1
|
Ishijima T, Nakajima K. Mechanisms of Microglia Proliferation in a Rat Model of Facial Nerve Anatomy. BIOLOGY 2023; 12:1121. [PMID: 37627005 PMCID: PMC10452325 DOI: 10.3390/biology12081121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/27/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023]
Abstract
Although microglia exist as a minor glial cell type in the normal state of the brain, they increase in number in response to various disorders and insults. However, it remains unclear whether microglia proliferate in the affected area, and the mechanism of the proliferation has long attracted the attention of researchers. We analyzed microglial mitosis using a facial nerve transection model in which the blood-brain barrier is left unimpaired when the nerves are axotomized. Our results showed that the levels of macrophage colony-stimulating factor (M-CSF), cFms (the receptor for M-CSF), cyclin A/D, and proliferating cell nuclear antigen (PCNA) were increased in microglia in the axotomized facial nucleus (axotFN). In vitro experiments revealed that M-CSF induced cFms, cyclin A/D, and PCNA in microglia, suggesting that microglia proliferate in response to M-CSF in vivo. In addition, M-CSF caused the activation of c-Jun N-terminal kinase (JNK) and p38, and the specific inhibitors of JNK and p38 arrested the microglial mitosis. JNK and p38 were shown to play roles in the induction of cyclins/PCNA and cFms, respectively. cFms was suggested to be induced through a signaling cascade of p38-mitogen- and stress-activated kinase-1 (MSK1)-cAMP-responsive element binding protein (CREB) and/or p38-activating transcription factor 2 (ATF2). Microglia proliferating in the axotFN are anticipated to serve as neuroprotective cells by supplying neurotrophic factors and/or scavenging excite toxins and reactive oxygen radicals.
Collapse
Affiliation(s)
- Takashi Ishijima
- Graduate School of Science and Engineering, Soka University, Tokyo 192-8577, Japan;
| | - Kazuyuki Nakajima
- Graduate School of Science and Engineering, Soka University, Tokyo 192-8577, Japan;
- Glycan & Life Systems Integration Center, Soka University, Tokyo 192-8577, Japan
| |
Collapse
|
2
|
Changes of signaling molecules in the axotomized rat facial nucleus. J Chem Neuroanat 2022; 126:102179. [DOI: 10.1016/j.jchemneu.2022.102179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 12/15/2022]
|
3
|
Baldacchino K, Peveler WJ, Lemgruber L, Smith RS, Scharler C, Hayden L, Komarek L, Lindsay SL, Barnett SC, Edgar JM, Linington C, Thümmler K. Myelinated axons are the primary target of hemin-mediated oxidative damage in a model of the central nervous system. Exp Neurol 2022; 354:114113. [PMID: 35569511 DOI: 10.1016/j.expneurol.2022.114113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/29/2022] [Accepted: 05/08/2022] [Indexed: 12/01/2022]
Abstract
Iron released from oligodendrocytes during demyelination or derived from haemoglobin breakdown products is believed to amplify oxidative tissue injury in multiple sclerosis (MS). However, the pathophysiological significance of iron-containing haemoglobin breakdown products themselves is rarely considered in the context of MS and their cellular specificity and mode of action remain unclear. Using myelinating cell cultures, we now report the cytotoxic potential of hemin (ferriprotoporphyrin IX chloride), a major degradation product of haemoglobin, is 25-fold greater than equimolar concentrations of free iron in myelinating cultures; a model that reproduces the complex multicellular environment of the CNS. At low micro molar concentrations (3.3 - 10 μM) we observed hemin preferentially binds to myelin and axons to initiate a complex detrimental response that results in targeted demyelination and axonal loss but spares neuronal cell bodies, astrocytes and the majority of oligodendroglia. Demyelination and axonal loss in this context are executed by a combination of mechanisms that include iron-dependent peroxidation by reactive oxygen species (ROS) and ferroptosis. These effects are microglial-independent, do not require any initiating inflammatory insult and represent a direct effect that compromises the structural integrity of myelinated axons in the CNS. Our data identify hemin-mediated demyelination and axonal loss as a novel mechanism by which intracerebral degradation of haemoglobin may contribute to lesion development in MS.
Collapse
Affiliation(s)
- Karl Baldacchino
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - William J Peveler
- WestCHEM, School of Chemistry, University of Glasgow, Joseph Black Building, G12 8QQ Glasgow, UK
| | - Leandro Lemgruber
- Glasgow Imaging Facility, Institute of Infection, Immunity and Inflammation, University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
| | - Rebecca Sherrard Smith
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Cornelia Scharler
- Institute of Experimental and Clinical Cell Therapy, Paracelsus Medical University, Salzburg, Austria
| | - Lorna Hayden
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Lina Komarek
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Susan L Lindsay
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Susan C Barnett
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Julia M Edgar
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Christopher Linington
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom
| | - Katja Thümmler
- Institute of Infection, Immunity and Inflammation, University of Glasgow, G12 8TA Glasgow, United Kingdom.
| |
Collapse
|
4
|
Yamamoto S, Matsui A, Ohyagi M, Kikutake C, Harada Y, Iizuka-Koga M, Suyama M, Yoshimura A, Ito M. In Vitro Generation of Brain Regulatory T Cells by Co-culturing With Astrocytes. Front Immunol 2022; 13:960036. [PMID: 35911740 PMCID: PMC9335882 DOI: 10.3389/fimmu.2022.960036] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022] Open
Abstract
Regulatory T cells (Tregs) are normally born in the thymus and activated in secondary lymphoid tissues to suppress immune responses in the lymph node and at sites of inflammation. Tregs are also resident in various tissues or accumulate in damaged tissues, which are now called tissue Tregs, and contribute to homeostasis and tissue repair by interacting with non-immune cells. We have shown that Tregs accumulate in the brain during the chronic phase in a mouse cerebral infarction model, and these Tregs acquire the characteristic properties of brain Tregs and contribute to the recovery of neurological damage by interacting with astrocytes. However, the mechanism of tissue Treg development is not fully understood. We developed a culture method that confers brain Treg characteristics in vitro. Naive Tregs from the spleen were activated and efficiently amplified by T-cell receptor (TCR) stimulation in the presence of primary astrocytes. Furthermore, adding IL-33 and serotonin could confer part of the properties of brain Tregs, such as ST2, peroxisome proliferator-activated receptor γ (PPARγ), and serotonin receptor 7 (Htr7) expression. Transcriptome analysis revealed that in vitro generated brain Treg-like Tregs (induced brain Tregs; iB-Tregs) showed similar gene expression patterns as those in in vivo brain Tregs, although they were not identical. Furthermore, in Parkinson’s disease models, in which T cells have been shown to be involved in disease progression, iB-Tregs infiltrated into the brain more readily and ameliorated pathological symptoms more effectively than splenic Tregs. These data indicate that iB-Tregs contribute to our understanding of brain Treg development and could also be therapeutic for inflammatory brain diseases.
Collapse
Affiliation(s)
- Shinichi Yamamoto
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Ako Matsui
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Masaki Ohyagi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Chie Kikutake
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Harada
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Mana Iizuka-Koga
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Mikita Suyama
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Minako Ito
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- *Correspondence: Minako Ito,
| |
Collapse
|
5
|
David S, Jhelum P, Ryan F, Jeong SY, Kroner A. Dysregulation of Iron Homeostasis in the Central Nervous System and the Role of Ferroptosis in Neurodegenerative Disorders. Antioxid Redox Signal 2022; 37:150-170. [PMID: 34569265 DOI: 10.1089/ars.2021.0218] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significance: Iron accumulation occurs in the central nervous system (CNS) in a variety of neurological conditions as diverse as spinal cord injury, stroke, multiple sclerosis, Parkinson's disease, and others. Iron is a redox-active metal that gives rise to damaging free radicals if its intracellular levels are not controlled or if it is not properly sequestered within cells. The accumulation of iron occurs due to dysregulation of mechanisms that control cellular iron homeostasis. Recent Advances: The molecular mechanisms that regulate cellular iron homeostasis have been revealed in much detail in the past three decades, and new advances continue to be made. Understanding which aspects of iron homeostasis are dysregulated in different conditions will provide insights into the causes of iron accumulation and iron-mediated tissue damage. Recent advances in iron-dependent lipid peroxidation leading to cell death, called ferroptosis, has provided useful insights that are highly relevant for the lipid-rich environment of the CNS. Critical Issues: This review examines the mechanisms that control normal cellular iron homeostasis, the dysregulation of these mechanisms in neurological disorders, and more recent work on how iron can induce tissue damage via ferroptosis. Future Directions: Quick and reliable tests are needed to determine if and when ferroptosis contributes to the pathogenesis of neurological disorders. In addition, there is need to develop better druggable agents to scavenge lipid radicals and reduce CNS damage for neurological conditions for which there are currently few effective treatments. Antioxid. Redox Signal. 37, 150-170.
Collapse
Affiliation(s)
- Samuel David
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Priya Jhelum
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Fari Ryan
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Suh Young Jeong
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Antje Kroner
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
6
|
Events Occurring in the Axotomized Facial Nucleus. Cells 2022; 11:cells11132068. [PMID: 35805151 PMCID: PMC9266054 DOI: 10.3390/cells11132068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 01/27/2023] Open
Abstract
Transection of the rat facial nerve leads to a variety of alterations not only in motoneurons, but also in glial cells and inhibitory neurons in the ipsilateral facial nucleus. In injured motoneurons, the levels of energy metabolism-related molecules are elevated, while those of neurofunction-related molecules are decreased. In tandem with these motoneuron changes, microglia are activated and start to proliferate around injured motoneurons, and astrocytes become activated for a long period without mitosis. Inhibitory GABAergic neurons reduce the levels of neurofunction-related molecules. These facts indicate that injured motoneurons somehow closely interact with glial cells and inhibitory neurons. At the same time, these events allow us to predict the occurrence of tissue remodeling in the axotomized facial nucleus. This review summarizes the events occurring in the axotomized facial nucleus and the cellular and molecular mechanisms associated with each event.
Collapse
|
7
|
Bi A, Wang Y, Chen L, Yin Z, Luo L. γ-Glutamylcysteine attenuates amyloid-β oligomers-induced neuroinflammation in microglia via blocking NF-κB signaling pathway. Chem Biol Interact 2022; 363:110019. [PMID: 35714925 DOI: 10.1016/j.cbi.2022.110019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 05/30/2022] [Accepted: 06/10/2022] [Indexed: 11/03/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurogenerative disease, characterized by progressive memory loss and cognitive deficits. Intracellular neurofibrillary tangles (NFTs) and amyloid-β (Aβ)-formed neuritic plaques are major pathological features of AD. Aβ evokes activation of microglia to release inflammatory mediators and ROS to induce neurotoxicity, leading to neurodegeneration. γ-Glutamylcysteine (γ-GC), an intermediate dipeptide of the GSH-synthesis pathway and possessing anti-inflammatory and anti-oxidative properties, represents a relatively unexplored option for AD treatment. In the present study, we investigated the anti-inflammatory effect of γ-GC on Aβ oligomer (AβO)-induced neuroinflammation and the associated molecular mechanism in microglia. The results showed that γ-GC reduced AβO-induced release of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and nitric oxide (NO), and the expression of inducible NO synthase (iNOS) and cyclooxygenase 2 (COX-2). γ-GC decreased ROS and MDA production and increased the GSH level, GSH/GSSG ratio, and SOD activity in AβO-treated microglia. Mechanistically, γ-GC inhibited activation of nuclear factor kappa B (NF-κB), and upregulated the nuclear receptor-related 1 (Nurr1) protein expression to suppress the transcriptional effect of NF-κB on the inflammatory genes. Besides, γ-GC suppressed the AβO-induced neuroinflammation in mice. These findings suggested that γ-GC might represent a potential therapeutic agent for anti-neuroinflammation.
Collapse
Affiliation(s)
- Aijing Bi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Yanan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Luyao Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, Jiangsu, China.
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
8
|
Ishijima T, Nakajima K. Inflammatory cytokines TNFα, IL-1β, and IL-6 are induced in endotoxin- stimulated microglia through different signaling cascades. Sci Prog 2021; 104:368504211054985. [PMID: 34821182 PMCID: PMC10450609 DOI: 10.1177/00368504211054985] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
By using an animal model in which inflammatory cytokines are induced in lipopolysaccharide (LPS)-injected rat brain, we investigated the induction of tumor necrosis factor alpha (TNFα), interleukin-1beta (IL-1β), and IL-6. Immunoblotting and immunohistochemistry revealed that all three cytokines were transiently induced in the cerebral cortex at about 12 h after LPS injection. To clarify which glial cell type induced the cytokines, we examined the respective abilities of astrocytes and microglia in vitro. Primary microglia largely induced TNFα, IL-1β and IL-6 in response to LPS, but primary astrocytes induced only limited levels of TNFα. Thus, we used specific inhibitors to focus on microglia in surveying signaling molecules involved in the induction of TNFα, IL-1β, and IL-6. The experiments using mitogen-activated protein kinases (MAPK) inhibitors revealed that c-Jun N-terminal kinase (JNK)/p38, external signal regulated kinase (ERK)/JNK, and ERK/JNK/p38 are necessary for the induction of TNFα, IL-1β, and IL-6, respectively. The experiments using protein kinase C (PKC) inhibitor clarified that PKCα is required for the induction of all these cytokines in LPS-stimulated microglia. Furthermore, LPS-dependent IL-1β/IL-6 induction was suppressed by pretreatment with a nitric oxide (NO) scavenger, suggesting that NO is involved in the signaling cascade of IL-1β/IL-6 induction. Thus, an inducible NO synthase induced in the LPS-injected cerebral cortex might be related to the induction of IL-1β/IL-6 through the production of NO in vivo. Taken together, these results demonstrated that microglia induce different kinds of inflammatory cytokine through specific combinations of MAPKs and by the presence or absence of NO.
Collapse
Affiliation(s)
- Takashi Ishijima
- Department of Science and Engineering for Sustainable Innovation, Faculty of Science and Engineering, Glycan & Life Systems Integration Center, Soka University, Tokyo, Japan
| | - Kazuyuki Nakajima
- Department of Science and Engineering for Sustainable Innovation, Faculty of Science and Engineering, Glycan & Life Systems Integration Center, Soka University, Tokyo, Japan
| |
Collapse
|
9
|
An X, Shi X. Effects of electroconvulsive shock on neuro-immune responses: Does neuro-damage occur? Psychiatry Res 2020; 292:113289. [PMID: 32702550 DOI: 10.1016/j.psychres.2020.113289] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 01/07/2023]
Abstract
Electroconvulsive therapy (ECT) is one of the most effective treatments for treatment-resistant depression. However, this treatment may produce memory impairment. The mechanisms of the cognitive adverse effects are not known. Neuroimmune response is related to the cognitive deficits. By reviewing the available animal literature, we examined the glia activation, inflammatory cytokines, neuron oxidative stress responses, and neural morphological changes following electroconvulsive shock (ECS) treatment. The studies showed that ECS activates microglia, upregulates neuro-inflammatory cytokines, and increases oxidative stress responses. But these effects are rapid and may be transient. They normalize as ECS treatment continues, suggesting endogenous neuroprotection may be mobilized. The transient changes are well in line with the clinical observations that ECT usually does not cause significant long-lasting retrograde amnesia. The longitudinal studies will be particularly important to explore the dynamic changes of neuroplasticity following ECT (Jonckheere et al., 2018). Investigating the neuroplasticity changes in animals that suffered chronic stress may also be crucial to giving support to the translation of preclinical research.
Collapse
Affiliation(s)
- Xianli An
- School of Educational Science, Yangzhou University, Yangzhou, JiangSu Province, China.
| | - Xiujian Shi
- School of Educational Science, Yangzhou University, Yangzhou, JiangSu Province, China
| |
Collapse
|
10
|
Askri D, Cunin V, Béal D, Berthier S, Chovelon B, Arnaud J, Rachidi W, Sakly M, Amara S, Sève M, Lehmann SG. Investigating the toxic effects induced by iron oxide nanoparticles on neuroblastoma cell line: an integrative study combining cytotoxic, genotoxic and proteomic tools. Nanotoxicology 2019; 13:1021-1040. [PMID: 31132913 DOI: 10.1080/17435390.2019.1621399] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Nanomaterials have gained much attention for their use and benefit in several fields. Iron Oxide Nanoparticles (IONPs) have been used in Biomedicine as contrast agents for imaging cancer cells. However, several studies reported the potential toxicity of those nanoparticles in different models, especially in cells. Therefore, in our present study, we investigated the effects of IONPs on the SH-SY5Y neuroblastoma cell line. We carried out cytotoxic and genotoxic studies to evaluate the phenotypic effects, and proteomic investigation to evaluate the molecular effects and the mechanisms by which this kind of NPs could induce toxicity. Our results showed that the use of three different sizes of IONPs (14, 22 and 30 nm) induced cell detachment, cell morphological changes, size, and concentration-dependent IONP internalization and cell mortality. IONPs induced slight genotoxic damage assayed by modified comet assay without affecting cell cycle, mitochondrial function, membrane integrity, intracellular calcium level, and without inducing ROS generation. All the studies were performed to compare also the effects of IONPs to the ferric iron by incubating cells with equivalent concentration of FeCl3. In all tests, the NPs exhibited more toxicity than the ferric iron. The proteomic analysis followed by gene ontology and pathway analysis evidenced the effects of IONPs on cytoskeleton, cell apoptosis, and cancer development. Our findings provided more information about IONP effects on human cells and especially on cancer cell line.
Collapse
Affiliation(s)
- Dalel Askri
- PROMETHEE Proteomic Platform, BEeSy, Grenoble Alpes University , Grenoble , France.,LBFA Inserm U1055, PROMETHEE Proteomic Platform , Grenoble , France.,CHU de Grenoble Alpes, Institut de Biologie et Pathologie , Grenoble, France.,Unit of Research in Integrated Physiology, College of Sciences of Bizerte, Carthage University , Bizerte , Tunisia
| | - Valérie Cunin
- PROMETHEE Proteomic Platform, BEeSy, Grenoble Alpes University , Grenoble , France.,LBFA Inserm U1055, PROMETHEE Proteomic Platform , Grenoble , France.,CHU de Grenoble Alpes, Institut de Biologie et Pathologie , Grenoble, France
| | - David Béal
- SyMMES/CIBEST UMR 5819 UGA-CNRS-CEA, INAC/CEA-Grenoble LAN, University Grenoble Alpes , Grenoble , France
| | - Sylvie Berthier
- Cytometry Platform, Pole Biology, University Grenoble Alpes , Grenoble , France
| | - Benoit Chovelon
- CHU de Grenoble Alpes, Institut de Biologie et Pathologie , Grenoble, France.,DPM UMR 5063, University Grenoble Alpes , Grenoble , France
| | - Josiane Arnaud
- LBFA Inserm U1055, PROMETHEE Proteomic Platform , Grenoble , France.,CHU de Grenoble Alpes, Institut de Biologie et Pathologie , Grenoble, France
| | - Walid Rachidi
- SyMMES/CIBEST UMR 5819 UGA-CNRS-CEA, INAC/CEA-Grenoble LAN, University Grenoble Alpes , Grenoble , France
| | - Mohsen Sakly
- Unit of Research in Integrated Physiology, College of Sciences of Bizerte, Carthage University , Bizerte , Tunisia
| | - Salem Amara
- Unit of Research in Integrated Physiology, College of Sciences of Bizerte, Carthage University , Bizerte , Tunisia
| | - Michel Sève
- PROMETHEE Proteomic Platform, BEeSy, Grenoble Alpes University , Grenoble , France.,LBFA Inserm U1055, PROMETHEE Proteomic Platform , Grenoble , France.,CHU de Grenoble Alpes, Institut de Biologie et Pathologie , Grenoble, France
| | - Sylvia G Lehmann
- PROMETHEE Proteomic Platform, BEeSy, Grenoble Alpes University , Grenoble , France.,LBFA Inserm U1055, PROMETHEE Proteomic Platform , Grenoble , France.,CHU de Grenoble Alpes, Institut de Biologie et Pathologie , Grenoble, France.,CNRS, IRD, IFSTTAR, ISTerre, University Grenoble Alpes , Grenoble , France.,CNRS, IRD, IFSTTAR, ISTerre, University Savoie Mont Blanc , Grenoble , France
| |
Collapse
|
11
|
Ndayisaba A, Kaindlstorfer C, Wenning GK. Iron in Neurodegeneration - Cause or Consequence? Front Neurosci 2019; 13:180. [PMID: 30881284 PMCID: PMC6405645 DOI: 10.3389/fnins.2019.00180] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/14/2019] [Indexed: 12/12/2022] Open
Abstract
Iron dyshomeostasis can cause neuronal damage to iron-sensitive brain regions. Neurodegeneration with brain iron accumulation reflects a group of disorders caused by iron overload in the basal ganglia. High iron levels and iron related pathogenic triggers have also been implicated in sporadic neurodegenerative diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), and multiple system atrophy (MSA). Iron-induced dyshomeostasis within vulnerable brain regions is still insufficiently understood. Here, we summarize the modes of action by which iron might act as primary or secondary disease trigger in neurodegenerative disorders. In addition, available treatment options targeting brain iron dysregulation and the use of iron as biomarker in prodromal stages are critically discussed to address the question of cause or consequence.
Collapse
Affiliation(s)
- Alain Ndayisaba
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Gregor K Wenning
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
12
|
Abstract
The key molecular events that provoke Parkinson's disease (PD) are not fully understood. Iron deposit was found in the substantia nigra pars compacta (SNpc) of PD patients and animal models, where dopaminergic neurons degeneration occurred selectively. The mechanisms involved in disturbed iron metabolism remain unknown, however, considerable evidence indicates that iron transporters dysregulation, activation of L-type voltage-gated calcium channel (LTCC) and ATP-sensitive potassium (KATP) channels, as well as N-methyl-D-aspartate (NMDA) receptors (NMDARs) contribute to this process. There is emerging evidence on the structural links and functional modulations between iron and α-synuclein, and the key player in PD which aggregates in Lewy bodies. Iron is believed to modulate α-synuclein synthesis, post-translational modification, and aggregation. Furthermore, glia, especially activated astroglia and microglia, are involved in iron deposit in PD. Glial contributions were largely dependent on the factors they released, e.g., neurotrophic factors, pro-inflammatory factors, lactoferrin, and those undetermined. Therefore, iron chelation using iron chelators, the extracts from many natural foods with iron chelating properties, may be an effective therapy for prevention and treatment of the disease.
Collapse
|
13
|
Ranjit S, Patters BJ, Gerth KA, Haque S, Choudhary S, Kumar S. Potential neuroprotective role of astroglial exosomes against smoking-induced oxidative stress and HIV-1 replication in the central nervous system. Expert Opin Ther Targets 2018; 22:703-714. [PMID: 30015535 DOI: 10.1080/14728222.2018.1501473] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION HIV-1-infected smokers are at risk of oxidative damage to neuronal cells in the central nervous system by both HIV-1 and cigarette smoke. Since neurons have a weak antioxidant defense system, they mostly depend on glial cells, particularly astrocytes, for protection against oxidative damage and neurotoxicity. Astrocytes augment the neuronal antioxidant system by supplying cysteine-containing products for glutathione synthesis, antioxidant enzymes such as SOD and catalase, glucose for antioxidant regeneration via the pentose-phosphate pathway, and by recycling of ascorbic acid. Areas covered: The transport of antioxidants and energy substrates from astrocytes to neurons could possibly occur via extracellular nanovesicles called exosomes. This review highlights the neuroprotective potential of exosomes derived from astrocytes against smoking-induced oxidative stress, HIV-1 replication, and subsequent neurotoxicity observed in HIV-1-positive smokers. Expert opinion: During stress conditions, the antioxidants released from astrocytes either via extracellular fluid or exosomes to neurons may not be sufficient to provide neuroprotection. Therefore, we put forward a novel strategy to combat oxidative stress in the central nervous system, using synthetically developed exosomes loaded with antioxidants such as glutathione and the anti-aging protein Klotho.
Collapse
Affiliation(s)
- Sabina Ranjit
- a Department of Pharmaceutical Sciences , University of Tennessee Health Science Center , Memphis , TN , United States
| | - Benjamin J Patters
- a Department of Pharmaceutical Sciences , University of Tennessee Health Science Center , Memphis , TN , United States
| | - Kelli A Gerth
- a Department of Pharmaceutical Sciences , University of Tennessee Health Science Center , Memphis , TN , United States
| | - Sanjana Haque
- a Department of Pharmaceutical Sciences , University of Tennessee Health Science Center , Memphis , TN , United States
| | - Sanjeev Choudhary
- b Department of Internal Medicine , University of Texas Medical Branch , Galveston , TX , United States
| | - Santosh Kumar
- a Department of Pharmaceutical Sciences , University of Tennessee Health Science Center , Memphis , TN , United States
| |
Collapse
|
14
|
Song N, Wang J, Jiang H, Xie J. Astroglial and microglial contributions to iron metabolism disturbance in Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2018; 1864:967-973. [PMID: 29317336 DOI: 10.1016/j.bbadis.2018.01.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/24/2017] [Accepted: 01/02/2018] [Indexed: 02/06/2023]
Abstract
Understandings of the disturbed iron metabolism in Parkinson's disease (PD) are largely from the perspectives of neurons. Neurodegenerative processes in PD trigger universal and conserved astroglial dysfunction and microglial activation. In this review, we start with astroglia and microglia in PD with an emphasis on their roles in spreading α-synuclein pathology, and then focus on their contributions in iron metabolism under normal conditions and the diseased state of PD. Elevated iron in the brain regions affects glial features, meanwhile, glial effects on neuronal iron metabolism are largely dependent on their releasing factors. These advances might be valuable for better understanding and modulating iron metabolism disturbance in PD.
Collapse
Affiliation(s)
- Ning Song
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao 266071, China.
| | - Jun Wang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao 266071, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao 266071, China
| | - Junxia Xie
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disease, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
15
|
Healy S, McMahon JM, FitzGerald U. Modelling iron mismanagement in neurodegenerative disease in vitro: paradigms, pitfalls, possibilities & practical considerations. Prog Neurobiol 2017; 158:1-14. [DOI: 10.1016/j.pneurobio.2017.08.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/27/2017] [Accepted: 08/23/2017] [Indexed: 01/26/2023]
|
16
|
Roth AD, Núñez MT. Oligodendrocytes: Functioning in a Delicate Balance Between High Metabolic Requirements and Oxidative Damage. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 949:167-181. [PMID: 27714689 DOI: 10.1007/978-3-319-40764-7_8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The study of the metabolic interactions between myelinating glia and the axons they ensheath has blossomed into an area of research much akin to the elucidation of the role of astrocytes in tripartite synapses (Tsacopoulos and Magistretti in J Neurosci 16:877-885, 1996). Still, unlike astrocytes, rich in cytochrome-P450 and other anti-oxidative defense mechanisms (Minn et al. in Brain Res Brain Res Rev 16:65-82, 1991; Wilson in Can J Physiol Pharmacol. 75:1149-1163, 1997), oligodendrocytes can be easily damaged and are particularly sensitive to both hypoxia and oxidative stress, especially during their terminal differentiation phase and while generating myelin sheaths. In the present review, we will focus in the metabolic complexity of oligodendrocytes, particularly during the processes of differentiation and myelin deposition, and with a specific emphasis in the context of oxidative stress and the intricacies of the iron metabolism of the most iron-loaded cells of the central nervous system (CNS).
Collapse
Affiliation(s)
- Alejandro D Roth
- Department of Biology, Faculty of Science, University of Chile, Santiago, Chile.
| | - Marco T Núñez
- Department of Biology, Faculty of Science, University of Chile, Santiago, Chile
| |
Collapse
|
17
|
Differences in vulnerability of neurons and astrocytes to heme oxygenase-1 modulation: Implications for mitochondrial ferritin. Sci Rep 2016; 6:24200. [PMID: 27097841 PMCID: PMC4838889 DOI: 10.1038/srep24200] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 03/22/2016] [Indexed: 11/08/2022] Open
Abstract
Induction of the antioxidant enzyme heme oxygenase-1 (HO-1) was observed in both astrocytes and neurons in the substantia nigra of patients with Parkinson's disease (PD). In the current study, we investigated whether HO-1 behaves differently between neurons and astrocytes under the condition of neurotoxicity related to PD. The results showed a time-dependent HO-1 upregulation in primary cultured ventral mesencephalon neurons and astrocytes treated with the mitochondria complex I inhibitor 1-methyl-4-phenylpyridinium (MPP(+)) or recombinant α-synuclein. However, HO-1 upregulation appeared much later in neurons than in astrocytes. The HO-1 inhibitor zinc protoporphyrin (ZnPP) aggravated MPP(+)- or α-synuclein-induced oxidative damage in both astrocytes and neurons, indicating that this HO-1 response was cytoprotective. For neurons, the HO-1 activator cobalt protoporphyrin IX (CoPPIX) exerted protective effects against MPP(+) or α-synuclein during moderate HO-1 upregulation, but it aggravated damage at the peak of the HO-1 response. For astrocytes, CoPPIXalways showed protective effects. Higher basal and CoPPIX-induced mitochondrial ferritin (MtFt) levels were detected in astrocytes. Lentivirus-mediated MtFt overexpression rescued the neuronal damage induced by CoPPIX, indicating that large MtFt buffering capacity contributes to pronounced HO-1 tolerance in astrocytes. Such findings suggest that astrocyte-targeted HO-1 interventions and MtFt modulations have potential as novel pharmacological strategies in PD.
Collapse
|
18
|
Jiang H, Wang J, Rogers J, Xie J. Brain Iron Metabolism Dysfunction in Parkinson's Disease. Mol Neurobiol 2016; 54:3078-3101. [PMID: 27039308 DOI: 10.1007/s12035-016-9879-1] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 03/21/2016] [Indexed: 12/15/2022]
Abstract
Dysfunction of iron metabolism, which includes its uptake, storage, and release, plays a key role in neurodegenerative disorders, including Parkinson's disease (PD), Alzheimer's disease, and Huntington's disease. Understanding how iron accumulates in the substantia nigra (SN) and why it specifically targets dopaminergic (DAergic) neurons is particularly warranted for PD, as this knowledge may provide new therapeutic avenues for a more targeted neurotherapeutic strategy for this disease. In this review, we begin with a brief introduction describing brain iron metabolism and its regulation. We then provide a detailed description of how iron accumulates specifically in the SN and why DAergic neurons are especially vulnerable to iron in PD. Furthermore, we focus on the possible mechanisms involved in iron-induced cell death of DAergic neurons in the SN. Finally, we present evidence in support that iron chelation represents a plausable therapeutic strategy for PD.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Medical College of Qingdao University, Qingdao, 266071, China.
| | - Jun Wang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Medical College of Qingdao University, Qingdao, 266071, China
| | - Jack Rogers
- Neurochemistry Laboratory, Division of Psychiatric Neurosciences and Genetics and Aging Research Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Junxia Xie
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Medical College of Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
19
|
Kwon SK, Ahn M, Song HJ, Kang SK, Jung SB, Harsha N, Jee S, Moon JY, Suh KS, Lee SD, Jeon BH, Kim DW, Kim CS. Nafamostat mesilate attenuates transient focal ischemia/reperfusion-induced brain injury via the inhibition of endoplasmic reticulum stress. Brain Res 2015; 1627:12-20. [PMID: 26390938 DOI: 10.1016/j.brainres.2015.09.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 08/18/2015] [Accepted: 09/10/2015] [Indexed: 01/22/2023]
Abstract
Nafamostat mesilate (NM), a serine protease inhibitor, has a broad range of clinical applications that include use as an anticoagulant during hemodialysis in cerebral hemorrhage patients, as a hemoperfusion anticoagulant for patients with intravascular coagulation, hemorrhagic lesions, and hemorrhagic tendencies, and for the improvement of acute pancreatitis. However, the effects of NM on acute cerebral ischemia have yet to be investigated. Thus, the present study utilized a rat model in which transient middle cerebral artery occlusion (MCAO) was used to induce ischemic injury to investigate the effects of NM on infarct volume and histological and biological changes. NM (1mg/kg) was intravenously administered prior to and after the MCAO procedure. Compared to control rats, the administration of NM significantly decreased infarct size and the extent of brain edema after the induction of focal ischemia via MCAO. Additionally, NM treatment attenuated MCAO-induced neuronal degeneration and activation of microglia and astrocytes. NM treatment also inhibited the MCAO-induced expression levels of glucose-regulated protein 78 (GRP78), CATT/EBP homologous protein (CHOP), and p-eukaryotic initiation factor 2α (eIF2α), which are endoplasmic reticulum (ER) stress markers, in the cerebral cortex. The present findings demonstrate that NM exerts neuroprotective effects in the brain following focal ischemia via, at least in part, the inhibition of ER stress.
Collapse
Affiliation(s)
- Sun Kwan Kwon
- Department of physiology, School of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Moonsang Ahn
- Department of Surgery, School of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Hee-Jung Song
- Department of Neurology, Chungnam National University Hospital, Daejeon 301-721, Republic of Korea
| | - Shin Kwang Kang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Saet-Byel Jung
- Department of Endocrinology, School of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Nagar Harsha
- Department of physiology, School of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Sungju Jee
- Department of Rehabilitation Medicine, Chungnam National University Hospital, Daejeon 301-721, Republic of Korea
| | - Jae Young Moon
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon 301-721, Republic of Korea
| | - Kwang-Sun Suh
- Department of Pathology, School of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Sang Do Lee
- Department of physiology, School of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Byeong Hwa Jeon
- Department of physiology, School of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea
| | - Dong Woon Kim
- Department of Anatomy, School of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea.
| | - Cuk-Seong Kim
- Department of physiology, School of Medicine, Chungnam National University, Daejeon 301-747, Republic of Korea.
| |
Collapse
|
20
|
Xu X, Song N, Wang R, Jiang H, Xie J. Preferential Heme Oxygenase-1 Activation in Striatal Astrocytes Antagonizes Dopaminergic Neuron Degeneration in MPTP-Intoxicated Mice. Mol Neurobiol 2015; 53:5056-65. [PMID: 26385576 DOI: 10.1007/s12035-015-9437-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/10/2015] [Indexed: 01/01/2023]
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) accompanied by increased oxidative damage. Astrocytes, which are the most abundant glial cell types in the brain, possess higher antioxidant potential partially due to preferentially activated nuclear factor E2-related factor 2 genes. Heme oxygenase isoform 1 (HO-1) is crucial for the response to oxidative stress via the catabolism of heme to carbon monoxide, bilirubin, and iron. In the present study, we aimed to investigate astroglial expression of HO-1 in the SNpc, especially in the striatum of a subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mouse model of PD, and to investigate the neuroprotective effects of intraventricularly administrated HO-1 activator cobalt protoporphyrin IX (CoPPIX). The results showed that HO-1 was faintly distributed in neurons but not astrocytes in the normal SNpc and striatum. MPTP triggered a robust HO-1 response in the astrocytes of the striatum after 1-day treatment, but the HO-1 levels declined dramatically at day 3 and were completely undetectable at day 5. Intraventricular administration of CoPPIX for 8 days could preferentially activate HO-1 in astrocytes in the striatum but not SNpc. The content of striatal dopamine and its derivatives was restored in the subacute MPTP models. CoPPIX also increased the number of dopaminergic neurons and the tyrosine hydroxylase levels in the SNpc. These results suggest that inadequate HO-1 in striatal astrocytes might contribute to the limited antioxidant defense and dopaminergic neuron degeneration in PD, and preferential HO-1 activation in striatal astrocytes might be neuroprotective. The study thus sheds light on the targeting of HO-1 in striatal astrocytes for PD therapeutics.
Collapse
Affiliation(s)
- Xiaofeng Xu
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao, 266071, China
| | - Ning Song
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao, 266071, China
| | - Ranran Wang
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao, 266071, China
| | - Hong Jiang
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China.,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao, 266071, China
| | - Junxia Xie
- Collaborative Innovation Center for Brain Science, Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, 266071, China. .,Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao, 266071, China.
| |
Collapse
|
21
|
Uptake and metabolism of iron and iron oxide nanoparticles in brain astrocytes. Biochem Soc Trans 2014; 41:1588-92. [PMID: 24256259 DOI: 10.1042/bst20130114] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Astrocytes are considered key regulators of the iron metabolism of the brain. These cells are able to rapidly accumulate iron ions and various iron-containing compounds, store iron efficiently in ferritin and also export iron. The present short review summarizes our current knowledge of the molecular mechanisms involved in the handling of iron by astrocytes. Cultured astrocytes efficiently take up iron as ferrous or ferric iron ions or as haem by specific iron transport proteins in their cell membrane. In addition, astrocytes accumulate large amounts of iron oxide nanoparticles by endocytotic mechanisms. Despite the rapid accumulation of high amounts of iron from various iron-containing sources, the viability of astrocytes is hardly affected. A rather slow liberation of iron from accumulated haem or iron oxide nanoparticles as well as the strong up-regulation of the synthesis of the iron storage protein ferritin are likely to contribute to the high resistance of astrocytes to iron toxicity. The efficient uptake of extracellular iron by cultured astrocytes as well as their strong up-regulation of ferritin after iron exposure also suggests that brain astrocytes deal well with an excess of iron and protect the brain against iron-mediated toxicity.
Collapse
|
22
|
Musci G, Polticelli F, Bonaccorsi di Patti MC. Ceruloplasmin-ferroportin system of iron traffic in vertebrates. World J Biol Chem 2014; 5:204-215. [PMID: 24921009 PMCID: PMC4050113 DOI: 10.4331/wjbc.v5.i2.204] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 02/19/2014] [Indexed: 02/05/2023] Open
Abstract
Safe trafficking of iron across the cell membrane is a delicate process that requires specific protein carriers. While many proteins involved in iron uptake by cells are known, only one cellular iron export protein has been identified in mammals: ferroportin (SLC40A1). Ceruloplasmin is a multicopper enzyme endowed with ferroxidase activity that is found as a soluble isoform in plasma or as a membrane-associated isoform in specific cell types. According to the currently accepted view, ferrous iron transported out of the cell by ferroportin would be safely oxidized by ceruloplasmin to facilitate loading on transferrin. Therefore, the ceruloplasmin-ferroportin system represents the main pathway for cellular iron egress and it is responsible for physiological regulation of cellular iron levels. The most recent findings regarding the structural and functional features of ceruloplasmin and ferroportin and their relationship will be described in this review.
Collapse
|
23
|
Luther EM, Petters C, Bulcke F, Kaltz A, Thiel K, Bickmeyer U, Dringen R. Endocytotic uptake of iron oxide nanoparticles by cultured brain microglial cells. Acta Biomater 2013; 9:8454-65. [PMID: 23727247 DOI: 10.1016/j.actbio.2013.05.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2012] [Revised: 05/17/2013] [Accepted: 05/21/2013] [Indexed: 11/29/2022]
Abstract
Microglia are the phagocytotic cells of the brain that respond rapidly to alterations in brain homeostasis. Since iron oxide nanoparticles (IONPs) are used for diagnostic and therapeutic applications in the brain, the consequences of an exposure of microglial cells to IONPs are of particular interest. To address this topic we have synthesized and characterized fluorescent BODIPY®-labelled IONPs (BP-IONPs). The average hydrodynamic diameter and the ζ-potential of BP-IONPs in water were ∼65 nm and -49 mV, respectively. Both values increased after dispersion of the particles in serum containing incubation medium to ∼130 nm and -8 mV. Exposure of cultured rat microglial cells with BP-IONPs caused a time-, concentration- and temperature-dependent uptake of the particles, as demonstrated by strong increases in cellular iron contents and cellular fluorescence. Incubation for 3h with 150 and 450 μM iron as BP-IONPs increased the cellular iron content from a low basal level of ∼50 nmol iron mg(-1) to 219±52 and 481±28 nmol iron (mg protein)(-1), respectively. These conditions did not affect cell viability, but exposure to higher concentrations of BP-IONPs or for longer incubation periods severely compromised cell viability. The BP-IONP fluorescence in viable microglial cells was co-localized with lysosomes. In addition, BP-IONP accumulation was lowered by 60% in the presence of the endocytosis inhibitors 5-(N-ethyl-N-isopropyl)amiloride, tyrphostin23 and chlorpromazin. These results suggest that the rapid accumulation of BP-IONPs by microglial cells is predominantly mediated by macropinocytosis and clathrin-mediated endocytosis, which direct the accumulated particles into the lysosomal compartment.
Collapse
Affiliation(s)
- Eva M Luther
- Center for Biomolecular Interactions Bremen, University of Bremen, P.O. Box 330440, D-28334 Bremen, Germany
| | | | | | | | | | | | | |
Collapse
|
24
|
Sozio P, Cerasa LS, Laserra S, Cacciatore I, Cornacchia C, Di Filippo ES, Fulle S, Fontana A, Di Crescenzo A, Grilli M, Marchi M, Di Stefano A. Memantine-sulfur containing antioxidant conjugates as potential prodrugs to improve the treatment of Alzheimer's disease. Eur J Pharm Sci 2013; 49:187-98. [PMID: 23454012 DOI: 10.1016/j.ejps.2013.02.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 02/04/2013] [Accepted: 02/05/2013] [Indexed: 11/30/2022]
Abstract
The approved treatments for Alzheimer's disease (AD) exploit mainly a symptomatic approach based on the use of cholinesterase inhibitors or N-methyl-D-aspartate (NMDA) receptor antagonists. Natural antioxidant compounds, able to pass through the blood-brain barrier (BBB), have been extensively studied as useful neuroprotective agents. A novel approach towards excitotoxicity protection and oxidative stress associated with excess β amyloid (Aβ) preservation in AD is represented by selective glutamatergic antagonists that possess as well antioxidant capabilities. In the present work, GSH (1) or (R)-α-lipoic acid (LA) (2) have been covalently linked with the NMDA receptor antagonists memantine (MEM). The new conjugates, proposed as potential antialzheimer drugs, should act both as glutamate receptor antagonists and radical scavenging agents. The physico-chemical properties and "in vitro" membrane permeability, the enzymatic and chemical stability, the demonstrated "in vitro" antioxidant activity associated to the capacity to inhibit Aβ(1-42) aggregation makes at least compound 2 a promising candidate for treatment of AD patients.
Collapse
Affiliation(s)
- Piera Sozio
- Dipartimento di Farmacia, Università G. D'Annunzio, Via dei Vestini 31, 66100 Chieti, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Li R, Xu X, Chen C, Yu X, Edin ML, Degraff LM, Lee CR, Zeldin DC, Wang DW. Cytochrome P450 2J2 is protective against global cerebral ischemia in transgenic mice. Prostaglandins Other Lipid Mediat 2012; 99:68-78. [PMID: 23041291 DOI: 10.1016/j.prostaglandins.2012.09.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 09/20/2012] [Accepted: 09/24/2012] [Indexed: 12/18/2022]
Abstract
Cytochrome P450 epoxygenase metabolites of arachidonic acid, EETs, have multiple cardiovascular effects, including reduction of blood pressure, protection against myocardial ischemia-reperfusion injury, and attenuation of endothelial apoptosis. This study investigated the hypothesis that transgenic mice with endothelial overexpression of CYP2J2 (Tie2-CYP2J2-Tr) would be protected against global cerebral ischemia induced by bilateral common carotid artery occlusion (BCCAO) and action mechanisms of EETs on cerebral ischemia in cultures of astrocytes exposed to oxygen-glucose deprivation (OGD). Tie2-CYP2J2-Tr mice had significantly increased CYP2J2 expression, increased 14,15-EET production, increases regional cerebral blood flow (rCBF) and microvascular density, decreased ROS production, decreased brain infarct size and apoptosis after ischemia compared to wild type mice, these were associated with increased activation of the PI3K/AKT and apoptosis-related protein in ischemic brain. Addition of exogenous EETs or CYP2J2 transfection attenuated OGD-induced apoptosis in astrocytes via activation of PI3K/AKT and anti-apoptosis pathways. However, these effects were reduced by pretreatments with inhibitor of the PI3K (LY294002) and 14,15-EET (14,15-EEZE), respectively. These results indicate that CYP2J2 overexpression exerts marked neuroprotective effects against ischemic injury by a mechanism linked to increased level of circulating EETs and increases CBF and reduction of apoptosis.
Collapse
Affiliation(s)
- Rui Li
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Halder SK, Matsunaga H, Ueda H. Neuron-specific non-classical release of prothymosin alpha: a novel neuroprotective damage-associated molecular patterns. J Neurochem 2012; 123:262-75. [PMID: 22853710 DOI: 10.1111/j.1471-4159.2012.07897.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 07/24/2012] [Accepted: 07/25/2012] [Indexed: 11/27/2022]
Abstract
Prothymosin alpha (ProTα), a nuclear protein devoid of signal sequence, has been shown to possess a number of cellular functions including cell survival. Most recently, we demonstrated that ProTα is localized in the nuclei of neurons, while it is found in both nuclei and cytoplasm in the astrocytes and microglia of adult brain. However, the cell type-specific non-classical release of ProTα under cerebral ischemia is yet unknown. In this study, we report that ProTα is non-classically released along with S100A13 from neurons in the hippocampus, striatum and somatosensory cortex at 3 h after cerebral ischemia, but amlexanox (an anti-allergic compound) reversibly blocks this neuronal ProTα release. We found that none of ProTα is released from astrocytes and microglia under ischemic stress. Indeed, ProTα intensity is increased gradually in astrocytes and microglia through 24 h after the cerebral ischemia. Interestingly, Z-Val-Ala-Asp fluoromethyl ketone, a caspase 3 inhibitor, pre-treatment induces ProTα release from astrocytes in the ischemic brain, but this release is reversibly blocked by amlexanox. However, Z-Val-Ala-Asp fluoromethyl ketone as well as amlexanox has no effect on ProTα distribution in microglia upon cerebral ischemia. Taken together, these results suggest that only neurons have machineries to release ProTα upon cerebral ischemic stress in vivo.
Collapse
Affiliation(s)
- Sebok Kumar Halder
- Department of Molecular Pharmacology and Neuroscience, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | |
Collapse
|
27
|
Dysregulation of iron metabolism in Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Adv Pharmacol Sci 2011; 2011:378278. [PMID: 22013437 PMCID: PMC3195304 DOI: 10.1155/2011/378278] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Revised: 07/09/2011] [Accepted: 07/25/2011] [Indexed: 12/22/2022] Open
Abstract
Dysregulation of iron metabolism has been observed in patients with neurodegenerative diseases (NDs). Utilization of several importers and exporters for iron transport in brain cells helps maintain iron homeostasis. Dysregulation of iron homeostasis leads to the production of neurotoxic substances and reactive oxygen species, resulting in iron-induced oxidative stress. In Alzheimer's disease (AD) and Parkinson's disease (PD), circumstantial evidence has shown that dysregulation of brain iron homeostasis leads to abnormal iron accumulation. Several genetic studies have revealed mutations in genes associated with increased iron uptake, increased oxidative stress, and an altered inflammatory response in amyotrophic lateral sclerosis (ALS). Here, we review the recent findings on brain iron metabolism in common NDs, such as AD, PD, and ALS. We also summarize the conventional and novel types of iron chelators, which can successfully decrease excess iron accumulation in brain lesions. For example, iron-chelating drugs have neuroprotective effects, preventing neural apoptosis, and activate cellular protective pathways against oxidative stress. Glial cells also protect neurons by secreting antioxidants and antiapoptotic substances. These new findings of experimental and clinical studies may provide a scientific foundation for advances in drug development for NDs.
Collapse
|
28
|
Yu X, Guo J, Fang H, Peng S. Basal metallothionein-I/II protects against NMDA-mediated oxidative injury in cortical neuron/astrocyte cultures. Toxicology 2011; 282:16-22. [PMID: 21215786 DOI: 10.1016/j.tox.2010.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 12/24/2010] [Indexed: 10/18/2022]
Abstract
N-Methyl-D-aspartate (NMDA) receptor overactivation-mediated oxidative stress has been proposed to contribute to brain injury. Metallothionein-I/II (MT-I/II), a member of cysteine-rich metalloproteins, has been found to express in the central nervous system primarily in cortical tissues and be upregulated following brain injury. To address the role of MT-I/II on NMDA-mediated oxidative injury, we established primary cortical neuron/astrocyte cultures from neonatal MT-I/II deficient (MT⁻/⁻) and wild type (MT+/+) mice to test whether basal MT-I/II protects cortical cultures against NMDA-mediated injury. We found that MT-I/II expression was increased by NMDA in MT+/+ cultures but was not detectable in MT⁻/⁻ cultures. NMDA concentration-dependently induced oxidative injury in both MT+/+ and MT⁻/⁻ cultures as evidenced by decrease of cell viability, increases of lipid peroxidation and DNA damage. However, these toxic effects were greater in MT⁻/⁻ than MT+/+ cultures. NMDA significantly increased reactive oxygen species (ROS) generation and disrupted mitochondrial membrane potential in neurons in MT+/+ cultures, and these effects were exaggerated in MT⁻/⁻ cultures. Our findings clearly show that basal MT-I/II provides protection against NMDA-mediated oxidative injury in cortical neuron/astrocyte cultures, and suggest that the protective effects are possibly associated with inhibition of ROS generation and preservation of mitochondrial membrane potential.
Collapse
Affiliation(s)
- Xiaoqian Yu
- Evaluation and Research Centre for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, 20# Dongdajie Rd, Fengtai District, Beijing 100071, PR China
| | | | | | | |
Collapse
|
29
|
Aluminium and Iron in Humans: Bioaccumulation, Pathology, and Removal. Rejuvenation Res 2010; 13:589-98. [DOI: 10.1089/rej.2009.0995] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
30
|
Bishop GM, Dang TN, Dringen R, Robinson SR. Accumulation of Non-Transferrin-Bound Iron by Neurons, Astrocytes, and Microglia. Neurotox Res 2010; 19:443-51. [DOI: 10.1007/s12640-010-9195-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 03/16/2010] [Accepted: 04/13/2010] [Indexed: 12/01/2022]
|
31
|
Tulpule K, Robinson SR, Bishop GM, Dringen R. Uptake of ferrous iron by cultured rat astrocytes. J Neurosci Res 2010; 88:563-71. [PMID: 19746426 DOI: 10.1002/jnr.22217] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Astrocytes are considered to play an important role in iron homeostasis of the brain, yet the mechanisms involved in the uptake of iron into astrocytes remain elusive. To investigate the uptake of iron into astrocytes, we have applied ferric ammonium citrate (FAC) to rat astrocyte-rich primary cultures. These cultures express the mRNAs of two membrane-bound ferric reductases, Dcytb and SDR2, and reduce extracellular ferric iron (100 muM) with a rate of 3.2 +/- 0.4 nmol/(hr x mg). This reduction rate is substantially lower than the rate of cellular iron accumulation from 100 muM FAC [24.7 +/- 8.9 nmol/(hr x mg)], which suggests that iron accumulation from FAC does at best partially depend on extracellular ferric reduction. Nonetheless, when the iron in FAC was almost completely reduced by an excess of exogenous ascorbate, astrocytes accumulated iron in a time- and concentration-dependent manner with specific iron accumulation rates that increased linearly for concentrations of up to 100 muM ferrous iron. This accumulation was attenuated by lowering the incubation temperature, by the presence of ferrous iron chelators, or by lowering the pH from 7.4 to 6.8. These data indicate that, in addition to the DMT1-mediated uptake of ferrous iron, astrocytes can accumulate ferric and ferrous iron by mechanisms that are independent of DMT1 or transferrin.
Collapse
Affiliation(s)
- Ketki Tulpule
- Center for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | | | | | |
Collapse
|
32
|
Nakajima K, Yamamoto S, Tohyama Y, Kohsaka S. Close association of p38 and JNK with plasminogen-dependent upregulation of PAI-1 in rat astrocytes in vitro. Neurosci Lett 2010; 471:66-9. [PMID: 20074614 DOI: 10.1016/j.neulet.2010.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 01/05/2010] [Accepted: 01/06/2010] [Indexed: 11/29/2022]
Abstract
As reported previously, stimulation of astrocytes with plasminogen (PLGn) remarkably enhances their production/release of plasminogen activator inhibitor-1 (PAI-1). In addition, both p38 mitogen-activated protein kinase (p38MAPK) and c-Jun N-terminal kinase (JNK) are activated in these astrocytes. However, it remains to be determined whether the MAPK activation is associated with the PAI-1 induction in PLGn-stimulated astrocytes. In the present study, we investigated the relationship between MAPK activity and PAI-1 induction in PLGn-stimulated astrocytes. PLGn stimulation led to definitive phosphorylation of three MAPKs: external signal regulated kinase (ERK), JNK and p38. These results suggest that all of these MAPKs, either alone or in combination, are involved in PAI-1 induction. To verify this association, an inhibition experiment was carried out by using inhibitors specific for each MAPK. The results of the immunoblotting analysis indicated that 20 microM SB203580 (the p38 inhibitor) or SP600125 (the JNK inhibitor) suppressed approximately 85% or 40% of PLGn-inducible PAI-1, respectively. Only 20% inhibition was achieved by pretreatment of astrocytes with 20 microM PD98059 (the inhibitor of MEK1/2, an upstream kinase of ERK). In conclusion, p38 and JNK were shown to be the major MAPKs involved in the signaling cascade leading to PAI-1 induction in astrocytes stimulated with PLGn.
Collapse
Affiliation(s)
- Kazuyuki Nakajima
- Department of Bioinformatics, Faculty of Engineering, Soka University, 1-236 Tangi-machi, Hachioji, Tokyo 192-8577, Japan.
| | | | | | | |
Collapse
|
33
|
Characteristic response of astrocytes to plasminogen/plasmin to upregulate transforming growth factor beta 3 (TGFbeta3) production/secretion through proteinase-activated receptor-1 (PAR-1) and the downstream phosphatidylinositol 3-kinase (PI3K)-Akt/PKB signaling cascade. Brain Res 2009; 1305:1-13. [PMID: 19765562 DOI: 10.1016/j.brainres.2009.09.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 08/27/2009] [Accepted: 09/08/2009] [Indexed: 01/31/2023]
Abstract
The effects of microglia-derived plasminogen (PLGn) on the neurotrophic role of astrocytes were investigated in vitro. The treatment of astrocytes with rat PLGn led to a significant increase in transforming growth factor beta3 (TGFbeta3) in the conditioned medium (CM). This response of astrocytes to PLGn was characteristic and different from that to other stimulators, including lipopolysaccharide, phorbol-12-myristate-13-acetate, interferon-gamma, and ATP. In surveying the signaling molecules that respond to PLGn in astrocytes, we found that Akt/PKB phosphorylation is promoted. The pretreatment of astrocytes with an Akt inhibitor prior to PLGn stimulation resulted in a significant decrease in TGFbeta3 amounts in the CM, suggesting an association of Akt with TGFbeta3 production/secretion. Further survey revealed that phosphatidylinositol 3 kinase (PI3K) is closely associated with TGFbeta3 production/secretion in astrocytes. In fact, PI3K inhibitor clearly depressed the phosphorylation of Akt, indicating that PI3K is localized upstream of Akt. Moreover, the effects of PLGn to increase TGFbeta3 were depressed by pretreatment with a proteinase-activated receptor-1 (PAR-1) inhibitor. Plasmin could mimic the PLGn effects to upregulate TGFbeta3, and the plasmin effects were suppressed by pretreatment with the PAR-1 inhibitor, suggesting the association of PLGn/plasmin effects with PAR-1. In addition, Akt phosphorylation caused by plasmin was inhibited in the presence of PAR-1 inhibitor. We have therefore demonstrated that PLGn/plasmin, probably plasmin, facilitates the production/secretion of TGFbeta3 in astrocytes through both PAR-1 and the subsequent signaling cascade including PI3K and Akt.
Collapse
|
34
|
Rhodes SL, Ritz B. Genetics of iron regulation and the possible role of iron in Parkinson's disease. Neurobiol Dis 2008; 32:183-95. [PMID: 18675357 PMCID: PMC3643980 DOI: 10.1016/j.nbd.2008.07.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 07/01/2008] [Accepted: 07/02/2008] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is acknowledged as the second most common neurodegenerative disorder after Alzheimer's Disease. Older age may be the only unequivocal risk factor for PD although the male to female ratio is consistently greater than 1 in populations of European ancestry. Characteristic features of PD include dopaminergic neuron death in the substantia nigra (SN) pars compacta, accumulation of alpha-synuclein inclusions known as Lewy bodies in the SN, and brain iron accumulation beyond that observed in non-PD brains of a similar age. In this review article, we will provide an overview of human and animal studies investigating the contributions of iron in PD, a summary of human studies of iron-related genes in PD, a review of the literature on the genetics of iron metabolism, and some hypotheses on possible roles for iron in the pathogenic processes of PD including potential interactions between iron and other factors associated with Parkinson's disease.
Collapse
Affiliation(s)
- Shannon L Rhodes
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | |
Collapse
|