, using a modified Perl's/DAB method, two types of iron plaques that showed age-dependent accumulation in the temporal GM and WM and the thalamus, along with the age-dependent increment in cerebellar-myelin-associated iron. We also demonstrated an age-dependent increase in MT-I/II immunoreactivity in the feline brain. In cats over 7 years old, Aβ immunoreactivity was detected in vessel walls and neuronal somata; extracellular Aβ deposits were also evident. Interestingly, Aβ-positive astrocytes were also observed in certain cases. ICP-MS analysis of brain content regarding iron and zinc concentrations showed no statistically significant association with age, but a mild increase in iron with age was noticed, while zinc levels were found to be higher in the Mature and Senior groups. Our findings reinforce the suggestion that cats could serve as a dependable natural animal model for brain aging and neurodegeneration; thus, they should be further investigated on the basis of metal ion concentration changes and their effects on aging.
Collapse
Affiliation(s)
- Emmanouela P Apostolopoulou
- Department of Pathology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54627 Thessaloniki, Greece
| | - Nikolaos Raikos
- Department of Forensic Medicine & Toxicology, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Ioannis Vlemmas
- Department of Pathology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54627 Thessaloniki, Greece
| | - Efstratios Michaelidis
- Laboratories of the 3rd Army Veterinary Hospital, Chemical Department, 57001 Thessaloniki, Greece
| | - Georgia D Brellou
- Department of Pathology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54627 Thessaloniki, Greece
| |
Collapse
|
11
|
Exploring Whether Iron Sequestration within the CNS of Patients with Alzheimer’s Disease Causes a Functional Iron Deficiency That Advances Neurodegeneration. Brain Sci 2023; 13:brainsci13030511. [PMID: 36979320 PMCID: PMC10046656 DOI: 10.3390/brainsci13030511] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/22/2023] Open
Abstract
The involvement of iron in the pathogenesis of Alzheimer’s disease (AD) may be multifaceted. Besides potentially inducing oxidative damage, the bioavailability of iron may be limited within the central nervous system, creating a functionally iron-deficient state. By comparing staining results from baseline and modified iron histochemical protocols, iron was found to be more tightly bound within cortical sections from patients with high levels of AD pathology compared to subjects with a diagnosis of something other than AD. To begin examining whether the bound iron could cause a functional iron deficiency, a protein-coding gene expression dataset of initial, middle, and advanced stages of AD from olfactory bulb tissue was analyzed for iron-related processes with an emphasis on anemia-related changes in initial AD to capture early pathogenic events. Indeed, anemia-related processes had statistically significant alterations, and the significance of these changes exceeded those for AD-related processes. Other changes in patients with initial AD included the expressions of transcripts with iron-responsive elements and for genes encoding proteins for iron transport and mitochondrial-related processes. In the latter category, there was a decreased expression for the gene encoding pitrilysin metallopeptidase 1 (PITRM1). Other studies have shown that PITRM1 has an altered activity in patients with AD and is associated with pathological changes in this disease. Analysis of a gene expression dataset from PITRM1-deficient or sufficient organoids also revealed statistically significant changes in anemia-like processes. These findings, together with supporting evidence from the literature, raise the possibility that a pathogenic mechanism of AD could be a functional deficiency of iron contributing to neurodegeneration.
Collapse
|
12
|
Bitonto V, Garello F, Scherberich A, Filippi M. Prussian Blue Staining to Visualize Iron Oxide Nanoparticles. Methods Mol Biol 2023; 2566:321-332. [PMID: 36152263 DOI: 10.1007/978-1-0716-2675-7_26] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Iron deposits in cells and tissues can be detected by ex vivo histological examination through the Prussian blue (PB) staining. This practical, inexpensive, and highly sensitive technique involves the treatment of fixed tissue sections and cells with acid solutions of ferrocyanides that combine with ferric ion forming a bright blue pigment (i.e., ferric ferrocyanide). The staining can be applied to visualize iron oxide nanoparticles (IONPs), versatile magnetic nanosystems that are used in various biomedical applications and whose localization is usually required at a higher resolution than that enabled by in vivo tracking techniques.
Collapse
Affiliation(s)
- Valeria Bitonto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Francesca Garello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Arnaud Scherberich
- Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland.
- Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland.
| | - Miriam Filippi
- Soft Robotics Laboratory, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
13
|
Woodman MF, Ozcan MCH, Gura MA, De La Cruz P, Gadson AK, Grive KJ. The Requirement of Ubiquitin C-Terminal Hydrolase L1 (UCHL1) in Mouse Ovarian Development and Fertility †. Biol Reprod 2022; 107:500-513. [PMID: 35512140 PMCID: PMC9382372 DOI: 10.1093/biolre/ioac086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/07/2022] [Accepted: 04/27/2022] [Indexed: 11/14/2022] Open
Abstract
Ubiquitin C-Terminal Hydrolase L1 (UCHL1) is a de-ubiquitinating enzyme enriched in neuronal and gonadal tissues known to regulate the cellular stores of mono-ubiquitin and protein turnover. While its function in maintaining proper motor neuron function is well-established, investigation into its role in the health and function of reproductive processes is only just beginning to be studied. Single-cell-sequencing analysis of all ovarian cells from the murine perinatal period revealed that Uchl1 is very highly expressed in the developing oocyte population, an observation which was corroborated by high levels of oocyte-enriched UCHL1 protein expression in oocytes of all stages throughout the mouse reproductive lifespan. To better understand the role UCHL1 may be playing in oocytes, we utilized a UCHL1-deficient mouse line, finding reduced number of litters, reduced litter sizes, altered folliculogenesis, morphologically abnormal oocytes, disrupted estrous cyclicity and apparent endocrine dysfunction in these animals compared to their wild-type and heterozygous littermates. These data reveal a novel role of UCHL1 in female fertility as well as overall ovarian function, and suggest a potentially essential role for the ubiquitin proteasome pathway in mediating reproductive health. Summary sentence: Ubiquitin C-Terminal Hydrolase L1 (UCHL1) is required for proper ovarian folliculogenesis, estrous cyclicity, and fertility in the female mouse.
Collapse
Affiliation(s)
- Morgan F Woodman
- Women and Infants Hospital of Rhode Island, Department of Obstetrics and Gynecology, Program in Women's Oncology, Providence, RI 02905
| | - Meghan C H Ozcan
- Women and Infants Hospital of Rhode Island, Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility Fellowship Program, Providence, RI 02905.,Warren Alpert Medical School of Brown University, Department of Obstetrics and Gynecology, Providence, RI 02905
| | - Megan A Gura
- Brown University, MCB Graduate Program and Department of Molecular Biology, Cell Biology, and Biochemistry, Providence, RI, 02906
| | - Payton De La Cruz
- Women and Infants Hospital of Rhode Island, Department of Obstetrics and Gynecology, Program in Women's Oncology, Providence, RI 02905.,Brown University, Pathobiology Graduate Program, Providence, RI, 02906
| | - Alexis K Gadson
- Women and Infants Hospital of Rhode Island, Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility Fellowship Program, Providence, RI 02905.,Warren Alpert Medical School of Brown University, Department of Obstetrics and Gynecology, Providence, RI 02905
| | - Kathryn J Grive
- Women and Infants Hospital of Rhode Island, Department of Obstetrics and Gynecology, Program in Women's Oncology, Providence, RI 02905.,Women and Infants Hospital of Rhode Island, Department of Obstetrics and Gynecology, Reproductive Endocrinology and Infertility Fellowship Program, Providence, RI 02905
| |
Collapse
|
14
|
Nazzal M, Madsen EC, Armstrong A, van Nispen J, Murali V, Song E, Voigt M, Madnawat H, Welu A, Manithody C, Suri A, Krebs J, Gilbert E, Samaddar A, Blackall D, Carpenter D, Varma C, Teckman J, Jain AK. Novel NMP split liver model recapitulates human IRI and demonstrates ferroptosis modulators as a new therapeutic strategy. Pediatr Transplant 2022; 26:e14164. [PMID: 34633130 DOI: 10.1111/petr.14164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/27/2021] [Accepted: 09/08/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Almost 9%of deceased donor livers are discarded as marginal donor livers (MDL) due to concern of severe ischemia reperfusion injury (IRI). Emerging data supports ferroptosis (iron regulated hepatocellular death) as an IRI driver, however lack of robust preclinical model limits therapeutic testing. In this manuscript we describe the development of a novel rigorous internal control system utilizing normothermic perfusion of split livers to test ferroptosis regulators modulating IRI. METHODS Upon institutional approval, split human MDLs were placed on our normothermic perfusion machine, Perfusion Regulated Organ Therapeutics with Enhanced Controlled Testing (PROTECT), pumping arterial and portal blood. Experiment 1 compared right (UR) and left (UL) lobes to validate PROTECT. Experiment 2 assessed ferroptosis regulator Deferoxamine in Deferoxamine Agent Treated (DMAT) vs. No Agent Internal Control (NAIC) lobes. Liver serology, histology, and ferroptosis genes were assessed. RESULTS Successful MDL perfusion validated PROTECT with no ALT or AST difference between UR and UL (∆ALT UR: 235, ∆ALT UL: 212; ∆AST UR: 576, ∆AST UL: 389). Liver injury markers increased in NAIC vs. DMAT (∆ALT NAIC: 586, ∆ALT DMAT: -405; ∆AST NAIC: 617, ∆AST DMAT: -380). UR and UL had similar expression of ferroptosis regulators RPL8,HO-1 and HIFα. Significantly decreased intrahepatic iron (p = .038), HO-1 and HIFα in DMAT (HO-1 NAIC: 6.93, HO-1 DMAT: 2.74; HIFαNAIC: 8.67, HIFαDMAT: 2.60)and no hepatocellular necrosis or immunohistochemical staining (Ki67/Cytokeratin-7) differences were noted. CONCLUSION PROTECT demonstrates the therapeutic utility of a novel normothermic perfusion split liver system for drug discovery and rapid translatability of therapeutics, driving a paradigm change in organ recovery and transplant medicine. Our study using human livers, provides preliminary proof of concept for the novel role of ferroptosis regulators in driving IRI.
Collapse
Affiliation(s)
- Mustafa Nazzal
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Erik C Madsen
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Austin Armstrong
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Johan van Nispen
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Vidul Murali
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Eric Song
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Marcus Voigt
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Himani Madnawat
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Adam Welu
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | | | - Anandini Suri
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Joseph Krebs
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Ester Gilbert
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Ashish Samaddar
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Douglas Blackall
- Department of Pathology, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Danielle Carpenter
- Department of Pathology, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Chintalapati Varma
- Department of Surgery, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Jeffrey Teckman
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA.,Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| | - Ajay Kumar Jain
- Department of Pediatrics, Saint Louis University School of Medicine, Saint Louis, Missouri, USA.,Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
15
|
Ellison G, Hollings AL, Hackett MJ. A review of the “metallome” within neurons and glia, as revealed by elemental mapping of brain tissue. BBA ADVANCES 2022; 2:100038. [PMID: 37082604 PMCID: PMC10074908 DOI: 10.1016/j.bbadva.2021.100038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 01/01/2023] Open
Abstract
It is now well established that transition metals, such as Iron (Fe), Copper (Cu), and Zinc (Zn) are necessary for healthy brain function. Although Fe, Cu, and Zn are essential to the brain, imbalances in the amount, distribution, or chemical form ("metallome") of these metals is linked to the pathology of numerous brain diseases or disorders. Despite the known importance of metal ions for both brain health and disease, the metallome that exists within specific types of brain cells is yet to be fully characterised. The aim of this mini-review is to present an overview of the current knowledge of the metallome found within specific brain cells (oligodendrocytes, astrocytes, microglia, and neurons), as revealed by direct elemental mapping techniques. It is hoped this review will foster continued research using direct elemental mapping techniques to fully characterise the brain cell metallome.
Collapse
Affiliation(s)
- Gaewyn Ellison
- School of Molecular and Life Sciences, Curtin University, Perth, WA 6845, Australia
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Ashley L. Hollings
- School of Molecular and Life Sciences, Curtin University, Perth, WA 6845, Australia
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
| | - Mark J. Hackett
- School of Molecular and Life Sciences, Curtin University, Perth, WA 6845, Australia
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia
- Corresponding author.
| |
Collapse
|
16
|
Huang T, Suen D. Iron insufficiency in floral buds impairs pollen development by disrupting tapetum function. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2021; 108:244-267. [PMID: 34310779 PMCID: PMC9292431 DOI: 10.1111/tpj.15438] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 06/25/2021] [Accepted: 07/20/2021] [Indexed: 06/13/2023]
Abstract
Reduction of crop yield due to iron (Fe) deficiency has always been a concern in agriculture. How Fe insufficiency in floral buds affects pollen development remains unexplored. Here, plants transferred to Fe-deficient medium at the reproductive stage had reduced floral Fe content and viable pollen and showed a defective pollen outer wall, all restored by supplying floral buds with Fe. A comparison of differentially expressed genes (DEGs) in Fe-deficient leaves, roots, and anthers suggested that changes in several cellular processes were unique to anthers, including increased lipid degradation. Co-expression analysis revealed that ABORTED MICROSPORES (AMS), DEFECTIVE IN TAPETAL DEVELOPMENT AND FUNCTION1, and BASIC HELIX-LOOP-HELIX 089/091/010 encode key upstream transcription factors of Fe deficiency-responsive DEGs involved in tapetum function and development, including tapetal ROS homeostasis, programmed cell death, and pollen outer wall formation-related lipid metabolism. Analysis of RESPIRATORY-BURST OXIDASE HOMOLOG E (RBOHE) gain- and loss-of-function under Fe deficiency indicated that RBOHE- and Fe-dependent regulation cooperatively control anther reactive oxygen species levels and pollen development. Since DEGs in Fe-deficient anthers were not significantly enriched in genes related to mitochondrial function, the changes in mitochondrial status under Fe deficiency, including respiration activity, density, and morphology, were probably because the Fe amount was insufficient to maintain proper mitochondrial protein function in anthers. To sum up, Fe deficiency in anthers may affect Fe-dependent protein function and impact upstream transcription factors and their downstream genes, resulting in extensively impaired tapetum function and pollen development.
Collapse
Affiliation(s)
- Tzu‐Hsiang Huang
- Agricultural Biotechnology Research CenterAcademia SinicaTaipei11529Taiwan
- Molecular and Biological Agricultural Sciences ProgramTaiwan International Graduate ProgramAcademia Sinica and National Chung‐Hsing UniversityTaipei11529Taiwan
- Graduate Institute of BiotechnologyNational Chung‐Hsing UniversityTaichung40227Taiwan
| | - Der‐Fen Suen
- Agricultural Biotechnology Research CenterAcademia SinicaTaipei11529Taiwan
- Molecular and Biological Agricultural Sciences ProgramTaiwan International Graduate ProgramAcademia Sinica and National Chung‐Hsing UniversityTaipei11529Taiwan
- Biotechnology CenterNational Chung‐Hsing UniversityTaichung40227Taiwan
| |
Collapse
|
17
|
Chen K, Jiang X, Wu M, Cao X, Bao W, Zhu LQ. Ferroptosis, a Potential Therapeutic Target in Alzheimer's Disease. Front Cell Dev Biol 2021; 9:704298. [PMID: 34422824 PMCID: PMC8374166 DOI: 10.3389/fcell.2021.704298] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/09/2021] [Indexed: 12/11/2022] Open
Abstract
Cell death is a common phenomenon in the progression of Alzheimer’s disease (AD). However, the mechanism of triggering the death of neuronal cells remains unclear. Ferroptosis is an iron-dependent lipid peroxidation-driven cell death and emerging evidences have demonstrated the involvement of ferroptosis in the pathological process of AD. Moreover, several hallmarks of AD pathogenesis were consistent with the characteristics of ferroptosis, such as excess iron accumulation, elevated lipid peroxides, and reactive oxygen species (ROS), reduced glutathione (GSH), and glutathione peroxidase 4 (GPX4) levels. Besides, some ferroptosis inhibitors can relieve AD-related pathological symptoms in AD mice and exhibit potential clinical benefits in AD patients. Therefore, ferroptosis is gradually being considered as a distinct cell death mechanism in the pathogenesis of AD. However, direct evidence is still lacking. In this review, we summarize the features of ferroptosis in AD, its underlying mechanisms in AD pathology, and review the application of ferroptosis inhibitors in both AD clinical trials and mice/cell models, to provide valuable information for future treatment and prevention of this devastating disease.
Collapse
Affiliation(s)
- Kai Chen
- Key Lab of Neurological Disorder of Education Ministry, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Neurosurgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Moxin Wu
- Department of Jiujiang Clinical Research Center for Precision Medicine, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Xianming Cao
- Department of Jiujiang Clinical Research Center for Precision Medicine, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Wendai Bao
- Key Lab of Neurological Disorder of Education Ministry, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Ling-Qiang Zhu
- Key Lab of Neurological Disorder of Education Ministry, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
LeVine SM, Zhu H, Tague SE. A Simplified Method for the Histochemical Detection of Iron in Paraffin Sections: Intracellular Iron Deposits in Central Nervous System Tissue. ASN Neuro 2021; 13:1759091420982169. [PMID: 33430620 PMCID: PMC7809306 DOI: 10.1177/1759091420982169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Although all cells contain iron, most histochemical methods fail to reveal the presence of iron within many cells of the central nervous system (CNS), particularly neurons. Previously, a sensitive method was developed that limited the extraction of iron in paraffin sections, and this method revealed staining within neurons. However, the staining was often too robust making it difficult to discern discrete intracellular structures. In 1970, a study incorporated acetone in an iron histochemical procedure to facilitate the demarcation of staining features. In the present study, both acetone and limits to iron extraction were included in a simplified staining procedure. This procedure was applied to paraffin sections of CNS tissue from CISD2 deficient and littermate control mice. Discrete nuclear and cytoplasmic staining features were detected in all mice. Although widely present in neurons, punctate cytoplasmic staining was particularly prominent in large neurons within the hindbrain. Evaluation of extended depth of focus images, from serial focal planes, revealed numerous stained cytoplasmic structures. Additionally, the simplified staining procedure was applied to paraffin sections from Alzheimer’s disease and control cases. Despite suboptimal processing conditions compared to mouse tissue, discrete staining of cytoplasmic structures was revealed in some neurons, although many other neurons had nondescript staining features. In addition, initial findings revealed iron deposited within some vessels from patients with Alzheimer’s disease. In summary, since paraffin sections are commonly used for histological preparations, this simplified histochemical procedure could facilitate the study of iron in various CNS conditions by revealing staining details often missed by other procedures.
Collapse
Affiliation(s)
- Steven M LeVine
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Hao Zhu
- Department of Clinical Laboratory Sciences, University of Kansas Medical Center, Kansas City, Kansas, United States.,Neuroscience Graduate Program, University of Kansas Medical Center, Kansas City, Kansas, United States.,Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Sarah E Tague
- Kansas Intellectual and Developmental Disabilities Research Center, University of Kansas Medical Center, Kansas City, Kansas, United States
| |
Collapse
|
19
|
Ejaz HW, Wang W, Lang M. Copper Toxicity Links to Pathogenesis of Alzheimer's Disease and Therapeutics Approaches. Int J Mol Sci 2020; 21:E7660. [PMID: 33081348 PMCID: PMC7589751 DOI: 10.3390/ijms21207660] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is an irreversible, age-related progressive neurological disorder, and the most common type of dementia in aged people. Neuropathological lesions of AD are neurofibrillary tangles (NFTs), and senile plaques comprise the accumulated amyloid-beta (Aβ), loaded with metal ions including Cu, Fe, or Zn. Some reports have identified metal dyshomeostasis as a neurotoxic factor of AD, among which Cu ions seem to be a central cationic metal in the formation of plaque and soluble oligomers, and have an essential role in the AD pathology. Cu-Aβ complex catalyzes the generation of reactive oxygen species (ROS) and results in oxidative damage. Several studies have indicated that oxidative stress plays a crucial role in the pathogenesis of AD. The connection of copper levels in AD is still ambiguous, as some researches indicate a Cu deficiency, while others show its higher content in AD, and therefore there is a need to increase and decrease its levels in animal models, respectively, to study which one is the cause. For more than twenty years, many in vitro studies have been devoted to identifying metals' roles in Aβ accumulation, oxidative damage, and neurotoxicity. Towards the end, a short review of the modern therapeutic approach in chelation therapy, with the main focus on Cu ions, is discussed. Despite the lack of strong proofs of clinical advantage so far, the conjecture that using a therapeutic metal chelator is an effective strategy for AD remains popular. However, some recent reports of genetic-regulating copper transporters in AD models have shed light on treating this refractory disease. This review aims to succinctly present a better understanding of Cu ions' current status in several AD features, and some conflicting reports are present herein.
Collapse
Affiliation(s)
- Hafza Wajeeha Ejaz
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, China;
| | - Wei Wang
- School of Medical and Health Sciences, Edith Cowan University, Perth WA6027, Australia;
| | - Minglin Lang
- CAS Center for Excellence in Biotic Interactions, College of Life Science, University of Chinese Academy of Sciences, Yuquan Road 19, Beijing 100049, China;
- College of Life Science, Agricultural University of Hebei, Baoding 071000, China
| |
Collapse
|
20
|
Burton LH, Radakovich LB, Marolf AJ, Santangelo KS. Systemic iron overload exacerbates osteoarthritis in the strain 13 guinea pig. Osteoarthritis Cartilage 2020; 28:1265-1275. [PMID: 32629162 PMCID: PMC7484276 DOI: 10.1016/j.joca.2020.06.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/05/2020] [Accepted: 06/16/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Iron is emerging as a key player in aging-associated diseases due to its propensity for driving free radical formation. Studies examining the role of iron in the pathogenesis of primary osteoarthritis (OA) are limited. Our objective was to establish a direct relationship between excess iron and OA by administering iron dextran to a guinea pig strain with decreased propensity for developing this disease. DESIGN Twenty, 12-week-old Strain 13 guinea pigs received either iron dextran or dextran control intraperitoneally once weekly for 4 weeks; termination occurred at 16 weeks of age. Iron levels were determined systemically (serum and liver) and within diarthrodial joints [femoral head articular cartilage and infrapatellar fat pads (IFPs) of knee joints]. One knee was collected to score structural changes associated with OA via microcomputed tomography (microCT) and histology using published grading schemes. Articular cartilage and IFPs were harvested from contralateral knees for gene expression analyses. RESULTS Iron overload was confirmed systemically via increased serum iron and liver iron concentration. Articular cartilage and IFPs in the iron dextran group also had higher levels of iron. Excess iron worsened knee OA using both microCT and histologic scoring systems. Gene analyses revealed that exogenous iron altered the expression of iron trafficking proteins, select cytokines, and structural components of cartilage. CONCLUSION These results demonstrate that systemic iron overload caused cellular iron accumulation in the knee joint. This excess iron is associated with increased expression of local inflammatory mediators and early onset and progression of knee joint OA in Strain 13 animals.
Collapse
Affiliation(s)
- Lindsey H. Burton
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Lauren B. Radakovich
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Angela J. Marolf
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Kelly S. Santangelo
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
21
|
Hackett MJ, Hollings A, Caine S, Bewer BE, Alaverdashvili M, Takechi R, Mamo JCL, Jones MWM, de Jonge MD, Paterson PG, Pickering IJ, George GN. Elemental characterisation of the pyramidal neuron layer within the rat and mouse hippocampus. Metallomics 2020; 11:151-165. [PMID: 30398510 DOI: 10.1039/c8mt00230d] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A unique combination of sensitivity, resolution, and penetration make X-ray fluorescence imaging (XFI) ideally suited to investigate trace elemental distributions in the biological context. XFI has gained widespread use as an analytical technique in the biological sciences, and in particular enables exciting new avenues of research in the field of neuroscience. In this study, elemental mapping by XFI was applied to characterise the elemental content within neuronal cell layers of hippocampal sub-regions of mice and rats. Although classical histochemical methods for metal detection exist, such approaches are typically limited to qualitative analysis. Specifically, histochemical methods are not uniformly sensitive to all chemical forms of a metal, often displaying variable sensitivity to specific "pools" or chemical forms of a metal. In addition, histochemical methods require fixation and extensive chemical treatment of samples, creating the strong likelihood for metal redistribution, leaching, or contamination. Direct quantitative elemental mapping of total elemental pools, in situ within ex vivo tissue sections, without the need for chemical fixation or addition of staining reagents is not possible with traditional histochemical methods; however, such a capability, which is provided by XFI, can offer an enormous analytical advantage. The results we report herein demonstrate the analytical advantage of XFI elemental mapping for direct, label-free metal quantification, in situ within ex vivo brain tissue sections. Specifically, we definitively characterise for the first time, the abundance of Fe within the pyramidal cell layers of the hippocampus. Localisation of Fe to this cell layer is not reproducibly achieved with classical Perls histochemical Fe stains. The ability of XFI to directly quantify neuronal elemental (P, S, Cl, K, Ca, Fe, Cu, Zn) distributions, revealed unique profiles of Fe and Zn within anatomical sub-regions of the hippocampus i.e., cornu ammonis 1, 2 or 3 (CA1, CA2 or CA3) sub-regions. Interestingly, our study reveals a unique Fe gradient across neuron populations within the non-degenerating and pathology free rat hippocampus, which curiously mirrors the pattern of region-specific vulnerability of the hippocampus that has previously been established to occur in various neurodegenerative diseases.
Collapse
Affiliation(s)
- M J Hackett
- Curtin Institute for Functional Molecules and Interfaces, School of Molecular and Life Sciences, Curtin University, GPOBox U1987, Bentley, WA 6845, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Rodriguez-Callejas JD, Fuchs E, Perez-Cruz C. Increased oxidative stress, hyperphosphorylation of tau, and dystrophic microglia in the hippocampus of aged Tupaia belangeri. Glia 2020; 68:1775-1793. [PMID: 32096580 DOI: 10.1002/glia.23804] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 12/26/2022]
Abstract
Aging is a major risk factor for the development of neurodegenerative diseases. Alzheimer's disease and other neurodegenerative diseases are characterized by abnormal and prominent protein aggregation in the brain, partially due to deficiency in protein clearance. It has been proposed that alterations in microglia phagocytosis and debris clearance hasten the onset of neurodegeneration. Dystrophic microglia are abundant in aged humans, and it has been associated with the onset of disease. Furthermore, alterations in microglia containing ferritin are associated with neurodegenerative conditions. To further understand the process of microglia dysfunction during the aging process, we used hippocampal sections from Tupaia belangeri (tree shrews). Adult (mean age 3.8 years), old (mean age 6 years), and aged (mean age 7.5 years) tree shrews were used for histochemical and immunostaining techniques to determine ferritin and Iba1 positive microglia, iron tissue content, tau hyperphosphorylation and oxidized-RNA in dentate gyrus, subiculum, and CA1-CA3 hippocampal regions. Our results indicated that aged tree shrews presented an increased number of activated microglia containing ferritin, but microglia labeled with Iba1 with a dystrophic phenotype was more abundant in aged individuals. With aging, oxidative damage to RNA (8OHG) increased significantly in all hippocampal regions, while tau hyperphosphorylation (AT100) was enhanced in DG, CA3, and SUB in aged animals. Phagocytic inclusions of 8OHG- and AT100-damaged cells were observed in activated M2 microglia in old and aged animals. These data indicate that aged tree shrew may be a suitable model for translational research to study brain and microglia alterations during the aging process.
Collapse
Affiliation(s)
| | - Eberhard Fuchs
- German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | | |
Collapse
|
23
|
Ring HL, Gao Z, Sharma A, Han Z, Lee C, Brockbank KGM, Greene ED, Helke KL, Chen Z, Campbell LH, Weegman B, Davis M, Taylor M, Giwa S, Fahy GM, Wowk B, Pagotan R, Bischof JC, Garwood M. Imaging the distribution of iron oxide nanoparticles in hypothermic perfused tissues. Magn Reson Med 2019; 83:1750-1759. [PMID: 31815324 DOI: 10.1002/mrm.28123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/01/2019] [Accepted: 11/20/2019] [Indexed: 12/23/2022]
Abstract
PURPOSE Herein, we evaluate the use of MRI as a tool for assessing iron oxide nanoparticle (IONP) distribution within IONP perfused organs and vascularized composite allografts (VCAs) (i.e., hindlimbs) prepared for cryopreservation. METHODS Magnetic resonance imaging was performed on room-temperature organs and VCAs perfused with IONPs and were assessed at 9.4 T. Quantitative T1 mapping and T 2 ∗ -weighted images were acquired using sweep imaging with Fourier transformation and gradient-echo sequences, respectively. Verification of IONP localization was performed through histological assessment and microcomputer tomography. RESULTS Quantitative imaging was achieved for organs and VCAs perfused with up to 642 mMFe (36 mgFe /mL), which is above previous demonstrations of upper limit detection in agarose (35.7mMFe [2 mgFe /mL]). The stability of IONPs in the perfusate had an effect on the quality of distribution and imaging within organs or VCA. Finally, MRI provided more accurate IONP localization than Prussian blue histological staining in this system, wherein IONPs remain primarily in the vasculature. CONCLUSION Using MRI, we were able to assess the distribution of IONPs throughout organs and VCAs varying in complexity. Additional studies are necessary to better understand this system and validate the calibration between T1 measurements and IONP concentration.
Collapse
Affiliation(s)
- Hattie L Ring
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minnesota
| | - Zhe Gao
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Anirudh Sharma
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Zonghu Han
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Charles Lee
- Department of Mechanical Engineering and Engineering Science, University of North Carolina, Charlotte, North Carolina
| | - Kelvin G M Brockbank
- Tissue Testing Technologies LLC, North Charleston.,Department of Bioengineering, Clemson University, Charleston, South Carolina.,Department of Comparative Medicine, Medical University of South Carolina, Charleston, South Carolina
| | | | - Kristi L Helke
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Zhen Chen
- Tissue Testing Technologies LLC, North Charleston
| | | | | | - Monica Davis
- Sylvatica Biotech, Inc., North Charleston, South Carolina
| | - Michael Taylor
- Sylvatica Biotech, Inc., North Charleston, South Carolina
| | - Sebastian Giwa
- Sylvatica Biotech, Inc., North Charleston, South Carolina
| | | | - Brian Wowk
- 21st Century Medicine, Inc., Fontana, California
| | | | - John C Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Michael Garwood
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
24
|
Rodríguez-Callejas JDD, Cuervo-Zanatta D, Rosas-Arellano A, Fonta C, Fuchs E, Perez-Cruz C. Loss of ferritin-positive microglia relates to increased iron, RNA oxidation, and dystrophic microglia in the brains of aged male marmosets. Am J Primatol 2019; 81:e22956. [DOI: 10.1002/ajp.22956] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 01/09/2019] [Accepted: 01/12/2019] [Indexed: 12/26/2022]
Affiliation(s)
| | - Daniel Cuervo-Zanatta
- Department of Pharmacology; Center of Research in Advance Studies; Mexico City Mexico
| | | | - Caroline Fonta
- Brain and Cognition Research Centre (CERCO); CNRS/University of Toulouse; Toulouse France
| | - Eberhard Fuchs
- German Primate Center; Leibniz Institute for Primate Research; Göttingen Germany
| | - Claudia Perez-Cruz
- Department of Pharmacology; Center of Research in Advance Studies; Mexico City Mexico
| |
Collapse
|
25
|
Dekens DW, Naudé PJW, Keijser JN, Boerema AS, De Deyn PP, Eisel ULM. Lipocalin 2 contributes to brain iron dysregulation but does not affect cognition, plaque load, and glial activation in the J20 Alzheimer mouse model. J Neuroinflammation 2018; 15:330. [PMID: 30501637 PMCID: PMC6267886 DOI: 10.1186/s12974-018-1372-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/18/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Lipocalin 2 (Lcn2) is an acute-phase protein implicated in multiple neurodegenerative conditions. Interestingly, both neuroprotective and neurodegenerative effects have been described for Lcn2. Increased Lcn2 levels were found in human post-mortem Alzheimer (AD) brain tissue, and in vitro studies indicated that Lcn2 aggravates amyloid-β-induced toxicity. However, the role of Lcn2 has not been studied in an in vivo AD model. Therefore, in the current study, the effects of Lcn2 were studied in the J20 mouse model of AD. METHODS J20 mice and Lcn2-deficient J20 (J20xLcn2 KO) mice were compared at the behavioral and neuropathological level. RESULTS J20xLcn2 KO and J20 mice presented equally strong AD-like behavioral changes, cognitive impairment, plaque load, and glial activation. Interestingly, hippocampal iron accumulation was significantly decreased in J20xLcn2 KO mice as compared to J20 mice. CONCLUSIONS Lcn2 contributes to AD-like brain iron dysregulation, and future research should further explore the importance of Lcn2 in AD.
Collapse
Affiliation(s)
- Doortje W. Dekens
- Department of Neurology and Alzheimer Research Center, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9713 GZ The Netherlands
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, Groningen, 9747 AG The Netherlands
| | - Petrus J. W. Naudé
- Department of Neurology and Alzheimer Research Center, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9713 GZ The Netherlands
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, Groningen, 9747 AG The Netherlands
| | - Jan N. Keijser
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, Groningen, 9747 AG The Netherlands
| | - Ate S. Boerema
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, Groningen, 9747 AG The Netherlands
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9713 GZ The Netherlands
| | - Peter P. De Deyn
- Department of Neurology and Alzheimer Research Center, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9713 GZ The Netherlands
- Laboratory of Neurochemistry and Behavior, Biobank, Institute Born-Bunge, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Ulrich L. M. Eisel
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, Groningen, 9747 AG The Netherlands
- University Center of Psychiatry & Interdisciplinary Center of Psychopathology of Emotion Regulation, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9713 GZ The Netherlands
| |
Collapse
|
26
|
Nixon AM, Meadowcroft MD, Neely EB, Snyder AM, Purnell CJ, Wright J, Lamendella R, Nandar W, Huang X, Connor JR. HFE Genotype Restricts the Response to Paraquat in a Mouse Model of Neurotoxicity. J Neurochem 2018; 145:299-311. [PMID: 29315562 DOI: 10.1111/jnc.14299] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/05/2017] [Accepted: 01/03/2018] [Indexed: 12/30/2022]
Abstract
Parkinson's disease is marked clinically by motor dysfunction and pathologically by dopaminergic cell loss in the substantia nigra and iron accumulation in the substantia nigra. The driver underlying iron accumulation remains unknown and could be genetic or environmental. The HFE protein is critical for the regulation of cellular iron uptake. Mutations within this protein are associated with increased iron accumulation including in the brain. We have focused on the commonly occurring H63D variant of the HFE gene as a disease modifier in a number of neurodegenerative diseases. To investigate the role of H63D HFE genotype, we generated a mouse model in which the wild-type (WT) HFE gene is replaced by the H67D gene variant (mouse homolog of the human H63D gene variant). Using paraquat toxicity as the model for Parkinson's disease, we found that WT mice responded as expected with significantly greater motor function, loss of tyrosine hydroxylase staining and increase microglial staining in the substantia nigra, and an increase in R2 relaxation rate within the substantia nigra of the paraquat-treated mice compared to their saline-treated counterparts. In contrast, the H67D mice showed a remarkable resistance to paraquat treatment; specifically differing from the WT mice with no changes in motor function or changes in R2 relaxation rates following paraquat exposure. At baseline, there were differences between the H67D HFE mice and WT mice in gut microbiome profile and increased L-ferritin staining in the substantia nigra that could account for the resistance to paraquat. Of particular note, the H67D HFE mice regardless of whether or not they were treated with paraquat had significantly less tyrosine hydroxylase immunostaining than WT. Our results clearly demonstrate that the HFE genotype impacts the expression of tyrosine hydroxylase in the substantia nigra, the gut microbiome and the response to paraquat providing additional support that the HFE genotype is a disease modifier for Parkinson's disease. Moreover, the finding that the HFE mutant mice are resistant to paraquat may provide a model in which to study resistant mechanisms to neurotoxicants.
Collapse
Affiliation(s)
- Anne M Nixon
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Mark D Meadowcroft
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Radiology, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Elizabeth B Neely
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Amanda M Snyder
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Carson J Purnell
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | | | - Regina Lamendella
- Wright Labs, Huntingdon, Pennsylvania, USA
- Department of Microbiology, Juniata College, Huntingdon, Pennsylvania, USA
| | - Wint Nandar
- Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xuemei Huang
- Department of Neurology, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - James R Connor
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| |
Collapse
|
27
|
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: 4.6] [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
|
28
|
Geldenhuys WJ, Benkovic SA, Lin L, Yonutas HM, Crish SD, Sullivan PG, Darvesh AS, Brown CM, Richardson JR. MitoNEET (CISD1) Knockout Mice Show Signs of Striatal Mitochondrial Dysfunction and a Parkinson's Disease Phenotype. ACS Chem Neurosci 2017; 8:2759-2765. [PMID: 28880525 DOI: 10.1021/acschemneuro.7b00287] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial dysfunction is thought to play a significant role in neurodegeneration observed in Parkinson's disease (PD), yet the mechanisms underlying this pathology remain unclear. Here, we demonstrate that loss of mitoNEET (CISD1), an iron-sulfur containing protein that regulates mitochondrial bioenergetics, results in mitochondrial dysfunction and loss of striatal dopamine and tyrosine hydroxylase. Mitochondria isolated from mice lacking mitoNEET were dysfunctional as revealed by elevated reactive oxygen species (ROS) and reduced capacity to produce ATP. Gait analysis revealed a shortened stride length and decreased rotarod performance in knockout mice, consistent with the loss of striatal dopamine. Together, these data suggest that mitoNEET KO mice exhibit many of the characteristics of early neurodegeneration in PD and may provide a novel drug discovery platform to evaluate compounds for enhancing mitochondrial function in neurodegenerative disorders.
Collapse
Affiliation(s)
- Werner J. Geldenhuys
- Department
of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Stanley A. Benkovic
- Department
of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Li Lin
- Department
of Pharmaceutical Sciences and Center for Neurodegenerative Disease
and Aging, Northeast Ohio Medical University, College of Pharmacy, Rootstown, Ohio 44272, United States
| | - Heather M. Yonutas
- Department
of Neuroscience; University of Kentucky Chandler College of Medicine, Lexington, Kentucky 40536, United States
| | - Samuel D. Crish
- Department
of Pharmaceutical Sciences and Center for Neurodegenerative Disease
and Aging, Northeast Ohio Medical University, College of Pharmacy, Rootstown, Ohio 44272, United States
| | - Patrick G. Sullivan
- Department
of Neuroscience; University of Kentucky Chandler College of Medicine, Lexington, Kentucky 40536, United States
| | - Altaf S. Darvesh
- Department
of Pharmaceutical Sciences and Center for Neurodegenerative Disease
and Aging, Northeast Ohio Medical University, College of Pharmacy, Rootstown, Ohio 44272, United States
| | - Candice M. Brown
- Department
of Microbiology, Immunology, and Cell Biology; School of Medicine, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Jason R. Richardson
- Department
of Pharmaceutical Sciences and Center for Neurodegenerative Disease
and Aging, Northeast Ohio Medical University, College of Pharmacy, Rootstown, Ohio 44272, United States
| |
Collapse
|
29
|
Peters DG, Purnell CJ, Haaf MP, Yang QX, Connor JR, Meadowcroft MD. Dietary lipophilic iron accelerates regional brain iron-load in C57BL6 mice. Brain Struct Funct 2017; 223:1519-1536. [DOI: 10.1007/s00429-017-1565-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/07/2017] [Indexed: 11/29/2022]
|
30
|
Li DD, Zhang W, Wang ZY, Zhao P. Serum Copper, Zinc, and Iron Levels in Patients with Alzheimer's Disease: A Meta-Analysis of Case-Control Studies. Front Aging Neurosci 2017; 9:300. [PMID: 28966592 PMCID: PMC5605551 DOI: 10.3389/fnagi.2017.00300] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 08/31/2017] [Indexed: 11/13/2022] Open
Abstract
Background: Many publications have investigated the association between metal ions and the risk of Alzheimer's disease (AD), but the results were ambiguous. Aims: The objective of this study was to assess the association between the serum levels of metals (copper/zinc/iron) and the risk of AD via meta-analysis of case-control studies. Methods: We screened literatures published after 1978 in the Pubmed, Embase, Cochrane library, Web of Science and ClinicalTrials.gov. Electronic databases. By using Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines, we performed a systematic review of the 407 publications, there are 44 of these publications met all inclusion criteria. The Review Manager 5.3 software was used to calculate available data from each study. Results: Consistent with the conclusions of other meta-analysis, our results demonstrated serum copper levels were significantly higher [MD = 9.27, 95% CI (5.02–13.52); p < 0.0001], and the serum zinc levels were significantly lower in AD patients than in healthy controls [MD = −6.12, 95% CI (−9.55, −2.69); p = 0.0005]. Serum iron levels were significantly lower in AD patients than in healthy controls after excluded two studies [MD = −13.01, 95% CI (−20.75, −5.27); p = 0.001]. Conclusion: The results of our meta-analysis provided rigorous statistical support for the association of the serum levels of metals and the risk of AD, suggesting a positive relationship between the serum copper levels and AD risk, and a negative relationship between the serum zinc/iron levels and AD risk.
Collapse
Affiliation(s)
- Dan-Dan Li
- College of Life and Health Sciences, Northeastern UniversityShenyang, China
| | - Wei Zhang
- Department of Hepatobiliary Surgery, General Hospital of Shenyang Military Area CommandShenyang, China
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern UniversityShenyang, China
| | - Pu Zhao
- College of Life and Health Sciences, Northeastern UniversityShenyang, China
| |
Collapse
|
31
|
Dietrich P, Johnson IM, Alli S, Dragatsis I. Elimination of huntingtin in the adult mouse leads to progressive behavioral deficits, bilateral thalamic calcification, and altered brain iron homeostasis. PLoS Genet 2017; 13:e1006846. [PMID: 28715425 PMCID: PMC5536499 DOI: 10.1371/journal.pgen.1006846] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 07/31/2017] [Accepted: 06/01/2017] [Indexed: 02/07/2023] Open
Abstract
Huntington's Disease (HD) is an autosomal dominant progressive neurodegenerative disorder characterized by cognitive, behavioral and motor dysfunctions. HD is caused by a CAG repeat expansion in exon 1 of the HD gene that is translated into an expanded polyglutamine tract in the encoded protein, huntingtin (HTT). While the most significant neuropathology of HD occurs in the striatum, other brain regions are also affected and play an important role in HD pathology. To date there is no cure for HD, and recently strategies aiming at silencing HTT expression have been initiated as possible therapeutics for HD. However, the essential functions of HTT in the adult brain are currently unknown and hence the consequence of sustained suppression of HTT expression is unpredictable and can potentially be deleterious. Using the Cre-loxP system of recombination, we conditionally inactivated the mouse HD gene homologue at 3, 6 and 9 months of age. Here we show that elimination of Htt expression in the adult mouse results in behavioral deficits, progressive neuropathological changes including bilateral thalamic calcification, and altered brain iron homeostasis.
Collapse
Affiliation(s)
- Paula Dietrich
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, Tennessee, United States of America
| | - Irudayam Maria Johnson
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, Tennessee, United States of America
| | - Shanta Alli
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, Tennessee, United States of America
| | - Ioannis Dragatsis
- Department of Physiology, The University of Tennessee, Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|