1
|
Wang Y, Li D, Xu K, Wang G, Zhang F. Copper homeostasis and neurodegenerative diseases. Neural Regen Res 2025; 20:3124-3143. [PMID: 39589160 PMCID: PMC11881714 DOI: 10.4103/nrr.nrr-d-24-00642] [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: 07/23/2024] [Revised: 07/27/2024] [Accepted: 10/14/2024] [Indexed: 11/27/2024] Open
Abstract
Copper, one of the most prolific transition metals in the body, is required for normal brain physiological activity and allows various functions to work normally through its range of concentrations. Copper homeostasis is meticulously maintained through a complex network of copper-dependent proteins, including copper transporters (CTR1 and CTR2), the two copper ion transporters the Cu -transporting ATPase 1 (ATP7A) and Cu-transporting beta (ATP7B), and the three copper chaperones ATOX1, CCS, and COX17. Disruptions in copper homeostasis can lead to either the deficiency or accumulation of copper in brain tissue. Emerging evidence suggests that abnormal copper metabolism or copper binding to various proteins, including ceruloplasmin and metallothionein, is involved in the pathogenesis of neurodegenerative disorders. However, the exact mechanisms underlying these processes are not known. Copper is a potent oxidant that increases reactive oxygen species production and promotes oxidative stress. Elevated reactive oxygen species levels may further compromise mitochondrial integrity and cause mitochondrial dysfunction. Reactive oxygen species serve as key signaling molecules in copper-induced neuroinflammation, with elevated levels activating several critical inflammatory pathways. Additionally, copper can bind aberrantly to several neuronal proteins, including alpha-synuclein, tau, superoxide dismutase 1, and huntingtin, thereby inducing neurotoxicity and ultimately cell death. This study focuses on the latest literature evaluating the role of copper in neurodegenerative diseases, with a particular focus on copper-containing metalloenzymes and copper-binding proteins in the regulation of copper homeostasis and their involvement in neurodegenerative disease pathogenesis. By synthesizing the current findings on the functions of copper in oxidative stress, neuroinflammation, mitochondrial dysfunction, and protein misfolding, we aim to elucidate the mechanisms by which copper contributes to a wide range of hereditary and neuronal disorders, such as Wilson's disease, Menkes' disease, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and multiple sclerosis. Potential clinically significant therapeutic targets, including superoxide dismutase 1, D-penicillamine, and 5,7-dichloro-2-[(dimethylamino)methyl]-8-hydroxyquinoline, along with their associated therapeutic agents, are further discussed. Ultimately, we collate evidence that copper homeostasis may function in the underlying etiology of several neurodegenerative diseases and offer novel insights into the potential prevention and treatment of these diseases based on copper homeostasis.
Collapse
Affiliation(s)
- Yuanyuan Wang
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Daidi Li
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Kaifei Xu
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Guoqing Wang
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Feng Zhang
- International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Ministry of Education, Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
2
|
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
|
3
|
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: 37] [Impact Index Per Article: 18.5] [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
|
4
|
Fujisawa C, Kodama H, Sato Y, Mimaki M, Yagi M, Awano H, Matsuo M, Shintaku H, Yoshida S, Takayanagi M, Kubota M, Takahashi A, Akasaka Y. Early clinical signs and treatment of Menkes disease. Mol Genet Metab Rep 2022; 31:100849. [PMID: 35242581 PMCID: PMC8861833 DOI: 10.1016/j.ymgmr.2022.100849] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Menkes disease (MD) is an X-linked recessive disorder caused by mutations in ATP7A. Patients with MD exhibit severe neurological and connective tissue disorders due to copper deficiency and typically die before 3 years of age. Early treatment with copper injections during the neonatal period, before the occurrence of neurological symptoms, can alleviate neurological disturbances to some degree. We investigated whether early symptoms can help in the early diagnosis of MD. Abnormal hair growth, prolonged jaundice, and feeding difficulties were observed during the neonatal period in 20 of 69, 16 of 67, and 3 of 18 patients, respectively. Only three patients visited a physician during the neonatal period; MD diagnosis was not made at that point. The mean age at diagnosis was 8.7 months. Seven patients, who were diagnosed in the prenatal stage or soon after birth, as they had a family history of MD, received early treatment. No diagnosis was made based on early symptoms, highlighting the difficulty in diagnosing MD based on symptoms observed during the neonatal period. Patients who received early treatment lived longer than their elderly relatives with MD. Three patients could walk and did not have seizures. Therefore, effective newborn screening for MD should be prioritized.
Collapse
Affiliation(s)
- Chie Fujisawa
- Department of Pediatrics, School of Medicine, Teikyo University, Itabashi-ku, Tokyo 173-8606, Japan
- Department of Research Unit, Faculty of Medicine, Toho University, Ota-ku, Tokyo 143-8540, Japan
- Corresponding authors at: Department of Research Unit, Faculty of Medicine, Toho University, Omori-Nishi, Ota-ku, Tokyo 143-8540, Japan.
| | - Hiroko Kodama
- Department of Pediatrics, School of Medicine, Teikyo University, Itabashi-ku, Tokyo 173-8606, Japan
- Graduate School of Health Sciences, Teikyo Heisei University, Toshima-ku, Tokyo 170-8445, Japan
- Corresponding authors at: Department of Research Unit, Faculty of Medicine, Toho University, Omori-Nishi, Ota-ku, Tokyo 143-8540, Japan.
| | - Yasuhiro Sato
- Department of Pediatrics, School of Medicine, Teikyo University, Itabashi-ku, Tokyo 173-8606, Japan
| | - Masakazu Mimaki
- Department of Pediatrics, School of Medicine, Teikyo University, Itabashi-ku, Tokyo 173-8606, Japan
| | - Mariko Yagi
- Department of Childhood Development and Education, Faculty of Human Science, Konan Women's University, Higashinada-ku, Kobe-shi, Hyogo 658-0001, Japan
| | - Hiroyuki Awano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Chuo-ku, Kobe-shi, Hyogo 650-0017, Japan
| | - Muneaki Matsuo
- Department of Pediatrics, Faculty of Medicine, Saga University, Nabeshima, Saga-shi, Saga 840-8502, Japan
| | - Haruo Shintaku
- Department of Pediatrics, Graduate School of Medicine, Osaka City University, Abeno-ku, Osaka-City, Osaka 545-8585, Japan
| | - Sayaka Yoshida
- Department of Pediatrics, Nara Prefecture General Medical Center, Nara-shi, Nara 630-8581, Japan
| | - Masaki Takayanagi
- Department of Pediatrics, Chiba Children's Hospital, Chiba-shi, Chiba 266-0007, Japan
| | - Mitsuru Kubota
- Department of General Pediatrics and Interdisciplinary Medicine, National Center for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
| | - Akihito Takahashi
- Department of Pediatrics, Kurashiki Central Hospital, Kurashiki, Okayama 710-8602, Japan
| | - Yoshikiyo Akasaka
- Unit of Regenerative Diseases Research, Division of Research Promotion and Development, Advanced Medical Research Center, Toho University Graduate School of Medicine, Ota-ku, Tokyo 143-8540, Japan
| |
Collapse
|
5
|
Firth G, Blower JE, Bartnicka JJ, Mishra A, Michaels AM, Rigby A, Darwesh A, Al-Salemee F, Blower PJ. Non-invasive radionuclide imaging of trace metal trafficking in health and disease: "PET metallomics". RSC Chem Biol 2022; 3:495-518. [PMID: 35656481 PMCID: PMC9092424 DOI: 10.1039/d2cb00033d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/07/2022] [Indexed: 12/05/2022] Open
Abstract
Several specific metallic elements must be present in the human body to maintain health and function. Maintaining the correct quantity (from trace to bulk) and location at the cell and tissue level is essential. The study of the biological role of metals has become known as metallomics. While quantities of metals in cells and tissues can be readily measured in biopsy and autopsy samples by destructive analytical techniques, their trafficking and its role in health and disease are poorly understood. Molecular imaging with radionuclides - positron emission tomography (PET) and single photon emission computed tomography (SPECT) - is emerging as a means to non-invasively study the acute trafficking of essential metals between organs, non-invasively and in real time, in health and disease. PET scanners are increasingly widely available in hospitals, and methods for producing radionuclides of some of the key essential metals are developing fast. This review summarises recent developments in radionuclide imaging technology that permit such investigations, describes the radiological and physicochemical properties of key radioisotopes of essential trace metals and useful analogues, and introduces current and potential future applications in preclinical and clinical investigations to study the biology of essential trace metals in health and disease.
Collapse
Affiliation(s)
- George Firth
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Julia E Blower
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Joanna J Bartnicka
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Aishwarya Mishra
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Aidan M Michaels
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Alex Rigby
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Afnan Darwesh
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Fahad Al-Salemee
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Philip J Blower
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| |
Collapse
|
6
|
Zirjacks L, Stransky N, Klumpp L, Prause L, Eckert F, Zips D, Schleicher S, Handgretinger R, Huber SM, Ganser K. Repurposing Disulfiram for Targeting of Glioblastoma Stem Cells: An In Vitro Study. Biomolecules 2021; 11:1561. [PMID: 34827559 PMCID: PMC8615869 DOI: 10.3390/biom11111561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/12/2021] [Accepted: 10/16/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal glioblastoma stem cells (GSCs), a subpopulation in glioblastoma that are responsible for therapy resistance and tumor spreading in the brain, reportedly upregulate aldehyde dehydrogenase isoform-1A3 (ALDH1A3) which can be inhibited by disulfiram (DSF), an FDA-approved drug formerly prescribed in alcohol use disorder. Reportedly, DSF in combination with Cu2+ ions exerts multiple tumoricidal, chemo- and radio-therapy-sensitizing effects in several tumor entities. The present study aimed to quantify these DSF effects in glioblastoma stem cells in vitro, regarding dependence on ALDH1A3 expression. To this end, two patient-derived GSC cultures with differing ALDH1A3 expression were pretreated (in the presence of CuSO4, 100 nM) with DSF (0 or 100 nM) and the DNA-alkylating agent temozolomide (0 or 30 µM) and then cells were irradiated with a single dose of 0-8 Gy. As read-outs, cell cycle distribution and clonogenic survival were determined by flow cytometry and limited dilution assay, respectively. As a result, DSF modulated cell cycle distribution in both GSC cultures and dramatically decreased clonogenic survival independently of ALDH1A3 expression. This effect was additive to the impairment of clonogenic survival by radiation, but not associated with radiosensitization. Of note, cotreatment with temozolomide blunted the DSF inhibition of clonogenic survival. In conclusion, DSF targets GSCs independent of ALDH1A3 expression, suggesting a therapeutic efficacy also in glioblastomas with low mesenchymal GSC populations. As temozolomide somehow antagonized the DSF effects, strategies for future combination of DSF with the adjuvant standard therapy (fractionated radiotherapy and concomitant temozolomide chemotherapy followed by temozolomide maintenance therapy) are not supported by the present study.
Collapse
Affiliation(s)
- Lisa Zirjacks
- Department of Radiation Oncology, Eberhard-Karls University, 72076 Tübingen, Germany; (L.Z.); (N.S.); (L.K.); (L.P.); (F.E.); (D.Z.); (K.G.)
| | - Nicolai Stransky
- Department of Radiation Oncology, Eberhard-Karls University, 72076 Tübingen, Germany; (L.Z.); (N.S.); (L.K.); (L.P.); (F.E.); (D.Z.); (K.G.)
| | - Lukas Klumpp
- Department of Radiation Oncology, Eberhard-Karls University, 72076 Tübingen, Germany; (L.Z.); (N.S.); (L.K.); (L.P.); (F.E.); (D.Z.); (K.G.)
| | - Lukas Prause
- Department of Radiation Oncology, Eberhard-Karls University, 72076 Tübingen, Germany; (L.Z.); (N.S.); (L.K.); (L.P.); (F.E.); (D.Z.); (K.G.)
| | - Franziska Eckert
- Department of Radiation Oncology, Eberhard-Karls University, 72076 Tübingen, Germany; (L.Z.); (N.S.); (L.K.); (L.P.); (F.E.); (D.Z.); (K.G.)
| | - Daniel Zips
- Department of Radiation Oncology, Eberhard-Karls University, 72076 Tübingen, Germany; (L.Z.); (N.S.); (L.K.); (L.P.); (F.E.); (D.Z.); (K.G.)
| | - Sabine Schleicher
- Department of Hematology and Oncology, University Hospital Tuebingen, Children’s Hospital, 72076 Tuebingen, Germany; (S.S.); (R.H.)
| | - Rupert Handgretinger
- Department of Hematology and Oncology, University Hospital Tuebingen, Children’s Hospital, 72076 Tuebingen, Germany; (S.S.); (R.H.)
| | - Stephan M. Huber
- Department of Radiation Oncology, Eberhard-Karls University, 72076 Tübingen, Germany; (L.Z.); (N.S.); (L.K.); (L.P.); (F.E.); (D.Z.); (K.G.)
| | - Katrin Ganser
- Department of Radiation Oncology, Eberhard-Karls University, 72076 Tübingen, Germany; (L.Z.); (N.S.); (L.K.); (L.P.); (F.E.); (D.Z.); (K.G.)
| |
Collapse
|
7
|
Jauregui-Osoro M, De Robertis S, Halsted P, Gould SM, Yu Z, Paul RL, Marsden PK, Gee AD, Fenwick A, Blower PJ. Production of copper-64 using a hospital cyclotron: targetry, purification and quality analysis. Nucl Med Commun 2021; 42:1024-1038. [PMID: 34397988 PMCID: PMC8357037 DOI: 10.1097/mnm.0000000000001422] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/09/2021] [Indexed: 12/31/2022]
Abstract
OBJECTIVES To construct and evaluate a 64Cu production system that minimises the amount of costly 64Ni, radionuclidic impurities and nonradioactive metal contamination and maximises radiochemical and radionuclidic purity and molar activity; and to report analytical and quality control methods that can be used within typical PET radiochemistry production facilities to measure metal ion concentrations and radiometal molar activities. METHODS Low volume was ensured by dissolving the irradiated nickel in a low volume of hydrochloric acid (<1 mL) using the concave gold target backing as a reaction vessel in a custom-built target holder. Removal of contaminating 55Co and nonradioactive trace metals was ensured by adding an intermediate hydrochloric acid concentration step during the conventional ion-exchange elution process. The radionuclidic purity of the product was determined by half-life measurements, gamma spectroscopy and ion radiochromatography. Trace metal contamination and molar activity were determined by ion chromatography. RESULTS AND CONCLUSIONS On a small scale, suitable for preclinical research, the process produced typically 3.2 GBq 64Cu in 2 mL solution from 9.4 ± 2.1 mg nickel-64 electroplated onto a gold target backing. The product had high molar activity (121.5 GBq/µmol), was free of trace metal contamination detectable by ion chromatography and has been used for many preclinical and clinical PET imaging applications.
Collapse
Affiliation(s)
- Maite Jauregui-Osoro
- School of Biomedical Engineering and Imaging Sciences, King’s College London, School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital
| | - Simona De Robertis
- School of Biomedical Engineering and Imaging Sciences, King’s College London, School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital
| | - Philip Halsted
- School of Biomedical Engineering and Imaging Sciences, King’s College London, School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital
| | - Sarah-May Gould
- School of Biomedical Engineering and Imaging Sciences, King’s College London, School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital
| | - Zilin Yu
- School of Biomedical Engineering and Imaging Sciences, King’s College London, School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital
| | - Rowena L Paul
- School of Biomedical Engineering and Imaging Sciences, King’s College London, School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital
| | - Paul K Marsden
- School of Biomedical Engineering and Imaging Sciences, King’s College London, School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital
| | - Antony D Gee
- School of Biomedical Engineering and Imaging Sciences, King’s College London, School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital
| | - Andrew Fenwick
- National Physical Laboratory, Teddington, Middlesex, London, UK
| | - Philip J. Blower
- School of Biomedical Engineering and Imaging Sciences, King’s College London, School of Biomedical Engineering and Imaging Sciences, St Thomas’ Hospital
| |
Collapse
|
8
|
Babak MV, Ahn D. Modulation of Intracellular Copper Levels as the Mechanism of Action of Anticancer Copper Complexes: Clinical Relevance. Biomedicines 2021; 9:biomedicines9080852. [PMID: 34440056 PMCID: PMC8389626 DOI: 10.3390/biomedicines9080852] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/29/2022] Open
Abstract
Copper (Cu) is a vital element required for cellular growth and development; however, even slight changes in its homeostasis might lead to severe toxicity and deleterious medical conditions. Cancer patients are typically associated with higher Cu content in serum and tumor tissues, indicating increased demand of cancer cells for this micronutrient. Cu is known to readily cycle between the +1 and +2 oxidation state in biological systems. The mechanism of action of Cu complexes is typically based on their redox activity and induction of reactive oxygen species (ROS), leading to deadly oxidative stress. However, there are a number of other biomolecular mechanisms beyond ROS generation that contribute to the activity of anticancer Cu drug candidates. In this review, we discuss how interfering with intracellular Cu balance via either diet modification or addition of inorganic Cu supplements or Cu-modulating compounds affects tumor development, progression, and sensitivity to treatment modalities. We aim to provide the rationale for the use of Cu-depleting and Cu-overloading conditions to generate the best possible patient outcome with minimal toxicity. We also discuss the advantages of the use of pre-formed Cu complexes, such as Cu-(bis)thiosemicarbazones or Cu-N-heterocyclic thiosemicarbazones, in comparison with the in situ formed Cu complexes with metal-binding ligands. In this review, we summarize available clinical and mechanistic data on clinically relevant anticancer drug candidates, including Cu supplements, Cu chelators, Cu ionophores, and Cu complexes.
Collapse
|
9
|
Nguyen MP, Goorden MC, Beekman FJ. EXIRAD-HE: multi-pinhole high-resolution ex vivo imaging of high-energy isotopes. ACTA ACUST UNITED AC 2020; 65:225029. [DOI: 10.1088/1361-6560/abbb77] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
10
|
Meggyesy PM, Masaldan S, Clatworthy SAS, Volitakis I, Eyckens DJ, Aston-Mourney K, Cater MA. Copper Ionophores as Novel Antiobesity Therapeutics. Molecules 2020; 25:E4957. [PMID: 33120881 PMCID: PMC7672559 DOI: 10.3390/molecules25214957] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/18/2020] [Accepted: 10/26/2020] [Indexed: 12/30/2022] Open
Abstract
The therapeutic utility of the copper ionophore disulfiram was investigated in a diet-induced obesity mouse model (C57BL/6J background), both through administration in feed (0.05 to 1% (w/w)) and via oral gavage (150 mg/kg) for up to eight weeks. Mice were monitored for body weight, fat deposition (perigonadal fat pads), metabolic changes (e.g., glucose dyshomeostasis) and pathologies (e.g., hepatic steatosis, hyperglycaemia and hypertriglyceridemia) associated with a high-fat diet. Metal-related pharmacological effects across major organs and serums were investigated using inductively coupled plasma mass spectrometry (ICP-MS). Disulfiram treatments (all modes) augmented hepatic copper in mice, markedly moderated body weight and abolished the deleterious systemic changes associated with a high-fat diet. Likewise, another chemically distinct copper ionophore H2(gtsm), administered daily (oral gavage), also augmented hepatic copper and moderated mouse body weight. Postmortem histological examinations of the liver and other major organs, together with serum aminotransferases, supported the reported therapeutic safety of disulfiram. Disulfiram specifically altered systemic copper in mice and altered hepatic copper metabolism, perturbing the incorporation of copper into ceruloplasmin (holo-ceruloplasmin biosynthesis) and subsequently reducing serum copper concentrations. Serum ceruloplasmin represents a biomarker for disulfiram activity. Our results establish copper ionophores as a potential class of antiobesity agents.
Collapse
Affiliation(s)
- Peter M. Meggyesy
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, Burwood, Victoria 3125, Australia; (P.M.M.); (S.M.); (S.A.S.C.)
| | - Shashank Masaldan
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, Burwood, Victoria 3125, Australia; (P.M.M.); (S.M.); (S.A.S.C.)
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia;
| | - Sharnel A. S. Clatworthy
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, Burwood, Victoria 3125, Australia; (P.M.M.); (S.M.); (S.A.S.C.)
| | - Irene Volitakis
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia;
| | - Daniel J. Eyckens
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Victoria 3216, Australia;
| | - Kathryn Aston-Mourney
- School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical~Translation, Deakin University, Geelong 3220, Australia;
| | - Michael A. Cater
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, Burwood, Victoria 3125, Australia; (P.M.M.); (S.M.); (S.A.S.C.)
- Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
11
|
Hoshina T, Nozaki S, Hamazaki T, Kudo S, Nakatani Y, Kodama H, Shintaku H, Watanabe Y. Disulfiram enhanced delivery of orally administered copper into the central nervous system in Menkes disease mouse model. J Inherit Metab Dis 2018; 41:1285-1291. [PMID: 30132231 DOI: 10.1007/s10545-018-0239-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/13/2018] [Accepted: 07/25/2018] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Menkes disease (MD) is an X-linked recessive disorder caused by dysfunction of a copper-transporting protein, leading to severe neurodegeneration in early childhood. We investigated whether a lipophilic copper chelator, disulfiram, could enhance copper absorption from the intestine and transport copper across the blood-brain barrier in MD model mice. METHODS Wild type and MD model mice were pretreated with disulfiram for 30 min before oral administration of 64CuCl2. Each organ was sequentially analyzed for radioactivity with γ counting. Copper uptake into the brain parenchyma was assessed by ex vivo autoradiography. RESULTS In wild type mice, orally administered copper was initially detected in the intestine within 2 h, reaching a maximum level in the liver (19.6 ± 3.8 percentage injected dose per gram [%ID/g]) at 6 h. In MD model mice, the copper reached the maximum level in the liver (5.3 ± 1.5 %ID/g) at 4 h, which was lower than that of wild type mice (19.0 ± 7.4 %ID/g) (P < 0.05). Pretreatment of disulfiram in MD model mice increased the copper level in the brain (0.59 ± 0.28 %ID/g) at 24 h compared with MD model mice without disulfiram (0.07 ± 0.05 %ID/g) (P < 0.05). Ex vivo autoradiography revealed that high levels of copper uptake was observed in the cerebral cortex upon disulfiram pretreatment. CONCLUSION Our data demonstrated that disulfiram enhanced the delivery of orally administered copper into the central nervous system in MD model mice. The administration of disulfiram will enable patients to avoid unpleasant subcutaneous copper injection in the future.
Collapse
Affiliation(s)
- Takao Hoshina
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan.
| | - Satoshi Nozaki
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research and Center for Life Science Technologies, Kobe, Hyogo, Japan
| | - Takashi Hamazaki
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Satoshi Kudo
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yuka Nakatani
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research and Center for Life Science Technologies, Kobe, Hyogo, Japan
| | - Hiroko Kodama
- Department of Pediatrics, Teikyo University School of Medicine, Tokyo, Japan
| | - Haruo Shintaku
- Department of Pediatrics, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research and Center for Life Science Technologies, Kobe, Hyogo, Japan
| |
Collapse
|
12
|
McInnes LE, Rudd SE, Donnelly PS. Copper, gallium and zirconium positron emission tomography imaging agents: The importance of metal ion speciation. Coord Chem Rev 2017. [DOI: 10.1016/j.ccr.2017.05.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
13
|
Liu X, Braun GB, Qin M, Ruoslahti E, Sugahara KN. In vivo cation exchange in quantum dots for tumor-specific imaging. Nat Commun 2017; 8:343. [PMID: 28839238 PMCID: PMC5571182 DOI: 10.1038/s41467-017-00153-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 06/02/2017] [Indexed: 01/30/2023] Open
Abstract
In vivo tumor imaging with nanoprobes suffers from poor tumor specificity. Here, we introduce a nanosystem, which allows selective background quenching to gain exceptionally tumor-specific signals. The system uses near-infrared quantum dots and a membrane-impermeable etchant, which serves as a cation donor. The etchant rapidly quenches the quantum dots through cation exchange (ionic etching), and facilitates renal clearance of metal ions released from the quantum dots. The quantum dots are intravenously delivered into orthotopic breast and pancreas tumors in mice by using the tumor-penetrating iRGD peptide. Subsequent etching quenches excess quantum dots, leaving a highly tumor-specific signal provided by the intact quantum dots remaining in the extravascular tumor cells and fibroblasts. No toxicity is noted. The system also facilitates the detection of peritoneal tumors with high specificity upon intraperitoneal tumor targeting and selective etching of excess untargeted quantum dots. In vivo cation exchange may be a promising strategy to enhance specificity of tumor imaging.The imaging of tumors in vivo using nanoprobes has been challenging due to the lack of sufficient tumor specificity. Here, the authors develop a tumor-specific quantum dot system that permits in vivo cation exchange to achieve selective background quenching and high tumor-specific imaging.
Collapse
Affiliation(s)
- Xiangyou Liu
- Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Road, La Jolla, California, 92037, USA
- Department of Surgery, Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, 1130 St Nicholas Avenue, New York, New York, 10032, USA
| | - Gary B Braun
- Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Road, La Jolla, California, 92037, USA.
| | - Mingde Qin
- Program of Materials Science and Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Erkki Ruoslahti
- Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Road, La Jolla, California, 92037, USA
- Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, 93106, USA
| | - Kazuki N Sugahara
- Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Road, La Jolla, California, 92037, USA.
- Department of Surgery, Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, 1130 St Nicholas Avenue, New York, New York, 10032, USA.
| |
Collapse
|
14
|
Yang J, Yang J, Wang L, Moore A, Liang SH, Ran C. Synthesis-free PET imaging of brown adipose tissue and TSPO via combination of disulfiram and 64CuCl 2. Sci Rep 2017; 7:8298. [PMID: 28811616 PMCID: PMC5557754 DOI: 10.1038/s41598-017-09018-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/14/2017] [Indexed: 12/21/2022] Open
Abstract
PET imaging is a widely applicable but a very expensive technology. On-site synthesis is one important contributor to the high cost. In this report, we demonstrated the feasibility of a synthesis-free method for PET imaging of brown adipose tissue (BAT) and translocator protein 18 kDa (TSPO) via a combination of disulfiram, an FDA approved drug for alcoholism, and 64CuCl2 (termed 64Cu-Dis). In this method, a step-wise injection protocol of 64CuCl2 and disulfiram was used to accomplish the purpose of synthesis-free. Specifically, disulfiram, an inactive 64Cu ligand, was first injected to allow it to metabolize into diethyldithiocarbamate (DDC), a strong 64Cu ligand, which can chelate 64CuCl2 from the following injection to form the actual PET tracer in situ. Our blocking studies, western blot, and tissue histological imaging suggested that the observed BAT contrast was due to 64Cu-Dis binding to TSPO, which was further confirmed as a specific biomarker for BAT imaging using [18F]-F-DPA, a TSPO-specific PET tracer. Our studies, for the first time, demonstrated that TSPO could serve as a potential imaging biomarker for BAT. We believe that our strategy could be extended to other targets while significantly reducing the cost of PET imaging.
Collapse
Affiliation(s)
- Jing Yang
- Molecular Imaging Laboratory, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston, MA, 01890, USA.,College of Pharmaceutical Sciences, Soochow University, Suzhou, 215006, China
| | - Jian Yang
- Molecular Imaging Laboratory, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston, MA, 01890, USA.,School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Lu Wang
- Division of Nuclear Medicine and Molecular Imaging & Gordon Center for Medical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Anna Moore
- Molecular Imaging Laboratory, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston, MA, 01890, USA
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging & Gordon Center for Medical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Chongzhao Ran
- Molecular Imaging Laboratory, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital/Harvard Medical School, Boston, MA, 01890, USA.
| |
Collapse
|
15
|
Cao B, Yang X, Chen Y, Huang Q, Wu Y, Gu Q, Xiao J, Yang H, Pan H, Chen J, Sun Y, Ren L, Zhao C, Deng Y, Yang Y, Chang X, Yang Z, Zhang Y, Niu Z, Wang J, Wu X, Wang J, Jiang Y. Identification of novel ATP7A mutations and prenatal diagnosis in Chinese patients with Menkes disease. Metab Brain Dis 2017; 32:1123-1131. [PMID: 28397151 DOI: 10.1007/s11011-017-9985-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/28/2017] [Indexed: 11/28/2022]
Abstract
Menkes disease (MD) is a fatal X-linked multisystem disease caused by mutations in ATP7A. In this study, clinical and genetic analysis was performed in 24 male MD patients. Development delay, seizures, kinky coarse hair, and dystonia were found in 24, 22, 24, and 24 patients, respectively. Serum ceruloplasmin/copper tested in 19 patients was low. Abnormal classic features of MD presented in the MRI/MRA of 19 patients. Seventeen mutations of ATP7A were identified in 22 patients. Twelve were novel mutations including three small deletion/insertion, one missense mutation, two nonsense mutations, three splicing-site mutations, and three gross deletions. Twenty-two patients were genetically diagnosed; neither point mutation nor deletion/duplication was found in two of them. c.2179G > A found in five patients might be a hot-spot mutation. Prenatal molecular diagnosis was performed for five unrelated fetuses (1 female and 4 male), which found four fetuses to be wild type and one male carried the same mutation as the proband. This study of the largest sample of Chinese MD patients examined to date discovered the unique phenotype and genotype spectrum in Chinese patients with 12 novel mutations of ATP7A, and that c.2179G > A might be a hot-spot mutation in MD patients. Five successful prenatal diagnosis contributed important information for MD families.
Collapse
Affiliation(s)
- Binbin Cao
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
| | - Xiaoping Yang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China
| | - Yinyin Chen
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China
| | - Qionghui Huang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
- Department of Pediatrics, Peking University People's Hospital, Beijing, 100044, China
| | - Ye Wu
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
| | - Qiang Gu
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
| | - Jiangxi Xiao
- Department of Radiology, Peking University First Hospital, Beijing, 100034, China
| | - Huixia Yang
- Department of Obstetrics, Peking University First Hospital, Beijing, 100034, China
| | - Hong Pan
- Department of Central Lab, Peking University First Hospital, Beijing, 100034, China
| | - Junya Chen
- Department of Obstetrics, Peking University First Hospital, Beijing, 100034, China
| | - Yu Sun
- Department of Obstetrics, Peking University First Hospital, Beijing, 100034, China
| | - Li Ren
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China
| | - Chengfeng Zhao
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
- Department of Epilepsy, Central Hospital of Jiamusi, Jiamusi University, Jiamusi, Heilongjiang Province, 154007, China
| | - Yanhua Deng
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China
| | - Yanling Yang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
| | - Xingzhi Chang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
| | - Zhixian Yang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
| | - Yuehua Zhang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
| | - Zhengping Niu
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China
| | - Juli Wang
- Department of Epilepsy, Central Hospital of Jiamusi, Jiamusi University, Jiamusi, Heilongjiang Province, 154007, China
| | - Xiru Wu
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China
| | - Jingmin Wang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China.
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, No. 1 Xi'anmen Street, West District, Beijing, 100034, China.
| |
Collapse
|
16
|
Xie F, Xi Y, Pascual JM, Muzik O, Peng F. Age-dependent changes of cerebral copper metabolism in Atp7b -/- knockout mouse model of Wilson's disease by [ 64Cu]CuCl 2-PET/CT. Metab Brain Dis 2017; 32:717-726. [PMID: 28130615 PMCID: PMC5573586 DOI: 10.1007/s11011-017-9956-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 01/18/2017] [Indexed: 12/29/2022]
Abstract
Copper is a nutritional metal required for brain development and function. Wilson's disease (WD), or hepatolenticular degeneration, is an inherited human copper metabolism disorder caused by a mutation of the ATP7B gene. Many WD patients present with variable neurological and psychiatric symptoms, which may be related to neurodegeneration secondary to copper metabolism imbalance. The objective of this study was to explore the feasibility and use of copper-64 chloride ([64C]CuCl2) as a tracer for noninvasive assessment of age-dependent changes of cerebral copper metabolism in WD using an Atp7b -/- knockout mouse model of WD and positron emission tomography/computed tomography (PET/CT) imaging. Continuing from our recent study of biodistribution and radiation dosimetry of [64C]CuCl2 in Atp7b -/- knockout mice, PET quantitative analysis revealed low 64Cu radioactivity in the brains of Atp7b -/- knockout mice at 7th weeks of age, compared with 64Cu radioactivity in the brains of age- and gender-matched wild type C57BL/6 mice, at 24 h (h) post intravenous injection of [64C]CuCl2 as a tracer. Furthermore, age-dependent increase of 64Cu radioactivity was detected in the brains of Atp7b -/- knockout mice from the 13th to 21th weeks of age, based on the data derived from a longitudinal [64C]CuCl2-PET/CT study of Atp7b -/- knockout mice with orally administered [64Cu]CuCl2 as a tracer. The findings of this study support clinical use of [64Cu]CuCl2-PET/CT imaging as a tool for noninvasive assessment of age-dependent changes of cerebral copper metabolism in WD patients presenting with variable neurological and psychiatric symptoms.
Collapse
Affiliation(s)
- Fang Xie
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-9140, USA
| | - Yin Xi
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-9140, USA
| | - Juan M Pascual
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Otto Muzik
- Carman & Ann Adams Department of Pediatrics, Wayne State University, Detroit, MI, USA
- Department of Radiology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Fangyu Peng
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-9140, USA.
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
17
|
Bremer PT, Pellett S, Carolan JP, Tepp WH, Eubanks LM, Allen KN, Johnson EA, Janda KD. Metal Ions Effectively Ablate the Action of Botulinum Neurotoxin A. J Am Chem Soc 2017; 139:7264-7272. [PMID: 28475321 PMCID: PMC5612488 DOI: 10.1021/jacs.7b01084] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Botulinum neurotoxin serotype A (BoNT/A) causes a debilitating and potentially fatal illness known as botulism. The toxin is also a bioterrorism threat, yet no pharmacological antagonist to counteract its effects has reached clinical approval. Existing strategies to negate BoNT/A intoxication have looked to antibodies, peptides, or organic small molecules as potential therapeutics. In this work, a departure from the traditional drug discovery mindset was pursued, in which the enzyme's susceptibility to metal ions was exploited. A screen of a series of metal salts showed marked inhibitory activity of group 11 and 12 metals against the BoNT/A light chain (LC) protease. Enzyme kinetics revealed that copper (I) and (II) cations displayed noncompetitive inhibition of the LC (Ki ≈ 1 μM), while mercury (II) cations were 10-fold more potent. Crystallographic and mutagenesis studies elucidated a key binding interaction between Cys165 on BoNT/A LC and the inhibitory metals. As potential copper prodrugs, ligand-copper complexes were examined in a cell-based model and were found to prevent BoNT/A cleavage of the endogenous protein substrate, SNAP-25, even at low μM concentrations of complexes. Further investigation of the complexes suggested a bioreductive mechanism causing intracellular release of copper, which directly inhibited the BoNT/A protease. In vivo experiments demonstrated that copper (II) dithiocarbamate and bis(thiosemicarbazone) complexes could delay BoNT/A-mediated lethality in a rodent model, indicating their potential for treating the harmful effects of BoNT/A intoxication. Our studies illustrate that metals can be therapeutically viable enzyme inhibitors; moreover, enzymes that share homology with BoNT LCs may be similarly targeted with metals.
Collapse
Affiliation(s)
- Paul T. Bremer
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sabine Pellett
- Department of Bacteriology, University of Wisconsin, 1550 Linden Drive, Madison, Wisconsin 53706, USA
| | - James P. Carolan
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, MA 02215, USA
| | - William H. Tepp
- Department of Bacteriology, University of Wisconsin, 1550 Linden Drive, Madison, Wisconsin 53706, USA
| | - Lisa M. Eubanks
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Karen N. Allen
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, MA 02215, USA
| | - Eric A. Johnson
- Department of Bacteriology, University of Wisconsin, 1550 Linden Drive, Madison, Wisconsin 53706, USA
| | - Kim D. Janda
- Departments of Chemistry and Immunology, The Skaggs Institute for Chemical Biology, Worm Institute of Research and Medicine (WIRM), The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
18
|
Bhattacharjee A, Yang H, Duffy M, Robinson E, Conrad-Antoville A, Lu YW, Capps T, Braiterman L, Wolfgang M, Murphy MP, Yi L, Kaler SG, Lutsenko S, Ralle M. The Activity of Menkes Disease Protein ATP7A Is Essential for Redox Balance in Mitochondria. J Biol Chem 2016; 291:16644-58. [PMID: 27226607 PMCID: PMC4974379 DOI: 10.1074/jbc.m116.727248] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Indexed: 11/13/2022] Open
Abstract
Copper-transporting ATPase ATP7A is essential for mammalian copper homeostasis. Loss of ATP7A activity is associated with fatal Menkes disease and various other pathologies. In cells, ATP7A inactivation disrupts copper transport from the cytosol into the secretory pathway. Using fibroblasts from Menkes disease patients and mouse 3T3-L1 cells with a CRISPR/Cas9-inactivated ATP7A, we demonstrate that ATP7A dysfunction is also damaging to mitochondrial redox balance. In these cells, copper accumulates in nuclei, cytosol, and mitochondria, causing distinct changes in their redox environment. Quantitative imaging of live cells using GRX1-roGFP2 and HyPer sensors reveals highest glutathione oxidation and elevation of H2O2 in mitochondria, whereas the redox environment of nuclei and the cytosol is much less affected. Decreasing the H2O2 levels in mitochondria with MitoQ does not prevent glutathione oxidation; i.e. elevated copper and not H2O2 is a primary cause of glutathione oxidation. Redox misbalance does not significantly affect mitochondrion morphology or the activity of respiratory complex IV but markedly increases cell sensitivity to even mild glutathione depletion, resulting in loss of cell viability. Thus, ATP7A activity protects mitochondria from excessive copper entry, which is deleterious to redox buffers. Mitochondrial redox misbalance could significantly contribute to pathologies associated with ATP7A inactivation in tissues with paradoxical accumulation of copper (i.e. renal epithelia).
Collapse
Affiliation(s)
| | | | - Megan Duffy
- the Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239
| | - Emily Robinson
- the Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239
| | - Arianrhod Conrad-Antoville
- the Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239
| | | | - Tony Capps
- the Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239
| | | | - Michael Wolfgang
- Cell Biology Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Michael P Murphy
- the Medical Research Council Mitochondrial Biology Unit, Cambridge CB2 0XY, United Kingdom, and
| | - Ling Yi
- the Section on Translational Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Stephen G Kaler
- the Section on Translational Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | | | - Martina Ralle
- the Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239,
| |
Collapse
|
19
|
Torres JB, Andreozzi EM, Dunn JT, Siddique M, Szanda I, Howlett DR, Sunassee K, Blowera PJ. PET Imaging of Copper Trafficking in a Mouse Model of Alzheimer Disease. J Nucl Med 2016; 57:109-14. [PMID: 26449834 PMCID: PMC6207347 DOI: 10.2967/jnumed.115.162370] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 09/28/2015] [Indexed: 01/23/2023] Open
Abstract
UNLABELLED Alzheimer disease (AD) is a fatal neurodegenerative disorder characterized by progressive neuronal loss and cognitive decline. The lack of reliable and objective diagnostic markers for AD hampers early disease detection and treatment. Growing evidence supports the existence of a dysregulation in brain copper trafficking in AD. The aim of this study was to investigate brain copper trafficking in a transgenic mouse model of AD by PET imaging with (64)Cu, to determine its potential as a diagnostic biomarker of the disorder. METHODS Brain copper trafficking was evaluated in 6- to 8-mo-old TASTPM transgenic mice and age-matched wild-type controls using the (64)Cu bis(thiosemicarbazone) complex (64)Cu-GTSM (glyoxalbis(N(4)-methyl-3-thiosemicarbazonato) copper(II)), which crosses the blood-brain barrier and releases (64)Cu bioreductively into cells. Animals were intravenously injected with (64)Cu-GTSM and imaged at 0-30 min and 24-25 h after injection. The images were analyzed by atlas-based quantification and texture analysis. Regional distribution of (64)Cu in the brain 24 h after injection was also evaluated via ex vivo autoradiography and compared with amyloid-β plaque deposition in TASTPM mice. RESULTS Compared with controls, in TASTPM mice PET image analysis demonstrated significantly increased (by a factor of ~1.3) brain concentration of (64)Cu at 30 min (P < 0.01) and 24 h (P < 0.05) after injection of the tracer and faster (by a factor of ~5) (64)Cu clearance from the brain (P < 0.01). Atlas-based quantification and texture analysis revealed significant differences in regional brain uptake of (64)Cu and PET image heterogeneity between the 2 groups of mice. Ex vivo autoradiography showed that regional brain distribution of (64)Cu at 24 h after injection did not correlate with amyloid-β plaque distribution in TASTPM mice. CONCLUSION The trafficking of (64)Cu in the brain after administration of (64)Cu-GTSM is significantly altered by AD-like pathology in the TASTPM mouse model, suggesting that (64)Cu-GTSM PET imaging warrants clinical evaluation as a diagnostic tool for AD and possibly other neurodegenerative disorders.
Collapse
Affiliation(s)
- Julia Baguña Torres
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, St. Thomas’ Hospital, London, SE1 7EH, UK
| | - Erica M. Andreozzi
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, St. Thomas’ Hospital, London, SE1 7EH, UK
| | - Joel T. Dunn
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, St. Thomas’ Hospital, London, SE1 7EH, UK
| | - Muhammad Siddique
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, St. Thomas’ Hospital, London, SE1 7EH, UK
| | - Istvan Szanda
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, St. Thomas’ Hospital, London, SE1 7EH, UK
| | - David R. Howlett
- King’s College London, Wolfson Centre for Age-Related Diseases, Hodgkin Building, Guy’s Campus, London, SE1 1UL, UK
| | - Kavitha Sunassee
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, St. Thomas’ Hospital, London, SE1 7EH, UK
| | - Philip J. Blowera
- King’s College London, Division of Imaging Sciences and Biomedical Engineering, St. Thomas’ Hospital, London, SE1 7EH, UK
| |
Collapse
|
20
|
Helsel ME, Franz KJ. Pharmacological activity of metal binding agents that alter copper bioavailability. Dalton Trans 2015; 44:8760-70. [PMID: 25797044 PMCID: PMC4425619 DOI: 10.1039/c5dt00634a] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Iron, copper and zinc are required nutrients for many organisms but also potent toxins if misappropriated. An overload of any of these metals can be cytotoxic and ultimately lead to organ failure, whereas deficiencies can result in anemia, weakened immune system function, and other medical conditions. Cellular metal imbalances have been implicated in neurodegenerative diseases, cancer and infection. It is therefore critical for living organisms to maintain careful control of both the total levels and subcellular distributions of these metals to maintain healthy function. This perspective explores several strategies envisioned to alter the bioavailability of metal ions by using synthetic metal-binding agents targeted for diseases where misappropriated metal ions are suspected of exacerbating cellular damage. Specifically, we discuss chemical properties that influence the pharmacological outcome of a subset of metal-binding agents known as ionophores, and review several examples that have shown multiple pharmacological activities in metal-related diseases, with a specific focus on copper.
Collapse
Affiliation(s)
- Marian E Helsel
- Duke University, Department of Chemistry, French Family Science Center, 124 Science Drive, 22708, Durham, NC, USA.
| | | |
Collapse
|
21
|
Hoshi Y, Tani N, Tabata H, Wakamatsu S, Munakata M, Maruyama K, Kodama H, Oshitari T, Natsugari H, Takahashi H. Development of a Therapeutic Agent for Menkes Disease: Solubilization of a Copper-Disulfiram Complex. YAKUGAKU ZASSHI 2015; 135:493-9. [DOI: 10.1248/yakushi.14-00188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
22
|
Guo W, Sun X, Jacobson O, Yan X, Min K, Srivatsan A, Niu G, Kiesewetter DO, Chang J, Chen X. Intrinsically radioactive [64Cu]CuInS/ZnS quantum dots for PET and optical imaging: improved radiochemical stability and controllable Cerenkov luminescence. ACS NANO 2015; 9:488-95. [PMID: 25549258 PMCID: PMC4310640 DOI: 10.1021/nn505660r] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 12/30/2014] [Indexed: 05/20/2023]
Abstract
Functionalized quantum dots (QDs) have been widely explored for multimodality bioimaging and proven to be versatile agents. Attaching positron-emitting radioisotopes onto QDs not only endows their positron emission tomography (PET) functionality, but also results in self-illuminating QDs, with no need for an external light source, by Cerenkov resonance energy transfer (CRET). Traditional chelation methods have been used to incorporate the radionuclide, but these methods are compromised by the potential for loss of radionuclide due to cleavage of the linker between particle and chelator, decomplexation of the metal, and possible altered pharmacokinetics of nanomaterials. Herein, we described a straightforward synthesis of intrinsically radioactive [(64)Cu]CuInS/ZnS QDs by directly incorporating (64)Cu into CuInS/ZnS nanostructure with (64)CuCl2 as synthesis precursor. The [(64)Cu]CuInS/ZnS QDs demonstrated excellent radiochemical stability with less than 3% free (64)Cu detected even after exposure to serum containing EDTA (5 mM) for 24 h. PEGylation can be achieved in situ during synthesis, and the PEGylated radioactive QDs showed high tumor uptake (10.8% ID/g) in a U87MG mouse xenograft model. CRET efficiency was studied as a function of concentration and (64)Cu radioactivity concentration. These [(64)Cu]CuInS/ZnS QDs were successfully applied as an efficient PET/self-illuminating luminescence in vivo imaging agents.
Collapse
Affiliation(s)
- Weisheng Guo
- School of Materials Science and Engineering, School of Life Sciences, Tianjin University, Tianjin 300072, China
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Xiaolian Sun
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
- Address correspondence to , ,
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Xuefeng Yan
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Kyunghyun Min
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Avinash Srivatsan
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Dale O. Kiesewetter
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Jin Chang
- School of Materials Science and Engineering, School of Life Sciences, Tianjin University, Tianjin 300072, China
- Address correspondence to , ,
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
- Address correspondence to , ,
| |
Collapse
|
23
|
Gao Y, Li Y, Yang X, He F, Huang J, Jiang M, Zhou Z, Chen H. Design, synthesis and biological evaluation of a novel Cu2+-selective fluorescence sensor for bio-detection and chelation. RSC Adv 2015. [DOI: 10.1039/c5ra12620g] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A new coumarin-based Cu2+-selective fluorescent sensor was designed and synthesized and the ability of this fluorescent sensor for the detection and chelation of Cu2+ in cultured cells was investigated.
Collapse
Affiliation(s)
- Yu Gao
- College of Chemistry
- Fuzhou University
- Fuzhou
- China
| | - Yazhen Li
- College of Chemistry
- Fuzhou University
- Fuzhou
- China
| | - Xiping Yang
- College of Chemistry
- Fuzhou University
- Fuzhou
- China
| | - Fangfei He
- College of Chemistry
- Fuzhou University
- Fuzhou
- China
| | - Jiamei Huang
- College of Chemistry
- Fuzhou University
- Fuzhou
- China
| | | | - Zaihui Zhou
- College of Chemistry
- Fuzhou University
- Fuzhou
- China
| | - Haijun Chen
- College of Chemistry
- Fuzhou University
- Fuzhou
- China
| |
Collapse
|