1
|
Leiva-Salcedo E, Riquelme D, Huidobro-Toro JP, Coddou C. Copper Increases the Cooperative Gating of Rat P2X2a Receptor Channels. Pharmaceuticals (Basel) 2024; 17:1590. [PMID: 39770432 PMCID: PMC11678522 DOI: 10.3390/ph17121590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: P2X receptor channels are widely expressed in the CNS, where they have multiple functions in health and disease. The rat P2X2a (rP2X2a) receptor channel is modulated by copper, an essential trace element that plays important roles in synaptic modulation and neurodegenerative disorders. Although essential extracellular amino acids that coordinate copper have been identified, the exact mechanism of copper-induced modulation has not been yet elucidated. Methods: We used HEK293T cells expressing rP2X2a channel(s) and performed outside-out single-channel and whole-cell recordings to explore copper's effects on rP2X2 currents and determine whether this metal can increase the cooperative gating of rP2X2a channel. Results: In whole-cell recordings and in patches containing 2 or 3 rP2X2a channels, copper enhanced the ATP-induced currents, significantly reducing the ATP EC50 and increasing the Hill coefficient. Moreover, copper increased the apparent Po in patches containing two or three channels. By contrast, in patches containing only one rP2X2a channel, we did not observe any significant changes in ATP EC50, the Hill coefficient, or Po. Conclusions: Copper modulates the gating of rP2X2a channels, enhancing interchannel cooperativity without altering single-channel conductance or Po. This novel regulatory mechanism could be relevant for understanding the role of P2X2 channels in physiological and pathological processes.
Collapse
Affiliation(s)
- Elias Leiva-Salcedo
- Department of Biology, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago 9170022, Chile; (E.L.-S.); (D.R.)
| | - Denise Riquelme
- Department of Biology, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago 9170022, Chile; (E.L.-S.); (D.R.)
| | - Juan Pablo Huidobro-Toro
- Department of Biology, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago 9170022, Chile; (E.L.-S.); (D.R.)
| | - Claudio Coddou
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo 1781421, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8331150, Chile
| |
Collapse
|
2
|
Behar AE, Maayan G. A cocktail of Cu 2+- and Zn 2+-peptoid-based chelators can stop ROS formation for Alzheimer's disease therapy. Chem Sci 2024:d4sc04313h. [PMID: 39464602 PMCID: PMC11503657 DOI: 10.1039/d4sc04313h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
The formation of reactive oxygen species (ROS) in the brain is a major cause of neuropathologic degradation associated with Alzheimer's Disease (AD). It has been suggested that the copper (Cu)-amyloid-β (Aβ) peptide complex can lead to ROS formation in the brain. An external chelator for Cu that can extract Cu from the CuAβ complex should inhibit the formation of ROS, making Cu chelation an excellent therapeutic approach for AD. Such a chelator should possess high selectivity for Cu over zinc (Zn), which is also present within the synaptic cleft. However, such selectivity is generally hard to achieve in one molecule due to the similarities in the binding preferences of these two metal ions. As an alternative to monotherapy (where Cu extraction is performed using a single chelator), herein we describe a variation of combination therapy - a novel cocktail approach, which is based on the co-administration of two structurally different peptidomimetic chelators, aiming to target both Cu2+ and Zn2+ ions simultaneously but independently from each other. Based on rigorous spectroscopic experiments, we demonstrate that our peptidomimetic cocktail allows, for the first time, the complete and immediate inhibition of ROS production by the CuAβ complex in the presence of Zn2+. In addition, we further demonstrate the high stability of the cocktail under simulated physiological conditions and its resistance to proteolytic degradation by trypsin and report the water/n-octanol partition coefficient, initially assessing the blood-brain barrier (BBB) permeability potential of the chelators.
Collapse
Affiliation(s)
- Anastasia E Behar
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Technion City 3200008 Haifa Israel
| | - Galia Maayan
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Technion City 3200008 Haifa Israel
| |
Collapse
|
3
|
Teng X, Stefaniak E, Willison KR, Ying L. Interplay between Copper, Phosphatidylserine, and α-Synuclein Suggests a Link between Copper Homeostasis and Synaptic Vesicle Cycling. ACS Chem Neurosci 2024; 15:2884-2896. [PMID: 39013013 PMCID: PMC11311125 DOI: 10.1021/acschemneuro.4c00280] [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: 05/04/2024] [Revised: 06/14/2024] [Accepted: 07/02/2024] [Indexed: 07/18/2024] Open
Abstract
Copper homeostasis is critical to the functioning of the brain, and its breakdown is linked with many brain diseases. Copper is also known to interact with the negatively charged lipid, phosphatidylserine (PS), as well as α-synuclein, an aggregation-prone protein enriched in the synapse, which plays a role in synaptic vesicle docking and fusion. However, the interplay between copper, PS lipid, and α-synuclein is not known. Herein, we report a detailed and predominantly kinetic study of the interactions among these three components pertinent to copper homeostasis and neurotransmission. We found that synaptic vesicle-mimicking small unilamellar vesicles (SUVs) can sequester any excess free Cu2+ within milliseconds, and bound Cu2+ on SUVs can be reduced to Cu+ by GSH at a nearly constant rate under physiological conditions. Moreover, we revealed that SUV-bound Cu2+ does not affect the binding between wild-type α-synuclein and SUVs but affect that between N-terminal acetylated α-synuclein and SUVs. In contrast, Cu2+ can effectively displace both types of α-synuclein from the vesicles. Our results suggest that synaptic vesicles may mediate copper transfer in the brain, while copper could participate in synaptic vesicle docking to the plasma membrane via its regulation of the interaction between α-synuclein and synaptic vesicle.
Collapse
Affiliation(s)
- Xiangyu Teng
- Department
of Chemistry, Imperial College London, Molecular
Sciences Research Hub, 82 Wood Lane, London W12
0BZ, U.K.
| | - Ewelina Stefaniak
- National
Heart and Lung Institute, Imperial College
London, Molecular Sciences Research Hub, 82 Wood Lane, London W12 0BZ, U.K.
| | - Keith R. Willison
- Department
of Chemistry, Imperial College London, Molecular
Sciences Research Hub, 82 Wood Lane, London W12
0BZ, U.K.
| | - Liming Ying
- National
Heart and Lung Institute, Imperial College
London, Molecular Sciences Research Hub, 82 Wood Lane, London W12 0BZ, U.K.
| |
Collapse
|
4
|
Gale J, Aizenman E. The physiological and pathophysiological roles of copper in the nervous system. Eur J Neurosci 2024; 60:3505-3543. [PMID: 38747014 PMCID: PMC11491124 DOI: 10.1111/ejn.16370] [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: 11/12/2023] [Revised: 02/28/2024] [Accepted: 04/10/2024] [Indexed: 07/06/2024]
Abstract
Copper is a critical trace element in biological systems due the vast number of essential enzymes that require the metal as a cofactor, including cytochrome c oxidase, superoxide dismutase and dopamine-β-hydroxylase. Due its key role in oxidative metabolism, antioxidant defence and neurotransmitter synthesis, copper is particularly important for neuronal development and proper neuronal function. Moreover, increasing evidence suggests that copper also serves important functions in synaptic and network activity, the regulation of circadian rhythms, and arousal. However, it is important to note that because of copper's ability to redox cycle and generate reactive species, cellular levels of the metal must be tightly regulated to meet cellular needs while avoiding copper-induced oxidative stress. Therefore, it is essential that the intricate system of copper transporters, exporters, copper chaperones and copper trafficking proteins function properly and in coordinate fashion. Indeed, disorders of copper metabolism such as Menkes disease and Wilson disease, as well as diseases linked to dysfunction of copper-requiring enzymes, such as SOD1-linked amyotrophic lateral sclerosis, demonstrate the dramatic neurological consequences of altered copper homeostasis. In this review, we explore the physiological importance of copper in the nervous system as well as pathologies related to improper copper handling.
Collapse
Affiliation(s)
- Jenna Gale
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Elias Aizenman
- Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
5
|
Kawahara M, Tanaka KI, Kato-Negishi M. Zinc, Copper, and Calcium: A Triangle in the Synapse for the Pathogenesis of Vascular-Type Senile Dementia. Biomolecules 2024; 14:773. [PMID: 39062487 PMCID: PMC11274390 DOI: 10.3390/biom14070773] [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: 06/05/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Zinc (Zn) and copper (Cu) are essential for normal brain functions. In particular, Zn and Cu are released to synaptic clefts during neuronal excitation. Synaptic Zn and Cu regulate neuronal excitability, maintain calcium (Ca) homeostasis, and play central roles in memory formation. However, in pathological conditions such as transient global ischemia, excess Zn is secreted to synaptic clefts, which causes neuronal death and can eventually trigger the pathogenesis of a vascular type of senile dementia. We have previously investigated the characteristics of Zn-induced neurotoxicity and have demonstrated that low concentrations of Cu can exacerbate Zn neurotoxicity. Furthermore, during our pharmacological approaches to clarify the molecular pathways of Cu-enhanced Zn-induced neurotoxicity, we have revealed the involvement of Ca homeostasis disruption. In the present review, we discuss the roles of Zn and Cu in the synapse, as well as the crosstalk between Zn, Cu, and Ca, which our study along with other recent studies suggest may underlie the pathogenesis of vascular-type senile dementia.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi 202-8585, Tokyo, Japan
| | | | | |
Collapse
|
6
|
Min JH, Sarlus H, Harris RA. Glycyl-l-histidyl-l-lysine prevents copper- and zinc-induced protein aggregation and central nervous system cell death in vitro. Metallomics 2024; 16:mfae019. [PMID: 38599632 PMCID: PMC11135135 DOI: 10.1093/mtomcs/mfae019] [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: 10/09/2023] [Accepted: 04/09/2024] [Indexed: 04/12/2024]
Abstract
Common features of neurodegenerative diseases are oxidative and inflammatory imbalances as well as the misfolding of proteins. An excess of free metal ions can be pathological and contribute to cell death, but only copper and zinc strongly promote protein aggregation. Herein we demonstrate that the endogenous copper-binding tripeptide glycyl-l-histidyl-l-lysine (GHK) has the ability to bind to and reduce copper redox activity and to prevent copper- and zinc-induced cell death in vitro. In addition, GHK prevents copper- and zinc-induced bovine serum albumin aggregation and reverses aggregation through resolubilizing the protein. We further demonstrate the enhanced toxicity of copper during inflammation and the ability of GHK to attenuate this toxicity. Finally, we investigated the effects of copper on enhancing paraquat toxicity and report a protective effect of GHK. We therefore conclude that GHK has potential as a cytoprotective compound with regard to copper and zinc toxicity, with positive effects on protein solubility and aggregation that warrant further investigation in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jin-Hong Min
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, S-171 76 Stockholm, Sweden
| | - Heela Sarlus
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, S-171 76 Stockholm, Sweden
| | - Robert A Harris
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, S-171 76 Stockholm, Sweden
| |
Collapse
|
7
|
Tian Y, Shang Q, Liang R, Viles JH. Copper(II) Can Kinetically Trap Arctic and Italian Amyloid-β 40 as Toxic Oligomers, Mimicking Cu(II) Binding to Wild-Type Amyloid-β 42: Implications for Familial Alzheimer's Disease. JACS AU 2024; 4:578-591. [PMID: 38425915 PMCID: PMC10900208 DOI: 10.1021/jacsau.3c00687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 03/02/2024]
Abstract
The self-association of amyloid-β (Aβ) peptide into neurotoxic oligomers is believed to be central to Alzheimer's disease (AD). Copper is known to impact Aβ assembly, while disrupted copper homeostasis impacts phenotype in Alzheimer's models. Here we show the presence of substoichiometric Cu(II) has very different impacts on the assembly of Aβ40 and Aβ42 isoforms. Globally fitting microscopic rate constants for fibril assembly indicates copper will accelerate fibril formation of Aβ40 by increasing primary nucleation, while seeding experiments confirm that elongation and secondary nucleation rates are unaffected by Cu(II). In marked contrast, Cu(II) traps Aβ42 as prefibrillar oligomers and curvilinear protofibrils. Remarkably, the Cu(II) addition to preformed Aβ42 fibrils causes the disassembly of fibrils back to protofibrils and oligomers. The very different behaviors of the two Aβ isoforms are centered around differences in their fibril structures, as highlighted by studies of C-terminally amidated Aβ42. Arctic and Italian familiar mutations also support a key role for fibril structure in the interplay of Cu(II) with Aβ40/42 isoforms. The Cu(II) dependent switch in behavior between nonpathogenic Aβ40 wild-type and Aβ40 Arctic or Italian mutants suggests heightened neurotoxicity may be linked to the impact of physiological Cu(II), which traps these familial mutants as oligomers and curvilinear protofibrils, which cause membrane permeability and Ca(II) cellular influx.
Collapse
Affiliation(s)
- Yao Tian
- School of Biological and Behavioral
Sciences, Queen Mary University of London, London E1 4NS, U.K.
| | - Qi Shang
- School of Biological and Behavioral
Sciences, Queen Mary University of London, London E1 4NS, U.K.
| | - Ruina Liang
- School of Biological and Behavioral
Sciences, Queen Mary University of London, London E1 4NS, U.K.
| | - John H. Viles
- School of Biological and Behavioral
Sciences, Queen Mary University of London, London E1 4NS, U.K.
| |
Collapse
|
8
|
do Amaral MJ, Mohapatra S, Passos AR, Lopes da Silva TS, Carvalho RS, da Silva Almeida M, Pinheiro AS, Wegmann S, Cordeiro Y. Copper drives prion protein phase separation and modulates aggregation. SCIENCE ADVANCES 2023; 9:eadi7347. [PMID: 37922348 PMCID: PMC10624353 DOI: 10.1126/sciadv.adi7347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/03/2023] [Indexed: 11/05/2023]
Abstract
Prion diseases are characterized by prion protein (PrP) transmissible aggregation and neurodegeneration, which has been linked to oxidative stress. The physiological function of PrP seems related to sequestering of redox-active Cu2+, and Cu2+ dyshomeostasis is observed in prion disease brain. It is unclear whether Cu2+ contributes to PrP aggregation, recently shown to be mediated by PrP condensation. This study indicates that Cu2+ promotes PrP condensation in live cells at the cell surface and in vitro through copartitioning. Molecularly, Cu2+ inhibited PrP β-structure and hydrophobic residues exposure. Oxidation, induced by H2O2, triggered liquid-to-solid transition of PrP:Cu2+ condensates and promoted amyloid-like PrP aggregation. In cells, overexpression of PrPC initially protected against Cu2+ cytotoxicity but led to PrPC aggregation upon extended copper exposure. Our data suggest that PrP condensates function as a buffer for copper that prevents copper toxicity but can transition into PrP aggregation at prolonged oxidative stress.
Collapse
Affiliation(s)
- Mariana Juliani do Amaral
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | | | - Aline Ribeiro Passos
- Brazilian Synchrotron Light Laboratory (LNLS), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | | | | | - Marcius da Silva Almeida
- Plataforma Avançada de Biomoléculas, Centro Nacional de Biologia Estrutural e Bioimagem, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anderson Sá Pinheiro
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Susanne Wegmann
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Yraima Cordeiro
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Han J. Copper trafficking systems in cells: insights into coordination chemistry and toxicity. Dalton Trans 2023; 52:15277-15296. [PMID: 37702384 DOI: 10.1039/d3dt02166a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Transition metal ions, such as copper, are indispensable components in the biological system. Copper ions which primarily exist in two major oxidation states Cu(I) and Cu(II) play crucial roles in various cellular processes including antioxidant defense, biosynthesis of neurotransmitters, and energy metabolism, owing to their inherent redox activity. The disturbance in copper homeostasis can contribute to the development of copper metabolism disorders, cancer, and neurodegenerative diseases, highlighting the significance of understanding the copper trafficking system in cellular environments. This review aims to offer a comprehensive overview of copper homeostatic machinery, with an emphasis on the coordination chemistry of copper transporters and trafficking proteins. While copper chaperones and the corresponding metalloenzymes are thoroughly discussed, we also explore the potential existence of low-molecular-mass metal complexes within cellular systems. Furthermore, we summarize the toxicity mechanisms originating from copper deficiency or accumulation, which include the dysregulation of oxidative stress, signaling pathways, signal transduction, and amyloidosis. This perspective review delves into the current knowledge regarding the intricate aspects of the copper trafficking system, providing valuable insights into potential treatment strategies from the standpoint of bioinorganic chemistry.
Collapse
Affiliation(s)
- Jiyeon Han
- Department of Applied Chemistry, University of Seoul, Seoul 02504, Republic of Korea.
| |
Collapse
|
10
|
Gielnik M, Szymańska A, Dong X, Jarvet J, Svedružić ŽM, Gräslund A, Kozak M, Wärmländer SKTS. Prion Protein Octarepeat Domain Forms Transient β-Sheet Structures upon Residue-Specific Binding to Cu(II) and Zn(II) Ions. Biochemistry 2023. [PMID: 37163663 DOI: 10.1021/acs.biochem.3c00129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Misfolding of the cellular prion protein (PrPC) is associated with the development of fatal neurodegenerative diseases called transmissible spongiform encephalopathies (TSEs). Metal ions appear to play a crucial role in PrPC misfolding. PrPC is a combined Cu(II) and Zn(II) metal-binding protein, where the main metal-binding site is located in the octarepeat (OR) region. Thus, the biological function of PrPC may involve the transport of divalent metal ions across membranes or buffering concentrations of divalent metal ions in the synaptic cleft. Recent studies have shown that an excess of Cu(II) ions can result in PrPC instability, oligomerization, and/or neuroinflammation. Here, we have used biophysical methods to characterize Cu(II) and Zn(II) binding to the isolated OR region of PrPC. Circular dichroism (CD) spectroscopy data suggest that the OR domain binds up to four Cu(II) ions or two Zn(II) ions. Binding of the first metal ion results in a structural transition from the polyproline II helix to the β-turn structure, while the binding of additional metal ions induces the formation of β-sheet structures. Fluorescence spectroscopy data indicate that the OR region can bind both Cu(II) and Zn(II) ions at neutral pH, but under acidic conditions, it binds only Cu(II) ions. Molecular dynamics simulations suggest that binding of either metal ion to the OR region results in the formation of β-hairpin structures. As the formation of β-sheet structures can be a first step toward amyloid formation, we propose that high concentrations of either Cu(II) or Zn(II) ions may have a pro-amyloid effect in TSE diseases.
Collapse
Affiliation(s)
- Maciej Gielnik
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, PL 61-614 Poznań, Poland
| | - Aneta Szymańska
- Department of Biomedical Chemistry, Faculty of Chemistry, Gdańsk University, PL 80-308 Gdańsk, Poland
| | - Xiaolin Dong
- Chemistry Section, Stockholm University, 10691 Stockholm, Sweden
| | - Jüri Jarvet
- Chemistry Section, Stockholm University, 10691 Stockholm, Sweden
- The National Institute of Chemical Physics and Biophysics, 12618 Tallinn, Estonia
| | - Željko M Svedružić
- Department of Biotechnology, University of Rijeka, HR 51000 Rijeka, Croatia
| | - Astrid Gräslund
- Chemistry Section, Stockholm University, 10691 Stockholm, Sweden
| | - Maciej Kozak
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, PL 61-614 Poznań, Poland
- National Synchrotron Radiation Centre SOLARIS, Jagiellonian University, PL 30-392 Kraków, Poland
| | | |
Collapse
|
11
|
Kawahara M, Kato-Negishi M, Tanaka KI. Dietary Trace Elements and the Pathogenesis of Neurodegenerative Diseases. Nutrients 2023; 15:2067. [PMID: 37432185 PMCID: PMC10180548 DOI: 10.3390/nu15092067] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 07/12/2023] Open
Abstract
Trace elements such as iron (Fe), zinc (Zn), copper (Cu), and manganese (Mn) are absorbed from food via the gastrointestinal tract, transported into the brain, and play central roles in normal brain functions. An excess of these trace elements often produces reactive oxygen species and damages the brain. Moreover, increasing evidence suggests that the dyshomeostasis of these metals is involved in the pathogenesis of neurodegenerative diseases, including Alzheimer's disease, prion diseases, and Lewy body diseases. The disease-related amyloidogenic proteins can regulate metal homeostasis at the synapses, and thus loss of the protective functions of these amyloidogenic proteins causes neurodegeneration. Meanwhile, metal-induced conformational changes of the amyloidogenic proteins contribute to enhancing their neurotoxicity. Moreover, excess Zn and Cu play central roles in the pathogenesis of vascular-type senile dementia. Here, we present an overview of the intake, absorption, and transport of four essential elements (Fe, Zn, Cu, Mn) and one non-essential element (aluminum: Al) in food and their connections with the pathogenesis of neurodegenerative diseases based on metal-protein, and metal-metal cross-talk.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| | - Midori Kato-Negishi
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| | - Ken-Ichiro Tanaka
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo 202-8585, Japan
| |
Collapse
|
12
|
Kim H, Jo S, Kim IG, Kim RK, Kahm YJ, Jung SH, Lee JH. Effect of Copper Chelators via the TGF-β Signaling Pathway on Glioblastoma Cell Invasion. Molecules 2022; 27:8851. [PMID: 36557987 PMCID: PMC9784955 DOI: 10.3390/molecules27248851] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a fast-growing and aggressive type of brain cancer. Unlike normal brain cells, GBM cells exhibit epithelial-mesenchymal transition (EMT), which is a crucial biological process in embryonic development and cell metastasis, and are highly invasive. Copper reportedly plays a critical role in the progression of a variety of cancers, including brain, breast, and lung cancers. However, excessive copper is toxic to cells. D-penicillamine (DPA) and triethylenetetramine (TETA) are well-known copper chelators and are the mainstay of treatment for copper-associated diseases. Following treatment with copper sulfate and DPA, GBM cells showed inhibition of proliferation and suppression of EMT properties, including reduced expression levels of N-cadherin, E-cadherin, and Zeb, which are cell markers associated with EMT. In contrast, treatment with copper sulfate and TETA yielded the opposite effects in GBM. Genes, including TGF-β, are associated with an increase in copper levels, implying their role in EMT. To analyze the invasion and spread of GBM, we used zebrafish embryos xenografted with the GBM cell line U87. The invasion of GBM cells into zebrafish embryos was markedly inhibited by copper treatment with DPA. Our findings suggest that treatment with copper and DPA inhibits proliferation and EMT through a mechanism involving TGF-β/Smad signaling in GBM. Therefore, DPA, but not TETA, could be used as adjuvant therapy for GBM with high copper concentrations.
Collapse
Affiliation(s)
- Heabin Kim
- Department of Genetic Resources, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea
| | - Seonmi Jo
- Department of Genetic Resources, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea
| | - In-Gyu Kim
- Department of Radiation Biology, Environmental Safety Assessment Research Division, Korea Atomic Energy Research Institute, Daejeon 34057, Republic of Korea
- Department of Radiation Science and Technology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Rae-Kwon Kim
- Department of Radiation Biology, Environmental Safety Assessment Research Division, Korea Atomic Energy Research Institute, Daejeon 34057, Republic of Korea
- Department of Radiation Science and Technology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Yeon-Jee Kahm
- Department of Radiation Biology, Environmental Safety Assessment Research Division, Korea Atomic Energy Research Institute, Daejeon 34057, Republic of Korea
- Department of Radiation Science and Technology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Seung-Hyun Jung
- Department of Genetic Resources, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea
| | - Jei Ha Lee
- Department of Genetic Resources, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea
| |
Collapse
|
13
|
Ng SW, Furman R, Axelsen PH, Shchepinov MS. Free Radical Chain Reactions and Polyunsaturated Fatty Acids in Brain Lipids. ACS OMEGA 2022; 7:25337-25345. [PMID: 35910174 PMCID: PMC9330197 DOI: 10.1021/acsomega.2c02285] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Polyunsaturated fatty acyl chains (PUFAs) concentrate in the brain and give rise to numerous oxidative chemical degradation products. It is widely assumed that these products are the result of free radical chain reactions, and reactions of this type have been demonstrated in preparations where a single PUFA substrate species predominates. However, it is unclear whether such reactions can occur in the biologically complex milieu of lipid membranes where PUFA substrates are a minority species, and where diverse free radical scavengers or other quenching mechanisms are present. It is of particular interest to know whether they occur in brain, where PUFAs are concentrated and where PUFA oxidation products have been implicated in the pathogenesis of neurodegenerative disorders. To ascertain whether free radical chain reactions can occur in a complex brain lipid mixture, mouse brain lipids were extracted, formed into vesicles, and treated with a fixed number of hydroxyl radicals under conditions wherein the concentrations and types of PUFA-containing phospholipids were varied. Specific phospholipid species in the mixture were assayed by tandem mass spectrometry to quantify the oxidative losses of endogenous PUFA-containing phospholipids. Results reveal crosstalk between the oxidative degradation of ω3 and ω6 PUFAs that can only be explained by the occurrence of free radical chain reactions. These results demonstrate that PUFAs in a complex brain lipid mixture can participate in free radical chain reactions wherein the extent of oxidative degradation is not limited by the number of reactive oxygen species available to initiate such reactions. These reactions may help explain otherwise puzzling in vivo interactions between ω3 and ω6 PUFAs in mouse brain.
Collapse
Affiliation(s)
- Sharon
C. W. Ng
- Department
of Pharmacology, 1009C Stellar Chance Laboratories, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6084, United States
| | - Ran Furman
- Department
of Pharmacology, 1009C Stellar Chance Laboratories, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6084, United States
| | - Paul H. Axelsen
- Department
of Pharmacology, 1009C Stellar Chance Laboratories, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6084, United States
| | | |
Collapse
|
14
|
Kawahara M, Tanaka KI, Kato-Negishi M. Crosstalk of copper and zinc in the pathogenesis of vascular dementia. J Clin Biochem Nutr 2022; 71:7-15. [PMID: 35903609 PMCID: PMC9309079 DOI: 10.3164/jcbn.22-40] [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: 04/01/2022] [Accepted: 05/12/2022] [Indexed: 11/23/2022] Open
Abstract
Copper and zinc are essential for normal brain functions. Both are localized in presynaptic vesicles and are secreted into synaptic clefts during neuronal excitation. Despite their significance, excesses of copper and zinc are neurotoxic. In particular, excess zinc after transient global ischemia plays a central role in the ischemia-induced neurodegeneration and pathogenesis of vascular type senile dementia. We previously found that sub-lethal concentrations of copper remarkably exacerbated zinc-induced neurotoxicity, and we investigated the molecular pathways of copper-enhanced zinc-induced neurotoxicity. The endoplasmic reticulum stress pathway, the stress-activated protein kinases/c-Jun amino-terminal kinases pathway, and mitochondrial energy production failure were revealed to be involved in the neurodegenerative processes. Regarding the upstream factors of these pathways, we focused on copper-derived reactive oxygen species and the disruption of calcium homeostasis. Because excess copper and zinc may be present in the synaptic clefts during ischemia, it is possible that secreted copper and copper-induced reactive oxygen species may enhance zinc neurotoxicity and eventually contribute to the pathogenesis of vascular type senile dementia.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Ken-Ichiro Tanaka
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Midori Kato-Negishi
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| |
Collapse
|
15
|
Pfalzer AC, Yan Y, Kang H, Totten M, Silverman J, Bowman AB, Erikson K, Claassen DO. Alterations in metal homeostasis occur prior to canonical markers in Huntington disease. Sci Rep 2022; 12:10373. [PMID: 35725749 PMCID: PMC9209499 DOI: 10.1038/s41598-022-14169-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 06/02/2022] [Indexed: 12/05/2022] Open
Abstract
The importance of metal biology in neurodegenerative diseases such as Huntingtin Disease is well documented with evidence of direct interactions between metals such as copper, zinc, iron and manganese and mutant Huntingtin pathobiology. To date, it is unclear whether these interactions are observed in humans, how this impacts other metals, and how mutant Huntington alters homeostatic mechanisms governing levels of copper, zinc, iron and manganese in cerebrospinal fluid and blood in HD patients. Plasma and cerebrospinal fluid from control, pre-manifest, manifest and late manifest HD participants were collected as part of HD-Clarity. Levels of cerebrospinal fluid and plasma copper, zinc, iron and manganese were measured as well as levels of mutant Huntingtin and neurofilament in a sub-set of cerebrospinal fluid samples. We find that elevations in cerebrospinal fluid copper, manganese and zinc levels are altered early in disease prior to alterations in canonical biomarkers of HD although these changes are not present in plasma. We also evidence that CSF iron is elevated in manifest patients. The relationships between plasma and cerebrospinal fluid metal are altered based on disease stage. These findings demonstrate that there are alterations in metal biology selectively in the CSF which occur prior to changes in known canonical biomarkers of disease. Our work indicates that there are pathological changes related to alterations in metal biology in individuals without elevations in neurofilament and mutant Huntingtin.
Collapse
Affiliation(s)
- Anna C. Pfalzer
- grid.412807.80000 0004 1936 9916Department of Neurology, Vanderbilt University Medical Center, 1611 21st Avenue South, Suite 1532, Nashville, TN 37232 USA
| | - Yan Yan
- grid.412807.80000 0004 1936 9916Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN USA
| | - Hakmook Kang
- grid.412807.80000 0004 1936 9916Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN USA
| | - Melissa Totten
- grid.266860.c0000 0001 0671 255XDepartment of Nutrition, University of North Carolina-Greensboro, Greensboro, NC USA
| | - James Silverman
- grid.412807.80000 0004 1936 9916Department of Neurology, Vanderbilt University Medical Center, 1611 21st Avenue South, Suite 1532, Nashville, TN 37232 USA
| | - Aaron B. Bowman
- grid.169077.e0000 0004 1937 2197School of Health Sciences, Purdue University, West Lafayette, IN USA
| | - Keith Erikson
- grid.266860.c0000 0001 0671 255XDepartment of Nutrition, University of North Carolina-Greensboro, Greensboro, NC USA
| | - Daniel O. Claassen
- grid.412807.80000 0004 1936 9916Department of Neurology, Vanderbilt University Medical Center, 1611 21st Avenue South, Suite 1532, Nashville, TN 37232 USA
| |
Collapse
|
16
|
Abstract
Amyloids are protein aggregates bearing a highly ordered cross β structural motif, which may be functional but are mostly pathogenic. Their formation, deposition in tissues and consequent organ dysfunction is the central event in amyloidogenic diseases. Such protein aggregation may be brought about by conformational changes, and much attention has been directed toward factors like metal binding, post-translational modifications, mutations of protein etc., which eventually affect the reactivity and cytotoxicity of the associated proteins. Over the past decade, a global effort from different groups working on these misfolded/unfolded proteins/peptides has revealed that the amino acid residues in the second coordination sphere of the active sites of amyloidogenic proteins/peptides cause changes in H-bonding pattern or protein-protein interactions, which dramatically alter the structure and reactivity of these proteins/peptides. These second sphere effects not only determine the binding of transition metals and cofactors, which define the pathology of some of these diseases, but also change the mechanism of redox reactions catalyzed by these proteins/peptides and form the basis of oxidative damage associated with these amyloidogenic diseases. The present review seeks to discuss such second sphere modifications and their ramifications in the etiopathology of some representative amyloidogenic diseases like Alzheimer's disease (AD), type 2 diabetes mellitus (T2Dm), Parkinson's disease (PD), Huntington's disease (HD), and prion diseases.
Collapse
Affiliation(s)
- Madhuparna Roy
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Arnab Kumar Nath
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Ishita Pal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Somdatta Ghosh Dey
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
17
|
Chin-Chan M, Montes S, Blanco-Álvarez VM, Aguirre-Alarcón HA, Hernández-Rodríguez I, Bautista E. Relevance of biometals during neuronal differentiation and myelination: in vitro and in vivo studies. Biometals 2022; 35:395-427. [DOI: 10.1007/s10534-022-00380-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 02/27/2022] [Indexed: 12/20/2022]
|
18
|
Wen L, Shen L. Effect of Surface-Chelated Cu 2+ on Amyloid-β Peptide Fibrillation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:174-181. [PMID: 34932369 DOI: 10.1021/acs.langmuir.1c02322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Abnormal interactions of copper (Cu) ions with amyloid-β (Aβ) peptides are believed to play an important role in the pathogenesis of Alzheimer's disease (AD). However, there is still debate as to the exact role of Cu ions in Aβ amyloidosis despite extensive studies on Aβ-Cu interactions. Unlike previously reported works, we herein study the effect of surface-chelated Cu2+, rather than the more usual solution-phase dissolved Cu2+, on Aβ aggregation. Through the combination of single molecule fluorescent tracking, atomic force microscopy imaging experiments, and all-atom molecular dynamic simulations, we show that the surface-chelated Cu2+ dynamically interacts with Aβ chains, restricts their 2D-diffusivity on the surface, and retards their fibrillation, while the designated surfaces without Cu2+ facilitate the 2D-diffusivity of Aβ chains for better interpeptide interaction and promote Aβ fibrillation. We offer a microscopic molecular insight into the retardation mechanism of surface-chelated Cu2+ on Aβ fibrillation, suggesting that the surface-bound pools of metal ions are critical in AD progression and drug design.
Collapse
Affiliation(s)
- Lisi Wen
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Lei Shen
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| |
Collapse
|
19
|
Probable Reasons for Neuron Copper Deficiency in the Brain of Patients with Alzheimer’s Disease: The Complex Role of Amyloid. INORGANICS 2022. [DOI: 10.3390/inorganics10010006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease is a progressive neurodegenerative disorder that eventually leads the affected patients to die. The appearance of senile plaques in the brains of Alzheimer’s patients is known as a main symptom of this disease. The plaques consist of different components, and according to numerous reports, their main components include beta-amyloid peptide and transition metals such as copper. In this disease, metal dyshomeostasis leads the number of copper ions to simultaneously increase in the plaques and decrease in neurons. Copper ions are essential for proper brain functioning, and one of the possible mechanisms of neuronal death in Alzheimer’s disease is the copper depletion of neurons. However, the reason for the copper depletion is as yet unknown. Based on the available evidence, we suggest two possible reasons: the first is copper released from neurons (along with beta-amyloid peptides), which is deposited outside the neurons, and the second is the uptake of copper ions by activated microglia.
Collapse
|
20
|
Prachayasittikul V, Pingaew R, Prachayasittikul S, Prachayasittikul V. 8-Hydroxyquinolines: A Promising Pharmacophore Potentially Developed as Disease-Modifying Agents for Neurodegenerative Diseases: A Review. HETEROCYCLES 2022. [DOI: 10.3987/rev-22-sr(r)6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
21
|
Kotuniak R, Bal W. Kinetics of Cu(II) complexation by ATCUN/NTS and related peptides: a gold mine of novel ideas for copper biology. Dalton Trans 2021; 51:14-26. [PMID: 34816848 DOI: 10.1039/d1dt02878b] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cu(II)-peptide complexes are intensely studied as models for biological peptides and proteins and for their direct importance in copper homeostasis and dyshomeostasis in human diseases. In particular, high-affinity ATCUN/NTS (amino-terminal copper and nickel/N-terminal site) motifs present in proteins and peptides are considered as Cu(II) transport agents for copper delivery to cells. The information on the affinities and structures of such complexes derived from steady-state methods appears to be insufficient to resolve the mechanisms of copper trafficking, while kinetic studies have recently shown promise in explaining them. Stopped-flow experiments of Cu(II) complexation to ATCUN/NTS peptides revealed the presence of reaction steps with rates much slower than the diffusion limit due to the formation of novel intermediate species. Herein, the state of the field in Cu(II)-peptide kinetics is reviewed in the context of physiological data, leading to novel ideas in copper biology, together with the discussion of current methodological issues.
Collapse
Affiliation(s)
- Radosław Kotuniak
- Institute of Biochemistry and Biophysics, Polish Academy of Science, Pawińskiego 5a, 02-106 Warsaw, Poland.
| | - Wojciech Bal
- Institute of Biochemistry and Biophysics, Polish Academy of Science, Pawińskiego 5a, 02-106 Warsaw, Poland.
| |
Collapse
|
22
|
Behar AE, Sabater L, Baskin M, Hureau C, Maayan G. A Water-Soluble Peptoid Chelator that Can Remove Cu 2+ from Amyloid-β Peptides and Stop the Formation of Reactive Oxygen Species Associated with Alzheimer's Disease. Angew Chem Int Ed Engl 2021; 60:24588-24597. [PMID: 34510664 DOI: 10.1002/anie.202109758] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/01/2021] [Indexed: 12/25/2022]
Abstract
Cu bound to amyloid-β (Aβ) peptides can act as a catalyst for the formation of reactive oxygen species (ROS), leading to neuropathologic degradation associated with Alzheimer's disease (AD). An excellent therapeutic approach is to use a chelator that can selectively remove Cu from Cu-Aβ. This chelator should compete with Zn2+ ions (Zn) that are present in the synaptic cleft while forming a nontoxic Cu complex. Herein we describe P3, a water-soluble peptidomimetic chelator that selectively removes Cu2+ from Cu-Aβ in the presence of Zn and prevent the formation of ROS even in a reductive environment. We demonstrate, based on extensive spectroscopic analysis, that although P3 extracts Zn from Cu,Zn-Aβ faster than it removes Cu, the formed Zn complexes are kinetic products that further dissociate, while CuP3 is formed as an exclusive stable thermodynamic product. Our unique findings, combined with the bioavailability of peptoids, make P3 an excellent drug candidate in the context of AD.
Collapse
Affiliation(s)
- Anastasia E Behar
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Technion City, 3200008, Haifa, Israel
| | - Laurent Sabater
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, 31077, Toulouse, France.,Université de Toulouse, 31077, Toulouse, France
| | - Maria Baskin
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Technion City, 3200008, Haifa, Israel
| | - Christelle Hureau
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, 31077, Toulouse, France.,Université de Toulouse, 31077, Toulouse, France
| | - Galia Maayan
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Technion City, 3200008, Haifa, Israel
| |
Collapse
|
23
|
Behar AE, Sabater L, Baskin M, Hureau C, Maayan G. A Water‐Soluble Peptoid Chelator that Can Remove Cu
2+
from Amyloid‐β Peptides and Stop the Formation of Reactive Oxygen Species Associated with Alzheimer's Disease. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202109758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Anastasia E. Behar
- Schulich Faculty of Chemistry Technion—Israel Institute of Technology Technion City 3200008 Haifa Israel
| | - Laurent Sabater
- CNRS LCC (Laboratoire de Chimie de Coordination) 205 route de Narbonne 31077 Toulouse France
- Université de Toulouse 31077 Toulouse France
| | - Maria Baskin
- Schulich Faculty of Chemistry Technion—Israel Institute of Technology Technion City 3200008 Haifa Israel
| | - Christelle Hureau
- CNRS LCC (Laboratoire de Chimie de Coordination) 205 route de Narbonne 31077 Toulouse France
- Université de Toulouse 31077 Toulouse France
| | - Galia Maayan
- Schulich Faculty of Chemistry Technion—Israel Institute of Technology Technion City 3200008 Haifa Israel
| |
Collapse
|
24
|
Nochebuena J, Quintanar L, Vela A, Cisneros GA. Structural and electronic analysis of the octarepeat region of prion protein with four Cu 2+ by polarizable MD and QM/MM simulations. Phys Chem Chem Phys 2021; 23:21568-21578. [PMID: 34550129 PMCID: PMC8497436 DOI: 10.1039/d1cp03187b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Prions have been linked to neurodegenerative diseases that affect various species of mammals including humans. The prion protein, located mainly in neurons, is believed to play the role of metal ion transporter. High levels of copper ions have been related to structural changes. A 32-residue region of the N-terminal domain, known as octarepeat, can bind up to four copper ions. Different coordination modes have been observed and are strongly dependent on Cu2+ concentration. Many theoretical studies carried out so far have focused on studying the coordination modes of a single copper ion. In this work we investigate the octarepeat region coordinated with four copper ions. Molecular dynamics (MD) and hybrid quantum mechanics/molecular mechanics (QM/MM) simulations using the polarizable AMOEBA force field have been carried out. The polarizable MD simulations starting from a fully extended conformation indicate that the tetra-Cu2+/octarepeat complex forms a globular structure. The globular form is stabilized by interactions between Cu2+ and tryptophan residues resulting in some coordination sites observed to be in close proximity, in agreement with experimental results. Subsequent QM/MM simulations on several snapshots suggests the system is in a high-spin quintet state, with all Cu2+ bearing one single electron, and all unpaired electrons are ferromagnetically coupled. NMR simulations on selected structures provides insights on the chemical shifts of the first shell ligands around the metals with respect to inter-metal distances.
Collapse
Affiliation(s)
- Jorge Nochebuena
- Department of Chemistry, University of North Texas, Denton, Texas, 76201, USA.
| | - Liliana Quintanar
- Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, 07360, Mexico
| | - Alberto Vela
- Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, 07360, Mexico
| | - G Andrés Cisneros
- Department of Chemistry, University of North Texas, Denton, Texas, 76201, USA.
| |
Collapse
|
25
|
Redox-Active Metal Ions and Amyloid-Degrading Enzymes in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22147697. [PMID: 34299316 PMCID: PMC8307724 DOI: 10.3390/ijms22147697] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/11/2021] [Accepted: 07/16/2021] [Indexed: 12/11/2022] Open
Abstract
Redox-active metal ions, Cu(I/II) and Fe(II/III), are essential biological molecules for the normal functioning of the brain, including oxidative metabolism, synaptic plasticity, myelination, and generation of neurotransmitters. Dyshomeostasis of these redox-active metal ions in the brain could cause Alzheimer’s disease (AD). Thus, regulating the levels of Cu(I/II) and Fe(II/III) is necessary for normal brain function. To control the amounts of metal ions in the brain and understand the involvement of Cu(I/II) and Fe(II/III) in the pathogenesis of AD, many chemical agents have been developed. In addition, since toxic aggregates of amyloid-β (Aβ) have been proposed as one of the major causes of the disease, the mechanism of clearing Aβ is also required to be investigated to reveal the etiology of AD clearly. Multiple metalloenzymes (e.g., neprilysin, insulin-degrading enzyme, and ADAM10) have been reported to have an important role in the degradation of Aβ in the brain. These amyloid degrading enzymes (ADE) could interact with redox-active metal ions and affect the pathogenesis of AD. In this review, we introduce and summarize the roles, distributions, and transportations of Cu(I/II) and Fe(II/III), along with previously invented chelators, and the structures and functions of ADE in the brain, as well as their interrelationships.
Collapse
|
26
|
Kawahara M, Tanaka KI, Kato-Negishi M. Copper as a Collaborative Partner of Zinc-Induced Neurotoxicity in the Pathogenesis of Vascular Dementia. Int J Mol Sci 2021; 22:ijms22147242. [PMID: 34298862 PMCID: PMC8305384 DOI: 10.3390/ijms22147242] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/17/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
Copper is an essential trace element and possesses critical roles in various brain functions. A considerable amount of copper accumulates in the synapse and is secreted in neuronal firings in a manner similar to zinc. Synaptic copper and zinc modulate neuronal transmission and contribute to information processing. It has been established that excess zinc secreted during transient global ischemia plays central roles in ischemia-induced neuronal death and the pathogenesis of vascular dementia. We found that a low concentration of copper exacerbates zinc-induced neurotoxicity, and we have demonstrated the involvement of the endoplasmic reticulum (ER) stress pathway, the stress-activated protein kinases/c-Jun amino-terminal kinases (SAPK/JNK) signaling pathway, and copper-induced reactive oxygen species (ROS) production. On the basis of our results and other studies, we discuss the collaborative roles of copper in zinc-induced neurotoxicity in the synapse and the contribution of copper to the pathogenesis of vascular dementia.
Collapse
|
27
|
Posadas Y, Parra-Ojeda L, Perez-Cruz C, Quintanar L. Amyloid β Perturbs Cu(II) Binding to the Prion Protein in a Site-Specific Manner: Insights into Its Potential Neurotoxic Mechanisms. Inorg Chem 2021; 60:8958-8972. [PMID: 34043332 DOI: 10.1021/acs.inorgchem.1c00846] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Amyloid β (Aβ) is a Cu-binding peptide that plays a key role in the pathology of Alzheimer's disease. A recent report demonstrated that Aβ disrupts the Cu-dependent interaction between cellular prion protein (PrPC) and N-methyl-d-aspartate receptor (NMDAR), inducing overactivation of NMDAR and neurotoxicity. In this context, it has been proposed that Aβ competes for Cu with PrPC; however, there is no spectroscopic evidence to support this hypothesis. Prion protein (PrP) can bind up to six Cu(II) ions: from one to four at the octarepeat (OR) region, producing low- and high-occupancy modes, and two at the His96 and His111 sites. Additionally, PrPC is cleaved by α-secretases at Lys110/His111, yielding a new Cu(II)-binding site at the α-cleaved His111. In this study, the competition for Cu(II) between Aβ(1-16) and peptide models for each Cu-binding site of PrP was evaluated using circular dichroism and electron paramagnetic resonance. Our results show that the impact of Aβ(1-16) on Cu(II) coordination to PrP is highly site-specific: Aβ(1-16) cannot effectively compete with the low-occupancy mode at the OR region, whereas it partially removes the metal ion from the high-occupancy modes and forms a ternary OR-Cu(II)-Aβ(1-16) complex. In contrast, Aβ(1-16) removes all Cu(II) ions from the His96 and His111 sites without formation of ternary species. Finally, at the α-cleaved His111 site, Aβ(1-16) yields at least two different ternary complexes depending on the ratio of PrP/Cu(II)/Aβ. Altogether, our spectroscopic results indicate that only the low-occupancy mode at the OR region resists the effect of Aβ, while Cu(II) coordination to the high-occupancy modes and all other tested sites of PrP is perturbed, by either removal of the metal ion or formation of ternary complexes. These results provide important insights into the intricate effect of Aβ on Cu(II) binding to PrP and the potential neurotoxic mechanisms through which Aβ might affect Cu-dependent functions of PrPC, such as NMDAR modulation.
Collapse
|
28
|
Nerve Growth Factor Peptides Bind Copper(II) with High Affinity: A Thermodynamic Approach to Unveil Overlooked Neurotrophin Roles. Int J Mol Sci 2021; 22:ijms22105085. [PMID: 34064906 PMCID: PMC8150721 DOI: 10.3390/ijms22105085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/16/2022] Open
Abstract
Nerve growth factor (NGF) is a protein essential to neurons survival, which interacts with its receptor as a non-covalent dimer. Peptides belonging to NGF N-terminal domain are able to mimic the activity of the whole protein. Such activity is affected by the presence of copper ions. The metal is released in the synaptic cleft where proteins, not yet identified, may bind and transfer to human copper transporter 1 (hCtr1), for copper uptake in neurons. The measurements of the stability constants of copper complexes formed by amyloid beta and hCtr1 peptide fragments suggest that beta-amyloid (Aβ) can perform this task. In this work, the stability constant values of copper complex species formed with the dimeric form of N-terminal domain, sequence 1–15 of the protein, were determined by means of potentiometric measurements. At physiological pH, NGF peptides bind one equivalent of copper ion with higher affinity of Aβ and lower than hCtr1 peptide fragments. Therefore, in the synaptic cleft, NGF may act as a potential copper chelating molecule, ionophore or chaperone for hCtr1 for metal uptake. Copper dyshomeostasis and mild acidic environment may modify the balance between metal, NGF, and Aβ, with consequences on the metal cellular uptake and therefore be among causes of the Alzheimer’s disease onset.
Collapse
|
29
|
La Mendola D, Arena G, Pietropaolo A, Satriano C, Rizzarelli E. Metal ion coordination in peptide fragments of neurotrophins: A crucial step for understanding the role and signaling of these proteins in the brain. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
30
|
Szabó Z, Péter M, Héja L, Kardos J. Dual Role for Astroglial Copper-Assisted Polyamine Metabolism during Intense Network Activity. Biomolecules 2021; 11:604. [PMID: 33921742 PMCID: PMC8073386 DOI: 10.3390/biom11040604] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 12/29/2022] Open
Abstract
Astrocytes serve essential roles in human brain function and diseases. Growing evidence indicates that astrocytes are central players of the feedback modulation of excitatory Glu signalling during epileptiform activity via Glu-GABA exchange. The underlying mechanism results in the increase of tonic inhibition by reverse operation of the astroglial GABA transporter, induced by Glu-Na+ symport. GABA, released from astrocytes, is synthesized from the polyamine (PA) putrescine and this process involves copper amino oxidase. Through this pathway, putrescine can be considered as an important source of inhibitory signaling that counterbalances epileptic discharges. Putrescine, however, is also a precursor for spermine that is known to enhance gap junction channel communication and, consequently, supports long-range Ca2+ signaling and contributes to spreading of excitatory activity through the astrocytic syncytium. Recently, we presented the possibility of neuron-glia redox coupling through copper (Cu+/Cu2+) signaling and oxidative putrescine catabolism. In the current work, we explore whether the Cu+/Cu2+ homeostasis is involved in astrocytic control on neuronal excitability by regulating PA catabolism. We provide supporting experimental data underlying this hypothesis. We show that the blockade of copper transporter (CTR1) by AgNO3 (3.6 µM) prevents GABA transporter-mediated tonic inhibitory currents, indicating causal relationship between copper (Cu+/Cu2+) uptake and the catabolism of putrescine to GABA in astrocytes. In addition, we show that MnCl2 (20 μM), an inhibitor of the divalent metal transporter DMT1, also prevents the astrocytic Glu-GABA exchange. Furthermore, we observed that facilitation of copper uptake by added CuCl2 (2 µM) boosts tonic inhibitory currents. These findings corroborate the hypothesis that modulation of neuron-glia coupling by copper uptake drives putrescine → GABA transformation, which leads to subsequent Glu-GABA exchange and tonic inhibition. Findings may in turn highlight the potential role of copper signaling in fine-tuning the activity of the tripartite synapse.
Collapse
Affiliation(s)
- Zsolt Szabó
- Functional Pharmacology Research Group, Research Centre for Natural Sciences, Institute of Organic Chemistry, H-1117 Budapest, Hungary; (Z.S.); (M.P.); (J.K.)
| | - Márton Péter
- Functional Pharmacology Research Group, Research Centre for Natural Sciences, Institute of Organic Chemistry, H-1117 Budapest, Hungary; (Z.S.); (M.P.); (J.K.)
- Hevesy György Ph.D. School of Chemistry, ELTE Eötvös Loránd University, H-1117 Budapest, Hungary
| | - László Héja
- Functional Pharmacology Research Group, Research Centre for Natural Sciences, Institute of Organic Chemistry, H-1117 Budapest, Hungary; (Z.S.); (M.P.); (J.K.)
| | - Julianna Kardos
- Functional Pharmacology Research Group, Research Centre for Natural Sciences, Institute of Organic Chemistry, H-1117 Budapest, Hungary; (Z.S.); (M.P.); (J.K.)
| |
Collapse
|
31
|
Sergeeva EG, Rosenberg PA, Benowitz LI. Non-Cell-Autonomous Regulation of Optic Nerve Regeneration by Amacrine Cells. Front Cell Neurosci 2021; 15:666798. [PMID: 33935656 PMCID: PMC8085350 DOI: 10.3389/fncel.2021.666798] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/19/2021] [Indexed: 11/13/2022] Open
Abstract
Visual information is conveyed from the eye to the brain through the axons of retinal ganglion cells (RGCs) that course through the optic nerve and synapse onto neurons in multiple subcortical visual relay areas. RGCs cannot regenerate their axons once they are damaged, similar to most mature neurons in the central nervous system (CNS), and soon undergo cell death. These phenomena of neurodegeneration and regenerative failure are widely viewed as being determined by cell-intrinsic mechanisms within RGCs or to be influenced by the extracellular environment, including glial or inflammatory cells. However, a new concept is emerging that the death or survival of RGCs and their ability to regenerate axons are also influenced by the complex circuitry of the retina and that the activation of a multicellular signaling cascade involving changes in inhibitory interneurons - the amacrine cells (AC) - contributes to the fate of RGCs. Here, we review our current understanding of the role that interneurons play in cell survival and axon regeneration after optic nerve injury.
Collapse
Affiliation(s)
- Elena G. Sergeeva
- Department of Neurology, Boston Children’s Hospital, Boston, MA, United States
- Kirby Center for Neuroscience, Boston Children’s Hospital, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - Paul A. Rosenberg
- Department of Neurology, Boston Children’s Hospital, Boston, MA, United States
- Kirby Center for Neuroscience, Boston Children’s Hospital, Boston, MA, United States
- Department of Neurology, Harvard Medical School, Boston, MA, United States
| | - Larry I. Benowitz
- Kirby Center for Neuroscience, Boston Children’s Hospital, Boston, MA, United States
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children’s Hospital, Boston, MA, United States
- Department of Neurosurgery, Boston Children’s Hospital, Boston, MA, United States
- Department of Neurosurgery, Harvard Medical School, Boston, MA, United States
- Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
32
|
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: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
Schoonover KE, Farmer CB, Morgan CJ, Sinha V, Odom L, Roberts RC. Abnormalities in the copper transporter CTR1 in postmortem hippocampus in schizophrenia: A subregion and laminar analysis. Schizophr Res 2021; 228:60-73. [PMID: 33434736 PMCID: PMC7987889 DOI: 10.1016/j.schres.2020.12.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/16/2020] [Accepted: 12/21/2020] [Indexed: 01/08/2023]
Abstract
Dysbindin-1 modulates copper transport, which is crucial for cellular homeostasis. Several brain regions implicated in schizophrenia exhibit decreased levels of dysbindin-1, which may affect copper homeostasis therein. Our recent study showed decreased levels of dysbindin-1, the copper transporter-1 (CTR1) and copper in the substantia nigra in schizophrenia, providing the first evidence of disrupted copper transport in schizophrenia. In the present study, we hypothesized that there would be lower levels of dysbindin-1 and CTR1 in the hippocampus in schizophrenia versus a comparison group. Using semi-quantitative immunohistochemistry for dysbindin1 and CTR1, we measured the optical density in a layer specific fashion in the hippocampus and entorhinal cortex in ten subjects with schizophrenia and ten comparison subjects. Both regions were richly immunolabeled for CTR1 and dysbindin1 in both groups. In the superficial layers of the entorhinal cortex, CTR1 immunolabeled neuropil and cells showed lower optical density values in patients versus the comparison group. In the molecular layer of the dentate gyrus, patients had higher optical density values of CTR1 versus the comparison group. The density and distribution of dysbindin-1 immunolabeling was similar between groups. These laminar specific alterations of CTR1 in schizophrenia suggest abnormal copper transport in those locations.
Collapse
Affiliation(s)
- Kirsten E. Schoonover
- Department of Psychology and Behavioral Neuroscience, University of Alabama at Birmingham
| | - Charlene B. Farmer
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham
| | - Charity J Morgan
- Department of Biostatistics, University of Alabama at Birmingham
| | - Vidushi Sinha
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham
| | - Laura Odom
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham
| | - Rosalinda C. Roberts
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham
| |
Collapse
|
34
|
Bakkar N, Starr A, Rabichow BE, Lorenzini I, McEachin ZT, Kraft R, Chaung M, Macklin-Isquierdo S, Wingfield T, Carhart B, Zahler N, Chang WH, Bassell GJ, Betourne A, Boulis N, Alworth SV, Ichida JK, August PR, Zarnescu DC, Sattler R, Bowser R. The M1311V variant of ATP7A is associated with impaired trafficking and copper homeostasis in models of motor neuron disease. Neurobiol Dis 2020; 149:105228. [PMID: 33359139 DOI: 10.1016/j.nbd.2020.105228] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/04/2020] [Accepted: 12/17/2020] [Indexed: 02/08/2023] Open
Abstract
Disruption in copper homeostasis causes a number of cognitive and motor deficits. Wilson's disease and Menkes disease are neurodevelopmental disorders resulting from mutations in the copper transporters ATP7A and ATP7B, with ATP7A mutations also causing occipital horn syndrome, and distal motor neuropathy. A 65 year old male presenting with brachial amyotrophic diplegia and diagnosed with amyotrophic lateral sclerosis (ALS) was found to harbor a p.Met1311Val (M1311V) substitution variant in ATP7A. ALS is a fatal neurodegenerative disease associated with progressive muscle weakness, synaptic deficits and degeneration of upper and lower motor neurons. To investigate the potential contribution of the ATP7AM1311V variant to neurodegeneration, we obtained and characterized both patient-derived fibroblasts and patient-derived induced pluripotent stem cells differentiated into motor neurons (iPSC-MNs), and compared them to control cell lines. We found reduced localization of ATP7AM1311V to the trans-Golgi network (TGN) at basal copper levels in patient-derived fibroblasts and iPSC-MNs. In addition, redistribution of ATP7AM1311V out of the TGN in response to increased extracellular copper was defective in patient fibroblasts. This manifested in enhanced intracellular copper accumulation and reduced survival of ATP7AM1311V fibroblasts. iPSC-MNs harboring the ATP7AM1311V variant showed decreased dendritic complexity, aberrant spontaneous firing, and decreased survival. Finally, expression of the ATP7AM1311V variant in Drosophila motor neurons resulted in motor deficits. Apilimod, a drug that targets vesicular transport and recently shown to enhance survival of C9orf72-ALS/FTD iPSC-MNs, also increased survival of ATP7AM1311V iPSC-MNs and reduced motor deficits in Drosophila expressing ATP7AM1311V. Taken together, these observations suggest that ATP7AM1311V negatively impacts its role as a copper transporter and impairs several aspects of motor neuron function and morphology.
Collapse
Affiliation(s)
- Nadine Bakkar
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Alexander Starr
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Benjamin E Rabichow
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Ileana Lorenzini
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Zachary T McEachin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Robert Kraft
- Departments of Molecular and Cellular Biology, Neuroscience, and Neurology, University of Arizona, Tucson, AZ 85721, USA
| | - Matthew Chaung
- Departments of Molecular and Cellular Biology, Neuroscience, and Neurology, University of Arizona, Tucson, AZ 85721, USA
| | - Sam Macklin-Isquierdo
- Departments of Molecular and Cellular Biology, Neuroscience, and Neurology, University of Arizona, Tucson, AZ 85721, USA
| | - Taylor Wingfield
- Departments of Molecular and Cellular Biology, Neuroscience, and Neurology, University of Arizona, Tucson, AZ 85721, USA
| | - Briggs Carhart
- Departments of Molecular and Cellular Biology, Neuroscience, and Neurology, University of Arizona, Tucson, AZ 85721, USA
| | | | | | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Nicholas Boulis
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | - Daniela C Zarnescu
- Departments of Molecular and Cellular Biology, Neuroscience, and Neurology, University of Arizona, Tucson, AZ 85721, USA
| | - Rita Sattler
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA.
| | - Robert Bowser
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA.
| |
Collapse
|
35
|
Cioffi F, Adam RHI, Broersen K. Molecular Mechanisms and Genetics of Oxidative Stress in Alzheimer's Disease. J Alzheimers Dis 2020; 72:981-1017. [PMID: 31744008 PMCID: PMC6971833 DOI: 10.3233/jad-190863] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alzheimer’s disease is the most common neurodegenerative disorder that can cause dementia in elderly over 60 years of age. One of the disease hallmarks is oxidative stress which interconnects with other processes such as amyloid-β deposition, tau hyperphosphorylation, and tangle formation. This review discusses current thoughts on molecular mechanisms that may relate oxidative stress to Alzheimer’s disease and identifies genetic factors observed from in vitro, in vivo, and clinical studies that may be associated with Alzheimer’s disease-related oxidative stress.
Collapse
Affiliation(s)
- Federica Cioffi
- Nanobiophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Rayan Hassan Ibrahim Adam
- Nanobiophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Kerensa Broersen
- Applied Stem Cell Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
36
|
Stefaniak E, Pushie MJ, Vaerewyck C, Corcelli D, Griggs C, Lewis W, Kelley E, Maloney N, Sendzik M, Bal W, Haas KL. Exploration of the Potential Role for Aβ in Delivery of Extracellular Copper to Ctr1. Inorg Chem 2020; 59:16952-16966. [PMID: 33211469 DOI: 10.1021/acs.inorgchem.0c02100] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Amyloid beta (Aβ) peptides are notorious for their involvement in Alzheimer's disease (AD), by virtue of their propensity to aggregate to form oligomers, fibrils, and eventually plaques in the brain. Nevertheless, they appear to be essential for correct neurophysiology on the synaptic level and may have additional functions including antimicrobial activity, sealing the blood-brain barrier, promotion of recovery from brain injury, and even tumor suppression. Aβ peptides are also avid copper chelators, and coincidentally copper is significantly dysregulated in the AD brain. Copper (Cu) is released in significant amounts during calcium signaling at the synaptic membrane. Aβ peptides may have a role in maintaining synaptic Cu homeostasis, including as a scavenger for redox-active Cu and as a chaperone for clearing Cu from the synaptic cleft. Here, we employed the Aβ1-16 and Aβ4-16 peptides as well-established non-aggregating models of major Aβ species in healthy and AD brains, and the Ctr1-14 peptide as a model for the extracellular domain of the human cellular copper transporter protein (Ctr1). With these model peptides and a number of spectroscopic techniques, we investigated whether the Cu complexes of Aβ peptides could provide Ctr1 with either Cu(II) or Cu(I). We found that Aβ1-16 fully and rapidly delivered Cu(II) to Ctr1-14 along the affinity gradient. Such delivery was only partial for the Aβ4-16/Ctr1-14 pair, in agreement with the higher complex stability for the former peptide. Moreover, the reaction was very slow and took ca. 40 h to reach equilibrium under the given experimental conditions. In either case of Cu(II) exchange, no intermediate (ternary) species were present in detectable amounts. In contrast, both Aβ species released Cu(I) to Ctr1-14 rapidly and in a quantitative fashion, but ternary intermediate species were detected in the analysis of XAS data. The results presented here are the first direct evidence of a Cu(I) and Cu(II) transfer between the human Ctr1 and Aβ model peptides. These results are discussed in terms of the fundamental difference between the peptides' Cu(II) complexes (pleiotropic ensemble of open structures of Aβ1-16 vs the rigid closed-ring system of amino-terminal Cu/Ni binding Aβ4-16) and the similarity of their Cu(I) complexes (both anchored at the tandem His13/His14, bis-His motif). These results indicate that Cu(I) may be more feasible than Cu(II) as the cargo for copper clearance from the synaptic cleft by Aβ peptides and its delivery to Ctr1. The arguments in favor of Cu(I) include the fact that cellular Cu export and uptake proteins (ATPase7A/B and Ctr1, respectively) specifically transport Cu(I), the abundance of extracellular ascorbate reducing agent in the brain, and evidence of a potential associative (hand-off) mechanism of Cu(I) transfer that may mirror the mechanisms of intracellular Cu chaperone proteins.
Collapse
Affiliation(s)
- Ewelina Stefaniak
- Department of Chemistry and Physics, Saint Mary's College, Notre Dame, Indiana 46556, United States.,Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland
| | - M Jake Pushie
- Department of Surgery, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Catherine Vaerewyck
- Department of Chemistry and Physics, Saint Mary's College, Notre Dame, Indiana 46556, United States
| | - David Corcelli
- Department of Chemistry and Physics, Saint Mary's College, Notre Dame, Indiana 46556, United States
| | - Chloe Griggs
- Department of Chemistry and Physics, Saint Mary's College, Notre Dame, Indiana 46556, United States
| | - Whitney Lewis
- Department of Chemistry and Physics, Saint Mary's College, Notre Dame, Indiana 46556, United States
| | - Emma Kelley
- Department of Chemistry and Physics, Saint Mary's College, Notre Dame, Indiana 46556, United States
| | - Noreen Maloney
- Department of Chemistry and Physics, Saint Mary's College, Notre Dame, Indiana 46556, United States
| | - Madison Sendzik
- Department of Chemistry and Physics, Saint Mary's College, Notre Dame, Indiana 46556, United States
| | - Wojciech Bal
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland
| | - Kathryn L Haas
- Department of Chemistry and Physics, Saint Mary's College, Notre Dame, Indiana 46556, United States
| |
Collapse
|
37
|
Krzywoszyńska K, Witkowska D, Świątek-Kozłowska J, Szebesczyk A, Kozłowski H. General Aspects of Metal Ions as Signaling Agents in Health and Disease. Biomolecules 2020; 10:biom10101417. [PMID: 33036384 PMCID: PMC7600656 DOI: 10.3390/biom10101417] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023] Open
Abstract
This review focuses on the current knowledge on the involvement of metal ions in signaling processes within the cell, in both physiological and pathological conditions. The first section is devoted to the recent discoveries on magnesium and calcium-dependent signal transduction-the most recognized signaling agents among metals. The following sections then describe signaling pathways where zinc, copper, and iron play a key role. There are many systems in which changes in intra- and extra-cellular zinc and copper concentrations have been linked to important downstream events, especially in nervous signal transduction. Iron signaling is mostly related with its homeostasis. However, it is also involved in a recently discovered type of programmed cell death, ferroptosis. The important differences in metal ion signaling, and its disease-leading alterations, are also discussed.
Collapse
Affiliation(s)
- Karolina Krzywoszyńska
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
- Correspondence: (K.K.); (D.W.); Tel.: +48-77-44-23-549 (K.K); +48-77-44-23-548 (D.W.)
| | - Danuta Witkowska
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
- Correspondence: (K.K.); (D.W.); Tel.: +48-77-44-23-549 (K.K); +48-77-44-23-548 (D.W.)
| | - Jolanta Świątek-Kozłowska
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
| | - Agnieszka Szebesczyk
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
| | - Henryk Kozłowski
- Institute of Health Sciences, University of Opole, 68 Katowicka St., 45-060 Opole, Poland; (J.Ś.-K.); (A.S.); (H.K.)
- Faculty of Chemistry, University of Wrocław, 14 F. Joliot-Curie St., 50-383 Wrocław, Poland
| |
Collapse
|
38
|
Moons R, Konijnenberg A, Mensch C, Van Elzen R, Johannessen C, Maudsley S, Lambeir AM, Sobott F. Metal ions shape α-synuclein. Sci Rep 2020; 10:16293. [PMID: 33004902 PMCID: PMC7529799 DOI: 10.1038/s41598-020-73207-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/27/2020] [Indexed: 12/24/2022] Open
Abstract
α-Synuclein is an intrinsically disordered protein that can self-aggregate and plays a major role in Parkinson's disease (PD). Elevated levels of certain metal ions are found in protein aggregates in neurons of people suffering from PD, and environmental exposure has also been linked with neurodegeneration. Importantly, cellular interactions with metal ions, particularly Ca2+, have recently been reported as key for α-synuclein's physiological function at the pre-synapse. Here we study effects of metal ion interaction with α-synuclein at the molecular level, observing changes in the conformational behaviour of monomers, with a possible link to aggregation pathways and toxicity. Using native nano-electrospray ionisation ion mobility-mass spectrometry (nESI-IM-MS), we characterize the heterogeneous interactions of alkali, alkaline earth, transition and other metal ions and their global structural effects on α-synuclein. Different binding stoichiometries found upon titration with metal ions correlate with their specific binding affinity and capacity. Subtle conformational effects seen for singly charged metals differ profoundly from binding of multiply charged ions, often leading to overall compaction of the protein depending on the preferred binding sites. This study illustrates specific effects of metal coordination, and the associated electrostatic charge patterns, on the complex structural space of the intrinsically disordered protein α-synuclein.
Collapse
Affiliation(s)
- Rani Moons
- Biomolecular and Analytical Mass Spectrometry Group, University of Antwerp, Antwerp, Belgium.,Receptor Biology Laboratory, University of Antwerp, Antwerp, Belgium
| | - Albert Konijnenberg
- Biomolecular and Analytical Mass Spectrometry Group, University of Antwerp, Antwerp, Belgium
| | - Carl Mensch
- Molecular Spectroscopy Group, University of Antwerp, Antwerp, Belgium.,Flemish Supercomputer Centre, Antwerp, Belgium
| | - Roos Van Elzen
- Laboratory of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | | | - Stuart Maudsley
- Receptor Biology Laboratory, University of Antwerp, Antwerp, Belgium
| | - Anne-Marie Lambeir
- Laboratory of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Frank Sobott
- Biomolecular and Analytical Mass Spectrometry Group, University of Antwerp, Antwerp, Belgium. .,Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK. .,School of Molecular and Cellular Biology, University of Leeds, Leeds, UK.
| |
Collapse
|
39
|
Mahmoud M, Zaitone S, Ammar A. Binary and ternary Cu(II) complexes of pregabalin with excitatory and inhibitory neurotransmitters and their antiepileptic effect. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110650. [DOI: 10.1016/j.msec.2020.110650] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/01/2019] [Accepted: 01/03/2020] [Indexed: 10/25/2022]
|
40
|
Wang L, Yin YL, Liu XZ, Shen P, Zheng YG, Lan XR, Lu CB, Wang JZ. Current understanding of metal ions in the pathogenesis of Alzheimer's disease. Transl Neurodegener 2020; 9:10. [PMID: 32266063 PMCID: PMC7119290 DOI: 10.1186/s40035-020-00189-z] [Citation(s) in RCA: 236] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 03/11/2020] [Indexed: 02/07/2023] Open
Abstract
Background The homeostasis of metal ions, such as iron, copper, zinc and calcium, in the brain is crucial for maintaining normal physiological functions. Studies have shown that imbalance of these metal ions in the brain is closely related to the onset and progression of Alzheimer's disease (AD), the most common neurodegenerative disorder in the elderly. Main body Erroneous deposition/distribution of the metal ions in different brain regions induces oxidative stress. The metal ions imbalance and oxidative stress together or independently promote amyloid-β (Aβ) overproduction by activating β- or γ-secretases and inhibiting α-secretase, it also causes tau hyperphosphorylation by activating protein kinases, such as glycogen synthase kinase-3β (GSK-3β), cyclin-dependent protein kinase-5 (CDK5), mitogen-activated protein kinases (MAPKs), etc., and inhibiting protein phosphatase 2A (PP2A). The metal ions imbalances can also directly or indirectly disrupt organelles, causing endoplasmic reticulum (ER) stress; mitochondrial and autophagic dysfunctions, which can cause or aggravate Aβ and tau aggregation/accumulation, and impair synaptic functions. Even worse, the metal ions imbalance-induced alterations can reversely exacerbate metal ions misdistribution and deposition. The vicious cycles between metal ions imbalances and Aβ/tau abnormalities will eventually lead to a chronic neurodegeneration and cognitive deficits, such as seen in AD patients. Conclusion The metal ions imbalance induces Aβ and tau pathologies by directly or indirectly affecting multiple cellular/subcellular pathways, and the disrupted homeostasis can reversely aggravate the abnormalities of metal ions transportation/deposition. Therefore, adjusting metal balance by supplementing or chelating the metal ions may be potential in ameliorating AD pathologies, which provides new research directions for AD treatment.
Collapse
Affiliation(s)
- Lu Wang
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Ya-Ling Yin
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Xin-Zi Liu
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Peng Shen
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Yan-Ge Zheng
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Xin-Rui Lan
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Cheng-Biao Lu
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Jian-Zhi Wang
- 2Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| |
Collapse
|
41
|
Witt B, Schaumlöffel D, Schwerdtle T. Subcellular Localization of Copper-Cellular Bioimaging with Focus on Neurological Disorders. Int J Mol Sci 2020; 21:ijms21072341. [PMID: 32231018 PMCID: PMC7178132 DOI: 10.3390/ijms21072341] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/17/2022] Open
Abstract
As an essential trace element, copper plays a pivotal role in physiological body functions. In fact, dysregulated copper homeostasis has been clearly linked to neurological disorders including Wilson and Alzheimer’s disease. Such neurodegenerative diseases are associated with progressive loss of neurons and thus impaired brain functions. However, the underlying mechanisms are not fully understood. Characterization of the element species and their subcellular localization is of great importance to uncover cellular mechanisms. Recent research activities focus on the question of how copper contributes to the pathological findings. Cellular bioimaging of copper is an essential key to accomplish this objective. Besides information on the spatial distribution and chemical properties of copper, other essential trace elements can be localized in parallel. Highly sensitive and high spatial resolution techniques such as LA-ICP-MS, TEM-EDS, S-XRF and NanoSIMS are required for elemental mapping on subcellular level. This review summarizes state-of-the-art techniques in the field of bioimaging. Their strengths and limitations will be discussed with particular focus on potential applications for the elucidation of copper-related diseases. Based on such investigations, further information on cellular processes and mechanisms can be derived under physiological and pathological conditions. Bioimaging studies might enable the clarification of the role of copper in the context of neurodegenerative diseases and provide an important basis to develop therapeutic strategies for reduction or even prevention of copper-related disorders and their pathological consequences.
Collapse
Affiliation(s)
- Barbara Witt
- Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114–116, 14558 Nuthetal, Germany;
- Correspondence: ; Tel.: +49-3320-088-5241
| | - Dirk Schaumlöffel
- Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux (IPREM), UMR 5254, CNRS/Université de Pau et des Pays de l’Adour/E2S UPPA, 64000 Pau, France;
| | - 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), Potsdam-Berlin-Jena, Germany
| |
Collapse
|
42
|
Lorentzon E, Kumar R, Horvath I, Wittung-Stafshede P. Differential effects of Cu 2+ and Fe 3+ ions on in vitro amyloid formation of biologically-relevant α-synuclein variants. Biometals 2020; 33:97-106. [PMID: 32170541 PMCID: PMC7295844 DOI: 10.1007/s10534-020-00234-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/04/2020] [Indexed: 01/26/2023]
Abstract
Alterations in metal ion homeostasis appear coupled to neurodegenerative disorders but mechanisms are unknown. Amyloid formation of the protein α-synuclein in brain cells is a hallmark of Parkinson's disease. α-Synuclein can bind several metal ions in vitro and such interactions may affect the assembly process. Here we used biophysical methods to study the effects of micromolar concentrations of Cu2+ and Fe3+ ions on amyloid formation of selected α-synuclein variants (wild-type and A53T α-synuclein, in normal and N-terminally acetylated forms). As shown previously, Cu2+ speeds up aggregation of normal wild-type α-synuclein, but not the acetylated form. However, Cu2+ has a minimal effect on (the faster) aggregation of normal A53T α-synuclein, despite that Cu2+ binds to this variant. Like Cu2+, Fe3+ speeds up aggregation of non-acetylated wild-type α-synuclein, but with acetylation, Fe3+ instead slows down aggregation. In contrast, for A53T α-synuclein, regardless of acetylation, Fe3+ slows down aggregation with the effect being most dramatic for acetylated A53T α-synuclein. The results presented here suggest a correlation between metal-ion modulation effect and intrinsic aggregation speed of the various α-synuclein variants.
Collapse
Affiliation(s)
- Emma Lorentzon
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.,Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Ranjeet Kumar
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Istvan Horvath
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Pernilla Wittung-Stafshede
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.
| |
Collapse
|
43
|
Peacey L, Elphick MR, Jones CE. Roles of copper in neurokinin B and gonadotropin-releasing hormone structure and function and the endocrinology of reproduction. Gen Comp Endocrinol 2020; 287:113342. [PMID: 31783025 DOI: 10.1016/j.ygcen.2019.113342] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/24/2019] [Accepted: 11/25/2019] [Indexed: 11/30/2022]
Abstract
Copper is a metal ion present in all organisms, where it has well-known roles in association with proteins and enzymes essential for cellular processes. In the early decades of the twentieth century copper was shown to influence mammalian reproductive biology, and it was subsequently shown to exert effects primarily at the level of the pituitary gland and/or hypothalamic regions of the brain. Furthermore, it has been reported that copper can interact with key neuropeptides in the hypothalamic-pituitary-gonadal axis, notably gonadotropin-releasing hormone (GnRH) and neurokinin B. Interestingly, recent phylogenetic analysis of the sequences of GnRH-related peptides indicates that copper binding is an evolutionarily ancient property of this neuropeptide family, which has been variously retained, modified or lost in the different taxa. In this mini-review the metal-binding properties of neuropeptides in the vertebrate reproductive pathway are reviewed and the evolutionary and functional significance of copper binding by GnRH-related neuropeptides in vertebrates and invertebrates are discussed.
Collapse
Affiliation(s)
- Lorraine Peacey
- School of Science and Health, The University of Western Sydney, Locked Bag 1797, Penrith, New South Wales, Australia
| | - Maurice R Elphick
- Queen Mary University of London, School of Biological and Chemical Sciences, Mile End Road, London E14NS, UK
| | - Christopher E Jones
- School of Science and Health, The University of Western Sydney, Locked Bag 1797, Penrith, New South Wales, Australia.
| |
Collapse
|
44
|
Magrì A, Tabbì G, Cucci LM, Satriano C, Pietropaolo A, Malgieri G, Isernia C, La Mendola D. The curious case of opossum prion: a physicochemical study on copper(ii) binding to the bis-decarepeat fragment from the protein N-terminal domain. Dalton Trans 2019; 48:17533-17543. [PMID: 31748763 DOI: 10.1039/c9dt02510c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The opossum is a peculiar model of immunity to prion diseases. Here we scrutinised the bis-decarepeat peptide sequence of the opossum prion (Op_bis-deca) protein by a multitechnique approach, with a combined experimental (potentiometry, UV-visible, circular dichroism, NMR and EPR spectroscopy, quartz crystal microbalance with dissipation monitoring and confocal microscopy) and simulation (DFT calculations) approach. Results showed that the macrochelate structures formed upon the binding to Cu(ii) by the analogous bis-octarepeat peptide sequence of human prion (Hu_bis-octa) are not found in the case of Op_bis-deca. At physiological pH and equimolar amount of copper ions, the [CuLH-2] is the major species formed by Op_bis-deca. In this species one imidazole and two amide nitrogen atoms are involved in metal coordination and its stability constant value is lower than that of the analogous species formed by Hu_bis-octa, due to the presence of an extra proline residue. Moreover, the study on the interaction of the peptides or the peptide/Cu(ii) complexes with the model cell membranes made of supported lipid bilayers disclosed different levels of interaction, monitored by the viscoelastic changes of the membranes, which exhibited a similar viscoelastic response at the interface of the two complexes, while in the absence of Cu(ii), the Hu_bis-octa/SLB interface was more viscoelastic than the Op_bis-deca one.
Collapse
Affiliation(s)
- Antonio Magrì
- Institute of Crystallography, National Research Council (CNR), S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Jayawardena BM, Jones MR, Hong Y, Jones CE. Copper ions trigger disassembly of neurokinin B functional amyloid and inhibit de novo assembly. J Struct Biol 2019; 208:107394. [DOI: 10.1016/j.jsb.2019.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/23/2019] [Accepted: 09/23/2019] [Indexed: 01/24/2023]
|
46
|
Tian Y, Stanyon HF, Barritt JD, Mayet U, Patel P, Karamani E, Fusco G, Viles JH. Copper2+ Binding to α-Synuclein. Histidine50 Can Form a Ternary Complex with Cu2+ at the N-Terminus but Not a Macrochelate. Inorg Chem 2019; 58:15580-15589. [DOI: 10.1021/acs.inorgchem.9b02644] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Yao Tian
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
| | - Helen F. Stanyon
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
| | - Joseph D Barritt
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Uroosa Mayet
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
| | - Pelak Patel
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
| | - Elena Karamani
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
| | - Giuliana Fusco
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge CB1 1EW, United Kingdom
| | - John H. Viles
- School of Biological and Chemical Sciences, Queen Mary, University of London Mile End Road, London E1 4NS, United Kingdom
| |
Collapse
|
47
|
Pushie MJ, Stefaniak E, Sendzik MR, Sokaras D, Kroll T, Haas KL. Using N-Terminal Coordination of Cu(II) and Ni(II) to Isolate the Coordination Environment of Cu(I) and Cu(II) Bound to His13 and His14 in Amyloid-β(4-16). Inorg Chem 2019; 58:15138-15154. [PMID: 31657204 DOI: 10.1021/acs.inorgchem.9b01940] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The amyloid-β (Aβ) peptide is a cleavage product of the amyloid precursor protein and has been implicated as a central player in Alzheimer's disease. The N-terminal end of Aβ is variable, and different proportions of these variable-length Aβ peptides are present in healthy individuals and those with the disease. The N-terminally truncated form of Aβ starting at position 4 (Aβ4-x) has a His residue as the third amino acid (His6 using the formal Aβ numbering). The N-terminal sequence Xaa-Xaa-His is known as an amino terminal copper and nickel binding motif (ATCUN), which avidly binds Cu(II). This motif is not present in the commonly studied Aβ1-x peptides. In addition to the ATCUN site, Aβ4-x contains an additional metal binding site located at the tandem His residues (bis-His at His13 and 14) which is also found in other isoforms of Aβ. Using the ATCUN and bis-His motifs, the Aβ4-x peptide is capable of binding multiple metal ions simultaneously. We confirm that Cu(II) bound to this particular ATCUN site is redox silent, but the second Cu(II) site is redox active and can be readily reduced with ascorbate. We have employed surrogate metal ions to block copper coordination at the ATCUN or the tandem His site in order to isolate spectral features of the copper coordination environment for structural characterization using extended X-ray absorption fine structure (EXAFS) spectroscopy. This approach reveals that each copper coordination environment is independent in the Cu2Aβ4-x state. The identification of two functionally different copper binding environments within the Aβ4-x sequence may have important implications for this peptide in vivo.
Collapse
Affiliation(s)
- M Jake Pushie
- Department of Surgery , University of Saskatchewan , Saskatoon , Saskatchewan S7N 5E5 , Canada
| | - Ewelina Stefaniak
- Department of Chemistry and Physics , Saint Mary's College , Notre Dame , Indiana 46556 , United States.,Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Pawińskiego 5a , 02-106 Warsaw , Poland
| | - Madison R Sendzik
- Department of Chemistry and Physics , Saint Mary's College , Notre Dame , Indiana 46556 , United States
| | - Dimosthenis Sokaras
- Stanford Synchrotron Radiation Lightsource , SLAC National Accelerator Laboratory , 2575 Sand Hill Road , Menlo Park , California 94025 , United States
| | - Thomas Kroll
- Stanford Synchrotron Radiation Lightsource , SLAC National Accelerator Laboratory , 2575 Sand Hill Road , Menlo Park , California 94025 , United States
| | - Kathryn L Haas
- Department of Chemistry and Physics , Saint Mary's College , Notre Dame , Indiana 46556 , United States
| |
Collapse
|
48
|
Nam E, Nam G, Lim MH. Synaptic Copper, Amyloid-β, and Neurotransmitters in Alzheimer's Disease. Biochemistry 2019; 59:15-17. [PMID: 31603659 DOI: 10.1021/acs.biochem.9b00775] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Eunju Nam
- Department of Chemistry , Korea Advanced Institute of Science and Technology (KAIST) , Daejeon 34141 , Republic of Korea
| | - Geewoo Nam
- Department of Chemistry , Korea Advanced Institute of Science and Technology (KAIST) , Daejeon 34141 , Republic of Korea.,Department of Chemistry , Ulsan National Institute of Science and Technology (UNIST) , Ulsan 44919 , Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry , Korea Advanced Institute of Science and Technology (KAIST) , Daejeon 34141 , Republic of Korea
| |
Collapse
|
49
|
Kepp KP, Squitti R. Copper imbalance in Alzheimer’s disease: Convergence of the chemistry and the clinic. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.06.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
50
|
Saito K, Watanabe K, Yanaoka R, Kageyama L, Miura T. Potential role of serotonin as a biological reductant associated with copper transportation. J Inorg Biochem 2019; 199:110770. [PMID: 31336257 DOI: 10.1016/j.jinorgbio.2019.110770] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/21/2019] [Accepted: 07/11/2019] [Indexed: 11/17/2022]
Abstract
Serotonin (5-HT) is a neurotransmitter that is derived from tryptophan. Owing to a hydroxyl group attached to the indole nucleus, 5-HT exhibits a considerably higher redox activity than tryptophan. To gain insight into the biological relevance of the redox activity of 5-HT, the effect of Cu(I)-binding ligands on the 5-HT-mediated copper reduction was investigated. The d-d transition band of Cu(II) complexed with glycine [Cu(II)-Gly2] was not affected by addition of 5-HT alone but was diminished when a thioether-containing compound coexists with 5-HT. Concomitant with disappearance of the d-d transition band of Cu(II)-Gly2, the π-π* transition band of 5-hydroxyindole of 5-HT exhibits a red-shift which is consistently explained by oxidation of 5-HT and subsequent formation of a dimeric species. The redox reactions between 5-HT and copper are also accelerated by a peptide composed of a methionine (Met)-rich region in the extracellular domain of an integral membrane protein, copper transporter 1 (Ctr1). Since Ctr1 transports copper across the plasma membrane with specificity for Cu(I), reduction of extracellular Cu(II) to Cu(I) is required for copper uptake by Ctr1. Metalloreductases that can donate Cu(I) for Ctr1 have been identified in yeast but not yet been found in mammals. The results of this study indicate that the Met-rich region in the N-terminal extracellular domain of Ctr1 promotes the 5-HT-mediated Cu(II) reduction in order to acquire Cu(I) via a non-enzymatic process.
Collapse
Affiliation(s)
- Kaede Saito
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Kitakanemaru 2600-1, Otawara, Tochigi 324-8501, Japan
| | - Kasumi Watanabe
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Kitakanemaru 2600-1, Otawara, Tochigi 324-8501, Japan
| | - Risa Yanaoka
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Kitakanemaru 2600-1, Otawara, Tochigi 324-8501, Japan
| | - Lisa Kageyama
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Kitakanemaru 2600-1, Otawara, Tochigi 324-8501, Japan
| | - Takashi Miura
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Kitakanemaru 2600-1, Otawara, Tochigi 324-8501, Japan.
| |
Collapse
|