1
|
Jing XZ, Li GY, Wu YP, Yuan XZ, Chen JL, Wang SH, Wang XP, Li JQ. Neuroimaging Correlates with Clinical Severity in Wilson Disease: A Multiparametric Quantitative Brain MRI. AJNR Am J Neuroradiol 2024:ajnr.A8479. [PMID: 39419533 DOI: 10.3174/ajnr.a8479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/12/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND AND PURPOSE Previous studies have reported metal accumulation and microstructure changes in deep gray nuclei (DGN) in Wilson disease (WD). However, there are limited studies that investigate whether there is metal accumulation and microstructure changes in DGN of patients with WD with normal-appearing routine MRI. This study aimed to evaluate multiparametric changes in DGN of WD and whether the findings correlate with clinical severity in patients with WD. MATERIALS AND METHODS The study enrolled 28 patients with WD (19 with neurologic symptoms) and 25 controls. Fractional anisotropy (FA), mean diffusivity (MD), and magnetic susceptibility in globus pallidus, pontine tegmentum, dentate nucleus, red nucleus, head of caudate nucleus, putamen, substantia nigra, and thalamus were extracted. Correlations between imaging data and the Unified Wilson's Disease Rating Scale (UWDRS) neurologic subitems were explored. RESULTS FA, MD, and susceptibility values were higher in multiple DGN of patients with WD than controls (P < .05). Patients with WD without abnormal signals in DGN on routine MRI also had higher FA, MD, and susceptibility values than controls (P < .017). We found that UWDRS neurologic subscores correlated with FA and susceptibility values of DGN (P < .05). In addition, we also found that FA and susceptibility values in specific structures correlated with specific neurologic symptoms of WD (ie, tremor, parkinsonism, dysarthria, dystonia, and ataxia) (P < .05). CONCLUSIONS Patients with WD have increased FA, MD, and susceptibility values even before the lesion is morphologically apparent on routine MRI. The increased FA and susceptibility values correlate with clinical severity of WD.
Collapse
Affiliation(s)
- Xiao-Zhong Jing
- From the Department of Neurology (X.-Z.J.), The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Neurology (X.-Z.J.), Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gai-Ying Li
- Shanghai Key Laboratory of Magnetic Resonance (G.-Y.L., Y-P.W., J-L.C., J-Q.L.), School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Yu-Peng Wu
- Shanghai Key Laboratory of Magnetic Resonance (G.-Y.L., Y-P.W., J-L.C., J-Q.L.), School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Xiang-Zhen Yuan
- Department of Neurology (X.-Z.Y.), Weifang People's Hospital, Weifang, China
| | - Jia-Lin Chen
- Shanghai Key Laboratory of Magnetic Resonance (G.-Y.L., Y-P.W., J-L.C., J-Q.L.), School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Shu-Hong Wang
- Department of Neurology (S.-H.W.), Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiao-Ping Wang
- Department of Neurology (X.-P.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Qi Li
- Shanghai Key Laboratory of Magnetic Resonance (G.-Y.L., Y-P.W., J-L.C., J-Q.L.), School of Physics and Electronic Science, East China Normal University, Shanghai, China
| |
Collapse
|
2
|
Fan SP, Chen YF, Li CH, Kuo YC, Lee NC, Chien YH, Hwu WL, Tseng TC, Su TH, Hsu CT, Chen HL, Lin CH, Ni YH. Topographical metal burden correlates with brain atrophy and clinical severity in Wilson's disease. Neuroimage 2024; 299:120829. [PMID: 39233127 DOI: 10.1016/j.neuroimage.2024.120829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Quantitative susceptibility mapping (QSM) is a post-processing technique that creates brain susceptibility maps reflecting metal burden through tissue magnetic susceptibility. We assessed topographic differences in magnetic susceptibility between participants with and without Wilson's disease (WD), correlating these findings with clinical severity, brain volume, and biofluid copper and iron indices. METHODS A total of 43 patients with WD and 20 unaffected controls, were recruited. QSM images were derived from a 3T MRI scanner. Clinical severity was defined using the minimal Unified Wilson's Disease Rating Scale (M-UWDRS) and Montreal Cognitive Assessment scoring. Differences in magnetic susceptibilities between groups were evaluated using general linear regression models, adjusting for age and sex. Correlations between the susceptibilities and clinical scores were analyzed using Spearman's method. RESULTS In age- and sex-adjusted analyses, magnetic susceptibility values were increased in WD patients compared with controls, including caudate nucleus, putamen, globus pallidus, and substantia nigra (all p < 0.01). Putaminal susceptibility was greater with an initial neuropsychiatric presentation (n = 25) than with initial hepatic dysfunction (n = 18; p = 0.04). Susceptibility changes correlated negatively with regional brain volume in almost all topographic regions. Serum ferritin, but not serum copper or ceruloplasmin, correlated positively with magnetic susceptibility level in the caudate nucleus (p = 0.04), putamen (p = 0.04) and the hippocampus (p = 0.03). The dominance of magnetic susceptibility in cortical over subcortical regions correlated with M-UWDRS scores (p < 0.01). CONCLUSION The magnetic susceptibility changes could serve as a surrogate marker for patients with WD.
Collapse
Affiliation(s)
- Sung-Pin Fan
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Fang Chen
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng-Hsuan Li
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan; Department of Neurology, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - Yih-Chih Kuo
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan; Department of Neurology, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - Ni-Chung Lee
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - Yin-Hsiu Chien
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - Wuh-Liang Hwu
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - Tai-Chung Tseng
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Tung-Hung Su
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chien-Ting Hsu
- Department of Pediatrics, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 100, Taiwan; Department of Pediatrics, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - Huey-Ling Chen
- Department of Pediatrics, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 100, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan; Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan; Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.
| | - Yen-Hsuan Ni
- Department of Pediatrics, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 100, Taiwan.
| |
Collapse
|
3
|
Dong J, Xiang G, Xia X, Xu L, Wen P, Xu C, Xu Y, Su Y, Song Y, Tong H, Zhu Q, Han Y, Han Y, Cheng N, Wang H, Zhou H. Aberrant copper metabolism and hepatic inflammation cause neurological manifestations in a mouse model of Wilson's disease. J Neuroinflammation 2024; 21:235. [PMID: 39334421 PMCID: PMC11437830 DOI: 10.1186/s12974-024-03178-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/16/2024] [Indexed: 09/30/2024] Open
Abstract
Pathogenic germline mutations in the P-type copper-transporting ATPase (ATP7B) gene cause Wilson's disease (WD), a hereditary disorder characterized by disrupted copper metabolism. The Arg778Leu (R778L) mutation in exon 8 is prevalent among individuals with WD in East Asia and is associated with more severe phenotypes. In this study, we generated a WD mouse model harboring R778L mutation (R778L mice) using CRISPR/Cas9. R778L mice exhibit a range of pathological characteristics resembling those of patients with WD and the same point mutations, including aberrant copper metabolism, pathological cellular injury, inflammation, and severe hepatic fibrosis. At 3-5 months of age, these mice started to display neurological deficits in motor coordination and cognitive dysfunction, accompanied by increased expression of inflammatory cytokines in the central nervous system. Microglia in the striatum and cortex exhibit significant activation, shorter processes, and decreased branch points. However, the Cu2+ levels in the brain tissue of R778L mice did not differ significantly from those of wild-type mice. Notably, inhibition of hepatic inflammation with PJ34 or siNfkb markedly alleviated the deficiencies in cognitive performance and improved locomotor activity in R778L mice. Thus, this study establishes a novel murine model to investigate the pathophysiology of WD, highlights the liver-brain crosstalk responsible for neurological manifestations in individuals with WD caused by the R778L point mutation, and demonstrates the potential of modulating liver inflammation as a therapeutic strategy for alleviating the neurological manifestations of WD.
Collapse
Affiliation(s)
- Jianjian Dong
- Institute of Neurology, Anhui University of Chinese Medicine, 357, Changjiang Rd. Middle, Hefei, Anhui, 230061, China
- Center for Xin-An Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Guanghai Xiang
- Key Laboratory of Immune Response and Immunotherapy, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China
| | - Xiaoxue Xia
- Institute of Neurology, Anhui University of Chinese Medicine, 357, Changjiang Rd. Middle, Hefei, Anhui, 230061, China
| | - Lewen Xu
- Institute of Neurology, Anhui University of Chinese Medicine, 357, Changjiang Rd. Middle, Hefei, Anhui, 230061, China
| | - Peihua Wen
- Institute of Neurology, Anhui University of Chinese Medicine, 357, Changjiang Rd. Middle, Hefei, Anhui, 230061, China
| | - Chenchen Xu
- Institute of Neurology, Anhui University of Chinese Medicine, 357, Changjiang Rd. Middle, Hefei, Anhui, 230061, China
| | - Yin Xu
- Institute of Neurology, Anhui University of Chinese Medicine, 357, Changjiang Rd. Middle, Hefei, Anhui, 230061, China
| | - Yushuang Su
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yanze Song
- Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 1 Beichen West Rd, Beijing, 100101, China
| | - Haiyang Tong
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Qingjun Zhu
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Yongzhu Han
- Institute of Neurology, Anhui University of Chinese Medicine, 357, Changjiang Rd. Middle, Hefei, Anhui, 230061, China
- Center for Xin-An Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yongsheng Han
- Institute of Neurology, Anhui University of Chinese Medicine, 357, Changjiang Rd. Middle, Hefei, Anhui, 230061, China
- Center for Xin-An Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Nan Cheng
- Institute of Neurology, Anhui University of Chinese Medicine, 357, Changjiang Rd. Middle, Hefei, Anhui, 230061, China.
- Center for Xin-An Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Haoyi Wang
- Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 1 Beichen West Rd, Beijing, 100101, China.
| | - Hong Zhou
- School of Life Sciences, Anhui Medical University, Hefei, 230032, China.
- School of Basic Medical Science, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
4
|
Ratner MH, Rutchik JS. A rare case of early onset lewy body dementia with parkinsonism associated with chronic exposure to copper contaminated drinking water. FRONTIERS IN TOXICOLOGY 2024; 6:1451235. [PMID: 39285928 PMCID: PMC11402898 DOI: 10.3389/ftox.2024.1451235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/07/2024] [Indexed: 09/19/2024] Open
Abstract
There is a well-recognized relationship between a person's body burden of essential trace elements such as copper and their neurological function in which both deficiencies and exposures to excessive concentrations are associated with adverse clinical outcomes. Preclinical studies indicate chronic excess copper exposure is associated with altered motor function, dopaminergic neuronal loss, astrocytosis, and microgliosis. Copper also promotes oligomerization and fibrilization of α-synuclein suggesting it may hasten the course of an α-synucleinopathy. Here we report a rare case of early onset Lewy Body Dementia with Parkinsonism in a 53-year-old Caucasian woman exposed to copper contaminated drinking water for more than 10 years. Her hair and that of her daughter had streaks of blue-green discoloration as did the porcelain sinks in their home. Testing confirmed copper contamination of the drinking water. A neurologist diagnosed her with Lewy Body Dementia with Parkinsonism. Skin biopsy for phosphorylated α was consistent with a diagnosis of an α-synucleinopathy. These findings suggest chronic exposure to excessive copper may act as disease modifying factor in Lewy Body Dementia with Parkinsonism. It has previously been recommended that individuals at risk of Alzheimer's disease (AD) avoid excessive intake of copper. Genetic studies indicate that Lewy Body Dementia shares risk factors and pathways with AD. Based on the observations in this patient we recommend that individuals at risk for an α-synucleinopathy based on a positive family history, genetic testing, and/or positive results on a skin biopsy for phosphorylated α-synuclein avoid exposure to excess copper.
Collapse
Affiliation(s)
- Marcia H Ratner
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Jonathan S Rutchik
- Neurology, Environmental and Occupational Medicine Associates, CA and Division of Medicine, Occupational Medicine, University of California at San Francisco, San Francisco, CA, United States
| |
Collapse
|
5
|
Sailer J, Nagel J, Akdogan B, Jauch AT, Engler J, Knolle PA, Zischka H. Deadly excess copper. Redox Biol 2024; 75:103256. [PMID: 38959622 PMCID: PMC11269798 DOI: 10.1016/j.redox.2024.103256] [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: 04/27/2024] [Revised: 06/13/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024] Open
Abstract
Higher eukaryotes' life is impossible without copper redox activity and, literally, every breath we take biochemically demonstrates this. However, this dependence comes at a considerable price to ensure target-oriented copper action. Thereto its uptake, distribution but also excretion are executed by specialized proteins with high affinity for the transition metal. Consequently, malfunction of copper enzymes/transporters, as is the case in hereditary Wilson disease that affects the intracellular copper transporter ATP7B, comes with serious cellular damage. One hallmark of this disease is the progressive copper accumulation, primarily in liver but also brain that becomes deadly if left untreated. Such excess copper toxicity may also result from accidental ingestion or attempted suicide. Recent research has shed new light into the cell-toxic mechanisms and primarily affected intracellular targets and processes of such excess copper that may even be exploited with respect to cancer therapy. Moreover, new therapies are currently under development to fight against deadly toxic copper.
Collapse
Affiliation(s)
- Judith Sailer
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Judith Nagel
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Banu Akdogan
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Adrian T Jauch
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Jonas Engler
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Percy A Knolle
- Institute of Molecular Immunology and Experimental Oncology, Technical University Munich, School of Medicine and Health, Munich, Germany
| | - Hans Zischka
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine and Health, Munich, Germany; Institute of Molecular Toxicology and Pharmacology, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
6
|
Gromadzka G, Czerwińska J, Krzemińska E, Przybyłkowski A, Litwin T. Wilson's Disease-Crossroads of Genetics, Inflammation and Immunity/Autoimmunity: Clinical and Molecular Issues. Int J Mol Sci 2024; 25:9034. [PMID: 39201720 PMCID: PMC11354778 DOI: 10.3390/ijms25169034] [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: 07/19/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
Wilson's disease (WD) is a rare, autosomal recessive disorder of copper metabolism caused by pathogenic mutations in the ATP7B gene. Cellular copper overload is associated with impaired iron metabolism. Oxidative stress, cuproptosis, and ferroptosis are involved in cell death in WD. The clinical picture of WD is variable. Hepatic/neuropsychiatric/other symptoms may manifest in childhood/adulthood and even old age. It has been shown that phenotypic variability may be determined by the type of ATP7B genetic variants as well as the influence of various genetic/epigenetic, environmental, and lifestyle modifiers. In 1976, immunological abnormalities were first described in patients with WD. These included an increase in IgG and IgM levels and a decrease in the percentage of T lymphocytes, as well as a weakening of their bactericidal effect. Over the following years, it was shown that there is a bidirectional relationship between copper and inflammation. Changes in serum cytokine concentrations and the relationship between cytokine gene variants and the clinical course of the disease have been described in WD patients, as well as in animal models of this disease. Data have also been published on the occurrence of antinuclear antibodies (ANAs), antineutrophil cytoplasmic antibodies (ANCAs), anti-muscle-specific tyrosine kinase antibodies, and anti-acetylcholine receptor antibodies, as well as various autoimmune diseases, including systemic lupus erythematosus (SLE), myasthenic syndrome, ulcerative colitis, multiple sclerosis (MS), polyarthritis, and psoriasis after treatment with d-penicillamine (DPA). The occurrence of autoantibodies was also described, the presence of which was not related to the type of treatment or the form of the disease (hepatic vs. neuropsychiatric). The mechanisms responsible for the occurrence of autoantibodies in patients with WD are not known. It has also not been clarified whether they have clinical significance. In some patients, WD was differentiated or coexisted with an autoimmune disease, including autoimmune hepatitis or multiple sclerosis. Various molecular mechanisms may be responsible for immunological abnormalities and/or the inflammatory processes in WD. Their better understanding may be important for explaining the reasons for the diversity of symptoms and the varied course and response to therapy, as well as for the development of new treatment regimens for WD.
Collapse
Affiliation(s)
- Grażyna Gromadzka
- Department of Biomedical Sciences, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University, Wóycickiego Street 1/3, 01-938 Warsaw, Poland
| | - Julia Czerwińska
- Students Scientific Association “Immunis”, Cardinal Stefan Wyszynski University, Dewajtis Street 5, 01-815 Warsaw, Poland
| | - Elżbieta Krzemińska
- Students Scientific Association “Immunis”, Cardinal Stefan Wyszynski University, Dewajtis Street 5, 01-815 Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland;
| | - Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego Street 9, 02-957 Warsaw, Poland;
| |
Collapse
|
7
|
Litwin T, Rędzia-Ogrodnik B, Antos A, Przybyłkowski A, Członkowska A, Bembenek JP. Brain Magnetic Resonance Imaging in Wilson's Disease-Significance and Practical Aspects-A Narrative Review. Brain Sci 2024; 14:727. [PMID: 39061467 PMCID: PMC11274939 DOI: 10.3390/brainsci14070727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Wilson's disease (WD) is a genetic disorder of copper metabolism with pathological copper accumulation in many organs, resulting in clinical symptoms, mostly hepatic and neuropsychiatric. As copper accumulates in the brain during WD, and almost 50% of WD patients at diagnosis present with neurological symptoms, neuroimaging studies (especially brain magnetic resonance imaging (MRI)) are part of WD diagnosis. The classical sequences (T1, T2, and fluid-attenuated inversion recovery) were used to describe brain MRI; however, with the development of neuroradiology, several papers proposed the use of new MRI sequences and techniques like susceptibility-weighted images, T2*, diffusion MRI, tractography, volumetric assessment and post-processing brain MRI analysis of paramagnetic accumulation-quantitative susceptibility mapping. Based on these neuroradiological data in WD, currently, brain MRI semiquantitative scale and the pathognomonic neuroradiological brain MRI signs in WD were proposed. Further, the volumetric studies and brain iron accumulation MRI analysis suggested brain atrophy and iron accumulation as biomarkers of neurological WD disease severity. All these results highlight the significance of brain MRI examinations in WD. Due to the extreme progress of these studies, based on the available literature, the authors present the current state of knowledge about the significance, practical aspects, and future directions of brain MRI in WD.
Collapse
Affiliation(s)
- Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland; (B.R.-O.); (A.A.); (A.C.)
| | - Barbara Rędzia-Ogrodnik
- Second Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland; (B.R.-O.); (A.A.); (A.C.)
| | - Agnieszka Antos
- Second Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland; (B.R.-O.); (A.A.); (A.C.)
| | - Adam Przybyłkowski
- Department of Gastroenterology, Medical University, Warsaw 02-097, Poland;
| | - Anna Członkowska
- Second Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland; (B.R.-O.); (A.A.); (A.C.)
| | - Jan Paweł Bembenek
- Department of Neurophysiology, Institute Psychiatry and Neurology, 02-957 Warsaw, Poland;
| |
Collapse
|
8
|
Gromadzka G, Wilkaniec A, Tarnacka B, Hadrian K, Bendykowska M, Przybyłkowski A, Litwin T. The Role of Glia in Wilson's Disease: Clinical, Neuroimaging, Neuropathological and Molecular Perspectives. Int J Mol Sci 2024; 25:7545. [PMID: 39062788 PMCID: PMC11276698 DOI: 10.3390/ijms25147545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Wilson's disease (WD) is inherited in an autosomal recessive manner and is caused by pathogenic variants of the ATP7B gene, which are responsible for impaired copper transport in the cell, inhibition of copper binding to apoceruloplasmin, and biliary excretion. This leads to the accumulation of copper in the tissues. Copper accumulation in the CNS leads to the neurological and psychiatric symptoms of WD. Abnormalities of copper metabolism in WD are associated with impaired iron metabolism. Both of these elements are redox active and may contribute to neuropathology. It has long been assumed that among parenchymal cells, astrocytes have the greatest impact on copper and iron homeostasis in the brain. Capillary endothelial cells are separated from the neuropil by astrocyte terminal legs, putting astrocytes in an ideal position to regulate the transport of iron and copper to other brain cells and protect them if metals breach the blood-brain barrier. Astrocytes are responsible for, among other things, maintaining extracellular ion homeostasis, modulating synaptic transmission and plasticity, obtaining metabolites, and protecting the brain against oxidative stress and toxins. However, excess copper and/or iron causes an increase in the number of astrocytes and their morphological changes observed in neuropathological studies, as well as a loss of the copper/iron storage function leading to macromolecule peroxidation and neuronal loss through apoptosis, autophagy, or cuproptosis/ferroptosis. The molecular mechanisms explaining the possible role of glia in copper- and iron-induced neurodegeneration in WD are largely understood from studies of neuropathology in Parkinson's disease and Alzheimer's disease. Understanding the mechanisms of glial involvement in neuroprotection/neurotoxicity is important for explaining the pathomechanisms of neuronal death in WD and, in the future, perhaps for developing more effective diagnostic/treatment methods.
Collapse
Affiliation(s)
- Grażyna Gromadzka
- Department of Biomedical Sciences, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University, Wóycickiego 1/3, 01-938 Warsaw, Poland
| | - Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland
| | - Beata Tarnacka
- Department of Rehabilitation, Medical University of Warsaw, Spartańska 1, 02-637 Warsaw, Poland
| | - Krzysztof Hadrian
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland (A.P.)
| | - Maria Bendykowska
- Students Scientific Association “Immunis”, Cardinal Stefan Wyszynski University, Dewajtis 5, 01-815 Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland (A.P.)
| | - Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957 Warsaw, Poland
| |
Collapse
|
9
|
Li S, Zhua Y, Liu X. Parkinsonism in liver diseases or dysfunction. Med Clin (Barc) 2024:S0025-7753(24)00356-7. [PMID: 38955605 DOI: 10.1016/j.medcli.2024.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 04/20/2024] [Indexed: 07/04/2024]
Abstract
Parkinsonism in liver diseases or dysfunction, mainly including neurological manifestations in hereditary liver diseases and neurological complications of advanced liver diseases, occur in isolation or in combination with other movement disorders, and progress along disease course. Prominent akinetic-rigidity syndrome, various onset and progression, poor levodopa response and metabolism abnormalities reflected by serum biomarkers and neuroimaging, make this atypical parkinsonism recognizable and notable in clinical practice. Different susceptibility of brain areas, especially in basal ganglia, to manganese, iron, copper, ammonia overload, together with subsequent oxidative stress, neurotransmitter alterations, disturbed glia-neuron homeostasis and eventually neurotoxicity, contribute to parkinsonism under the circumstances of insufficient liver clearance ability. These mechanisms are interrelated and may interact collectively, adding to the complexity of clinical manifestations and treatment responses. This review summarizes shared clinical features of parkinsonism in liver diseases or dysfunction, depicts their underlying mechanisms and suggests practical flowchart for differential diagnosis.
Collapse
Affiliation(s)
- Sichen Li
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxia Zhua
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xi Liu
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
10
|
Su D, Zhang Z, Zhang Z, Zheng S, Yao T, Dong Y, Zhu W, Wei N, Suo Y, Liu X, Zhao H, Wang Z, Ma H, Li W, Zhou J, Lam JST, Wu T, Dusek P, Stoessl AJ, Wang X, Jing J, Feng T. Distinctive Pattern of Metal Deposition in Neurologic Wilson Disease: Insights From 7T Susceptibility-Weighted Imaging. Neurology 2024; 102:e209478. [PMID: 38830145 PMCID: PMC11244749 DOI: 10.1212/wnl.0000000000209478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/11/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Noninvasive and accurate biomarkers of neurologic Wilson disease (NWD), a rare inherited disorder, could reduce diagnostic error or delay. Excessive subcortical metal deposition seen on susceptibility imaging has suggested a characteristic pattern in NWD. With submillimeter spatial resolution and increased contrast, 7T susceptibility-weighted imaging (SWI) may enable better visualization of metal deposition in NWD. In this study, we sought to identify a distinctive metal deposition pattern in NWD using 7T SWI and investigate its diagnostic value and underlying pathophysiologic mechanism. METHODS Patients with WD, healthy participants with monoallelic ATP7B variant(s) on a single chromosome, and health controls (HCs) were recruited. NWD and non-NWD (nNWD) were defined according to the presence or absence of neurologic symptoms during investigation. Patients with other diseases with comparable clinical or imaging manifestations, including early-onset Parkinson disease (EOPD), multiple system atrophy (MSA), progressive supranuclear palsy (PSP), and neurodegeneration with brain iron accumulation (NBIA), were additionally recruited and assessed for exploratory comparative analysis. All participants underwent 7T T1, T2, and high-resolution SWI scanning. Quantitative susceptibility mapping and principal component analysis were performed to illustrate metal distribution. RESULTS We identified a linear signal intensity change consisting of a hyperintense strip at the lateral border of the globus pallidus in patients with NWD. We termed this feature "hyperintense globus pallidus rim sign." This feature was detected in 38 of 41 patients with NWD and was negative in all 31 nNWD patients, 15 patients with EOPD, 30 patients with MSA, 15 patients with PSP, and 12 patients with NBIA; 22 monoallelic ATP7B variant carriers; and 41 HC. Its sensitivity to differentiate between NWD and HC was 92.7%, and specificity was 100%. Severity of the hyperintense globus pallidus rim sign measured by a semiquantitative scale was positively correlated with neurologic severity (ρ = 0.682, 95% CI 0.467-0.821, p < 0.001). Patients with NWD showed increased susceptibility in the lenticular nucleus with high regional weights in the lateral globus pallidus and medial putamen. DISCUSSION The hyperintense globus pallidus rim sign showed high sensitivity and excellent specificity for diagnosis and differential diagnosis of NWD. It is related to a special metal deposition pattern in the lenticular nucleus in NWD and can be considered as a novel neuroimaging biomarker of NWD. CLASSIFICATION OF EVIDENCE The study provides Class II evidence that the hyperintense globus pallidus rim sign on 7T SWI MRI can accurately diagnose neurologic WD.
Collapse
Affiliation(s)
- Dongning Su
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Zhijin Zhang
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Zhe Zhang
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Sujun Zheng
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Tingyan Yao
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yi Dong
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Wanlin Zhu
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Ning Wei
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yue Suo
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Xinyao Liu
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Huiqing Zhao
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Zhan Wang
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Huizi Ma
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Wei Li
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Junhong Zhou
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Joyce S T Lam
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Tao Wu
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Petr Dusek
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - A Jon Stoessl
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Xiaoping Wang
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Jing Jing
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Tao Feng
- From the Department of Neurology (D.S., Zhijin Zhang, H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.), Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.S., Zhijin Zhang, Zhe Zhang, W.Z., N.W., Y.S., X.L., H.Z., Z.W., H.M., W.L., T.W., J.J., T.F.); Tiantan Neuroimaging Center of Excellence (Zhe Zhang, W.Z., N.W., Y.S., X.L., J.J.), and Department of Hepatology (S.Z.), Beijing Youan Hospital, Capital Medical University; Department of Neurology (T.Y.), Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Disorders; Senior Department of Hepatology (Y.D.), the Fifth Medical Center of PLA General Hospital, Beijing, China; Hinda and Arthur Marcus Institute for Aging Research (J.Z.), Hebrew SeniorLife, Roslindale; Harvard Medical School (J.Z.), Boston, MA; Pacific Parkinson's Research Centre (J.S.T.L., A.J.S.), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Neurology and Centre of Clinical Neuroscience (P.D.), First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Division of Neurology (A.J.S.), Department of Medicine, University of British Columbia, Vancouver, Canada; and Department of Neurology (X.W.), Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
11
|
Hu S, Wei T, Li C, Wang X, Nguchu BA, Wang Y, Dong T, Yang Y, Ding Y, Qiu B, Yang W. Abnormalities in subcortical function and their treatment response in Wilson's disease. Neuroimage Clin 2024; 43:103618. [PMID: 38830274 PMCID: PMC11180346 DOI: 10.1016/j.nicl.2024.103618] [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: 01/22/2024] [Revised: 04/22/2024] [Accepted: 05/10/2024] [Indexed: 06/05/2024]
Abstract
Extensive neuroimaging abnormalities in subcortical regions build the pathophysiological basis of Wilson's disease (WD). Yet, subcortical topographic organization fails to articulate, leaving a huge gap in understanding the neural mechanism of WD. Thus, how functional abnormalities of WD subcortical regions influence complex clinical symptoms and response to treatment remain unknown. Using resting-state functional MRI data from 232 participants (including 130 WD patients and 102 healthy controls), we applied a connectivity-based parcellation technique to develop a subcortical atlas for WD. The atlas was further used to investigate abnormalities in subcortical function (ASF) by exploring intrasubcortical functional connectivity (FC) and topographic organization of cortico-subcortical FC. We further used support vector machine (SVM) to integrate these functional abnormalities into the ASF score, which serves as a biomarker for characterizing individual subcortical dysfunction for WD. Finally, the baseline ASF score and one-year treatment data of the follow-up WD patients were used to assess treatment response. A group set of subcortical parcellations was evaluated, in which 26 bilateral regions well recapitulated the anatomical nuclei of the subcortical areas of WD. The results of cortico-subcortical FC and intrasubcortical FC reveal that dysfunction of the somatomotor networks-lenticular nucleus-thalamic pathways is involved in complex symptoms of WD. The ASF score was able to characterize disease progression and was significantly associated with treatment response of WD. Our findings provide a comprehensive elaboration of functional abnormalities of WD subcortical regions and reveal their association with clinical presentations, improving our understanding of the functional neural underpinnings in WD. Furthermore, abnormalities in subcortical function could serve as a potential biomarker for understanding the disease progression and evaluating treatment response of WD.
Collapse
Affiliation(s)
- Sheng Hu
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China; Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 2300026, China; School of Medical Information Engineering, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230012, China
| | - Taohua Wei
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China; Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
| | - Chuanfu Li
- Medical Imaging Center, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China.
| | - Xiaoxiao Wang
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 2300026, China
| | | | - Yanming Wang
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 2300026, China
| | - Ting Dong
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China; Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
| | - Yulong Yang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
| | - Yufeng Ding
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China
| | - Bensheng Qiu
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 2300026, China.
| | - Wenming Yang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China; Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, 230031, China.
| |
Collapse
|
12
|
Zhong G, Wang X, Li J, Xie Z, Wu Q, Chen J, Wang Y, Chen Z, Cao X, Li T, Liu J, Wang Q. Insights Into the Role of Copper in Neurodegenerative Diseases and the Therapeutic Potential of Natural Compounds. Curr Neuropharmacol 2024; 22:1650-1671. [PMID: 38037913 PMCID: PMC11284712 DOI: 10.2174/1570159x22666231103085859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/23/2023] [Accepted: 06/05/2023] [Indexed: 12/02/2023] Open
Abstract
Neurodegenerative diseases encompass a collection of neurological disorders originating from the progressive degeneration of neurons, resulting in the dysfunction of neurons. Unfortunately, effective therapeutic interventions for these diseases are presently lacking. Copper (Cu), a crucial trace element within the human body, assumes a pivotal role in various biological metabolic processes, including energy metabolism, antioxidant defense, and neurotransmission. These processes are vital for the sustenance, growth, and development of organisms. Mounting evidence suggests that disrupted copper homeostasis contributes to numerous age-related neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), Wilson's disease (WD), Menkes disease (MD), prion diseases, and multiple sclerosis (MS). This comprehensive review investigates the connection between the imbalance of copper homeostasis and neurodegenerative diseases, summarizing pertinent drugs and therapies that ameliorate neuropathological changes, motor deficits, and cognitive impairments in these conditions through the modulation of copper metabolism. These interventions include Metal-Protein Attenuating Compounds (MPACs), copper chelators, copper supplements, and zinc salts. Moreover, this review highlights the potential of active compounds derived from natural plant medicines to enhance neurodegenerative disease outcomes by regulating copper homeostasis. Among these compounds, polyphenols are particularly abundant. Consequently, this review holds significant implications for the future development of innovative drugs targeting the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Guangcheng Zhong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Wang
- The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiaqi Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhouyuan Xie
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiqing Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaxin Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yiyun Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziying Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyue Cao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianyao Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinman Liu
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
13
|
Mariño Z, Berenguer M, Peña-Quintana L, Olveira A, Miralpeix A, Sastre I, Reyes-Domínguez A, Castillo P, García-Solà C, Bono A, Romero M, Pérez-Sádaba FJ, Aceituno S, Anguera A. Health-Related Quality of Life in Patients Living with Wilson Disease in Spain: A Cross-Sectional Observational Study. J Clin Med 2023; 12:4823. [PMID: 37510937 PMCID: PMC10381913 DOI: 10.3390/jcm12144823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Wilson disease (WD) is a rare copper metabolism disorder caused by mutations in the ATP7B gene. It usually affects young individuals and can produce hepatic and/or neurological involvement, potentially affecting health-related quality of life (HRQoL). We assessed HRQoL in a cohort of Spanish patients with WD and evaluated disease impact on several domains of patients' lives, treatment adherence, drug preference and satisfaction, and healthcare resource utilisation in a cross-sectional, retrospective, multicentric, observational study. A total of 102 patients were included: 81.4% presented isolated liver involvement (group H) and 18.6% presented neurological or mixed involvement (group EH). Up to 30% of patients reported a deteriorated emotional status with anxiety and depression, which was greater in the EH subgroup; the use of neuropsychiatric drugs was high. Over 70% of the patients were satisfied with their current treatment but complained about taking too many pills, stating they would consider switching to another more patient-friendly treatment if available. The Simplified Medication Adherence Questionnaire revealed only 22.5% of patients were fully adherent to therapy, suggesting that alternative therapies are needed. This real-world study, even though is highly enriched with hepatic patients and mild disease, shows that WD impacts patients' HRQoL, especially in the emotional domain.
Collapse
Affiliation(s)
- Zoe Mariño
- Liver Unit, Hospital Clínic Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), European Reference Networks (ERN)-RARE Liver, University of Barcelona, 08007 Barcelona, Spain
| | - Marina Berenguer
- Department of Gastroenterology and Hepatology, Hospital Universitari i Politècnic La Fe, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Investigación Sanitaria (IIS La Fe), 46026 Valencia, Spain
| | - Luis Peña-Quintana
- Pediatric Gastroenterology, Hepatoloy and Nutrition Unit, Complejo Hospitalario Universitario Insular Materno-Infantil, 35016 Las Palmas de Gran Canaria, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, University of Las Palmas de Gran Canaria, 35001 Las Palmas de Gran Canaria, Spain
| | - Antonio Olveira
- Department of Gastroenterology, Hospital La Paz, 28046 Madrid, Spain
| | - Anna Miralpeix
- Liver Unit, Hospital Clínic Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), European Reference Networks (ERN)-RARE Liver, University of Barcelona, 08007 Barcelona, Spain
| | - Isabel Sastre
- Department of Gastroenterology and Hepatology, Hospital Universitari i Politècnic La Fe, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Investigación Sanitaria (IIS La Fe), 46026 Valencia, Spain
| | - Ana Reyes-Domínguez
- Pediatric Gastroenterology, Hepatoloy and Nutrition Unit, Complejo Hospitalario Universitario Insular Materno-Infantil, 35016 Las Palmas de Gran Canaria, Spain
| | - Pilar Castillo
- Department of Gastroenterology, Hospital La Paz, 28046 Madrid, Spain
| | - Clàudia García-Solà
- Liver Unit, Hospital Clínic Barcelona, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), European Reference Networks (ERN)-RARE Liver, University of Barcelona, 08007 Barcelona, Spain
| | - Ariadna Bono
- Department of Gastroenterology and Hepatology, Hospital Universitari i Politècnic La Fe, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Investigación Sanitaria (IIS La Fe), 46026 Valencia, Spain
| | - Miriam Romero
- Department of Gastroenterology, Hospital La Paz, 28046 Madrid, Spain
| | | | | | - Anna Anguera
- Medical Department, Alexion, AstraZeneca Rare Diseases, 08028 Barcelona, Spain
| |
Collapse
|
14
|
Nguyen HH, Swain MG. Avenues within the gut-liver-brain axis linking chronic liver disease and symptoms. Front Neurosci 2023; 17:1171253. [PMID: 37521690 PMCID: PMC10372440 DOI: 10.3389/fnins.2023.1171253] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/09/2023] [Indexed: 08/01/2023] Open
Abstract
Symptoms of fatigue, social withdrawal and mood disturbances are commonly encountered in patients with chronic liver disease and have a detrimental effect on patient quality of life. Treatment options for these symptoms are limited and a current area of unmet medical need. In this review, we will evaluate the potential mechanistic avenues within the gut-liver-brain axis that may be altered in the setting of chronic liver disease that drive the development of these symptoms. Both clinical and pre-clinical studies will be highlighted as we discuss how perturbations in host immune response, microbiome, neural responses, and metabolites composition can affect the central nervous system.
Collapse
Affiliation(s)
- Henry H. Nguyen
- University of Calgary Liver Unit, Departments of Medicine and Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mark G. Swain
- University of Calgary Liver Unit, Department of Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
15
|
Einer C, Munk DE, Park E, Akdogan B, Nagel J, Lichtmannegger J, Eberhagen C, Rieder T, Vendelbo MH, Michalke B, Wimmer R, Blutke A, Feuchtinger A, Dershwitz P, DiSpirito AM, Islam T, Castro RE, Min BK, Kim T, Choi S, Kim D, Jung C, Lee H, Park D, Im W, Eun SY, Cho YH, Semrau JD, Rodrigues CMP, Hohenester S, Damgaard Sandahl T, DiSpirito AA, Zischka H. ARBM101 (Methanobactin SB2) Drains Excess Liver Copper via Biliary Excretion in Wilson's Disease Rats. Gastroenterology 2023; 165:187-200.e7. [PMID: 36966941 DOI: 10.1053/j.gastro.2023.03.216] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND & AIMS Excess copper causes hepatocyte death in hereditary Wilson's disease (WD). Current WD treatments by copper-binding chelators may gradually reduce copper overload; they fail, however, to bring hepatic copper close to normal physiological levels. Consequently, lifelong daily dose regimens are required to hinder disease progression. This may result in severe issues due to nonadherence or unwanted adverse drug reactions and also due to drug switching and ultimate treatment failures. This study comparatively tested bacteria-derived copper binding agents-methanobactins (MBs)-for efficient liver copper depletion in WD rats as well as their safety and effect duration. METHODS Copper chelators were tested in vitro and in vivo in WD rats. Metabolic cage housing allowed the accurate assessment of animal copper balances and long-term experiments related to the determination of minimal treatment phases. RESULTS We found that copper-binding ARBM101 (previously known as MB-SB2) depletes WD rat liver copper dose dependently via fecal excretion down to normal physiological levels within 8 days, superseding the need for continuous treatment. Consequently, we developed a new treatment consisting of repetitive cycles, each of ∼1 week of ARBM101 applications, followed by months of in-between treatment pauses to ensure a healthy long-term survival in WD rats. CONCLUSIONS ARBM101 safely and efficiently depletes excess liver copper from WD rats, thus allowing for short treatment periods as well as prolonged in-between rest periods.
Collapse
Affiliation(s)
- Claudia Einer
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ditte Emilie Munk
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Eok Park
- R&D Center, ArborMed Company Ltd, Pangyo, Seongnam, Gyeonggi-do, Republic of Korea; Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
| | - Banu Akdogan
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Judith Nagel
- Institute of Toxicology and Environmental Hygiene, School of Medicine, Technical University Munich, Munich, Germany
| | - Josef Lichtmannegger
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Carola Eberhagen
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Tamara Rieder
- Institute of Toxicology and Environmental Hygiene, School of Medicine, Technical University Munich, Munich, Germany
| | - Mikkel H Vendelbo
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Bernhard Michalke
- Research Unit Analytical BioGeoChemistry, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ralf Wimmer
- Department of Medicine II, Ludwig Maximilian University Munich, Munich, Germany
| | - Andreas Blutke
- Research Unit Analytical Pathology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Philip Dershwitz
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa
| | - Ana M DiSpirito
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa
| | - Tawhidul Islam
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Rui E Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Byong-Keol Min
- R&D Center, ArborMed Company Ltd, Pangyo, Seongnam, Gyeonggi-do, Republic of Korea
| | - TaeWon Kim
- R&D Center, ArborMed Company Ltd, Pangyo, Seongnam, Gyeonggi-do, Republic of Korea
| | - Seoyoung Choi
- R&D Center, ArborMed Company Ltd, Pangyo, Seongnam, Gyeonggi-do, Republic of Korea
| | - Dasol Kim
- R&D Center, ArborMed Company Ltd, Pangyo, Seongnam, Gyeonggi-do, Republic of Korea
| | - Chunwon Jung
- R&D Center, ArborMed Company Ltd, Pangyo, Seongnam, Gyeonggi-do, Republic of Korea
| | - Hongjae Lee
- R&D Center, ArborMed Company Ltd, Pangyo, Seongnam, Gyeonggi-do, Republic of Korea
| | - Dongsik Park
- R&D Center, ArborMed Company Ltd, Pangyo, Seongnam, Gyeonggi-do, Republic of Korea
| | - Weonbin Im
- R&D Center, ArborMed Company Ltd, Pangyo, Seongnam, Gyeonggi-do, Republic of Korea
| | - So-Young Eun
- R&D Center, ArborMed Company Ltd, Pangyo, Seongnam, Gyeonggi-do, Republic of Korea
| | - You-Hee Cho
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
| | - Jeremy D Semrau
- Department of Civil and Environmental Engineering, University of Michigan, Ann Arbor, Michigan
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Simon Hohenester
- Department of Medicine II, Ludwig Maximilian University Munich, Munich, Germany
| | | | - Alan A DiSpirito
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa
| | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany; Institute of Toxicology and Environmental Hygiene, School of Medicine, Technical University Munich, Munich, Germany.
| |
Collapse
|
16
|
Huang Z, Yang J, Chen D, Zhou X, Xiao X, Wang J, Wang M, Zhao J, Chu J. Metal deposits associated with brain atrophy in the deep gray matter nucleus in Wilson's disease. Cereb Cortex 2023:bhad182. [PMID: 37365842 DOI: 10.1093/cercor/bhad182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/03/2023] [Indexed: 06/28/2023] Open
Abstract
Regional atrophy and metal deposition are typical manifestations in Wilson's disease, but their relationship has not been systematically investigated. We aim to investigate the association of regional brain atrophy and metal deposition in the deep gray matter nucleus at MRI in Wilson's disease. We acquired the structural and susceptibility mapping and performed a cross-sectional comparison of volume and susceptibility in deep gray matter nucleus. The most extensive and severe atrophy was detected in brain regions in neuro-Wilson's disease, as well as the most widespread and heaviest metal deposits. Metal deposits were significantly negatively correlated with volume in the bilateral thalamus, caudate, and putamen. None of correlation was found between the clinical score with volume or susceptibility in the focused regions. In the 1-year follow-up analysis, the volume of right thalamus, globus pallidus, and brainstem and the susceptibility of the left caudate have decreased significantly as the symptom improvement. In Wilson's disease, phenotypes have varied scope and extend of volumetric atrophy and metal deposits. This study is expected to take the lead in revealing that in neuro-Wilson's disease, greater regional atrophy associated with heavier metal deposits in Wilson's disease. Moreover, after 1-year treatment, the imaging data have changed as the patient's condition improvement.
Collapse
Affiliation(s)
- Zihuan Huang
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| | - Jie Yang
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 518000, Guangdong Province, China
| | - Dingbang Chen
- Department of Neurology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| | - Xiangxue Zhou
- Department of Neurology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| | - Xia Xiao
- Department of Neurology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| | - Junqiao Wang
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| | - Mengzhu Wang
- Department of MR Scientific Marketing, Siemens Healthineers, Guangzhou 510120, Guangdong Province, China
| | - Jing Zhao
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| | - Jianping Chu
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
| |
Collapse
|
17
|
Sajjad H, Sajjad A, Haya RT, Khan MM, Zia M. Copper oxide nanoparticles: In vitro and in vivo toxicity, mechanisms of action and factors influencing their toxicology. Comp Biochem Physiol C Toxicol Pharmacol 2023; 271:109682. [PMID: 37328134 DOI: 10.1016/j.cbpc.2023.109682] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/21/2023] [Accepted: 06/11/2023] [Indexed: 06/18/2023]
Abstract
Copper oxide nanoparticles (CuO NPs) have received increasing interest due to their distinctive properties, including small particle size, high surface area, and reactivity. Due to these properties, their applications have been expanded rapidly in various areas such as biomedical properties, industrial catalysts, gas sensors, electronic materials, and environmental remediation. However, because of these widespread uses, there is now an increased risk of human exposure, which could lead to short- and long-term toxicity. This review addresses the underlying toxicity mechanisms of CuO NPs in cells which include reactive oxygen species generation, leaching of Cu ion, coordination effects, non-homeostasis effect, autophagy, and inflammation. In addition, different key factors responsible for toxicity, characterization, surface modification, dissolution, NPs dose, exposure pathways and environment are discussed to understand the toxicological impact of CuO NPs. In vitro and in vivo studies have shown that CuO NPs cause oxidative stress, cytotoxicity, genotoxicity, immunotoxicity, neurotoxicity, and inflammation in bacterial, algal, fish, rodents, and human cell lines. Therefore, to make CuO NPs a more suitable candidate for various applications, it is essential to address their potential toxic effects, and hence, more studies should be done on the long-term and chronic impacts of CuO NPs at different concentrations to assure the safe usage of CuO NPs.
Collapse
Affiliation(s)
- Humna Sajjad
- Department of Biotechnology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Anila Sajjad
- Department of Biotechnology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Rida Tul Haya
- Department of Biotechnology, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | | | - Muhammad Zia
- Department of Biotechnology, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|
18
|
Raschke S, Bornhorst J, Schwerdtle T. Se supplementation to an in vitro blood-brain barrier does not affect Cu transfer into the brain. J Trace Elem Med Biol 2023; 78:127180. [PMID: 37201367 DOI: 10.1016/j.jtemb.2023.127180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/26/2023] [Accepted: 04/26/2023] [Indexed: 05/20/2023]
Abstract
BACKGROUND Dyshomeostasis of copper (Cu) accompanied by Cu accumulation in certain brain areas has been associated with neurodegenerative diseases. One proposed toxic mode of action following Cu overload is oxidative stress associated with neuronal damage, whereas Selenium (Se) is assumed to play here a protective role. This study investigates the relationship between adequate Se supplementation and the respective consequences for Cu transfer into the brain applying an in vitro model of the blood-brain barrier (BBB). METHODS Primary porcine brain capillary endothelial cells (PBCECs) seeded on Transwell® inserts were supplemented with selenite starting at cultivation in both compartments. After apical application of 15 or 50 µM CuSO4, transfer of Cu to the basolateral compartment, the brain facing side, was assessed by ICP-MS/MS. RESULTS Incubation with Cu did not negatively affect the barrier properties, whereas Se had a positive effect. Additionally, Se status improved after selenite supplementation. Transfer of Cu was not affected by selenite supplementation. Under Se-deficient conditions, Cu permeability coefficients decreased with increasing Cu concentrations. CONCLUSION The results of this study do not indicate that under suboptimal Se supplementation more Cu transfers across the BBB to the brain.
Collapse
Affiliation(s)
- Stefanie Raschke
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Julia Bornhorst
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstraße 20, 42119 Wuppertal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin, Germany
| | - Tanja Schwerdtle
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin, Germany; German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany.
| |
Collapse
|
19
|
Vielee ST, Wise JP. Among Gerontogens, Heavy Metals Are a Class of Their Own: A Review of the Evidence for Cellular Senescence. Brain Sci 2023; 13:500. [PMID: 36979310 PMCID: PMC10046019 DOI: 10.3390/brainsci13030500] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Advancements in modern medicine have improved the quality of life across the globe and increased the average lifespan of our population by multiple decades. Current estimates predict by 2030, 12% of the global population will reach a geriatric age and live another 3-4 decades. This swelling geriatric population will place critical stress on healthcare infrastructures due to accompanying increases in age-related diseases and comorbidities. While much research focused on long-lived individuals seeks to answer questions regarding how to age healthier, there is a deficit in research investigating what aspects of our lives accelerate or exacerbate aging. In particular, heavy metals are recognized as a significant threat to human health with links to a plethora of age-related diseases, and have widespread human exposures from occupational, medical, or environmental settings. We believe heavy metals ought to be classified as a class of gerontogens (i.e., chemicals that accelerate biological aging in cells and tissues). Gerontogens may be best studied through their effects on the "Hallmarks of Aging", nine physiological hallmarks demonstrated to occur in aged cells, tissues, and bodies. Evidence suggests that cellular senescence-a permanent growth arrest in cells-is one of the most pertinent hallmarks of aging and is a useful indicator of aging in tissues. Here, we discuss the roles of heavy metals in brain aging. We briefly discuss brain aging in general, then expand upon observations for heavy metals contributing to age-related neurodegenerative disorders. We particularly emphasize the roles and observations of cellular senescence in neurodegenerative diseases. Finally, we discuss the observations for heavy metals inducing cellular senescence. The glaring lack of knowledge about gerontogens and gerontogenic mechanisms necessitates greater research in the field, especially in the context of the global aging crisis.
Collapse
Affiliation(s)
- Samuel T. Vielee
- Pediatrics Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - John P. Wise
- Pediatrics Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
20
|
Diao SP, Zhuang YS, Huang YQ, Zhou ZH, Liu AQ, Hong MF. Analysis of risk factors for neurological symptoms in patients with purely hepatic Wilson's disease at diagnosis. BMC Neurol 2023; 23:89. [PMID: 36855079 PMCID: PMC9972690 DOI: 10.1186/s12883-023-03105-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/02/2023] [Indexed: 03/02/2023] Open
Abstract
OBJECTIVE To analyze and explore the risk factors for neurological symptoms in patients with purely hepatic Wilson's disease (WD) at diagnosis. METHODS This retrospective study was conducted at the First Affiliated Hospital of the Guangdong Pharmaceutical University on 68 patients with purely hepatic WD aged 20.6 ± 7.2 years. The physical examinations, laboratory tests, color Doppler ultrasound of the liver and spleen, and magnetic resonance imaging (MRI) of the brain were performed. RESULTS The elevated alanine transaminase (ALT) and aspartate transaminase (AST) levels and 24-h urinary copper level were higher in the purely hepatic WD who developed neurological symptoms (NH-WD) group than those in the purely hepatic WD (H-WD) group. Adherence to low-copper diet, and daily oral doses of penicillamine (PCA) and zinc gluconate (ZG) were lower in the NH-WD group than those in the H-WD group. Logistic regression analysis showed that insufficient doses of PCA and ZG were associated with the development of neurological symptoms in patients with purely hepatic WD at diagnosis. CONCLUSION The development of neurological symptoms in patients with purely hepatic WD was closely associated with insufficient doses of PCA and ZG, and the inferior efficacy of copper-chelating agents. During the course of anti-copper treatment, the patient's medical status and the efficacy of copper excretion should be closely monitored.
Collapse
Affiliation(s)
- Sheng-Peng Diao
- grid.412601.00000 0004 1760 3828Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, 610630 Guangdong China ,grid.477976.c0000 0004 1758 4014Department of Neurology, College of Clinical Medicine, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, 510062 Guangdong China
| | - Yang-Sha Zhuang
- grid.477976.c0000 0004 1758 4014Department of Neurology, College of Clinical Medicine, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, 510062 Guangdong China
| | - Ye-Qing Huang
- grid.477976.c0000 0004 1758 4014Department of Neurology, College of Clinical Medicine, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, 510062 Guangdong China
| | - Zhi-Hua Zhou
- grid.477976.c0000 0004 1758 4014Department of Neurology, College of Clinical Medicine, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, 510062 Guangdong China
| | - Ai-Qun Liu
- grid.477976.c0000 0004 1758 4014Department of Neurology, College of Clinical Medicine, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, 510062 Guangdong China
| | - Ming-Fan Hong
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, 610630, Guangdong, China. .,Department of Neurology, College of Clinical Medicine, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, 510062, Guangdong, China.
| |
Collapse
|
21
|
Maryam T, Rana NF, Alshahrani SM, Batool F, Fatima M, Tanweer T, Alrdahe SS, Alanazi YF, Alsharif I, Alaryani FS, Kashif AS, Menaa F. Silymarin Encapsulated Liposomal Formulation: An Effective Treatment Modality against Copper Toxicity Associated Liver Dysfunction and Neurobehavioral Abnormalities in Wistar Rats. Molecules 2023; 28:molecules28031514. [PMID: 36771180 PMCID: PMC9920678 DOI: 10.3390/molecules28031514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 02/09/2023] Open
Abstract
Wilson's disease causes copper accumulation in the liver and extrahepatic organs. The available therapies aim to lower copper levels by various means. However, a potent drug that can repair the damaged liver and brain tissue is needed. Silymarin has hepatoprotective, antioxidant, and cytoprotective properties. However, poor oral bioavailability reduces its efficacy. In this study, a "thin film hydration method" was used for synthesizing silymarin-encapsulated liposome nanoparticles (SLNPs) and evaluated them against copper toxicity, associated liver dysfunction and neurobehavioral abnormalities in Wistar rats. After copper toxicity induction, serological and behavioral assays were conducted to evaluate treatment approaches. Histological examination of the diseased rats revealed severe hepatocyte necrosis and neuronal vacuolation. These cellular degenerations were mild in rats treated with SLNPs and a combination of zinc and SLNPs (ZSLNPs). SLNPs also decreased liver enzymes and enhanced rats' spatial memory significantly (p = 0.006) in the diseased rats. During forced swim tests, SLNPs treated rats exhibited a 60-s reduction in the immobility period, indicating reduced depression. ZSLNPs were significantly more effective than traditional zinc therapy in decreasing the immobility period (p = 0.0008) and reducing liver enzymes, but not in improving spatial memory. Overall, SLNPs enhanced oral silymarin administration and managed copper toxicity symptoms.
Collapse
Affiliation(s)
- Tuba Maryam
- School of Mechanical & Manufacturing Engineering (SMME), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Nosheen Fatima Rana
- School of Mechanical & Manufacturing Engineering (SMME), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
- Correspondence: (N.F.R.); (F.M.)
| | - Sultan M. Alshahrani
- Clinical Pharmacy Department, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia
| | - Farhat Batool
- School of Mechanical & Manufacturing Engineering (SMME), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Misha Fatima
- School of Mechanical & Manufacturing Engineering (SMME), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Tahreem Tanweer
- School of Mechanical & Manufacturing Engineering (SMME), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Salma Saleh Alrdahe
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Yasmene F. Alanazi
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Ifat Alsharif
- Department of Biology, Jamoum University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Fatima S. Alaryani
- Department of Biology, College of Science, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Amer Sohail Kashif
- School of Mechanical & Manufacturing Engineering (SMME), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Farid Menaa
- Departments of Internal Medicine and Nanomedicine, California Innovations Corporation, 9, San Diego, CA 92037, USA
- Correspondence: (N.F.R.); (F.M.)
| |
Collapse
|
22
|
Jing XZ, Li GY, Wu YP, Yuan XZ, Luo XG, Chen JL, Taximaimaiti R, Wang XP, Li JQ. Free water imaging as a novel biomarker in Wilson's disease: A cross-sectional study. Parkinsonism Relat Disord 2023; 106:105234. [PMID: 36481719 DOI: 10.1016/j.parkreldis.2022.105234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND The bi-tensor free water imaging may provide more specific information in detecting microstructural brain tissue alterations than conventional single tensor diffusion tensor imaging. The study aimed to investigate microstructural changes in deep gray matter (DGM) nuclei of Wilson's disease (WD) using a bi-tensor free water imaging and whether the findings correlate with the neurological impairment in WD patients. METHODS The study included 29 WD patients and 25 controls. Free water and free water corrected fractional anisotropy (FAT) in DGM nuclei of WD patients were calculated. The correlations of free water and FAT with the Unified WD Rating Scale (UWDRS) neurological subscale of WD patients were performed. RESULTS Free water and FAT values were significantly increased in multiple DGM nuclei of neurological WD patients compared to controls. WD patients with normal appearing on conventional MRI also had significantly higher free water and FAT values in multiple DGM nuclei than controls. Positive correlations were noted between the UWDRS neurological subscores and free water values of the putamen and pontine tegmentum as well as FAT values of the dentate nucleus, red nucleus, and globus pallidus. In addition, the measured free water and FAT values of specific structures also showed a positive correlation with specific clinical symptoms in neurological WD patients, such as dysarthria, parkinsonian signs, tremor, dystonia, and ataxia. CONCLUSIONS Free water imaging detects microstructural changes in both normal and abnormal appearing DGM nuclei of WD patients. Free water imaging indices were correlated with the severity of neurological impairment in WD patients.
Collapse
Affiliation(s)
- Xiao-Zhong Jing
- Department of Neurology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Neurology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Gai-Ying Li
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China.
| | - Yu-Peng Wu
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China.
| | - Xiang-Zhen Yuan
- Department of Neurology, Weifang People's Hospital, Weifang, Shandong, China.
| | - Xing-Guang Luo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
| | - Jia-Lin Chen
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China.
| | - Reyisha Taximaimaiti
- Department of Neurology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiao-Ping Wang
- Department of Neurology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Neurology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jian-Qi Li
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China.
| |
Collapse
|
23
|
Avan A, Członkowska A, Gaskin S, Granzotto A, Sensi SL, Hoogenraad TU. The Role of Zinc in the Treatment of Wilson’s Disease. Int J Mol Sci 2022; 23:ijms23169316. [PMID: 36012580 PMCID: PMC9409413 DOI: 10.3390/ijms23169316] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/09/2022] [Accepted: 08/15/2022] [Indexed: 02/06/2023] Open
Abstract
Wilson’s disease (WD) is a hereditary disorder of copper metabolism, producing abnormally high levels of non-ceruloplasmin-bound copper, the determinant of the pathogenic process causing brain and hepatic damage and dysfunction. Although the disease is invariably fatal without medication, it is treatable and many of its adverse effects are reversible. Diagnosis is difficult due to the large range and severity of symptoms. A high index of suspicion is required as patients may have only a few of the many possible biomarkers. The genetic prevalence of ATP7B variants indicates higher rates in the population than are currently diagnosed. Treatments have evolved from chelators that reduce stored copper to zinc, which reduces the toxic levels of circulating non-ceruloplasmin-bound copper. Zinc induces intestinal metallothionein, which blocks copper absorption and increases excretion in the stools, resulting in an improvement in symptoms. Two meta-analyses and several large retrospective studies indicate that zinc is equally effective as chelators for the treatment of WD, with the advantages of a very low level of toxicity and only the minor side effect of gastric disturbance. Zinc is recommended as a first-line treatment for neurological presentations and is gaining acceptance for hepatic presentations. It is universally recommended for lifelong maintenance therapy and for presymptomatic WD.
Collapse
Affiliation(s)
- Abolfazl Avan
- Department of Public Health, School of Medicine, Mashhad University of Medical Sciences, Mashhad 93518-88415, Iran
- Correspondence:
| | - Anna Członkowska
- 2nd Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | - Susan Gaskin
- Department of Civil Engineering, McGill University, Montreal, QC H3A 0C3, Canada
| | - Alberto Granzotto
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences (DNISC), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Sue and Bill Gross Stem Cell Research Center, University of California-Irvine, Irvine, CA 92697, USA
| | - Stefano L. Sensi
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences (DNISC), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Tjaard U. Hoogenraad
- Department of Neurology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
24
|
Shribman S, Burrows M, Convery R, Bocchetta M, Sudre CH, Acosta-Cabronero J, Thomas DL, Gillett GT, Tsochatzis EA, Bandmann O, Rohrer JD, Warner TT. Neuroimaging Correlates of Cognitive Deficits in Wilson's Disease. Mov Disord 2022; 37:1728-1738. [PMID: 35723521 PMCID: PMC9542291 DOI: 10.1002/mds.29123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Cognitive impairment is common in neurological presentations of Wilson's disease (WD). Various domains can be affected, and subclinical deficits have been reported in patients with hepatic presentations. Associations with imaging abnormalities have not been systematically tested. OBJECTIVE The aim was to determine the neuroanatomical basis for cognitive deficits in WD. METHODS We performed a 16-item neuropsychological test battery and magnetic resonance brain imaging in 40 patients with WD. The scores for each test were compared between patients with neurological and hepatic presentations and with normative data. Associations with Unified Wilson's Disease Rating Scale neurological examination subscores were examined. Quantitative, whole-brain, multimodal imaging analyses were used to identify associations with neuroimaging abnormalities in chronically treated stable patients. RESULTS Abstract reasoning, executive function, processing speed, calculation, and visuospatial function scores were lower in patients with neurological presentations than in those with hepatic presentations and correlated with neurological examination subscores. Deficits in abstract reasoning and phonemic fluency were associated with lower putamen volumes even after controlling for neurological severity. About half of patients with hepatic presentations had poor performance in memory for faces, cognitive flexibility, or associative learning relative to normative data. These deficits were associated with widespread cortical atrophy and/or white matter diffusion abnormalities. CONCLUSIONS Subtle cognitive deficits in patients with seemingly hepatic presentations represent a distinct neurological phenotype associated with diffuse cortical and white matter pathology. This may precede the classical neurological phenotype characterized by movement disorders and executive dysfunction and be associated with basal ganglia damage. A binary phenotypic classification for WD may no longer be appropriate. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Samuel Shribman
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London
| | - Maggie Burrows
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London
| | - Rhian Convery
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Martina Bocchetta
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Carole H Sudre
- MRC Unit for Lifelong Health and Ageing, University College London, London, United Kingdom.,Centre for Medical Image Computing, University College London, London, United Kingdom.,Biomedical Engineering & Imaging Sciences, King's College London, London, United Kingdom
| | | | - David L Thomas
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom.,Neuroradiological Academic Unit, UCL Queen Square Institute of Neurology, London, United Kingdom.,Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Godfrey T Gillett
- Department of Clinical Chemistry, Northern General Hospital, Sheffield, United Kingdom
| | - Emmanuel A Tsochatzis
- UCL Institute of Liver and Digestive Health and Royal Free Hospital, London, United Kingdom
| | - Oliver Bandmann
- Sheffield Institute of Translational Neuroscience, Sheffield, United Kingdom
| | - Jonathan D Rohrer
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Thomas T Warner
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London
| |
Collapse
|
25
|
Jing XZ, Yuan XZ, Li GY, Chen JL, Wu R, Yang LL, Zhang SY, Wang XP, Li JQ. Increased Magnetic Susceptibility in the Deep Gray Matter Nuclei of Wilson's Disease: Have We Been Ignoring Atrophy? Front Neurosci 2022; 16:794375. [PMID: 35720701 PMCID: PMC9198485 DOI: 10.3389/fnins.2022.794375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 05/04/2022] [Indexed: 11/25/2022] Open
Abstract
Background Histopathological studies in Wilson's disease (WD) have revealed increased copper and iron concentrations in the deep gray matter nuclei. However, the commonly used mean bulk susceptibility only reflects the regional metal concentration rather than the total metal content, and regional atrophy may affect the assessment of mean bulk susceptibility. Our study aimed to quantitatively assess the changes of metal concentration and total metal content in deep gray matter nuclei by quantitative susceptibility mapping to distinguish patients with neurological and hepatic WD from healthy controls. Methods Quantitative susceptibility maps were obtained from 20 patients with neurological WD, 10 patients with hepatic WD, and 25 healthy controls on a 3T magnetic resonance imaging system. Mean bulk susceptibility, volumes, and total susceptibility of deep gray matter nuclei in different groups were compared using a linear regression model. The area under the curve (AUC) was calculated by receiver characteristic curve to analyze the diagnostic capability of mean bulk susceptibility and total susceptibility. Results Mean bulk susceptibility and total susceptibility of multiple deep gray matter nuclei in patients with WD were higher than those in healthy controls. Compared with patients with hepatic WD, patients with neurological WD had higher mean bulk susceptibility but similar total susceptibility in the head of the caudate nuclei, globus pallidus, and putamen. Mean bulk susceptibility of putamen demonstrated the best diagnostic capability for patients with neurological WD, the AUC was 1, and the sensitivity and specificity were all equal to 1. Total susceptibility of pontine tegmentum was most significant for the diagnosis of patients with hepatic WD, the AUC was 0.848, and the sensitivity and specificity were 0.7 and 0.96, respectively. Conclusion Brain atrophy may affect the assessment of mean bulk susceptibility in the deep gray matter nuclei of patients with WD, and total susceptibility should be an additional metric for total metal content assessment. Mean bulk susceptibility and total susceptibility of deep gray matter nuclei may be helpful for the early diagnosis of WD.
Collapse
Affiliation(s)
- Xiao-Zhong Jing
- Department of Neurology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang-Zhen Yuan
- Department of Neurology, Weifang People's Hospital, Weifang, China
| | - Gai-Ying Li
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Jia-Lin Chen
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Rong Wu
- Department of Neurology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling-Li Yang
- Department of Neurology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu-Yun Zhang
- Department of Neurology, Weifang People's Hospital, Weifang, China
| | - Xiao-Ping Wang
- Department of Neurology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Neurology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Qi Li
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| |
Collapse
|
26
|
Shribman S, Bocchetta M, Sudre CH, Acosta-Cabronero J, Burrows M, Cook P, Thomas DL, Gillett GT, Tsochatzis EA, Bandmann O, Rohrer JD, Warner TT. Neuroimaging correlates of brain injury in Wilson's disease: a multimodal, whole-brain MRI study. Brain 2022; 145:263-275. [PMID: 34289020 PMCID: PMC8967100 DOI: 10.1093/brain/awab274] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/25/2021] [Accepted: 07/04/2021] [Indexed: 11/23/2022] Open
Abstract
Wilson's disease is an autosomal-recessive disorder of copper metabolism with neurological and hepatic presentations. Chelation therapy is used to 'de-copper' patients but neurological outcomes remain unpredictable. A range of neuroimaging abnormalities have been described and may provide insights into disease mechanisms, in addition to prognostic and monitoring biomarkers. Previous quantitative MRI analyses have focused on specific sequences or regions of interest, often stratifying chronically treated patients according to persisting symptoms as opposed to initial presentation. In this cross-sectional study, we performed a combination of unbiased, whole-brain analyses on T1-weighted, fluid-attenuated inversion recovery, diffusion-weighted and susceptibility-weighted imaging data from 40 prospectively recruited patients with Wilson's disease (age range 16-68). We compared patients with neurological (n = 23) and hepatic (n = 17) presentations to determine the neuroradiological sequelae of the initial brain injury. We also subcategorized patients according to recent neurological status, classifying those with neurological presentations or deterioration in the preceding 6 months as having 'active' disease. This allowed us to compare patients with active (n = 5) and stable (n = 35) disease and identify imaging correlates for persistent neurological deficits and copper indices in chronically treated, stable patients. Using a combination of voxel-based morphometry and region-of-interest volumetric analyses, we demonstrate that grey matter volumes are lower in the basal ganglia, thalamus, brainstem, cerebellum, anterior insula and orbitofrontal cortex when comparing patients with neurological and hepatic presentations. In chronically treated, stable patients, the severity of neurological deficits correlated with grey matter volumes in similar, predominantly subcortical regions. In contrast, the severity of neurological deficits did not correlate with the volume of white matter hyperintensities, calculated using an automated lesion segmentation algorithm. Using tract-based spatial statistics, increasing neurological severity in chronically treated patients was associated with decreasing axial diffusivity in white matter tracts whereas increasing serum non-caeruloplasmin-bound ('free') copper and active disease were associated with distinct patterns of increasing mean, axial and radial diffusivity. Whole-brain quantitative susceptibility mapping identified increased iron deposition in the putamen, cingulate and medial frontal cortices of patients with neurological presentations relative to those with hepatic presentations and neurological severity was associated with iron deposition in widespread cortical regions in chronically treated patients. Our data indicate that composite measures of subcortical atrophy provide useful prognostic biomarkers, whereas abnormal mean, axial and radial diffusivity are promising monitoring biomarkers. Finally, deposition of brain iron in response to copper accumulation may directly contribute to neurodegeneration in Wilson's disease.
Collapse
Affiliation(s)
- Samuel Shribman
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
| | - Martina Bocchetta
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3AR, UK
| | - Carole H Sudre
- MRC Unit for Lifelong Health and Ageing, University College London, London WC1E 7HB, UK
- Centre for Medical Image Computing, University College London, London WC1V 6LJ, UK
- Biomedical Engineering and Imaging Sciences, King’s College London, London WC2R 2LS, UK
| | | | - Maggie Burrows
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
| | - Paul Cook
- Department of Clinical Biochemistry, Southampton General Hospital, Southampton SO16 6YD, UK
| | - David L Thomas
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3AR, UK
- Neuroradiological Academic Unit, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, London WC1N 3AR, UK
| | - Godfrey T Gillett
- Department of Clinical Chemistry, Northern General Hospital, Sheffield S5 7AU, UK
| | - Emmanuel A Tsochatzis
- UCL Institute of Liver and Digestive Health and Royal Free Hospital, London NW3 2PF, UK
| | - Oliver Bandmann
- Sheffield Institute of Translational Neuroscience, Sheffield S10 2HQ, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London WC1N 3AR, UK
| | - Thomas T Warner
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
| |
Collapse
|
27
|
Borchard S, Raschke S, Zak KM, Eberhagen C, Einer C, Weber E, Müller SM, Michalke B, Lichtmannegger J, Wieser A, Rieder T, Popowicz GM, Adamski J, Klingenspor M, Coles AH, Viana R, Vendelbo MH, Sandahl TD, Schwerdtle T, Plitz T, Zischka H. Bis-choline tetrathiomolybdate prevents copper-induced blood-brain barrier damage. Life Sci Alliance 2021; 5:5/3/e202101164. [PMID: 34857647 PMCID: PMC8675913 DOI: 10.26508/lsa.202101164] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 11/24/2022] Open
Abstract
The blood–brain barrier endothelial cell monolayer becomes permeable to elevated copper loosely bound to albumin, which can be avoided by a high-affinity copper chelator but not by D-penicillamine. In Wilson disease, excessive copper accumulates in patients’ livers and may, upon serum leakage, severely affect the brain according to current viewpoints. Present remedies aim at avoiding copper toxicity by chelation, for example, by D-penicillamine (DPA) or bis-choline tetrathiomolybdate (ALXN1840), the latter with a very high copper affinity. Hence, ALXN1840 may potentially avoid neurological deterioration that frequently occurs upon DPA treatment. As the etiology of such worsening is unclear, we reasoned that copper loosely bound to albumin, that is, mimicking a potential liver copper leakage into blood, may damage cells that constitute the blood-brain barrier, which was found to be the case in an in vitro model using primary porcine brain capillary endothelial cells. Such blood–brain barrier damage was avoided by ALXN1840, plausibly due to firm protein embedding of the chelator bound copper, but not by DPA. Mitochondrial protection was observed, a prerequisite for blood–brain barrier integrity. Thus, high-affinity copper chelators may minimize such deterioration in the treatment of neurologic Wilson disease.
Collapse
Affiliation(s)
- Sabine Borchard
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Stefanie Raschke
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.,TraceAge-Deutsche Forschungsgemeinschaft Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (Forschungsgruppe 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
| | - Krzysztof M Zak
- Institute of Structural Biology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Carola Eberhagen
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Claudia Einer
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Elisabeth Weber
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Sandra M Müller
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Bernhard Michalke
- Research Unit Analytical BioGeoChemistry, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Josef Lichtmannegger
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Albrecht Wieser
- Institute of Radiation Medicine, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Tamara Rieder
- Technical University Munich, School of Medicine, Institute of Toxicology and Environmental Hygiene, Munich, Germany
| | - Grzegorz M Popowicz
- Institute of Structural Biology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Jerzy Adamski
- Research Unit Molecular Endocrinology and Metabolism, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.,Lehrstuhl für Experimentelle Genetik, Technical University Munich, Freising-Weihenstephan, Germany.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, Technical University of Munich, School of Life Sciences Weihenstephan, Freising, Germany.,Else-Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
| | | | - Ruth Viana
- Alexion AstraZeneca Rare Disease, Boston, MA, USA
| | - Mikkel H Vendelbo
- Department of Nuclear Medicine and Positron Emission Tomography Centre, Aarhus University Hospital, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Thomas D Sandahl
- Medical Department Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Tanja Schwerdtle
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.,TraceAge-Deutsche Forschungsgemeinschaft Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (Forschungsgruppe 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
| | | | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany .,Technical University Munich, School of Medicine, Institute of Toxicology and Environmental Hygiene, Munich, Germany
| |
Collapse
|
28
|
Osterode W, Falkenberg G, Wrba F. Copper and Trace Elements in Gallbladder form Patients with Wilson's Disease Imaged and Determined by Synchrotron X-ray Fluorescence. J Imaging 2021; 7:jimaging7120261. [PMID: 34940728 PMCID: PMC8705686 DOI: 10.3390/jimaging7120261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 12/16/2022] Open
Abstract
Investigations about suspected tissue alterations and the role of gallbladder in Wilson’s disease (WD)—an inherited genetic disease with impaired copper metabolism—are rare. Therefore, tissue from patients with genetically characterised WD was investigated by microscopic synchrotron X-ray fluorescence (µSRXRF). For two-dimensional imaging and quantification of elements, X-ray spectra were peak-fitted, and the net peak intensities were normalised to the intensity of the incoming monochromatic beam intensity. Concentrations were calculated by fundamental parameter-based program quant and external standardisation. Copper (Cu), zinc (Zn) and iron (Fe) along with sulphur (S) and phosphorus (P) mappings could be demonstrated in a near histological resolution. All these elements were increased compared to gallbladder tissue from controls. Cu and Zn and Fe in WD-GB were mostly found to be enhanced in the epithelium. We documented a significant linear relationship with Cu, Zn and sulphur. Concentrations of Cu/Zn were roughly 1:1 while S/Cu was about 100:1, depending on the selected areas for investigation. The significant linear relationship with Cu, Zn and sulphur let us assume that metallothioneins, which are sulphur-rich proteins, are increased too. Our data let us suggest that the WD gallbladder is the first in the gastrointestinal tract to reabsorb metals to prevent oxidative damage caused by metal toxicity.
Collapse
Affiliation(s)
- Wolf Osterode
- Universitätsklinik für Innere Medizin II, Medical University of Vienna, A-1090 Vienna, Austria
- Correspondence:
| | - Gerald Falkenberg
- Deutsches Elektronen-Synchrotron (DESY), Photon Science, D-22603 Hamburg, Germany;
| | - Fritz Wrba
- Klinisches Institut für Klinische Pathologie, Medical University of Vienna, A-1090 Vienna, Austria;
| |
Collapse
|
29
|
Blink reflex in newly diagnosed and treated patients with Wilson's disease. J Neural Transm (Vienna) 2021; 128:1873-1880. [PMID: 34669020 PMCID: PMC8571127 DOI: 10.1007/s00702-021-02432-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/14/2021] [Indexed: 01/02/2023]
Abstract
Abnormal blink reflex (BR) results mainly from the dysfunction of reticular brainstem pathways and is one of the features of degenerative brain disorders. We aimed to investigate whether patients with Wilson’s disease (WD) have abnormal BR. This was a prospective, observational, single-center study. BR was assessed in accordance with generally accepted standards in 44 newly diagnosed treatment-naïve and 66 treated patients with WD. Any abnormal parameters in BR were observed in 45.5% treatment-naïve patients and 37.9% treated patients (p = 0.429). We also did not observe significant differences in BR parameters and frequency of abnormal findings between treated and treatment naïve patients. Abnormal findings in any of the BR parameters were more frequent in patients with neurological vs. non-neurological presentation (57.5 vs. 28.6%, p = 0.002), present vs. absent Kayser–Fleischer ring (73 vs. 21.5%, p < 0.001), and typical vs. no typical WD abnormalities in brain MRI (50% vs. 24.4%, p = 0.009). In addition, longer median R1 and R2 latencies, both ipsilateral and contralateral, were significantly more frequent in neurological than non-neurological WD patients, those with Kayser–Fleischer rings, and those with abnormal MRI findings typical of WD. Our results confirm frequent BR abnormalities in WD, which may be explained by the pathological influence of copper deposits in the circuit linking the basal ganglia, cerebellum and brainstem.
Collapse
|
30
|
Squitti R, Ventriglia M, Granzotto A, Sensi SL, Rongioletti MCA. Non-Ceruloplasmin Copper as a Stratification Biomarker of Alzheimer's Disease Patients: How to Measure and Use It. Curr Alzheimer Res 2021; 18:533-545. [PMID: 34674622 DOI: 10.2174/1567205018666211022085755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/19/2021] [Accepted: 06/30/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a type of dementia very common in the elderly. A growing body of recent evidence has linked AD pathogenesis to copper (Cu) dysmetabolism in the body. In fact, a subset of patients affected either by AD or by its prodromal form known as Mild Cognitive Impairment (MCI) have been observed to be unable to maintain a proper balance of Cu metabolism and distribution and are characterized by the presence in their serum of increased levels of Cu not bound to ceruloplasmin (non-ceruloplasmin Cu). Since serum non-ceruloplasmin Cu is a biomark- er of Wilson's disease (WD), a well-known condition of Cu-driven toxicosis, in this review, we pro- pose that in close analogy with WD, the assessment of non-ceruloplasmin Cu levels can be exploit- ed as a cost-effective stratification and susceptibility/risk biomarker for the identification of some AD/MCI individuals. The approach can also be used as an eligibility criterion for clinical trials aim- ing at investigating Cu-related interventions against AD/MCI.
Collapse
Affiliation(s)
- Rosanna Squitti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia. Italy
| | - Mariacarla Ventriglia
- Fatebenefratelli Foundation for Health Research and Education, AFaR Division, San Giovanni Calibita Fatebene-fratelli Hospital, Isola Tiberina, Rome. Italy
| | - Alberto Granzotto
- Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti. Italy
| | - Stefano L Sensi
- Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, Chieti. Italy
| | - Mauro Ciro Antonio Rongioletti
- Department of Laboratory Medicine, Research and Development Division, San Giovanni Calibita Fatebenefratelli Hospital, Isola Tiberina, Rome. Italy
| |
Collapse
|
31
|
Shribman S, Poujois A, Bandmann O, Czlonkowska A, Warner TT. Wilson's disease: update on pathogenesis, biomarkers and treatments. J Neurol Neurosurg Psychiatry 2021; 92:1053-1061. [PMID: 34341141 DOI: 10.1136/jnnp-2021-326123] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/08/2021] [Indexed: 12/22/2022]
Abstract
Wilson's disease is an autosomal-recessive disorder of copper metabolism caused by mutations in ATP7B and associated with neurological, psychiatric, ophthalmological and hepatic manifestations. Decoppering treatments are used to prevent disease progression and reduce symptoms, but neurological outcomes remain mixed. In this article, we review the current understanding of pathogenesis, biomarkers and treatments for Wilson's disease from the neurological perspective, with a focus on recent advances. The genetic and molecular mechanisms associated with ATP7B dysfunction have been well characterised, but despite extensive efforts to identify genotype-phenotype correlations, the reason why only some patients develop neurological or psychiatric features remains unclear. We discuss pathological processes through which copper accumulation leads to neurodegeneration, such as mitochondrial dysfunction, the role of brain iron metabolism and the broader concept of selective neuronal vulnerability in Wilson's disease. Delayed diagnoses continue to be a major problem for patients with neurological presentations. We highlight limitations in our current approach to making a diagnosis and novel diagnostic biomarkers, including the potential for newborn screening programmes. We describe recent progress in developing imaging and wet (fluid) biomarkers for neurological involvement, including findings from quantitative MRI and other neuroimaging studies, and the development of a semiquantitative scoring system for assessing radiological severity. Finally, we cover the use of established and novel chelating agents, paradoxical neurological worsening, and progress developing targeted molecular and gene therapy for Wilson's disease, before discussing future directions for translational research.
Collapse
Affiliation(s)
- Samuel Shribman
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Aurelia Poujois
- Department of Neurology, National Reference Centre for Wilson's Disease, Rothschild Foundation Hospital, Paris, France
| | - Oliver Bandmann
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, Sheffield, UK
| | - Anna Czlonkowska
- Second Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Thomas T Warner
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
32
|
Lei C, Liao J, Li Q, Shi J, Zhang H, Guo J, Han Q, Hu L, Li Y, Pan J, Tang Z. Copper induces mitochondria-mediated apoptosis via AMPK-mTOR pathway in hypothalamus of Pigs. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 220:112395. [PMID: 34102394 DOI: 10.1016/j.ecoenv.2021.112395] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/27/2021] [Accepted: 05/30/2021] [Indexed: 06/12/2023]
Abstract
Copper (Cu), one of the heavy metals, is far beyond the carrying capacity of the environment with Cu mining, industrial wastewater discharging and the use of Cu-containing pesticides. Intaking excess Cu can cause toxic effects on liver, kidney, heart, but few studies report Cu toxicity on brain tissue. It is noteworthy that most toxicity tests are based on rodent models, but large mammals chosen as animal models has no reported. To explore the relationship of the Cu toxicity and mitochondria-mediated apoptosis on hypothalamus in pigs, the content of Cu, histomorphology, mitochondrial related indicators, apoptosis, and AMPK-mTOR signaling pathway were detected. Results showed that Cu could accumulate in hypothalamus and lead to mitochondrial dysfunction, evidenced by the decrease of ATP production, activities of respiratory chain complex I-IV, and mitochondrial respiratory function in Cu-treated groups. Additionally, the genes and proteins expression of Bax, Caspase-3, Cytc in treatment group were higher than control group. Furthermore, the protein level of p-AMPK was enhanced significantly and p-mTOR was declined, which manifested that AMPK-mTOR signaling pathway was activated in Cu-treated groups. In conclusion, this study illuminated that the accumulation of Cu could cause mitochondrial dysfunction, induce mitochondria-mediated apoptosis and activate AMPK-mTOR pathway in hypothalamus.
Collapse
Affiliation(s)
- Chaiqin Lei
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Quanwei Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Jian Shi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Jianying Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Qingyue Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Jiaqiang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| |
Collapse
|
33
|
Falcone E, Okafor M, Vitale N, Raibaut L, Sour A, Faller P. Extracellular Cu2+ pools and their detection: From current knowledge to next-generation probes. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2020.213727] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
34
|
Copper Toxicity Is Not Just Oxidative Damage: Zinc Systems and Insight from Wilson Disease. Biomedicines 2021; 9:biomedicines9030316. [PMID: 33804693 PMCID: PMC8003939 DOI: 10.3390/biomedicines9030316] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/13/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Essential metals such as copper (Cu) and zinc (Zn) are important cofactors in diverse cellular processes, while metal imbalance may impact or be altered by disease state. Cu is essential for aerobic life with significant functions in oxidation-reduction catalysis. This redox reactivity requires precise intracellular handling and molecular-to-organismal levels of homeostatic control. As the central organ of Cu homeostasis in vertebrates, the liver has long been associated with Cu storage disorders including Wilson Disease (WD) (heritable human Cu toxicosis), Idiopathic Copper Toxicosis and Endemic Tyrolean Infantile Cirrhosis. Cu imbalance is also associated with chronic liver diseases that arise from hepatitis viral infection or other liver injury. The labile redox characteristic of Cu is often discussed as a primary mechanism of Cu toxicity. However, work emerging largely from the study of WD models suggests that Cu toxicity may have specific biochemical consequences that are not directly attributable to redox activity. This work reviews Cu toxicity with a focus on the liver and proposes that Cu accumulation specifically impacts Zn-dependent processes. The prospect that Cu toxicity has specific biochemical impacts that are not entirely attributable to redox may promote further inquiry into Cu toxicity in WD and other Cu-associated disorders.
Collapse
|
35
|
Ravanfar P, Loi SM, Syeda WT, Van Rheenen TE, Bush AI, Desmond P, Cropley VL, Lane DJR, Opazo CM, Moffat BA, Velakoulis D, Pantelis C. Systematic Review: Quantitative Susceptibility Mapping (QSM) of Brain Iron Profile in Neurodegenerative Diseases. Front Neurosci 2021; 15:618435. [PMID: 33679303 PMCID: PMC7930077 DOI: 10.3389/fnins.2021.618435] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Iron has been increasingly implicated in the pathology of neurodegenerative diseases. In the past decade, development of the new magnetic resonance imaging technique, quantitative susceptibility mapping (QSM), has enabled for the more comprehensive investigation of iron distribution in the brain. The aim of this systematic review was to provide a synthesis of the findings from existing QSM studies in neurodegenerative diseases. We identified 80 records by searching MEDLINE, Embase, Scopus, and PsycInfo databases. The disorders investigated in these studies included Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Wilson's disease, Huntington's disease, Friedreich's ataxia, spinocerebellar ataxia, Fabry disease, myotonic dystrophy, pantothenate-kinase-associated neurodegeneration, and mitochondrial membrane protein-associated neurodegeneration. As a general pattern, QSM revealed increased magnetic susceptibility (suggestive of increased iron content) in the brain regions associated with the pathology of each disorder, such as the amygdala and caudate nucleus in Alzheimer's disease, the substantia nigra in Parkinson's disease, motor cortex in amyotrophic lateral sclerosis, basal ganglia in Huntington's disease, and cerebellar dentate nucleus in Friedreich's ataxia. Furthermore, the increased magnetic susceptibility correlated with disease duration and severity of clinical features in some disorders. Although the number of studies is still limited in most of the neurodegenerative diseases, the existing evidence suggests that QSM can be a promising tool in the investigation of neurodegeneration.
Collapse
Affiliation(s)
- Parsa Ravanfar
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
| | - Samantha M Loi
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia.,Neuropsychiatry, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Warda T Syeda
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia
| | - Tamsyn E Van Rheenen
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia.,Centre for Mental Health, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Patricia Desmond
- Melbourne Brain Centre Imaging Unit, Department of Medicine and Radiology, The University of Melbourne, Parkville, VIC, Australia.,Department of Radiology, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Vanessa L Cropley
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia.,Centre for Mental Health, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Carlos M Opazo
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Bradford A Moffat
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia.,Melbourne Brain Centre Imaging Unit, Department of Medicine and Radiology, The University of Melbourne, Parkville, VIC, Australia
| | - Dennis Velakoulis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia.,Neuropsychiatry, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne and Melbourne Health, Carlton South, VIC, Australia.,Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
36
|
Gromadzka G, Wierzbicka D, Litwin T, Przybyłkowski A. Iron metabolism is disturbed and anti-copper treatment improves but does not normalize iron metabolism in Wilson's disease. Biometals 2021; 34:407-414. [PMID: 33555495 PMCID: PMC7940312 DOI: 10.1007/s10534-021-00289-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 01/26/2021] [Indexed: 12/23/2022]
Abstract
Wilson's disease (WD) is a rare hereditary disorder of copper metabolism. Some data suggest that iron metabolism is disturbed in WD and this may affect the course of the disease. The current study aimed to determine whether anti-copper treatment could affect iron metabolism in WD. One hundred thirty-eight WD patients and 102 controls were examined. Serum ceruloplasmin and copper were measured by colorimetric enzyme assay or atomic adsorption spectroscopy, respectively. Routine and non-routine parameters of iron metabolism were measured by standard laboratory methods or enzyme immunoassay, respectively. WD patients, both newly diagnosed and treated, had less serum copper and ceruloplasmin than controls (90.0, 63.0, 22.0 mg/dL, respectively, p < 0.001); in the treated patients blood copper and ceruloplasmin were lower than in untreated patients (p < 0.001). Untreated patients (n = 39) had a higher median blood iron (126.0 vs 103.5 ug/dL, p < 0.05), ferritin (158.9 vs 47.5 ng/mL, p < 0.001), hepcidin (32, 6 vs 12.1 ng/mL, p < 0.001) and sTfR (0.8 vs. 0.7 ug/mL, p < 0.001) and lower blood transferrin (2.4 vs. 2.7 g/L, p < 0.001), TIBC (303.0 vs 338.0 ug/dL, p < 0.001), hemoglobin (13.1 vs 13.9 g/dL, p < 0.01) and RBC (4.3 vs. 4.6, p < 0.002) than controls. Treated patients (n = 99) had a significantly lower median iron (88.0 vs. 126.0 ug/dL, p < 0.001), ferritin (77.0 vs. 158.9 ng/mL, p < 0.005) and hepcidin (16.7 vs. 32.6 ng/mL, p < 001) and higher transferrin (2.8 vs. 2.4 g/L, p < 0.005), TIBC (336.0 vs 303.0 ug/dL, p < 0.001), RBC (4.8 vs. 4.3 M/L, p < 0.001) and hemoglobin (14.4 vs. 13.1 g/dL, p < 0.001) than untreated; the median iron (p < 0.005) was lower, and ferritin (p < 0.005), RBC (p < 0.005) and hepcidin (p < 0.002) were higher in them than in the control group. Changes in copper metabolism are accompanied by changes in iron metabolism in WD. Anti-copper treatment improves but does not normalize iron metabolism.
Collapse
Affiliation(s)
- Grażyna Gromadzka
- Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszyński University in Warsaw, Wóycickiego Str. 1/3, 01-938, Warsaw, Poland
| | - Diana Wierzbicka
- Second Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego Str. 9, 02-957, Warsaw, Poland
| | - Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego Str. 9, 02-957, Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Banacha Str. 1a, 02-097, Warsaw, Poland.
| |
Collapse
|
37
|
Yuan XZ, Yang RM, Wang XP. Management Perspective of Wilson's Disease: Early Diagnosis and Individualized Therapy. Curr Neuropharmacol 2021; 19:465-485. [PMID: 32351182 PMCID: PMC8206458 DOI: 10.2174/1570159x18666200429233517] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/13/2020] [Accepted: 04/24/2020] [Indexed: 02/05/2023] Open
Abstract
Wilson's disease (WD) is an inherited disease caused by mutations in ATP7B and is characterized by the pathological accumulation of copper in the liver and brain. Common clinical manifestations of WD include a wide range of liver disease and neurological symptoms. In some patients, psychiatric symptoms may be the only manifestation at the time of diagnosis. The clinical features of WD are highly variable and can mimic any disease of internal medicine. Therefore, for unexplained medical diseases, the possibility of WD should not be ignored. Early diagnosis and treatment can improve the prognosis of WD patients and reduce disability and early death. Gene sequencing is becoming a valuable method to diagnose WD, and if possible, all WD patients and their siblings should be genetically sequenced. Copper chelators including D-penicillamine, trientine, and dimercaptosuccinic acid can significantly improve the liver injury and symptoms of WD patients but may have a limited effect on neurological symptoms. Zinc salts may be more appropriate for the treatment of asymptomatic patients or for the maintenance treatment of symptomatic patients. High-quality clinical trials for the drug treatment of WD are still lacking, therefore, individualized treatment options for patients are recommended. Individualized treatment can be determined based on the clinical features of the WD patients, efficacy and adverse effects of the drugs, and the experience of the physician. Liver transplantation is the only effective method to save patients with acute liver failure or with severe liver disease who fail drug treatment.
Collapse
Affiliation(s)
| | | | - Xiao-Ping Wang
- Address correspondence to this author at the Department of Neurology, TongRen Hospital, Shanghai Jiao Tong University School of Medicine, No.1111 Xianxia Road, 200336, Shanghai, China; Tel: +86-021-52039999-72223; Fax: +86-021-52039999-72223; E-mail:
| |
Collapse
|
38
|
Gromadzka G, Wierzbicka D, Litwin T, Przybyłkowski A. Difference in iron metabolism may partly explain sex-related variability in the manifestation of Wilson's disease. J Trace Elem Med Biol 2020; 62:126637. [PMID: 32937238 DOI: 10.1016/j.jtemb.2020.126637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/05/2020] [Accepted: 08/26/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND/AIM Wilson's disease (WD) is a hereditary disorder characterized by abnormal metabolism of copper. For unknown reasons, the clinical picture of this disease appears to be sex-dependent. Because the metabolism of copper and iron is interrelated, we aimed to evaluate whether the variability in the clinical picture of WD could be explained by the sex difference in iron metabolism. METHODS A total of 138 WD patients were examined in this study: 39 newly diagnosed, treatment naive patients and 99 individuals already treated with decoppering drugs. The serum concentration of ceruloplasmin (Cp) and copper were measured using an enzymatic colorimetric assay and by atomic absorption spectroscopy, respectively. The parameters of iron metabolism were determined by using standard laboratory methods and enzyme immunoassays. RESULTS In the treatment naive group men had a higher median serum concentration of ferritin (290.5 vs. 81.0 ng/mL, p < 10-4), and hepcidin (Hepc) (55.4 vs. 22.8 ng/mL, p < 10-3) compared to women, and tended to have higher concentration of iron, hemoglobin (HGB) and number of red blood cells (RBC). In the treated group men had higher median ferritin (122.0 vs. 46.0 ng/mL, p < 10-4), Hepc (23.5 vs. 10.8 ng/mL, p < 10-4), iron (102.5 vs. 68.0 μg/dL, p < 10-4), HGB (15.0 vs. 13.2 g/dL, p < 10-4), and RBC (5.0 vs. 4.5 M/L, p < 10-4) than women. CONCLUSION Iron metabolism differs between men and women with WD, which may partly explain the sex difference noted in the disease manifestation.
Collapse
Affiliation(s)
- Grażyna Gromadzka
- Cardinal Stefan Wyszyński University, Faculty of Medical Science, Collegium Medicum, Warsaw, Poland
| | - Diana Wierzbicka
- Institute of Psychiatry and Neurology, Second Department of Neurology, Warsaw, Poland
| | - Tomasz Litwin
- Institute of Psychiatry and Neurology, Second Department of Neurology, Warsaw, Poland
| | - Adam Przybyłkowski
- Medical University in Warsaw, Department of Gastroenterology and Internal Medicine, Warsaw, Poland.
| |
Collapse
|
39
|
Timoshenko RV, Vaneev AN, Savin NA, Klyachko NL, Parkhomenko YN, Salikhov SV, Majouga AG, Gorelkin PV, Erofeev AS. Promising Approaches for Determination of Copper Ions in Biological Systems. ACTA ACUST UNITED AC 2020. [DOI: 10.1134/s1995078020020196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
40
|
Gromadzka G, Wierzbicka DW, Przybyłkowski A, Litwin T. Effect of homeostatic iron regulator protein gene mutation on Wilson's disease clinical manifestation: original data and literature review. Int J Neurosci 2020; 132:894-900. [PMID: 33175593 DOI: 10.1080/00207454.2020.1849190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Wilson's disease (WD) is a hereditary disorder of copper metabolism. The metabolic pathways of copper and iron are interrelated. Our goal was to determine the frequency of the two most common mutations in the coding region of the human iron homeostatic protein gene (HFE) in Europe: C282Y (rs1800562) and H63D (rs1799945) in WD patients, as well as to analyze their relation with WD phenotypic traits. MATERIAL AND METHODS HFE mutations were studied by PCR RFLP method in 445 WD patients and 102 controls. All patients met the diagnostic criteria of WD 8th International Conference on Wilson Disease and Menkes Disease. RESULTS HFE C282Y heterozygotes, both women and men, showed WD symptoms earlier than patients with wild-type HFE genotype. HFE 63HD heterozygous men presented symptoms later than HFE 63HH homozygotes, but HFE 63HD women manifested symptoms later than those with HFE 63HH genotype. CONCLUSIONS HFE genotype seems to be one of the factors modifying Wilson's disease phenotype.
Collapse
Affiliation(s)
- Grażyna Gromadzka
- Faculty of Medicine (Collegium Medicum), Cardinal Stefan Wyszyński University in Warsaw, Warsaw, Poland
| | | | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University in Warsaw, Warsaw, Poland
| | - Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| |
Collapse
|
41
|
Yuan XZ, Li GY, Chen JL, Li JQ, Wang XP. Paramagnetic Metal Accumulation in the Deep Gray Matter Nuclei Is Associated With Neurodegeneration in Wilson's Disease. Front Neurosci 2020; 14:573633. [PMID: 33041766 PMCID: PMC7525019 DOI: 10.3389/fnins.2020.573633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/27/2020] [Indexed: 02/05/2023] Open
Abstract
Background Neuropathological studies have revealed copper and iron accumulation in the deep gray matter (DGM) nuclei of patients with Wilson’s disease (WD). However, the association between metal accumulation and neurodegeneration in WD has not been well studied in vivo. The study was aimed to investigate whether metal accumulation in the DGM was associated with the structural and functional changes of DGM in neurological WD patients. Methods Seventeen neurological WD patients and 20 healthy controls were recruited for the study. Mean bulk susceptibility values and volumes of DGM were obtained from quantitative susceptibility mapping (QSM). Regions of interest including the head of the caudate nucleus, globus pallidus, putamen, thalamus, substantia nigra, red nucleus, and dentate nucleus were manually segmented. The susceptibility values and volumes of DGM in different groups were compared using a linear regression model. Correlations between susceptibility values and volumes of DGM and Unified Wilson’s Disease Rating Scale (UWDRS) neurological subscores were investigated. Results The susceptibility values of all examined DGM in WD patients were higher than those in healthy controls (P < 0.05). Volume reductions were observed in the head of the caudate nucleus, globus pallidus, putamen, thalamus, and substantia nigra of WD patients (P < 0.001). Susceptibility values were negatively correlated with the volumes of the head of the caudate nucleus (rp = −0.657, P = 0.037), putamen (rp = −0.667, P = 0.037), and thalamus (rp = −0.613, P = 0.046) in WD patients. UWDRS neurological subscores were positively correlated with the susceptibility values of all examined DGM. The susceptibility values of putamen, head of the caudate nucleus, and dentate nucleus could well predict UWDRS neurological subscores. Conclusion Our study provided in vivo evidence that paramagnetic metal accumulation in the DGM was associated with DGM atrophy and neurological impairment. The susceptibility of DGM could be used as a biomarker to assess the severity of neurodegeneration in WD.
Collapse
Affiliation(s)
- Xiang-Zhen Yuan
- Department of Neurology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gai-Ying Li
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Jia-Lin Chen
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Jian-Qi Li
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Xiao-Ping Wang
- Department of Neurology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
42
|
Li G, Wu R, Tong R, Bo B, Zhao Y, Gillen KM, Spincemaille P, Ku Y, Du Y, Wang Y, Wang X, Li J. Quantitative Measurement of Metal Accumulation in Brain of Patients With Wilson's Disease. Mov Disord 2020; 35:1787-1795. [PMID: 32681698 DOI: 10.1002/mds.28141] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 05/12/2020] [Accepted: 05/18/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Currently, no study has evaluated metal accumulation in the brains of patients with Wilson's disease by using quantitative susceptibility mapping at 3T MRI. The objectives of this study were to qualitatively and quantitatively evaluate changes in magnetic susceptibility and R2* maps in deep gray matter nuclei to discriminate Wilson's disease patients from healthy controls and to evaluate their sensitivities in diagnosing Wilson's disease. METHODS Magnetic susceptibility and R2* maps and conventional T1-weighted, T2-weighted, and T2-weighted fluid-attenuated inversion recovery images were obtained from 17 Wilson's disease patients and 14 age-matched healthy controls on a 3T MRI scanner. Differences between Wilson's disease and healthy control groups in susceptibility and R2* values in multiple deep nuclei were evaluated using a Mann-Whitney U test and receiver operating characteristic curves. The correlations of susceptibility and R2* values with Unified Wilson's Disease Rating Scale score were also performed. RESULTS Magnetic susceptibility and R2* can effectively distinguish different types of signal abnormalities. Magnetic susceptibility and R2* values in multiple deep nuclei of Wilson's disease patients were significantly higher than those in healthy controls. Magnetic susceptibility value in the substantia nigra had the highest area under the curve (0.888). There were positive correlations of the Unified Wilson's Disease Rating Scale score with susceptibility values in the caudate nucleus (r = 0.757, P = 0.011), putamen (r = 0.679, P = 0.031), and red nucleus (r = 0.638, P = 0.047), as well as R2* values in the caudate nucleus (r = 0.754, P = 0.012). CONCLUSIONS Quantitative susceptibility mapping at 3T could be a useful tool to evaluate metal accumulation in deep gray matter nuclei of Wilson's disease patients. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Gaiying Li
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Rong Wu
- Department of Neurology, Shanghai Tong-Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Tong
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Binshi Bo
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Yu Zhao
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| | - Kelly M Gillen
- Department of Radiology, Weill Medical College of Cornell University, New York, New York, USA
| | - Pascal Spincemaille
- Department of Radiology, Weill Medical College of Cornell University, New York, New York, USA
| | - Yixuan Ku
- Department of Psychology, Sun Yat-sen University, Guangzhou Higher Education Mega Center, Guangzhou, China
| | - Yasong Du
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Wang
- Department of Radiology, Weill Medical College of Cornell University, New York, New York, USA
| | - Xiaoping Wang
- Department of Neurology, Shanghai Tong-Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianqi Li
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, China
| |
Collapse
|
43
|
Wu H, Guo H, Liu H, Cui H, Fang J, Zuo Z, Deng J, Li Y, Wang X, Zhao L. Copper sulfate-induced endoplasmic reticulum stress promotes hepatic apoptosis by activating CHOP, JNK and caspase-12 signaling pathways. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 191:110236. [PMID: 32001424 DOI: 10.1016/j.ecoenv.2020.110236] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/16/2020] [Accepted: 01/21/2020] [Indexed: 06/10/2023]
Abstract
Copper (Cu), a transition metal, is an essential trace element in human and animal nutrition at low concentration, but Cu has toxic effects on tissues and organs at high concentration. Endoplasmic reticulum (ER) is a toxicological target in Cu poison. Thus far, no studies have focused on the relationship among copper, endoplasmic reticulum (ER) stress and apoptosis in animal and human livers. In the present study, mice treated with copper sulfate (CuSO4) were used to assess the impacts of copper on ER stress and hepatic apoptosis. A total of 240 mice were orally administered with 0 (control), 10, 20 and 40 mg/kg of CuSO4 for 42 days. The results indicated that CuSO4 at 10 mg/kg markedly induced hepatocyte apoptosis and ER stress. In addition, ER stress was characterized by the increased mRNA and protein levels of glucose-regulated protein 78 (GRP78) and 94 (GRP94). Furthermore, ER stress-triggered 3 apoptotic pathways were also activated by the increased intracellular calcium and up-regulated expression levels of genes involved in growth arrest- and DNA damage-inducible gene 153 (Gadd153/CHOP), c-Jun N-terminal kinase (JNK) and cysteine aspartate-specific protease 12 (caspase-12) signaling pathways in CuSO4-treated mice. In conclusion, CuSO4-induced ER stress can promote hepatic apoptosis in mice by activating CHOP, JNK and caspase-12 signaling pathways.
Collapse
Affiliation(s)
- Hongbin Wu
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Hongrui Guo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Huan Liu
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Agricultural information engineering of Sichuan Province, Sichuan Agriculture University, Yaan, Sichuan, 625014, China.
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Xun Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| |
Collapse
|
44
|
Bisaglia M, Bubacco L. Copper Ions and Parkinson's Disease: Why Is Homeostasis So Relevant? Biomolecules 2020; 10:biom10020195. [PMID: 32013126 PMCID: PMC7072482 DOI: 10.3390/biom10020195] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/20/2020] [Accepted: 01/28/2020] [Indexed: 12/18/2022] Open
Abstract
The involvement of copper in numerous physiological processes makes this metal ion essential for human life. Alterations in copper homeostasis might have deleterious consequences, and several neurodegenerative disorders, including Parkinson’s disease (PD), have been associated with impaired copper levels. In the present review, we describe the molecular mechanisms through which copper can exert its toxicity, by considering how it can interfere with other cellular processes known to play a role in PD, such as dopamine metabolism, oxidative stress, and α-synuclein aggregation. The recent experimental evidence that associates copper deficiency and the formation of superoxide dismutase 1 (SOD1) aggregates with the progression of PD is also discussed together with its therapeutic implication. Overall, the recent discoveries described in this review show how either copper deficiency or excessive levels can promote detrimental effects, highlighting the importance of preserving copper homeostasis and opening unexplored therapeutic avenues in the definition of novel disease-modifying drugs.
Collapse
|
45
|
Kalita J, Kumar V, Misra UK, Bora HK. Movement Disorder in Copper Toxicity Rat Model: Role of Inflammation and Apoptosis in the Corpus Striatum. Neurotox Res 2019; 37:904-912. [PMID: 31811585 DOI: 10.1007/s12640-019-00140-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022]
Abstract
The pattern of copper (Cu) toxicity in humans is similar to Wilson disease, and they have movement disorders and frequent involvement of corpus striatum. The extent of cell deaths in corpus striatum may be the basis of movement disorder and may be confirmed in the experimental study. To evaluate the extent of apoptosis and glial activation in corpus striatum following Cu toxicity in a rat model, and correlate these with spontaneous locomotor activity (SLA), six male Wistar rats were fed normal saline (group I) and another six were fed copper sulfate 100 mg/kgBWt/daily orally (group II). At 1 month, neurobehavioral studies including SLA, rotarod, and grip strength were done. Corpus striatum was removed and was subjected to glial fibrillary acidic protein (GFAP) and caspase-3 immunohistochemistry. The concentration of tissue Cu, total antioxidant capacity (TAC), glutathione (GSH), malondialdehyde (MDA), and glutamate were measured. Group II rats had higher expression of caspase-3 (Mean ± SEM 32.67 ± 1.46 vs 4.47 ± 1.08; p < 0.01) and GFAP (41.81 ± 1.68 vs 31.82 ± 1.27; p < 0.01) compared with group I. Neurobehavioral studies revealed reduced total distance traveled, time moving, the number of rearing, latency to fall on the rotarod, grip strength, and increased resting time compared with group I. The expression of GFAP and caspase-3 correlated with SLA parameters, tissue Cu, GSH, MDA, TAC, and glutamate levels. The impaired locomotor activity in Cu toxicity rats is due to apoptotic and inflammatory-mediated cell death in the corpus striatum because of Cu-mediated oxidative stress and excitotoxicity.
Collapse
Affiliation(s)
- Jayantee Kalita
- Department of Neurology, Sanjay Gandhi Post Graduate Medical Sciences, Raebareily Road, Lucknow, 226014, India.
| | - Vijay Kumar
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - Usha K Misra
- Department of Neurology, Sanjay Gandhi Post Graduate Medical Sciences, Raebareily Road, Lucknow, 226014, India
| | - Himangsu K Bora
- National Laboratory Animal Centre, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
46
|
Güngör Ş, Selimoğlu MA, Varol Fİ, Güngör S, Üremiş MM. The effects of iron and zinc status on prognosis in pediatric Wilson's disease. J Trace Elem Med Biol 2019; 55:33-38. [PMID: 31345362 DOI: 10.1016/j.jtemb.2019.05.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Wilson's disease (WD) is a metabolic disorder leading to hepatic and extrahepatic copper deposition. Several studies have reported that besides copper (Cu), iron (Fe) and zinc (Zn) are also accumulated at varying levels in various tissues in WD. However, there is not an adequate number of studies investigating the effects of Fe and Zn status on WD presentation and prognosis. We aimed to evaluate serum levels of ferritin (SFr), copper (SCu), and zinc (SZn) in WD and determine their role in disease presentation and prognosis. MATERIALS-METHOD We retrospectively reviewed the medical records of 97 pediatric patients with WD who were diagnosed and followed at İnönü University Pediatric Gastroenterology, Hepatology and Nutrition Department between January 2006 and May 2017. Serum Cu and Zn levels were analyzed by using flame atomic absorption spectrophotometer. Ferritin was analyzed by chemiluminescence immunoassay method. RESULTS Analysis of serum levels of the elements according to the type of presentation, there was no significant difference between the groups for ceruloplasmin. However, SCU, FSCu, SFr and 24 h urinary copper levels were significantly higher (p = 0.002, p = 0.003, p = 0.023 and p < 0.001, respectively) and SZn and CSZn levels were significantly lower (fulminant WD). p < 0.001, p < 0.001). There was a positive correlation between SFr, SCu serum levels and mortality scores (respectively, r: 0.501, 0.564 for PELD, r:0.490, 0.504 for MELD, r: 0.345, 0.374 for Dhwan), and a negative correlation between SZn level and mortality scores. (r:-0.650 for PELD, r:-0.703 for MELD, r:-0.642 for Dhwan) We used the ROC curves to determine the worst prognosis for fulminant Wilson disease. According to these limit values, we found that the sensitivity and specificity of FWD development was significantly higher. (for SZn sensitivity of 91.5%, a specificity of 100%, p=<0,001, for SCu predicted FWD development with a sensitivity of 100%, a specificity of 73.7%, p=<0,001, for SFr predicted FWD development with a sensitivity of 92.9%, a specificity of 66.2%, p < 0,001) CONCLUSION: Our study suggests that SFr, SCu, SZn levels might have prognostic importance for WD.
Collapse
Affiliation(s)
- Şükrü Güngör
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, İnönü University, Faculty of Medicine, Malatya,Turkey.
| | - Mukadder Ayşe Selimoğlu
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, İnönü University, Faculty of Medicine, Malatya,Turkey.
| | - Fatma İlknur Varol
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, İnönü University, Faculty of Medicine, Malatya,Turkey.
| | - Serdal Güngör
- Department of Pediatric Neurology, İnönü University, Faculty of Medicine, Malatya,Turkey.
| | - Muhammed Mehdi Üremiş
- Department of Medicinal Biochemistry, İnönü University, Faculty of Medicine, Malatya, Turkey.
| |
Collapse
|
47
|
Brain volume is related to neurological impairment and to copper overload in Wilson's disease. Neurol Sci 2019; 40:2089-2095. [PMID: 31147855 PMCID: PMC6745045 DOI: 10.1007/s10072-019-03942-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 05/16/2019] [Indexed: 12/16/2022]
Abstract
Introduction To determine whether brain volume was associated with functional and neurological impairments and with copper overload markers in patients with Wilson’s disease. Methods In 48 treatment-naïve patients, we assessed functional and neurological impairments with the Unified Wilson’s Disease Rating Scale, measured normalized brain volumes based on magnetic resonance images, and assessed concentration of non-ceruloplasmin-bound copper. We correlated brain volume measures with functional and neurological impairment scores and copper overload indices. Results Functional and neurological impairments correlated with all brain volume measures, including the total brain volume and the volumes of white matter and gray matter (both peripheral gray matter and deep brain nuclei). Higher non-ceruloplasmin-bound copper concentrations were associated with greater functional and neurological impairments and lower brain volumes. Conclusions Our findings provided the first in vivo evidence that the severity of brain atrophy is a correlate of functional and neurological impairments in patients with Wilson’s disease and that brain volume could serve as a marker of neurodegeneration induced by copper. Electronic supplementary material The online version of this article (10.1007/s10072-019-03942-z) contains supplementary material, which is available to authorized users.
Collapse
|
48
|
Dusek P, Litwin T, Członkowska A. Neurologic impairment in Wilson disease. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S64. [PMID: 31179301 DOI: 10.21037/atm.2019.02.43] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neurologic symptoms in Wilson disease (WD) appear at an older age compared to hepatic symptoms and manifest in patients with misdiagnosed liver disease, in patients when the hepatic stage is clinically silent, in the case of non-compliance with anti-copper treatment, or with treatment failure. Neurologic symptoms in WD are caused by nervous tissue damage that is primarily a consequence of extrahepatic copper toxicity. Copper levels in brain tissues as well as cerebrospinal fluid (CSF) are diffusely increased by a factor of 10 and its toxicity involves various mechanisms such as mitochondrial toxicity, oxidative stress, cell membrane damage, crosslinking of DNA, and inhibition of enzymes. Excess copper is initially taken-up and buffered by astrocytes and oligodendrocytes but ultimately causes dysfunction of blood-brain-barrier and demyelination. Most severe neuropathologic abnormalities, including tissue rarefaction, reactive astrogliosis, myelin palor, and presence of iron-laden macrophages, are typically present in the putamen while other basal ganglia, thalami, and brainstem are usually less affected. The most common neurologic symptoms of WD are movement disorders including tremor, dystonia, parkinsonism, ataxia and chorea which are associated with dysphagia, dysarthria and drooling. Patients usually manifest with various combinations of these symptoms while purely monosymptomatic presentation is rare. Neurologic symptoms are largely reversible with anti-copper treatment, but a significant number of patients are left with residual impairment. The approach for symptomatic treatment in WD is based on guidelines for management of common movement disorders. The vast majority of WD patients with neurologic symptoms have abnormalities on brain magnetic resonance imaging (MRI). Pathologic MRI changes include T2 hyperintensities in the basal ganglia, thalami and white matter, T2 hypointensities in the basal ganglia, and atrophy. Most importantly, brain damage and neurologic symptoms can be prevented with an early initiation of anti-copper treatment. Introducing population WD screening, e.g., by exome sequencing genetic methods, would allow early treatment and decrease the neurologic burden of WD.
Collapse
Affiliation(s)
- Petr Dusek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia.,Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Tomasz Litwin
- 2nd Department of Neurology, Institute Psychiatry and Neurology, Warsaw, Poland
| | - Anna Członkowska
- 2nd Department of Neurology, Institute Psychiatry and Neurology, Warsaw, Poland
| |
Collapse
|
49
|
The exceptional sensitivity of brain mitochondria to copper. Toxicol In Vitro 2018; 51:11-22. [PMID: 29715505 DOI: 10.1016/j.tiv.2018.04.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/27/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023]
Abstract
Wilson disease (WD) is characterized by a disrupted copper homeostasis resulting in dramatically increased copper levels, mainly in liver and brain. While copper damage to mitochondria is an established feature in WD livers, much less is known about such detrimental copper effects in other organs. We therefore assessed the mitochondrial sensitivity to copper in a tissue specific manner, namely of isolated rat liver, kidney, heart, and brain mitochondria. Brain mitochondria presented with exceptional copper sensitivity, as evidenced by a comparatively early membrane potential loss, profound structural changes already at low copper dose, and a dose-dependent reduced capacity to produce ATP. This sensitivity was likely due to a copper-dependent attack on free protein thiols and due to a decreased copper reactive defense system, as further evidenced in neuroblastoma SHSY5Y cells. In contrast, an increased production of reactive oxygen species was found to be a late-stage event, only occurring in destroyed mitochondria. We therefore propose mitochondrial protein thiols as major targets of mitochondrial copper toxicity.
Collapse
|
50
|
Peng F, Xie F, Muzik O. Alteration of Copper Fluxes in Brain Aging: A Longitudinal Study in Rodent Using 64CuCl 2-PET/CT. Aging Dis 2018; 9:109-118. [PMID: 29392086 PMCID: PMC5772849 DOI: 10.14336/ad.2017.1025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 10/25/2017] [Indexed: 12/20/2022] Open
Abstract
Brain aging is associated with changes of various metabolic pathways. Copper is required for brain development and function, but little is known about changes in copper metabolism during brain aging. The objective of this study was to investigate alteration of copper fluxes in the aging mouse brain with positron emission tomography/computed tomography using 64CuCl2 as a radiotracer (64CuCl2-PET/CT). A longitudinal study was conducted in C57BL/6 mice (n = 5) to measure age-dependent brain and whole-body changes of 64Cu radioactivity using PET/CT after oral administration of 64CuCl2 as a radiotracer. Cerebral 64Cu uptake at 13 months of age (0.17 ± 0.05 %ID/g) was higher than the cerebral 64Cu uptake at 5 months of age (0.11 ± 0.06 %ID/g, p < 0.001), followed by decrease to (0.14 ± 0.04 %ID/g, p = 0.02) at 26 months of age. In contrast, cerebral 18F-FDG uptake was highest at 5 months of age (7.8 ± 1.2 %ID/g) and decreased to similar values at 12 (5.2 ± 1.1 %ID/g, p < 0.001) and 22 (5.6 ± 1.1 %ID/g, p < 0.001) months of age. The findings demonstrated alteration of copper fluxes associated with brain aging and the time course of brain changes in copper fluxes differed from changes in brain glucose metabolism across time, suggesting independent underlying physiological processes. Hence, age-dependent changes of cerebral copper fluxes might represent a novel metabolic biomarker for assessment of human brain aging process with PET/CT using 64CuCl2 as a radiotracer.
Collapse
Affiliation(s)
- Fangyu Peng
- 1Department of Radiology, and.,2Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX75390, USA
| | | | - Otto Muzik
- 3Department of Pediatrics and.,4 Department of Radiology, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|