1
|
Dluhosch D, Kersten LS, Schott-Verdugo S, Hoppen C, Schwarten M, Willbold D, Gohlke H, Groth G. Structure and dimerization properties of the plant-specific copper chaperone CCH. Sci Rep 2024; 14:19099. [PMID: 39154065 PMCID: PMC11330527 DOI: 10.1038/s41598-024-69532-y] [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: 05/02/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024] Open
Abstract
Copper chaperones of the ATX1 family are found in a wide range of organisms where these essential soluble carriers strictly control the transport of monovalent copper across the cytoplasm to various targets in diverse cellular compartments thereby preventing detrimental radical formation catalyzed by the free metal ion. Notably, the ATX1 family in plants contains two distinct forms of the cellular copper carrier. In addition to ATX1 having orthologs in other species, they also contain the copper chaperone CCH. The latter features an extra C-terminal extension whose function is still unknown. The secondary structure of this extension was predicted to be disordered in previous studies, although this has not been experimentally confirmed. Solution NMR studies on purified CCH presented in this study disclose that this region is intrinsically disordered regardless of the chaperone's copper loading state. Further biophysical analyses of the purified metallochaperone provide evidence that the C-terminal extension stabilizes chaperone dimerization in the copper-free and copper-bound states. A variant of CCH lacking the C-terminal extension, termed CCHΔ, shows weaker dimerization but similar copper binding. Computational studies further corroborate the stabilizing role of the C-terminal extension in chaperone dimerization and identify key residues that are vital to maintaining dimer stability.
Collapse
Affiliation(s)
- Dominik Dluhosch
- Institute of Biochemical Plant Physiology, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
| | - Lisa Sophie Kersten
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
| | - Stephan Schott-Verdugo
- Institute of Bio- and Geosciences: Bioinformatics (IBG-4), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Claudia Hoppen
- Institute of Biochemical Plant Physiology, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
| | - Melanie Schwarten
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Dieter Willbold
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52425, Jülich, Germany
- Institut Für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
| | - Holger Gohlke
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
- Institute of Bio- and Geosciences: Bioinformatics (IBG-4), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Georg Groth
- Institute of Biochemical Plant Physiology, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany.
| |
Collapse
|
2
|
El-Sayed A, Ebissy E, Ateya A. Positive impacts of Nannochloropsis oculata supplementation on gene expression of immune and antioxidant markers and metabolic profile of Barki sheep in the transition period and lipogenic effects on progeny. Vet Res Commun 2024; 48:2207-2226. [PMID: 38702499 PMCID: PMC11315803 DOI: 10.1007/s11259-024-10392-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 04/20/2024] [Indexed: 05/06/2024]
Abstract
Nannochloropsis species should be given priority when it comes to microalgae that should be added to feed since they are suitable for intense culture and have a high concentration of PUFAs (especially EPA), antioxidants, and certain vitamins. This study investigated the possible immune and antioxidant impacts of Nannochloropsis supplementation on Barki ewes during transition period and their newly born lambs. Three weeks prior to the expected time of lambing, the researched ewes were divided into two equal groups of thirty ewes each. The second group, on the other hand, was fed the same base diet as the first group plus 10 g of commercially available Nannochloropsis powder per kg of concentrate, given daily to each ewe's concentrate. Findings revealed that supplementation of ewes with Nannochloropsis significantly up-regulated the expression pattern of immune (NFKB, RANTES, HMGB1, TNF-α, IRF4, TLR7, CLA-DRB3.2, IL1B, IL6, CXCL8, S-LZ, and Cathelicidin), and antioxidant (SOD1, CAT, GPX1, GST, ATOX1, Nrf2 and AhpC/TSA) markers in ewes post-lambing and their newly born lambs. Additionally, mRNA levels of lipogenic (ACACA, FASN SCD, LPL, and BTN1A) markers were significantly up-regulated in lambs from supplemented ewes than control ones. There was a significant increase in the WBCs, Hb, RBc count, serum level of glucose, total protein, triacylglycerol and total cholesterol, GPx, catalase, IL1α and IL6 with significantly decreased serum level of TNF-α and MDA in supplemented ewes after lambing as compared with control ones. There was also a significant increase in WBCs, Hb, RBc count, birth weight and body temperature with significantly decreased in the serum levels of TNF-α and stillbirth of newly born lambs from supplemented ewes as compared to other lambs from control ones.
Collapse
Affiliation(s)
- Ahmed El-Sayed
- Department of Animal Health and Poultry, Animal and Poultry Production Division, Desert Research Center (DRC), Cairo, Egypt
| | - Eman Ebissy
- Department of Animal Health and Poultry, Animal and Poultry Production Division, Desert Research Center (DRC), Cairo, Egypt
| | - Ahmed Ateya
- Department of Development of Animal Wealth, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
3
|
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
|
4
|
Feng Y, Yang Z, Wang J, Zhao H. Cuproptosis: unveiling a new frontier in cancer biology and therapeutics. Cell Commun Signal 2024; 22:249. [PMID: 38693584 PMCID: PMC11064406 DOI: 10.1186/s12964-024-01625-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024] Open
Abstract
Copper plays vital roles in numerous cellular processes and its imbalance can lead to oxidative stress and dysfunction. Recent research has unveiled a unique form of copper-induced cell death, termed cuproptosis, which differs from known cell death mechanisms. This process involves the interaction of copper with lipoylated tricarboxylic acid cycle enzymes, causing protein aggregation and cell death. Recently, a growing number of studies have explored the link between cuproptosis and cancer development. This review comprehensively examines the systemic and cellular metabolism of copper, including tumor-related signaling pathways influenced by copper. It delves into the discovery and mechanisms of cuproptosis and its connection to various cancers. Additionally, the review suggests potential cancer treatments using copper ionophores that induce cuproptosis, in combination with small molecule drugs, for precision therapy in specific cancer types.
Collapse
Affiliation(s)
- Ying Feng
- Department of Emergency, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266005, Shandong, China
| | - Zhibo Yang
- Department of Neurosurgery, 3201 Hospital of Xi'an Jiaotong University Health Science Center, Hanzhong, 723000, Shaanxi, China
| | - Jianpeng Wang
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266005, Shandong, China
| | - Hai Zhao
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266005, Shandong, China.
| |
Collapse
|
5
|
Liu WQ, Lin WR, Yan L, Xu WH, Yang J. Copper homeostasis and cuproptosis in cancer immunity and therapy. Immunol Rev 2024; 321:211-227. [PMID: 37715546 DOI: 10.1111/imr.13276] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/17/2023]
Abstract
Copper is an essential nutrient for maintaining enzyme activity and transcription factor function. Excess copper results in the aggregation of lipoylated dihydrolipoamide S-acetyltransferase (DLAT), which correlates to the mitochondrial tricarboxylic acid (TCA) cycle, resulting in proteotoxic stress and eliciting a novel cell death modality: cuproptosis. Cuproptosis exerts an indispensable role in cancer progression, which is considered a promising strategy for cancer therapy. Cancer immunotherapy has gained extensive attention owing to breakthroughs in immune checkpoint blockade; furthermore, cuproptosis is strongly connected to the modulation of antitumor immunity. Thus, a thorough recognition concerning the mechanisms involved in the modulation of copper metabolism and cuproptosis may facilitate improvement in cancer management. This review outlines the cellular and molecular mechanisms and characteristics of cuproptosis and the links of the novel regulated cell death modality with human cancers. We also review the current knowledge on the complex effects of cuproptosis on antitumor immunity and immune response. Furthermore, potential agents that elicit cuproptosis pathways are summarized. Lastly, we discuss the influence of cuproptosis induction on the tumor microenvironment as well as the challenges of adding cuproptosis regulators to therapeutic strategies beyond traditional therapy.
Collapse
Affiliation(s)
- Wei-Qing Liu
- Department of Internal Medicine-Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wan-Rong Lin
- Department of Surgical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li Yan
- Department of Internal Medicine-Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wen-Hao Xu
- Department of Internal Medicine-Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jun Yang
- Department of Surgical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
6
|
Fodor I, Yañez-Guerra LA, Kiss B, Büki G, Pirger Z. Copper-transporting ATPases throughout the animal evolution - From clinics to basal neuron-less animals. Gene 2023; 885:147720. [PMID: 37597707 DOI: 10.1016/j.gene.2023.147720] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/01/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
Copper-transporting ATPases are a group of heavy metal-transporting proteins and which can be found in all living organisms. In animals, they are generally referred to as ATP7 proteins and are involved in many different physiological processes including the maintaining of copper homeostasis and the supply of copper to cuproenzymes. A single ATP7 gene is present in non-chordate animals while it is divided into ATP7A and ATP7B in chordates. In humans, dysfunction of ATP7 proteins can lead to severe genetic disorders, such as, Menkes disease and Wilson's disease, which are characterized by abnormal copper transport and accumulation, causing significant health complications. Therefore, there is a substantial amount of research on ATP7 genes and ATP7 proteins in humans and mice to understand pathophysiological conditions and find potential therapeutic interventions. Copper-transporting ATPases have also been investigated in some non-mammalian vertebrates, protostomes, single-cellular eukaryotes, prokaryotes, and archaea to gain useful evolutionary insights. However, ATP7 function in many animals has been somewhat neglected, particularly in non-bilaterians. Previous reviews on this topic only broadly summarized the available information on the function and evolution of ATP7 genes and ATP7 proteins and included only the classic vertebrate and invertebrate models. Given this, and the fact that a considerable amount of new information on this topic has been published in recent years, the present study was undertaken to provide an up-to-date, comprehensive summary of ATP7s/ATP7s and give new insights into their evolutionary relationships. Additionally, this work provides a framework for studying these genes and proteins in non-bilaterians. As early branching animals, they are important to understand the evolution of function of these proteins and their important role in copper homeostasis and neurotransmission.
Collapse
Affiliation(s)
- István Fodor
- Ecophysiological and Environmental Toxicological Research Group, Balaton Limnological Research Institute, H-8237 Tihany, Hungary.
| | | | - Bence Kiss
- Institute of Biochemistry and Medical Chemistry, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Gergely Büki
- Department of Medical Genetics, Medical School, University of Pécs, H-7624 Pécs, Hungary
| | - Zsolt Pirger
- Ecophysiological and Environmental Toxicological Research Group, Balaton Limnological Research Institute, H-8237 Tihany, Hungary
| |
Collapse
|
7
|
Kunkle DE, Skaar EP. Moving metals: How microbes deliver metal cofactors to metalloproteins. Mol Microbiol 2023; 120:547-554. [PMID: 37408317 PMCID: PMC10592388 DOI: 10.1111/mmi.15117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 07/07/2023]
Abstract
First row d-block metal ions serve as vital cofactors for numerous essential enzymes and are therefore required nutrients for all forms of life. Despite this requirement, excess free transition metals are toxic. Free metal ions participate in the production of noxious reactive oxygen species and mis-metalate metalloproteins, rendering enzymes catalytically inactive. Thus, bacteria require systems to ensure metalloproteins are properly loaded with cognate metal ions to maintain protein function, while avoiding metal-mediated cellular toxicity. In this perspective we summarize the current mechanistic understanding of bacterial metallocenter maturation with specific emphasis on metallochaperones; a group of specialized proteins that both shield metal ions from inadvertent reactions and distribute them to cognate target metalloproteins. We highlight several recent advances in the field that have implicated new classes of proteins in the distribution of metal ions within bacterial proteins, while speculating on the future of the field of bacterial metallobiology.
Collapse
Affiliation(s)
- Dillon E. Kunkle
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
8
|
Rivera PL, Li WT, Bhogal S, Mandell JB, Belayneh R, Hankins ML, Payne JT, Watters RJ, Weiss KR. Antioxidant 1 copper chaperone gene expression and copper levels in dog osteosarcoma patients. Vet Comp Oncol 2023; 21:559-564. [PMID: 37148200 PMCID: PMC11231990 DOI: 10.1111/vco.12903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/30/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023]
Abstract
Twenty-four dogs with OS underwent limb amputation. Serum, OS tumour, and normal bone were harvested at time of surgery. RNA was extracted and gene expression was performed using quantitative polymerase chain reaction (qPCR). Tissue and blood copper concentrations were also determined with spectrophotometry. Compared to bone, tumour samples had significantly higher expressions of antioxidant 1 copper chaperone (ATOX1, p = .0003). OS tumour copper levels were significantly higher than that of serum (p < .010) and bone (p = .038). Similar to our previous observations in mouse and human OS, dog OS demonstrates overexpression of genes that regulate copper metabolism (ATOX1), and subsequent copper levels. Dogs with OS may provide a robust comparative oncology platform for the further study of these factors, as well as potential pharmacologic interventions.
Collapse
Affiliation(s)
- Pedro L. Rivera
- Department of Surgery, Pittsburgh Veterinary Specialty & Emergency Center-BluePearl, Pittsburgh, Pennsylvania, USA
| | - William T. Li
- Musculoskeletal Oncology Laboratory, Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sumail Bhogal
- Musculoskeletal Oncology Laboratory, Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jonathan B. Mandell
- Musculoskeletal Oncology Laboratory, Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rebekah Belayneh
- Musculoskeletal Oncology Laboratory, Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Margaret L. Hankins
- Musculoskeletal Oncology Laboratory, Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John T. Payne
- Department of Surgery, Pittsburgh Veterinary Specialty & Emergency Center-BluePearl, Pittsburgh, Pennsylvania, USA
| | - Rebecca J. Watters
- Musculoskeletal Oncology Laboratory, Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kurt R. Weiss
- Musculoskeletal Oncology Laboratory, Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
9
|
Yang D, Xiao P, Qiu B, Yu HF, Teng CB. Copper chaperone antioxidant 1: multiple roles and a potential therapeutic target. J Mol Med (Berl) 2023; 101:527-542. [PMID: 37017692 DOI: 10.1007/s00109-023-02311-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/23/2023] [Accepted: 03/26/2023] [Indexed: 04/06/2023]
Abstract
Copper (Cu) was recently demonstrated to play a critical role in cellular physiological and biochemical processes, including energy production and maintenance, antioxidation and enzymatic activity, and signal transduction. Antioxidant 1 (ATOX1), a chaperone of Cu previously named human ATX1 homologue (HAH1), has been found to play an indispensable role in maintaining cellular Cu homeostasis, antioxidative stress, and transcriptional regulation. In the past decade, it has also been found to be involved in a variety of diseases, including numerous neurodegenerative diseases, cancers, and metabolic diseases. Recently, increasing evidence has revealed that ATOX1 is involved in the regulation of cell migration, proliferation, autophagy, DNA damage repair (DDR), and death, as well as in organism development and reproduction. This review summarizes recent advances in the research on the diverse physiological and cytological functions of ATOX1 and the underlying mechanisms of its action in human health and diseases. The potential of ATOX1 as a therapeutic target is also discussed. This review aims to pose unanswered questions related to ATOX1 biology and explore the potential use of ATOX1 as a therapeutic target.
Collapse
Affiliation(s)
- Dian Yang
- Animal Development Biology Laboratory, College of Life Science, Northeast Forestry University, Harbin, 150040, People's Republic of China
| | - Pengyu Xiao
- Animal Development Biology Laboratory, College of Life Science, Northeast Forestry University, Harbin, 150040, People's Republic of China
| | - Botao Qiu
- Animal Development Biology Laboratory, College of Life Science, Northeast Forestry University, Harbin, 150040, People's Republic of China
| | - Hai-Fan Yu
- Animal Development Biology Laboratory, College of Life Science, Northeast Forestry University, Harbin, 150040, People's Republic of China.
| | - Chun-Bo Teng
- Animal Development Biology Laboratory, College of Life Science, Northeast Forestry University, Harbin, 150040, People's Republic of China.
| |
Collapse
|
10
|
Camponeschi F, Banci L. Metal trafficking in the cell: Combining atomic resolution with cellular dimension. FEBS Lett 2023; 597:122-133. [PMID: 36285633 DOI: 10.1002/1873-3468.14524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 01/14/2023]
Abstract
Metals are widely present in biological systems as simple ions or complex cofactors, and are involved in a variety of processes essential for life. Their transport inside cells and insertion into the binding sites of the proteins that need metals to function occur through complex and selective pathways involving dedicated multiprotein machineries specifically and transiently interacting with each other, often sharing the coordination of metal ions and/or cofactors. The understanding of these machineries requires integrated approaches, ranging from bioinformatics to experimental investigations, possibly in the cellular context. In this review, we report two case studies where the use of integrated in vitro and in cellulo approaches is necessary to clarify at atomic resolution essential aspects of metal trafficking in cells.
Collapse
Affiliation(s)
- Francesca Camponeschi
- Magnetic Resonance Center CERM, University of Florence, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Florence, Italy
| | - Lucia Banci
- Magnetic Resonance Center CERM, University of Florence, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Florence, Italy.,Department of Chemistry, University of Florence, Italy
| |
Collapse
|
11
|
Schwartz R, Ruthstein S, Major DT. Copper coordination states affect the flexibility of copper Metallochaperone Atox1: Insights from molecular dynamics simulations. Protein Sci 2022; 31:e4464. [PMID: 36208051 PMCID: PMC9667823 DOI: 10.1002/pro.4464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/15/2022] [Accepted: 10/04/2022] [Indexed: 12/13/2022]
Abstract
Copper is an essential element in nature but in excess, it is toxic to the living cell. The human metallochaperone Atox1 participates in copper homeostasis and is responsible for copper transmission. In a previous multiscale simulation study, we noticed a change in the coordination state of the Cu(I) ion, from 4 bound cysteine residues to 3, in agreement with earlier studies. Here, we perform and analyze classical molecular dynamic simulations of various coordination states: 2, 3, and 4. The main observation is an increase in protein flexibility as a result of a decrease in the coordination state. In addition, we identified several populated conformations that correlate well with double electron-electron resonance distance distributions or an X-ray structure of Cu(I)-bound Atox1. We suggest that the increased flexibility might benefit the process of ion transmission between interacting proteins. Further experiments can scrutinize this hypothesis and shed additional light on the mechanism of action of Atox1.
Collapse
Affiliation(s)
- Renana Schwartz
- Department of Chemistry and Institute for Nanotechnology and Advanced MaterialsBar‐Ilan UniversityRamat‐GanIsrael
| | - Sharon Ruthstein
- Department of Chemistry and Institute for Nanotechnology and Advanced MaterialsBar‐Ilan UniversityRamat‐GanIsrael
| | - Dan Thomas Major
- Department of Chemistry and Institute for Nanotechnology and Advanced MaterialsBar‐Ilan UniversityRamat‐GanIsrael
| |
Collapse
|
12
|
Pham KLJ, Schmollinger S, Merchant SS, Strenkert D. Chlamydomonas ATX1 is essential for Cu distribution to multiple cupro-enzymes and maintenance of biomass in conditions demanding cupro-enzyme-dependent metabolic pathways. PLANT DIRECT 2022; 6:e383. [PMID: 35141461 PMCID: PMC8814560 DOI: 10.1002/pld3.383] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/13/2021] [Accepted: 01/10/2022] [Indexed: 06/01/2023]
Abstract
Copper (Cu) chaperones, of which yeast ATX1 is a prototype, are small proteins with a Cu(I) binding MxCxxC motif and are responsible for directing intracellular Cu toward specific client protein targets that use Cu as a cofactor. The Chlamydomonas reinhardtii ATX1 (CrATX1) was identified by its high sequence similarity with yeast ATX1. Like the yeast homologue, CrATX1 accumulates in iron-deficient cells (but is not impacted by other metal-deficiencies). N- and C-terminally YFP-ATX1 fusion proteins are distributed in the cytoplasm. Reverse genetic analysis using artificial microRNA (amiRNA) to generate lines with reduced CrATX1 abundance and CRISPR/Cpf1 to generate atx1 knockout lines validated a function for ATX1 in iron-poor cells, again reminiscent of yeast ATX1, most likely because of an impact on metalation of the multicopper oxidase FOX1, which is an important component in high-affinity iron uptake. We further identify other candidate ATX1 targets owing to reduced growth of atx1 mutant lines on guanine as a sole nitrogen source, which we attribute to loss of function of UOX1, encoding a urate oxidase, a cupro-enzyme involved in guanine assimilation. An impact of ATX1 on Cu distribution in atx1 mutants is strikingly evident by a reduced amount of intracellular Cu in all conditions probed in this work.
Collapse
Affiliation(s)
- Keegan L. J. Pham
- Department of Plant and Microbial BiologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Stefan Schmollinger
- California Institute for Quantitative BiosciencesUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Sabeeha S. Merchant
- Department of Plant and Microbial BiologyUniversity of CaliforniaBerkeleyCaliforniaUSA
- California Institute for Quantitative BiosciencesUniversity of CaliforniaBerkeleyCaliforniaUSA
- Department of Molecular & Cell BiologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Daniela Strenkert
- California Institute for Quantitative BiosciencesUniversity of CaliforniaBerkeleyCaliforniaUSA
| |
Collapse
|
13
|
Furukawa Y. A pathological link between dysregulated copper binding in Cu/Zn-superoxide dismutase and amyotrophic lateral sclerosis. J Clin Biochem Nutr 2022; 71:73-77. [PMID: 36213785 PMCID: PMC9519421 DOI: 10.3164/jcbn.22-42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/17/2022] [Indexed: 11/22/2022] Open
Abstract
Mutations in the gene coding Cu/Zn-superoxide dismutase (SOD1) are linked to a familial form of amyotrophic lateral sclerosis (ALS), and its pathological hallmark includes abnormal accumulation of mutant SOD1 proteins in spinal motorneurons. Mutant SOD1 proteins are considered to be susceptible to misfolding, resulting in the accumulation as oligomers/aggregates. While it remains obscure how and why SOD1 becomes misfolded under pathological conditions in vivo, the failure to bind a copper and zinc ion in SOD1 in vitro leads to the significant destabilization of its natively folded structure. Therefore, genetic and pharmacological attempts to promote the metal binding in mutant SOD1 could serve as an effective treatment of ALS. Here, I briefly review the copper and zinc binding process of SOD1 in vivo and discuss a copper chaperone for SOD1 as a potential target for developing ALS therapeutics.
Collapse
|
14
|
Copper in tumors and the use of copper-based compounds in cancer treatment. J Inorg Biochem 2021; 226:111634. [PMID: 34740035 DOI: 10.1016/j.jinorgbio.2021.111634] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022]
Abstract
Copper homeostasis is strictly regulated by protein transporters and chaperones, to allow its correct distribution and avoid uncontrolled redox reactions. Several studies address copper as involved in cancer development and spreading (epithelial to mesenchymal transition, angiogenesis). However, being endogenous and displaying a tremendous potential to generate free radicals, copper is a perfect candidate, once opportunely complexed, to be used as a drug in cancer therapy with low adverse effects. Copper ions can be modulated by the organic counterpart, after complexed to their metalcore, either in redox potential or geometry and consequently reactivity. During the last four decades, many copper complexes were studied regarding their reactivity toward cancer cells, and many of them could be a drug choice for phase II and III in cancer therapy. Also, there is promising evidence of using 64Cu in nanoparticles as radiopharmaceuticals for both positron emission tomography (PET) imaging and treatment of hypoxic tumors. However, few compounds have gone beyond testing in animal models, and none of them got the status of a drug for cancer chemotherapy. The main challenge is their solubility in physiological buffers and their different and non-predictable mechanism of action. Moreover, it is difficult to rationalize a structure-based activity for drug design and delivery. In this review, we describe the role of copper in cancer, the effects of copper-complexes on tumor cell death mechanisms, and point to the new copper complexes applicable as drugs, suggesting that they may represent at least one component of a multi-action combination in cancer therapy.
Collapse
|
15
|
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: 27] [Impact Index Per Article: 6.8] [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
|
16
|
Jastrzab R, Nowak M, Zabiszak M, Odani A, Kaczmarek MT. Significance and properties of the complex formation of phosphate and polyphosphate groups in particles present in living cells. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Zhang X, Blockhuys S, Devkota R, Pilon M, Wittung-Stafshede P. The Caenorhabditis elegans homolog of human copper chaperone Atox1, CUC-1, aids in distal tip cell migration. Biometals 2020; 33:147-157. [PMID: 32506305 PMCID: PMC7295847 DOI: 10.1007/s10534-020-00239-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/30/2020] [Indexed: 12/01/2022]
Abstract
Cell migration is a fundamental biological process involved in for example embryonic development, immune system and wound healing. Cell migration is also a key step in cancer metastasis and the human copper chaperone Atox1 was recently found to facilitate this process in breast cancer cells. To explore the role of the copper chaperone in other cell migration processes, we here investigated the putative involvement of an Atox1 homolog in Caenorhabditis elegans, CUC-1, in distal tip cell migration, which is a key process during the development of the C. elegans gonad. Using knock-out worms, in which the cuc-1 gene was removed by CRISPR-Cas9 technology, we probed life span, brood size, as well as distal tip cell migration in the absence or presence of supplemented copper. Upon scoring of gonads, we found that cuc-1 knock-out, but not wild-type, worms exhibited distal tip cell migration defects in approximately 10–15% of animals and, had a significantly reduced brood size. Importantly, the distal tip cell migration defect was rescued by a wild-type cuc-1 transgene provided to cuc-1 knock-out worms. The results obtained here for C. elegans CUC-1 imply that Atox1 homologs, in addition to their well-known cytoplasmic copper transport, may contribute to developmental cell migration processes.
Collapse
Affiliation(s)
- Xiaolu Zhang
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96, Gothenburg, Sweden
| | - Stéphanie Blockhuys
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96, Gothenburg, Sweden
| | - Ranjan Devkota
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41390, Gothenburg, Sweden
| | - Marc Pilon
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41390, Gothenburg, Sweden
| | - Pernilla Wittung-Stafshede
- Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96, Gothenburg, Sweden.
| |
Collapse
|
18
|
Liu T, Karlsen M, Karlberg AM, Redalen KR. Hypoxia imaging and theranostic potential of [ 64Cu][Cu(ATSM)] and ionic Cu(II) salts: a review of current evidence and discussion of the retention mechanisms. EJNMMI Res 2020; 10:33. [PMID: 32274601 PMCID: PMC7145880 DOI: 10.1186/s13550-020-00621-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 03/19/2020] [Indexed: 02/07/2023] Open
Abstract
Background Tumor hypoxia (low tissue oxygenation) is an adverse condition of the solid tumor environment, associated with malignant progression, radiotherapy resistance, and poor prognosis. One method to detect tumor hypoxia is by positron emission tomography (PET) with the tracer [64Cu][Cu-diacetyl-bis(N(4)-methylthiosemicarbazone)] ([64Cu][Cu(ATSM)]), as demonstrated in both preclinical and clinical studies. In addition, emerging studies suggest using [64Cu][Cu(ATSM)] for molecular radiotherapy, mainly due to the release of therapeutic Auger electrons from copper-64, making [64Cu][Cu(ATSM)] a “theranostic” agent. However, the radiocopper retention based on a metal-ligand dissociation mechanism under hypoxia has long been controversial. Recent studies using ionic Cu(II) salts as tracers have raised further questions on the original mechanism and proposed a potential role of copper itself in the tracer uptake. We have reviewed the evidence of using the copper radiopharmaceuticals [60/61/62/64Cu][Cu(ATSM)]/ionic copper salts for PET imaging of tumor hypoxia, their possible therapeutic applications, issues related to the metal-ligand dissociation mechanism, and possible explanations of copper trapping based on studies of the copper metabolism under hypoxia. Results We found that hypoxia selectivity of [64Cu][Cu(ATSM)] has been clearly demonstrated in both preclinical and clinical studies. Preclinical therapeutic studies in mice have also demonstrated promising results, recently reporting significant tumor volume reductions and improved survival in a dose-dependent manner. Cu(II)-[Cu(ATSM)] appears to be accumulated in regions with substantially higher CD133+ expression, a marker for cancer stem cells. This, combined with the reported requirement of copper for activation of the hypoxia inducible factor 1 (HIF-1), provides a possible explanation for the therapeutic effects of [64Cu][Cu(ATSM)]. Comparisons between [64Cu][Cu(ATSM)] and ionic Cu(II) salts have showed similar results in both imaging and therapeutic studies, supporting the argument for the central role of copper itself in the retention mechanism. Conclusions We found promising evidence of using copper-64 radiopharmaceuticals for both PET imaging and treatment of hypoxic tumors. The Cu(II)-[Cu(ATSM)] retention mechanism remains controversial and future mechanistic studies should be focused on understanding the role of copper itself in the hypoxic tumor metabolism.
Collapse
Affiliation(s)
- Tengzhi Liu
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491, Trondheim, Norway.,Department of Radiology and Nuclear Medicine, St. Olavs hospital, Trondheim University Hospital, Trondheim, Norway
| | - Morten Karlsen
- Department of Radiology and Nuclear Medicine, St. Olavs hospital, Trondheim University Hospital, Trondheim, Norway
| | - Anna Maria Karlberg
- Department of Radiology and Nuclear Medicine, St. Olavs hospital, Trondheim University Hospital, Trondheim, Norway.,Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kathrine Røe Redalen
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491, Trondheim, Norway.
| |
Collapse
|
19
|
Liu M, Yu W, Jin J, Ma M, An T, Nie Y, Teng CB. Copper promotes sheep pancreatic duct organoid growth by activation of an antioxidant protein 1-dependent MEK-ERK pathway. Am J Physiol Cell Physiol 2020; 318:C806-C816. [PMID: 32130071 DOI: 10.1152/ajpcell.00509.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Proper amounts of copper supplemented in livestock feed improve the physical growth and traits of farm animals. The pancreas is an important organ with both exocrine and endocrine portions. To investigate the role and mechanism of copper in the sheep pancreas, we first established sheep pancreatic duct organoids (sPDOs). We found that an appropriate amount of copper benefited the formation and growth of sPDOs, whereas excess or deficient copper damaged sPDOs. We found that the proliferation-stimulating effect of copper was related to the copper chaperone antioxidant protein 1 (ATOX1)-dependent activation of MEK-ERK1/2 signaling. Atox1 knockdown suppressed the cell proliferation of sPDOs, even in the presence of the MEK activator. These results indicate that moderate concentrations of copper promote sPDO growth through ATOX1-regulated cell proliferation by activation of MEK-ERK. Moreover, our study indicates that organoids may be a useful model to study organ growth mechanisms in livestock.
Collapse
Affiliation(s)
- Miao Liu
- Animal Development Biology Laboratory, College of Life Science, Northeast Forestry University, Harbin, China
| | - Wen Yu
- Animal Development Biology Laboratory, College of Life Science, Northeast Forestry University, Harbin, China
| | - Jing Jin
- Animal Development Biology Laboratory, College of Life Science, Northeast Forestry University, Harbin, China
| | - Mingjun Ma
- Animal Development Biology Laboratory, College of Life Science, Northeast Forestry University, Harbin, China
| | - Tiezhu An
- Animal Development Biology Laboratory, College of Life Science, Northeast Forestry University, Harbin, China
| | - Yuzhe Nie
- Animal Development Biology Laboratory, College of Life Science, Northeast Forestry University, Harbin, China.,Key Laboratory of Saline-alkali Vegetation Ecology Restoration, College of Life Science, Northeast Forestry University, Harbin, China
| | - Chun-Bo Teng
- Animal Development Biology Laboratory, College of Life Science, Northeast Forestry University, Harbin, China.,Key Laboratory of Saline-alkali Vegetation Ecology Restoration, College of Life Science, Northeast Forestry University, Harbin, China
| |
Collapse
|
20
|
Puchkova LV, Broggini M, Polishchuk EV, Ilyechova EY, Polishchuk RS. Silver Ions as a Tool for Understanding Different Aspects of Copper Metabolism. Nutrients 2019; 11:E1364. [PMID: 31213024 PMCID: PMC6627586 DOI: 10.3390/nu11061364] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/08/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022] Open
Abstract
In humans, copper is an important micronutrient because it is a cofactor of ubiquitous and brain-specific cuproenzymes, as well as a secondary messenger. Failure of the mechanisms supporting copper balance leads to the development of neurodegenerative, oncological, and other severe disorders, whose treatment requires a detailed understanding of copper metabolism. In the body, bioavailable copper exists in two stable oxidation states, Cu(I) and Cu(II), both of which are highly toxic. The toxicity of copper ions is usually overcome by coordinating them with a wide range of ligands. These include the active cuproenzyme centers, copper-binding protein motifs to ensure the safe delivery of copper to its physiological location, and participants in the Cu(I) ↔ Cu(II) redox cycle, in which cellular copper is stored. The use of modern experimental approaches has allowed the overall picture of copper turnover in the cells and the organism to be clarified. However, many aspects of this process remain poorly understood. Some of them can be found out using abiogenic silver ions (Ag(I)), which are isoelectronic to Cu(I). This review covers the physicochemical principles of the ability of Ag(I) to substitute for copper ions in transport proteins and cuproenzyme active sites, the effectiveness of using Ag(I) to study copper routes in the cells and the body, and the limitations associated with Ag(I) remaining stable in only one oxidation state. The use of Ag(I) to restrict copper transport to tumors and the consequences of large-scale use of silver nanoparticles for human health are also discussed.
Collapse
Affiliation(s)
- Ludmila V Puchkova
- Laboratory of Trace elements metabolism, ITMO University, Kronverksky av., 49, St.-Petersburg 197101, Russia.
- Department of Molecular Genetics, Research Institute of Experimental Medicine, Acad. Pavlov str., 12, St.-Petersburg 197376, Russia.
- Department of Biophysics, Peter the Great St. Petersburg Polytechnic University, Politekhnicheskaya str., 29, St.-Petersburg 195251, Russia.
| | - Massimo Broggini
- Laboratory of Trace elements metabolism, ITMO University, Kronverksky av., 49, St.-Petersburg 197101, Russia.
- Laboratory of molecular pharmacology, Istituto di Ricerche Farmacologiche "Mario Negri" IRCCS, Via La Masa, 19, Milan 20156, Italy.
| | - Elena V Polishchuk
- Laboratory of Trace elements metabolism, ITMO University, Kronverksky av., 49, St.-Petersburg 197101, Russia.
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (NA) 80078, Italy.
| | - Ekaterina Y Ilyechova
- Laboratory of Trace elements metabolism, ITMO University, Kronverksky av., 49, St.-Petersburg 197101, Russia.
| | - Roman S Polishchuk
- Telethon Institute of Genetics and Medicine, Via Campi Flegrei 34, Pozzuoli (NA) 80078, Italy.
| |
Collapse
|
21
|
Naletova I, Satriano C, Curci A, Margiotta N, Natile G, Arena G, La Mendola D, Nicoletti VG, Rizzarelli E. Cytotoxic phenanthroline derivatives alter metallostasis and redox homeostasis in neuroblastoma cells. Oncotarget 2018; 9:36289-36316. [PMID: 30555630 PMCID: PMC6284747 DOI: 10.18632/oncotarget.26346] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/01/2018] [Indexed: 02/06/2023] Open
Abstract
Copper homeostasis is generally investigated focusing on a single component of the metallostasis network. Here we address several of the factors controlling the metallostasis for neuroblastoma cells (SH-SY5Y) upon treatment with 2,9-dimethyl-1,10-phenanthroline-5,6-dione (phendione) and 2,9-dimethyl-1,10-phenanthroline (cuproindione). These compounds bind and transport copper inside cells, exert their cytotoxic activity through the induction of oxidative stress, causing apoptosis and alteration of the cellular redox and copper homeostasis network. The intracellular pathway ensured by copper transporters (Ctr1, ATP7A), chaperones (CCS, ATOX, COX 17, Sco1, Sco2), small molecules (GSH) and transcription factors (p53) is scrutinised.
Collapse
Affiliation(s)
- Irina Naletova
- Department of Chemical Sciences, University of Catania, Catania, Italy
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB), Bari, Italy
| | - Cristina Satriano
- Department of Chemical Sciences, University of Catania, Catania, Italy
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB), Bari, Italy
| | - Alessandra Curci
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB), Bari, Italy
- Department of Chemistry, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Nicola Margiotta
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB), Bari, Italy
- Department of Chemistry, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Giovanni Natile
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB), Bari, Italy
- Department of Chemistry, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Giuseppe Arena
- Department of Chemical Sciences, University of Catania, Catania, Italy
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB), Bari, Italy
| | - Diego La Mendola
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB), Bari, Italy
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Vincenzo Giuseppe Nicoletti
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB), Bari, Italy
- Section of Medical Biochemistry, Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Catania, Italy
| | - Enrico Rizzarelli
- Department of Chemical Sciences, University of Catania, Catania, Italy
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB), Bari, Italy
| |
Collapse
|
22
|
Baker ZN, Jett K, Boulet A, Hossain A, Cobine PA, Kim BE, El Zawily AM, Lee L, Tibbits GF, Petris MJ, Leary SC. The mitochondrial metallochaperone SCO1 maintains CTR1 at the plasma membrane to preserve copper homeostasis in the murine heart. Hum Mol Genet 2018; 26:4617-4628. [PMID: 28973536 PMCID: PMC5886179 DOI: 10.1093/hmg/ddx344] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/31/2017] [Indexed: 11/14/2022] Open
Abstract
SCO1 is a ubiquitously expressed, mitochondrial protein with essential roles in cytochrome c oxidase (COX) assembly and the regulation of copper homeostasis. SCO1 patients present with severe forms of early onset disease, and ultimately succumb from liver, heart or brain failure. However, the inherent susceptibility of these tissues to SCO1 mutations and the clinical heterogeneity observed across SCO1 pedigrees remain poorly understood phenomena. To further address this issue, we generated Sco1hrt/hrt and Sco1stm/stm mice in which Sco1 was specifically deleted in heart and striated muscle, respectively. Lethality was observed in both models due to a combined COX and copper deficiency that resulted in a dilated cardiomyopathy. Left ventricular dilation and loss of heart function was preceded by a temporal decrease in COX activity and copper levels in the longer-lived Sco1stm/stm mice. Interestingly, the reduction in copper content of Sco1stm/stm cardiomyocytes was due to the mislocalisation of CTR1, the high affinity transporter that imports copper into the cell. CTR1 was similarly mislocalized to the cytosol in the heart of knockin mice carrying a homozygous G115S substitution in Sco1, which in humans causes a hypertrophic cardiomyopathy. Our current findings in the heart are in marked contrast to our prior observations in the liver, where Sco1 deletion results in a near complete absence of CTR1 protein. These data collectively argue that mutations perturbing SCO1 function have tissue-specific consequences for the machinery that ultimately governs copper homeostasis, and further establish the importance of aberrant mitochondrial signaling to the etiology of copper handling disorders.
Collapse
Affiliation(s)
- Zakery N Baker
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Kimberly Jett
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Aren Boulet
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Amzad Hossain
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Paul A Cobine
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Byung-Eun Kim
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Amr M El Zawily
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Ling Lee
- Department of Cardiovascular Sciences, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Glen F Tibbits
- Department of Cardiovascular Sciences, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Michael J Petris
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Scot C Leary
- Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| |
Collapse
|
23
|
Ogórek M, Lenartowicz M, Starzyński R, Jończy A, Staroń R, Doniec A, Krzeptowski W, Bednarz A, Pierzchała O, Lipiński P, Rajfur Z, Baster Z, Gibas-Tybur P, Grzmil P. Atp7a and Atp7b regulate copper homeostasis in developing male germ cells in mice. Metallomics 2018; 9:1288-1303. [PMID: 28820536 DOI: 10.1039/c7mt00134g] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The maintenance of copper homeostasis is critical for all cells. As learned from mice with disturbed copper metabolism, this trace element is also important for spermatogenesis. The experiments conducted in yeasts have demonstrated that appropriate copper level must be preserved to enable meiosis progression; however, increased copper level is toxic for cells. This study aims to analyze the expression profile of Atp7a and Atp7b and other genes encoding copper-related proteins during spermatogenesis in mice. Using the transcripts and protein detection techniques, we demonstrate that within seminiferous tubuli, ATP7A is mainly present in early meiotic germ cells (leptotene to pachytene spermatocytes) and in Sertoli cells (SCs). During spermatogenesis, the progression Atp7a expression profile corresponds to Slc31a1 (encoding copper importer CTR1) and Atox1 (encoding chaperon protein, which delivers copper from CTR1 to ATP7A and ATP7B) expression, suggesting that male germ cells retrieve copper and ATP7A protects them from copper overdose. In contrast, ATP7B protein is observed in SCs and near elongated spermatids; thus, its function seems to be related to copper extraction during spermiogenesis. This is the first study to give a comprehensive view on the activity of copper-related genes during spermatogenesis in mice.
Collapse
Affiliation(s)
- Mateusz Ogórek
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University Kraków, Gronostajowa 9, 30-387 Kraków, Poland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Desferal regulates hCtr1 and transferrin receptor expression through Sp1 and exhibits synergistic cytotoxicity with platinum drugs in oxaliplatin-resistant human cervical cancer cells in vitro and in vivo. Oncotarget 2018; 7:49310-49321. [PMID: 27384479 PMCID: PMC5226510 DOI: 10.18632/oncotarget.10336] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/13/2016] [Indexed: 01/05/2023] Open
Abstract
The development of resistance to platinum drugs in cancer cells severely reduces the efficacy of these drugs. Thus, the discovery of novel drugs or combined strategies to overcome drug resistance is imperative. In addition to our previous finding that combined D-penicillamine with platinum drugs exerts synergistic cytotoxicity, we recently identified a novel therapeutic strategy by combining an iron chelating agent desferal with platinum drugs to overcome platinum resistance in an oxaliplatin-resistant human cervical cancer cell line, S3. Further study demonstrated that the level of platinum–DNA adduct formation positively correlated with cell death in combination of desferal with platinums than that of each drug alone in S3 cells. Decrement of human copper transporter 1 (hCtr1) and transferrin receptor 1 (TfR1) expression involved in the development of platinum resistance in S3 cells. Moreover, desferal promoted the expression of hCtr1 through the upregulation of Sp1. The overexpression of Sp1 increased the expression of NF-κB and translocated it into the nucleus to bind to the TfR1 promoter region, which subsequently increased the expression of TfR1. Importantly, the cotreatment of oxaliplatin with desferal significantly potentiated the oxaliplatin-elicited antitumoral effect in the oxaliplatin-resistant xenograft animal model without any toxic effect observed. Taken together, these results demonstrated that the combination of desferal with oxaliplatin can overcome oxaliplatin resistance through the regulation of hCtr1 and TfR1, and may have beneficial effect for treatment of patient with oxaliplatin-refractory tumors.
Collapse
|
25
|
Tarenzi T, Calandrini V, Potestio R, Giorgetti A, Carloni P. Open Boundary Simulations of Proteins and Their Hydration Shells by Hamiltonian Adaptive Resolution Scheme. J Chem Theory Comput 2017; 13:5647-5657. [PMID: 28992702 DOI: 10.1021/acs.jctc.7b00508] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The recently proposed Hamiltonian adaptive resolution scheme (H-AdResS) allows the performance of molecular simulations in an open boundary framework. It allows changing, on the fly, the resolution of specific subsets of molecules (usually the solvent), which are free to diffuse between the atomistic region and the coarse-grained reservoir. So far, the method has been successfully applied to pure liquids. Coupling the H-AdResS methodology to hybrid models of proteins, such as the molecular mechanics/coarse-grained (MM/CG) scheme, is a promising approach for rigorous calculations of ligand binding free energies in low-resolution protein models. Toward this goal, here we apply for the first time H-AdResS to two atomistic proteins in dual-resolution solvent, proving its ability to reproduce structural and dynamic properties of both the proteins and the solvent, as obtained from atomistic simulations.
Collapse
Affiliation(s)
- Thomas Tarenzi
- Computation-Based Science and Technology Research Center CaSToRC, The Cyprus Institute , 20 Konstantinou Kavafi Street, 2121, Aglantzia, Nicosia, Cyprus
- Department of Physics, Faculty of Mathematics, Computer Science and Natural Sciences, Aachen University , Otto-Blumenthal-Straße, 52074 Aachen, Germany
| | - Vania Calandrini
- Computational Biomedicine, Institute for Advanced Simulation IAS-5, and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich , 52425 Jülich, Germany
| | - Raffaello Potestio
- Max Planck Institute for Polymer Research , Ackermannweg 10, 55128 Mainz, Germany
| | - Alejandro Giorgetti
- Computational Biomedicine, Institute for Advanced Simulation IAS-5, and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich , 52425 Jülich, Germany
- Department of Biotechnology, University of Verona , Ca' Vignal 1, Strada Le Grazie 15, 37134 Verona, Italy
| | - Paolo Carloni
- Department of Physics, Faculty of Mathematics, Computer Science and Natural Sciences, Aachen University , Otto-Blumenthal-Straße, 52074 Aachen, Germany
- Computational Biomedicine, Institute for Advanced Simulation IAS-5, and Institute of Neuroscience and Medicine INM-9, Forschungszentrum Jülich , 52425 Jülich, Germany
| |
Collapse
|
26
|
Abstract
Alzheimer's disease (AD) is the most common form of adult neurode-generation and is characterised by progressive loss of cognitive function leading to death. The neuropathological hallmarks include extracellular amyloid plaque accumulation in affected regions of the brain, formation of intraneuronal neurofibrillary tangles, chronic neuroinflammation, oxidative stress, and abnormal biometal homeostasis. Of the latter, major changes in copper (Cu) levels and localisation have been identified in AD brain, with accumulation of Cu in amyloid deposits, together with deficiency of Cu in some brain regions. The amyloid precursor protein (APP) and the amyloid beta (Aβ) peptide both have Cu binding sites, and interaction with Cu can lead to potentially neurotoxic outcomes through generation of reactive oxygen species. In addition, AD patients have systemic changes to Cu metabolism, and altered Cu may also affect neuroinflammatory outcomes in AD. Although we still have much to learn about Cu homeostasis in AD patients and its role in disease aetiopathology, therapeutic approaches for regulating Cu levels and interactions with Cu-binding proteins in the brain are currently being developed. This review will examine how Cu is associated with pathological changes in the AD brain and how these may be targeted for therapeutic intervention.
Collapse
|
27
|
Changes in intracellular copper concentration and copper-regulating gene expression after PC12 differentiation into neurons. Sci Rep 2016; 6:33007. [PMID: 27623342 PMCID: PMC5020689 DOI: 10.1038/srep33007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/18/2016] [Indexed: 12/02/2022] Open
Abstract
It is suspected that some neurodegenerative diseases are a result of the disturbance of copper (Cu) homeostasis, although it remains unclear whether the disturbance of Cu homeostasis has aberrant effects on neurons. Herein, we investigated Cu metabolism specifically in neurons in terms of changes in the intracellular Cu concentration and the expression of Cu-regulating genes, such as Cu transporters and metallothioneins (MTs), before and after the differentiation of rat pheochromocytoma cells (PC12 cells) into neurons. After the differentiation, Cu and Zn imaging with fluorescent probes revealed an increase in intracellular Cu concentration. The concentrations of other essential metals, which were determined by an inductively coupled plasma mass spectrometer, were not altered. The mRNA expression of the Cu influx transporter, Ctr1, was decreased after the differentiation, and the differentiated cells acquired tolerance to Cu and cisplatin, another substrate of Ctr1. In addition, the expression of MT-3, a brain-specific isoform, was increased, contrary to the decreased expression of MT-1 and MT-2. Taken together, the differentiation of PC12 cells into neurons induced MT-3 expression, thereby resulting in intracellular Cu accumulation. The decrease in Ctr1 expression was assumed to be a response aimed at abolishing the physiological accumulation of Cu after the differentiation.
Collapse
|
28
|
Barresi V, Trovato-Salinaro A, Spampinato G, Musso N, Castorina S, Rizzarelli E, Condorelli DF. Transcriptome analysis of copper homeostasis genes reveals coordinated upregulation of SLC31A1,SCO1, and COX11 in colorectal cancer. FEBS Open Bio 2016; 6:794-806. [PMID: 27516958 PMCID: PMC4971835 DOI: 10.1002/2211-5463.12060] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 02/06/2016] [Accepted: 03/17/2016] [Indexed: 12/12/2022] Open
Abstract
Copper homeostasis and distribution is strictly regulated by a network of transporters and intracellular chaperones encoded by a group of genes collectively known as copper homeostasis genes (CHGs). In this work, analysis of The Cancer Genome Atlas database for somatic point mutations in colorectal cancer revealed that inactivating mutations are absent or extremely rare in CHGs. Using oligonucleotide microarrays, we found a strong increase in mRNA levels of the membrane copper transporter 1 protein [CTR1; encoded by the solute carrier family 31 member 1 gene (SLC31A1 gene)] in our series of colorectal carcinoma samples. CTR1 is the main copper influx transporter and changes in its expression are able to induce modifications of cellular copper accumulation. The increased SLC31A1 mRNA level is accompanied by a parallel increase in transcript levels for copper efflux pump ATP7A, copper metabolism Murr1 domain containing 1 (COMMD1), the cytochrome C oxidase assembly factors [synthesis of cytochrome c oxidase 1 (SCO1) and cytochrome c oxidase copper chaperone 11 (COX11)], the cupric reductase six transmembrane epithelial antigen of the prostate (STEAP3), and the metal‐regulatory transcription factors (MTF1, MTF2) and specificity protein 1 (SP1). The significant correlation between SLC31A1,SCO1, and COX11 mRNA levels suggests that this transcriptional upregulation might be part of a coordinated program of gene regulation. Transcript‐level upregulation of SLC31A1,SCO1, and COX11 was also confirmed by the analysis of different colon carcinoma cell lines (Caco‐2, HT116, HT29) and cancer cell lines of different tissue origin (MCF7, PC3). Finally, exon‐level expression analysis of SLC31A1 reveals differential expression of alternative transcripts in colorectal cancer and normal colonic mucosa.
Collapse
Affiliation(s)
- Vincenza Barresi
- Section of Medical Biochemistry Department of Biomedical and Biotechnological Sciences University of Catania Italy
| | - Angela Trovato-Salinaro
- Section of Medical Biochemistry Department of Biomedical and Biotechnological Sciences University of Catania Italy
| | - Giorgia Spampinato
- Section of Medical Biochemistry Department of Biomedical and Biotechnological Sciences University of Catania Italy
| | - Nicolò Musso
- Section of Medical Biochemistry Department of Biomedical and Biotechnological Sciences University of Catania Italy
| | - Sergio Castorina
- Section of Human Anatomy Department of Biomedical and Biotechnological Sciences University of Catania Italy
| | - Enrico Rizzarelli
- Institute of Biostructures and Bioimaging National Council of Research UOS Catania Italy
| | - Daniele Filippo Condorelli
- Section of Medical Biochemistry Department of Biomedical and Biotechnological Sciences University of Catania Italy
| |
Collapse
|
29
|
Belviso BD, Galliani A, Lasorsa A, Mirabelli V, Caliandro R, Arnesano F, Natile G. Oxaliplatin Binding to Human Copper Chaperone Atox1 and Protein Dimerization. Inorg Chem 2016; 55:6563-73. [DOI: 10.1021/acs.inorgchem.6b00750] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Benny D. Belviso
- Institute of Crystallography, Consiglio Nazionale delle Ricerche, via Amendola 122/o, 70126 Bari, Italy
| | - Angela Galliani
- Department
of Chemistry, University of Bari “A. Moro”, via E.
Orabona 4, 70125 Bari, Italy
| | - Alessia Lasorsa
- Department
of Chemistry, University of Bari “A. Moro”, via E.
Orabona 4, 70125 Bari, Italy
| | - Valentina Mirabelli
- Institute of Crystallography, Consiglio Nazionale delle Ricerche, via Amendola 122/o, 70126 Bari, Italy
- Department of Economics, University of Foggia, Via A. Gramsci 89/91, 71122 Foggia, Italy
| | - Rocco Caliandro
- Institute of Crystallography, Consiglio Nazionale delle Ricerche, via Amendola 122/o, 70126 Bari, Italy
| | - Fabio Arnesano
- Department
of Chemistry, University of Bari “A. Moro”, via E.
Orabona 4, 70125 Bari, Italy
| | - Giovanni Natile
- Department
of Chemistry, University of Bari “A. Moro”, via E.
Orabona 4, 70125 Bari, Italy
| |
Collapse
|
30
|
Ahn EH, Kim DW, Shin MJ, Ryu EJ, Yong JI, Chung SY, Cha HJ, Kim SJ, Choi YJ, Kim DS, Cho SW, Lee K, Cho YS, Kwon HY, Park J, Eum WS, Choi SY. Tat-ATOX1 inhibits streptozotocin-induced cell death in pancreatic RINm5F cells and attenuates diabetes in a mouse model. Int J Mol Med 2016; 38:217-24. [PMID: 27222268 DOI: 10.3892/ijmm.2016.2599] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 04/27/2016] [Indexed: 11/06/2022] Open
Abstract
Antioxidant 1 (ATOX1) functions as an antioxidant against hydrogen peroxide and superoxide, and therefore may play a significant role in many human diseases, including diabetes mellitus (DM). In the present study, we examined the protective effects of Tat-ATOX1 protein on streptozotocin (STZ)-exposed pancreatic insulinoma cells (RINm5F) and in a mouse model of STZ-induced diabetes using western blot analysis, immunofluorescence staining and MTT assay, as well as histological and biochemical analysis. Purified Tat-ATOX1 protein was efficiently transduced into RINm5F cells in a dose- and time-dependent manner. Additionally, Tat-ATOX1 protein markedly inhibited reactive oxygen species (ROS) production, DNA damage and the activation of Akt and mitogen activated protein kinases (MAPKs) in STZ-exposed RINm5F cells. In addition, Tat-ATOX1 protein transduced into mice pancreatic tissues and significantly decreased blood glucose and hemoglobin A1c (HbA1c) levels as well as the body weight changes in a model of STZ-induced diabetes. These results indicate that transduced Tat-ATOX1 protein protects pancreatic β-cells by inhibiting STZ-induced cellular toxicity in vitro and in vivo. Based on these findings, we suggest that Tat-ATOX1 protein has potential applications as a therapeutic agent for oxidative stress-induced diseases including DM.
Collapse
Affiliation(s)
- Eun Hee Ahn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, Gangwon-do 25457, Republic of Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| | - Eun Ji Ryu
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| | - Ji In Yong
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| | - Seok Young Chung
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| | - Hyun Ju Cha
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| | - Sang Jin Kim
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| | - Yeon Joo Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| | - Duk-Soo Kim
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan-Si, Chungnam 31538, Republic of Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Keunwook Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| | - Yoon Shin Cho
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| | - Hyeok Yil Kwon
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon‑do 24252, Republic of Korea
| |
Collapse
|
31
|
Canine Models for Copper Homeostasis Disorders. Int J Mol Sci 2016; 17:196. [PMID: 26861285 PMCID: PMC4783930 DOI: 10.3390/ijms17020196] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 01/21/2016] [Accepted: 01/25/2016] [Indexed: 12/23/2022] Open
Abstract
Copper is an essential trace nutrient metal involved in a multitude of cellular processes. Hereditary defects in copper metabolism result in disorders with a severe clinical course such as Wilson disease and Menkes disease. In Wilson disease, copper accumulation leads to liver cirrhosis and neurological impairments. A lack in genotype-phenotype correlation in Wilson disease points toward the influence of environmental factors or modifying genes. In a number of Non-Wilsonian forms of copper metabolism, the underlying genetic defects remain elusive. Several pure bred dog populations are affected with copper-associated hepatitis showing similarities to human copper metabolism disorders. Gene-mapping studies in these populations offer the opportunity to discover new genes involved in copper metabolism. Furthermore, due to the relatively large body size and long life-span of dogs they are excellent models for development of new treatment strategies. One example is the recent use of canine organoids for disease modeling and gene therapy of copper storage disease. This review addresses the opportunities offered by canine genetics for discovery of genes involved in copper metabolism disorders. Further, possibilities for the use of dogs in development of new treatment modalities for copper storage disorders, including gene repair in patient-derived hepatic organoids, are highlighted.
Collapse
|
32
|
Barresi V, Spampinato G, Musso N, Trovato Salinaro A, Rizzarelli E, Condorelli DF. ATOX1 gene silencing increases susceptibility to anticancer therapy based on copper ionophores or chelating drugs. J Inorg Biochem 2016; 156:145-52. [PMID: 26784148 DOI: 10.1016/j.jinorgbio.2016.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 12/28/2015] [Accepted: 01/07/2016] [Indexed: 11/17/2022]
Abstract
Copper is a catalytic cofactor required for the normal function of many enzymes involved in fundamental biological processes but highly cytotoxic when in excess. Therefore its homeostasis and distribution is strictly regulated by a network of transporters and intracellular chaperones. ATOX1 (antioxidant protein 1) is a copper chaperone that plays a role in copper homeostasis by binding and transporting cytosolic copper to ATPase proteins in the trans-Golgi network. In the present study the Caco-2 cell line, a colon carcinoma cell line, was used as an in vitro model to evaluate if ATOX1 deficiency could affect sensitivity to experimentally induced copper dyshomeostasis. Silencing of ATOX1 increased toxicity of a short treatment with a high concentration of Cu(2+). Copper ionophores, such as 5-chloro-8-hydroxyquinoline, induced a copper-dependent cell toxicity which was significantly potentiated after ATOX1 silencing. The copper chelator TPEN (N,N,N',N'-tetrakis (2-pyridylmethyl) ethylenediamine) produced a form of cell toxicity that was reversed by the addition of Cu(2+). ATOX1 silencing increased Caco-2 cell sensitivity to TPEN toxicity. Our results suggest the possibility of a therapy with copper-chelating or ionophore drugs in subtypes of tumors showing specific alterations in ATOX1 expression.
Collapse
Affiliation(s)
- Vincenza Barresi
- Department of Biomedical and Biotechnological Sciences, Unit of Medical Biochemistry, University of Catania, Italy
| | - Giorgia Spampinato
- Department of Biomedical and Biotechnological Sciences, Unit of Medical Biochemistry, University of Catania, Italy
| | - Nicolò Musso
- Department of Biomedical and Biotechnological Sciences, Unit of Medical Biochemistry, University of Catania, Italy
| | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, Unit of Medical Biochemistry, University of Catania, Italy
| | - Enrico Rizzarelli
- Institute of Biostructures and Bioimaging, National Council of Research, UOS Catania, Italy
| | - Daniele Filippo Condorelli
- Department of Biomedical and Biotechnological Sciences, Unit of Medical Biochemistry, University of Catania, Italy.
| |
Collapse
|
33
|
Urso E, Maffia M. Behind the Link between Copper and Angiogenesis: Established Mechanisms and an Overview on the Role of Vascular Copper Transport Systems. J Vasc Res 2015; 52:172-96. [PMID: 26484858 DOI: 10.1159/000438485] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 07/07/2015] [Indexed: 11/19/2022] Open
Abstract
Angiogenesis critically sustains the progression of both physiological and pathological processes. Copper behaves as an obligatory co-factor throughout the angiogenic signalling cascades, so much so that a deficiency causes neovascularization to abate. Moreover, the progress of several angiogenic pathologies (e.g. diabetes, cardiac hypertrophy and ischaemia) can be tracked by measuring serum copper levels, which are being increasingly investigated as a useful prognostic marker. Accordingly, the therapeutic modulation of body copper has been proven effective in rescuing the pathological angiogenic dysfunctions underlying several disease states. Vascular copper transport systems profoundly influence the activation and execution of angiogenesis, acting as multi-functional regulators of apparently discrete pro-angiogenic pathways. This review concerns the complex relationship among copper-dependent angiogenic factors, copper transporters and common pathological conditions, with an unusual accent on the multi-faceted involvement of the proteins handling vascular copper. Functions regulated by the major copper transport proteins (CTR1 importer, ATP7A efflux pump and metallo-chaperones) include the modulation of endothelial migration and vascular superoxide, known to activate angiogenesis within a narrow concentration range. The potential contribution of prion protein, a controversial regulator of copper homeostasis, is discussed, even though its angiogenic involvement seems to be mainly associated with the modulation of endothelial motility and permeability.
Collapse
Affiliation(s)
- Emanuela Urso
- Department of Biological and Environmental Science and Technologies, University of Salento, Lecce, Italy
| | | |
Collapse
|
34
|
Ronca F, Raggi A. Structure-function relationships in mammalian histidine-proline-rich glycoprotein. Biochimie 2015; 118:207-20. [PMID: 26409900 DOI: 10.1016/j.biochi.2015.09.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/22/2015] [Indexed: 02/01/2023]
Abstract
Histidine-proline-rich glycoprotein (HPRG), or histidine-rich glycoprotein (HRG), is a serum protein that is synthesized in the liver and is actively internalised by different cells, including skeletal muscle. The multidomain arrangement of HPRG comprises two modules at the N-terminus that are homologous to cystatin but void of cysteine proteinase inhibitor function, and a second half consisting of a histidine-proline-rich region (HPRR) located between two proline-rich regions (PRR1 and PRR2), and a C-terminus domain. HPRG has been reported to bind various ligands and to modulate angiogenesis via the histidine residues of the HPRR. However, the secondary structure prediction of the HPRR reveals that more than 98% is disordered and the structural basis of the hypothesized functions remains unclear. Comparison of the PRR1 of several mammalian species indicates the presence of a conserved binding site that might coordinate the Zn(2+) ion with an amino acid arrangement compatible with the cysteine-containing site that has been identified experimentally for rabbit HPRG. This observation provides a structural basis to the function of HPRG as an intracellular zinc chaperone which has been suggested by the involvement of the protein in the maintenance of the quaternary structure of skeletal muscle AMP deaminase (AMPD). During Anthropoidea evolution, a change of the primary structure of the PRR1 Zn(2+) binding site took place, giving rise to the sequence M-S-C-S/L-S/R-C that resembles the MxCxxC motif characteristic of metal transporters and metallochaperones.
Collapse
Affiliation(s)
- Francesca Ronca
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Antonio Raggi
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, Via Roma 55, 56126 Pisa, Italy.
| |
Collapse
|
35
|
Abstract
Fungal infections are responsible for millions of human deaths annually. Copper, an essential but toxic trace element, plays an important role at the host-pathogen axis during infection. In this review, we describe how the host uses either Cu compartmentalization within innate immune cells or Cu sequestration in other infected host niches such as in the brain to combat fungal infections. We explore Cu toxicity mechanisms and the Cu homeostasis machinery that fungal pathogens bring into play to succeed in establishing an infection. Finally, we address recent approaches that manipulate Cu-dependent processes at the host-pathogen axis for antifungal drug development.
Collapse
Affiliation(s)
| | - Dennis J Thiele
- From the Departments of Pharmacology & Cancer Biology and Biochemistry, Duke University, School of Medicine, Durham, North Carolina 27710
| |
Collapse
|
36
|
Mechanistic basis of a combination D-penicillamine and platinum drugs synergistically inhibits tumor growth in oxaliplatin-resistant human cervical cancer cells in vitro and in vivo. Biochem Pharmacol 2015; 95:28-37. [PMID: 25801007 DOI: 10.1016/j.bcp.2015.03.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 03/13/2015] [Indexed: 12/12/2022]
Abstract
The platinum-based regimen is the front-line treatment of chemotherapy. However, development of platinum resistance often causes therapeutic failure in this disease. We previously have generated an oxaliplatin-resistant subline, named S3, from human cervical carcinoma SiHa cells, and its resistant phenotype was well-characterized. In the present study, we aimed to identify the novel therapeutic strategy by combining copper chelator D-penicillamine with oxaliplatin, and to elucidate the underlying mechanisms for overcoming oxaliplatin resistance. As the result, D-penicillamine exerted synergistic killing effects only in S3 cells when combined with oxaliplatin and cisplatin by using Chou-Talalay method. Further study showed that the amounts of platinum DNA adduct formed were positively correlated to the percentage of cell death in S3 cells when co-treated D-penicillamine with oxaliplatin and cisplatin. D-penicillamine promoted copper influx transporter hCtr1 expression through upregulation of Sp1. Sp1 overexpression induced p53 translocation from nucleus to cytosol and caused p53 degradation through ubiquitination, which subsequently suppressed the expression of the copper efflux transporter ATP7A. Importantly, co-treatment of cisplatin with D-penicillamine enhanced oxaliplatin-elicited antitumor effect in the oxalipatin-resistant S3 xenograft tumors, but not found in SiHa xenograft model. Notably, Mice received D-penicillamine alone or in combination of D-penicillamine ad oxalipatin, increased hCtrl protein level in S3 xenograft tumor, however, the protein level of ATP7A was decreased. Taken together, this study provides insight into that the co-manipulation of hCtrl and ATP7A by D-penicillamine could increase the therapeutic efficacy of platinum drugs in oxaliplatin resistant tumors, especially in resistant phenotype with downexpression of hCtrl and overexpression of ATP7A.
Collapse
|
37
|
Kim SM, Hwang IK, Yoo DY, Eum WS, Kim DW, Shin MJ, Ahn EH, Jo HS, Ryu EJ, Yong JI, Cho SW, Kwon OS, Lee KW, Cho YS, Han KH, Park J, Choi SY. Tat-antioxidant 1 protects against stress-induced hippocampal HT-22 cells death and attenuate ischaemic insult in animal model. J Cell Mol Med 2015; 19:1333-45. [PMID: 25781353 PMCID: PMC4459847 DOI: 10.1111/jcmm.12513] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/14/2014] [Indexed: 11/30/2022] Open
Abstract
Oxidative stress-induced reactive oxygen species (ROS) are responsible for various neuronal diseases. Antioxidant 1 (Atox1) regulates copper homoeostasis and promotes cellular antioxidant defence against toxins generated by ROS. The roles of Atox1 protein in ischaemia, however, remain unclear. In this study, we generated a protein transduction domain fused Tat-Atox1 and examined the roles of Tat-Atox1 in oxidative stress-induced hippocampal HT-22 cell death and an ischaemic injury animal model. Tat-Atox1 effectively transduced into HT-22 cells and it protected cells against the effects of hydrogen peroxide (H2O2)-induced toxicity including increasing of ROS levels and DNA fragmentation. At the same time, Tat-Atox1 regulated cellular survival signalling such as p53, Bad/Bcl-2, Akt and mitogen-activate protein kinases (MAPKs). In the animal ischaemia model, transduced Tat-Atox1 protected against neuronal cell death in the hippocampal CA1 region. In addition, Tat-Atox1 significantly decreased the activation of astrocytes and microglia as well as lipid peroxidation in the CA1 region after ischaemic insult. Taken together, these results indicate that transduced Tat-Atox1 protects against oxidative stress-induced HT-22 cell death and against neuronal damage in animal ischaemia model. Therefore, we suggest that Tat-Atox1 has potential as a therapeutic agent for the treatment of oxidative stress-induced ischaemic damage.
Collapse
Affiliation(s)
- So Mi Kim
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Kangnung-Wonju National University, Kangneung, Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Eun Hee Ahn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Hyo Sang Jo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Eun Ji Ryu
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Ji In Yong
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, Korea
| | - Oh-Shin Kwon
- Department of Biochemistry, School of Life Sciences & Biotechnology, Kyungpook National University, Taegu, Korea
| | - Keun Wook Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Yoon Shin Cho
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Kyu Hyung Han
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chunchon, Korea
| |
Collapse
|
38
|
Ogra Y. Molecular mechanisms underlying copper homeostasis in Mammalian cells. Nihon Eiseigaku Zasshi 2015; 69:136-45. [PMID: 24858509 DOI: 10.1265/jjh.69.136] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Copper (Cu) is an essential metal for living organisms that utilize oxygen for respiration and is required as a cofactor of redox-regulating enzymes, such as superoxide dismutase, ceruloplasmin, lysyl oxidase, tyrosinase, and dopamine β-hydroxylase. However, the redox-active property of this metal may have toxic effects on cells due to the generation of harmful reactive oxygen species. Given these circumstances, it is said that cells have a dependable system for Cu homeostasis that efficiently distributes this essential metal to cuproenzymes, thereby preventing damage to proteins, nucleic acids, sugars, and lipids. In particular, influx, efflux, and intracellular distribution with maintenance of the oxidation state of Cu are strictly regulated. Several groups of Cu-regulating factors have been identified in mammalian cells, i.e., Cu transporters, Cu chaperones, Cu-binding proteins/peptides, and others. In this review, the features of the Cu-regulating factors are concisely examined in terms of molecular mechanisms underlying Cu homeostasis in cells.
Collapse
Affiliation(s)
- Yasumitsu Ogra
- Laboratory of Chemical Toxicology and Environmental Health, Showa Pharmaceutical University
| |
Collapse
|
39
|
Lenartowicz M, Kennedy C, Hayes H, McArdle HJ. Transcriptional regulation of copper metabolism genes in the liver of fetal and neonatal control and iron-deficient rats. Biometals 2014; 28:51-9. [PMID: 25349135 PMCID: PMC4300417 DOI: 10.1007/s10534-014-9802-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 10/16/2014] [Indexed: 12/25/2022]
Abstract
Copper and iron metabolism have been known to interact for many years. We have previously shown, during pregnancy, that copper levels in the maternal liver rise as a consequence of iron deficiency, but that levels in the fetal liver decrease. In this paper, we measure expression of genes involved in copper metabolism in fetal and postnatal liver, to test whether alterations can explain this observation. Additionally, we study the extent to which gene expression changes in the latter stages of pregnancy and in the perinatal period. Ctr1 expression levels dropped to term, rising again thereafter. There was no difference in gene expression between control and iron deficient animals. Atox1 expression remained approximately stable until term, and then there was a rise to a maximum at about Day 8. Atp7a expression levels remained constant, except for a brief drop at term. Atp7b levels, in contrast, decreased from a maximum early in gestation to low levels in the term and post-natal livers. Ceruloplasmin expression appeared to be diametrically opposite to Atp7b. The other two metallochaperones showed the same pattern of expression as Atox1, with a decrease to term, a rise at Day 1, or a rise after birth followed by a brief decrease at about Day 3. None of the genes were significantly affected by iron deficiency, suggesting that changes in expression cannot explain the altered copper levels in the fetal and neonatal liver.
Collapse
Affiliation(s)
- Malgorzata Lenartowicz
- Department of Genetics and Evolution, Institute of Zoology, Jagiellonian University, Gronostajowa 9, 30-387, Kraków, Poland
| | | | | | | |
Collapse
|
40
|
Balter V, Lamboux A, Zazzo A, Télouk P, Leverrier Y, Marvel J, Moloney AP, Monahan FJ, Schmidt O, Albarède F. Contrasting Cu, Fe, and Zn isotopic patterns in organs and body fluids of mice and sheep, with emphasis on cellular fractionation. Metallomics 2014; 5:1470-82. [PMID: 23963064 DOI: 10.1039/c3mt00151b] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We report Cu, Fe, and Zn natural isotope compositions in organs, body fluids, diets and feces of mice and sheep. Large and systematic isotope variability is observed, notably in the δ(66)Zn in liver and δ(65)Cu in kidneys, but significant differences exist between mice, sheep and humans, especially in the δ(66)Zn value of blood. The results are interpreted with reference to current knowledge of metal trafficking and redox conditions in cells. In general, the light isotopes preferentially fractionate into 'softer' bonds involving sulfur such as cysteine and glutathione, whereas heavy isotopes fractionate into 'harder' bonds involving nitrogen (histidine) and even more oxygen, notably hydroxides, phosphates, and carbonates. Bonds involving the reduced forms Cu(+) and Fe(2+) are enriched in the light isotopes relative to bonds involving the oxidized Cu(2+) and Fe(3+) forms. Differences in blood Zn isotope abundances between mice, sheep and humans may reflect a different prevalence of Zn ZIP transporters. The isotopically heavy Cu in the kidneys may reflect isotope fractionation during redox processes and may be relevant to ascorbate degradation into oxalate.
Collapse
Affiliation(s)
- Vincent Balter
- CNRS UMR 5276 "Laboratoire de Géologie de Lyon", Ecole Normale Supérieure. 46, Allée d'Italie, 69634 Lyon, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ranieri-Raggi M, Moir AJG, Raggi A. The role of histidine-proline-rich glycoprotein as zinc chaperone for skeletal muscle AMP deaminase. Biomolecules 2014; 4:474-97. [PMID: 24970226 PMCID: PMC4101493 DOI: 10.3390/biom4020474] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 04/08/2014] [Accepted: 04/10/2014] [Indexed: 11/19/2022] Open
Abstract
Metallochaperones function as intracellular shuttles for metal ions. At present, no evidence for the existence of any eukaryotic zinc-chaperone has been provided although metallochaperones could be critical for the physiological functions of Zn2+ metalloenzymes. We propose that the complex formed in skeletal muscle by the Zn2+ metalloenzyme AMP deaminase (AMPD) and the metal binding protein histidine-proline-rich glycoprotein (HPRG) acts in this manner. HPRG is a major plasma protein. Recent investigations have reported that skeletal muscle cells do not synthesize HPRG but instead actively internalize plasma HPRG. X-ray absorption spectroscopy (XAS) performed on fresh preparations of rabbit skeletal muscle AMPD provided evidence for a dinuclear zinc site in the enzyme compatible with a (μ-aqua)(μ-carboxylato)dizinc(II) core with two histidine residues at each metal site. XAS on HPRG isolated from the AMPD complex showed that zinc is bound to the protein in a dinuclear cluster where each Zn2+ ion is coordinated by three histidine and one heavier ligand, likely sulfur from cysteine. We describe the existence in mammalian HPRG of a specific zinc binding site distinct from the His-Pro-rich region. The participation of HPRG in the assembly and maintenance of skeletal muscle AMPD by acting as a zinc chaperone is also demonstrated.
Collapse
Affiliation(s)
- Maria Ranieri-Raggi
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, via Roma 55, Pisa 56126, Italy.
| | - Arthur J G Moir
- Department of Molecular Biology and Biotechnology, Krebs Institute, University of Sheffield, Sheffield S10 2UH, UK.
| | - Antonio Raggi
- Laboratory of Biochemistry, Department of Pathology, University of Pisa, via Roma 55, Pisa 56126, Italy.
| |
Collapse
|
42
|
Scheiber IF, Mercer JF, Dringen R. Metabolism and functions of copper in brain. Prog Neurobiol 2014; 116:33-57. [DOI: 10.1016/j.pneurobio.2014.01.002] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 01/08/2014] [Accepted: 01/08/2014] [Indexed: 12/15/2022]
|
43
|
|
44
|
Öhrvik H, Thiele DJ. The role of Ctr1 and Ctr2 in mammalian copper homeostasis and platinum-based chemotherapy. J Trace Elem Med Biol 2014; 31:178-82. [PMID: 24703712 PMCID: PMC4175275 DOI: 10.1016/j.jtemb.2014.03.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 03/13/2014] [Indexed: 12/31/2022]
Abstract
Copper (Cu) is an essential metal for growth and development that has the potential to be toxic if levels accumulate beyond the ability of cells to homeostatically balance uptake with detoxification. One system for Cu acquisition is the integral membrane Cu(+) transporter, Ctr1, which has been quite well characterized in terms of its function and physiology. The mammalian Ctr2 protein has been a conundrum for the copper field, as it is structurally closely related to the high affinity Cu transporter Ctr1, sharing important motifs for Cu transport activity. However, in contrast to mammalian Ctr1, Ctr2 fails to suppress the Cu-dependent growth phenotype of yeast cells defective in Cu(+) import, nor does it appreciably stimulate Cu acquisition when over-expressed in mammalian cells, underscoring important functional dissimilarities between the two proteins. Several roles for the mammalian Ctr2 have been suggested both in vitro and in vivo. Here, we summarize and discuss current insights into the Ctr2 protein and its interaction with Ctr1, its functions in mammalian Cu homeostasis and platinum-based chemotherapy.
Collapse
Affiliation(s)
- Helena Öhrvik
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Dennis J Thiele
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
45
|
Chaturvedi KS, Henderson JP. Pathogenic adaptations to host-derived antibacterial copper. Front Cell Infect Microbiol 2014; 4:3. [PMID: 24551598 PMCID: PMC3909829 DOI: 10.3389/fcimb.2014.00003] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 01/06/2014] [Indexed: 12/11/2022] Open
Abstract
Recent findings suggest that both host and pathogen manipulate copper content in infected host niches during infections. In this review, we summarize recent developments that implicate copper resistance as an important determinant of bacterial fitness at the host-pathogen interface. An essential mammalian nutrient, copper cycles between copper (I) (Cu(+)) in its reduced form and copper (II) (Cu(2+)) in its oxidized form under physiologic conditions. Cu(+) is significantly more bactericidal than Cu(2+) due to its ability to freely penetrate bacterial membranes and inactivate intracellular iron-sulfur clusters. Copper ions can also catalyze reactive oxygen species (ROS) generation, which may further contribute to their toxicity. Transporters, chaperones, redox proteins, receptors and transcription factors and even siderophores affect copper accumulation and distribution in both pathogenic microbes and their human hosts. This review will briefly cover evidence for copper as a mammalian antibacterial effector, the possible reasons for this toxicity, and pathogenic resistance mechanisms directed against it.
Collapse
Affiliation(s)
- Kaveri S Chaturvedi
- Division of Infectious Diseases, Department of Internal Medicine, Center for Women's Infectious Diseases Research, Washington University School of Medicine St. Louis, MO, USA
| | - Jeffrey P Henderson
- Division of Infectious Diseases, Department of Internal Medicine, Center for Women's Infectious Diseases Research, Washington University School of Medicine St. Louis, MO, USA
| |
Collapse
|
46
|
Copper and copper proteins in Parkinson's disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:147251. [PMID: 24672633 PMCID: PMC3941957 DOI: 10.1155/2014/147251] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 12/09/2013] [Indexed: 02/07/2023]
Abstract
Copper is a transition metal that has been linked to pathological and beneficial effects in neurodegenerative diseases. In Parkinson's disease, free copper is related to increased oxidative stress, alpha-synuclein oligomerization, and Lewy body formation. Decreased copper along with increased iron has been found in substantia nigra and caudate nucleus of Parkinson's disease patients. Copper influences iron content in the brain through ferroxidase ceruloplasmin activity; therefore decreased protein-bound copper in brain may enhance iron accumulation and the associated oxidative stress. The function of other copper-binding proteins such as Cu/Zn-SOD and metallothioneins is also beneficial to prevent neurodegeneration. Copper may regulate neurotransmission since it is released after neuronal stimulus and the metal is able to modulate the function of NMDA and GABA A receptors. Some of the proteins involved in copper transport are the transporters CTR1, ATP7A, and ATP7B and the chaperone ATOX1. There is limited information about the role of those biomolecules in the pathophysiology of Parkinson's disease; for instance, it is known that CTR1 is decreased in substantia nigra pars compacta in Parkinson's disease and that a mutation in ATP7B could be associated with Parkinson's disease. Regarding copper-related therapies, copper supplementation can represent a plausible alternative, while copper chelation may even aggravate the pathology.
Collapse
|
47
|
Flores AG, Unger VM. Atox1 contains positive residues that mediate membrane association and aid subsequent copper loading. J Membr Biol 2013; 246:903-13. [PMID: 24036897 DOI: 10.1007/s00232-013-9592-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 08/23/2013] [Indexed: 12/20/2022]
Abstract
Copper chaperones bind intracellular copper and ensure proper trafficking to downstream targets via protein-protein interactions. In contrast to the mechanisms of copper binding and transfer to downstream targets, the mechanisms of initial copper loading of the chaperones are largely unknown. Here, we demonstrate that antioxidant protein 1 (Atox1 in human cells), the principal cellular copper chaperone responsible for delivery of copper to the secretory pathway, possesses the ability to interact with negatively charged lipid headgroups via distinct surface lysine residues. Moreover, loss of these residues lowers the efficiency of copper loading of Atox1 in vivo, suggesting that the membrane may play a scaffolding role in copper distribution to Atox1. These findings complement the recent discovery that the membrane also facilitates copper loading of the copper chaperone for superoxide dismutase 1 and provide further support for the emerging paradigm that the membrane bilayer plays a central role in cellular copper acquisition and distribution.
Collapse
Affiliation(s)
- Adrian G Flores
- Department of Molecular Biosciences and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | | |
Collapse
|
48
|
Ayub A, Ashfaq UA, Haque A. HBV induced HCC: major risk factors from genetic to molecular level. BIOMED RESEARCH INTERNATIONAL 2013; 2013:810461. [PMID: 23991421 PMCID: PMC3749539 DOI: 10.1155/2013/810461] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 07/09/2013] [Indexed: 12/15/2022]
Abstract
Hepatocellular carcinoma (HCC) is a deadly and emerging disease leading to death in Asian countries. High hepatitis B virus (HBV) load and chronic hepatitis B (CHB) infection increase the risk of developing HCC. HBV is a DNA virus that can integrate DNA into host genome thereby increase the yield of transactivator protein HBxAg that may deregulate many pathways involving in metabolism of cells. Several monogenic and polygenic risk factors are also involved in HCC development. This review summarizes the mechanism involved in HCC development and discusses some promising therapies to make HCC curative.
Collapse
Affiliation(s)
- Ambreen Ayub
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad 38000, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad 38000, Pakistan
| | - Asma Haque
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad (GCUF), Faisalabad 38000, Pakistan
| |
Collapse
|
49
|
CAI HUAWEI, PENG FANGYU. Knockdown of copper chaperone antioxidant-1 by RNA interference inhibits copper-stimulated proliferation of non-small cell lung carcinoma cells. Oncol Rep 2013; 30:269-275. [PMID: 23624903 PMCID: PMC3729205 DOI: 10.3892/or.2013.2436] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 03/26/2013] [Indexed: 12/11/2022] Open
Abstract
Copper is required for cell proliferation and tumor angiogenesis. Cellular copper metabolism is regulated by a network of copper transporters and chaperones. Antioxidant-1 (ATOX1) is a cytosolic copper chaperone important for intracellular copper transport, which plays a role in the regulation of cell proliferation by functioning as a transcription factor in cell growth signal-transduction pathways. The present study aimed to explore the role of ATOX1 in the copper-related regulation of lung cancer cell proliferation by immunohistochemical (IHC) analysis of ATOX1 expression in non-small cell lung cancer (NSCLC) tissue samples and by assessing the effects of RNA interference (RNAi)-mediated knockdown of ATOX1 on copper-stimulated proliferation of NSCLC cells. Overexpression of ATOX1 was detected in NSCLC by IHC analysis of the tissue samples from patients diagnosed with NSCLC when compared with expression of ATOX1 in non-malignant lung tissue samples. Knockdown of ATOX1 in the NSCLC cells transduced by a lentiviral vector encoding short hairpin RNA (shRNA) specific for ATOX1 was associated with reduction in copper-stimulated cell proliferation. These findings suggest that ATOX1 plays an important role in copper-stimulated proliferation of NSCLC cells and ATOX1 holds potential as a therapeutic target for lung cancer therapy targeting copper metabolism.
Collapse
Affiliation(s)
- HUAWEI CAI
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8542, USA
| | - FANGYU PENG
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390-8542, USA
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390-8542, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390-8542, USA
| |
Collapse
|
50
|
Davies KM, Hare DJ, Cottam V, Chen N, Hilgers L, Halliday G, Mercer JFB, Double KL. Localization of copper and copper transporters in the human brain. Metallomics 2013; 5:43-51. [PMID: 23076575 DOI: 10.1039/c2mt20151h] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Disturbances in brain copper result in rare and severe neurological disorders and may play a role in the pathogenesis and progression of multiple neurodegenerative diseases. Our current understanding of mammalian brain copper transport is based on model systems outside the central nervous system and no data are available regarding copper transport systems in the human brain. To address this deficit, we quantified regional copper concentrations and examined the distribution and cellular localization of the copper transport proteins Copper transporter 1, Atox1, ATP7A, and ATP7B in multiple regions of the human brain using inductively coupled plasma-mass spectrometry, Western blot and immunohistochemistry. We identified significant relationships between copper transporter levels and brain copper concentrations, supporting a role for these proteins in copper transport in the human brain. Interestingly, the substantia nigra contained twice as much copper than that in other brain regions, suggesting an important role for copper in this brain region. Furthermore, ATP7A levels were significantly greater in the cerebellum, compared with other brain regions, supporting an important role for ATP7A in cerebellar neuronal health. This study provides novel data regarding copper regulation in the human brain, critical to understand the mechanisms by which brain copper levels can be altered, leading to neurological disease.
Collapse
Affiliation(s)
- Katherine M Davies
- Neuroscience Research Australia and The University of New South Wales, Randwick, NSW 2031, Australia.
| | | | | | | | | | | | | | | |
Collapse
|