1
|
Gao G, You L, Zhang J, Chang YZ, Yu P. Brain Iron Metabolism, Redox Balance and Neurological Diseases. Antioxidants (Basel) 2023; 12:1289. [PMID: 37372019 DOI: 10.3390/antiox12061289] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The incidence of neurological diseases, such as Parkinson's disease, Alzheimer's disease and stroke, is increasing. An increasing number of studies have correlated these diseases with brain iron overload and the resulting oxidative damage. Brain iron deficiency has also been closely linked to neurodevelopment. These neurological disorders seriously affect the physical and mental health of patients and bring heavy economic burdens to families and society. Therefore, it is important to maintain brain iron homeostasis and to understand the mechanism of brain iron disorders affecting reactive oxygen species (ROS) balance, resulting in neural damage, cell death and, ultimately, leading to the development of disease. Evidence has shown that many therapies targeting brain iron and ROS imbalances have good preventive and therapeutic effects on neurological diseases. This review highlights the molecular mechanisms, pathogenesis and treatment strategies of brain iron metabolism disorders in neurological diseases.
Collapse
Affiliation(s)
- Guofen Gao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| | - Linhao You
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| | - Jianhua Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| | - Yan-Zhong Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| | - Peng Yu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| |
Collapse
|
2
|
Mamais A, Kluss JH, Bonet-Ponce L, Landeck N, Langston RG, Smith N, Beilina A, Kaganovich A, Ghosh MC, Pellegrini L, Kumaran R, Papazoglou I, Heaton GR, Bandopadhyay R, Maio N, Kim C, LaVoie MJ, Gershlick DC, Cookson MR. Mutations in LRRK2 linked to Parkinson disease sequester Rab8a to damaged lysosomes and regulate transferrin-mediated iron uptake in microglia. PLoS Biol 2021; 19:e3001480. [PMID: 34914695 PMCID: PMC8675653 DOI: 10.1371/journal.pbio.3001480] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/10/2021] [Indexed: 01/09/2023] Open
Abstract
Mutations in leucine-rich repeat kinase 2 (LRRK2) cause autosomal dominant Parkinson disease (PD), while polymorphic LRRK2 variants are associated with sporadic PD. PD-linked mutations increase LRRK2 kinase activity and induce neurotoxicity in vitro and in vivo. The small GTPase Rab8a is a LRRK2 kinase substrate and is involved in receptor-mediated recycling and endocytic trafficking of transferrin, but the effect of PD-linked LRRK2 mutations on the function of Rab8a is poorly understood. Here, we show that gain-of-function mutations in LRRK2 induce sequestration of endogenous Rab8a to lysosomes in overexpression cell models, while pharmacological inhibition of LRRK2 kinase activity reverses this phenotype. Furthermore, we show that LRRK2 mutations drive association of endocytosed transferrin with Rab8a-positive lysosomes. LRRK2 has been nominated as an integral part of cellular responses downstream of proinflammatory signals and is activated in microglia in postmortem PD tissue. Here, we show that iPSC-derived microglia from patients carrying the most common LRRK2 mutation, G2019S, mistraffic transferrin to lysosomes proximal to the nucleus in proinflammatory conditions. Furthermore, G2019S knock-in mice show a significant increase in iron deposition in microglia following intrastriatal LPS injection compared to wild-type mice, accompanied by striatal accumulation of ferritin. Our data support a role of LRRK2 in modulating iron uptake and storage in response to proinflammatory stimuli in microglia. Brain iron deposition is a feature of Parkinson’s disease pathology, but how this contributes to neurodegeneration is unclear. This study show that Parkinson’s disease-linked mutations in LRRK2 cause aberrant brain iron accumulation in vivo and iron dyshomeostasis in vitro, supporting a role of LRRK2 in modulating iron uptake and storage in response to proinflammatory stimuli in microglia.
Collapse
Affiliation(s)
- Adamantios Mamais
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Maryland, United States of America
- Department of Neurology, University of Florida, Gainesville, Florida, United States of America
| | - Jillian H. Kluss
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Maryland, United States of America
| | - Luis Bonet-Ponce
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Maryland, United States of America
| | - Natalie Landeck
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Maryland, United States of America
| | - Rebekah G. Langston
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Maryland, United States of America
| | - Nathan Smith
- Department of Biochemistry and the Redox Biology Center, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Alexandra Beilina
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Maryland, United States of America
| | - Alice Kaganovich
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Maryland, United States of America
| | - Manik C. Ghosh
- Molecular Medicine Branch, ‘Eunice Kennedy Shriver’ National Institute of Child Health and Human Development, Bethesda, Maryland, United States of America
| | | | - Ravindran Kumaran
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Maryland, United States of America
| | - Ioannis Papazoglou
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - George R. Heaton
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Maryland, United States of America
| | - Rina Bandopadhyay
- UCL Institute of Neurology and Reta Lila Weston Institute of Neurological Studies, University College London, London, United Kingdom
| | - Nunziata Maio
- Molecular Medicine Branch, ‘Eunice Kennedy Shriver’ National Institute of Child Health and Human Development, Bethesda, Maryland, United States of America
| | - Changyoun Kim
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Matthew J. LaVoie
- Department of Neurology, University of Florida, Gainesville, Florida, United States of America
| | - David C. Gershlick
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Maryland, United States of America
- * E-mail:
| |
Collapse
|
3
|
D’Mello SR, Kindy MC. Overdosing on iron: Elevated iron and degenerative brain disorders. Exp Biol Med (Maywood) 2020; 245:1444-1473. [PMID: 32878460 PMCID: PMC7553095 DOI: 10.1177/1535370220953065] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IMPACT STATEMENT Brain degenerative disorders, which include some neurodevelopmental disorders and age-associated diseases, cause debilitating neurological deficits and are generally fatal. A large body of emerging evidence indicates that iron accumulation in neurons within specific regions of the brain plays an important role in the pathogenesis of many of these disorders. Iron homeostasis is a highly complex and incompletely understood process involving a large number of regulatory molecules. Our review provides a description of what is known about how iron is obtained by the body and brain and how defects in the homeostatic processes could contribute to the development of brain diseases, focusing on Alzheimer's disease and Parkinson's disease as well as four other disorders belonging to a class of inherited conditions referred to as neurodegeneration based on iron accumulation (NBIA) disorders. A description of potential therapeutic approaches being tested for each of these different disorders is provided.
Collapse
Affiliation(s)
| | - Mark C Kindy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
- James A. Haley Veterans Affairs Medical Center, Tampa, FL 33612, USA
| |
Collapse
|
4
|
Cai R, Zhang Y, Simmering JE, Schultz JL, Li Y, Fernandez-Carasa I, Consiglio A, Raya A, Polgreen PM, Narayanan NS, Yuan Y, Chen Z, Su W, Han Y, Zhao C, Gao L, Ji X, Welsh MJ, Liu L. Enhancing glycolysis attenuates Parkinson's disease progression in models and clinical databases. J Clin Invest 2020; 129:4539-4549. [PMID: 31524631 DOI: 10.1172/jci129987] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/23/2019] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease that lacks therapies to prevent progressive neurodegeneration. Impaired energy metabolism and reduced ATP levels are common features of PD. Previous studies revealed that terazosin (TZ) enhances the activity of phosphoglycerate kinase 1 (PGK1), thereby stimulating glycolysis and increasing cellular ATP levels. Therefore, we asked whether enhancement of PGK1 activity would change the course of PD. In toxin-induced and genetic PD models in mice, rats, flies, and induced pluripotent stem cells, TZ increased brain ATP levels and slowed or prevented neuron loss. The drug increased dopamine levels and partially restored motor function. Because TZ is prescribed clinically, we also interrogated 2 distinct human databases. We found slower disease progression, decreased PD-related complications, and a reduced frequency of PD diagnoses in individuals taking TZ and related drugs. These findings suggest that enhancing PGK1 activity and increasing glycolysis may slow neurodegeneration in PD.
Collapse
Affiliation(s)
- Rong Cai
- Institute of Hypoxia Medicine, Xuanwu Hospital and Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, and.,Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yu Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
| | | | - Jordan L Schultz
- Departments of Pharmaceutical Care and Neurology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Yuhong Li
- Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Irene Fernandez-Carasa
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, Hospitalet de Llobregat and Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Antonella Consiglio
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, Hospitalet de Llobregat and Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Angel Raya
- Center of Regenerative Medicine in Barcelona (CMRB) and Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Hospital Duran i Reynals, Hospitalet de Llobregat, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | | | - Nandakumar S Narayanan
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Yanpeng Yuan
- Institute of Hypoxia Medicine, Xuanwu Hospital and Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, and
| | - Zhiguo Chen
- Institute of Hypoxia Medicine, Xuanwu Hospital and Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, and
| | - Wenting Su
- Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yanping Han
- Institute of Hypoxia Medicine, Xuanwu Hospital and Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, and
| | - Chunyue Zhao
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China
| | - Lifang Gao
- Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Xunming Ji
- Institute of Hypoxia Medicine, Xuanwu Hospital and Key Laboratory for Neurodegenerative Diseases of the Ministry of Education, and.,Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Michael J Welsh
- Howard Hughes Medical Institute, Departments of Internal Medicine, Neurology, and Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Lei Liu
- Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China
| |
Collapse
|
5
|
Abstract
The key molecular events that provoke Parkinson's disease (PD) are not fully understood. Iron deposit was found in the substantia nigra pars compacta (SNpc) of PD patients and animal models, where dopaminergic neurons degeneration occurred selectively. The mechanisms involved in disturbed iron metabolism remain unknown, however, considerable evidence indicates that iron transporters dysregulation, activation of L-type voltage-gated calcium channel (LTCC) and ATP-sensitive potassium (KATP) channels, as well as N-methyl-D-aspartate (NMDA) receptors (NMDARs) contribute to this process. There is emerging evidence on the structural links and functional modulations between iron and α-synuclein, and the key player in PD which aggregates in Lewy bodies. Iron is believed to modulate α-synuclein synthesis, post-translational modification, and aggregation. Furthermore, glia, especially activated astroglia and microglia, are involved in iron deposit in PD. Glial contributions were largely dependent on the factors they released, e.g., neurotrophic factors, pro-inflammatory factors, lactoferrin, and those undetermined. Therefore, iron chelation using iron chelators, the extracts from many natural foods with iron chelating properties, may be an effective therapy for prevention and treatment of the disease.
Collapse
|
6
|
Mouhape C, Costa G, Ferreira M, Abin-Carriquiry JA, Dajas F, Prunell G. Nicotine-Induced Neuroprotection in Rotenone In Vivo and In Vitro Models of Parkinson’s Disease: Evidences for the Involvement of the Labile Iron Pool Level as the Underlying Mechanism. Neurotox Res 2018; 35:71-82. [DOI: 10.1007/s12640-018-9931-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/18/2018] [Accepted: 07/05/2018] [Indexed: 11/29/2022]
|
7
|
An altered blood-brain barrier contributes to brain iron accumulation and neuroinflammation in the 6-OHDA rat model of Parkinson's disease. Neuroscience 2017; 362:141-151. [PMID: 28842186 DOI: 10.1016/j.neuroscience.2017.08.023] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/09/2017] [Accepted: 08/11/2017] [Indexed: 11/20/2022]
Abstract
Brain iron accumulation is a common feature shared by several neurodegenerative disorders including Parkinson's disease. However, what produces this accumulation of iron is still unknown. In this study, the 6-hydroxydopamine (6-OHDA) hemi-parkinsonian rat model was used to investigate abnormal iron accumulation in substantia nigra. We investigated three possible causes of iron accumulation; a compromised blood-brain barrier (BBB), abnormal expression of ferritin, and neuroinflammation. We identified alterations in the BBB subsequent to the injection of 6-OHDA using gadolinium-enhanced magnetic resonance imaging (MRI). Moreover, detection of extravasated IgG suggested that peripheral components are able to enter the brain through a leaky BBB. Presence of iron following dopamine cell degeneration was studied by MRI, which revealed hypointense signals in the substantia nigra. The presence of iron deposits was further validated in histological evaluations. Furthermore, iron inclusions were closely associated with active microglia and with increased levels of L-ferritin indicating a putative role for microglia and L-ferritin in brain iron accumulation and dopamine neurodegeneration.
Collapse
|
8
|
Gao G, You LH, Chang YZ. Iron Metabolism in Parkinson’s Disease. OXIDATIVE STRESS AND REDOX SIGNALLING IN PARKINSON’S DISEASE 2017. [DOI: 10.1039/9781782622888-00255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In the central nervous system, iron is involved in many biologically important processes such as oxygen transport and storage, electron transport, energy metabolism, and antioxidant and DNA synthesis. Parkinson’s disease (PD) is a common neurodegenerative disease characterized by loss of dopaminergic neurons in the substantia nigra. Extensive research has reported that iron is heavily accumulated in the dopaminergic neurons in substantia nigra (SN) of PD patients. Changes in the expression of key iron transporters have also been observed in PD patients. Excessive iron accumulation can induce neuronal damage through reactive oxygen species production, which can cause oxidative stress increased membrane lipid peroxidation, DNA damage and protein oxidation and misfolding. This chapter provides a review about brain iron metabolism in PD, the role of iron transporters expression and function on brain iron homeostasis and distribution of intracellular iron. This knowledge will be of benefit to novel therapeutic targets for PD.
Collapse
Affiliation(s)
- Guofen Gao
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University Shijiazhuang Hebei Province 050024 China
| | - Lin-Hao You
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University Shijiazhuang Hebei Province 050024 China
| | - Yan-Zhong Chang
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University Shijiazhuang Hebei Province 050024 China
| |
Collapse
|
9
|
Virel A, Rehnmark A, Orädd G, Olmedo-Díaz S, Faergemann E, Strömberg I. Magnetic resonance imaging as a tool to image neuroinflammation in a rat model of Parkinson's disease--phagocyte influx to the brain is promoted by bilberry-enriched diet. Eur J Neurosci 2015; 42:2761-71. [PMID: 26273789 DOI: 10.1111/ejn.13044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 08/05/2015] [Accepted: 08/11/2015] [Indexed: 12/22/2022]
Abstract
Neuroinflammation is a chronic event in neurodegenerative disorders. In the rat model of Parkinson's disease, including a striatal injection of the neurotoxin 6-hydroxydopamine (6-OHDA), antioxidant treatment affects the inflammatory process. Despite a heavy accumulation of microglia early after the injury, dopamine nerve fibre regeneration occurs. It remains unclear why this heavy accumulation of microglia is found early after the lesion in antioxidant-treated animals, or even more, what is the origin of these microglia. In this study magnetic resonance imaging (MRI) was used to elucidate whether the inflammatory response was generated from the blood or from activated brain microglia. Superparamagnetic iron oxide (SPIO) nanoparticles were injected intravenously prior to a striatal 6-OHDA injection to tag phagocytes in the blood. Rats were fed either with bilberry-enriched or control diet. T2*-weighted MRI scans were performed 1 week after the lesion, and hypointense areas were calculated from T2*-weighted images, to monitor the presence of SPIO particles. The results revealed that feeding the animals with bilberries significantly promoted accumulation of blood-derived immune cells. Gadolinium-enhanced MRI demonstrated no difference in leakage of the blood-brain barrier independent of diets. To conclude, bilberry-enriched diet promotes an influx of periphery-derived immune cells to the brain early after injury.
Collapse
Affiliation(s)
- Ana Virel
- Integrative Medical Biology, Umeå University, SE 901 87, Umeå, Sweden
| | - Anna Rehnmark
- Integrative Medical Biology, Umeå University, SE 901 87, Umeå, Sweden
| | - Greger Orädd
- Department of Radiation Sciences, Umeå University, SE 901 87 Umeå, Sweden
| | - Sonia Olmedo-Díaz
- Integrative Medical Biology, Umeå University, SE 901 87, Umeå, Sweden
| | - Erik Faergemann
- Integrative Medical Biology, Umeå University, SE 901 87, Umeå, Sweden
| | - Ingrid Strömberg
- Integrative Medical Biology, Umeå University, SE 901 87, Umeå, Sweden
| |
Collapse
|
10
|
Marei HES, Lashen S, Farag A, Althani A, Afifi N, A AE, Rezk S, Pallini R, Casalbore P, Cenciarelli C. Human olfactory bulb neural stem cells mitigate movement disorders in a rat model of Parkinson's disease. J Cell Physiol 2015; 230:1614-29. [PMID: 25536543 DOI: 10.1002/jcp.24909] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 12/18/2014] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is a neurological disorder characterized by the loss of midbrain dopaminergic (DA) neurons. Neural stem cells (NSCs) are multipotent stem cells that are capable of differentiating into different neuronal and glial elements. The production of DA neurons from NSCs could potentially alleviate behavioral deficits in Parkinsonian patients; timely intervention with NSCs might provide a therapeutic strategy for PD. We have isolated and generated highly enriched cultures of neural stem/progenitor cells from the human olfactory bulb (OB). If NSCs can be obtained from OB, it would alleviate ethical concerns associated with the use of embryonic tissue, and provide an easily accessible cell source that would preclude the need for invasive brain surgery. Following isolation and culture, olfactory bulb neural stem cells (OBNSCs) were genetically engineered to express hNGF and GFP. The hNFG-GFP-OBNSCs were transplanted into the striatum of 6-hydroxydopamin (6-OHDA) Parkinsonian rats. The grafted cells survived in the lesion environment for more than eight weeks after implantation with no tumor formation. The grafted cells differentiated in vivo into oligodendrocyte-like (25 ± 2.88%), neuron-like (52.63 ± 4.16%), and astrocyte -like (22.36 ± 1.56%) lineages, which we differentiated based on morphological and immunohistochemical criteria. Transplanted rats exhibited a significant partial correction in stepping and placing in non-pharmacological behavioral tests, pole and rotarod tests. Taken together, our data encourage further investigations of the possible use of OBNSCs as a promising cell-based therapeutic strategy for Parkinson's disease.
Collapse
Affiliation(s)
- Hany E S Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Virel A, Faergemann E, Orädd G, Strömberg I. Magnetic resonance imaging (MRI) to study striatal iron accumulation in a rat model of Parkinson's disease. PLoS One 2014; 9:e112941. [PMID: 25398088 PMCID: PMC4232582 DOI: 10.1371/journal.pone.0112941] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/22/2014] [Indexed: 12/12/2022] Open
Abstract
Abnormal accumulation of iron is observed in neurodegenerative disorders. In Parkinson's disease, an excess of iron has been demonstrated in different structures of the basal ganglia and is suggested to be involved in the pathogenesis of the disease. Using the 6-hydroxydopamine (6-OHDA) rat model of Parkinson's disease, the edematous effect of 6-OHDA and its relation with striatal iron accumulation was examined utilizing in vivo magnetic resonance imaging (MRI). The results revealed that in comparison with control animals, injection of 6-OHDA into the rat striatum provoked an edematous process, visible in T2-weighted images that was accompanied by an accumulation of iron clearly detectable in T2*-weighted images. Furthermore, Prussian blue staining to detect iron in sectioned brains confirmed the existence of accumulated iron in the areas of T2* hypointensities. The presence of ED1-positive microglia in the lesioned striatum overlapped with this accumulation of iron, indicating areas of toxicity and loss of dopamine nerve fibers. Correlation analyses demonstrated a direct relation between the hyperintensities caused by the edema and the hypointensities caused by the accumulation of iron.
Collapse
Affiliation(s)
- Ana Virel
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Erik Faergemann
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Greger Orädd
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Ingrid Strömberg
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
12
|
Nigral iron elevation is an invariable feature of Parkinson's disease and is a sufficient cause of neurodegeneration. BIOMED RESEARCH INTERNATIONAL 2014; 2014:581256. [PMID: 24527451 PMCID: PMC3914334 DOI: 10.1155/2014/581256] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 10/28/2013] [Indexed: 12/22/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor deficits accompanying degeneration of substantia nigra pars compactor (SNc) neurons. Although familial forms of the disease exist, the cause of sporadic PD is unknown. Symptomatic treatments are available for PD, but there are no disease modifying therapies. While the neurodegenerative processes in PD may be multifactorial, this paper will review the evidence that prooxidant iron elevation in the SNc is an invariable feature of sporadic and familial PD forms, participates in the disease mechanism, and presents as a tractable target for a disease modifying therapy.
Collapse
|
13
|
Striatal TH-immunopositive fibers recover after an intrastriatal injection of 6-hydroxydopamine in golden hamsters treated with prednisolone: roles of tumor necrosis factor-α and inducible nitric oxide synthase in neurodegeneration. Neurosci Res 2013; 76:83-92. [PMID: 23471013 DOI: 10.1016/j.neures.2013.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 02/20/2013] [Indexed: 11/23/2022]
Abstract
Neuroinflammation has been implicated in the pathology of neurodegenerative processes such as Parkinson's disease (PD). Using the golden hamster (GH) 6-hydroxydopamine (6-OHDA) model, we investigated whether the attenuation of neuroinflammation influences the onset and progression of dopamine cell degeneration. 6-OHDA-injected GH received a treatment of minocycline (MINO), prednisolone (Pred) or a combination of minocycline and prednisolone (MINO+Pred). Immunohistochemistry for tyrosine hydroxylase (TH), Iba-1 and glial fibrillary acidic protein (GFAP) was used to evaluate lesions in the nigrostriatal axis and the amount of activated microglia and astroglia, respectively. RT-PCR was used to measure mRNA levels of cytokines and trophic neuroprotective factors. The three anti-inflammatory treatments dramatically reduced activated microglia in the nigrostriatal axis. In addition, TH-immunostaining showed that the positive areas in the ipsilateral striatum of either MINO or Pred groups were higher than that of control. However, only in Pred group this recovery was significant. mRNA measurements demonstrated lower levels of TNF-α, iNOS, BDNF and GDNF in Pred group when compared with controls. The results suggest that TH-immunopositive fibers have the ability to recover after 6-OHDA-induced toxicity of dopaminergic neurons, and this recovery may be due to a decrease in the microglial production of TNF-α and iNOS.
Collapse
|
14
|
Li WJ, Jiang H, Song N, Xie JX. Dose- and time-dependent alpha-synuclein aggregation induced by ferric iron in SK-N-SH cells. Neurosci Bull 2010; 26:205-10. [PMID: 20502498 DOI: 10.1007/s12264-010-1117-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Intracellular formation of Lewy body (LB) is one of the hallmarks of Parkinson's disease. The main component of LB is aggregated alpha-synuclein, present in the substantia nigra where iron accumulation also occurs. The present study aimed to study the relationship between iron and alpha-synuclein aggregation. METHODS SK-N-SH cells were treated with different concentrations of ferric iron for 24 h or 48 h. MTT assay was conducted to determine the cell viability. Thioflavine S staining was used to detect alpha-synuclein aggregation. RESULTS With the increase of iron concentration, the cell viability decreased significantly. At the concentrations of 5 mmol/L and 10 mmol/L, iron induced alpha-synuclein aggregation more severely than at the concentration of 1 mmol/L. Besides, 48-h treatment-induced aggregation was more severe than that induced by 24-h treatment, at the corresponding iron concentrations. CONCLUSION Ferric iron can induce alpha-synuclein aggregation, which is toxic to the cells, in a dose- and time-dependent way.
Collapse
Affiliation(s)
- Wen-Jing Li
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China
| | | | | | | |
Collapse
|
15
|
Lv Z, Jiang H, Xu H, Song N, Xie J. Increased iron levels correlate with the selective nigral dopaminergic neuron degeneration in Parkinson's disease. J Neural Transm (Vienna) 2010; 118:361-9. [PMID: 20556443 DOI: 10.1007/s00702-010-0434-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Accepted: 05/31/2010] [Indexed: 12/01/2022]
Abstract
The staging of Lewy-related pathology in sporadic Parkinson's disease (PD) reveals that many brain nuclei are affected in PD during different stages, except the ventral tegmental area (VTA), which is close related to the substantia nigra (SN) and enriched in dopamine (DA) neurons. Why DA neurons are selectively degenerated in the SN of PD is far from known. In the present study, we observed that the number of tyrosine hydroxylase immunoreactive neurons decreased and iron-staining positive cells increased in the SN, but not in the VTA, in the chronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated PD mice. Increased expression of divalent metal transporter 1 and decreased expression of ferroportin 1 might associate with this increased nigral iron levels. Lipofuscin granular aggregations and upregulation of alpha-synuclein (α-synuclein) were also observed only in the SN. These results suggest that increased iron levels associate with the selective degeneration of DA neurons in the SN. The intracellular regulation mechanisms for the iron transporters may be different in the SN and VTA under the same conditions. Moreover, the lipofuscin granular aggregations and upregulation of α-synuclein were also involved in the selective degeneration of dopaminergic neurons in the SN.
Collapse
Affiliation(s)
- Zhanyun Lv
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Medical College of Qingdao University, Qingdao, 266071, China
| | | | | | | | | |
Collapse
|
16
|
Kobayashi H, Oikawa S, Umemura S, Hirosawa I, Kawanishi S. Mechanism of metal-mediated DNA damage and apoptosis induced by 6-hydroxydopamine in neuroblastoma SH-SY5Y cells. Free Radic Res 2009; 42:651-60. [DOI: 10.1080/10715760802270334] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
17
|
Datla KP, Zbarsky V, Rai D, Parkar S, Osakabe N, Aruoma OI, Dexter DT. Short-term supplementation with plant extracts rich in flavonoids protect nigrostriatal dopaminergic neurons in a rat model of Parkinson's disease. J Am Coll Nutr 2007; 26:341-9. [PMID: 17906186 DOI: 10.1080/07315724.2007.10719621] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Antioxidants from plants were known to reduce the oxidative stress by scavenging free radicals, chelating metal ions and reducing inflammation. As increased oxidative stress was implicated in the nigrostriatal dopaminergic neuronal loss in Parkinson's disease (PD), we have assessed whether the plant extracts protects the nigrostriatal dopaminergic neurons in the animal model of PD. METHODS Male adult Sprague-Dawley rats were treated orally between 10 am-11 am each day with the extracts from tangerine peel, grape seeds, cocoa and red clover for four days. One hour after the final dosing, the left medial forebrain bundle was lesioned by infusing the dopaminergic neurotoxin 6-hydroxydopamine (6-OHDA; 12 microg) under anaesthesia. Seven days post-lesion, the number of dopaminergic cells in the substantia nigra pars compacta and the levels of dopamine and its metabolites 3, 4-dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA) in the striata were quantified and compared with the vehicle-treated groups. RESULTS Compared to the unlesioned side, 6-OHDA lesions significantly reduced the number of dopaminergic cells and the levels of dopamine and its metabolites DOPAC and HVA in the vehicle-treated animals. Pretreatment of animals with extracts of tangerine peel (rich in polymethoxylated flavones; 35 mg/kg/day), cocoa-2 (rich in procyanidins; 100 mg/kg/day) and red clover (rich in isoflavones; 200 mg/kg/day) significantly attenuated the 6-OHDA-induced dopaminergic loss. However, no significant protection was seen in animals supplemented with red and white grape seeds (rich in catechins; 100 mg/kg/day), and cocoa-1 (rich in catechins; 100 mg/kg/day). CONCLUSIONS Pre-treatment of plant extracts rich in polymethoxylated flavones, procyanidins and isoflavones but not catechins protected the nigrostriatal dopaminergic neurons in the rat model of PD.
Collapse
Affiliation(s)
- Krishna P Datla
- Department of Cellular and Molecular Neuroscience, Division of Neuroscience and Mental Health, Faculty of Medicine, Imperial College, Charing Cross Campus, St. Dunstan's Road, London W6 8RP, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
18
|
|
19
|
Quintero EM, Willis L, Singleton R, Harris N, Huang P, Bhat N, Granholm AC. Behavioral and morphological effects of minocycline in the 6-hydroxydopamine rat model of Parkinson's disease. Brain Res 2006; 1093:198-207. [PMID: 16712819 DOI: 10.1016/j.brainres.2006.03.104] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2005] [Revised: 03/17/2006] [Accepted: 03/20/2006] [Indexed: 12/21/2022]
Abstract
The neuropathology in many neurodegenerative diseases is mediated by inflammatory cascades that influence neuronal dysfunction and death. Minocycline reduces the neurodegeneration observed in various models of Parkinson's. We exploited the unilateral 6-hydroxydopamine (6-OHDA) lesion model to assess the effect of minocycline on related neurodegeneration. Thirty Fisher 344 rats were divided into three daily treatment groups: (1) after: 45 mg/kg of minocycline beginning 24 h after lesioning; (2) before: 45 mg/kg of minocycline beginning 3 days before 6-OHDA lesioning; (3) control: corresponding saline-treated controls. Animals were assessed for apomorphine-induced rotations for 4 weeks. A longitudinal model for repeated measures showed that both after and before groups had significantly lower rotations than controls (P < 0.001 for both comparisons). Pair-wise group comparisons showed that the before animals rotated less compared to controls (mean rotations: 164 +/- 38 versus 386 +/- 49, respectively, P = 0.001). After animals also rotated significantly less then controls (mean rotations: 125 +/- 41 versus 386 +/- 49, respectively, P < 0.001). Animals receiving minocycline displayed reduced tyrosine hydroxylase-positive cell loss in the lesioned nigra versus contralateral nonlesioned nigra, compared to controls (mean differences: 5065 for after, 3550 for before, and 6483 for controls; P = 0.158 for after versus controls, P = 0.019 for before versus controls). The remaining lesioned nigral cells of both minocycline-treated groups were larger than controls, with the most robust cell size and fiber density observed in the after group. These data suggest that the therapeutic potential of minocycline may depend on the time of drug administration relative to neuropathogenic event.
Collapse
Affiliation(s)
- Elias Matthew Quintero
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, Suite 403, Charleston, SC 29425, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
Huang E, Ong WY. Distribution of ferritin in the rat hippocampus after kainate-induced neuronal injury. Exp Brain Res 2004; 161:502-11. [PMID: 15747160 DOI: 10.1007/s00221-004-2110-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2004] [Accepted: 08/28/2004] [Indexed: 11/25/2022]
Abstract
A gradual increase in iron occurs in the lesioned hippocampus after neuronal injury induced by the excitotoxin kainate, and the present study was carried out to investigate whether this increase in iron might be associated with changes in expression of the iron binding protein, ferritin. An increase in ferritin immunoreactivity was observed in glial cells of the hippocampus, as early as three days after intracerebroventricular injections of kainate. The number of ferritin positive cells peaked four weeks after the kainate injection, and decreased eight and twelve weeks after injection. They were found to be mostly microglia and oligodendrocytes by double immunofluorescence labeling with glial markers. A number of ferritin-labeled endothelial cells were also observed via electron microscopy. The decline in ferritin immunoreactivity four weeks after the injection of kainate is accompanied by an increase in the number of ferric and ferrous iron positive cells in the lesioned tissue. A substantial non-overlap between ferritin and iron-containing cells was observed. In particular, spherical ferric or ferrous iron-laden cells in the degenerating hippocampus were unlabeled for ferritin for long time periods after the kainate injection. An increase in iron, together with a reduced expression of iron binding proteins such as ferritin at long time intervals after kainate lesions, could result in a relative decrease in ferritin-induced ferroxidase activity and the presence of some of the iron in the ferrous form. It is postulated that this may contribute to chronic neuronal injury, following acute kainate-induced neurodegeneration.
Collapse
Affiliation(s)
- En Huang
- Department of Anatomy, National University of Singapore, 119260 Singapore
| | | |
Collapse
|
21
|
He Y, Thong PS, Lee T, Leong SK, Mao BY, Dong F, Watt F. Dopaminergic cell death precedes iron elevation in MPTP-injected monkeys. Free Radic Biol Med 2003; 35:540-7. [PMID: 12927603 DOI: 10.1016/s0891-5849(03)00385-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Though increasing lines of evidence suggest that iron accumulation and iron-induced oxidative stress might be important pathological factors responsible for substantia nigra (SN) cell death in Parkinson's disease (PD), it is still unknown whether iron accumulation is a primary cause or consequence of nigral cell death. Using nuclear microscopy, iron histochemistry, TUNEL method for apoptosis detection, and tyrosine hydroxylase (TH) immunohistochemistry, the present study investigated possible changes in iron contents in the SN and correlations of dopaminergic cell death progression with the process of iron accumulation in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine(MPTP)-induced parkinsonian monkey from 1 d to 18 months after MPTP administration. Our study demonstrated that apoptosis occurred in the ipsilateral SN at 1 d after MPTP injection and the number of TH-positive cells decreased significantly from 1 week onward. However, iron content was significantly increased in the ipsilateral SN from 4.5 months to 18 months after MPTP injection, and the iron increase was significantly correlated to the extent of dopaminergic cell death. These results suggest that dopaminergic cell death induced by MPTP administration might lead to iron accumulation in the monkey SN, and increased iron might contribute to the progression of nigral degeneration.
Collapse
Affiliation(s)
- Yi He
- Department of Surgery, National University of Singapore, Singapore, Singapore.
| | | | | | | | | | | | | |
Collapse
|
22
|
Teismann P, Tieu K, Cohen O, Choi DK, Wu DC, Marks D, Vila M, Jackson-Lewis V, Przedborski S. Pathogenic role of glial cells in Parkinson's disease. Mov Disord 2003; 18:121-9. [PMID: 12539204 DOI: 10.1002/mds.10332] [Citation(s) in RCA: 197] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the progressive loss of the dopaminergic neurons in the substantia nigra pars compacta (SNpc). The loss of these neurons is associated with a glial response composed mainly of activated microglial cells and, to a lesser extent, of reactive astrocytes. This glial response may be the source of trophic factors and can protect against reactive oxygen species and glutamate. Alternatively, this glial response can also mediate a variety of deleterious events related to the production of pro-oxidant reactive species, and pro-inflammatory prostaglandin and cytokines. We discuss the potential protective and deleterious effects of glial cells in the SNpc of PD and examine how those factors may contribute to the pathogenesis of this disease.
Collapse
Affiliation(s)
- Peter Teismann
- Neuroscience Research, Movement Disorder Division, Department of Neurology, Columbia University, New York, New York 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Przedborski S, E. Goldman J. Pathogenic role of glial cells in Parkinson's disease. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s1569-2558(03)31043-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
24
|
Gerlach M, Double KL, Ben-Shachar D, Zecca L, Youdim MBH, Riederer P. Neuromelanin and its interaction with iron as a potential risk factor for dopaminergic neurodegeneration underlying Parkinson's disease. Neurotox Res 2003; 5:35-44. [PMID: 12832223 DOI: 10.1007/bf03033371] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Neuromelanin (NM) is a granular, dark brown pigment produced in some but not all of the dopaminergic neurons of the human substantia nigra (SN). In Parkinson's disease (PD) the pigmented dopaminergic neurons of the SN degenerate, suggesting that this process is related to the presence of NM. As yet it is unknown whether NM in the parkinsonian brain differs from that found in healthy tissue and thus may fulfil a different role within this tissue. The function of NM within the pigmented neurons is unknown but other melanins are believed to play a protective role via attenuation of free radical damage. Experimental evidence suggests that NM may also exhibit this characteristic, possibly by direct inactivation of free radical species or via its ability to chelate transition metals, such as iron. NM has the ability to bind a variety of metals, seven per cent of isolated NM is reported to consist of Fe, Cu, Zn and Cr. Iron is of particular interest as this metal is highly concentrated within the SN. Up to 20 per cent of the total iron contained in the SN from normal subjects is bound within NM. Further, it was demonstrated that NM contains a protein component and that iron is bound to NM in the ferric form. Increased tissue iron found in the parkinsonian SN may saturate iron-chelating sites on NM, and a looser association between iron and NM may result in an increased, rather than decreased, production of free radical species. It is hypothesized that this redox-active iron could be released and involved in a Fenton-like reaction leading to an increased production of oxidative radicals. The resultant radical-mediated cytotoxicity may contribute to cellular damage observed in PD.
Collapse
Affiliation(s)
- Manfred Gerlach
- Clinical Neurochemistry, Department of Child and Youth Psychiatry and Psychotherapy, University of Würzburg, Füchsleinstrasse 15, D-97080 Würzburg, Germany.
| | | | | | | | | | | |
Collapse
|
25
|
Double KL, Ben-Shachar D, Youdim MBH, Zecca L, Riederer P, Gerlach M. Influence of neuromelanin on oxidative pathways within the human substantia nigra. Neurotoxicol Teratol 2002; 24:621-8. [PMID: 12200193 DOI: 10.1016/s0892-0362(02)00218-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neuromelanin (NM) is a dark-coloured pigment produced in the dopaminergic neurons of the human substantia nigra (SN). The function of NM within the pigmented neurons is unknown but other melanins are believed to play a protective role via attenuation of free radical damage. Experimental evidence suggests that NM may also exhibit this characteristic, possibly by directly inactivating free radical species or via its ability to chelate transition metals, such as iron. Increased tissue iron, however, may saturate iron-chelating sites on NM and a looser association between iron and NM may result in an increased, rather than decreased, production of free radical species. The death of NM-pigmented neurons in Parkinson's disease (PD) is associated with both a measurable increase in tissue iron concentrations and indices of free radical mediated damage, suggesting that NM is involved in the aetiology of this disorder. As yet, it is unknown whether NM in the parkinsonian brain differs to that found in healthy tissue and thus may fulfil a different role within this tissue.
Collapse
Affiliation(s)
- K L Double
- Prince of Wales Medical Research Institute, Barker Street, Randwick, Sydney NSW 2031, Australia.
| | | | | | | | | | | |
Collapse
|
26
|
He Y, Appel S, Le W. Minocycline inhibits microglial activation and protects nigral cells after 6-hydroxydopamine injection into mouse striatum. Brain Res 2001; 909:187-93. [PMID: 11478935 DOI: 10.1016/s0006-8993(01)02681-6] [Citation(s) in RCA: 258] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
To determine the role of immune/inflammatory factors in dopaminergic cell degeneration in parkinsonian substantia nigra, we assayed tyrosine hydroxylase (TH)-positive immunoreactive neuronal numbers with stereologic techniques and CD11b-positive immunoreactive microglial profiles following 6-hydroxydopamine (6-OHDA) injection into ipsilateral striatum of mice. We further investigated the effect of minocycline on the inhibition of microglial activation and subsequent protection of nigral cells. The relative number of microglial profiles in the substantia nigra (SN) ipsilateral to the injection increased from 31 to 32% 1-3 days after injection, and increased further to 55% by 7 days and 59% by 14 days, compared with the contralateral SN. These changes started prior to the decrease of TH immunoreactivity of 34% on day 7 and of 42% by day 14. In animals treated with minocycline, microglial activation was inhibited by 47%, and TH positive cells were protected by 21% at day 14 after 6-OHDA injection, compared with those parkinsonian animals without minocycline treatment. All these results suggest that microglial activation may be involved in the nigral cell degeneration in 6-OHDA induced parkinsonian mice.
Collapse
Affiliation(s)
- Y He
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | | | |
Collapse
|
27
|
Double KL, Gerlach M, Youdim MB, Riederer P. Impaired iron homeostasis in Parkinson's disease. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2001:37-58. [PMID: 11205155 DOI: 10.1007/978-3-7091-6301-6_3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Despite physiological systems designed to achieve iron homeostasis, increased concentrations of brain iron have been demonstrated in a range of neurodegenerative diseases. These including the parkinsonian syndromes, the trinucleotide repeat disorders and the dementia syndromes. The increased brain iron is confined to those brain regions most affected by the degeneration characteristic of the particular disorder and is suggested to stimulate cell damage via oxidative mechanisms. Changes in central iron homeostasis have been most closely investigated in PD, as this disorder is well characterised both clinically and pathologically. PD is associated with a significant increase in iron in the degenerating substantia nigra (SN) and is measureable in living PD patients and in post-mortem brain. This increase, however, occurs only in the advanced stages of the disease, suggesting that this phenonoma may be a secondary, rather than a primary initiating event, a hypothesis also supported by evidence from animal experiments. The source of the increased iron is unknown but a variety of changes in iron homeostasis have been identified in PD, both in the brain and in the periphery. The possibility that an increased amount of iron may be transported into the SN is supported by data demonstrating that one form of the iron-binding glycoprotein transferrin family, lactotransferrin, is increased in surviving neurons in the SN in the PD brain and that this change is associated with increased numbers of lactotransferrin receptors on neurons and microvessels in the parkinsonian SN. These changes could represent one mechanism by which iron might concentrate within the PD SN. Alternatively, the measured increased in iron might result from a redistribution of ferritin iron stores. Ferritin is located in glial cells while the degenerating neurons do not stain positive for ferritin. As free radicals are highly reactive, it is unlikely that glial-derived free radicals diffuse across the intracellular space in sufficent quantities to damage neuronal constituents. If intracellular iron release contributes to neuronal damage it seems more probable that an intraneuronal iron source is responsible for oxidant-mediated damage. Such a iron source is neuromelanin (NM), a dark-coloured pigment found in the dopaminergic neurons of the human SN. In the normal brain, NM has the ability to bind a variety of metals, including iron, and increased NM-bound iron is reported in the parkinsonian SN. The consequences of these phenomena for the cell have not yet been clarified. In the absence of significant quantities of iron NM can act as an antioxidant, in that it can interact with and inactivate free radicals. On the other hand, in the presence of iron NM appears to act as a proxidant, increasing the rate of free radical production and thus the oxidative load within the vulnerable neurons. Given that increased iron is only apparent in the advanced stages of the disease it is unlikely that NM is of importance for the primary aetiology of PD. A localised increase in tissue iron and its interaction with NM may be, however, important as a secondary mechanism by increasing the oxidative load on the cell, thereby driving neurodegeneration.
Collapse
Affiliation(s)
- K L Double
- Prince of Wales Medical Research Institute, Sydney, NSW, Australia.
| | | | | | | |
Collapse
|