1
|
Lei L, Yuan J, Dai Z, Xiang S, Tu Q, Cui X, Zhai S, Chen X, He Z, Fang B, Xu Z, Yu H, Tang L, Zhang C. Targeting the Labile Iron Pool with Engineered DFO Nanosheets to Inhibit Ferroptosis for Parkinson's Disease Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409329. [PMID: 39221531 DOI: 10.1002/adma.202409329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/31/2024] [Indexed: 09/04/2024]
Abstract
Ferroptosis in neurons is considered one of the key factors that induces Parkinson's disease (PD), which is caused by excessive iron accumulation in the intracellular labile iron pool (LIP). The iron ions released from the LIP lead to the aberrant generation of reactive oxygen species (ROS) to trigger ferroptosis and exacerbate PD progression. Herein, a pioneering design of multifunctional nanoregulator deferoxamine (DFO)-integrated nanosheets (BDPR NSs) is presented that target the LIP to restrict ferroptosis and protect against PD. The BDPR NSs are constructed by incorporating a brain-targeting peptide and DFO into polydopamine-modified black phosphorus nanosheets. These BDPR NSs can sequester free iron ions, thereby ameliorating LIP overload and regulating iron metabolism. Furthermore, the BDPR NSs can decrease lipid peroxidation generation by mitigating ROS accumulation. More importantly, BDPR NSs can specifically accumulate in the mitochondria to suppress ROS generation and decrease mitochondrial iron accumulation. In vivo experiments demonstrated that the BDPR NSs highly efficiently mitigated dopaminergic neuronloss and its associated behavioral disorders by modulating the LIP and inhibiting ferroptosis. Thus, the BDPR-based nanovectors holds promise as a potential avenue for advancing PD therapy.
Collapse
Affiliation(s)
- Li Lei
- Department of Chemistry, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
- Department of Neurosurgery, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, China
| | - Jiali Yuan
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
| | - Zhijun Dai
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
| | - Song Xiang
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
| | - Qiuxia Tu
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
| | - Xing Cui
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, College of Pharmacy, Guizhou Medical University, Guiyang, 550025, China
| | - Suzhen Zhai
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
| | - Xiaozhong Chen
- The Jinyang Hospital Affiliated to Guizhou Medical University: The Second People's Hospital of Guiyang, Guiyang, 550025, China
| | - Zhixu He
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, 550004, China
| | - Boyan Fang
- Parkinson Medical Center, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, 100144, China
| | - Zhiai Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Haijun Yu
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lei Tang
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, College of Pharmacy, Guizhou Medical University, Guiyang, 550025, China
| | - Chunlin Zhang
- Department of Biology, Engineering Research Center for Molecular Medicine, College of Basic Medical Science, Guizhou Medical University, Guiyang, 550025, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang, 550004, China
| |
Collapse
|
2
|
Corona-Trejo A, Gonsebatt ME, Trejo-Solis C, Campos-Peña V, Quintas-Granados LI, Villegas-Vázquez EY, Daniel Reyes-Hernández O, Hernández-Abad VJ, Figueroa-González G, Silva-Adaya D. Transsulfuration pathway: a targeting neuromodulator in Parkinson's disease. Rev Neurosci 2023; 34:915-932. [PMID: 37409540 DOI: 10.1515/revneuro-2023-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/04/2023] [Indexed: 07/07/2023]
Abstract
The transsulfuration pathway (TSP) is a metabolic pathway involving sulfur transfer from homocysteine to cysteine. Transsulfuration pathway leads to many sulfur metabolites, principally glutathione, H2S, taurine, and cysteine. Key enzymes of the TSP, such as cystathionine β-synthase and cystathionine γ-lyase, are essential regulators at multiple levels in this pathway. TSP metabolites are implicated in many physiological processes in the central nervous system and other tissues. TSP is important in controlling sulfur balance and optimal cellular functions such as glutathione synthesis. Alterations in the TSP and related pathways (transmethylation and remethylation) are altered in several neurodegenerative diseases, including Parkinson's disease, suggesting their participation in the pathophysiology and progression of these diseases. In Parkinson's disease many cellular processes are comprised mainly those that regulate redox homeostasis, inflammation, reticulum endoplasmic stress, mitochondrial function, oxidative stress, and sulfur content metabolites of TSP are involved in these damage processes. Current research on the transsulfuration pathway in Parkinson's disease has primarily focused on the synthesis and function of certain metabolites, particularly glutathione. However, our understanding of the regulation of other metabolites of the transsulfuration pathway, as well as their relationships with other metabolites, and their synthesis regulation in Parkinson´s disease remain limited. Thus, this paper highlights the importance of studying the molecular dynamics in different metabolites and enzymes that affect the transsulfuration in Parkinson's disease.
Collapse
Affiliation(s)
- Andrea Corona-Trejo
- Carrera de Biología, Laboratorio de Farmacogenética, Unidad Multidisciplinaria de Investigación Experimental Zaragoza, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico
| | - María E Gonsebatt
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Cristina Trejo-Solis
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Mexico, 14269, Mexico
| | - Victoria Campos-Peña
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Mexico, 14269, Mexico
| | | | - Edgar Yebrán Villegas-Vázquez
- Laboratorio de Farmacogenética, Unidad Multidisciplinaria de Investigación Experimental Zaragoza, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, 09230 Mexico City, Mexico
| | - Octavio Daniel Reyes-Hernández
- Laboratorio de Biología Molecular del Cáncer, Unidad Multidisciplinaria de Investigación Experimental Zaragoza, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México 09230, Mexico
| | - Vicente Jesús Hernández-Abad
- Laboratorio de Investigación Farmacéutica, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Batalla de 5 de mayo s/n, Col, Ejército de Oriente, 09230 Mexico City, Mexico
| | - Gabriela Figueroa-González
- Laboratorio de Farmacogenética, Unidad Multidisciplinaria de Investigación Experimental Zaragoza, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, 09230 Mexico City, Mexico
| | - Daniela Silva-Adaya
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Mexico, 14269, Mexico
| |
Collapse
|
3
|
Warming H, Deinhardt K, Garland P, More J, Bulters D, Galea I, Vargas-Caballero M. Functional effects of haemoglobin can be rescued by haptoglobin in an in vitro model of subarachnoid haemorrhage. J Neurochem 2023; 167:90-103. [PMID: 37702203 DOI: 10.1111/jnc.15936] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 09/14/2023]
Abstract
During subarachnoid haemorrhage, a blood clot forms in the subarachnoid space releasing extracellular haemoglobin (Hb), which causes oxidative damage and cell death in surrounding tissues. High rates of disability and cognitive decline in SAH survivors are attributed to loss of neurons and functional connections during secondary brain injury. Haptoglobin sequesters Hb for clearance, but this scavenging system is overwhelmed after a haemorrhage. Whilst exogenous haptoglobin application can attenuate cytotoxicity of Hb in vitro and in vivo, the functional effects of sub-lethal Hb concentrations on surviving neurons and whether cellular function can be protected with haptoglobin treatment remain unclear. Here we use cultured neurons to investigate neuronal health and function across a range of Hb concentrations to establish the thresholds for cellular damage and investigate synaptic function. Hb impairs ATP concentrations and cytoskeletal structure. At clinically relevant but sub-lethal Hb concentrations, we find that synaptic AMPAR-driven currents are reduced, accompanied by a reduction in GluA1 subunit expression. Haptoglobin co-application can prevent these deficits by scavenging free Hb to reduce it to sub-threshold concentrations and does not need to be present at stoichiometric amounts to achieve efficacy. Haptoglobin itself does not impair measures of neuronal health and function at any concentration tested. Our data highlight a role for Hb in modifying synaptic function in surviving neurons, which may link to impaired cognition or plasticity after SAH and support the development of haptoglobin as a therapy for subarachnoid haemorrhage.
Collapse
Affiliation(s)
- Hannah Warming
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Katrin Deinhardt
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | | | - John More
- Bio Products Laboratory Limited, Elstree, UK
| | - Diederik Bulters
- Department of Neurosurgery, Wessex Neurological Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Mariana Vargas-Caballero
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
4
|
Zhang Q, Zhao H, Luo M, Cheng X, Li Y, Li Q, Wang Z, Niu Q. The Classification and Prediction of Ferroptosis-Related Genes in ALS: A Pilot Study. Front Genet 2022; 13:919188. [PMID: 35873477 PMCID: PMC9305067 DOI: 10.3389/fgene.2022.919188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive muscle paralysis, which is followed by degeneration of motor neurons in the motor cortex of the brainstem and spinal cord. The etiology of sporadic ALS (sALS) is still unknown, limiting the exploration of potential treatments. Ferroptosis is a new form of cell death and is reported to be closely associated with Alzheimer’s disease (AD), Parkinson’s disease (PD), and ALS. In this study, we used datasets (autopsy data and blood data) from Gene Expression Omnibus (GEO) to explore the role of ferroptosis and ferroptosis-related gene (FRG) alterations in ALS. Gene set enrichment analysis (GSEA) found that the activated ferroptosis pathway displayed a higher enrichment score, and the expression of 26 ferroptosis genes showed obvious group differences between ALS and controls. Using weighted gene correlation network analysis (WGCNA), we identified FRGs associated with ALS, of which the Gene Ontology (GO) analysis displayed that the biological process of oxidative stress was the most to be involved in. KEGG pathway analysis revealed that the FRGs were enriched not only in ferroptosis pathways but also in autophagy, FoxO, and mTOR signaling pathways. Twenty-one FRGs (NR4A1, CYBB, DRD4, SETD1B, LAMP2, ACSL4, MYB, PROM2, CHMP5, ULK1, AKR1C2, TGFBR1, TMBIM4, MLLT1, PSAT1, HIF1A, LINC00336, AMN, SLC38A1, CISD1, and GABARAPL2) in the autopsy data and 16 FRGs (NR4A1, DRD4, SETD1B, MYB, PROM2, CHMP5, ULK1, AKR1C2, TGFBR1, TMBIM4, MLLT1, HIF1A, LINC00336, IL33, SLC38A1, and CISD1) in the blood data were identified as target genes by least absolute shrinkage and selection operator analysis (LASSO), in which gene signature could differentiate ALS patients from controls. Finally, the higher the expression of CHMP5 and SLC38A1 in whole blood, the shorter the lifespan of ALS patients will be. In summary, our study presents potential biomarkers for the diagnosis and prognosis of ALS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Qi Niu
- *Correspondence: Qi Niu, ; Zheng Wang,
| |
Collapse
|
5
|
Caioni G, Cimini A, Benedetti E. Food Contamination: An Unexplored Possible Link between Dietary Habits and Parkinson’s Disease. Nutrients 2022; 14:nu14071467. [PMID: 35406080 PMCID: PMC9003245 DOI: 10.3390/nu14071467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 12/10/2022] Open
Abstract
Importance of a healthy lifestyle in maintaining the population’s well-being and health, especially in terms of balanced nutrition, is well known. Food choice of and dieting habits could impact disease management, which is especially true for Parkinson’s disease (PD). However, nowadays, it is not that simple to maintain a balance in nutrition, and the idea of a healthy diet tends to fade as the consequence of a western lifestyle. This should not only be dealt with in the context of food choice, but also from an environmental point of view. What we put into our bodies is strictly related to the quality of ecosystems we live in. For these reasons, attention should be directed to all the pollutants, which in many cases, we unknowingly ingest. It will be necessary to explore the interaction between food and environment, since human activity also influences the raw materials destined for consumption. This awareness can be achieved by means of an innovative scientific approach, which involves the use of new models, in order to overcome the traditional scientific investigations included in the study of Parkinson’s disease.
Collapse
Affiliation(s)
- Giulia Caioni
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.C.)
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.C.)
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (G.C.); (A.C.)
- Correspondence: ; Tel.: +39-086-243-3267
| |
Collapse
|
6
|
Wang S, Jiang Y, Liu Y, Liu Q, Sun H, Mei M, Liao X. Ferroptosis promotes microtubule-associated protein tau aggregation via GSK-3β activation and proteasome inhibition. Mol Neurobiol 2022; 59:1486-1501. [PMID: 34997541 DOI: 10.1007/s12035-022-02731-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/30/2021] [Indexed: 12/17/2022]
Abstract
Ferroptosis is a form of regulated cell death resulting from iron accumulation and lipid peroxidation. Iron dyshomeostasis and peroxidation damage of neurons in some particular brain regions are closely related to a wide range of neurodegenerative diseases known as "tauopathies," in which intracellular aggregation of microtubule-associated protein tau is the common neuropathological feature. However, the relationship between ferroptosis and tau aggregation is not well understood. The current study demonstrates that erastin-induced ferroptosis can promote tau hyperphosphorylation and aggregation in mouse neuroblastoma cells (N2a cells). Moreover, ferroptosis inhibitor ferrostatin-1 can alleviate tau aggregation effectively. In-depth mechanism research indicates that activated glycogen synthase kinase-3β (GSK-3β) is responsible for the abnormal hyperphosphorylation of tau. More importantly, proteasome inhibition can exacerbate tau degradation obstacle and accelerate tau aggregation in the process of ferroptosis. Our results indicate that ferroptosis can lead to abnormal aggregation of tau protein and might be a promising therapeutic target of tauopathies.
Collapse
Affiliation(s)
- Shaohui Wang
- Hubei Key Lab of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, People's Republic of China
| | - Yao Jiang
- Hubei Key Lab of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, People's Republic of China
| | - Yabo Liu
- Hubei Key Lab of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, People's Republic of China
| | - Qianhui Liu
- Hubei Key Lab of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, People's Republic of China
| | - Hongwei Sun
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Institute of Environmental & Applied Chemistry, College of Chemistry, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Mengjie Mei
- Hubei Key Lab of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, People's Republic of China
| | - Xiaomei Liao
- Hubei Key Lab of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, People's Republic of China.
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Institute of Environmental & Applied Chemistry, College of Chemistry, Central China Normal University, Wuhan, 430079, People's Republic of China.
| |
Collapse
|
7
|
Maung MT, Carlson A, Olea-Flores M, Elkhadragy L, Schachtschneider KM, Navarro-Tito N, Padilla-Benavides T. The molecular and cellular basis of copper dysregulation and its relationship with human pathologies. FASEB J 2021; 35:e21810. [PMID: 34390520 DOI: 10.1096/fj.202100273rr] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/23/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022]
Abstract
Copper (Cu) is an essential micronutrient required for the activity of redox-active enzymes involved in critical metabolic reactions, signaling pathways, and biological functions. Transporters and chaperones control Cu ion levels and bioavailability to ensure proper subcellular and systemic Cu distribution. Intensive research has focused on understanding how mammalian cells maintain Cu homeostasis, and how molecular signals coordinate Cu acquisition and storage within organs. In humans, mutations of genes that regulate Cu homeostasis or facilitate interactions with Cu ions lead to numerous pathologic conditions. Malfunctions of the Cu+ -transporting ATPases ATP7A and ATP7B cause Menkes disease and Wilson disease, respectively. Additionally, defects in the mitochondrial and cellular distributions and homeostasis of Cu lead to severe neurodegenerative conditions, mitochondrial myopathies, and metabolic diseases. Cu has a dual nature in carcinogenesis as a promotor of tumor growth and an inducer of redox stress in cancer cells. Cu also plays role in cancer treatment as a component of drugs and a regulator of drug sensitivity and uptake. In this review, we provide an overview of the current knowledge of Cu metabolism and transport and its relation to various human pathologies.
Collapse
Affiliation(s)
- May T Maung
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Alyssa Carlson
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Monserrat Olea-Flores
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Guerrero, Mexico
| | - Lobna Elkhadragy
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA.,National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Napoleon Navarro-Tito
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Guerrero, Mexico
| | | |
Collapse
|
8
|
Chen J, Gong NJ, Chaim KT, Otaduy MCG, Liu C. Decompose quantitative susceptibility mapping (QSM) to sub-voxel diamagnetic and paramagnetic components based on gradient-echo MRI data. Neuroimage 2021; 242:118477. [PMID: 34403742 PMCID: PMC8720043 DOI: 10.1016/j.neuroimage.2021.118477] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/13/2021] [Indexed: 12/31/2022] Open
Abstract
PURPOSE A method named DECOMPOSE-QSM is developed to decompose bulk susceptibility measured with QSM into sub-voxel paramagnetic and diamagnetic components based on a three-pool complex signal model. METHODS Multi-echo gradient echo signal is modeled as a summation of three weighted exponentials corresponding to three types of susceptibility sources: reference susceptibility, diamagnetic and paramagnetic susceptibility relative to the reference. Paramagnetic component susceptibility (PCS) and diamagnetic component susceptibility (DCS) maps are constructed to represent the sub-voxel compartments by solving for linear and nonlinear parameters in the model. RESULTS Numerical forward simulation and phantom validation confirmed the ability of DECOMPOSE-QSM to separate the mixture of paramagnetic and diamagnetic components. The PCS obtained from temperature-variant brainstem imaging follows the Curie's Law, which further validated the model and the solver. Initial in vivo investigation of human brain images showed the ability to extract sub-voxel PCS and DCS sources that produce visually enhanced contrast between brain structures comparing to threshold QSM.
Collapse
Affiliation(s)
- Jingjia Chen
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA, USA
| | - Nan-Jie Gong
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA, USA; Vector Lab for Intelligent Medical Imaging and Neural Engineering, International Innovation Center of Tsinghua University, Shanghai, China
| | - Khallil Taverna Chaim
- LIM44, Instituto e Departamento de Radiologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
9
|
Ma L, Gholam Azad M, Dharmasivam M, Richardson V, Quinn RJ, Feng Y, Pountney DL, Tonissen KF, Mellick GD, Yanatori I, Richardson DR. Parkinson's disease: Alterations in iron and redox biology as a key to unlock therapeutic strategies. Redox Biol 2021; 41:101896. [PMID: 33799121 PMCID: PMC8044696 DOI: 10.1016/j.redox.2021.101896] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
A plethora of studies indicate that iron metabolism is dysregulated in Parkinson's disease (PD). The literature reveals well-documented alterations consistent with established dogma, but also intriguing paradoxical observations requiring mechanistic dissection. An important fact is the iron loading in dopaminergic neurons of the substantia nigra pars compacta (SNpc), which are the cells primarily affected in PD. Assessment of these changes reveal increased expression of proteins critical for iron uptake, namely transferrin receptor 1 and the divalent metal transporter 1 (DMT1), and decreased expression of the iron exporter, ferroportin-1 (FPN1). Consistent with this is the activation of iron regulator protein (IRP) RNA-binding activity, which is an important regulator of iron homeostasis, with its activation indicating cytosolic iron deficiency. In fact, IRPs bind to iron-responsive elements (IREs) in the 3ꞌ untranslated region (UTR) of certain mRNAs to stabilize their half-life, while binding to the 5ꞌ UTR prevents translation. Iron loading of dopaminergic neurons in PD may occur through these mechanisms, leading to increased neuronal iron and iron-mediated reactive oxygen species (ROS) generation. The "gold standard" histological marker of PD, Lewy bodies, are mainly composed of α-synuclein, the expression of which is markedly increased in PD. Of note, an atypical IRE exists in the α-synuclein 5ꞌ UTR that may explain its up-regulation by increased iron. This dysregulation could be impacted by the unique autonomous pacemaking of dopaminergic neurons of the SNpc that engages L-type Ca+2 channels, which imparts a bioenergetic energy deficit and mitochondrial redox stress. This dysfunction could then drive alterations in iron trafficking that attempt to rescue energy deficits such as the increased iron uptake to provide iron for key electron transport proteins. Considering the increased iron-loading in PD brains, therapies utilizing limited iron chelation have shown success. Greater therapeutic advancements should be possible once the exact molecular pathways of iron processing are dissected.
Collapse
Affiliation(s)
- L Ma
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - M Gholam Azad
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - M Dharmasivam
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - V Richardson
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - R J Quinn
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - Y Feng
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - D L Pountney
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - K F Tonissen
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - G D Mellick
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - I Yanatori
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - D R Richardson
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| |
Collapse
|
10
|
Kim S, Lee Y, Jeon CY, Jin YB, Oh S, Lee C. Observation of magnetic susceptibility changes within the thalamus: a comparative study between healthy and Parkinson’s disease afflicted cynomolgus monkeys using 7 T MRI. J Anal Sci Technol 2019. [DOI: 10.1186/s40543-019-0199-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Although the thalamus is known to modulate basal ganglia function related to motor control activity, the abnormal changes within the thalamus during distinct medical complications have been scarcely investigated. In order to explore the feasibility of assessing iron accumulation in the thalamus as an informative biomarker for Parkinson’s disease (PD), this study was designed to employ quantitative susceptibility mapping using a 7 T magnetic resonance imaging system in cynomolgus monkeys. A 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-injected cynomolgus monkey and a healthy control (HC) were examined by 7 T magnetic resonance imaging. Positron emission tomography with 18F-N-(3-fluoro propyl)-2ß-carboxymethoxy-3ß-(4-iodophenyl) nortropane was also employed to identify the relationship between iron deposits and dopamine depletion. All acquired values were averaged within the volume of interest of the nigrostriatal pathway.
Findings
Compared with the HC, the overall elevation of iron deposition within the thalamus in the Parkinson’s disease model (about 53.81% increase) was similar to that in the substantia nigra (54.81%) region. Substantial susceptibility changes were observed in the intralaminar part of the thalamus (about 70.78% increase). Additionally, we observed that in the Parkinson’s disease model, binding potential values obtained from positron emission tomography were considerably decreased in the thalamus (97.51%) and substantia nigra (92.48%).
Conclusions
The increased iron deposition in the thalamus showed negative correlation with dopaminergic activity in PD, supporting the idea that iron accumulation affects glutaminergic inputs and dopaminergic neurons. This investigation indicates that the remarkable susceptibility changes in the thalamus could be an initial major diagnostic biomarker for Parkinson’s disease-related motor symptoms.
Collapse
|
11
|
Khan AI, Liu J, Dutta P. Bayesian inference for parameter estimation in lactoferrin-mediated iron transport across blood-brain barrier. Biochim Biophys Acta Gen Subj 2019; 1864:129459. [PMID: 31682896 DOI: 10.1016/j.bbagen.2019.129459] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 10/11/2019] [Accepted: 10/22/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND In neurodegenerative diseases such as Alzheimer's and Parkinson's, excessive irons as well as lactoferrin (Lf), but not transferrin (Tf), have been found in and around the affected regions of the brain. These evidences suggest that lactoferrin plays a critical role during neurodegenerative diseases, although Lf-mediated iron transport across blood-brain barrier (BBB) is negligible compared to that of transferrin in normal condition. However, the kinetics of lactoferrins and lactoferrin-mediated iron transport are still unknown. METHOD To determine the kinetic rate constants of lactoferrin-mediated iron transport through BBB, a mass-action based ordinary differential equation model has been presented. A Bayesian framework is developed to estimate the kinetic rate parameters from posterior probability density functions. The iron transport across BBB is studied by considering both Lf- and Tf-mediated pathways for both normal and pathologic conditions. RESULTS Using the point estimates of kinetic parameters, our model can effectively reproduce the experimental data of iron transport through BBB endothelial cells. The robustness of the model and parameter estimation process are further verified by perturbation of kinetic parameters. Our results show that surge in high-affinity receptor density increases lactoferrin as well as iron in the brain. CONCLUSIONS Due to the lack of a feedback loop such as iron regulatory proteins (IRPs) for lactoferrin, iron can transport to the brain continuously, which might increase brain iron to pathological levels and can contribute to neurodegeneration. GENERAL SIGNIFICANCE This study provides an improved understanding of presence of lactoferrin and iron in the brain during neurodegenerative diseases.
Collapse
Affiliation(s)
- Aminul Islam Khan
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164-2920, United States of America
| | - Jin Liu
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164-2920, United States of America
| | - Prashanta Dutta
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164-2920, United States of America.
| |
Collapse
|
12
|
Guan X, Zhang Y, Wei H, Guo T, Zeng Q, Zhou C, Wang J, Gao T, Xuan M, Gu Q, Xu X, Huang P, Pu J, Zhang B, Liu C, Zhang M. Iron-related nigral degeneration influences functional topology mediated by striatal dysfunction in Parkinson's disease. Neurobiol Aging 2019; 75:83-97. [PMID: 30554085 PMCID: PMC6538269 DOI: 10.1016/j.neurobiolaging.2018.11.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 12/14/2022]
Abstract
In Parkinson's disease (PD), iron accumulation in the substantia nigra (SN) exacerbates oxidative stress and α-synuclein aggregation, leading to neuronal death. However, the influence of iron-related nigral degeneration on the subcortical function and global network configuration in PD remains unknown. Ninety PD patients and 38 normal controls underwent clinical assessments and multimodality magnetic resonance imaging scans. Iron accumulation in the inferior SN and disrupted functional connectivity between the bilateral striatums were observed in PD, and negative correlation between them was found in the whole population. The binarized functional network exhibited enhanced global efficiency and reduced local efficiency while the weighted functional network exhibited reduction in both, and both changes were correlated with nigral iron accumulation in PD. Mediation analysis demonstrated that the functional connectivity between bilateral striatums was a mediator between the nigral iron accumulation and weighted functional network alterations. In conclusion, our findings reveal that iron-related nigral degeneration possibly influences the functional topology mediated by striatal dysfunction, which extends the scientific understanding of PD pathogenesis.
Collapse
Affiliation(s)
- Xiaojun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, USA
| | - Yuyao Zhang
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, USA
| | - Hongjiang Wei
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, USA
| | - Tao Guo
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiaoling Zeng
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng Zhou
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqiu Wang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Gao
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Xuan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Quanquan Gu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peiyu Huang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiali Pu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA.
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
13
|
Núñez MT, Hidalgo C. Noxious Iron-Calcium Connections in Neurodegeneration. Front Neurosci 2019; 13:48. [PMID: 30809110 PMCID: PMC6379295 DOI: 10.3389/fnins.2019.00048] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/18/2019] [Indexed: 12/26/2022] Open
Abstract
Iron and calcium share the common feature of being essential for normal neuronal function. Iron is required for mitochondrial function, synaptic plasticity, and the development of cognitive functions whereas cellular calcium signals mediate neurotransmitter exocytosis, axonal growth and synaptic plasticity, and control the expression of genes involved in learning and memory processes. Recent studies have revealed that cellular iron stimulates calcium signaling, leading to downstream activation of kinase cascades engaged in synaptic plasticity. The relationship between calcium and iron is Janus-faced, however. While under physiological conditions iron-mediated reactive oxygen species generation boosts normal calcium-dependent signaling pathways, excessive iron levels promote oxidative stress leading to the upsurge of unrestrained calcium signals that damage mitochondrial function, among other downstream targets. Similarly, increases in mitochondrial calcium to non-physiological levels result in mitochondrial dysfunction and a predicted loss of iron homeostasis. Hence, if uncontrolled, the iron/calcium self-feeding cycle becomes deleterious to neuronal function, leading eventually to neuronal death. Here, we review the multiple cell-damaging responses generated by the unregulated iron/calcium self-feeding cycle, such as excitotoxicity, free radical-mediated lipid peroxidation, and the oxidative modification of crucial components of iron and calcium homeostasis/signaling: the iron transporter DMT1, plasma membrane, and intracellular calcium channels and pumps. We discuss also how iron-induced dysregulation of mitochondrial calcium contributes to the generation of neurodegenerative conditions, including Alzheimer’s disease (AD) and Parkinson’s disease (PD).
Collapse
Affiliation(s)
- Marco Tulio Núñez
- Iron and Neuroregeneration Laboratory, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- Calcium Signaling Laboratory, Biomedical Research Institute, CEMC, Physiology and Biophysics Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
14
|
Riboflavin in Human Health: A Review of Current Evidences. ADVANCES IN FOOD AND NUTRITION RESEARCH 2018; 83:57-81. [PMID: 29477226 DOI: 10.1016/bs.afnr.2017.11.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Riboflavin is a water-soluble vitamin, which was initially isolated from milk. There are two coenzyme forms of riboflavin, flavin mononucleotide and flavin adenine dinucleotide, in which riboflavin plays important roles in the enzymatic reactions. Riboflavin is found in a wide variety of animal and plant foods. Meat and dairy products are the major contributors of riboflavin dietary intake. In this chapter, the latest evidence on the relationship between riboflavin status and specific health risks will be reviewed. Also, some of the mechanisms by which riboflavin exerts its roles will be discussed. The evidence accrued suggests that riboflavin is an antioxidant nutrient which may prevent lipid peroxidation and reperfusion oxidative injury. Moreover, riboflavin deficiency may increase the risk of some cancers. Riboflavin may also exert a neuroprotective effects in some neurological disorders (e.g., Parkinson disease, migraine, and multiple sclerosis) through its role in some pathways that are hypothesized to be impaired in neurological disorders such as antioxidation, myelin formation, mitochondrial function, and iron metabolism.
Collapse
|
15
|
Ma RE, Ward EJ, Yeh CL, Snyder S, Long Z, Gokalp Yavuz F, Zauber SE, Dydak U. Thalamic GABA levels and occupational manganese neurotoxicity: Association with exposure levels and brain MRI. Neurotoxicology 2018; 64:30-42. [PMID: 28873337 PMCID: PMC5891096 DOI: 10.1016/j.neuro.2017.08.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/30/2017] [Accepted: 08/30/2017] [Indexed: 01/08/2023]
Abstract
Excessive occupational exposure to Manganese (Mn) has been associated with clinical symptoms resembling idiopathic Parkinson's disease (IPD), impairing cognitive and motor functions. Several studies point towards an involvement of the brain neurotransmitter system in Mn intoxication, which is hypothesized to be disturbed prior to onset of symptoms. Edited Magnetic Resonance Spectroscopy (MRS) offers the unique possibility to measure γ-amminobutyric acid (GABA) and other neurometabolites in vivo non-invasively in workers exposed to Mn. In addition, the property of Mn as Magnetic Resonance Imaging (MRI) contrast agent may be used to study Mn deposition in the human brain. In this study, using MRI, MRS, personal air sampling at the working place, work history questionnaires, and neurological assessment (UPDRS-III), the effects of chronic Mn exposure on the thalamic GABAergic system was studied in a group of welders (N=39) with exposure to Mn fumes in a typical occupational setting. Two subgroups of welders with different exposure levels (Low: N=26; mean air Mn=0.13±0.1mg/m3; High: N=13; mean air Mn=0.23±0.18mg/m3), as well as unexposed control workers (N=22, mean air Mn=0.002±0.001mg/m3) were recruited. The group of welders with higher exposure showed a significant increase of thalamic GABA levels by 45% (p<0.01, F(1,33)=9.55), as well as significantly worse performance in general motor function (p<0.01, F(1,33)=11.35). However, welders with lower exposure did not differ from the controls in GABA levels or motor performance. Further, in welders the thalamic GABA levels were best predicted by past-12-months exposure levels and were influenced by the Mn deposition in the substantia nigra and globus pallidus. Importantly, both thalamic GABA levels and motor function displayed a non-linear pattern of response to Mn exposure, suggesting a threshold effect.
Collapse
Affiliation(s)
- Ruoyun E Ma
- School of Health Sciences, Purdue University, West Lafayette, IN, USA; Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Eric J Ward
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Chien-Lin Yeh
- School of Health Sciences, Purdue University, West Lafayette, IN, USA; Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sandy Snyder
- School of Health Sciences, Purdue University, West Lafayette, IN, USA; Department of Speech, Language and Hearing Sciences, Purdue University, West Lafayette, IN, USA
| | - Zaiyang Long
- School of Health Sciences, Purdue University, West Lafayette, IN, USA; Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Fulya Gokalp Yavuz
- Department of Statistics, Purdue University, IN, USA; Yildiz Technical University, Istanbul, Turkey
| | - S Elizabeth Zauber
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ulrike Dydak
- School of Health Sciences, Purdue University, West Lafayette, IN, USA; Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Speech, Language and Hearing Sciences, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
16
|
Amireddy N, Puttapaka SN, Vinnakota RL, Ravuri HG, Thonda S, Kalivendi SV. The unintended mitochondrial uncoupling effects of the FDA-approved anti-helminth drug nitazoxanide mitigates experimental parkinsonism in mice. J Biol Chem 2017; 292:15731-15743. [PMID: 28798236 DOI: 10.1074/jbc.m117.791863] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/31/2017] [Indexed: 01/04/2023] Open
Abstract
Mitochondria play a primary role in the pathophysiology of Parkinson's disease (PD), and small molecules that counteract the initial stages of disease may offer therapeutic benefit. In this regard, we have examined whether the off-target effects of the Food and Drug Administration (FDA)-approved anti-helminth drug nitazoxanide (NTZ) on mitochondrial respiration could possess any therapeutic potential for PD. Results indicate that MPP+-induced loss in oxygen consumption rate (OCR) and ATP production by mitochondria were ameliorated by NTZ in real time by virtue of its mild uncoupling effect. Pretreatment of cells with NTZ mitigated MPP+-induced loss in mitochondrial OCR and reactive oxygen species (ROS). Similarly, addition of NTZ to cells pretreated with MPP+ could reverse block in mitochondrial OCR and reactive oxygen species induced by MPP+ in real time. The observed effects of NTZ were found to be transient and reversible as removal of NTZ from incubation medium restored the mitochondrial respiration to that of controls. Apoptosis induced by MPP+ was ameliorated by NTZ in a dose-dependent manner. In vivo results demonstrated that oral administration of NTZ (50 mg/kg) in an acute MPTP mouse model of PD conferred significant protection against the loss of tyrosine hydroxylase (TH)-positive neurons of substantia nigra. Based on the above observations we believe that repurposing of NTZ for PD may offer therapeutic benefit.
Collapse
Affiliation(s)
| | | | | | - Halley G Ravuri
- Pharmacology and Toxicology, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad - 500 007, Telangana State, India
| | | | | |
Collapse
|
17
|
Meng FX, Hou JM, Sun TS. In vivo evaluation of microglia activation by intracranial iron overload in central pain after spinal cord injury. J Orthop Surg Res 2017; 12:75. [PMID: 28521818 PMCID: PMC5437601 DOI: 10.1186/s13018-017-0578-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/24/2017] [Indexed: 11/23/2022] Open
Abstract
Background Central pain (CP) is a common clinical problem in patients with spinal cord injury (SCI). Recent studies found the pathogenesis of CP was related to the remodeling of the brain. We investigate the roles of iron overload and subsequent microglia activate in the remodeling of the brain after SCI. Methods An SCI-induced CP model was established in Sprague-Dawley rats that were randomly assigned to SCI, sham operation, deferoxamine (DFX), minocycline, and nitric oxide synthase inhibitor treatment groups. At 12 weeks, pain behavior and thermal pain threshold were evaluated in each group, and iron transferrin receptor (TfR)1 and ferritin (Fn) mRNA, as well as iron-regulatory protein (IRP)1, FN, lactoferrin, and nuclear factor (NF)-κB protein levels in the rat brains were measured. Microglia proliferation and differentiation and IRP1 expression were evaluated by immunohistochemistry. Results Autophagy was observed in rats after SCI, accompanied by reduced latency of thermal pain, increased iron content and IRP1 and NF-κB levels in the hindlimb sensory area, hippocampus, and thalamus, and decreased Fn levels in the hindlimb sensory area. TfR1 mRNA expression was upregulated in activated microglia. Treatment with an iron-chelating agent, or inhibitors of nitric oxide synthase or microglia suppressed microglia proliferation. Conclusions SCI may induce intracranial iron overload, which activates microglia via NF-κB signaling. Microglia secrete inflammatory factors that induce neuronal damage and lead to CP. Treatment with an iron-chelating agent or NF-κB or microglia inhibitors can relieve CP resulting from SCI.
Collapse
Affiliation(s)
- Fan Xing Meng
- Third Military Medical University, No. 30 Gaotanyan Street, Chongqing, 400038, China.,Department of Orthopedics, Chinese PLA Army General Hospital, Dongcheng District, Nanmencang No. 5, Beijing, 100700, China
| | - Jing Ming Hou
- Department of Orthopedics, Chinese PLA Army General Hospital, Dongcheng District, Nanmencang No. 5, Beijing, 100700, China.,Department of Rehabilitation, Southwest Hospital, Third Military Medical University, No. 30 Gaotanyan Street, Chongqing, 400038, China
| | - Tian Sheng Sun
- Third Military Medical University, No. 30 Gaotanyan Street, Chongqing, 400038, China. suntiansheng-@163.com.,Department of Orthopedics, Chinese PLA Army General Hospital, Dongcheng District, Nanmencang No. 5, Beijing, 100700, China. suntiansheng-@163.com
| |
Collapse
|
18
|
Meng FX, Hou JM, Sun TS. Effect of oxidative stress induced by intracranial iron overload on central pain after spinal cord injury. J Orthop Surg Res 2017; 12:24. [PMID: 28178997 PMCID: PMC5299723 DOI: 10.1186/s13018-017-0526-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 01/28/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Central pain (CP) is a common clinical problem in patients with spinal cord injury (SCI). Recent studies found the pathogenesis of CP was related to the remodeling of the brain. We investigate the roles of iron overload and subsequent oxidative stress in the remodeling of the brain after SCI. METHODS We established a rat model of central pain after SCI. Rats were divided randomly into four groups: SCI, sham operation, SCI plus deferoxamine (DFX) intervention, and SCI plus nitric oxide synthase (NOS) inhibitor treatment. Pain behavior was observed and thermal pain threshold was measured regularly, and brain levels of iron, transferrin receptor 1 (TfR1), ferritin (Fn), and lactoferrin (Lf), were detected in the different groups 12 weeks after establishment of the model. RESULTS Rats demonstrated self-biting behavior after SCI. Furthermore, the latent period of thermal pain was reduced and iron levels in the hind limb sensory area, hippocampus, and thalamus increased after SCI. Iron-regulatory protein (IRP) 1 levels increased in the hind limb sensory area, while Fn levels decreased. TfR1 mRNA levels were also increased and oxidative stress was activated. Oxidative stress could be inhibited by ferric iron chelators and NOS inhibitors. CONCLUSIONS SCI may cause intracranial iron overload through the NOS-iron-responsive element/IRP pathway, resulting in central pain mediated by the oxidative stress response. Iron chelators and oxidative stress inhibitors can effectively relieve SCI-associated central pain.
Collapse
Affiliation(s)
- Fan Xing Meng
- Third Military Medical University, No. 30 Gaotanyan Street, 400038 Chongqing, China
- Department of Orthopedics, Chinese PLA Army General Hospital, Dongcheng District, Nanmencang No. 5, 100700 Beijing, China
| | - Jing Ming Hou
- Department of Orthopedics, Chinese PLA Army General Hospital, Dongcheng District, Nanmencang No. 5, 100700 Beijing, China
- Southwest Hospital, Third Military Medical University, No. 30 Gaotanyan Street, 400038 Chongqing, China
| | - Tian Sheng Sun
- Third Military Medical University, No. 30 Gaotanyan Street, 400038 Chongqing, China
- Department of Orthopedics, Chinese PLA Army General Hospital, Dongcheng District, Nanmencang No. 5, 100700 Beijing, China
| |
Collapse
|
19
|
Morgan LA, Grundmann O. Preclinical and Potential Applications of Common Western Herbal Supplements as Complementary Treatment in Parkinson's Disease. J Diet Suppl 2017; 14:453-466. [DOI: 10.1080/19390211.2016.1263710] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Luke A. Morgan
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Oliver Grundmann
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| |
Collapse
|
20
|
You LH, Li Z, Duan XL, Zhao BL, Chang YZ, Shi ZH. Mitochondrial ferritin suppresses MPTP-induced cell damage by regulating iron metabolism and attenuating oxidative stress. Brain Res 2016; 1642:33-42. [DOI: 10.1016/j.brainres.2016.03.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 02/22/2016] [Accepted: 03/15/2016] [Indexed: 01/22/2023]
|
21
|
Shirai Y, Mori A, Nakahara T, Sakamoto K, Ishii K. Deferiprone Protects against Photoreceptor Degeneration Induced by Tunicamycin in the Rat Retina. Biol Pharm Bull 2016; 38:1076-80. [PMID: 26133718 DOI: 10.1248/bpb.b15-00185] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endoplasmic reticulum stress has been reported to be involved in the pathogenesis of retinitis pigmentosa, macular degeneration and diabetic retinopathy. In the present study, we examined the effects of deferiprone, an iron chelator, on photoreceptor degeneration induced by tunicamycin (300 nmol/eye), an endoplasmic reticulum stress inducer, in the rat retina. Scotopic electroretinogram measurement and morphometric evaluation were done 7 d after the injection of tunicamycin. In the scotopic electroretinogram, intravitreal deferiprone (5 nmol/eye) injected simultaneously with tunicamycin significantly reduced the decreases in a- and b-wave amplitudes induced by tunicamycin. Morphometric evaluation showed that deferiprone significantly reduced thinning of the outer nuclear layer, the inner segment and the outer segment. These results suggest that iron chelation therapy may be a good candidate for the treatment of eye diseases related to endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Yousuke Shirai
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences
| | | | | | | | | |
Collapse
|
22
|
The novel mitochondrial iron chelator 5-((methylamino)methyl)-8-hydroxyquinoline protects against mitochondrial-induced oxidative damage and neuronal death. Biochem Biophys Res Commun 2015; 463:787-92. [DOI: 10.1016/j.bbrc.2015.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/02/2015] [Indexed: 11/21/2022]
|
23
|
Manganese-Induced Parkinsonism and Parkinson's Disease: Shared and Distinguishable Features. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:7519-40. [PMID: 26154659 PMCID: PMC4515672 DOI: 10.3390/ijerph120707519] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/12/2014] [Accepted: 01/06/2015] [Indexed: 11/30/2022]
Abstract
Manganese (Mn) is an essential trace element necessary for physiological processes that support development, growth and neuronal function. Secondary to elevated exposure or decreased excretion, Mn accumulates in the basal ganglia region of the brain and may cause a parkinsonian-like syndrome, referred to as manganism. The present review discusses the advances made in understanding the essentiality and neurotoxicity of Mn. We review occupational Mn-induced parkinsonism and the dynamic modes of Mn transport in biological systems, as well as the detection and pharmacokinetic modeling of Mn trafficking. In addition, we review some of the shared similarities, pathologic and clinical distinctions between Mn-induced parkinsonism and Parkinson’s disease. Where possible, we review the influence of Mn toxicity on dopamine, gamma aminobutyric acid (GABA), and glutamate neurotransmitter levels and function. We conclude with a survey of the preventive and treatment strategies for manganism and idiopathic Parkinson’s disease (PD).
Collapse
|
24
|
Bar-Am O, Amit T, Kupershmidt L, Aluf Y, Mechlovich D, Kabha H, Danovitch L, Zurawski VR, Youdim MB, Weinreb O. Neuroprotective and neurorestorative activities of a novel iron chelator-brain selective monoamine oxidase-A/monoamine oxidase-B inhibitor in animal models of Parkinson's disease and aging. Neurobiol Aging 2015; 36:1529-42. [DOI: 10.1016/j.neurobiolaging.2014.10.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/19/2014] [Accepted: 10/19/2014] [Indexed: 10/24/2022]
|
25
|
Ghadery C, Pirpamer L, Hofer E, Langkammer C, Petrovic K, Loitfelder M, Schwingenschuh P, Seiler S, Duering M, Jouvent E, Schmidt H, Fazekas F, Mangin JF, Chabriat H, Dichgans M, Ropele S, Schmidt R. R2* mapping for brain iron: associations with cognition in normal aging. Neurobiol Aging 2015; 36:925-32. [DOI: 10.1016/j.neurobiolaging.2014.09.013] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 09/11/2014] [Accepted: 09/15/2014] [Indexed: 11/26/2022]
|
26
|
Lorio S, Lutti A, Kherif F, Ruef A, Dukart J, Chowdhury R, Frackowiak RS, Ashburner J, Helms G, Weiskopf N, Draganski B. Disentangling in vivo the effects of iron content and atrophy on the ageing human brain. Neuroimage 2014; 103:280-289. [PMID: 25264230 PMCID: PMC4263529 DOI: 10.1016/j.neuroimage.2014.09.044] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 09/15/2014] [Accepted: 09/17/2014] [Indexed: 01/06/2023] Open
Abstract
Evidence from magnetic resonance imaging (MRI) studies shows that healthy aging is associated with profound changes in cortical and subcortical brain structures. The reliable delineation of cortex and basal ganglia using automated computational anatomy methods based on T1-weighted images remains challenging, which results in controversies in the literature. In this study we use quantitative MRI (qMRI) to gain an insight into the microstructural mechanisms underlying tissue ageing and look for potential interactions between ageing and brain tissue properties to assess their impact on automated tissue classification. To this end we acquired maps of longitudinal relaxation rate R1, effective transverse relaxation rate R2* and magnetization transfer - MT, from healthy subjects (n=96, aged 21-88 years) using a well-established multi-parameter mapping qMRI protocol. Within the framework of voxel-based quantification we find higher grey matter volume in basal ganglia, cerebellar dentate and prefrontal cortex when tissue classification is based on MT maps compared with T1 maps. These discrepancies between grey matter volume estimates can be attributed to R2* - a surrogate marker of iron concentration, and further modulation by an interaction between R2* and age, both in cortical and subcortical areas. We interpret our findings as direct evidence for the impact of ageing-related brain tissue property changes on automated tissue classification of brain structures using SPM12. Computational anatomy studies of ageing and neurodegeneration should acknowledge these effects, particularly when inferring about underlying pathophysiology from regional cortex and basal ganglia volume changes.
Collapse
Affiliation(s)
- S Lorio
- LREN, Dept. of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland
| | - A Lutti
- LREN, Dept. of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland
| | - F Kherif
- LREN, Dept. of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland
| | - A Ruef
- LREN, Dept. of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland
| | - J Dukart
- LREN, Dept. of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland
| | - R Chowdhury
- Wellcome Trust Centre for Neuroimaging, UCL Institute of Neurology, UCL, London, UK
| | - R S Frackowiak
- LREN, Dept. of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland
| | - J Ashburner
- Wellcome Trust Centre for Neuroimaging, UCL Institute of Neurology, UCL, London, UK
| | - G Helms
- University Medical Centre, UMG, Dept. of Cognitive Neurology, Göttingen, Germany
| | - N Weiskopf
- Wellcome Trust Centre for Neuroimaging, UCL Institute of Neurology, UCL, London, UK
| | - B Draganski
- LREN, Dept. of Clinical Neurosciences, CHUV, University of Lausanne, Lausanne Switzerland; Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| |
Collapse
|
27
|
Adachi Y, Sato N, Saito Y, Kimura Y, Nakata Y, Ito K, Kamiya K, Matsuda H, Tsukamoto T, Ogawa M. Usefulness of SWI for the Detection of Iron in the Motor Cortex in Amyotrophic Lateral Sclerosis. J Neuroimaging 2014; 25:443-51. [PMID: 24888543 DOI: 10.1111/jon.12127] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 01/07/2014] [Accepted: 03/02/2014] [Indexed: 11/28/2022] Open
Abstract
PURPOSE The purpose of the present retrospective study was to evaluate the sensitivity of susceptibility-weighted imaging (SWI) compared to conventional spin-echo T2-weighted and T2*-weighted images in detecting iron deposition in the motor cortex of amyotrophic lateral sclerosis (ALS) patients in comparison with age-matched normal controls. We also investigated the etiology of the low signal referring to the pathology of one autopsy case. METHODS This retrospective magnetic resonance (MR) study included 23 ALS patients and 28 age-matched normal controls. The signal intensity of the motor cortex was scored by SWI, conventional T2-weighted images and T2*-weighted images. A postmortem study of one patient was also performed. RESULTS On SWI, there was a significant difference between the precentral cortical signal intensity scores in the ALS patients and the controls (P < .0001). The total scores of signal intensities of the precentral cortex were positively correlated with age in the normal controls (r = .494), but no correlation was observed in the ALS patients. The postmortem study showed intensely stained microglias and macrophages after antiferritin antibody staining in the precentral cortices. CONCLUSIONS Decreased signal intensity of the motor cortex on SWI may serve a useful role in ALS diagnoses, particularly in young patients. MR images were also helpful for speculating on the etiology of ALS.
Collapse
Affiliation(s)
- Yuko Adachi
- National Center Hospital of Neurology and Psychiatry Radiology, Kodaira, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Muller M, Leavitt BR. Iron dysregulation in Huntington's disease. J Neurochem 2014; 130:328-50. [PMID: 24717009 DOI: 10.1111/jnc.12739] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 03/19/2014] [Accepted: 04/07/2014] [Indexed: 12/13/2022]
Abstract
Huntington's disease (HD) is one of many neurodegenerative diseases with reported alterations in brain iron homeostasis that may contribute to neuropathogenesis. Iron accumulation in the specific brain areas of neurodegeneration in HD has been proposed based on observations in post-mortem tissue and magnetic resonance imaging studies. Altered magnetic resonance imaging signal within specific brain regions undergoing neurodegeneration has been consistently reported and interpreted as altered levels of brain iron. Biochemical studies using various techniques to measure iron species in human samples, mouse tissue, or in vitro has generated equivocal data to support such an association. Whether elevated brain iron occurs in HD, plays a significant contributing role in HD pathogenesis, or is a secondary effect remains currently unclear.
Collapse
Affiliation(s)
- Michelle Muller
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, University of British Columbia and Children's and Women's Hospital, Vancouver, British Columbia, Canada
| | | |
Collapse
|
29
|
Mot AI, Wedd AG, Sinclair L, Brown DR, Collins SJ, Brazier MW. Metal attenuating therapies in neurodegenerative disease. Expert Rev Neurother 2014; 11:1717-45. [DOI: 10.1586/ern.11.170] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
30
|
Weinreb O, Mandel S, Youdim MBH, Amit T. Targeting dysregulation of brain iron homeostasis in Parkinson's disease by iron chelators. Free Radic Biol Med 2013; 62:52-64. [PMID: 23376471 DOI: 10.1016/j.freeradbiomed.2013.01.017] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 01/09/2013] [Accepted: 01/14/2013] [Indexed: 10/27/2022]
Abstract
Brain iron accumulation has been implicated in a host of chronic neurological diseases, including Parkinson's disease (PD). The elevated iron levels observed in the substantia nigra of PD subjects have been suggested to incite the generation of reactive oxygen species and intracellular α-synuclein aggregation, terminating in the oxidative neuronal destruction of this brain area. Thus, elucidation of the molecular mechanisms involved in iron dysregulation and oxidative stress-induced neurodegeneration is a crucial step in deciphering PD pathology and in developing novel iron-complexing compounds aimed at restoring brain iron homeostasis and attenuating neurodegeneration. This review discusses the involvement of dysregulation of brain iron homeostasis in PD pathology, with an emphasis on the potential effectiveness of naturally occurring compounds and novel iron-chelating/antioxidant therapeutic hybrid molecules, exerting a spectrum of neuroprotective interrelated activities: antioxidant/monoamine oxidase inhibition, activation of the hypoxia-inducible factor (HIF)-1 signaling pathway, induction of HIF-1 target iron-regulatory and antioxidative genes, and inhibition of α-synuclein accumulation and aggregation.
Collapse
Affiliation(s)
- Orly Weinreb
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research, Department of Pharmacology, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel.
| | - Silvia Mandel
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research, Department of Pharmacology, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Moussa B H Youdim
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research, Department of Pharmacology, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Tamar Amit
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research, Department of Pharmacology, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|
31
|
Cattani D, Goulart PB, Cavalli VLDLO, Winkelmann-Duarte E, Dos Santos AQ, Pierozan P, de Souza DF, Woehl VM, Fernandes MC, Silva FRMB, Gonçalves CA, Pessoa-Pureur R, Zamoner A. Congenital hypothyroidism alters the oxidative status, enzyme activities and morphological parameters in the hippocampus of developing rats. Mol Cell Endocrinol 2013; 375:14-26. [PMID: 23693027 DOI: 10.1016/j.mce.2013.05.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 04/17/2013] [Accepted: 05/01/2013] [Indexed: 11/23/2022]
Abstract
Congenital hypothyroidism is associated with delay in cell migration and proliferation in brain tissue, impairment of synapse formation, misregulation of neurotransmitters, hypomyelination and mental retardation. However, the mechanisms underlying the neuropsychological deficits observed in congenital hypothyroidism are not completely understood. In the present study we proposed a mechanism by which hypothyroidism leads to hippocampal neurotoxicity. Congenital hypothyroidism induces c-Jun-N-terminal kinase (JNK) pathway activation leading to hyperphosphorylation of the glial fibrillary acidic protein (GFAP), vimentin and neurofilament subunits from hippocampal astrocytes and neurons, respectively. Moreover, hyperphosphorylation of the cytoskeletal proteins was not reversed by T3 and poorly reversed by T4. In addition, congenital hypothyroidism is associated with downregulation of astrocyte glutamate transporters (GLAST and GLT-1) leading to decreased glutamate uptake and subsequent influx of Ca(2+) through N-methyl-D-aspartate (NMDA) receptors. The Na(+)-coupled (14)C-α-methyl-amino-isobutyric acid ((14)C-MeAIB) accumulation into hippocampal cells also might cause an increase in the intracellular Ca(2+) concentration by opening voltage-dependent calcium channels (VDCC). The excessive influx of Ca(2+) through NMDA receptors and VDCCs might lead to an overload of Ca(2+) within the cells, which set off glutamate excitotoxicity and oxidative stress. The inhibited acetylcholinesterase (AChE) activity might also induce Ca(2+) influx. The inhibited glucose-6-phosphate dehydrogenase (G6PD) and gamma-glutamyl transferase (GGT) activities, associated with altered glutamate and neutral amino acids uptake could somehow affect the GSH turnover, the antioxidant defense system, as well as the glutamate-glutamine cycle. Reduced levels of S100B and glial fibrillary acidic protein (GFAP) take part of the hypothyroid condition, suggesting a compromised astroglial/neuronal neurometabolic coupling which is probably related to the neurotoxic damage in hypothyroid brain.
Collapse
Affiliation(s)
- Daiane Cattani
- Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Jellinger KA. The relevance of metals in the pathophysiology of neurodegeneration, pathological considerations. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2013; 110:1-47. [PMID: 24209432 DOI: 10.1016/b978-0-12-410502-7.00002-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurodegenerative disorders are featured by a variety of pathological conditions that share similar critical processes, such as oxidative stress, free radical activity, proteinaceous aggregations, mitochondrial dysfunctions, and energy failure. They are mediated or triggered by an imbalance of metal ions leading to changes of critical biological systems and initiating a cascade of events finally leading to neurodegeneration and cell death. Their causes are multifactorial, and although the source of the shift in oxidative homeostasis is still unclear, current evidence points to changes in the balance of redox transition metals, especially iron, copper, and other trace metals. They are present at elevated levels in Alzheimer disease, Parkinson disease, multisystem atrophy, etc., while in other neurodegenerative disorders, copper, zinc, aluminum, and manganese are involved. This chapter will review the recent advances of the role of metals in the pathogenesis and pathophysiology of major neurodegenerative diseases and discuss the use of chelating agents as potential therapies for metal-related disorders.
Collapse
|
33
|
Hare DJ, Adlard PA, Doble PA, Finkelstein DI. Metallobiology of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine neurotoxicity. Metallomics 2013; 5:91-109. [DOI: 10.1039/c2mt20164j] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
34
|
Farina M, Avila DS, da Rocha JBT, Aschner M. Metals, oxidative stress and neurodegeneration: a focus on iron, manganese and mercury. Neurochem Int 2012; 62:575-94. [PMID: 23266600 DOI: 10.1016/j.neuint.2012.12.006] [Citation(s) in RCA: 364] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 02/08/2023]
Abstract
Essential metals are crucial for the maintenance of cell homeostasis. Among the 23 elements that have known physiological functions in humans, 12 are metals, including iron (Fe) and manganese (Mn). Nevertheless, excessive exposure to these metals may lead to pathological conditions, including neurodegeneration. Similarly, exposure to metals that do not have known biological functions, such as mercury (Hg), also present great health concerns. This review focuses on the neurodegenerative mechanisms and effects of Fe, Mn and Hg. Oxidative stress (OS), particularly in mitochondria, is a common feature of Fe, Mn and Hg toxicity. However, the primary molecular targets triggering OS are distinct. Free cationic iron is a potent pro-oxidant and can initiate a set of reactions that form extremely reactive products, such as OH. Mn can oxidize dopamine (DA), generating reactive species and also affect mitochondrial function, leading to accumulation of metabolites and culminating with OS. Cationic Hg forms have strong affinity for nucleophiles, such as -SH and -SeH. Therefore, they target critical thiol- and selenol-molecules with antioxidant properties. Finally, we address the main sources of exposure to these metals, their transport mechanisms into the brain, and therapeutic modalities to mitigate their neurotoxic effects.
Collapse
Affiliation(s)
- Marcelo Farina
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88040-900 Florianópolis, SC, Brazil
| | | | | | | |
Collapse
|
35
|
NGP1-01, a multi-targeted polycyclic cage amine, attenuates brain endothelial cell death in iron overload conditions. Brain Res 2012; 1489:133-9. [DOI: 10.1016/j.brainres.2012.10.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/12/2012] [Accepted: 10/14/2012] [Indexed: 11/23/2022]
|
36
|
The hemochromatosis proteins HFE, TfR2, and HJV form a membrane-associated protein complex for hepcidin regulation. J Hepatol 2012; 57:1052-60. [PMID: 22728873 DOI: 10.1016/j.jhep.2012.06.015] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 06/11/2012] [Accepted: 06/13/2012] [Indexed: 12/29/2022]
Abstract
BACKGROUND & AIMS The hereditary hemochromatosis-associated membrane proteins HFE, TfR2, and HJV are required for adequate hepatic expression of the iron hormone hepcidin. While the genetic interactions are clear, it remains elusive how bone morphogenetic protein co-receptor HJV functions together with HFE and TfR2 to activate hepcidin transcription via the BMP-SMAD signaling pathway. Here, we investigate whether HFE, TfR2, and HJV physically interact on the surface of hepatocytes. METHODS We explore protein-protein interactions by glycerol gradient sedimentation assays and co-immunoprecipitation analyses in transfected HuH7 hepatoma-derived cells. RESULTS Our data demonstrate that HFE and TfR2 bind HJV in a non-competitive manner. Co-immunoprecipitation analyses provide direct experimental evidence that HFE, TfR2, and HJV form a multi-protein membrane complex. Our experiments show that like TfR2, HJV competes with TfR1 for binding to HFE, indicating that the expression of TfR2 and HJV may be critical for iron sensing. We identify residues 120-139 of the TfR2 extra-cellular domain as the critical amino acids required for the binding of both HFE and HJV. Interestingly, RGMA, a central nervous system homolog, can substitute for HJV in the complex and promote hepcidin transcription, implicating RGMA in the local control of hepcidin in the CNS. CONCLUSIONS Taken together, our findings provide a biochemical basis for hepcidin control by HFE, TfR2, and HJV.
Collapse
|
37
|
Menzie J, Pan C, Prentice H, Wu JY. Taurine and central nervous system disorders. Amino Acids 2012; 46:31-46. [DOI: 10.1007/s00726-012-1382-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 07/27/2012] [Indexed: 01/28/2023]
|
38
|
Mounsey RB, Teismann P. Chelators in the treatment of iron accumulation in Parkinson's disease. Int J Cell Biol 2012; 2012:983245. [PMID: 22754573 PMCID: PMC3382398 DOI: 10.1155/2012/983245] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 03/18/2012] [Indexed: 12/21/2022] Open
Abstract
Iron is an essential element in the metabolism of all cells. Elevated levels of the metal have been found in the brains of patients of numerous neurodegenerative disorders, including Parkinson's disease (PD). The pathogenesis of PD is largely unknown, although it is thought through studies with experimental models that oxidative stress and dysfunction of brain iron homeostasis, usually a tightly regulated process, play significant roles in the death of dopaminergic neurons. Accumulation of iron is present at affected neurons and associated microglia in the substantia nigra of PD patients. This additional free-iron has the capacity to generate reactive oxygen species, promote the aggregation of α-synuclein protein, and exacerbate or even cause neurodegeneration. There are various treatments aimed at reversing this pathologic increase in iron content, comprising both synthetic and natural iron chelators. These include established drugs, which have been used to treat other disorders related to iron accumulation. This paper will discuss how iron dysregulation occurs and the link between increased iron and oxidative stress in PD, including the mechanism by which these processes lead to cell death, before assessing the current pharmacotherapies aimed at restoring normal iron redox and new chelation strategies undergoing research.
Collapse
Affiliation(s)
- Ross B. Mounsey
- School of Medical Sciences, College of Life Sciences and Medicine Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Peter Teismann
- School of Medical Sciences, College of Life Sciences and Medicine Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| |
Collapse
|
39
|
Kwan JY, Jeong SY, Van Gelderen P, Deng HX, Quezado MM, Danielian LE, Butman JA, Chen L, Bayat E, Russell J, Siddique T, Duyn JH, Rouault TA, Floeter MK. Iron accumulation in deep cortical layers accounts for MRI signal abnormalities in ALS: correlating 7 tesla MRI and pathology. PLoS One 2012; 7:e35241. [PMID: 22529995 PMCID: PMC3328441 DOI: 10.1371/journal.pone.0035241] [Citation(s) in RCA: 204] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 03/11/2012] [Indexed: 11/21/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by cortical and spinal motor neuron dysfunction. Routine magnetic resonance imaging (MRI) studies have previously shown hypointense signal in the motor cortex on T2-weighted images in some ALS patients, however, the cause of this finding is unknown. To investigate the utility of this MR signal change as a marker of cortical motor neuron degeneration, signal abnormalities on 3T and 7T MR images of the brain were compared, and pathology was obtained in two ALS patients to determine the origin of the motor cortex hypointensity. Nineteen patients with clinically probable or definite ALS by El Escorial criteria and 19 healthy controls underwent 3T MRI. A 7T MRI scan was carried out on five ALS patients who had motor cortex hypointensity on the 3T FLAIR sequence and on three healthy controls. Postmortem 7T MRI of the brain was performed in one ALS patient and histological studies of the brains and spinal cords were obtained post-mortem in two patients. The motor cortex hypointensity on 3T FLAIR images was present in greater frequency in ALS patients. Increased hypointensity correlated with greater severity of upper motor neuron impairment. Analysis of 7T T2*-weighted gradient echo imaging localized the signal alteration to the deeper layers of the motor cortex in both ALS patients. Pathological studies showed increased iron accumulation in microglial cells in areas corresponding to the location of the signal changes on the 3T and 7T MRI of the motor cortex. These findings indicate that the motor cortex hypointensity on 3T MRI FLAIR images in ALS is due to increased iron accumulation by microglia.
Collapse
Affiliation(s)
- Justin Y Kwan
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Huang K, Li H, Shen H, Li M. Psychological stress expands low molecular weight iron pool in cerebral cortex, hippocampus, and striatum of rats. Biol Trace Elem Res 2012; 146:79-85. [PMID: 21993966 DOI: 10.1007/s12011-011-9230-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 10/04/2011] [Indexed: 01/30/2023]
Abstract
We have previously demonstrated that psychological stress (PS) can cause iron to accumulate in the cerebral cortex, hippocampus, and striatum of rats. However, why iron accumulates and in what oxidation state iron it accumulates in the brain of PS-exposed rats has not been well elucidated. In the present study, we investigated the influence of PS on the low molecular weight iron pool (LMWIP) in the rat brain. The results showed that: (1) PS significantly expanded LMWIP in the cerebral cortex, hippocampus, and striatum in rats; (2) PS caused derangement of pyramidal cells and reduced the layers of pyramidal CA1 and CA2 neurons; (3) PS exposure greatly lowered the expression of ferritin (Fn) and hephaestin (HP) in the rat cortex and hippocampus; and (4) PS decreased superoxide dismutase, glutathione peroxidase, and glutathione level and increased malondialdehyde level in the cerebral cortex, hippocampus, and striatum in rats. These results indicated that PS could expand LMWIP significantly, which may be attributed to PS-induced decrease in Fn, HP expression, and the subsequent reduction in iron storage and utilization, and expansion of LMWIP could in turn lead to aggravation of oxidative damage.
Collapse
Affiliation(s)
- Kai Huang
- Department of Military Hygiene, Second Military Medical University, No. 800 Xiangyin Road, Yangpu District, Shanghai, 200433, China
| | | | | | | |
Collapse
|
41
|
|
42
|
Borgonovo J, Capella P, Seltzer A, Sosa MA. Expression of coat proteins changes during postnatal development in selected areas of the rat brain. Int J Dev Neurosci 2012; 30:333-41. [PMID: 22306374 DOI: 10.1016/j.ijdevneu.2012.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 01/03/2012] [Accepted: 01/17/2012] [Indexed: 11/25/2022] Open
Abstract
It is well known that clathrin-mediated endocytosis is crucial for the normal functioning and integrity of neurons in the central nervous system. In this study we attempted to correlate the expression of coat proteins with development in different areas of rat brain. By Western blot, we studied the expression of AP-2, GGA1 and GGA2 in striatum, cerebellum, brain stem, cerebral cortex and hippocampus of newborn rats and during post-natal development; 5, 15, 30, 60, 90 or 150 days after birth. We observed that the expression of the α2 subunit of AP-2 increased substantially between the 15th and 30th day after birth in all areas studied, excepting the cerebellum and cortex. On the other hand, the expression of the α1 subunit does not change significantly during the development in any of the areas under study. We also noted that the expression of the μ2 subunit did not follow the pattern of α2 during development. In general terms, the expression of GGA1 and GGA2 followed a similar pattern to that of AP-2, although these proteins increased significantly in the cerebral cortex from the 15th day after birth. Moreover, presenilin-1, a protein associated with aging and neurodegeneration, shows an expression pattern similar to coat proteins in the striatum and cortex. These results suggest that proteins that conform the intracellular transport machinery in the brain cells seems to accompany development, according to the maturation of the different brain areas.
Collapse
Affiliation(s)
- Janina Borgonovo
- Laboratorio de Biología y Fisiología Celular "Dr. Francisco Bertini", Instituto de Histología y Embriología, FCM, Universidad Nacional de Cuyo, Mendoza, Argentina
| | | | | | | |
Collapse
|
43
|
Weinreb O, Mandel S, Bar-Am O, Amit T. Iron-chelating backbone coupled with monoamine oxidase inhibitory moiety as novel pluripotential therapeutic agents for Alzheimer's disease: a tribute to Moussa Youdim. J Neural Transm (Vienna) 2011; 118:479-92. [PMID: 21360301 DOI: 10.1007/s00702-011-0597-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 01/27/2011] [Indexed: 12/13/2022]
Abstract
It is for these authors a great privilege to dedicate this review article to Moussa Youdim, who is one of the most imperative pharmacologists and pioneer investigators in the search and development of novel therapeutics for neurodegenerative diseases. 40 years ago, Moussa Youdim has started studying brain iron, catecholamine receptor and monoamine oxidase (MAO)-A and -B functions. Although Moussa Youdim succeeded in exploring the novel anti-Parkinsonian, selective MAO-B inhibitor drug, rasagiline (Azilect, Teva Pharmaceutical Co.), he did not stop searching for superior therapeutic approaches for neurodegenerative disorders. To date, Moussa Youdim and his research group are designing and synthesizing pluripotential drug candidates possessing diverse pharmacological properties that can act on multiple targets and pathological features ascribed to Parkinson's disease, Alzheimer's disease (AD) and amyotrophic lateral sclerosis. One such example is the multimodal non-toxic, brain-permeable iron-chelating compound, M30 (5-[N-methyl-N-propargylaminomethyl]-8-hydroxyquinoline), which amalgamates the propargyl moiety of rasagiline with the backbone of the potent iron chelator, VK28. This review discusses the multiple effects of several leading compounds of this series, concerning their neuroprotective/neurorestorative molecular mechanisms in vivo and in vitro, with a special focus on the pathological features ascribed to AD, including antioxidant and iron chelating activities, regulation of amyloid precursor protein and amyloid β peptide expression processing, activation of pro-survival signaling pathways and regulation of cell cycle and neurite outgrowth.
Collapse
Affiliation(s)
- Orly Weinreb
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research and Department of Pharmacology, Rappaport Family Research Institute, Technion-Faculty of Medicine, P.O.B. 9697, 31096, Haifa, Israel.
| | | | | | | |
Collapse
|
44
|
Nishida Y. The chemical process of oxidative stress by copper(II) and iron(III) ions in several neurodegenerative disorders. MONATSHEFTE FUR CHEMIE 2011. [DOI: 10.1007/s00706-010-0444-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
45
|
Hider RC, Roy S, Ma YM, Le Kong X, Preston J. The potential application of iron chelators for the treatment of neurodegenerative diseases. Metallomics 2011; 3:239-49. [DOI: 10.1039/c0mt00087f] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
46
|
Garcia TB, Oliveira KRM, do Nascimento JLM, Crespo-López ME, Picanço-Diniz DLW, Mota TC, Herculano AM. Glutamate induces glutathione efflux mediated by glutamate/aspartate transporter in retinal cell cultures. Neurochem Res 2010; 36:412-8. [PMID: 21161593 DOI: 10.1007/s11064-010-0356-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2010] [Indexed: 11/29/2022]
Abstract
This study was undertaken in order to characterize the role of the glutamate/aspartate transporter (GLAST) in the glutathione (GSH) efflux induced by glutamate. Our results demonstrated that retinal cell cultures exhibit two mechanisms of GSH release, one Na(+)-independent and other Na(+)-dependent. Glutamate and aspartate induced GSH efflux only in presence of Na(+). Treatment with PCD (L-trans-Pyrrolidine-2,4-dicarboxylate), a transportable glutamate uptake blocker, increased GSH release indicating that GSH can be carried by glutamate transporters in retinal cell cultures. Added to this, treatment with zinc ion cultures, a recognized inhibitor of GLAST blocked GSH efflux evoked by glutamate. Treatment with NMDA antagonist (MK-801) did not have any effect on the GSH release induced by glutamate. These results suggest that glutamate induces GLAST-mediated release of GSH from retinal cell cultures and this could represent an important mechanism of cellular protection against glutamate toxicity in the CNS.
Collapse
Affiliation(s)
- T B Garcia
- Laboratório de Neuroendocrinologia, Instituto de Ciências Biológicas, Universidade Federal do Pará, Campus do Guamá, Av. Augusto Corrêa, 01, Belém, Pará, 66075-900, Brazil
| | | | | | | | | | | | | |
Collapse
|
47
|
Zhou ZD, Lan YH, Tan EK, Lim TM. Iron species-mediated dopamine oxidation, proteasome inhibition, and dopaminergic cell demise: implications for iron-related dopaminergic neuron degeneration. Free Radic Biol Med 2010; 49:1856-71. [PMID: 20854902 DOI: 10.1016/j.freeradbiomed.2010.09.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 08/19/2010] [Accepted: 09/13/2010] [Indexed: 12/31/2022]
Abstract
Iron species have been suggested to be highly involved in the pathogenesis of Parkinson disease. However, the detailed mechanism of iron-induced dopaminergic degeneration is still unclear. In this study, we demonstrate that free iron ions (trivalent or bivalent) and iron ions in stable complex with cyanide ions (K(4)Fe(CN)(6) and K(3)Fe(CN)(6)) can induce dopamine (DA) oxidation with different profiles and subsequently lead to proteasome inhibition and even dopaminergic MN9D cell demise via different mechanisms. The free iron ions could mediate extensive DA oxidation in an iron-DA complex-dependent manner. However, iron ions in stable complex with cyanide ions could not induce, or could induce only brief, DA oxidation. Deferoxamine, a specific iron ion chelator, could disrupt iron-DA complex formation and thus abrogate free iron ion-catalyzed DA oxidation and subsequent cell toxicity. Glutathione could neither disrupt iron-DA complex formation nor influence free iron ion-catalyzed DA oxidation but could protect against iron-mediated toxicity via detoxification of toxic by-products of iron-mediated DA oxidation. The resulting DA oxidation could inhibit chymotrypsin-like, trypsin-like, and caspase-like proteasome activities. However, we demonstrated that oxidative damage was not the major toxic mechanism of MN9D cell degeneration, but it was the DA quinones derived from iron-induced DA oxidation that contributed significantly to proteasome inhibition and even dopaminergic cell demise.
Collapse
Affiliation(s)
- Zhi Dong Zhou
- Department of Biological Science, National University of Singapore, Singapore
| | | | | | | |
Collapse
|
48
|
Kell DB. Towards a unifying, systems biology understanding of large-scale cellular death and destruction caused by poorly liganded iron: Parkinson's, Huntington's, Alzheimer's, prions, bactericides, chemical toxicology and others as examples. Arch Toxicol 2010; 84:825-89. [PMID: 20967426 PMCID: PMC2988997 DOI: 10.1007/s00204-010-0577-x] [Citation(s) in RCA: 265] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 07/14/2010] [Indexed: 12/11/2022]
Abstract
Exposure to a variety of toxins and/or infectious agents leads to disease, degeneration and death, often characterised by circumstances in which cells or tissues do not merely die and cease to function but may be more or less entirely obliterated. It is then legitimate to ask the question as to whether, despite the many kinds of agent involved, there may be at least some unifying mechanisms of such cell death and destruction. I summarise the evidence that in a great many cases, one underlying mechanism, providing major stresses of this type, entails continuing and autocatalytic production (based on positive feedback mechanisms) of hydroxyl radicals via Fenton chemistry involving poorly liganded iron, leading to cell death via apoptosis (probably including via pathways induced by changes in the NF-κB system). While every pathway is in some sense connected to every other one, I highlight the literature evidence suggesting that the degenerative effects of many diseases and toxicological insults converge on iron dysregulation. This highlights specifically the role of iron metabolism, and the detailed speciation of iron, in chemical and other toxicology, and has significant implications for the use of iron chelating substances (probably in partnership with appropriate anti-oxidants) as nutritional or therapeutic agents in inhibiting both the progression of these mainly degenerative diseases and the sequelae of both chronic and acute toxin exposure. The complexity of biochemical networks, especially those involving autocatalytic behaviour and positive feedbacks, means that multiple interventions (e.g. of iron chelators plus antioxidants) are likely to prove most effective. A variety of systems biology approaches, that I summarise, can predict both the mechanisms involved in these cell death pathways and the optimal sites of action for nutritional or pharmacological interventions.
Collapse
Affiliation(s)
- Douglas B Kell
- School of Chemistry and the Manchester Interdisciplinary Biocentre, The University of Manchester, Manchester M1 7DN, UK.
| |
Collapse
|
49
|
Fikre A, Van Moorhem M, Ahmed S, Lambein F, Gheysen G. Studies on neurolathyrism in Ethiopia: dietary habits, perception of risks and prevention. Food Chem Toxicol 2010; 49:678-84. [PMID: 20950665 DOI: 10.1016/j.fct.2010.09.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 09/14/2010] [Accepted: 09/22/2010] [Indexed: 10/19/2022]
Abstract
This study describes the correlation of traditional perceptions and dietary habits with the incidence of neurolathyrism to propose preventive measures. Therefore, 118 households of South Wollo and North Gondar (Amhara Regional State, Ethiopia), of which one third had at least one neurolathyrism affected member, were interviewed. Most of the affected families in this study had one neurolathyrism victim, being predominantly male and of younger age. The incidence among youngsters (boys and girls) was significantly correlated with the consumption of green unripe seeds (eshet), confirming this as a risk factor for developing neurolathyrism. The consumption of other popular grass pea preparations was not age related. Neurolathyrism patients did not attempt any medication as most people knew that neurolathyrism is incurable, but the consumption of grass pea was abandoned after developing the disease. The minority 'Woito' tribe was virtually unaffected. They were the only people reporting to consume fish which is rich in amino acids such as methionine and using metallic kitchen utensils in addition to clay pots. This observation points to the correlation between low neurolathyrism incidence and a better balanced diet as well as metallic kitchen utensils, suggesting a new approach for neurolathyrism prevention.
Collapse
Affiliation(s)
- A Fikre
- Ethiopian Institute for Agricultural Research, Debre Zeit Center, PO Box 32, Debre Zeit, Ethiopia
| | | | | | | | | |
Collapse
|
50
|
Weinreb O, Amit T, Mandel S, Kupershmidt L, Youdim MBH. Neuroprotective multifunctional iron chelators: from redox-sensitive process to novel therapeutic opportunities. Antioxid Redox Signal 2010; 13:919-49. [PMID: 20095867 DOI: 10.1089/ars.2009.2929] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Accumulating evidence suggests that many cytotoxic signals occurring in the neurodegenerative brain can initiate neuronal death processes, including oxidative stress, inflammation, and accumulation of iron at the sites of the neuronal deterioration. Neuroprotection by iron chelators has been widely recognized with respect to their ability to prevent hydroxyl radical formation in the Fenton reaction by sequestering redox-active iron. An additional neuroprotective mechanism of iron chelators is associated with their ability to upregulate or stabilize the transcriptional activator, hypoxia-inducible factor-1alpha (HIF-1alpha). HIF-1alpha stability within the cells is under the control of a class of iron-dependent and oxygen-sensor enzymes, HIF prolyl-4-hydroxylases (PHDs) that target HIF-1alpha for degradation. Thus, an emerging novel target for neuroprotection is associated with the HIF system to promote stabilization of HIF-1alpha and increase transcription of HIF-1-related survival genes, which have been reported to be regulated in patient's brains afflicted with diverse neurodegenerative diseases. In accordance, a new potential therapeutic strategy for neurodegenerative diseases is explored, by which iron chelators would inhibit PHDs, target the HIF-1-signaling pathway and ultimately activate HIF-1-dependent neuroprotective genes. This review discusses two interrelated approaches concerning therapy targets in neurodegeneration, sharing in common the implementation of iron chelation activity: antioxidation and HIF-1-pathway activation.
Collapse
Affiliation(s)
- Orly Weinreb
- Eve Topf Centers of Excellence for Neurodegenerative Diseases Research and Department of Pharmacology, Rappaport Family Research Institute, Technion-Faculty of Medicine, Haifa, Israel.
| | | | | | | | | |
Collapse
|